# About Name: BugSpeaks® Description: BugSpeaks®, developed by Leucine Rich Bio Pvt Ltd, South Asia’s first microbiome company, is headquartered in Bengaluru, India. Since 2014, the company has pioneered advanced analytics to analyze complex genomics data. Collaborating with leading research institutes globally, Leucine Rich Bio has leveraged its expertise to create BugSpeaks®, South Asia’s first gut microbiome test. URL: https://www.bugspeaks.com/blog # Navigation Menu - Home: https://www.bugspeaks.com/ - Test: https://www.bugspeaks.com/test/ - RychBiome: https://www.bugspeaks.com/rychbiome-probiotic-capsules-gut-health/ - Company: # - Our Science: https://www.bugspeaks.com/science/ - About Us: https://www.bugspeaks.com/about-us/ - Experts: https://www.bugspeaks.com/experts/ - Blogs: https://www.bugspeaks.com/blog/ - Contact Us: https://www.bugspeaks.com/contact-us/ - Shop: https://www.bugspeaks.com/shop/ - login: https://www.bugspeaks.com/login/ # Blog Posts ## How to Restore Gut Health After Antibiotics: Causes, Effects & Proven Tips Author: BugSpeaks Published: 2025-04-18 Category: Gut Health Meta Title: How to Restore Gut Health After Antibiotics: Causes, Effects & Proven Recovery Tips Meta Description: Discover how antibiotics disrupt your gut microbiome and learn science-backed ways to restore gut health, prevent dysbiosis, and fight antibiotic resistance. Tags: Antibiotics, AMR, Gut Health, Gut Dysbiosis URL: https://www.bugspeaks.com/blog/restore-gut-health-after-antibiotics-causes-effects-tips Just as an ocean nurtures marine life, human gut harbours trillions of microorganisms that form a complex ecosystem. The microbiome starts to develop before birth during foetal development and continues to diversify and establish its richness as one grows, thus playing a prominent role in shaping our unique existence. The past decade has witnessed significant research in the field of the impact of the gut microbiome on health, and its role in many chronic and inflammatory diseases. Several studies emphasise the importance of restoring gut imbalances for better management of overall health and improving one’s quality of life. Alongside restoring gut health, researchers across the globe have also studied various environmental, physical, chemical and biological factors that could lead to gut imbalances and poor microbial diversities among individuals. One of these being antibiotics. Antibiotics, which are common and often life-saving drugs, can also have the adverse effects of disturbing the gut microbiome and giving rise to antibiotic resistant bacteria. This has led to the problem of antimicrobial resistance, now a crucial field to be studied by the next generation of researchers. The Journey of Antibiotics -------------------------- In 1928, Alexander Fleming returned to his laboratory after time away. While looking through old experiments, he discovered that a particular mold was able to kill all the bacteria around it on an old petri plate. He went on to find that this secretion from the mold was capable of destroying multiple bacterial pathogens. He named the killing compound penicillin, after the mold itself- Pencillium notatum. This breakthrough revolutionised medicine at the time, eventually saving countless patients from death by infection, and paved the path for the discovery of hundreds of antibiotics in the 21st century. But as time passed, and more antibiotics entered therapeutic circulation, the dark side of this practice emerged. Overusage, improper administration, lack of disposal awareness, and self-prescription of antibiotics have few immediate effects, but introduce higher and higher levels of antibiotics into our bodies and surroundings. Besides this, antibiotics are now extensively used in agriculture and dairy sectors to ensure the health of animals and to prevent crop failures. Due to the persistence of these antibiotics in the environment, established through several routes, the bacteria too evolved- developing various methods of survival, known now as “resistance”. Today many bacterial strains like E. coli, Staphylococcus aureus, Enterococcus have become resistant to common antibiotics resulting in recurrent infections in the blood stream, lungs, urinary tract (UTI), and causing diabetic ulcers and wounds in individuals, which are difficult to treat due to a lack of effective options and sometimes fatal.  Since the human gut affects nearly every metabolic process that occurs in the body, gut health is of utmost importance. The naturally occurring diversity of gut microbes is frequently disrupted by the inappropriate use of antibiotics, leading to the emergence of difficult-to-treat resistant and dangerous bacterial strains, as well as a disturbance in the ecology of the gut itself. Antibiotic associated Gut Dysbiosis ----------------------------------- Today, the silent pandemic of AMR (antimicrobial resistance) remains massively consequential on systemic and personal scales. Vulnerable populations like neonates, elderly, chronically ill, and immunocompromised individuals are under increasing risk of infection and comorbidities. On an individual level, short-term antibiotic use can cause diarrhoea and infections, while long-term effects include allergies and obesity. Studies reveal that early life exposure to low-dose antibiotics can have significant long-term effects on gut health. Antibiotics can disrupt the intestinal ecosystem, promoting conditions favourable for growth of harmful bacteria. [It has been](https://www.sciencedirect.com/science/article/pii/S2212877820300491?via%3Dihub) shown that overuse of antibiotics could disrupt individual immune responses resulting in metabolic disorders. Developments in genomics, metabolomics, and bioinformatics are rapidly changing the way the gut microbiome can be profiled, and how its interactions with other bodily systems can be understood and established. The influence of external factors on the gut microbiome is varied, therefore the reaction of this entity to antibiotics is expected to vary with the type of drug used, its concentration, and the route by which it enters the body.  Several [studies](https://doi.org/10.1080/19490976.2020.1870402) have been undertaken to identify the changes caused by different ubiquitously used and prescribed antibiotics to the gut microbiome. Azithromycin (a macrolide class of antibiotic) is known for its significant influence on the microbial diversity and richness of the gut microbiome. Macrolides, the structural class of antibiotics it belongs to, disrupt the microbiome longer, and cause a decrease in its diversity. The analysis of Penicillin antibiotic class also revealed that exposure reduced the number of beneficial gut bacteria, including Lactobacillus and Bifidobacteria. These beneficial bacteria are part of the healthy gut flora, and help in fibre digestion, produce anti-inflammatory molecules, promote immune responses to fight off infections, and produce short chain fatty acids that help to maintain gut membrane integrity and prevent leaky gut. Amoxicillin, a penicillin class antibiotic, negatively impacts the levels of Bacteroides fragilis and the beneficial Bifidobacteria in infants. A similar effect is seen in children exposed to Ampicillin (penicillin class) and Gentamycin (aminoglycoside class) where gut commensals Bifidobacteria and Lactobacillus are depleted, instead increasing others like Proteobacteria and Enterobacterisceae, and the genus Clostridium, of which several pathogens are part. ### A few prevalent conditions that are linked to intestinal dysbiosis include: * Increased risk of infections- gut dysbiosis often leads to growth of pathogenic bacteria like Clostridium difficile that causes recurrent colon infections that might at times lead to life threatening conditions. * Metabolic disorders- Dysbiosis often leads to altered metabolism, increased fat storage, and tissue inflammation that results in obesity, diabetes and atherosclerosis. * Allergic conditions- Dysbiosis leads to poor immune system responses, predisposing individuals to allergic responses. * Inflammatory diseases- Dysbiosis often leads to leaky gut and intestinal inflammation leading to various inflammatory diseases like IBD and autoimmune diseases. * Neurological effects- Recent studies show that gut imbalances affect gut-brain axis bidirectional communications and functioning. * Impact on drug efficacy- Dysbiosis can alter the metabolism of drugs, affecting their efficacy and safety. For instance, [antibiotics can change the gut microbiome](https://www.bugspeaks.com/blog/antibiotics:-clearing-the-infection-or-clearing-out-your-gut-navigating-the-impact-on-gut-health), which may influence the response to other medications, including cancer therapies Combatting antimicrobial resistance and related dysbiosis- you and the world ---------------------------------------------------------------------------- In the field of AMR, prevention is certainly better than cure. The biggest driver of prevention is adequate awareness, both for practitioners and the general public. Healthcare providers need to be cautious about prescribing antibiotics especially for non-serious infections to prevent unnecessary exposure. This includes evaluating the necessity of antibiotics and considering alternative treatments when appropriate to reduce the risk of metabolic disorders and developing resistance later in life. Parents must also be careful to adhere to prescribed courses, and must avoid self-prescribing antibiotics without clinical consultation. ### Individual interventions for antibiotic-related dysbiosis: Dietary interventions- Following a holistic lifestyle and including prebiotic and probiotic rich foods post antibiotic treatments could help in restoring the gut imbalances. Fecal Microbiota Transplantation (FMT) – FMT along with probiotic supplementation is one of the most researched approaches to restore gut imbalances. As the name suggests, the healthy gut flora from a stool sample of a suitable donor is transferred to the individual who is in need of it. Though in its infancy, further research in this direction could pave the way for more efficient therapies for gut dysbiosis. ### Systemic changes to reduce antibiotic prevalence: Waste treatment- [Since](https://www.sciencedirect.com/science/article/pii/S2213716523000851) wastewater and sewage plants are hotbeds of bacteria, proper decontamination and antibiotic removal must be ensured to reduce bioaccumulation. Hospital protocols- [Hospitals](https://pmc.ncbi.nlm.nih.gov/articles/PMC4420185/#:~:text=Antimicrobial%20resistance%20rates%20are%20highest,and%20nasogastric%20tubes%2C%20urinary%20catheters%2C) are a common site of acquiring resistance due to shared equipment, care givers, and a large flux of vulnerable populations. Proper protocols must be put in place to avoid the transmission of resistant infections. Public health policies- Public health campaigns can concentrate on informing the public about the possible dangers of excessive antibiotic usage. Campaigns could raise awareness of the importance of the gut microbiota to health and promote the judicious use of antibiotics. Control use of Antibiotics in Animal Feed- Antibiotics reach humans not only through direct intake but also via bioaccumulation in food. Work must be done to reduce the dependence of the agriculture industry on feed antibiotics, such as improving access to vaccines, clean water, and adequate information. ### Future Perspectives Antibiotics, though indispensable in modern medicine today, have serious ramifications on an individual and collective, even worldwide level. While it is getting easier to understand the impact of individual antibiotics on the human body and the gut microbiome, the underlying takeaway is this- overuse of antibiotics can lead to gut dysbiosis and the development of resistance which leads to both ineffectiveness of a particular drug, and the development of hard to kill pathogens. Scaling this problem across various intersecting social, political, and economic layers, AMR worldwide must be controlled and reduced through robust regulations, international cooperation, and widespread awareness. [Research](https://www.sciencedirect.com/science/article/pii/S2949916X24000343) for alternative methods of handling infections is a rapidly booming area, from the discovery of novel antibiotics, to combination therapy, immunomodulatory routes, drug repurposing and phage steering. This science must also translate from the bench to the field, requiring collaboration between academia, authority, and the industry. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Sample Page Author: BugSpeaks Published: 2025-03-01 URL: https://www.bugspeaks.com/blog/sample-page This is a page. Notice how there are no elements like author, date, social sharing icons? Yes, this is the page format. You can create a whole website using Superblog if you wish to do so! --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Antibiotics: Clearing the Infection or Clearing Out Your Gut? Navigating the Impact on Gut Health Author: BugSpeaks Published: 2024-04-10 Category: Microbiome and Disease Meta Title: Antibiotics: Clearing the Infection or Clearing Out Your Gut? Navigating the Impact on Gut Health Meta Description:
Antibiotics revolutionized the fight against infectious diseases, altering the landscape of modern medicine. Their disco URL: https://www.bugspeaks.com/blog/antibiotics:-clearing-the-infection-or-clearing-out-your-gut-navigating-the-impact-on-gut-health Antibiotics revolutionized the fight against infectious diseases, altering the landscape of modern medicine. Their discovery, which provided a powerful weapon against bacterial illnesses that once posed serious dangers to human life, represented a turning point in the history of healthcare. Antibiotics have made diseases such as pneumonia, TB, and many others more treatable, lowering mortality rates and improving public health worldwide. The capacity to rapidly eliminate bacterial pathogens and efficiently cure infections transformed medical procedures, resulting in notable improvements in patient care and results. The widespread use of antibiotics raises new concerns, including antibiotic resistance as bacteria develop defenses against them. Additionally, antibiotics can harm the gut flora, disrupting its delicate balance. This ecosystem in our digestive tract plays a crucial role in immune function, food absorption, and digestion, affecting overall health. Broad-spectrum antibiotics, while targeting harmful bacteria, can also disturb the gut microbiome, potentially impacting long-term human health.  **Impact of Antibiotics on Gut Health** **On Mothers and Baby:** Antibiotics during pregnancy and breastfeeding can harm both mother and baby by upsetting the balance of beneficial gut bacteria. Research suggests that these medications can alter the baby's gut flora, potentially leading to digestive issues like colitis later in life. Additionally, administering antibiotics during childbirth can compromise the baby's immune system and overall health. Antibiotics are frequently given to premature babies in the NICU because they are more susceptible to contracting infections. However, administering these strong antibiotics can alter the variety of beneficial bacteria in the baby's gut, making it less diverse and reducing the number of different types of bacteria. Also, this may increase the bacteria's resistance to drugs, which could be dangerous. These alterations in gut flora have been connected to severe health issues in preterm infants, such as necrotizing enterocolitis, a dangerous intestinal disorder, and late-onset sepsis, a form of infection. Other than that, it can increase Enterococcus and decrease beneficial bacteria like Bifidobacteria and Bacteroidetes. **On Adults:** The use of antibiotics in adults can cause notable disruptions in the makeup of the gut microbiota, which lowers the diversity and richness of microbial communities. It has been demonstrated that even brief antibiotic courses have long-lasting impacts on the gut microbiota, with the establishment of antibiotic-resistant strains and an incomplete restoration of the microbial makeup. Furthermore, different antibiotics have distinct effects on the gut microbiota; bacteriostatic medications encourage the growth of Gram-negative bacteria, whereas bactericidal medications encourage the growth of Gram-positive bacteria. Overall, indiscriminate antibiotic use across life stages offers severe health risks to humans by altering the delicate balance of the microbiota and encouraging the emergence of antibiotic resistance. **Clinical Consequences of Antibiotic:** **Short-Term Consequences:** Excessive antibiotic use can lead to immediate consequences like antibiotic-associated diarrhea (AAD), caused by disrupting the balance of microorganisms in the stomach. This disruption can persist after stopping the medication, potentially leading to Clostridium difficile-associated diarrhea (CDAD), a more serious condition that can be harmful, especially in elderly individuals with weakened immune systems. **Long-Term Consequences:** Misusing antibiotics, especially in infancy and childhood, can have significant long-term health consequences. It disrupts the gut microbiota, increasing the risk of obesity, asthma, allergies, and inflammatory bowel disease (IBD) later in life. Studies show that children given antibiotics at a young age are more likely to develop these illnesses compared to those who are not. **Effect of antibiotic-induced gut alternation on Immune and Metabolic Health:** Infection Susceptibility- Changes in the gut microbiota enhance infection susceptibility, which is a major concern. An imbalance in the gut's bacterial population can increase the risk of intestinal illnesses. This may occur as a result of opportunistic organisms becoming more aggressive or dangerous pathogens taking control. For instance, diarrhea brought on by antibiotics, which is frequently brought on by Clostridium difficile, can get severe and recurrent. Changes in the gut flora can also make bloodstream infections more common in people with compromised immunity. Premature infants, who are frequently given antibiotics, are more vulnerable to sepsis, a life-threatening illness linked to changes in their gut microbes. These dangers emphasize how crucial it is to maintain a balanced gut microbiota to prevent infections. **Impaired Tolerance and Immune Homeostasis:** Antibiotic usage can alter the gut flora, disrupt immunological homeostasis, and have long-term health implications. Early exposure to antibiotics has been associated with a higher incidence of autoimmune, inflammatory, and atopic illnesses. Research indicates correlations between the use of antibiotics and ailments such as allergic reactions, inflammatory bowel illnesses, and asthma. However, new studies show that exposure to antibiotics, especially broad-spectrum antibiotics, might cause problems like asthma in a dose-dependent manner. Furthermore, a mother's use of antibiotics throughout her pregnancy may raise her child's risk of developing atopic illnesses. Microbiota changes are also associated with irritable bowel syndrome (IBS), which frequently occurs after gastrointestinal illnesses and may be impacted by antibiotic use. **Deregulated Metabolism:** Many metabolic problems can arise from disruptions in host metabolism brought on by antibiotic-induced changes in the gut microbiota. Obesity has been linked to changes in the gut microbiota, including decreased diversity and changes in bacterial genes and metabolic pathways that promote energy extraction from food. By increasing persistent low-grade inflammation, antibiotics can worsen the development of metabolic syndrome, a group of disorders that includes fatty liver disease, cardiovascular disease, and type 2 diabetes. Moreover, epidemiological research and mice models have demonstrated that antibiotics raise the risk of type 1 diabetes, an autoimmune illness. These results emphasize the intricate connection between gut microbiota, antibiotic usage, and metabolic health and highlight the significance of sparingly using antibiotics to reduce metabolic risks. **How do you keep your gut healthy while taking antibiotics?** **  ** **Img-1: Precautions to be taken while taking antibiotics.** _**When consuming antibiotics prescribed by a doctor, it is crucial to take sufficient care of your gut. You can add probiotic supplements if necessary and consume fermented foods to maintain gut health. Additionally, staying hydrated and maintaining a balanced diet are important. If any side effects of antibiotics are observed, please seek expert advice.**_ Maintaining a healthy gut when taking antibiotics is critical to minimizing the alteration of the gut microbiota and lowering the possibility of antibiotic-related adverse effects. The following advice will help you maintain a healthy stomach while taking antibiotics: **Take Probiotics:** Consider taking probiotic supplements containing helpful bacteria like Lactobacillus and Bifidobacterium. The good bacteria in your stomach that antibiotics may have destroyed can be restored with the use of probiotics. To make sure probiotics are safe and suitable for you, consult with your doctor before beginning any supplementation. **Consume Fermented Foods:** Include foods like yoghurt, kefir, sauerkraut, kimchi, and kombucha in your diet that have undergone fermentation. Natural probiotics found in these foods can aid in maintaining a balanced population of good gut flora. **Stay Hydrated:** Since antibiotics can occasionally result in dehydration, make sure you drink lots of water to stay hydrated. Drinking enough water promotes intestinal health in general and aids in the removal of toxins from the body. **Eat a Balanced Diet:** Pay attention to consuming a diet high in whole grains, fruits, vegetables, lean meats, and healthy fats. Essential minerals and fiber from various foods improve intestinal health and encourage the growth of good bacteria. **Eat Less Sugary and Processed Foods:** Consuming too much sugary and processed food might upset your stomach's natural balance and encourage the growth of dangerous bacteria. To support a healthy gut environment, choose whole foods that are high in nutrients instead. **Take Antibiotics as Directed:** Follow your doctor's instructions for taking antibiotics exactly as prescribed. Even if you feel better before finishing the course of antibiotics, take them all to be sure the infection is completely cleared. **Address Side Effects:** Talk to your doctor if you have any gastrointestinal side effects after taking antibiotics, such as nausea, diarrhoea, or upset stomach. To treat these symptoms, they could advise changing your antibiotic treatment or recommending extra medications. **Promote Digestive Health:** Include foods high in prebiotic fibres, which support the growth of good gut bacteria, such as onions, garlic, bananas, asparagus, and oats. Consider using digestive enzymes or herbal supplements to support digestive function and reduce gastrointestinal discomfort. **Conclusion** In conclusion, the widespread and often indiscriminate use of antibiotics presents serious concerns despite their undeniable contributions to modern medicine. Antibiotic resistance poses a significant threat to their effectiveness in treating infections, while the collateral harm to the gut microbiota underscores the importance of responsible antibiotic use to mitigate long-term health risks. These gut modifications extend beyond immediate health concerns, affecting metabolic function and immune response. Strategies such as probiotic supplements and a balanced diet are crucial for maintaining gut health during antibiotic use. However, addressing antibiotic misuse requires coordinated efforts to promote alternative infection control methods, raise public awareness, and foster responsible antibiotic practices. By prioritizing both the therapeutic benefits of antibiotics and the preservation of gut health, we can combat antibiotic resistance and safeguard public health for future generations. **References** 1. [https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8756738/](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8756738/) 2. [https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7732679/](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7732679/) 3. [https://www.the-scientist.com/what-happens-to-the-gut-microbiome-after-taking-antibiotics-69970](https://www.the-scientist.com/what-happens-to-the-gut-microbiome-after-taking-antibiotics-69970) 4. [https://www.kemin.com/in/en/blog/animal/managing-gut-health-with-antibiotic-alternatives](https://www.kemin.com/in/en/blog/animal/managing-gut-health-with-antibiotic-alternatives) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## How Gut Health Impacts Anemia: Exploring the Vital Connection Author: BugSpeaks Published: 2024-03-21 Category: Microbiome and Disease Meta Title: How Gut Health Impacts Anemia: Exploring the Vital Connection Meta Description:
In India, alarming percentages of women (57%), female infants (67%), males (25%), and teenage boys (31%) battle anemia. URL: https://www.bugspeaks.com/blog/how-gut-health-impacts-anemia:-exploring-the-vital-connection In India, alarming percentages of women (57%), female infants (67%), males (25%), and teenage boys (31%) battle anemia. On this National Anemia Day, let's explore how gut health influences this condition and work towards solutions for better health. Anemia, characterized by a deficiency of healthy red blood cells or hemoglobin, disrupts the vital process of oxygen transportation throughout the body. This deficiency can stem from various causes, including blood loss, impaired red blood cell synthesis, or accelerated destruction. Symptoms of anemia are weariness, weakness, and a pale complexion, all indicative of insufficient oxygen-carrying capacity in the bloodstream. Breathing difficulties, dizziness upon sudden movements, and palpitations are common experiences, highlighting the profound impact of oxygen deprivation on bodily functions. Additionally, headaches, cold extremities, and difficulty concentrating are typical signs of anemia, while severe cases may lead to leg cramps, chest discomfort, and memory loss. Due to the variability in symptoms and underlying causes, a comprehensive medical examination is essential for accurate diagnosis and personalized treatment strategies.  **Types of Anemia** There are three types of anemia, mainly known: **1\. Iron-Deficiency Anemia:** Iron deficiency anemia arises when the body lacks a sufficient amount of iron to produce hemoglobin, the vital protein found in red blood cells responsible for ferrying oxygen from the lungs to every corner of the body. Iron plays a pivotal role in the synthesis of hemoglobin, and its scarcity can stem from a multitude of factors. These may include inadequate consumption of iron-rich foods, such as meat, beans, and fortified cereals, or chronic blood loss, which can occur due to heavy menstrual periods, gastrointestinal bleeding, or other sources. Additionally, conditions like celiac disease or gastric bypass surgery can impair iron absorption in the gut, further exacerbating the deficiency. Furthermore, periods of high iron demand, such as pregnancy or menstruation, can deplete iron storage, worsening the problem. A range of symptoms, including weakness, exhaustion, headaches, pale yellow skin, and cold extremities, are symptoms of iron deficiency anemia and show how difficult it is for the body to withstand oxygen deprivation. **2\. Vitamin B12-deficiency anemia:** Vitamin B12 deficiency Anemia stems from an insufficient supply of vitamin B12, which is crucial for the synthesis of red blood cells and the proper functioning of the nervous system. Primary sources of vitamin B12 include animal-derived foods such as meat, fish, poultry, eggs, and dairy products. However, deficiencies may arise due to various factors, including inadequate dietary intake, impaired absorption in the gut, gastrointestinal surgeries, or deficiencies in intrinsic factors, like a stomach-produced protein essential for vitamin B12 absorption. Symptoms of vitamin B12 deficiency anemia include fatigue and weakness, as well as a pale yellow complexion. Additionally, individuals may experience shortness of breath, tingling or numbness in the extremities, difficulties in walking, and cognitive impairments, underscoring the profound impact of vitamin B12 deficiency on both physical and neurological well-being. **3\. Hemolytic Anemia:** Hemolytic anemia occurs when the rate of red blood cell loss exceeds the rate of production, resulting in a depletion of these vital cells within the bloodstream. Hemolysis, the term describing this accelerated destruction, can be instigated by a diverse number of factors. These may include autoimmune diseases, where the body erroneously attacks its red blood cells, as well as inherited disorders like sickle cell disease or thalassemia. Infections, exposure to certain drugs or toxins, and mechanical anomalies such as blood vessel abnormalities can also precipitate hemolysis. Symptoms of hemolytic anemia include weakness, splenomegaly (enlargement of the liver), and a pale yellow complexion. Darkened urine, a rapid heartbeat, and difficulty breathing may also occur, underscoring the variable nature of this condition's presentation.  **Img-1: Symptoms of Anemia** _The pictures above show typical signs and symptoms of anemia. Continuous signs include headaches, cognitive decline, fatigue and weakness, shortness of breath, pale or yellowish skin, and dizziness, which can result from iron deficiency. These symptoms underline the serious impact of iron deficiency on both physical and cognitive function, highlighting the significance of early detection and management techniques to reduce its impacts on individual health._ **Importance of Iron** Iron serves as a crucial micronutrient vital for numerous metabolic processes within the body. Primarily, it functions as an integral component of proteins and enzymes responsible for oxygen transport, notably myoglobin and hemoglobin. Furthermore, iron plays pivotal roles in DNA processing, cellular development, proliferation, and mitochondrial energy metabolism. The body's iron stores experience depletion primarily through the shedding of enterocytes, epidermal cells, and menstrual blood in women, with lesser losses occurring through urine and feces. Despite these losses, the body efficiently renews its iron stores internally through the recycling of senescent and damaged erythrocytes. A daily intake of 1 to 2 mg is typically sufficient to maintain optimal iron levels, ensuring concentrations remain within the range of 3 to 5 grams. Iron absorption predominantly occurs in the duodenum and proximal section of the jejunum, facilitated by the high expression of proteins involved in non-heme iron absorption in these regions. However, despite its essential role, iron can pose toxicity risks when its binding protein capacity is compromised. This metal exhibits unique properties that render it potentially harmful; it can generate free radicals and also serves as a critical growth factor for numerous pathogenic bacteria, fungi, protozoa, and cancerous cells. Thus, maintaining a delicate balance of iron within the body is imperative to harness its benefits while mitigating the risks associated with its toxicity.  **Img-2: Anemia blood type.** _The graphic illustrates the difference in red blood cell (RBC) count between a healthy person and a person who has anemia. A healthy human has an abundance of red blood cells in their blood, while an anemic person has fewer red blood cells in their blood. The key feature of anemia is highlighted by this graphic, which highlights the blood's decreased ability to carry oxygen because of low RBC counts._ **Revealing the role of the gut in anemia** Iron supplementation exerts a profound influence on the delicate balance of the intestinal microbiota, creating a complex and bidirectional relationship between iron levels and gut health. Experimental evidence suggests that an excess of iron in the intestinal lumen can compromise the integrity of the intestinal mucous membrane, posing potential harm. Despite its crucial role in various biological functions, excessive free iron can generate harmful free radicals within the colon, leading to oxidative stress and direct damage to the intestinal epithelium. This disruption of the intestinal barrier can result in increased permeability, commonly referred to as "leaky gut," thereby increasing susceptibility to bacterial endotoxins and culminating in metabolic endotoxemia and microinflammation. Furthermore, the symbiotic relationship between gut microbes and iron levels further complicates the intricate interplay. Studies indicate that both excessive and insufficient iron levels significantly influence the composition of gutthe gut microbiota, initiating and exacerbating conditions of inflammation and even contributing to the development of colorectal cancer. Certain bacteria, such as Streptococcus bovis, Enterococcus faecalis, and Clostridia, are implicated in the pathogenesis of intestinal diseases, producing substances like secondary biliary salts and hydrogen sulfide that fuel inflammation and carcinogenesis. Conversely, beneficial bacteria like Lactobacillus acidophilus and Bifidobacterium longum play a protective role, mitigating intestinal inflammation and maintaining gut health. The gut serves as the primary site for the absorption of essential nutrients, including iron and vitamin B12, which are crucial for the synthesis of red blood cells. Disruptions in gut health, stemming from gastrointestinal illnesses, dietary deficiencies, or alterations in gut microbiota composition, can significantly impair the absorption of these vital nutrients, potentially resulting in various forms of anemia. Conditions such as celiac disease, inflammatory bowel disease, and chronic gastritis further worsen the challenge by limiting the body's capacity to absorb iron or vitamin B12. Thus, understanding and addressing the intricate relationship between gut health and nutrient absorption is paramount in the management and prevention of anemia and associated conditions. **Solution for Anemia** 1. Addressing Gut Infections: Get tested for intestinal infections due to dysbiosis. You can visit our website for more details on the [test](https://www.bugspeaks.com/pricing). 2. Gut Healing: Use targeted therapies, such as dietary adjustments, gut-healing nutrients, and modifications to your lifestyle, to address any underlying problems causing gut inflammation or permeability. 3. Frequent Observation: Collaborate with a healthcare expert to monitor gut health parameters and adapt treatment approaches depending on individual requirements and responses. 4. Balancing Stress: To promote gut health, engage in stress-reduction practices including deep breathing exercises, mindfulness, meditation, and regular physical activity. 5. Preventing Excessive Medication Use: Reduce your usage of medications available without a prescription, especially those that might affect gut health, like nonsteroidal anti-inflammatory drugs. 6. Dietary Modifications: Consume a diet high in whole foods, such as fruits, vegetables, lean proteins, healthy fats, and fiber, to reduce inflammation. To encourage the development of good gut flora, include foods high in prebiotics, such as whole grains, garlic, onions, and bananas. Eat foods high in probiotics, such as fermented food, buttermilk, and yogurt, to help maintain a balanced gut microbiome. **Conclusion** The complex interrelationship between gut health and anemia emphasizes how crucial it is to take care of both for general well-being. Anemia is a medical condition that is common in India and across the world. It can have serious effects on one's health and quality of life. Understanding the many forms, symptoms, and fundamental reasons for Anemia is essential for a suitable diagnosis and management. Furthermore, the correlation between gut health and the absorption of nutrients, including iron and vitamin B12, highlights the need for prioritizing gut health therapies in conjunction with the treatment of anemia. People can strive towards regaining balance and health by using a comprehensive strategy that addresses gut infections, promotes gut healing, and lowers inflammation in the body. By taking preventative action and working together with medical experts, we can work to lessen the impact of Anemia and enhance general health outcomes for people as well as communities. **References** 1. Malesza, I. J., Bartkowiak-Wieczorek, J., Winkler-Galicki, J., Nowicka, A., Dzieciolowska, D., Baszczyk, M., ... & Madry, E. (2022). The dark side of iron: the relationship between iron, inflammation and gut microbiota in selected diseases associated with iron deficiency Anemia - a narrative review. Nutrients, 14(17), 3478. 2. Rusu, I. G., Suharoschi, R., Vodnar, D. C., Pop, C. R., Socaci, S. A., Vulturar, R., ... & Pop, O. L. (2020). Iron supplementation influence on the gut microbiota and probiotic intake effect in iron deficiency - A literature-based review. Nutrients, 12(7), 1993. 3. Yilmaz, B., & Li, H. (2018). Gut microbiota and iron: the crucial actors in health and disease. Pharmaceuticals, 11(4), 98. 4. [https://www.nhlbi.nih.gov/health/anemia](https://www.nhlbi.nih.gov/health/anemia) 5. [https://economictimes.indiatimes.com/magazines/panache/health/making-anemia-a-visible-problem-in-india/articleshow/100819443.cms?from=mdr](https://economictimes.indiatimes.com/magazines/panache/health/making-anemia-a-visible-problem-in-india/articleshow/100819443.cms?from=mdr) 6. [https://my.clevelandclinic.org/health/diseases/3929-anemia](https://my.clevelandclinic.org/health/diseases/3929-anemia) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Postpartum Gut Health: A Guide for New Mothers Author: BugSpeaks Published: 2024-03-15 Category: Gestational and Neonatal Meta Title: Postpartum Gut Health: A Guide for New Mothers Meta Description:
While new moms are filled with excitement and surprise at the birth of their child, the postpartum phase frequently causes a flurr URL: https://www.bugspeaks.com/blog/postpartum-gut-health:-a-guide-for-new-mothers While new moms are filled with excitement and surprise at the birth of their child, the postpartum phase frequently causes a flurry of changes in their lives. It's a period of great physical and mental change, from enduring restless nights to getting used to the responsibilities of caring for a newborn. The effect postpartum recovery has on gut health is one area that is sometimes disregarded but has benefits if given careful consideration. Understanding and promoting gut health becomes essential for the long-term health of the mother as well as that of her child, as the body experiences extraordinary changes following childbirth.  **Changes in a woman's body after the birth of a baby** During the postpartum period, a woman's body undergoes significant changes as it transitions from pregnancy to motherhood. Uterine involution, marked by the contraction and shedding of the uterine lining, occurs, along with hormonal fluctuations affecting mood, lactation, and gut diversity. Breast changes prepare for lactation, while vaginal changes like discharge and swelling are common. Side effects such as tiredness, mood shifts, and weight loss are experienced, alongside pelvic floor alterations and postpartum hair loss.  **Postpartum Changes in Mother After Delivery** _After giving birth, the woman experiences several physical and emotional changes. Constant mood fluctuations, exhaustion, lack of sleep at night, and hair loss. Numerous problems, including cramping, accompany uterine involution and gut alteration._ **Understanding Postpartum Gut Health** **1\. Physical Changes and Gut Microbiota** Physical changes, including uterine involution, hormonal fluctuations, breast changes, and vaginal alterations, are accompanied by shifts in gut microbiota composition. These changes can affect gut motility, digestion, and overall gut health, often leading to symptoms like constipation or diarrhea. **2\. Sleep disturbance and Gut Health** Sleep disruption and deprivation are major problems for women in the postpartum period as they balance the responsibilities of infant care. Getting enough sleep is essential for the immunological and neurological systems to operate properly and for the postpartum healing process. However, sleep disturbances are often linked to depression and exhaustion, which presents more difficulties for new moms. Long-term sleep disturbances can have profound effects on metabolism, hormone release, and the risk of cardiometabolic diseases, including diabetes and obesity. Research has revealed fascinating connections between sleep, circadian rhythm, chronic fatigue syndrome, and the gut microbiota. Research has revealed that individuals suffering from chronic fatigue syndrome frequently display a less diverse gut microbiota, which can result in dysbiosis and heightened gut permeability. The disruption of the gut flora may worsen long-term inflammation and make it more difficult for new mothers to heal from childbirth. **3\. Mental Health and Gut Health** Gut health and mental health problems, especially postpartum depression (PPD), are closely related throughout the postpartum period. Research has shown that changes in the variety and composition of the gut microbiome can affect the synthesis of neurotransmitters and other neuroactive substances, which can then affect the control of emotions and brain function. Additionally, it is becoming more widely acknowledged that a major role in mental health is played by the gut-brain axis, a bidirectional communication link between the gut and the brain. Disruptions in this axis, such as gut microbiota imbalances or increased intestinal permeability, may lead to the emergence or worsening of mental health issues, including postpartum depression. Furthermore, gut health and microbiome composition can be further influenced by hormonal changes that occur during pregnancy and the postpartum period, which may increase a person's vulnerability to mental health problems. In general, there is a complicated and varied association between gut health and mental health throughout the postpartum phase.  **Image no. 2- Stress and Gut Relation** _The gut and the brain talk to each other in many ways. One way is through the vagus nerve, which connects them directly. Another is through substances produced by the gut bacteria that can affect the immune system and the gut lining. Also, gut bacteria create chemicals called short-chain fatty acids (SCFAs) that can influence the brain. Additionally, cells lining the gut release hormones that can send signals to the brain. These signals control important functions like mood, inflammation, and stress response. When the gut bacteria get out of balance, causing dysbiosis, it can disrupt these signals and contribute to stress-related problems._ **Digestive issues and gut health** Many women have digestive problems during the postpartum period, including bloating, constipation, and pain, which can have a serious negative effect on their health. These problems are frequently associated with modifications to gut health, such as adjustments to the makeup and functionality of the gut microbiota. The delicate balance of bacteria in the gut becomes disturbed during pregnancy and delivery, which can result in dysbiosis and digestive issues. These problems can be made worse by elements including dietary modifications, stress, hormone swings, and postpartum drugs. **Breast infection and gut health** Breast inflammation, or mastitis, can have several reasons and is most common in the postpartum phase. 3–33% of nursing mothers experience it, usually in the first few phases of lactation. A prominent explanation for bacterial mastitis is an overabundance of pathogenic strains in the mammary gland, such as group B streptococci and Staphylococcus spp. A series of immunological and metabolic alterations that result in the clinical signs and symptoms of mastitis are caused by this excess, disrupting the balance of beneficial bacteria. Unfortunately, the discomfort, fever, and malaise associated with mastitis frequently discourage breastfeeding. Misconceptions regarding the transmission of infection to the newborn while breastfeeding may also deter moms from nursing their babies while undergoing therapy. Despite the challenges posed by these modifications, they are all integral aspects of the body's natural recovery and adaptation to the demands of motherhood. **How can you take care of your gut during postpartum?** **Stay Hydrated:** Maintaining gut health and preventing constipation may be achieved by drinking enough fluids, especially warm beverages, first thing in the morning. This also helps keep stools soft and encourages bowel movements. **Boost Dietary Fiber:** Include naturally occurring dietary fiber sources in your diet, such as fruits, vegetables, nuts, almonds, and flaxseeds, to maintain a healthy colon by encouraging regularity and avoiding constipation. **Regular Exercise:** Physical activity on a regular basis enhances gut health and digestive function by stimulating bowel movement and improving circulation. Walking and yoga are examples of gentle workouts that might be very helpful in the postpartum phase. **Consume Probiotics:** Using probiotics to repopulate a healthy gut flora aids in optimal nutrient absorption and improves digestive health in general. Incorporating probiotics into the diet through foods like yogurt (curd), buttermilk (chaas), and fermented dishes like idli, dosa, or dhokla helps improve gut health in the postpartum phase. **Make Sleep a Priority:** During the postpartum phase, good sleep is essential for gut health and general well-being. Improved sleep quality may be achieved by establishing a regular bedtime, creating a sleep-friendly atmosphere, and asking partners or family for assistance with nocturnal baby care. **Control Mental Stress:** During the postpartum phase, gut health and general wellness depend on the control of mental stress. Techniques that can help lower stress and improve emotional well-being include mindfulness meditation, deep breathing exercises, and asking for help from loved ones or mental health specialists. **How can you enhance your baby’s gut health?** Throughout infancy and beyond, you should support the healthy development of your baby's microbiome. Vaginal birth introduces a broad range of microorganisms, encouraging the early development of the baby's microbiome. Breastfeeding encourages the development of a varied microbiome in your child by giving them vital nutrients and beneficial probiotics. If nursing isn't an option, you could choose to use screened milk from milk banks. In the first few weeks, embrace skin-to-skin contact and refrain from using harsh cleaning solutions to support your baby's developing microbiome and impart the advantages of your mature microbiota. Prioritize a diet high in whole, natural foods when your baby starts to eat solid meals to help the diversity and health of the microbiome.  **Image no.3- Healthier Gut Health Ways of Baby** _The newborn baby is completely sterile when entering this world. So the first exposure to bacteria is during birth through the vagina. Breastfeeding and skin-to-skin contact, which involve a lot of hugs and kisses for the baby, help introduce mature microbiota. After some time, enhance the baby's diet with a probiotic-rich natural diet._ **Conclusion** New moms experience a variety of physical and psychological changes throughout the postpartum phase. It's a period of major adjustment and adaptation, from the amazing changes in the body to the difficulties in managing sleep, emotional stress, and digestive concerns. Understanding the significance of gut health throughout this stage is critical since it affects both the mother's and her child's well-being. Women may maintain their gut health and go through the postpartum journey more easily and resiliently by placing a high priority on water, dietary fiber, exercise, probiotics, restful sleep, and mental stress management throughout their pregnancy and beyond. Remember, dear moms, that in the middle of all the change and responsibility, taking care of yourself is just as crucial as caring for your child. You are strong and capable, and you deserve love and support as you embark on the amazing path of parenthood! **References** 1. Weerasuriya, W., Saunders, J. E., Kis, L., Ho, T. T., Xu, K., Lemas, D. J., ... & Louis-Jacques, A. F. (2023). Maternal gut microbiota in the postpartum Period: A Systematic review. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2. Mutic, A. D., Jordan, S., Edwards, S. M., Ferranti, E. P., Thul, T. A., & Yang, I. (2017). The postpartum maternal and newborn microbiomes. MCN: The American Journal of Maternal/Child Nursing, 42(6), 326-331. 3. [https://www.biocodexmicrobiotainstitute.com/en/postpartum-depression-changes-gut-microbiota-under-spotlight](https://www.biocodexmicrobiotainstitute.com/en/postpartum-depression-changes-gut-microbiota-under-spotlight) 4. [https://www.mothernutrient.com/blogs/mother-nutrient-blog/how-to-ease-common-postpartum-digestive-issues](https://www.mothernutrient.com/blogs/mother-nutrient-blog/how-to-ease-common-postpartum-digestive-issues) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## How Your Microbiome Influences Cravings and Eating Disorders Author: BugSpeaks Published: 2024-02-27 Category: Microbiome and Disease Meta Title: How Your Microbiome Influences Cravings and Eating Disorders Meta Description:
Craving for a pizza, burger, fries, cold drinks, panipuri, samosa, or vadapav (add your favorite snack of all time)? Hav URL: https://www.bugspeaks.com/blog/how-your-microbiome-influences-cravings-and-eating-disorders Craving for a pizza, burger, fries, cold drinks, panipuri, samosa, or vadapav (add your favorite snack of all time)? Have you ever felt like eating too much food without knowing how much you ate? Or have you ever felt like eating nothing because you might gain weight? So, this emotional eating, or eating according to your thoughts, is normal for all of us. But what if we told you that this emotional eating could be an eating disorder? Eating disorders are severe psychiatric conditions, primarily encompassing mood and anxiety disorders along with obsessive-compulsive disorders, characterized by extreme weight fluctuations and disruptions in hunger regulation. These illnesses, which can be grave and even fatal, manifest through significant disruptions in eating patterns alongside corresponding thoughts and emotions. It affects up to 5% of the population and typically begins in adolescence or young adulthood. These can be extremely dangerous conditions that interfere with social, psychological, and physical functioning. **What are the causes?** While precise causes remain elusive, experts posit that a blend of factors, including genetics, social dynamics, physical health, and psychological factors, contribute to the development of eating disorders. Psychological factors such as being more susceptible to depression and anxiety, having trouble dealing with stress, worrying excessively about the future, having perfectionistic tendencies, having trouble controlling one's emotions, feeling obsessive or compulsive, and being afraid of being called "fat" or overweight lead to interrupted eating habits. Genome-wide association scientific studies (which help to identify genes related to disease) have discovered distinct genetic polymorphisms linked to eating disorder risk, emphasizing the intricate interaction of genetics and environment in their development. However, the gut microbiome research on this topic has caught the attention of the researchers as it is related to neuropsychiatric disorders, metabolic disorders, and immune-mediated diseases, which indirectly are related to the factors affecting eating disorders. The alterations in gastrointestinal functioning disrupt the circadian rhythms (the body's internal clock) by influencing the timing and release of hormones involved in digestion, such as leptin and ghrelin, which control hunger and the metabolism of energy. The synchronization of peripheral clocks in the stomach with the gut circadian clock can be disrupted by irregular eating patterns. This can result in disruptions to general metabolic processes, nutrition absorption, and digestion. Excessive dietary restriction prolongs the time that food remains in the stomach by slowing down the process of gastric emptying. Prolonged transit times might cause constipation by interfering with regular bowel motions. It also modifies the makeup of the gut flora, giving preference to microorganisms linked to diet restrictions. These modifications to the gut flora may help to develop or exacerbate restrictive eating behaviors by extending the cycle of food restriction. Gut microorganisms may have an impact on hunger and satiety hormones such as leptin, ghrelin, peptide YY (PYY), and neuropeptide Y (NPY), which are implicated in eating disorders. Under normal physiological circumstances, leptin suppresses appetite, while ghrelin suppresses hunger. Different conditions cause various types of eating disorders, specifying different gut conditions. **Types of Eating Disorders** **  ** **Img-1: Potential signs of an Eating disorder.** _A few indicators of eating disorders are shown in the image above, including worrying excessively about one's weight and visual appeal, exercising excessively because one is preoccupied with being overweight and maintaining abnormally high electrolyte levels. Consuming unusually large amounts of food without tracking how much is consumed. Experiencing occasional anxiety and food avoidance._ International disease classification systems recognize seven distinct eating disorders. However, the three most prevalent among humans globally are **Anorexia Nervosa (AN)**, **Bulimia Nervosa (BN)**, and **Binge Eating Disorder (BED)**, while others include rumination disorder, pica, avoidant food intake disorder, and avoidant/restrictive food intake disorder (ARFID). **Anorexia Nervosa (AN)** is a severe eating disorder characterized by severe dietary restriction, which leads to dangerously low body weight and a deep-seated fear of gaining weight. A psychological profile characterized by a strong desire to be skinny and unhappiness with one's physical appearance is typically the core cause of this condition. Furthermore, about 40% of individuals with AN experience a substantial drop in their bone mineral density (BMD), which increases their lifelong fracture risk by three times. According to epidemiological statistics and genome-wide association studies, people with anxiety and depressive illnesses may be susceptible to AN. Symptoms of extreme dehydration include dizziness or fainting, as well as brittle hair and nails, cold sensitivity, and muscular weakness. Vomiting may cause heartburn and reflux, and severe constipation and bloating which are frequently followed by a feeling of fullness after meals are typical gastrointestinal symptoms. Obsessive exercisers run the risk of stress fractures and bone loss, which can develop into osteopenia or osteoporosis. The psychological effects of eating disorders might also include exhaustion, melancholy, irritability, anxiety, and poor focus. **Bulimia Nervosa (BN)** is characterized by the disappearance of enormous amounts of food, persistent sore throats, and enlarged salivary glands. The term "binge eating" refers to eating a lot of food quickly and feeling out of control of what or how much is consumed. Binge behavior is frequently hidden and accompanied by emotions of guilt or humiliation. Food is frequently ingested quickly, beyond fullness, to the point of nausea and pain, and binges can be quite enormous. At least once a week, binges happen, and in order to avoid gaining weight, "compensatory behaviors" are usually engaged thereafter. Fasting, vomiting, laxative abuse, and obsessive exercise are examples of such behaviors. People who suffer from bulimia nervosa may be underweight, average-weight, overweight, or even obese. Heartburn, laxative or diuretic misconduct, and dental deterioration due to erosion caused by stomach acid are other prevalent symptoms. Frequent, unexplained diarrhea and dehydration from excessive purging behaviors may be signs of borderline personality disorder (BN). **Binge Eating Disorder (BED):** Like bulimia nervosa, patients suffer phases of binge eating during which they eat a lot of food in a short amount of time, feel as though they have lost control over their eating, and are upset by their excessive behavior. They do not, however, frequently engage in compensatory behaviors to get rid of the food, such as forced vomiting, fasting, exercising, or abusing laxatives, unlike those who suffer from bulimia nervosa. Serious health issues, including obesity, diabetes, hypertension, and cardiovascular disorders, can be brought on by binge eating disorders. The diagnosis of binge eating disorder involves frequent binges (at least once a week for three months), along with three or more of the following features: 1. Eating until their stomachs hurt. 2. Consuming a lot of food even when not hungry. 3. Eating alone due to embarrassment about one's appetite. 4. Feeling extremely guilty, unhappy, or dissatisfied with oneself following a binge. Here are a few other eating disorders, explained briefly - A recently characterized eating disorder called **Avoidant Restrictive Food Intake Disorder (ARFID)** is characterized by high pickiness and a chronic inability to achieve nutritional demands due to an eating disturbance. An individual with **Pica**, an eating problem, would often consume non-food items that are devoid of nutrients. Depending on the age and accessibility, common materials consumed might be anything from paper to paint chips, soap, fabric, hair, string, chalk, metal, stones, charcoal, or clay. **Rumination Disorder** is characterized by recurrent episodes of food regurgitation and rechewing after eating, in which the food is willingly pulled back up into the mouth, chewed again, swallowed again, or spit out. Rumination disorder can strike throughout early life, childhood, adolescence, or even as an adult. **How are these eating disorders related to the gut?** Every person's gut microbiota is different and is impacted by a variety of factors, including geography, age, sex, food, health, and drug exposure. An imbalance in microbial species, known as microbial dysbiosis, is frequently associated with several diseases. The gut microbiota plays an important role in drug detoxification, immune system training, and vitamin synthesis. It also affects host behavior modification, diet-induced energy extraction, and weight management via direct and indirect pathways. Overall, it is an essential component in preserving many aspects of health and well-being. Dysregulated calorie and nutrition intake may impact gut bacteria metabolism in eating disorder individuals. Research on animals indicates a link between the gut microbiota and characteristics of eating disorders, such as dysregulated behavior and energy balance. α-diversity refers to the diversity of bacteria in an individual ecological community. Research has shown that higher α-diversity correlates with better health. A reduction in gut microbiota diversity, particularly among butyrate-producing bacterial species, is linked to increased anxiety, depression, and eating disorder (ED) psychopathology. This shows that the gut microbiota may be the missing link in our knowledge of eating disorders (EDs) by having a significant impact on eating behavior and mental health. Research findings reveal a negative correlation between α-diversity and ED psychopathology, encompassing depression and body image issues. Poor nutrition in anorexia nervosa (AN) can alter the internal structure of the small intestine, decreasing its capacity to absorb nutrients and making weight gain more difficult. Purging behaviors, such as vomiting, can damage the stomach lining and alter its function, resulting in stomach disorders. Misusing laxatives in AN or bulimia nervosa (BN) can result in chronic diarrhea, electrolyte abnormalities, and bowel movement problems. In AN and BN populations, a lower BMI is linked to a greater prevalence of E. coli in the gut. **Treatments** **  ** **Img-2: Therapies for Eating Disorders.** _The picture above shows us many approaches to managing eating problems. It is always preferable to consult specialists to prevent future issues. Among the therapies used to treat include psychotherapy, family-based therapy, nutritional therapy, and occupational therapy. Engaging in physical activity is crucial for maintaining stability and improving health._ Starting with basic treatment, i.e., treating your gut first to improve and stabilize all the affecting factors of mental and physical health, To increase gut diversity, simply by following a few habits like: 1. Consuming a wide range of fruits, vegetables, whole grains, legumes, nuts, and seeds. Including probiotics like yogurt, buttermilk, fermented foods like Idli, Dosa, pickles, etc. Also, eat prebiotic-rich foods like onions, garlic, bananas, asparagus, and whole grains. 2. Staying hydrated is very important to maintain good health. 3. Managing stress and balancing mental health through yoga, meditation, and deep breathing exercises. 4. Staying active involves regular exercise and physical activities. 5. Consuming limited processed foods and sugars. 6. Getting plenty of sleep for up to 6-7 hours. Repopulating the gut microbiome with organisms that reduce Eating disorder-related symptoms, such as Lactobacilli, Bifidobacterium spp., and Enterococcus spp., may result in improved recovery rates. However, increasing the variety of bacteria may affect food choices and habits, resulting in good weight. Restoring the gut microbiota may also address dysfunctional physical alterations (such as a reduced ability to absorb nutrients) that impede recovery and lead to long-term weight gain and better results. Regular exercise may increase α-diversity in the gut microbiota. Treatment for anorexia nervosa focuses on helping individuals normalize their eating habits and restore a healthy weight. This includes addressing any other medical or mental health conditions. The treatment plan often involves a balanced nutritional approach to ease anxiety around food and ensure regular, spaced-out meals. In adolescents, involving parents in meal support is effective. Body image concerns are also addressed, but they may take longer to improve than eating habits. Severe cases may require admission to an inpatient or residential programme, which can be effective in restoring weight and eating behaviors, though the relapse risk remains high after discharge. The most evidence-based treatment for bulimia nervosa is outpatient cognitive-behavioral therapy. It assists patients in controlling the thoughts and emotions that feed the condition as well as normalizing their eating habits. The desire to binge and throw up can also be lessened by antidepressants (fluoxetine, for example). Similar to bulimia nervosa, cognitive behavioral psychotherapy, either on an individual or group basis, is the most successful treatment for binge eating disorder. Research has demonstrated the efficacy of interpersonal therapy and several antidepressant drugs. **Conclusion** To sum up, eating disorders are complex mental illnesses with a variety of underlying causes and manifestations. From emotional eating habits to genetic predispositions, a variety of variables contribute to the development and recurrence of these disorders. New research highlights the connection between physiological and psychological variables in eating disorders by indicating a major role of the gut microbiota in affecting eating behaviors and mental health. Gaining knowledge of the gut-brain axis and how eating disorder pathophysiology is affected creates novel therapy options, such as probiotic supplements and dietary changes. Comprehensive treatment options, however, are necessary to improve general well-being and support long-term recovery from eating disorders by addressing both their psychological and physical components. Meanwhile, ignore your self-defeating thoughts about your weight and looks; instead, follow your heart and adopt a healthy diet. Happy eating! **References** 1. Feng, B., Harms, J., Chen, E., Gao, P., Xu, P., & He, Y. (2023). Current Discoveries and Future Implications of Eating Disorders. International Journal of Environmental Research and Public Health, 20(14), 6325. 2. Xu, J., Carroll, I. M., & Huckins, L. M. (2023). Eating disorders: are gut microbiota to blame?. Trends in Molecular Medicine. 3. Terry, S. M., Barnett, J. A., & Gibson, D. L. (2022). A critical analysis of eating disorders and the gut microbiome. Journal of Eating Disorders, 10(1), 154. 4. Thaiss, C. A., Zeevi, D., Levy, M., Zilberman-Schapira, G., Suez, J., Tengeler, A. C., ... & Elinav, E. (2014). Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell, 159(3), 514-529. 5. Oliphant, K., & Allen-Vercoe, E. (2019). Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome, 7(1), 1-15. 6. Lucka, A., & Wysokinski, A. (2019). Association between adiposity and fasting serum levels of appetite-regulating peptides: Leptin, neuropeptide Y, desacyl ghrelin, peptide YY (1-36), obestatin, cocaine and amphetamine-regulated transcript, and agouti-related protein in nonobese participants. Journal of Physiology Investigation, 62(5), 217-225. 7. [https://www.nimh.nih.gov/health/topics/eating-disorders](https://www.nimh.nih.gov/health/topics/eating-disorders) 8. [https://www.psychiatry.org/patients-families/eating-disorders/what-are-eating-disorders](https://www.psychiatry.org/patients-families/eating-disorders/what-are-eating-disorders) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## How and Why to Cultivate a Healthy Gut During Pregnancy Author: BugSpeaks Published: 2024-02-05 Category: Gestational and Neonatal Meta Title: How and Why to Cultivate a Healthy Gut During Pregnancy Meta Description:
Pregnancy is a wonderful time in a woman's life when her body transforms into a safe haven and she writes a story about love, URL: https://www.bugspeaks.com/blog/how-and-why-to-cultivate-a-healthy-gut-during-pregnancy Pregnancy is a wonderful time in a woman's life when her body transforms into a safe haven and she writes a story about love, excitement, and the joyful birth of a tiny miracle. Every heartbeat, a gentle note creating the harmony of a fresh existence, is created with unwavering love and care as she nourishes the priceless gift inside her womb.  Ensuring the health and nutrition of mothers is critical to ensuring the survival of their babies. Poor maternal health and illnesses can significantly increase the risk of negative outcomes for newborns, including stillbirths, neonatal deaths, and other clinical complications. It becomes clear that promoting the health of moms and their infants requires an integrated approach to maternal health. Pregnancy brings about intentional changes in the body and mind, beyond hormonal fluctuations and uterus growth; these changes facilitate women's smooth and easy transition into the complex role of motherhood on all levels-physical, emotional, and psychological. It brings about several adaptations, such as:- 1. An abrupt increase in the mother's cardiac output, 2. Immunological modifications such as increased T-regulatory cells, which peak in the second trimester, and 3. Modifications to the gut microbiome, which represent the dynamic changes that occur throughout her body. Even though the main factors that significantly shape the microbial makeup of a baby are the mode of birth, breastfeeding, and early-life antibiotic therapy, pregnancy-related maternal variables like nutrition, weight, and antibiotic use are the prenatal factors in the microbiome of the unborn child.  **Image 1- Factors affecting gut health at different stages** The above picture depicts the different factors that affect a newborn’s microbiome. Gestation is the prenatal stage where maternal nutrition, the use of antibiotics, and the placenta affect the gut health of the baby. In the neonatal stage, the gut health of a newborn depends on the mode of birth and gestational age of the mother. Later in the postnatal stage, i.e., the childhood of the baby, multiple factors like milk, geography, diet, host interaction, family, weaning, and exercise or play shape the gut microbiome. Changes in the gut microbiota during infancy can affect long-term risks including obesity, diabetes, and neurological diseases, as well as health outcomes like necrotizing enterocolitis in infancy, which can be impacted by maternal factors, delivery methods, and surroundings components. The makeup of the woman's gut microbiome also has a substantial impact as a result of childbirth, with long-term health consequences for both the mother and the infant. Let us look at some of the significant changes that occur in a mother's body while the fetus develops. **What are the changes taking place in the mother's body?** **Hormonal changes** The delicate balance of hormones is essential for the successful outcome of pregnancy since they are the body's complex messengers, coordinating reactions across different tissues and organs. Many hormones alter significantly throughout pregnancy, each with a specific function. 1. Human Chorionic Gonadotropin hormone (hCG), which is solely generated in the placenta, increases significantly during the first trimester (first 12 weeks), probably contributing to nausea and vomiting. 2. Human Placental Lactogen (hPL) has two functions: it feeds the fetus and stimulates breast milk glands in preparation for future nursing. To maintain a healthy pregnancy, the placenta also produces estrogen, which is essential for the development of female sexual characteristics. 3. Furthermore, progesterone, which is produced by the placenta and the ovaries throughout pregnancy, aids in the thickening of the uterine lining and creates an environment that is favorable for the implantation of a fertilized egg. Significant hormonal changes throughout the first few weeks of pregnancy cause distinct immunological and inflammatory alterations that affect gut microbiota and function. Estrogen and progesterone influence the makeup of the gut microbiome by influencing the bacterial metabolism, proliferation, and virulence of pathogenic bacteria. Gut contractility and transit are also impacted by significant ovarian hormone fluctuations that occur throughout the prenatal and postpartum phases. During pregnancy, the body may release hormones that slow down the rate at which food passes through the stomach and intestines. This might be a beneficial reaction since it gives the body more time to absorb nutrients, allowing the pregnant woman and baby to gain weight. **Metabolic changes** Pregnancy causes significant alterations in the mother's metabolism. Early gestation can be considered an anabolic condition (the building of complex molecules from numerous simple ones) in the mother, with an increase in maternal fat storage and minor changes in insulin sensitivity. As a result, nutrients conserved throughout early pregnancy fulfill the placental and maternal demands of late gestation and nursing. Late pregnancy, on the other hand, is best characterized as a catabolic condition (involving the breakdown of complex molecules into simpler forms) with lower insulin sensitivity (increasing insulin resistance). Maternal glucose and free fatty acid concentrations rise as insulin resistance increases, providing more substrate availability for fetal development. The microbial diversity in the gut during the commencement of pregnancy appears to be comparable to that of non-pregnant women. However, in over 70% of women, the quantity of gut bacteria linked to inflammatory conditions rises as pregnancy progresses. The ratio of specific important bacteria (Firmicutes:Bacteroidetes) in the gut microbiota changes most, resembling the increased concentrations of Firmicutes associated with obesity. Serum levels of proinflammatory cytokines (such as TNF-a, IL-2, IL-6, and IFN-y) also increase, and the mucosal surfaces of the gastrointestinal system exhibit low-grade inflammation. **Immunologic Changes** The mother's immune system undergoes significant changes throughout pregnancy to defend against infections and prevent harmful immunological reactions against the allogeneic fetus (the fetus carrying distinct genetic material from the mother or the combination of genetic material from the mother and father). While there is little evidence that the maternal immune system is universally reduced during pregnancy, elevated risks for some types of illnesses imply significant qualitative immunological alterations. A better understanding of immunological changes during pregnancy may also be essential in determining effective techniques for using vaccinations, such as influenza and pertussis, to protect both the pregnant mother and the newborn. There are many inflammatory states throughout pregnancy, ranging from increased inflammation during implantation and labor to decreased inflammation in the middle of the pregnancy. Although the placental bed has anti-inflammatory qualities that protect the fetus from rejection, the mucosal surfaces of the intestinal tract and other tissues suffer low-grade inflammation with increased amounts of pro-inflammatory cytokines and white cells as the pregnancy progresses. Beyond these few major modifications, women's bodies have undergone several more changes as well. Both the mother and the infant go through a lot of changes from the first day until the baby is born, which affects each differently. While most of the changes are positive for both individuals, a small number of them may increase risk or cause disease. **Risk of developing various ailments during pregnancy due to dysbiosis** 1. The gut microbiome plays a critical role in the renewal of epithelial cells and the maintenance of intestinal integrity, which in turn affects an individual's systemic and intestinal immune systems. Bacteria go from the gut into the bloodstream when the intestinal wall is unhealthy, which worsens systemic inflammation. This movement is caused by an intestinal permeability process known as "leaky gut." * Dysbiosis is thought to be connected to low-grade inflammatory illnesses such as obesity and related ailments, insulin resistance, and Type 2 diabetes, through increased intestinal permeability. High permeability makes it possible for bacteria to seep out and/or for bacterial components, such as the lipopolysaccharides (LPS) found in gram-negative bacteria's cell walls, to cause metabolic endotoxemia, a harmful systemic inflammatory disease. 2. Preeclampsia is a serious problem in obstetrics, affecting 2-8% of pregnancies globally. It is among the most frequent reasons for morbidity and death among expectant women and their children throughout the perinatal stage. Preeclampsia's process is still not entirely understood. Researchers discovered that the abnormal shape of the placenta might harm blood vessels, resulting in preeclampsia during pregnancy. Preeclampsia may be associated with metabolic syndrome, a disorder characterized by impaired glucose and lipid metabolism, insulin resistance, and damage to the blood vessel lining. * Some research suggests a relationship between preeclampsia and the mother's gut flora, with decreased levels of particular beneficial bacteria like Prevotella, Porphyromonas, Varibaculum, and Lactobacillus in women with preeclampsia. Prevotella is a multipurpose bacteria found in the human digestive system. It acts as a defense against microbial illnesses. 3. Fetal Growth Restriction (FGR), also known as Intrauterine Growth Restriction (IUGR), is a common pregnancy issue. It happens when the baby doesn't grow properly in the womb due to reasons like infections, advanced maternal age, malnourishment, genetic problems, or a placenta that can't provide enough nutrients. * Some studies suggest a potential link between the gut bacteria (microbiota) and FGR. In one study of 6,000 pregnant women, Helicobacter pylori was connected to FGR. * Stool samples from pregnant women with FGR showed differences in the proliferation of Bacteroides, Faecalibacterium, and Lachnospira compared to a control group, indicating a possible role of gut bacteria in FGR. 4. In addition to increasing the risk of obesity, type 2 diabetes, and metabolic syndrome, gestational diabetes mellitus (GDM) is the most prevalent metabolic consequence of pregnancy. Blood glucose levels are impacted by metabolic changes that are mediated by the gut bacteria. For many experts, the question of whether intestinal dysbiosis contributes to the development of GDM remains controversial. * As the pregnancy proceeds, the kinds of bacteria in the stomach change, particularly in the middle and late stages. Research on Gestational Diabetes Mellitus (GDM) discovered that people with GDM had more of a kind of bacteria called Firmicutes and fewer of another type called Bacteroidetes. In addition, in the third trimester of pregnancy, persons with GDM had a larger ratio of Firmicutes to Bacteroidetes (F/B) than those without.  **Image 2- Major risk in pregnancy due toincreased pathobionts** The above images tell us about risky conditions for pregnant ladies when there is an increase in pathobionts. Pathobionts are microorganisms such as bacteria, viruses, or fungi that are normally found in the human microbiome but can become pathogenic in particular situations. Insulin resistance, gestational diabetes mellitus, preeclampsia, obesity, and fetal growth restriction are some of the commonly observed risks during dysbiosis. Preventive measures to be taken for a healthier baby's gut 1. Increase your intake of prebiotic foods, including bananas, oats, onions, asparagus, legumes, etc. Additionally, meals containing chemicals, artificial sweeteners, and food colors are bad for your gut health since they interact with probiotics. 2. Reduce your coffee intake in the late afternoon and make time for restful sleep. Establish a nightly routine that communicates to your body when it's time to relax and regenerate. Additionally, try to go to bed and wake up at the same time every day. These techniques, commonly called "sleep hygiene," have a beneficial effect on weariness and stress. 3. Lower your stress levels by engaging in regular exercise, deep breathing, and meditation. It seems to make sense that pregnant women may feel a stronger desire to remain still. Some tend to induce constant sleepiness. While these changes are to be expected, remaining active is crucial for your health and that of the developing fetus. 1. Pregnancy-related exercise may have a positive prenatal impact, fostering a healthy environment in the womb during a crucial stage of organ development. The benefits to offspring can be observed in terms of body weight and composition, cardiovascular health, and nervous system development. 2. Women can adopt healthy lifestyle practices that can improve postnatal health and lower their child's chance of acquiring chronic illnesses, including obesity, diabetes, and cardiovascular disease (CVD). 3. Exercise is excellent for both mental and physical health, and it also improves gut health by lowering inflammation and changing the way the body is built. 4. Exercise also encourages favorable alterations in the makeup of the gut microbiota that are created in the gastrointestinal system. 5. It’s believed that exercise during pregnancy can lead to larger placentas, providing the fetus with more oxygen and nutrients. 6. It’s important to remember that everyone is unique and needs individualized care. The key is to maintain physical activity through exercises that are gentle and that your body is familiar with.  **Image 3- A few things to remember during pregnancy** The above picture talks about some important reminders for the pregnant lady. It is very important to follow a healthy diet routine and avoid processed foods. Daily exercise, meditation, and mindful activities are also necessary to live a happy and peaceful life. Have a good 6-7 hours of sleep and follow up with regular checkups.. **Conclusion** In conclusion, a woman's body undergoes a symphony of changes throughout pregnancy, including modifications to her metabolism, major immunological changes, and hormonal shifts, all intricately linked to the health of both mother and child. The gut microbiome emerges as a pivotal player, influencing long-term health implications. The potential influence of the gut microbiota on maternal and fetal health is highlighted by the connections between dysbiosis and diseases such as gestational diabetes mellitus, fetal growth restriction, and preeclampsia. As we navigate these transformations, embracing preventive measures such as a balanced diet, adequate sleep, stress reduction, and mindful activities becomes paramount. By nurturing gut health during pregnancy, we not only promote a smoother journey into motherhood but also contribute to the well-being of generations to come. So, dear ladies, as you embark on the adventure of motherhood or intend to bring a new life into the world, are you ready to protect not only your health but also that of the precious life growing inside of you by nurturing your gut health? **References** 1\. Lothian, J. A. (2008). The journey of becoming a mother. The Journal of perinatal education, 17(4), 43-47. 2\. Edwards, S. M., Cunningham, S. A., Dunlop, A. L., & Corwin, E. J. (2017). The maternal gut microbiome during pregnancy. MCN. The American journal of maternal child nursing, 42(6), 310. 3\. Gorczyca, K., Obuchowska, A., Kimber-Trojnar, Z., Wierzchowska-Opoka, M., & Leszczynska-Gorzelak, B. (2022). Changes in the gut microbiome and pathologies in pregnancy. International journal of environmental research and public health, 19(16), 9961. 4\. Yao, Y., Cai, X., Chen, C., Fang, H., Zhao, Y., Fei, W., ... & Zheng, C. (2020). The role of microbiomes in pregnant women and offspring: research progress of recent years. Frontiers in Pharmacology, 11, 643. 5. Lain, K. Y., & Catalano, P. M. (2007). Metabolic changes in pregnancy. Clinical obstetrics and gynecology, 50(4), 938-948. 6\. Abu-Raya, B., Michalski, C., Sadarangani, M., & Lavoie, P. M. (2020). Maternal immunological adaptation during normal pregnancy. Frontiers in immunology, 2627. 7.https://www.niddk.nih.gov/health-information/weight-management/healthy-eating-physical-activity-for-life/health-tips-for-pregnant-women --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Unity in Every Bite: A Culinary Exploration of India's Gut Diversity Author: BugSpeaks Published: 2024-01-25 Category: On Diversity Debate Meta Title: Unity in Every Bite: A Culinary Exploration of India's Gut Diversity Meta Description:
In the heart of our old Indian home, filled with colorful flowers and the soothing sounds of nature, Grandma fondly reca URL: https://www.bugspeaks.com/blog/unity-in-every-bite:-a-culinary-exploration-of-india's-gut-diversity In the heart of our old Indian home, filled with colorful flowers and the soothing sounds of nature, Grandma fondly recalled, "Back then, our mornings were like a mix of languages and different traditions. Our meals were a blend of flavors that represented India's diverse culture, keeping us healthy in so many delicious ways." Grandpa, with a hint of nostalgia, added, "We cherished our differences and found togetherness through traditions like Ayurveda, guiding us to a balanced life." And that's why the saying "Anekta me ekta" holds so true for all Indians. Our country is like a colorful mix of cuisines, languages, faiths, and more-a beautiful mosaic that makes it wonderfully diverse. Our daily lives are a celebration of these differences, from the spices in our curries to the various languages telling our stories. But it's not just about what you see on the outside; even our stomachs tell a tale of India's many flavors. The food we enjoy adapts to our different habits, showing the unique variety in our digestive systems. This internal harmony highlights our strength and proves that diversity doesn't break unity; it thrives within it. In every taste, language, and heartbeat, we find the essence of our incredible country. **Gut microbiome profile of Indians:** The "Landscape of Gut Microbiome - Pan-India Exploration" (LogMPIE) project is the first large-scale, national record of the Indian gut microbiome. Determining and mapping the baseline Indian gut microbiota was the main goal of the research. This observational research was carried out in 14 different Indian regions. The participants who enrolled were evenly dispersed in terms of body mass index (obese and non-obese) and geographic regions (north, east, west, and south). In addition, the research design took into account variables like age and physical activity that affect the microbiome. The LogMPIE study collected data from 1004 eligible participants and identified 993 distinct microbes from the Indian microbiome diaspora. The predominant species of microbes found in the Indian gut are Prevotella copri and Faecalibacterium prausnitzii. Other highly abundant microbes in Indians compared with people living elsewhere in the world include Mitsuokella, Lactobacillus, and Megasphaera. The Prevotella were found in the gut of people who consumed plant-based diets. Lactobacillus, Megasphaera, and Mitsuokella were dominant in people whose diets contained fermented foods and dairy products. The information serves as a valuable resource for research, analysis, and identification of signs defining the participants' physiological dispositions, in addition to mapping the baseline gut microbiota of India. Moreover, they shed light on the distinctive characteristics that describe the Indian microbiome. **Region-wise gut microbiome difference** In North India, indulgence in fluffy naan, creamy dal makhani, and spicy tandoori delights creates a gastronomic landscape rich in probiotics and prebiotics. The gut health of North Indians oscillates between climatic problems and spicy strengths. It feeds on foods high in fiber and strong spices to produce good bacteria. Summer heat and water problems, however, are hazards. The gut microbiota of North Indians is shaped by a special combination of dietary diversity and environmental challenges, which makes it an interesting field for future study and supports customs for gut health. * They primarily consume a plant-based diet, which is associated with Prevotella and shows enrichment of branched-chain amino acid (BCAA) and lipopolysaccharide biosynthesis pathways. A multicenter study involving healthy individuals from urban northern India (Delhi) and adjacent rural regions suggested 50 core operational taxonomy units (OTUs) among which Prevotella, Megasphaera, Faecalibacterium, Lactobacillus, Ruminococcus, and Roseburia were the most dominant. Venturing southward, the lush embrace of Kerala unfolds a culinary narrative featuring dosa and sambar infused with the tropical essence of coconut. Fermented rice dishes like idli and appam contribute prebiotics, fostering a healthy gut microbiota. However, the melody isn't always perfect. The microbial equilibrium can be upset by water problems in some places, and the urban beat of processed meals jeopardizes the peace. Nonetheless, the South Indian stomach still has a lot going for it in terms of its traditional tastes and abundance of plant-based foods; it just needs to be deeply studied and supported for best health. * They mainly consume an omnivorous diet, which shows associations with Bacteroides, Ruminococcus, and Faecalibacterium and has an enrichment of short chain fatty acid (SCFA) biosynthesis pathway and branched-chain amino acid (BCAA) transporters. The concentration of BCAAs was found to be higher in the fecal metabolome of the Southern India cohort and shows a positive correlation with the higher abundance of BCAA transporters. Another study found that the microbiome of participants from Kerala was enriched in species of Bifidobacterium, Ruminococcus, Clostridium, and Faecalibacterium. The Western tales, from Goa's sun-kissed coasts to Gujarat's arid expanses, showcase a fresh turn in the gut health saga. Fresh fish from Goa and Gujarat offer omega-3 fats and fiber, while besan meals contribute beneficial bacteria and protein. Although very popular, the spicy food of Gujarat may cause upset stomachs, and consuming too much oil might worsen digestive problems. In Maharashtra, the stress and sedentary lifestyles of cities like Mumbai can worsen the effects of urbanization on diets, problems with water quality, and some cultural practices on gut health. * The gut microbiome of individuals from the Western region of India, specifically Ahmedabad city, is dominated by a Prevotella microbial community. The typical diet included carbohydrate and fiber-rich components (predominantly whole grains and legume-based preparations), which appear to validate the proposed correlation between diet, geography and microbiome composition. Journeying eastward, fermented foods and spice-infused music set the beat for East Indian intestinal wellness. The delicious chorus of Dahi restores intestinal flora and strengthens immunity. Turmeric adds anti-inflammatory overtones to fish curries like Machher Jhol, balancing intestinal health. The fiber bassline of Litti Chokha keeps you full and facilitates easy digestion. It might be difficult for East Indian people to keep their gut flora healthy due to the restricted availability of clean water. Moreover, access to foods that are good for the stomach is hampered by socioeconomic gaps, which are characterized by poverty and malnutrition and exacerbate gut health problems in these areas. Along with the food, the climatic conditions and lifestyle of the people also affect the gut health of people in all these regions. **  ** **Img 1- Indian diversity in food** **The image above describes some of the most well-known foods from the various Indian states.** **[https://www.humanium.org/en/indian-food-diversity/](https://www.humanium.org/en/indian-food-diversity/)** **Why are Indians facing more gut issues now than ever?** From birth, the gut microbiome undergoes a complex journey influenced by various factors, with minimal diversity in early life giving way to stability and variety in adulthood. * Contemporary challenges, including a rising BMI (Body Mass Index) due to processed food consumption influenced by Western fast-food chains and sedentary lifestyles, disrupt the delicate balance of the microbiota. This disruption results in an imbalance of harmful and beneficial bacteria, altering gut flora, leading to dysbiosis, and impacting metabolism, food absorption, and overall digestive health. The prevalence of constipation, bloating, and diarrhea underscores the toll of low-fiber, processed foods on gut functionality. * The prevalence of gut health issues among Indians is a significant concern, as evidenced by a surge in non-communicable diseases such as diabetes, cancer, cardiovascular diseases, stroke, and hypertension. The roots of these issues delve into lifestyle choices, heredity, and the evolving gut microbiome. It is surprising to note that over 61% (5.87 million) of all deaths in India are due to lifestyle & and non-communicable diseases. **What’s the solution?** Addressing the escalating gut health challenges among Indians necessitates a return to ancient food techniques and the incorporation of daily practices such as increased physical activities, mindfulness, and self-care activities following regular routines. Traditional Indian meals, abundant in prebiotics, serve as nutrition for specific gut bacteria, fostering the growth of beneficial microflora. This, in turn, positively impacts the individual, leading to the production of metabolic products that lower blood cholesterol, regulate stomach pH, stimulate the immune system, and enhance mineral absorption. To understand whether we have enough of the beneficial microbes, getting a gut health test becomes important in checking the microbiome composition. Fortunately, Leucine Rich Bio has South Asia’s first microbiome-based test called Bugspeaks, which identifies the microbiota profiles and predicts susceptibility to over 20 diseases by analyzing the stool samples and giving detailed personalized nutritional recommendations. In the fast-paced modern era, where time is a luxury, it is crucial to recognize the role of exercise as an environmental component influencing gut microbial composition. Eating a diverse range of foods increases the diversity of the microflora and improves the Bacteroidetes-Firmicutes ratio. It emerges as a potent tool to mitigate weight-related pathologies, gastrointestinal disorders, and the incidence of metabolic and obesity-related diseases. Regular physical activity is a therapeutic measure to maintain microbial balance, correct dysbiosis, and improve overall health. **  ** **Img 2- Healthy Indian Thali** Behold the gastronomic masterpiece! This picture encapsulates the epitome of an Indian feast, a symphony for a flourishing gut microbiota. Every morsel tells a tale of nutrient harmony, a culinary orchestra ensuring balance in the body's nutritional saga. It's not just a meal; it's the art of wellness on a plate, where diversity reigns supreme, promising a saga of robust health through the magic of varied, wholesome Indian delicacies. [https://www.bing.com/images/create/indian-diet/1-65b15e6261bf4b33bf52c95d3c5e842b?id=9ZYuBB%2fXZSDeCw%2fVcUlFsQ%3d%3d&view=detailv2&idpp=genimg&noidpclose=1&FORM=SYDBIC](https://www.bing.com/images/create/indian-diet/1-65b15e6261bf4b33bf52c95d3c5e842b?id=9ZYuBB%2fXZSDeCw%2fVcUlFsQ%3d%3d&view=detailv2&idpp=genimg&noidpclose=1&FORM=SYDBIC) **Conclusion** The rich mosaic of India's many civilizations, climates, and culinary traditions greatly adds to the country's uniqueness. Every region has a unique variety of meals that affect the gut microbiota, from the rich and spicy cuisines of the North to the coconut-infused delicacies of the South, and from the seafood joys of the West to the rice-centric pleasures of the East. However, the increasing number of non-communicable illnesses among Indians highlights how urgent it is to address gut health concerns. The makeup of the gut microbiome is seriously threatened by the trend towards processed meals, reduced physical activity, and the ensuing rise in BMI. An imbalance or dysbiosis can occur, which can cause inflammation, metabolic issues, and digestive difficulties. To overcome these obstacles, Indians must return to their traditional diets, with a focus on foods high in prebiotics and getting their gut tested. Including regular exercise in daily routines can also have a favorable impact on gut bacteria diversity, which in turn promotes general well-being. So, don’t you think the best way to embrace the uniqueness of India is to incorporate healthy food choices from different regions into our diets because increasing diversity in our plates ensures diversity in our gut and lessens the increasing number of Indians suffering from digestive disorders? Therefore, this Republic Day, let's get back to our traditional nutrition and healthy lifestyle practices and enjoy this rich and diverse India every day! **References** * Shetty, S. A., Marathe, N. P., & Shouche, Y. S. (2013). Opportunities and challenges for gut microbiome studies in the Indian population. Microbiome, 1(1), 1-12. * Dhakan, D. B., Maji, A., Sharma, A. K., Saxena, R., Pulikkan, J., Grace, T., ... & Sharma, V. K. (2019). The unique composition of Indian gut microbiome, gene catalogue, and associated fecal metabolome deciphered using multi-omics approaches. Gigascience, 8(3), giz004. * Kedia, S., Rampal, R., Paul, J., & Ahuja, V. (2016). Gut microbiome diversity in acute infective and chronic inflammatory gastrointestinal diseases in North India. Journal of gastroenterology, 51, 660-671. * Samanta, A. K., Kolte, A. P., Senani, S., Sridhar, M., & Jayapal, N. (2011). Prebiotics in ancient Indian diets. Current Science, 43-46. * Monda, V., Villano, I., Messina, A., Valenzano, A., Esposito, T., Moscatelli, F., ... & Messina, G. (2017). Exercise modifies the gut microbiota with positive health effects. Oxidative medicine and cellular longevity, 2017. * Dubey, A. K., Uppadhyaya, N., Nilawe, P., Chauhan, N., Kumar, S., Gupta, U. A., & Bhaduri, A. (2018). LogMPIE, pan-India profiling of the human gut microbiome using 16S rRNA sequencing. Scientific data, 5(1), 1-10. * [https://timesofindia.indiatimes.com/blogs/voices/gut-health-matters-tackling-indias-alarming-rise-in-non-communicable-diseases-from-the-inside-out/](https://timesofindia.indiatimes.com/blogs/voices/gut-health-matters-tackling-indias-alarming-rise-in-non-communicable-diseases-from-the-inside-out/) * [https://www.hindustantimes.com/fitness/a-guide-to-caring-for-the-indian-gut/story-vdbrFbXP6ZQac5ioDnePnI.html](https://www.hindustantimes.com/fitness/a-guide-to-caring-for-the-indian-gut/story-vdbrFbXP6ZQac5ioDnePnI.html) * [https://himalayarestaurant.com.au/the-health-benefits-of-traditional-indian-food/](https://himalayarestaurant.com.au/the-health-benefits-of-traditional-indian-food/) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## New Year's resolution for a healthier gut Author: BugSpeaks Published: 2024-01-10 Category: Microbiome and Lifestyle Meta Title: New Year's resolution for a healthier gut Meta Description:
The year 2024 has already kick-started and we're sure there must be so many of you who have already made your New Ye URL: https://www.bugspeaks.com/blog/new-year's-resolution-for-a-healthier-gut The year 2024 has already kick-started and we're sure there must be so many of you who have already made your New Year's resolutions and are ready to have an amazing year ahead! A New Year's resolution is a promise to yourself to do something differently, isn't it? Many of us set the resolution to make better lifestyle choices, to learn something new, to lose weight, to spend more time with family, and the list goes on. But we all commonly need to set a New Year's resolution for a healthier gut! Gut health plays a vital role in everyone's life, as it impacts our overall health and well-being. So let's dive in to see why we need to make such a resolution in 2024. **Why make it?** Our gut microbiota - the system of microorganisms in a person's gastrointestinal tract-has been identified as an essential 'super-organism' in numerous scientific and clinical studies. This 'super-organism' is the diverse and dynamic range of the microbial ecosystem within our gut. Every individual has a unique gut ecosystem consisting of more than 1000 species of microorganisms, predominantly bacterial species. This builds right after the birth of a child when the colonization of the gastrointestinal (GI) tract starts, and it evolves and changes over the individual's life. The adult human GI tract consists of several billion microbes, varying in composition along the length of the GI tract, demonstrating that certain bacteria tend to adhere to the mucosal surface while others are in the lumen (inside the tract). The bacteria at the mucosal surface, or what can be said as within the mucus layer, participate in the interactions with the host immune system, and those in the lumen are more likely to be involved in the metabolic interactions that are products of digestion. Our microbes are responsible for the metabolism of undigested food remnants as they help to digest dietary fiber and polyphenols. Also, they can supply essential nutrients, synthesize vitamin K, aid in the digestion of cellulose, and promote angiogenesis - the growth of blood vessels from the existing blood vessels. Thus, our unique gut composition supports our body's functions by breaking down the food we eat and absorbing the nutrients from it. To know your unique mixture of microorganisms in the gut, learn about our [gut microbiome test,](https://www.bugspeaks.com/pricing) understand several factors about your health and wellness, and take advantage of our new year's 15% offer using coupon code: [Flat15](https://www.bugspeaks.com/pricing) !! Sometimes, our gut may undergo abnormal changes or imbalances between the good and bad microbes, called dysbiosis, leading to many diseases with initial symptoms like bloating, abdominal pain, and diarrhea. The bacteria _Anaerotruncus colihominis, Ruminococcus callidus,_ and _Lachnospira pectinoschiza_ were highly observed in the bloating cases. An abundance of Bifidobacteria may have a negative correlation with abdominal pain. When there is a reduction in the number of members of the genus _Streptococcus_, diarrhea is observed. There are a few other highly observed diseases, like inflammatory bowel diseases, obesity, diabetes, liver diseases, chronic diseases, cancers, HIV, and autism, that show a correlation with the microbial composition of our gut.  **Source :- Figure 1 - Gut microbial strains and negative health outcomes of gut microbial dysbiosis.** **Source : Reference Paper \[8\]** **The image above depicts the many species of microorganisms found in various parts of the GI tract. An individual's gut health is closely linked to the function of the heart, brain, kidney, muscle, bones, liver, lung, pancreas, and skin, among other vital organs. The image explains how the dysbiosis of the gut microbiota can impact each organ in the body, resulting in various issues and illnesses.** **Benefits of a healthy gut** As we discussed, gut health not only affects your digestion activities but also your whole body. So, we can say that a healthier gut microbiome: 1. Aids Digestion: Diverse gut bacteria facilitate complete digestion and nutrient utilization from complex carbs and proteins. 2. Promotes Immune Health: A healthy gut flora acts as a defense against pathogens, releasing antimicrobial substances and preventing infections. 3. Supports Bone Health: Beneficial bacteria in a healthy gut may enhance calcium absorption, contributing to overall bone health. 4. Maintains Healthy Weight: A balanced microbiome plays a role in supporting a healthy weight, associating a diverse microbial composition with weight management. 5. Promotes Brain Health: The gut-brain axis communication is influenced by certain gut microbes producing neurotransmitters, potentially impacting mood and cognition. **How do I plan the resolution?** Before planning for a healthier gut, we need to know what factors affect gut health. It is commonly known that our diet majorly affects the gut microbiome-not just diet, but our lifestyle, physical activities, genetics, age, gender, mental health, the medicines we consume, and the environment we live in can all have an impact on our gut. **Healthy food consumption: - Prebiotics and Probiotics** It is highly recommended to consume probiotics or live beneficial microorganisms for a healthier gut by the Food and Agricultural Organisation of the United Nations and the World Health Organization. Probiotics balance the gut microbiome and help stabilize microbial communities. Fermented foods and drinks are considered sources of probiotics, for example, yogurt, kefir, sauerkraut, tempeh, kimchi, miso, kombucha, kanji, some types of cheese, idli, dosa, buttermilk, pickles, and chutneys, the rare plant-based probiotic green peas, etc. According to many studies, fiber, a nutrient derived from plants, lowers the risk of metabolic illnesses by promoting the diversity and proliferation of beneficial bacteria in the gut. Foods like beets, carrots, garlic, banana, oats, onions, sweet potatoes, spinach, etc. are natural sources of fiber, also called prebiotics, that improve the gut flora. Whole grains are a great source of fiber, in addition to fruits and vegetables. **Exercise** Exercise benefits the microflora by enriching its diversity. The Firmicutes/Bacteroidetes ratio reflects the balance between two major bacterial phyla in the gut microbiome, influencing aspects of human physiology and gut health. Exercise improves the Bacteroidetes-Firmicutes ratio, which may help reduce weight and gastrointestinal disorders; encourages the growth of bacteria that can modulate mucosal immunity and improve intestinal barrier functions, which may help reduce the incidence of metabolic and obesity-related diseases; and encourages the growth of bacteria that can produce compounds like short-chain fatty acids that protect against gastrointestinal disorders and colon cancer. As a result, exercise may be used as a treatment to keep the gut in balance or to correct any dysbiosis that may develop, improving overall health. **Mental Health** Our mental health plays a very significant role in improving our overall well-being. A person with balanced mental health can be 10 times more efficient in their daily activities and can do better in their lifestyle. Research indicates a bidirectional communication between the central nervous system and gut microbiota, revealing the relationship between the gut microorganisms and the gut-brain axis. Anxiety and depression, two conditions that are becoming common in today's world, have been related to dysbiosis and inflammation of the stomach. The release of hormones during fear or anxiety can affect the gut flora, potentially aiding digestion while suppressing antibody production. So, hormonal imbalances can lead to various gastrointestinal disorders. Foods containing fiber, vitamin D, proteins, and omega-3s can help boost the gut as well as mental health. Along with that, practicing mindfulness activities like meditation and yoga; mind-relaxing activities like music therapy, breathing exercises, and talking to our loved ones; having a good sleep schedule; and maintaining a work-life balance are important for good mental health. Therefore, these three key elements-consumption of healthy food, regular exercise, and keeping our mental health a priority-all work together to support a healthy lifestyle and a longer, more productive life. Based on these factors, you can plan for a healthier gut. 1. Eating mindfully and maintaining a balanced diet with optimum amounts of good fats, plant carbohydrates and protein, prebiotics, and probiotics. Our BugSpeaks dietary recommendations are completely personalized for your gut, which ensures better healing of the gut. 2. Avoid emotional eating. You can maintain a diary of how you feel before and after eating any food; that way, you can keep track of your cravings vs. your actual hunger! 3. Avoid high-sugar, processed, and packaged food items. They may affect gut microbial balance, contribute to inflammation, and disrupt intestinal lining integrity, resulting in digestive disorders and a weakened immune system. 4. Create a good schedule for each task you want to complete, and manage your time, as it promotes better digestion, lowers stress, and permits regular eating patterns and consistent meal planning. An organized schedule makes it easier to find time for rest and exercise, which supports a balanced lifestyle and intestinal health in general. 5. Including some kind of physical activity in everyday routines as it improves gut health by increasing the diversity and number of healthy gut bacteria. Exercise increases the formation of short-chain fatty acids, which promotes a healthy gut environment, increases blood flow to the intestines, and aids in the regulation of bowel motions. 6. Start journaling to calm your mind. Journaling offers a therapeutic outlet by recording thoughts and feelings, promoting emotional well-being that is linked to a better gut-brain connection. 7. Getting at least 6-7 hours of sleep because it corresponds to the circadian rhythms of gut flora. A healthy sleep schedule helps the body's internal clock stay in sync, which controls a number of physiological functions, including the activity of gut microorganisms. Our living conditions should allow us to preserve the harmony of the elements influencing our gut health.  **Image 2- To do list for Healthier gut Resolution 2024** **The above image displays the To Do List for the Healthier gut resolution of this year. Hydration and Exercise are a must for a good start. The diet should include the regular consumption of Prebiotics and Probiotics. Add the habit of Daily journaling and 6-7 hours of good sleep for Better Mental health.** **Conclusion** Adopting a New Year's resolution for a healthy gut is a critical step that may have a big influence on overall well-being. The gut is home to a diverse microbial population that affects a wide range of human functions. Formulating a successful resolution requires an understanding of the components that impact gut health. In addition to food preferences, other important aspects include heredity, age, gender, physical activity, mental health, medicines, and environmental circumstances. For the best outcomes, a comprehensive strategy must be put into practice. Beyond improved digestion, a healthy gut also affects immune system function, bone health, weight control, and even cognitive abilities. Making gut health a priority can benefit people's general health and wellbeing. Let our goals for 2024 represent a dedication to nourishing our "super organism" and discovering the numerous advantages that a healthy gut has to offer. Let us cheer for a year full of energy, perseverance, and success! **References** 1\. Quigley, E. M. (2013). Gut bacteria in health and disease. Gastroenterology & hepatology, 9(9), 560. 2\. Aziz, Q., Doré, J., Emmanuel, A., Guarner, F., & Quigley, E. M. M. (2013). Gut microbiota and gastrointestinal health: current concepts and future directions. Neurogastroenterology & Motility, 25(1), 4-15. 3\. Flint, H. J., Scott, K. P., Louis, P., & Duncan, S. H. (2012). The role of the gut microbiota in nutrition and health. Nature reviews Gastroenterology & hepatology, 9(10), 577-589. 4\. Zmora, N., Suez, J., & Elinav, E. (2019). You are what you eat: diet, health and the gut microbiota. Nature reviews Gastroenterology & hepatology, 16(1), 35-56. 5\. Jovel, J., Dieleman, L. A., Kao, D., Mason, A. L., & Wine, E. (2018). The human gut microbiome in health and disease. Metagenomics, 197-213. 6\. Monda, V., Villano, I., Messina, A., Valenzano, A., Esposito, T., Moscatelli, F., ... & Messina, G. (2017). Exercise modifies the gut microbiota with positive health effects. Oxidative medicine and cellular longevity, 2017. 7\. Clapp, M., Aurora, N., Herrera, L., Bhatia, M., Wilen, E., & Wakefield, S. (2017). Gut microbiota's effect on mental health: The gut-brain axis. Clinics and practice, 7(4), 987. 8\. Afzaal M, Saeed F, Shah YA, Hussain M, Rabail R, Socol CT, Hassoun A, Pateiro M, Lorenzo JM, Rusu AV and Aadil RM (2022) Human gut microbiota in health and disease: Unveiling the relationship. Front. Microbiol. 13:999001. doi: 10.3389/fmicb.2022.999001 --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## The Year of Millets: Reflecting on Why Millets Took Over All Other Grains in 2023 Author: BugSpeaks Published: 2023-12-29 Category: Nutrition Meta Title: The Year of Millets: Reflecting on Why Millets Took Over All Other Grains in 2023 Meta Description:
In the dynamic realm of dietary trends, millets have seized the spotlight, emerging as a nutritional powerhouse with myriad benefi URL: https://www.bugspeaks.com/blog/the-year-of-millets:-reflecting-on-why-millets-took-over-all-other-grains-in-2023 In the dynamic realm of dietary trends, millets have seized the spotlight, emerging as a nutritional powerhouse with myriad benefits for gut health. As we delve into the numerous advantages of millets, explore the most consumed varieties in 2023, and understand their pivotal role in addressing prevalent health conditions, it becomes evident that millets are not merely a food choice but a key player in fostering overall well-being.  **Discover the joy of a happy gut through Super Food - Millets. Explore the main millets and their benefits. #HealthyLiving** The image showcases key millets that are commonly consumed and readily available, highlighting their benefits for gut health. 1. Foxtail Millet: Boosts digestion with fiber, preventing constipation, and nurturing gut health. 2. Little Millet: maintains a healthy gut with high fiber, reducing digestive risks. 3. Barnyard Millet: Fosters beneficial gut bacteria for enhanced digestive well-being. 4. Pearl Millet: Supports gut health with insoluble fiber, promoting regular bowel movements. 5. Proso Millet: Enhances digestion, prevents constipation, and maintains a balanced gut microbiota. 6. Finger Millet: Promotes gut health with abundant fiber and essential nutrients, preventing gastrointestinal issues. Include these millets for a healthier gut and balanced microbiome. **What are Millets?** Millets are a group of small-seeded grasses widely grown around the world as cereal crops or grains for fodder and human food. They belong to the Poaceae family, commonly known as the grass family. Millets are highly tolerant of extreme weather conditions and have a similar nutrient content to other major cereals. They can be grown even in regions experiencing drought since they require less water for growing, also it is naturally pest-resistant, hence not many chemicals are used to grow them. **Most Consumed Millets** In 2023, the most consumed millets were Bajra (Pearl millet), Ragi (Finger millets), Kangni (Foxtail Millet), Kodra (Kodo millet), Jowar (Sorghum Millet), Kutki (Little Millet), Sanwa (Barnyard Millet). Each variety brings unique health benefits to the table, from the antioxidant properties of Kodo millet to the blood sugar-regulating effects of foxtail millet. Pearl millet's triglyceride-lowering capabilities and finger millet's role as a gluten-free substitute showcase the diverse nutritional profile of millets. **The Global Recognition of Millets** With the United Nations designating 2023 as the International Year of Millets, these ancient grains enjoyed a renaissance on the global stage.. Originating in China over 5,000 years ago, millets have been a staple in India and parts of Africa for generations. The resilience of millets as a staple diet for tribal and rural populations is evident in traditional dishes like jowar roti (sorghum flatbread) and ragi mudde (steamed balls made from finger millet), reflecting a rich culinary heritage. **Nutritional Prowess of Millets** Millets stand out for their nutritional density, boasting fiber, protein, vitamins, and essential minerals like calcium, zinc, and iron. Bajra, in particular, outshines traditional grains like rice, wheat, or maize in protein, iron, and zinc content. This nutritional richness makes millets a valuable addition to diets, especially in regions grappling with vitamin deficiencies. **  ** **Millets: Rich in antioxidant phenolic compounds, gluten-free proteins, and heart-healthy fatty acids. Harness their dietary fibre, vitamins, and essential minerals to stave off deficiencies and lower the Glycemic Index. Elevate your nutritional intake with these scientifically proven benefits.** This image highlights the nutritional content of millets. 1. Lipids: Abundant in heart-healthy unsaturated fatty acids. 2. Proteins: Gluten-free and replete with essential amino acids. 3. Phenolic Compounds: Exhibit antioxidant, antiviral, and anti-inflammatory properties, mitigating risks of Cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. 4. Minerals and Vitamins: Effectively address deficiencies in calcium, iron, and zinc. 5. Dietary Fibres: Reduce the Glycemic Index, serving as prebiotics and fostering the production of short-chain fatty acids. **Millets and Gut Health: A Symbiotic Relationship** The increase in millet production worldwide is a result of nutritional needs as well as a proactive effort toward improving global health. The nutritional value of millets is influenced by the processing procedures used, such as decortication, soaking, malting, milling, and fermentation. Hence, millets have a good impact on gut microbiota composition, which is an important factor in gut health. Millets function as prebiotics, providing nourishment to the beneficial bacteria in our gut. They foster a healthy digestive system by promoting the growth of beneficial gut bacteria like Lactobacilli and Bifidobacteria. Additionally, millets have been found to dose-dependently increase levels of Bifidobacterium and Lactobacillus and decrease levels of Escherichia coli, Enterococcus, and Bacteroides in the intestine. This balance is essential for optimal digestive function and overall health. So, Millets' bioactive compounds influence the development and diversity of gut flora. They are an excellent choice for promoting intestinal health, minimizing the risk of diseases such as colorectal cancer, and alleviating constipation due to their high fiber content, prebiotic properties, and resistant starch. Millets are a gluten-free alternative for people with celiac disease or gluten intolerance, which broadens their appeal. **Millets’ role in improving overall health** As we reflect on prevalent health issues in 2023, including mental health concerns, chronic diseases, and infectious diseases, millets emerge as versatile allies. The slow-digesting carbs in millet contribute to stable blood sugar levels, potentially reducing anxiety. The fiber and antioxidant-rich composition aid in regulating diabetes, with the low glycemic index providing additional benefits for heart health. Millets, enriched with zinc and iron, actively contribute to bolstering the immune system to combat infectious diseases. **  ** **A diagram of the human body with organs made with millets, showcasing its golden grains and highlighting the nutritious and versatile nature of this gluten-free grain in promoting overall human health.** This image highlights the various health benefits of millets. 1. High Nutritional Value: Rich in protein, fiber, and essential minerals, it has a low glycemic index, making it suitable for stable blood sugar levels. 2. Boosts Immunity: The protein content supports immune system development, reducing the risk of diseases. 3. Reduces Cardiovascular Risks: Essential fats in millets prevent excess fat buildup, lowering the risk of high cholesterol and heart problems. 4. Aids Weight Loss: Low-calorie content and prolonged satiety help in weight management. 5. Prevents Asthma: Magnesium in millets can lower migraine and asthma symptoms, free from allergens causing asthma. 6. Acts as an Antioxidant: Millets' antioxidant properties help detoxify the body and neutralize enzymatic activity. 7. Prebiotic Properties: Millet's prebiotics promote the growth of good gut bacteria, enhancing overall gut health. 8. Helps in Digestion: High fiber content aids digestion, reducing bloating, gas, and constipation for a healthy digestive system. As we reflect on prevalent health issues in 2023, including mental health concerns, chronic diseases, and infectious diseases, millets emerge as versatile allies. The slow-digesting carbs in millets contribute to stable blood sugar levels, potentially reducing anxiety. The fiber and antioxidant-rich composition aid in regulating diabetes, with the low glycemic index providing additional benefits for heart health. Millets, enriched with zinc and iron, actively contribute to bolstering the immune system to combat infectious diseases. **Conclusion** In conclusion, the popularity of millets like pearl, foxtail, kodo, sorghum, small, barnyard, and finger millet in 2023 signifies a shift towards a more conscious and nutritious dietary choice. As we bid farewell to traditional dietary norms and welcome the new year 2024, millets emerge as a beacon of nutrition, offering an array of health benefits. From fostering gut health to addressing prevalent health concerns, millets are more than just a culinary delight; they are a nutritional revolution that reminds us to embrace a healthier, more sustainable lifestyle. So, let millets grace your plate and pave the way for a future where well-being is not just a choice but a way of life. Here's to millets, here's to digestive vitality, and here's to the promise of a nourished and harmonious tomorrow! **References** 1. Mohod, N. B., Borah, A., Goswami, P., Koshariya, A. K., & Sahoo, S. (2023). The International Year of Millet 2023: A Global Initiative for Sustainable Food Security and Nutrition. International Journal of Plant & Soil Science, 35(19), 1204-1211. 2. Sabuz, A. A., Rana, M. R., Ahmed, T., Molla, M. M., Islam, N., Khan, H. H., ... & Shen, Q. (2023). Health-Promoting Potential of Millet: A Review. Separations, 10(2), 80. 3. Singh, V., Lee, G., Son, H., Amani, S., Baunthiyal, M., & Shin, J. H. (2022). Anti-diabetic prospects of dietary bio-actives of millets and the significance of the gut microbiota: A case of finger millet. Frontiers in Nutrition, 9, 1056445. 4. https://www.potsandpans.in/blogs/articles/millets-and-the-gut-microbiome-a-healthier-digestive-system#:~:text=Millets%20play%20a%20role%20in,and%20support%20overall%20gut%20health 5. https://www.hindustantimes.com/lifestyle/health/superfood-in-news-7-millets-that-were-all-the-rage-in-2023-101702901188058.html --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Environmental Effects on Microbes and Consequences of Gut Health Author: BugSpeaks Published: 2023-10-06 Category: Microbiome Meta Title: Environmental Effects on Microbes and Consequences of Gut Health Meta Description:
As India is a culturally diverse nation with a myriad of anthropological findings, the natural environment that encompasses it is URL: https://www.bugspeaks.com/blog/environmental-effects-on-microbes-and-consequences-of-gut-health As India is a culturally diverse nation with a myriad of anthropological findings, the natural environment that encompasses it is home to a vast number of microorganisms. Each environment is unique towards how an individual's health is affected as a result of different factors. This can be observed as certain environments can cause diseases to an individual who grew up in a different place as opposed to an individual who has been living in that environment their whole life. Likewise, certain organs in the human body tend to be affected the most when compared to others due to being the target site of either a pathogen or microbe. While individuals are affected differently as a result of their environment, how each individual is affected as well as the effects they suffer is also related to genetics. This can be observed comparatively with smokers and nonsmokers. Despite proof from scientific research that smoking indeed causes cancer, some individuals are seen to be chain-smokers and live a long life without any severe health complications. On the other hand, individuals who are observed to be maintaining a healthy lifestyle through incorporating exercise and diet in their daily lives, have been reported to suffer various diseases and even cancer (Kusnierczyk 2023).  An individual's genetic makeup is composed of their DNA and which genes are expressed while others are not. For instance, receptors for a certain disease will render an individual unaffected due to not being expressed. Due to that specific gene not being expressed, there is no receptor for a certain disease and the body is not applicable towards being infected. Moreover, gene expression serves as an example of intraspecies variability when certain microbes exist in an individual and not in another despite living in the same environment. When an individual is exposed to food or water in an environment through ingestion, the organ most affected is the gut. For example, individuals who have ingested water from an open source tend to experience symptoms of diarrhea as well as stomach indigestion. Through affecting the gut and exposing it to certain conditions, microorganisms will remain there and the body will eventually adapt to microbes of the same species that exist in that environment. Nevertheless, microbiomes in the gut contain a mutualistic relationship without the human body as they are important in carrying out several body processes and in turn gain a home to thrive and survive while living in the gut. Some of these processes carried out by microbes in the gut include biotransformation of conjugated bile acids, synthesis of certain vitamins, and degradation of dietary oxalates (Kelsen et al 2012). However, there are several occasions in which microbes can cause complications, requiring physicians to know which specific microbe is problematic to provide remedial solutions. Many microbes can thrive in certain conditions while others are unable to due to their genetic makeup. Similar to humans, while many have adapted to living in cold climates, individuals who grew up in warm arid climates are unable to adapt effectively to their surroundings and environment. Conditions that determine whether a microbe is able to effectively thrive in their environment include humidity, temperature, oxygen content and various pollutants (Singh et al 2016). As discussed earlier, regardless of environmental conditions, microbe type and specificity play a significant factor in how they exist in their environment. Moreover, this is detrimental to the gut of an individual predisposed to an environment their entire life since birth. For example, despite living in a humid environment with a warm climate, many individuals are seen to have been observed suffering from a disease that was believed to be less effective. This is due to the fact that the microbe contains several adaptable characteristics allowing it to survive in environments that prove otherwise fatal (Singh et al 2016). Due to the variability of different microbes as well as the conditions they are able to effectively survive, this creates the necessity of remedies from pharmaceutical companies. Many healthcare companies are able to determine remedies for microbes through surveying the different complications and symptoms an individual may be experiencing. For example, Leucine Rich Bio, a healthcare company based in India aims to determine remedies for people experiencing complications from the microbiome centered in the gut. Leucine Rich bio was able to implement a platform called BugSpeaks that is able to effectively analyze gut microbial sequencing data and create actionable solutions. BugSpeaks functions by primarily determining what microbe is causing complications in the gut as well as serving as a potential index for which microbe physicians and pharmacists would be potentially dealing with (Leucine Rich Bio 2023). This drastically reduces the need for more invasive tests being conducted when doctors test patients from symptoms they encounter. In addition to the microbe index, BugSpeaks also provides a disease risk index and three phase nutritional recommendations. The disease risk index acts as a measure for individuals to determine the severity of their condition and encourage them to seek medical assistance sooner before complications worsen (Leucine Rich Bio 2023). As discussed earlier, the three phase nutritional recommendations work hand in hand with the disease risk index and represent actionable solutions which can be implemented. The three phase nutritional recommendations monitor the individual over a course of time and determines how a certain solution or remedy should be altered. Genome sequencing is a procedure first conducted from a sample obtained by an individual in order to determine the exact problem they may be facing. Genome sequencing is an effective tool which helps with finding the genetic signature of a certain microbe. After this is done, physicians and pharmacists are able to efficiently determine what problems and complications arise from the specific microbe the individual encounters. They can then provide medications and treatment plans accommodating the individual. Due to the ability of microbes to exist in environments that would otherwise prove fatal to them, BugSpeaks provides a manner of identifying these microbes efficiently. Furthermore, through properly identifying microbes, individuals will maintain a faster recovery period without further complications. Overall, despite both environment and genetics playing a factor as to how the gut of an individual is affected, microbes that exist within the gut are similarly affected by both genetics and conditions that allow them to thrive. Despite their importance when carrying out bodily functions, several microbes have the potential to cause severe health complications. Due to research from pharmaceutical companies such as Leucine Rich Bio, platforms such as BugSpeaks allow for healthy aging and presents a platform for encouraging health education. **Sources** * [Gupta, A., Gupta, R., & Singh, R. L. (2016, October 15). Microbes and environment. Principles and Applications of Environmental Biotechnology for a Sustainable Future.](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7189961/#:~:text=However%2C%20many%20other%20factors%20also,%2Dassociated%20factors%20\(AOFs\).) * [Kelsen, J. R., & Wu, G. D. (2012). The gut microbiota, environment and diseases of modern society. Gut Microbes, 3(4), 374–382.](https://doi.org/10.4161/gmic.21333) * [Kusnierczyk, P. (2023b). Genetic differences between smokers and never-smokers with lung cancer. Frontiers in Immunology, 14.](https://doi.org/10.3389/fimmu.2023.1063716) * Leucine Rich Bio is South Asia’s first microbiome company. [Leucine Rich Bio](https://www.leucinerichbio.com/). (n.d.). date accessed: 09/12/2023 --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Gut Microbiota Analysis for Diagnosing and Treating Diseases Author: BugSpeaks Published: 2023-07-18 Category: Microbiome and Disease Meta Title: Gut Microbiota Analysis for Diagnosing and Treating Diseases Meta Description:
The human gut is home to trillions of microorganisms, collectively known as the gut microbiota. Emerging research suggests that URL: https://www.bugspeaks.com/blog/gut-microbiota-analysis-for-diagnosing-and-treating-diseases The human gut is home to trillions of microorganisms, collectively known as the gut microbiota. Emerging research suggests that these microbes play a crucial role in maintaining overall health and can significantly impact various aspects of our well-being. In recent years, the analysis of gut microbiota has gained significant attention as a powerful tool for diagnosing and treating diseases. Here we will explore the importance of gut microbiota analysis and how it can revolutionize healthcare.  **Understanding Gut Microbiota** The gut microbiota refers to the diverse community of microorganisms residing in the gastrointestinal tract. This ecosystem primarily consists of bacteria, but also includes viruses, fungi, and other microorganisms. These microbes have a symbiotic relationship with the human body, influencing digestion, immune function, metabolism, and even brain health. **Link Between Gut Microbiota and Diseases** Research has uncovered a strong association between imbalances or disruptions in gut microbiota and various diseases. Conditions such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), obesity, diabetes, cardiovascular diseases, and even mental health disorders have been linked to alterations in the gut microbiota. These can we solved with the help of [gut microbiome test](https://www.bugspeaks.com/) **Gut Microbiota Analysis Techniques** Advancements in technology and research methodologies have enabled scientists to analyze the gut microbiota in detail. Several techniques are employed for gut microbiota analysis, including: **Shotgun metagenomic Sequencing:** This technique involves the extraction of DNA from the gut microbiota samples to identify and quantify the genetic material of the microorganisms. It provides a comprehensive view of the microbial composition and functional potential. **16S rRNA Sequencing:** This technique targets a specific region of the bacterial DNA called 16S ribosomal RNA. It provides information about the bacterial diversity and community structure. **Metabolomics:** Metabolomics focuses on analyzing the small molecules produced by the gut microbiota. It helps in understanding the metabolic activities and interactions between the host and microbiota. **Role of Gut Microbiota Analysis in Disease Diagnosis** Gut microbiota analysis has the potential to revolutionize disease diagnosis by serving as a non-invasive and highly informative tool. The microbial composition and functional capabilities obtained through analysis can provide valuable insights into disease mechanisms and help identify biomarkers for early detection. For instance, studies have found distinct microbial signatures in patients with IBD compared to healthy individuals. By analyzing the gut microbiota, healthcare professionals can potentially predict disease progression and develop personalized treatment plans. **Gut Microbiota Analysis for Disease Treatment** The ability to modulate the gut microbiota opens up new avenues for disease treatment and management. Targeted interventions, such as probiotics, prebiotics, and postbiotics, can be used to restore a healthy gut microbiota composition. Probiotics are live microorganisms that, when administered in adequate amounts, confer health benefits to the host. Prebiotics, on the other hand, are non-digestible fibers that selectively stimulate the growth of beneficial bacteria in the gut. Postbiotics are the metabolites produced by the gut microbiota that exhibit various bioactive properties. **Conclusion** Gut microbiota analysis has emerged as a powerful tool for diagnosing and treating diseases. By understanding the complex interplay between the gut microbiota and human health, healthcare professionals can potentially identify early disease markers, predict disease progression, and develop targeted treatment strategies. As the field of gut microbiota analysis continues to advance, it holds immense promise for personalized medicine and improving patient outcomes. By leveraging the knowledge gained from gut microbiota analysis, companies like Bugspeaks are at the forefront of revolutionizing healthcare and transforming the way we approach disease diagnosis and treatment. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Leaky Gut Syndrome: Causes, Symptoms, and Treatment Options Author: BugSpeaks Published: 2023-07-18 Category: Microbiome and Disease Meta Title: Leaky Gut Syndrome: Causes, Symptoms, and Treatment Options Meta Description:
In recent years, there has been growing recognition of the importance of the gut microbiome in maintaining overall h URL: https://www.bugspeaks.com/blog/leaky-gut-syndrome:-causes,-symptoms,-and-treatment-options In recent years, there has been growing recognition of the importance of the [gut microbiome](https://www.bugspeaks.com/) in maintaining overall health and well-being. One condition that has gained significant attention is leaky gut syndrome. Also known as increased intestinal permeability, leaky gut syndrome refers to a condition where the lining of the intestinal wall becomes compromised, allowing substances to leak into the bloodstream that would normally be filtered out. In this blog post, we will delve into the causes, symptoms, and treatment options for leaky gut syndrome, shedding light on this complex condition.  **Causes of Leaky Gut Syndrome:** Leaky gut syndrome can be caused by a variety of factors, including: **Poor Diet:** Consuming a diet high in processed foods, sugars, and unhealthy fats can contribute to inflammation and damage to the intestinal lining. **Chronic Stress:** Prolonged stress can disrupt the balance of the gut microbiome, leading to increased intestinal permeability. **Environmental Factors:** Exposure to environmental toxins, such as pesticides, heavy metals, and pollutants, can damage the intestinal lining and contribute to leaky gut. **Medications:** Certain medications, such as non-steroidal anti-inflammatory drugs (NSAIDs), antibiotics, and corticosteroids, can disrupt the gut microbiome and contribute to intestinal permeability. **Imbalance in Gut Microbiome:** An imbalance in the gut microbiome, with an overgrowth of harmful bacteria and a depletion of beneficial bacteria, can contribute to the development of leaky gut syndrome. **Symptoms of Leaky Gut Syndrome:** The symptoms of leaky gut syndrome can vary from person to person, and they may manifest in different ways. Some common symptoms include: **Digestive Issues:** This can include bloating, gas, diarrhea, constipation, and abdominal pain. **Food Sensitivities:** Leaky gut syndrome can lead to an increased immune response to certain foods, causing food sensitivities or allergies. **Fatigue and Low Energy:** As the body becomes less efficient at absorbing nutrients, fatigue and low energy levels may arise. **Skin Problems:** Leaky gut syndrome has been linked to skin conditions such as eczema, acne, and psoriasis. **Joint Pain and Inflammation:** Increased intestinal permeability can trigger an immune response, leading to joint pain and inflammation. **Treatment Options for Leaky Gut Syndrome:** **Dietary Modifications:** Adopting an anti-inflammatory diet rich in whole foods, such as fruits, vegetables, lean proteins, and healthy fats, can help reduce gut inflammation and support the healing process. **Probiotics and Prebiotics:** Consuming probiotic-rich foods or taking probiotic supplements can help restore a healthy balance of beneficial bacteria in the gut. Prebiotic foods, such as onions, garlic, and bananas, can also nourish the beneficial bacteria. **Gut-Healing Supplements:** Certain supplements, such as L-glutamine, zinc, and omega-3 fatty acids, can aid in repairing the intestinal lining and reducing inflammation. **Stress Management:** Incorporating stress management techniques, such as meditation, yoga, and regular exercise, can help reduce stress and support gut health. **Avoiding Triggers:** Identifying and avoiding potential triggers, such as processed foods, alcohol, and certain medications, can help prevent further damage to the gut lining. **Conclusion:** Leaky gut syndrome is a complex condition that can have a significant impact on overall health and well-being. Understanding the causes, symptoms, and treatment options is crucial for those seeking to address this condition. By adopting a holistic approach that includes dietary modifications, gut-healing supplements, stress management, and the restoration of a healthy gut microbiome, individuals can take important steps towards repairing the intestinal lining and improving their overall health. Remember, it is always advisable to consult with a healthcare professional before making any significant changes to your diet or treatment plan. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## How to Improve Your Gut Health Based on Your Microbiome Analysis Results Author: BugSpeaks Published: 2023-07-18 Category: Microbiome Meta Title: How to Improve Your Gut Health Based on Your Microbiome Analysis Results Meta Description:
Your gut health is essential for your overall well-being. The gut microbiome, which is the community of bacteria that live in your gut, plays a vit URL: https://www.bugspeaks.com/blog/how-to-improve-your-gut-health-based-on-your-microbiome-analysis-results Your gut health is essential for your overall well-being. The gut microbiome, which is the community of bacteria that live in your gut, plays a vital role in digestion, immunity, and even mental health. If you're interested in improving your gut health, you may be considering getting a [gut microbiome test](https://www.bugspeaks.com/). This test can give you insights into the diversity and balance of your gut bacteria, which can help you identify areas where you can make improvements. We'll discuss how to interpret your microbiome analysis results and how to use them to improve your gut health. We'll also introduce Bugspeaks, a gut microbiome test that provides [personalized nutritional recommendations](https://www.bugspeaks.com/) based on your analysis results.  **Understanding the Gut Microbiome** Understanding the importance of gut health and its impact on overall well-being has gained significant attention in recent years. The human gut harbors a diverse community of microorganisms known as the gut microbiome. Advancements in technology have allowed us to analyze and interpret the composition of our microbiome, providing valuable insights into our health. In this blog post, we will explore how you can improve your gut health based on the results of your microbiome analysis. By leveraging the power of personalized data, you can make informed choices to optimize your gut health and enhance your overall quality of life. The gut microbiome is a complex ecosystem that contains trillions of bacteria. These bacteria play a variety of roles in your health, including: 1\. Digesting food and absorbing nutrients 2\. Regulating the immune system 3\. Producing vitamins 4\. Protecting against harmful bacteria 5\. Maintaining a healthy gut lining The composition of your gut microbiome is influenced by a variety of factors, including your diet, lifestyle, and genetics. A healthy gut microbiome is diverse and balanced, with a variety of different types of bacteria. **Introducing Gut Microbiome Analysis** Microbiome analysis involves sequencing and analyzing the DNA of microorganisms present in your gut. The results of this analysis provide a detailed breakdown of the types and relative abundances of different microbial species residing in your gut. These results can help identify imbalances or dysbiosis in your gut microbiome, which can be associated with various health issues such as digestive disorders, immune dysfunction, and mental health conditions. By understanding your microbiome analysis results, you can gain insights into the specific microbial populations that may require attention. Look for key indicators such as decreased microbial diversity, overgrowth of certain species, or the presence of potentially harmful bacteria. This information forms the foundation for personalized interventions to improve your gut health. Gut microbiome analysis is a test that can assess the diversity and balance of your gut bacteria. This test is typically done by collecting a stool sample and sending it to a laboratory for analysis. The results of a microbiome analysis can provide you with insights into your gut health, including: 1\. The overall diversity of your gut bacteria 2\. The relative abundance of different types of bacteria 3\. The presence of any harmful bacteria **Importance of Interpreting Microbiome Analysis Results** The results of a microbiome analysis can be complex, but it's important to understand them so that you can make informed decisions about your gut health. Bugspeaks can help you interpret your results and develop a personalized nutritional plan for improving your gut health. **Tailoring Your Diet for Optimal Gut Health** One of the most impactful ways to improve your gut health is through dietary modifications. Your microbiome analysis results can guide you in making informed decisions about your food choices. Incorporate a variety of whole foods rich in fiber, such as fruits, vegetables, legumes, and whole grains. Fiber acts as a prebiotic, providing nourishment for beneficial bacteria in your gut. Aim for a diverse range of plant-based foods to promote gut microbial diversity. Additionally, consider incorporating fermented foods into your diet. These include yogurt, sauerkraut, kefir, and kimchi. Fermented foods contain probiotics, live microorganisms that confer health benefits when consumed. Probiotics can help restore and maintain a healthy balance of bacteria in your gut. **Bugspeaks Recommendations for Gut Health Enhancement** Bugspeaks is a gut microbiome test that provides personalized recommendations based on your analysis results. The report provides nutritional recommendations and meal plans based on the choice of the customer. The plan may include dietary changes, lifestyle modifications, or the use of supplements. **Conclusion** Improving your gut health can have a positive impact on your overall well-being. By understanding your microbiome analysis results and making changes to your diet and lifestyle, you can improve the diversity and balance of your gut bacteria, which can lead to improved digestion, immunity, and mental health. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Gut Microbiome in Cancer: Exploring the Latest Developments Author: BugSpeaks Published: 2023-06-27 Category: Microbiome and Disease Meta Title: Gut Microbiome in Cancer: Exploring the Latest Developments Meta Description:
The gut microbiome has emerged as a crucial player in various aspects of human health, including cancer development and progressio URL: https://www.bugspeaks.com/blog/gut-microbiome-in-cancer:-exploring-the-latest-developments The gut microbiome has emerged as a crucial player in various aspects of human health, including cancer development and progression. The trillions of microorganisms inhabiting the human gut play an essential role in _modulating the immune system and metabolism_. Recent research has uncovered fascinating insights into the _complex interplay between the gut microbiome and cancer_, thereby opening new avenues for targeted therapies and preventative measures. This article delves into the latest findings in _gut microbiome research_, highlighting key _areas of interest_ and _potential clinical applications_.  **1\. The Role of Gut Microbiome in Cancer Development** **1.1 Microbial Dysbiosis and Inflammation:** _Chronic inflammation_ is a well-known risk factor for various cancers, including _colorectal cancer._ Emerging evidence suggests that an imbalance in the gut microbiota composition, known as _dysbiosis_, may _contribute to chronic inflammation and cancer development._ Dysbiotic gut microbiota can stimulate _pro-inflammatory immune responses,_ leading to _DNA damage_ and increased susceptibility to cancer. Moreover, certain pathogens such as _Helicobacter pylori_ and _Fusobacterium nucleatum_ have been implicated in promoting inflammation and tumour growth. **1.2. Genotoxic Bacterial Metabolites:** Some gut bacteria produce _genotoxic metabolites_ that can directly _damage host DNA_, increasing the risk of cancer. For example, _Escherichia coli_ strains carrying the _pks pathogenicity island_ produce a genotoxin called _colibactin_, which induces _DNA double-strand breaks in host cells_. Similarly, the _Cytolethal Distending Toxin (CDT)_ produced by certain _Proteobacteria_ can cause _genomic instability_ and _promote tumorigenesis_. **1.3. Altered Metabolism and Cancer Risk:** The gut microbiome plays a significant role in modulating host metabolism, and _alterations in microbial metabolic pathways_ have been linked to cancer risk. For instance, _gut bacteria can convert dietary components_ such as red meat and dietary fibres _into potentially carcinogenic_ or tumor-suppressive metabolites, respectively. High consumption of red meat has been associated with an increased risk of colorectal cancer, partly due to the production of _harmful secondary bile acids_ and _genotoxic N-nitroso_ compounds by gut bacteria. **2\. Gut Microbiome and Cancer Therapy** **2.1. Impact on Immunotherapy:** Recent studies have shown that the gut microbiome can influence the effectiveness of cancer immunotherapy, particularly _immune checkpoint inhibitors_. Specific bacterial species, such as _Bacteroides fragilis_ and _Enterococcus hirae_, have been found to enhance _the antitumor efficacy of immune checkpoint inhibitors_ by modulating immune cell activity and promoting a favourable immune response against tumors. Thus, manipulating the gut microbiota may offer a novel strategy to improve the effectiveness of cancer immunotherapy. **2.2. Influence on Chemotherapy:** Gut microbiota can also _impact the host response to conventional chemotherapeutic drugs_. For example, _the anticancer effects of the platinum chemotherapeutic oxaliplatin_ and _the alkylating agent cyclophosphamide are attenuated in germ-free_ or antibiotic-treated mice, highlighting the importance of gut microbiota in mediating the therapeutic response to these drugs. Furthermore, targeting specific bacterial enzymes, such as _β-glucuronidases_, may help _reduce chemotherapy-induced toxicity_ and improve treatment outcomes. **3\. Targeting the Gut Microbiome for Cancer Prevention and Treatment** **3.1. Probiotics and Prebiotics:** The use of probiotics (beneficial live bacteria) and prebiotics (non-digestible fibers that promote the growth of beneficial bacteria) has gained considerable attention as a potential strategy to modulate the gut microbiome and reduce cancer risk. Some studies have shown that the _consumption of probiotics and prebiotics can enhance the production of short-chain fatty acids, such as butyrate, which exhibit tumor-suppressive effects_. Moreover, specific probiotic strains, such as _Lactobacillus spp_., have been found to _improve immune function_ and _reduce inflammation, potentially reducing the risk of cancer._ **3.2. Fecal Microbiota Transplantation (FMT):** FMT involves _transferring the fecal microbiota_ from a healthy _donor to a patient_ with a _dysbiotic gut microbiome_, aiming to _restore eubiosis_ and improve health outcomes. While FMT has shown promising results _in treating recurrent Clostridium difficile infection_, its potential application in cancer prevention and treatment is still under investigation. Some preclinical studies suggest that _FMT may help reduce inflammation_ and _improve the response to cancer therapies_; however, more research is needed to establish the safety and efficacy of this approach in cancer patients. **3.3. Personalized Microbiome-based Interventions:** Given the _inter-individual variability_ in gut microbiome composition and response to therapies, personalized microbiome-based interventions hold promise for _improving cancer prevention_ and treatment outcomes. _By integrating information_ on an individual's gut microbiota composition, genetic background, and environmental factors, _personalized strategies_, such as targeted dietary modifications, probiotics, or prebiotics, _could be developed to modulate the gut microbiome and reduce cancer risk or enhance treatment response_. **4\. Challenges and Future Directions** Despite the significant progress made in understanding the role of the gut microbiome in cancer, several challenges remain. These include: **4.1. Causality and Mechanistic Insights:** Establishing causality between gut microbiota alterations and cancer development is challenging, as most studies are observational and cannot distinguish between cause and effect. Moreover, _understanding the complex mechanisms_ by which gut microbiota _influence cancer risk_ and therapy response requires further research _using advanced experimental models_ and multi-omics approaches. **4.2. Standardization of Methodologies:** There is a need for standardized methodologies in gut microbiome research, including _sample collection, processing, sequencing, and data analysis_. This will facilitate the comparison of results across studies and enable the identification of consistent patterns and biomarkers associated with cancer risk and treatment response. **4.3. Clinical Translation:** Translating findings from _preclinical studies to clinical practice_ is a major challenge, as many factors can influence the gut microbiome and its interaction with the _host, including diet, lifestyle, genetics, and environmental exposures_. _Large-scale, well-designed clinical trials_ are needed to establish the safety and efficacy of microbiome-based interventions in cancer prevention and treatment. **Conclusion** In conclusion, the gut microbiome plays a pivotal role in cancer development and therapy response. Further research is needed to unravel the complex interplay between gut microbiota, host factors, and environmental exposures, and to develop _effective microbiome-based strategies_ for cancer prevention and treatment. With advances in _sequencing technologies, bioinformatics, and experimental models, the field of gut microbiome research holds immense potential for improving our understanding and management of cancer_. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## How Gut Microbiome Testing Can Improve Digestive Health Author: BugSpeaks Published: 2023-06-26 Category: Process Meta Title: How Gut Microbiome Testing Can Improve Digestive Health Meta Description:
In recent years, there has been a growing interest in the role of gut microbiome in maintaining overall health. The gut microbiome URL: https://www.bugspeaks.com/blog/how-gut-microbiome-testing-can-improve-digestive-health In recent years, there has been a growing interest in the role of gut microbiome in maintaining overall health. The gut microbiome refers to the trillions of microorganisms that reside in our digestive tract, playing a crucial role in various bodily functions. Research suggests that imbalances in the gut microbiome can contribute to digestive health issues such as bloating, constipation, diarrhea, and even more serious conditions like inflammatory bowel disease (IBD). This is where gut microbiome testing comes into the picture. **What is Gut Microbiome Testing?** Gut microbiome testing, also known as microbiome analysis or gut microbiota profiling, is a process that involves analyzing the composition and diversity of microorganisms present in the gut. The tests are usually conducted using a **[stool test sample](https://www.bugspeaks.com/howitworks)**, which contains a rich collection of bacteria, fungi, viruses, and other microorganisms. By examining the genetic material of these microorganisms, scientists can gain valuable insights into the individual's gut health. **The Importance of Gut Microbiome Testing** **1\. Identifying Imbalances:** Gut microbiome testing can help identify imbalances in the microbial composition of the gut. This information is crucial in understanding the root cause of digestive issues and developing targeted treatment plans. By identifying specific bacteria or pathogens that are overgrown or lacking, healthcare providers can recommend personalized dietary changes, probiotic supplementation, or other interventions to restore balance. **2\. Personalized Approach:** Each individual's gut microbiome is unique, influenced by factors such as genetics, diet, lifestyle, and environment. **[Gut microbiome testing](https://www.bugspeaks.com/product)** allows for a personalized approach to improving digestive health. Instead of adopting a one-size-fits-all approach, healthcare providers can tailor interventions based on an individual's specific microbiome composition, leading to more effective outcomes. **3\. Preventive Care:** Gut microbiome testing not only helps in identifying existing imbalances but also plays a significant role in preventive care. By understanding an individual's gut by gut health test,-potential issues can be detected early on, allowing for proactive measures to maintain a healthy microbiome. This can help prevent the development of chronic digestive conditions and promote overall well-being. **The Gut Health and Digestive Health** The gut microbiome health plays a vital role in digestive health through various mechanisms: **1\. Nutrient Absorption:** The microorganisms in the gut assist in breaking down and absorbing nutrients from the food we consume. They help break down complex carbohydrates, fiber, and other substances that our body cannot digest on its own. Imbalances in the gut microbiome can hinder proper nutrient absorption, leading to deficiencies and digestive discomfort. **2\. Immune Function:** A significant portion of our immune system is located in the gut. The gut microbiome helps regulate immune responses, protecting against harmful pathogens while maintaining tolerance to beneficial bacteria. Imbalances in the gut microbiome can disrupt this delicate balance, leading to inflammation and increased susceptibility to infections. **3\. Intestinal Barrier Integrity:** The gut lining acts as a barrier, preventing harmful substances from entering the bloodstream. The gut microbiome helps maintain the integrity of this barrier by promoting the production of mucus and tightening the junctions between intestinal cells. Imbalances in the gut microbiome can compromise the gut barrier, leading to a condition known as "leaky gut" and allowing toxins and bacteria to enter the bloodstream, triggering inflammation and digestive issues. **Taking Steps Towards a Healthy Gut Microbiome** Improving your gut microbiome starts with adopting healthy lifestyle habits: **1\. Dietary Modifications:** Include a variety of fiber-rich fruits, vegetables, whole grains, and fermented foods in your diet. These provide nourishment to beneficial gut bacteria. Limit the consumption of processed foods, sugary snacks, and artificial additives, as they can negatively impact the gut microbiome. **2\. Probiotic Supplementation:** Probiotics are beneficial bacteria that can support a healthy gut microbiome. Consider taking a high-quality probiotic supplement to introduce and maintain a diverse range of beneficial bacteria in your gut. **3\. Stress Management:** Chronic stress can disrupt the gut-brain axis, affecting the gut microbiome. Engage in stress-reducing activities such as meditation, exercise, and adequate sleep to promote a healthy gut. **4\. Avoid Overuse of Antibiotics:** While antibiotics are necessary for treating bacterial infections, overuse can disrupt the gut microbiome. Only take antibiotics when prescribed by a healthcare professional and consider probiotic supplementation during and after the course to support microbial balance. **Conclusion** Gut microbiome testing offers valuable insights into an individual's gut health test and serves as a powerful tool in improving digestive health. By identifying imbalances and personalizing interventions, healthcare providers can help individuals restore balance in their gut microbiome, alleviate digestive issues, and promote overall well-being. Adopting a holistic approach that includes dietary modifications, probiotic supplementation, stress management, and responsible antibiotic use can further support a healthy gut microbiome. Remember, maintaining a balanced gut microbiome is key to optimal digestive health and overall vitality. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Gut Microbiome in Maternal Health: An In-depth Exploration Author: BugSpeaks Published: 2023-06-13 Category: Microbiome and Lifestyle Meta Title: Gut Microbiome in Maternal Health: An In-depth Exploration Meta Description:
The gut microbiome, a complex ecosystem of microorganisms residing in the human digestive tract, plays a crucial role in URL: https://www.bugspeaks.com/blog/gut-microbiome-in-maternal-health:-an-in-depth-exploration The gut microbiome, a complex ecosystem of microorganisms residing in the human digestive tract, plays a crucial role in maintaining overall health. Recent research has highlighted the significance of the gut microbiome in various physiological processes, including maternal health. This aims to provide an in-depth exploration of the relationship between the gut microbiome and maternal health, focusing on its influence during pregnancy, labour, and the postpartum period. Understanding the dynamic interplay between the gut microbiome and maternal health holds promising implications for optimizing pregnancy outcomes and promoting maternal well-being.  **1\. Gut Microbiome Composition During Pregnancy:** **1.1 - Hormonal Influences:** a. Oestrogen and progesterone levels during pregnancy affect the gut microbiome composition. b. Hormonal fluctuations create an environment that supports specific bacterial growth. **1.2 - Dietary Modifications:** a. Pregnancy induces changes in dietary habits, impacting the gut microbiome. b. Increased intake of certain nutrients and fibre affects bacterial diversity. **1.3 - Immune System Adaptations:** a. Maternal immune system undergoes adaptations to support foetal development. b. These changes influence the gut microbiome composition during pregnancy. **1.4 - Increased Microbial Diversity:** a. Studies suggest that microbial diversity increases during pregnancy. b. Higher diversity may be beneficial for maternal and foetal health. **2\. Impact of Gut Microbiome on Pregnancy Outcome:** **2.1 - Gestational Weight Gain:** a. Dysbiosis, an imbalance in gut microbial composition, may contribute to excessive weight gain. b. Dysbiosis leads to inflammation and insulin resistance, affecting metabolic health. **2.2 - Gestational Diabetes:** a. Imbalances in gut microbial composition are associated with an increased risk of gestational diabetes. b. Dysbiosis contributes to inflammation and insulin resistance, impairing glucose metabolism. **2.3 - Preterm Birth:** a. Certain gut bacteria produce metabolites that influence uterine contractions and cervical ripening. b. Dysbiosis may contribute to an increased risk of preterm birth. **3\. Gut Microbiome and Maternal Immune System:** **3.1 - Immune Tolerance:** a. Gut bacteria interact with immune cells, contributing to immune tolerance. b. Immune tolerance is crucial during pregnancy to prevent adverse immune responses against the foetus. **3.2 - Preeclampsia:** a. Dysbiosis has been linked to an increased risk of preeclampsia. b. Dysbiosis leads to immune dysregulation and inflammation, contributing to preeclampsia. **3.3 - Maternal-Foetal Immune Activation:** a. Altered gut microbiome is associated with maternal-fetal immune activation. b. Maternal immune activation can increase the risk of neurodevelopmental disorders in the offspring. **4\. Gut Microbiome in Postpartum Health:** **4.1 - Hormonal Fluctuations and Mood:** a. Hormonal changes in the postpartum period influence the gut microbiome. b. Gut microbiome affects the production of neurotransmitters, impacting maternal mood. **4.2 - Lactation:** a. Gut microbiome influences breast milk composition. b. Beneficial bacteria in the gut contribute to the synthesis of important nutrients in breast milk. **4.3 - Mode of Delivery:** a. Vaginal delivery and caesarean section have differing impacts on the establishment of the infant's gut microbiome. b. Mode of delivery influences the transmission of maternal microbiota to the newborn. **4.4 - Postpartum Recovery:** a. A healthy gut microbiome is essential for postpartum recovery. b. Promoting a healthy gut microbiome through dietary interventions and probiotics can aid in recovery. **Conclusion** The gut microbiome exerts a profound influence on maternal health throughout pregnancy, labor, and the postpartum period. Understanding the intricate relationship between the gut microbiome and maternal health holds promising potential for optimizing pregnancy outcomes, preventing complications, and promoting overall well-being. Future research should focus on elucidating the specific mechanisms underlying these associations and exploring targeted interventions, such as probiotic supplementation and personalized dietary recommendations, to modulate the gut microbiome and improve maternal health. By harnessing the power of the gut microbiome, we can enhance maternal care and ensure better maternal and foetal outcomes. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## The Gut Feeling: Unlocking the Secrets to Optimal Health Author: BugSpeaks Published: 2023-05-23 Category: Microbiome Meta Title: The Gut Feeling: Unlocking the Secrets to Optimal Health Meta Description:
In recent years, gut health has emerged as a hot topic in the world of wellness, and for good reason. Our gut, often ref URL: https://www.bugspeaks.com/blog/the-gut-feeling:-unlocking-the-secrets-to-optimal-health In recent years, gut health has emerged as a hot topic in the world of wellness, and for good reason. Our gut, often referred to as our "second brain," plays a pivotal role in our health and well-being. From digestion and nutrient absorption to immune function and mental health, the gut is a complex ecosystem that deserves our attention. In this blog, we'll delve into the importance of gut health and provide practical tips for nurturing a healthy gut. The gut is home to trillions of microorganisms, collectively known as the gut microbiota. These microscopic inhabitants, include bacteria, fungi, and viruses which work in harmony to perform essential functions that impact our physical and mental health. A balanced gut microbiome is crucial for optimal digestion, nutrient absorption, and immune system function. It also influences our mood, cognitive abilities, and even our weight. So, what happens when our gut health is compromised? An imbalance in the gut microbiota, known as dysbiosis, can lead to a host of issues, such as digestive problems, weakened immunity, and chronic inflammation. Research has also linked poor gut health to mental health disorders, including anxiety and depression, as well as chronic conditions like obesity, diabetes, and autoimmune diseases.  Now that we understand the significance of gut health, let's explore some practical ways to nurture a healthy gut: * **Eat a diverse, fibre-rich diet:** A varied diet rich in fibre is essential for maintaining a balanced gut microbiome. Incorporate a wide range of fruits, vegetables, whole grains, legumes, nuts, and seeds into your meals. These foods provide the necessary nutrients and prebiotics that promote the growth of beneficial gut bacteria. * **Include probiotics and fermented foods:** Probiotics are live microorganisms that can help restore balance in the gut microbiome. You can find them in supplements or naturally occurring in fermented foods like yogurt, kefir, sauerkraut, kimchi, and kombucha. Including these foods in your diet can support a healthy gut ecosystem. * **Limit processed foods and added sugars:** Processed foods and added sugars can negatively impact gut health by promoting the growth of harmful bacteria. Aim to reduce your intake of these foods and opt for whole, unprocessed options whenever possible. * **Stay hydrated:** Drinking enough water is essential for maintaining a healthy gut. Adequate hydration helps to promote regular bowel movements and supports the mucosal lining of the intestines, which acts as a barrier against harmful bacteria. * **Manage stress:** Chronic stress can wreak havoc on your gut health, leading to an imbalance in the gut microbiota. Incorporate stress-reducing practices like meditation, yoga, or deep breathing exercises into your daily routine to support a healthy gut. Gut health is not only about the balance of microorganisms in our digestive system; it also involves the integrity of the gut lining and the efficiency of the gut-brain communication. Let's explore these aspects further: 1. **Gut lining integrity:** The gut lining acts as a barrier between the contents of the gut and the rest of the body. It selectively allows nutrients to pass through while preventing harmful substances and pathogens from entering the bloodstream. A compromised gut lining, often referred to as "leaky gut," can lead to increased intestinal permeability, allowing toxins and undigested food particles to enter the bloodstream. This can trigger an immune response, resulting in inflammation and various health issues, such as food sensitivities, skin conditions, and autoimmune disorders. To maintain a healthy gut lining, focus on consuming nutrient-dense, anti-inflammatory foods, and avoid irritants like alcohol, caffeine, and certain medications. 2. **Gut-brain communication:** The gut and brain are in constant communication through the gut-brain axis, a complex network of nerves, hormones, and neurotransmitters. This connection allows the gut to influence our mood, appetite, and stress response. For example, the gut produces around 90% of the body's serotonin, a neurotransmitter that regulates mood, sleep, and appetite. Disruptions in the gut-brain axis have been linked to mental health issues, such as anxiety, depression, and even neurodegenerative diseases like Parkinson's and Alzheimer's. To support healthy gut-brain communication, engage in regular physical activity, practice stress management techniques, and consume a balanced diet rich in omega-3 fatty acids, antioxidants, and other brain-boosting nutrients. In conclusion, gut health is a multifaceted concept that encompasses the balance of microorganisms, the integrity of the gut lining, and healthy gut-brain communication. By nurturing a healthy gut through practical tips and focusing on these key aspects, we can optimize our overall well-being and unlock the secrets to optimal health. Remember, a happy gut serves as the foundation for a happier, healthier life. Trust your gut feeling and make prioritizing your gut health an essential part of your daily routine. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Gut Microbiome And Depression Author: BugSpeaks Published: 2023-01-04 Category: Microbiome and Lifestyle Meta Title: Gut Microbiome And Depression Meta Description:
Depression is a common and complex mental health disorder that affects millions of people around the world. Despite its prevalence URL: https://www.bugspeaks.com/blog/gut-microbiome-and-depression Depression is a common and complex mental health disorder that affects millions of people around the world. Despite its prevalence, the causes of depression are still not fully understood, making it difficult to effectively diagnose and treat. However, recent research has begun to shed light on some of the underlying mechanisms behind the disease, and one promising area of investigation is the gut microbiome. The gut microbiome is the collection of microorganisms living in our digestive tract, and it has been found to play a role in a variety of health conditions, including mental health disorders. A growing body of evidence suggests that there may be a link between the gut microbiome and depression, with some research finding that people with depression have different gut microbiomes than those without the disorder. A new study published in the journal Nature Communications adds to this growing body of evidence by investigating the relationship between the gut microbiome and depression in two large cohort studies. The researchers analyzed the fecal microbiomes of over 2,500 individuals and found that certain microbial taxa (types of bacteria) were more common in people with symptoms of depression. These bacteria are known to be involved in the synthesis of neurotransmitters like glutamate, butyrate, serotonin, and GABA, which play a role in mood regulation. The study suggests that the composition of the gut microbiome may be an important factor in the development of depression and that targeting the microbiome could potentially be a new approach to treating the disorder.  In addition to the previously mentioned microbial taxa, the intestinal bacterial strains Eggerthella, Subdoligranulum, Coprococcus, and Ruminococcaceae have also been reported to be associated with major depression in earlier studies. Eggerthella has been consistently found to be increased in cases of depression and anxiety in 8 different studies, which is in line with the findings of the current study. There is evidence to suggest a causal link between major depressive disorder (MDD) and Eggerthella, but further investigation is needed to confirm this. Similarly, Subdoligranulum and Coprococcus have been consistently found to be depleted in individuals with generalized anxiety disorder and depression in several studies. In a recent study, Coprococcus was depleted in rats that exhibited depressed behavior following a fecal transplant from depressed human subjects, suggesting that Coprococcus may have a causal impact on depression. Both Subdoligranulum and Coprococcus are involved in the production of butyrate, and Subdoligranulum was found to be increased in a diet rich in omega-3 fatty acids. A previous meta-analysis has shown that omega-3 fatty acid supplementation, specifically eicosapentaenoic acid (EPA), is beneficial for depression. At the genus and family levels, Ruminococcaceae have been found to be depleted in cases of both unipolar and bipolar depression. A similar pattern was observed in a study by Bosch et al., with several genera belonging to the Ruminococcaceae family being depleted in those reporting higher levels of depressive symptoms, which is consistent with the results of the current study. While more research is needed to fully understand the gut-brain connection and the potential role of the microbiome in depression, this study adds to the growing body of evidence supporting the importance of gut health in mental well-being. Taking care of our gut health, through a healthy diet and lifestyle, may have important implications for our mental health and well-being. Link to the study: [https://www.nature.com/articles/s41467-022-34502-3](https://www.nature.com/articles/s41467-022-34502-3) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## A gradual shift to agriculture Author: BugSpeaks Published: 2022-12-30 Category: Microbiome Meta Title: A gradual shift to agriculture Meta Description:
A gradual shift to agriculture – An ancient oral microbiome study
The gut microbiome is the community of microorganisms that live in the digestive tract and play a vital role in the overall heal URL: https://www.bugspeaks.com/blog/gut-microbiome-profiling The gut microbiome is the community of microorganisms that live in the digestive tract and play a vital role in the overall health and wellness of an individual. These microorganisms, which include bacteria, fungi, and viruses, are essential for digestion and absorption of nutrients, immune system function, and overall health.  Gut microbiome profiling is a process that involves the identification and analysis of the specific types of microorganisms present in the gut, as well as their relative abundance and functional capabilities. This can be done through various techniques such as DNA sequencing, metagenomic analysis, and functional assays. **  ** There is growing evidence that the gut microbiome plays a role in a wide range of health conditions, including obesity, inflammatory bowel disease, diabetes, and even mental health disorders. An unhealthy gut microbiome, characterized by an imbalance of microorganisms or a lack of diversity, has been linked to a higher risk of these and other conditions. **  ** On the other hand, a healthy gut microbiome, characterized by a diverse and balanced community of microorganisms, has been associated with better overall health and wellness. A healthy gut microbiome can help to support digestion and absorption of nutrients, regulate the immune system, and protect against infections and other diseases. **  ** Gut microbiome profiling can provide valuable insights into the composition and function of the gut microbiome and can be used to identify imbalances or other abnormalities that may be contributing to health issues. This information can be used to develop targeted interventions, such as dietary changes or probiotic supplementation, to restore balance and support overall health and wellness.  While gut microbiome profiling can provide valuable insights into the role of the gut microbiome in health and wellness, it is important to keep in mind that this field is still relatively new and more research is needed to fully understand the relationship between the gut microbiome and various health conditions. The key now is to re-balance the gut microbiome. BugSpeaks aims to do that through nutritional and probiotic interventions. **  ** --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## The Secret to Glowing Skin: A Healthy Gut Author: BugSpeaks Published: 2022-12-09 Category: Microbiome and Lifestyle Meta Title: The Secret to Glowing Skin: A Healthy Gut Meta Description:
Are you tired of trying every skincare product on the market, but still not achieving the glowing, radiant skin you desire? The secret to healthy, URL: https://www.bugspeaks.com/blog/the-secret-to-glowing-skin:-a-healthy-gut Are you tired of trying every skincare product on the market, but still not achieving the glowing, radiant skin you desire? The secret to healthy, glowing skin might be closer than you think: your gut! You might be wondering, what does my gut have to do with my skin? The truth is, our gut health plays a crucial role in the health and appearance of our skin. Here's how: 1. The gut is responsible for digesting and absorbing the nutrients from the food we eat. These nutrients, such as vitamins and minerals, are essential for maintaining healthy skin. For example, vitamin A is necessary for cell growth and repair, while vitamin C helps to protect the skin from damage caused by the sun and other environmental factors. 2. A healthy gut can also help to regulate the immune system, which can protect the skin from damage and inflammation. When our gut is healthy, it can produce antibodies that help to fight off harmful bacteria and other invaders that can cause skin irritation and inflammation. 3. A healthy gut microbiome (the collection of bacteria that live in the gut) is essential for good overall health, including the health of our skin. A balanced microbiome can help to break down and eliminate toxins from the body, which can help to prevent skin problems such as acne and other skin conditions.  So, what can you do to maintain a healthy gut and achieve glowing skin? Here are a few simple tips: * Eat a balanced diet that includes plenty of fruits, vegetables, whole grains, and lean proteins. These foods are rich in the nutrients your skin needs to stay healthy, and they can also help to promote a healthy gut microbiome. * Stay hydrated by drinking plenty of water. Water is essential for good digestion and can also help to flush toxins from the body, which can help to prevent skin problems. * Avoid processed, sugary foods and drinks that can disrupt the balance of bacteria in the gut and lead to inflammation. * Consider taking a daily probiotic supplement, which can help to maintain a healthy balance of bacteria in the gut and support overall gut health. By following these simple tips and focusing on maintaining a healthy gut, you can achieve glowing, radiant skin that will have everyone asking for your skincare secrets! --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Allergies, Intolerances And The Microbes In Our Gut Author: BugSpeaks Published: 2021-07-20 Category: Nutrition Meta Title: Allergies, Intolerances And The Microbes In Our Gut Meta Description:
Candida overgrowth in the gut is caused by an overly tolerant immune system. Chronic fatigue is caused by a suppressed i URL: https://www.bugspeaks.com/blog/allergies,-intolerances-and-the-microbes-in-our-gut Candida overgrowth in the gut is caused by an overly tolerant immune system. Chronic fatigue is caused by a suppressed immune system. Arthritis is caused by a faulty immune system that has lost its ability to generate antibodies to attack tumor cells and other foreign substances. Autoimmunity can result from an overactive, hypersensitive immune system or a missing or poorly functioning adaptive immune response. The immune system, as magnificent as it is, is not without flaws! Allergies are caused by a highly sensitive immune system. Before we get into how gut microorganisms and allergic reactions are linked, it's important to understand what an allergic reaction is. Lymphocytes are white blood cells that form an important part of our immune system. B-lymphocytes and T-lymphocytes are two types of lymphocytes which aid in the defence of our body against foreign invaders such as bacteria, viruses, and other foreign toxins like poisons. When a lymphocyte comes into contact with a particle or cell that has surface marking molecules that identify it as a foreign invader (antigen), it does a tiny version of fingerprinting. B-cell recognizes that antigen and produces antibodies that are particularly built to combat the threat. There are five fundamental forms of antibodies called Immunoglobulins (Igs) - IgA, IgD, IgE, IgG and IgM. IgE is the antibody that is produced in allergic responses. In a healthy immune system, Ig antibodies bind to disease cell surface antigens and destroy them, which is fine. But in case of an allergic reaction, the B-cells, are genetically "misinformed," about a generally harmless protein, produce high amounts of IgE antibodies and attach themselves to mast cells and basophils throughout the body, resulting in a reaction called ‘sensitising exposure.’ The mast cells (connective tissue cells) and basophils (white blood cells) both contain histamine, an important anti-infective weapon in the body's arsenal. Unfortunately, histamine is a potentially dangerous substance when it is released into the body in an incorrect or excessive amount. So, when the Ig antibodies destroy these cells, their reserves of histamine and other allergy mediators are released into the tissues and bloodstream. This causes a variety of allergic symptoms. So, when an allergy-sufferer ingests a particular allergen (the antigen) for the first time, the immune system tries to fight what it perceives as an invader. Swelling occurs and if left untreated, it may lead to a severe allergic reaction. It is critical to understand that allergies are not the same as intolerances. As we have seen, allergies are immunological reactions of the body. Whereas, intolerances are non-immunological, non-allergic hypersensitivities. It's a physical reaction to some foods that aren't adequately digested or cause our digestive system to malfunction in some way. It happens when the digestive system lacks the enzymes or chemicals needed to break down and digest the food. As a result, if a person's body lacks the enzymes required to break down a particular food, he or she may develop food ‘intolerance’ after eating that item and not food ‘allergy’.  Both food allergies and intolerances have a substantial impact on the quality of life. They negatively impact physical health, reduce the enjoyment of eating, and cause a condition of hypervigilance for probable allergens in the food. Food allergies and intolerances have become much more common in the last ten years. There have been several attempts to explain why, but only recent studies have been able to pinpoint the critical factor, i.e our microbiota. Our microbes help to grow the gut immune system and generate immunological tolerance. In fact, it wouldn't be wrong to say that the immune system is trained by our gut microbiome! Immunological tolerance refers to the host's ability to differentiate harmless ingested or inhaled antigens from those that trigger an immune response. The less the inflammation, the more tolerant the immune system. The root cause of numerous diseases is heavily influenced by the breakdown of this tolerance. The adaptive immune system is important for distinguishing between self and foreign antigens, but the intestinal microbiota poses a challenge as it contains a large amount of foreign antigens like dietary substances and commensal bacteria which must be ignored or tolerated by the immune cells, in order to maintain health. Our microbes have evolved methods to decrease undesired inflammation. There is growing evidence that the mechanisms that regulate commensal bacteria tolerance are also important for maintaining immunological tolerance, which can prevent allergy and autoimmune disorders. A rise in immunologically induced illnesses could be explained by a delay in immune function development during childhood, leading to a loss of immune tolerance. The interaction with the commensal microbiota of the gut helps promote the development and maturation of T-cells (like Th1,Th2, Treg). Low or no early exposure to foreign antigens, according to the "hygiene hypothesis," may raise the risk of allergy disorders. The “old friends hypothesis” (a revised version of the “hygiene hypothesis”) claims that the presence of commensal bacteria is critical for stimulating Treg cells maturation, which is another type of T-cell called regulatory T (Treg) cell, having immunomodulatory or immunosuppressive capabilities. They are crucial for the maintenance of immune tolerance and play a key role in the control of inflammatory responses thereby enabling better processing of self or foreign antigens, and the induction of immunological tolerance. Furthermore, allergic and non-allergic people have different gut flora, implying that changes in the normal human gut flora have a role in the development of allergies and intolerances. Bacteroides, Enterobacteria, Bifidobacteria, and Lactobacilli are the commensal bacteria that work with the gut's immune system to promote immunological tolerance. Lifestyle factors that reduce the amount of beneficial gut microorganisms can compromise this tolerance, leading to allergy sensitization and food intolerances. For instance, egg and milk allergies are linked to a decrease in Lactobacilli and an increase in Staphylococcus aureus in children. During the first two months of life, children with lower levels of Lactobacillus rhamnosus, Lactobacillus casei, Lactobacillus paracasei, and Bifidobacterium adolescentis were found to have a higher chance of developing allergy sensitization to cow's milk, egg white, and inhalant allergens. Food allergen sensitivity in children is also linked to lower levels of Bacteroides, Proteobacteria, and Actinobacteria. A lack of microbial variety also may predispose to the non-immunologic food intolerances such as gluten, FODMAP and histamine intolerance.  _Factors responsible for changes in the normal gut flora, leading to dysbiosis which in turn increases the risk of acquiring allergic diseases :- A. Genetics ; B. Less breastfeeding ; C. Transition from rural to urban and suburban living (less contact with nature) ; D. Smaller families ; E. High fat, low fibre diet ; F. Antibiotics ; G. More cesarean sections ; H. Over-sanitization_ Therefore, it is clear that our microbes are vital for immunological tolerance and changes in the composition of the microbiota can lead to allergies and intolerances. We should be cautious about things that can greatly influence our gut flora such as antibiotic use, chemical exposure, nutrition and lifestyle. After all, prevention is better than cure. But sometimes there is little we can do about the way a baby is born, how they are nursed, or how much medication they are given. Synbiotics, which are a combination of pre and probiotics, have been shown to protect the microbiomes of newborns who are prone to cow's milk allergy. Hence, supplementing with probiotics, prebiotics and synbiotics can help repair the gut microbiota and increase the immune system's resistance to allergies and intolerances, among many other health benefits. You can discover our range of supplements here at [Rychbiome](https://rychbiome.com/)! **References:** 1. _McLoughlin, Rachel M.; Mills, Kingston H.G. (2011). “Influence of gastrointestinal commensal bacteria on the immune responses that mediate allergy and asthma.” Journal of Allergy and Clinical Immunology, 127(5), 1097–1107. doi:10.1016/j.jaci.2011.02.012_ 2. _Wu, Hsin-Jung, and Eric Wu. “The role of gut microbiota in immune homeostasis and autoimmunity.” Gut microbes vol. 3,1 (2012): 4-14. doi:10.4161/gmic.19320_ 3. _Yoo, Ji Youn et al. “Gut Microbiota and Immune System Interactions.” Microorganisms vol. 8,10 1587. 15 Oct. 2020, doi:10.3390/microorganisms8101587_ 4. _Kau, Andrew L et al. \`\`Human nutrition, the gut microbiome and the immune system.” Nature vol. 474,7351 327-36. 15 Jun. 2011, doi:10.1038/nature10213_ 5. _Liu, Andrew H. “Revisiting the hygiene hypothesis for allergy and asthma.” The Journal of allergy and clinical immunology vol. 136,4 (2015): 860-5. doi:10.1016/j.jaci.2015.08.012_ 6. _Castellazzi, Anna Maria et al. “Probiotics and food allergy.” Italian journal of pediatrics vol. 39 47. 29 Jul. 2013, doi:10.1186/1824-7288-39-47_ 7. _Björkstén, B et al. “The intestinal microflora in allergic Estonian and Swedish 2-year-old children.” Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology vol. 29,3 (1999): 342-6. doi:10.1046/j.1365-2222.1999.00560.x_ 8. _Sjögren, Y.M., Jenmalm, M.C., Böttcher, M.F., Björkstén, B. and Sverremark-Ekström, E. (2009), “Altered early infant gut microbiota in children developing allergy up to 5 years of age.” Clinical & Experimental Allergy, 39: 518-526. [https://doi.org/10.1111/j.1365-2222.2008.03156.x](https://doi.org/10.1111/j.1365-2222.2008.03156.x)_ 9. _Ling, Zongxin et al. “Altered fecal microbiota composition associated with food allergy in infants.” Applied and environmental microbiology vol. 80,8 (2014): 2546-54. doi:10.1128/AEM.00003-14_ 10. _Caminero, Alberto et al. “Diversity of the cultivable human gut microbiome involved in gluten metabolism: isolation of microorganisms with potential interest for coeliac disease.” FEMS microbiology ecology vol. 88,2 (2014): 309-19. doi:10.1111/1574-6941.12295_ 11. _Smolinska, S et al. “Histamine and gut mucosal immune regulation.” Allergy vol. 69,3 (2014): 273-81. doi:10.1111/all.12330_ 12. _Pascal M, Pérez-Gordo M, Caballero T et al. “[Microbiome and Allergic Diseases](https://www.frontiersin.org/article/10.3389/fimmu.2018.01584).” Frontiers in Immunology. 2018; DOI=10.3389/fimmu.2018.01584_ --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Super Sanitization In The Corona Era - Stop And Ponder Author: BugSpeaks Published: 2021-06-28 Category: Microbiome and Lifestyle Meta Title: Super Sanitization In The Corona Era - Stop And Ponder Meta Description:
Sanitization, undoubtedly, is a proactive and preventative approach to contain the spread of SARS-CoV2. Because there is no surefi URL: https://www.bugspeaks.com/blog/super-sanitization-in-the-corona-era---stop-and-ponder Sanitization, undoubtedly, is a proactive and preventative approach to contain the spread of SARS-CoV2. Because there is no surefire way to completely combat the COVID-19 outbreak, sanitization is crucial. While there is nothing wrong with using sanitizing chemicals in general, too many of us may have gone too far with the advice. Homemakers, understandably, want to make sure that their homes are clean and free of microorganisms now more than ever. However, it appears that the solution may be more dangerous than the problem. Triclosan and triclocarban are two antimicrobial compounds that are often found in many household cleaning products and disinfectants. These two compounds have been linked to skin irritation, allergy susceptibility, bacterial and endocrine disruption at high enough concentrations. They are resistant to treatment, which is concerning because triclosan can transform into a poison called dioxin when exposed to sunlight and water. Dioxin can cause skin sores and even affect liver function at high amounts. Long-term dioxin exposure has the potential to harm the immune, neurological, endocrine, and reproductive systems of the body. Hand sanitizers with an ethanol or alcohol base, on the other hand, are an efficient way to reduce the number of microbes on our hands. Alcohol and ethanol destroy all microbes, good or bad, by denaturing or breaking down their proteins. It is known that a balanced microbiome is involved in a variety of critical functions, including immune system training and digestion, biosynthesis of vitamins K and B in the gut, and much more. When children’s bodies are not being exposed to as many different types of microorganisms, their immune systems become weaker and weaker, resulting in more illnesses later in life, leading to allergies, asthma, autoimmune disorders, obesity, type 2 diabetes, and other chronic diseases. All of these conditions are correlated to our microbiomes. The most well-supported explanation for understanding this trend is the “hygiene hypothesis”. The hypothesis is founded on the idea that early life exposure to microbes, including viruses, is advantageous to the training and development of the human immune system. Highly hygienic circumstances in westernised countries impedes our natural immunity.  The microbiomes of the very young and the elderly are the most labile, whereas older children and adults' microbiomes are resilient, and any harm caused by the pandemic can be readily restored. Microbiome variety can be improved by eating a high-fiber diet, spending time outdoors, interacting with animals, and, of course, returning to regular life when it is safe to do so, mixing freely with others and hugging and kissing dear ones. The fact that over-70s were the first to get vaccinated, allowed them to re-establish normal microbiomes through earlier exposure to visitors and open places. It's not that using sanitizing items is inherently wrong, but balance is crucial here. It is not necessary to sanitize every inch of the house in order to stay away from corona. Hand sanitizers were designed to reduce the risk of cross-contamination in hospitals when patients' immune systems are weak or non-existent. Instead of relying completely on hand sanitizers, follow the guidelines for good hygiene, such as sneezing into elbows rather than the hand, wearing masks properly, following proper hand washing steps and staying at home while sick. When we are all vaccinated and the pandemic is gone, it will be critical to reestablish normal social interactions, while taking extra measures during flu season. Given the growing awareness of the microbiome's impact on human health, it's important to learn how the microbiome influences susceptibility to the coronavirus, as well as how COVID-19 disease and prevention methods may alter the microbiome. This understanding is critical in prevention and treatment of the pandemic, as well as long-term biological and social consequences. **References:** 1. Vatanen T, Kostic A, d’Hennezel E, et al. [Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans](http://www.cell.com/cell/fulltext/S0092-8674\(16\)30398-1). Cell. Online April 28, 2016. DOI: 10.1016/j.cell.2016.04.007 2. B. Brett Finlay et al. The hygiene hypothesis, the COVID pandemic, and consequences for the human microbiome. Proceedings of the National Academy of Sciences Feb 2021, 118 (6) e2010217118; DOI: 10.1073/pnas.2010217118 3. Rook GA. Hygiene and other early childhood influences on the subsequent function of the immune system. Dig Dis. 2011;29(2):144-53. Epub 2011 Jul 5. PMID: 21734378. doi: 10.1159/000323877. 4. Chatterjee Bithika et al. The mortality due to COVID-19 in different nations is associated with the demographic character of nations and the prevalence of autoimmunity. medRxiv 2020.07.31.20165696; doi: https://doi.org/10.1101/2020.07.31.20165696 5. Okada, H et al. “The 'hygiene hypothesis' for autoimmune and allergic diseases: an update.” Clinical and experimental immunology vol. 160,1 (2010): 1-9. doi:10.1111/j.1365-2249.2010.04139.x --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Gut Bugs: A Butyrate producing factory! Author: BugSpeaks Published: 2021-06-25 Category: Nutrition Meta Title: Gut Bugs: A Butyrate producing factory! Meta Description:
Trillions of microorganisms live within you (and even on you) and call your body home. There are bacteria, viruses, and URL: https://www.bugspeaks.com/blog/gut-bugs:-a-butyrate-producing-factory Trillions of microorganisms live within you (and even on you) and call your body home. There are bacteria, viruses, and fungi in many parts of your body, but the gut microbiome is the largest and most important. The gut microbiome is made up of all the microbes and genes found in the colon. In your intestine, there are approximately 1,000 different bacterial species. So, your body is not solely yours! Human health is strongly influenced by gut microbes. The good bacteria in our gut perform vital functions such as producing metabolites like short-chain fatty acids, vitamins and amino acids. These have a variety of health benefits, ranging from promoting a healthy immune system to maintaining gut lining integrity. One of the important aspects of the gut microbiota is “fermentation”. Dietary fibres that cannot be broken by the body are “fermented” by the gut resident bacteria producing a variety of metabolites including butyrate, an organic compound that belongs to the group of “short-chain fatty acids”. This metabolite has many important functions within the human body, particularly for digestive health, as well as supporting brain health and protecting against disease. These health-promoting molecules are an important source of energy for the body, accounting for anywhere from 5% to 15% of a person's daily caloric needs.  **Structure of butyrate and butyrate acid** Butyrate, the primary fuel for the cells that line the gut also referred to as "colonocytes," fulfils 90% of the energy requirements of those cells. Another important role of butyrate is that it is a Histone deacetylase inhibitor or "HDAC inhibitor,” thereby promoting anticancer activity. Furthermore, by lowering the gut's inflammatory capacity, it creates an environment in which the microbiome can thrive without triggering an acute immune response. This short-chain fatty acid also functions as an antioxidant and aids in gut health. It boosts the production of mucin, a gel-like substance that coats the inside of the gut, and promotes the growth of villi, microscopic finger-like extrusions that line the intestines. These mechanisms explain how it contributes to the integrity of the gut wall, also known as the "epithelial defence barrier," which prevents opportunistic pathogens, toxins and other harmful substances from entering the bloodstream. So, now that you're aware of the health benefits that butyrate can provide, we're sure you're eager to learn how to boost butyrate levels in the gut. You do not need to eat foods high in butyrate; instead, you should eat foods that feed your gut bacteria. Because butyrate is produced when your gut microbes convert the dietary fibre (also called prebiotic) that your body cannot digest into those incredibly beneficial compounds. A balanced diet with decent amounts of carb, Low-fat and protein can enhance the activity of the gut bacteria. Thereby, increasing butyrate level. Prebiotic dietary fibers are the best food source for our microbiome, which is found in plant based food like fruits, vegetables and pulses. Although many fad diets claim to have some benefits, they can also be harmful to your gut microbiome and its ability to produce butyrate. Hence, think twice before following any strict diet because our microbiome is variable and requires a balanced diet.  **Butyrate is produced by gut bacteria from prebiotic fibers in plant foods** So, it is to be remembered that a good, wholesome and a balanced diet can help us in feeding our microbes but also improve our overall well being in many folds. Whereas, a rigid and a wrong diet can have the potential to affect us in many possible ways. So, choose wisely what goes inside your body! **References:** 1. Guilloteau P, Martin L, Eeckhaut V, Ducatelle R, Zabielski R, Van Immerseel F. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutr Res Rev. 2010;23:366–384. \[[PubMed](https://www.ncbi.nlm.nih.gov/pubmed/20937167)\] 2. Rabbani GH, Teka T, Zaman B, Majid N, Khatun M, Fuchs GJ. Clinical studies in persistent diarrhea: dietary management with green banana or pectin in Bangladeshi children. Gastroenterology. 2001;121:554–560. \[[PubMed](https://www.ncbi.nlm.nih.gov/pubmed/11522739)\] 3. Chang, P. V., Hao, L., Offermanns, S., and Medzhitov, R. (2014). The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. U.S.A. 111, 2247–2252. doi: 10.1073/pnas.1322269111 4. [Cani, P, D. Human gut microbiome: hopes, threats and promises, 2018](https://gut.bmj.com/content/67/9/1716?aff=68) --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Increasing importance of Gut Health in the aftermath of COVID-19 Author: BugSpeaks Published: 2021-06-14 Category: Microbiome and Disease Meta Title: Increasing importance of Gut Health in the aftermath of COVID-19 Meta Description:
Since last year, when the entire world was struck by a massive health crisis as a result of the Coronavirus illness, the URL: https://www.bugspeaks.com/blog/increasing-importance-of-gut-health-in-the-aftermath-of-covid-19 Since last year, when the entire world was struck by a massive health crisis as a result of the Coronavirus illness, there has been an increasing focus on people's nutrition and lifestyle choices, compelling everyone to prioritize their health and well-being. COVID-19 has affected [over 100 million people](https://covid19.who.int/) worldwide, with the worst-affected countries being the United States, India, Brazil, Russia, the United Kingdom, and France. Among the many [symptoms of COVID-19](https://www.cdc.gov/coronavirus/2019-ncov/symptoms-testing/symptoms.html), some notable ones are nausea or vomiting, diarrhea and intestinal discomfort. This very fact has arisen the quest to find an answer to the following question, “What if SARS-CoV-2 and the microbes living inside our intestines have a significant link and whether this connection can be used as a potential tool to combat further infections ?” In fact, many studies are being conducted to understand this very link. People with underlying medical disorders such as high blood pressure, diabetes, and obesity have a higher chance of severe COVID-19. And the risk of major consequences and hospitalization increases with age, with the older population being the most vulnerable. However, both of these characteristics, old age and chronic illnesses, have been linked to a change in gut microbiota i.e., the different types of microbes present inside our gut. Alterations in the gut microbiota, also referred to as "gut dysbiosis," have been linked to a variety of long-term diseases and disorders, including Irritable Bowel Disorder, type 2 diabetes, depression, and cardiovascular disease.  Many people who survive their first encounter with the disease go on to have puzzling and sometimes crippling symptoms called "long COVID" for months. As a result, it's proposed that people who have COVID-19 and leave the hospital require continued treatment and rehabilitation to assist them recover. Long COVID can be exacerbated by abnormalities in the type of bacteria prevalent in the gut, according to studies. The gateway to our cells for the entry of the coronavirus is the protein called angiotensin converting enzyme 2 or ACE-2. This ACE-2 receptor protein is found in the lungs as well as in our intestines, indicating that the virus might have a significant relation with the gut microbiota. A study was conducted to see the changes in the types of microorganisms present in the faeces of 15 Hong Kong patients with SARS-CoV-2 infection during hospitalization and its relationship with severity and presence of the viral particles in their faeces. Their samples had significant alterations compared with controls(healthy individuals), that means they had more opportunistic or ‘bad’ microbes than the beneficial ones and this imbalance persisted even after SARS-CoV-2 was no longer present in their bodies and resolution of respiratory symptoms. This indicated that even after recovery, there was a pronounced imbalance in the gut, which can potentially impact the lungs, but also other organs as well as the immune system. So, it becomes important to keep higher levels of good bacteria in our bodies than the pathogenic ones. Diet, unsurprisingly, has a substantial impact on the composition of bacterial communities in the intestine. It is known from several studies that plant based foods, containing high fibres, have a beneficial impact on our gut health. Another new study has discovered a relation between plant-based and pescatarian diets and COVID-19 milder cases. Healthcare practitioners who ate a plant-based diet had a 73% lower risk of moderate to severe COVID-19, and those who ate a plant or fish based diet had a 59% lower risk of being very ill. The study also argued, despite having efficient healthcare facilities, the severely affected countries in the pandemic are the western countries, indicating that one of the underlying reasons behind this is the type of diet and lifestyle choices. Western diets containing processed foods with high fats, oils and refined sugars are major triggers for poor gut health. As a result, eating a more fibre diet, supplementing with probiotics, prebiotics, or synbiotics on a regular basis, exercising regularly, and most importantly, having our gut health examined becomes vital. Understanding the situations from the ground up is the need of the hour. Many investigations have yielded significant results regarding this continually changing and evolving pathogen. When a virus replicates in a host cell, it has a chance of mutating, which means it can make minor changes that affect the expression of certain proteins. Those changes can sometimes become fixed traits in the viral population, which helps them to survive in the host population. This suggests that the more copies the virus makes, the more it spreads and the more it can alter, adapt, and learn to thrive in that environment in the form of variants. And therefore, people are constantly encouraged to get vaccinated and continue maintaining all the necessary protocols and not let "pandemic fatigue" set in at any cost. With all the growing evidence pointing towards a link between host immunity, the microbes living inside us and the covid-19 , it is of utmost importance for everyone right now to practise towards making our gut healthy and strong. **References :-** 1. _Debojyoti Dhar, Abhishek Mohanty, “Gut microbiota and Covid-19- possible link and implications”, Virus Research, [https://doi.org/10.1016/j.virusres.2020.198018](https://doi.org/10.1016/j.virusres.2020.198018)._ 2. _Quigley, Eamonn M M. “Gut bacteria in health and disease.” Gastroenterology & hepatology vol. 9,9 (2013): 560-9._ 3. _Vasile Coman, Dan Cristian Vodnar, “Gut microbiota and old age: Modulating factors and interventions for healthy longevity”, Experimental Gerontology, Vol. 141 (2020), 0531-5565, https://doi.org/10.1016/j.exger.2020.111095._ 4. _Zuo, Tao et al. “Alterations in Gut Microbiota of Patients With COVID-19 During Time of Hospitalization.” Gastroenterology vol. 159,3 (2020): 944-955.e8. doi:10.1053/j.gastro.2020.05.048_ 5. _Kim H, Rebholz CM, Hegde S, et alPlant-based diets, pescatarian diets and COVID-19 severity: a population-based case–control study in six countriesBMJ Nutrition, Prevention & Health 2021 doi: 10.1136/bmjnph-2021-000272_ 6. _Zhang, Yu-Jie et al. “Impacts of gut bacteria on human health and diseases.” International journal of molecular sciences vol. 16,4 7493-519. 2 Apr. 2015, doi:10.3390/ijms16047493_ --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Microbiome Gazers Episode 5 Author: BugSpeaks Published: 2020-10-30 Category: General Meta Title: Microbiome Gazers Episode 5 Meta Description:
This is the 5th episode of the 'Microbiome Gazers'. Proudly hosted and organized by BugSpeaks® an URL: https://www.bugspeaks.com/blog/microbiome-gazers-episode-5 This is the 5th episode of the 'Microbiome Gazers'. Proudly hosted and organized by BugSpeaks® and Leucine Rich Bio. In yet another special episode, we were glad to have Nimrod Mon Brokman. Mon is the co-founder of Behavioural Foresight - a company in the field of Behavioural Science and Psychology that provides a holistic approach to the health of body and mind. His mental approach been shaped while working with Olympic athletes and different types of performers- from the encounters in the Israel Defence Forces and as a former professional athlete himself. **Highlights:** * Mon begins by sharing his own \[fascinating\] life experience as a fencer for Israel Olympic team to his work as a rescue worker for defence services to his academic orientation towards psychology. * With this experience, he brings his unique view on the mind and body and how to explore & exploit it further for personal development. * Later, Mon delves deeper into definitions of behavioural psychology and behavioural economics. * He differentiates the aspects of behavioural psychology into two approaches, a top down approach \[mind to body\] and bottom up approach \[body to mind\], and how microbiome plays a significant role in the bottom up approach. * Further, he adds as to how objective neurological measurements like biofeedback and neural feedback can be used to make subjective observations about an individual’s behaviour, to increase their potential – physically, mentally, and emotionally. * Given the pandemic and its side effects, Mon touches upon the aspects of stress, how crucial it is cope and manage it well, why it is critical to push us towards a favourable outcome, instead of a unfavourable outcome, which we are usually inclined towards. * The stress and its management bring them to the bottom up approach, where the microbiome, influencing the gut-brain axis, has a crucial role to play. * Kumar expands on this aspect, and stresses on the notion that “root of all disease is inflammation” and “root of all inflammation is in the gut”. * Mon and Kumar later talk about breathing, breathing exercises, gut microbiome and GBA, along with the criticality of respiratory system \[gut-lung-axis\]. * The focus later shifts to neurological measurements of brain waves and how its function can be used for making objective and subjective observations about behavioural diseases. * The most interesting aspect of these measurements, in context of gut and microbiome, comes with the idea of managing anything from acidity to constipation to anxiety and depression, by understanding the triggers for these biofeedback measurements. * Mon also demonstrates some aspects of breathing exercise, more specifically on the resonance breathing which can be adopted by almost everyone. * Kumar tops all this with some details on how the gut microbiome controls the behaviour from a simple hunger to a craving to much deeper behavioural patterns. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Microbiome Gazers Episode 4 Author: BugSpeaks Published: 2020-10-19 Category: General Meta Title: Microbiome Gazers Episode 4 Meta Description:
This is the 4th episode of the 'Microbiome Gazers'. Proudly hosted and organized by BugSpeaks® URL: https://www.bugspeaks.com/blog/microbiome-gazers-episode-4 This is the 4th episode of the 'Microbiome Gazers'. Proudly hosted and organized by BugSpeaks® and Leucine Rich Bio. This was a special episode where we were honoured to have Dr Shabnam Das Kar, a world-renowned functional medicine specialist, talking on “Role of Microbiome – A Clinician’s Perspective”. Dr Debojyoti Dhar, our co-founder and Director, hosted and coordinated this discussion. **Highlights:** * Dr Kar begins with her introduction and with an interesting point as to how the perspective of microbes have changed since the time when they were considered to be absolute bad to now being considered as an integrated part of our bodies. * She also briefly introduces the audience to the controllable or influenceable factors of developing once microbiome, from birth modes, diet and beyond. * She goes into details of birth modes and how some of the most fascinating studies conducted recently have unequivocally proved the importance of vaginal delivery over C-section delivery. * Later she moves onto diet, which is the most fascinating part for her, given her functional medicine expertise. * Diet is also the most important aspect for her in context of gut microbiome, since she believes that diet, not only has the biggest impact on gut microbiome, but its also the most under our control, as compared to that of geography, genetics and other factors. * She talks about the specific example of insulin resistance, diabetes, and microbiome to stress the importance of diet and microbiome, along with some more disease examples. * She also touches upon the interrelationship between immune system, microbiome, health, and medicine. * Then she describes the impact of medicines on gut microbiome, beginning with antibiotics to PPIs \[proton pump inhibitors\] to metformin \[antidiabetics\] to statins. * She briefly touches upon probiotics and faecal microbiota transplantation \[FMT\] as other methods of manipulating the gut microbiome. * The discussion later moves on to the kind or types of individuals, consulting for a functional medicine approach, should get their microbiome profiles, and how it aids the to improve their health. * Key shift to “predictive personalized medicine” and “patient participatory medicine”, and she shared many observations, experience, and insights she has gained during her consults. * Dr Dhar also shares his own experience and insights, which he gained through BugSpeaks® testing, and he stresses on the significance of clinician in this whole scenario. * They also discuss on the importance of building a community of clinicians/physicians, which understand the importance of microbiome and its impact on health. * Dr Kar shares her view on the challenges of using a microbiome test as a diagnostic method for diseases. * They wrap up by answering questions from the attendees. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Microbiome Gazers Episode 3 Author: BugSpeaks Published: 2020-10-14 Category: General Meta Title: Microbiome Gazers Episode 3 Meta Description:
This is the third episode of the 'Microbiome Gazers'. Proudly hosted and organized by BugSpeaks® and Leucine URL: https://www.bugspeaks.com/blog/microbiome-gazers-episode-3 This is the third episode of the 'Microbiome Gazers'. Proudly hosted and organized by BugSpeaks® and Leucine Rich Bio. Here Co-founders of Leucine Rich Bio Dr Debojyoti Dhar \[as guest\] and Mr Kumar Sankaran \[as host\] talk about the technologies – both sequencing and computational analysis – used within the field of microbiome. **Highlights:** * They begin with a brief comparison between the traditional microbiology / culture based approached and modern microbiome-based methodology. * Factors involved in culturing-based methods and the associated drawbacks, especially regarding the inability to culture close to 2000 plus species that might thrive in a gut. * Early technological walkthrough from Fluorescent In Situ Hybridization (FISH) to early PCR based amplicon sequencing, and other DNA based sequencing methods. * About current technologies used – Amplicon \[mostly rRNA\] sequencing, whole genome metagenomics. * Quick insights on meta-transcriptomics technology. * Differences between using short read vs long read sequencing technologies. * Details on various analysis platforms or suites available for analysing the metagenomic data, with a general follow through of major steps involved during a metagenomics analysis. * Thoughts on the criticality of sample collection and extraction of DNA/RNA, and its impact on bioinformatic analysis. * Thoughts on the criticality of reference databases. * Thoughts on MetaRich - Leucine Rich Bio’s suite of metagenomic analysis suite, with specific focus on MetaRef \[Microorganism reference database\], MicroByte \[database of associations between diseases and microbiome\] and NutriBite \[ database of associations between nutrition and microbiome\]. * They wrap up by answering questions from attendees. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Microbiome Gazers Episode 2 Author: BugSpeaks Published: 2020-10-05 Category: General Meta Title: Microbiome Gazers Episode 2 Meta Description:
This is the 2nd episode, of the 'Microbiome Gazers'. Proudly presented by BugSpeaks® and Leucine URL: https://www.bugspeaks.com/blog/microbiome-gazers-episode-2 This is the 2nd episode, of the 'Microbiome Gazers'. Proudly presented by BugSpeaks® and Leucine Rich Bio. In this interactive session, under the aegis of BugSpeaks live stream series, co-founders of Leucine Rich Bio and developers of South Asia's first gut microbiome test - BugSpeaks® - Dr Debojyoti Dhar and Mr Kumar Sankaran talk about the evolution of microbiome reporting with respect to BugSpeaks®. The speakers discuss about the various facets of BugSpeaks® report. The key highlight was the "question-answer" session between the attendees and the panellists. **Highlights:** * Observations about microbiome reporting in general and how the BugSpeaks® report have evolved in the last 2-3 years. * How the transition from "one-page clinical report" to "an elaborate report" happened, both with BugSpeaks® and other microbiome companies across the world. * How the intriguing nature of microbiome, which has been interesting even to the lay person, have driven a detailed BugSpeaks® report over the years. * How balancing the aspects of microbiome that is relevant to both clinical doctor and common man was key to this reporting. * What kind of diseases would BugSpeaks® analysis, reporting and recommendations would be most impactful? * The causation vs correlation/association debate and how our disease risk estimation has been conceptualized. * Other salient features of BugSpeaks® report, including the Gut Microbiome Index \[GMI\], pathogen characterization, and most importantly the personalized 3-Phase dietary recommendations. * How BugSpeaks® GMI is different from that of Bacteroidetes to Firmicutes ratio or any other metric generally used and how we are on a quest for a universal standard for representing the extent of dysbiosis of gut microbiome. * More on the direction of personalized nutrition and diet and how BugSpeaks® can contribute to this area. * Why a "single time, eat or don’t eat kind of diet" has been unsuccessful and why multiphase dietary recommendations is the way to go forward. * Detailed description of the 3-Phase Diet and importance of Supplements during Phase 2 and why going natural is important than finding off the shelf probiotics. * Why achieving personalized nutrition is extremely hard. * How all of these would aid everyone from individuals to nutritionists to clinicians to take actionable efforts towards better health. * They wrap up the discussion by answering questions from the attendees. --- This blog is powered by Superblog. Visit https://superblog.ai to know more. --- ## Microbiome Gazers Episode-1 Author: BugSpeaks Published: 2020-10-02 Category: General Meta Title: Microbiome Gazers Episode-1 Meta Description:
This is the very first, of the many episodes, of the 'Microbiome Gazers'. One of our own and proudly hosted and organized by BugSpeaks
URL: https://www.bugspeaks.com/blog/microbiome-gazers-episode-1
This is the very first, of the many episodes, of the 'Microbiome Gazers'. One of our own and proudly hosted and organized by BugSpeaks® and Leucine Rich Bio.
Here Co-founders of Leucine Rich Bio Dr Debojyoti Dhar \[as guest\] and Mr Kumar Sankaran \[as host\] talk about their journey and the challenges in the field of microbiome, developing BugSpeaks® - South Asia's first gut microbiome test and more.
**Highlights:**
* How did the microbiome journey begin for Leucine Rich Bio and Kumar?
* What are strengths of Leucine Rich Bio in the field of microbiome, and how the diverse team of microbiologists and computational biologists working on genomics to metabolomics, statistics, machine learning and beyond, are key contributors to developing BugSpeaks®.
* Memories of the first microbiome or BugSpeaks® client we had.
* What are the challenges in the microbiome field in general and with specific to India?
* What is the right or balanced team composition to come up with tools and tests that can give key solutions through microbiome analysis?
* First \[microbiome\] clinical study partnership/collaboration and its experience.
* How logistics is one of the key aspects of this whole testing, how it can make or mar the whole development and how we cracked the logistics of BugSpeaks® and how this helped us to be a truly global company.
* Key points on awareness of microbiome in general and how the awareness of BugSpeaks® as a test have brought about some key improvements.
* How writing our own blogs, starting YouTube channel, constant presence on twitter and other social media platforms and now this, the microbiome gazers, have all added up to the awareness.
* Which are the domains in the medical field that Leucine Rich Bio is perusing with BugSpeaks®
* They end with talking about, what is in store for Leucine Rich Bio and BugSpeaks® the future.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Bite Sized BugSpeaks Ep-2
Author: BugSpeaks
Published: 2020-08-07
Category: General
Meta Title: Bite Sized BugSpeaks Ep-2
Meta Description:
Once upon a time, being chubby and obese was a mark of affluence. As in earlier times, food was often insufficient, and
URL: https://www.bugspeaks.com/blog/being-chubby---mark-of-affluence-or-aberration
Once upon a time, being chubby and obese was a mark of affluence. As in earlier times, food was often insufficient, and famines were common obesity meant prosperity. But the world changed, and food became abundant in many regions of the world. So, what was once a mark of prosperity, has now become a sign of an unhealthy lifestyle. Obesity involves the accumulation of a considerable amount of body fat, which intensifies the risk of additional health hazards—for example, diabetes, heart disorders, liver diseases, high blood pressure, and certain cancers.
Yet, over the past few decades, obesity and related co-morbidities have reached epidemic magnitudes. People continue to eat more. The modern industrial world considers it a [crisis of communal](https://www.nejm.org/doi/full/10.1056/NEJMoa1614362) wellbeing. Generally, obesity is a result of an amalgamation of a few factors like heredity, lifestyle, and environment. Among the environmental elements, the gut microbiome flourishing in our bodies seems to play a vital role.

**Our gut microbiome**
As our gut plays home to a vast and diverse array of microorganisms, they, in return, play an essential role in several vital processes, like, digestion, metabolism, and energy production. Five bacteria, Bacteroidetes, Firmicutes, Actinobacteria, Proteobacteria, and Verrucomicrobia, are predominant in the gut flora of an average healthy individual, with Bacteroidetes and Firmicutes, making around [90%](https://www.ncbi.nlm.nih.gov/pubmed/15505215) of the total. Our gut flora, while regulating numerous bodily processes, also influences the production, metabolism, storage, and [breakdown of lipids](https://www.ncbi.nlm.nih.gov/pubmed/31391921). Lipids are the small molecules that also makeup fat.
**How microscopic gut microbes influence macroscopic bodyweight?**
Like in other metabolic diseases, the gut microbiota in over-weight people shows misbalance as compared to normal [healthy individuals](https://www.ncbi.nlm.nih.gov/pubmed/15505215). Dysbiosis, the imbalance in the proportion of good and bad gut microbes, causes varied digestion of otherwise indigestible complex carbohydrates. Such digestion of carbohydrates contributes to the risk of developing obesity.
Digestive juice of the upper GI tract cannot digest complex dietary carbohydrates, including polysaccharides, oligosaccharides, and starches of plant origin. Only the microbiota harbouring our gut can ferment them and produce Short Chain Fatty Acids (SCFAs). [SFCAs](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5867888/) act as the primary energy source to support both the microbial growth and the gut cells. It also regulates optimal lipid and cholesterol metabolism besides colonic immunity.
But, when you continuously eat food low in complex carbohydrates, it alters gut pH and modifies the structure and metabolism of gut microbiota—reduced dietary carbohydrate results in the reduction of total SCFA. Also, an alteration in gut microbiome profile characterized by decreased Roseburia, the Eubacterium, and Bifidobacteria occurs.
Additionally, a protein-rich diet with a low proportion of carbohydrates also alters gut microbiome [composition](https://www.ncbi.nlm.nih.gov/pubmed/24999296). The ratio of the abundance of Gram+ and Gram− bacteria gets ultimately reverses in the gut of both the obese and [anorexic people](https://www.ncbi.nlm.nih.gov/pubmed/28636668).
**Gut crisis when good microbes crash**
Health remains the most valuable currency. If not realized on time, it can cost you a lot of your actual money. So, how the good microbes go up and down in an obese gut? Gut microbiome composition is extremely diverse between healthy individuals, lean, and overweight ones, characterized by the low [bacterial richness](https://www.nature.com/articles/nature12506) in obese subjects.
High numbers of Firmicutes and low numbers of [Bacteroidetes](https://www.ncbi.nlm.nih.gov/pubmed/23719380) occur in obese people. The proportion of Bacteroidetes increases with low-calorie diet-based [weight loss](https://www.ncbi.nlm.nih.gov/pubmed/17183309), thus, suggesting a tight association between the gut microbiota and body [weight regulation](https://www.ncbi.nlm.nih.gov/pubmed/6834773). Less diversified gut flora during obesity becomes inefficient in consuming energy produced from [metabolic processes](https://www.ncbi.nlm.nih.gov/pubmed/23478685). Altered gut flora in overweight people thus causes amplified production of energy from food. It also promotes the storage of fats by encouraging the [formation of biological](https://www.ncbi.nlm.nih.gov/pubmed/27703805) products required for the same.
Obesity-induced transformed intestinal flora also produces bacterial endotoxins, which pass into our circulation and cause inflammation and insulin resistance. Finally, leading to enhanced fat deposition and related [complications](https://www.ncbi.nlm.nih.gov/pubmed/27616451). Alterations of the gut microbiome in initial life also boost the onset of obesity due to altered mechanisms regulating [satiety/appetite](https://www.ncbi.nlm.nih.gov/pubmed/25126780).
Gram-negative bacteria, abundant in the gut of obese people, damage the gut epithelium and enhance the permeability of the intestinal wall. As a consequence, loads of bacterial endotoxins enter the blood circulation stimulating the production of molecules associated with [obesity](https://www.ncbi.nlm.nih.gov/pubmed/20508158). Moreover, excessive gut barrier disruption can also result due to the activation of metabolic pathways that affect adipose tissue. This also aggravates the initial disorders and leads to a vicious cycle of [detrimental processes](https://www.ncbi.nlm.nih.gov/pubmed/20664638).
**Giving the microbes healthy nutrition**
One can correct dysbiosis induced obesity by tracing the path back to healthy gut microbiota. Just correcting the proportions of healthy microbes can help you become lean. One can reform gut dysbiosis by several months of the regular low-energy diet, including intake of some micronutrients, prebiotics, and probiotics. Altered diet impacts the regulation of gene expression of the liver, muscle, and adipose tissue along with the alteration of [gut microbiome composition](https://www.ncbi.nlm.nih.gov/pubmed/25361999). Regular intake of probiotics improves gut health by restoring the gut epithelium, healthy bloodstream, and controlling [local inflammation](https://www.ncbi.nlm.nih.gov/pubmed/28792488)19.
Antioxidants present in tea, like oolong tea polyphenols (OTP), mainly increase gut bacterial biodiversity with a higher number of Bacteroidetes and a low number of Firmicutes. They are thereby helping your gut experience the healthy flow of SCFAs once again. Intake of another prebiotic, [galacto-oligosaccharide (GOS)](https://www.ncbi.nlm.nih.gov/pubmed/29465126), encourages the abundance of Bacteroides, Ruminococcaceae, and Oscillibacter. It not only reduces the production of harmful bacterial metabolites but also brings down blood triglyceride levels.
Thus, probiotics and prebiotics like, GOS and OTP can be used to prevent obesity-related metabolic disorders through modification of the [intestinal microbiota](https://www.ncbi.nlm.nih.gov/pubmed/29355278). Additionally, regular physical exercise also helps to lose weight. It also restores gut health by encouraging beneficial gut flora, which synthesizes SCFAs and controls the [local inflammation](https://www.ncbi.nlm.nih.gov/pubmed/31380886). Another therapeutic option to treat obesity is through various surgical techniques, known as Bariatric surgery, which find a relation with alteration of gut flora and weight reduction and other [health benefits](https://www.ncbi.nlm.nih.gov/pubmed/28359101).
**Gut microbiome test: a helpful tool**
The fact is that even modest weight loss can improve or prevent obesity-associated complications. For that, it is a prerequisite to have a ‘Gut microbiome test.’ It identifies your unique microbiome. State-of-the-art, ‘DNA sequencing technology,’ and algorithm-based data acquisition are used in this process. A prescription diet is formulated comprising of probiotics, and prebiotics based on the test results. Dietary changes increased physical activity, and lifestyle is the key to lose weight.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Gut Microbiome in Atherosclerosis: Unclogging the Bloody Pipes
Author: BugSpeaks
Published: 2020-07-10
Category: Microbiome and Disease
Meta Title: Gut Microbiome in Atherosclerosis: Unclogging the Bloody Pipes
Meta Description: All the blood in the human body flows through a network of arteries, just like the drainage system in house. When not cl
URL: https://www.bugspeaks.com/blog/gut-microbiome-in-atherosclerosis:-unclogging-the-bloody-pipes
All the blood in the human body flows through a network of arteries, just like the drainage system in house. When not cleaned regularly, sometimes the pipes in the system get blocked as the dirt and filth collect. Similarly, your arteries may get blocked if not cared for properly, as the fatty matter deposits in them. The arteries become narrow leading to the grave disease called atherosclerosis. So why the arteries get clogged? The sedentary lifestyles are one to blame for.
Moreover, elevated blood cholesterol, high blood pressure, diabetes, obesity, family history, an unhealthy lifestyle, and abnormal gut microbes are blamed for the [disease](https://www.ncbi.nlm.nih.gov/pubmed/23618829). Yes, you read it right. The microbes residing in your gut, which make up the gut microbiome can also place you at atherosclerosis risk. Your gut microbiome plays a vital role in controlling the metabolism and health of organs. Hence, it is quite relevant to explore the purpose of this diverse ecosystem of microorganisms in causing atherosclerosis.

**Gut microbiome dysbiosis: Cause of blockage**
Atherosclerosis is a classical chronic inflammatory disease. The healthy gut flora supports lipid (small molecules which makeup fat) metabolism and prevents the development of lesions on [arteries](https://www.nature.com/articles/s41467-017-00900-1). But [gut dysbiosis](https://www.ncbi.nlm.nih.gov/pubmed/23212374), characterized by maladaptation of gut flora, proves to be a significant player in the event of atherosclerosis. Under this condition, a relatively lower abundance of beneficial microbes such as Roseburia and Eubacterium, and overpopulation of harmful ones such as Collinsella occurs. Not only this, but dysbiosis also damages the [gut wall](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4590619/) and impairs its integrity. It gives the microbial components like lipopolysaccharide (LPS) and peptidoglycan a chance to escape into the blood circulation and reach arteries. Upon entering the arteries, these components cause inflammation resulting in the development of [atherosclerosis](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4456003/)9.
**What all microbial materials block your arteries?**
It is not only the components of gut microbes wreaking havoc in arteries. Specific metabolites produced by gut microbes also control inflammation and atherosclerosis. These metabolites include short-chain fatty acids (SCFAs), methylamines, polyamines, trimethylamine N-oxide (TMAO), and secondary bile acids (BAs). All of them play an essential role in [fat metabolism](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5579652/#!po=34.0000).
SCFAs arise from microbial fermentation of starches and complex dietary fibres in the intestine. Taking part in further metabolic events in the body, the SCFAs inhibit the accumulation of lipid and inflammation in the arterial wall. Also, [SFCAs](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5579652/#!po=34.0000) induce secretion of gut hormones, which reduce food intake. But in a dysbiotic gut, [butyrate](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5579652/#!po=34.0000), a critical SCFA occurs in low amounts, as butyrate-producing bacteria such as Roseburia, occur in small numbers. Likewise, the inflammatory insults on the arterial walls increases.
TMAO comes from the oxidation of trimethylamine (TMA), a product of gut microbe activity in the liver. Varying compositions of bacteria pose differential abilities to generate TMAO. Thus, a dysbiotic gut containing higher numbers of TMA-producing bacteria can produce higher levels of [TMAO](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5579652/#B58-nutrients-09-00859). A higher level of TMAO causes atherosclerosis and larger [plaque area](https://www.nature.com/articles/nature09922). Even low levels of circulating TMAO attenuates atherosclerosis. Studies have found a correlation between TMAO levels and certain human gut microbes’ types. For example, [Prevotella](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5579652/#B59-nutrients-09-00859) finds a relation with high TMAO levels while Bacteroides produce lower levels.
Bile acids (BA) form another group of metabolites produced from cholesterol. BA helps absorb dietary lipids and fat-soluble vitamins. Gut microbiota-derived enzymes [convert](https://www.ncbi.nlm.nih.gov/pubmed/26939849) primary BAs into secondary BAs. Secondary BA influences cholesterol metabolism in host and energy expenditure. But when in a dysbiotic gut, the enzyme activity reduces the gut does not reabsorb secondary BAs that occur in large numbers.
So, these secondary BAs enter the blood circulation, impair elimination of cholesterol, and signal deposition of fats, leading to the development of atherosclerosis. [Methanobrevibacter smithii](https://www.ncbi.nlm.nih.gov/pubmed/22972297), Clostridium, and Enterococcus find an association with reduced enzyme activity.14 Additionally, the [BA receptors](https://www.ncbi.nlm.nih.gov/pubmed/27045028) in inactivated form inhibit atherosclerosis formation by reducing macrophage inflammation and lipid loading15.
**How to manage and correct the blockage??**
To correct the blockage, we just need to treat the source, meaning the gut microbiome. If one can restore the balance in the gut, the balance in arteries may also be restored. Therefore, the intestinal microbiome offers a new potential target for the treatment of atherosclerosis.
**Probiotics**
Just like pouring a cleaning solution down the drainage pipe helps remove collected material, similarly, pouring probiotics in your body can help. [Probiotics](https://www.ncbi.nlm.nih.gov/pubmed/25529048) contain beneficial live microorganisms. Ingestion of viable microorganisms, like, Lactobacillus rhamnosus GR-1, helps hydrolyse bile salts, lower blood cholesterol, and reduce the risk of developing atherosclerosis. In essence, the beneficial microbes can help restore healthy metabolism and removal of plaques in arteries.
**Healthy diet**
Increased consumption of [whole-plant](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4315380/) foods, including fruits, vegetables, and wholegrain [cereals](https://www.ncbi.nlm.nih.gov/pubmed/22360862), helps modulate the gut microbiota. These foods share an inverse correlation with the risk of atherosclerosis development. Antioxidant abundant plant polyphenols prevent the oxidation of low-density lipoprotein. [Cranberry extract](https://www.ncbi.nlm.nih.gov/pubmed/25080446) (CE), a rich source of polyphenols, reduces fatty deposits on arterial walls. Additionally, the intake of CE increases the number of beneficial bacterium Akkermansia. It helps reduce fat storage and the size of plaques.
Intake of fibre-rich wholegrain foods, on the other hand, associates with lower blood glucose and total lipid. Dietary fibres also aid in the proliferation of [gut-friendly flora](https://openheart.bmj.com/content/6/1/e000993#ref-48): Bifidobacterium, Lactobacillus, and Enterococcus. Thus, a diet based on healthy foods promotes a happy and healthy microbiome. Strategies that modulate the gut microbiota and their metabolic activities by whole-plant foods, probiotics, and prebiotics may be at the base of healthy eating pyramids. They can decrease the risk of atherosclerosis and related co-morbidities21.
The [modern-day diet](https://www.ncbi.nlm.nih.gov/pubmed/22468338) remains rich in high calorie processed food with high sugar and fat content. While raw and whole food and dietary fibre and fermentable substrates are relatively low, this type of urban diet with high saturated and trans fatty acids relates to the depletion of good metabolic fuels. All this negatively alters the gut microbial profile contributing to increased cholesterol levels[.](https://openheart.bmj.com/content/6/1/e000993#ref-53) A shift towards a plant-based diet may confer protective effects against atherosclerosis by promoting protective factors and reducing harmful ones.
**Cow Milk**
Drinking' [bovine colostrum'](https://www.ncbi.nlm.nih.gov/pubmed/22236001), the milk produced by cow up to a few days after the birth of calf remains beneficial. The colostrum contains anti-LPS compounds, which alleviate hyperlipidaemia and atherosclerosis. These compounds also help in healing 'leaky' gut mucosal membranes by removing harmful microbial toxins circulating in the blood. Thus, these compounds reduce local inflammation and prevent disease progression.
**Surgery & Exercise**
[Bariatric procedures](https://pubs.acs.org/doi/abs/10.1021/pr400748f), mainly used to lose weight, also help improve obesity-associated metabolic disorders such as atherosclerosis. It also helps in the alteration of the gut microbiota as the body experiences a significant shift in metabolic requirements. Regular physical exercise also alters gut microbial composition. [Exercise](https://www.ncbi.nlm.nih.gov/pubmed/28862530) increases the number of SCFA producing bacteria, thus helping in preventing atherosclerosis and related complication[25](https://openheart.bmj.com/content/6/1/e000993#ref-64).
**Summing Up**
Atherosclerosis is a prevalent disease, and surely, you will accept that the health of your gut microbiome and your lipid metabolism are entangled. So, to cure it, you need a 'Gut Microbiome Test.' It uses sophisticated 'DNA sequencing technology' in stool samples to give a snapshot of the exclusive assortment of microorganisms present in your gut. Then this data will then be used to prepare your individualized diet plan, comprising probiotics, prebiotics, and regular physical exercise. In this way, you can tackle your atherosclerosis.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Urinary Metabolites & Microbiome
Author: BugSpeaks
Published: 2020-06-23
Category: Microbiome and Disease
Meta Title: Urinary Metabolites & Microbiome
Meta Description: "I'm not just your PEE but your METABOLIC INDEX" From the t
URL: https://www.bugspeaks.com/blog/urinary-metabolites-&-microbiome
"I'm not just your PEE but your METABOLIC INDEX"
From the time immemorial 'PEE or Urine' has been regarded as a vital biofluid with substantial biological information and details and an indicator to the one’s "Pink of Health"and "Striking vigour".
Does it remind you of our Grandma’s Traditional knowledge and Home remedies …??
Of Course!!
It is well established fact that the properties of urine have long been regarded as a yardstick for wellness, a fact well substantiated in its mention in scriptures of Shushrutha in Indian Alternative Medicine like Ayurveda and Naturopathy. The use of urine in medical analysis dates to ancient Egypt, Byzantine Era, and the middle ages. Hippocrates largely legitimized the medical practice of Uroscopy, even the use of urine colour wheels for diagnosis of various diseases was much in vogue.
Thus, the Colour, Odour and the Frequency of urination can give a hint of what is going on in the body... so let’s face it... Do check it out before you flush it out of sight...

Urine being the favoured biofluid clinically is a storehouse of metabolic by products such as Urea, Uric Acid, Creatinine, Inorganic Ions, several pigments and the remnants of food, drinks, drugs, chemicals, environmental contaminants, and microbial intermediates. The presence of significant levels of proteins and sugars indicate potential health concerns. However, it is sterile, obtained in large volumes and largely free from proteins and lipids.
Being chemically diverse, it is always a challenge to characterise chemical composition of the “Water of Life” as it is also referred to, while the newly advent technologies like NMR Spectroscopy, Gas Chromatography Mass Spectroscopy, High Performance Liquid Chromatography, Direct Flow Injection Mass Spectrometry, Inductively coupled plasma mass spectrometry comes in handy yielding a comprehensive, quantitative and metabolome wide elucidation of urine. These multiple metabolic platforms have helped to identify and annotate around 3079 urinary metabolite species in total. Thus, Urine stands foremost as the highly preferred biological sample for metabolomics research.
Metabolomics stand as relatively new field in comparison with its sister “omics”” (Genomics and Proteomics) yet highly promising, for it being “the study of Metabolites” which are the downstream products of various genome wide and Proteome wide interactions, is essentially a sensitive measure of an organism’s phenotype. With its far-reaching impacts as Disease Biomarkers and as Indicators of Environment gene Interactions and Drug Toxicity ,metabolomics warrants its potential by taking a step further in predicting the microbial health of an individual.
**Microbial Metabolomics:**
The Gut Microbiota consists of 10 trillion microbial cells and is a dynamic stage for small molecules and other bioactive compounds that can trigger both host metabolic and immune interplay. The microbial ecosystems in the gut contain about 1000 different bacterial species with a close-knit homeostasis established among them through cell-cell signalling and production of anti-microbial peptides. Along with achieving community dynamics with other neighbouring microbes, the gut microflora also communicates with the human host either in a symbiotic or deleterious fashion which in turn contributes to human diseases. With the advances in metagenomics and Metabolomics ,particularly 16sRNA amplicon sequencing and Shot Gun Sequencing has led to the identification of previously unidentified members of gut microbial community.
**Urinary Microbial Metabolites as Disease Markers:**
The diagnosis of gastrointestinal (GI) diseases is usually based on techniques such as upper or lower GI endoscopy, while extremely sensitive and specific non-invasive diagnostic or screening tools are usually lacking. Hence, it is imperative for the development of novel technologies for the detection of these disorders at an early stage.
In this regard, Urinary Microbial Metabolites as the non-invasive biomarkers of the gastrointestinal disorders is highly convincing.
**Microbial Metabolites in IBD:**

Hallmark of IBD is the microbial dysbiosis characterized by the decrease in commensals versus the pathogenic organisms. Commensals are protective in function against the pathogens through the secretion of bioactive substances as SCFAs (Short Chain Fatty Acids) which maintains the epithelial integrity and improves the barrier function are produced from the microbial fermentation of Dietary fibre by anaerobic microbiota in the intestine and colon. SCFAs activate specific G protein coupled receptors specifically GPR 43 and GPR41 which in turn mediate cell activation, proliferation, differentiation, and production of other bioactive molecules. The other mechanisms of SCFA include inhibition of HDAC, through improving the acetylation of histones impacts several genes and proteins. SCFAs also regulate T-cell differentiation and response through mTOR activation.
There is a marked depletion in urinary microbial metabolites such as SCFA, Methylamine, Trimethylamine, Fucose, Citrate, Hippurate, Taurine, Succinate, 2-Hydroxy-Isobutyrate levels during the progression of IBD.

**Microbial Metabolites in Neurological Behaviour:**
Tryptophan ,the least abundant amino acid in proteins is necessary for protein biosynthesis and synthesis of neurotransmitters like Serotonin and Melatonin. The metabolic activity of Gut microbiota influences the availability of Tryptophan, consequently the serotonergic signalling, which further emphasises the significance of gut microbiota in modulation of human behaviour.
Tryptophan is metabolised by microbial enzymes to a range of indole derived intermediates such as Indole 3 propionate and Indole-3-Acetate which are characterized in the biofluids. Tryptophanase in Indole producing bacteria brings about the formation of Indole from Tryptophan, further, Oxygenases in the non-indole producing bacteria catalyse the production of Indole-3-Propionate (IPA). The commensal bacteria primarily Clostridium sporogenes largely demonstrates the synthesis of IPA from Tryptophan. The detection of the novel metabolites like Methyl Indole 3 Acetate and Methyl Indole 3 Propionate in urine warrants the potential role of gut microbiota on serotonin related gut brain axis disorders.
**Arabinitol:**
Several Candida species like C. albicans, C. tropicalis. C. parapsilosis, C. pseudotropicalis, C. lusitaniae, C. guilliermondii produce D. Arabinitol in cultures. The elevated D-Arabinitol and L arabinitol ratios confers a serious threat to the susceptibility to Invasive candiasis (grave infection caused by the species of Candida) giving early warning signs for a potential candidate to set out on an anti-fungal therapy at the earliest.
**Microbial Phenolic metabolites & Inflammation:**
Several low molecular weight phenolic acids of microbial origin are produced in the blood of septic patients at high levels. Bifidobacteria, lactobacilli, produced in vitro considerable amounts of phenyllactic and p-hydroxyphenyllactic acids, Clostridia s. produced great quantities of phenylpropionic and p-hydroxyphenylpropionic acids, p-hydroxyphenylacetic acid was produced by Pseudomonas aeruginosa and Acinetobacter baumanii; and benzoic acid, by Serratia marcescens.
Influence of Phenolic acids on ROS production in mitochondria and neutrophils has been established. Low-molecular weight phenolic acids of microbial origin participate in the regulation of the ROS production in both the circulation and tissues, thereby affecting the level of oxidative stress in sepsis and inflammatory syndrome... The mechanism of sepsis includes the excessive production of defensive and inflammatory responses like increased generation of ROS,NO and inflammatory cytokines. The level of oxidative stress is crucial in the genesis and outcome of sepsis.
Pathogen-associated molecular patterns such as bacterial lipopolysaccharides and lipoteichoic acids, which are recognized by immune cells and initiate an acute response to pathogens; microbial phenolic acids may be assumed to regulate the magnitude of this response. Phenolic acids act on targets that produce ROS in the organism. The mechanism underlying the action of Phenyllactic acid and Para Hydroxy Phenyllactic acid is related to scavenging superoxide anion. On the contrary, cinnamic and benzoic acids do not act as ROS scavengers but exhibit the prooxidant activity, interacting with thiol groups. Presumably, their effect can be explained by the inhibition of NADPH-oxidase. Some phenolic acids, which decrease ROS production in both mitochondria and neutrophils, can play a role of natural antioxidants. By affecting neutrophils, they retard the immune response, whereas while acting on mitochondria, they prevent the development of multiple organ failure or reduce it.
Thus, microbial metabolites can either increase or reduce the inflammatory syndrome. The high levels of phenyllactic and p- hydroxyphenyllactate acids correlate with the development of sepsis and the risk of lethal outcome.
**References:**
1\. Bouatra S, Aziat F, Mandal R, Guo AC, Wilson MR, Knox C, et al. (2013) The Human Urine Metabolome. PLoS ONE 8(9): e73076. [https://doi.org/10.1371/journal.pone.0073076](https://doi.org/10.1371/journal.pone.0073076)
2\. Martinez, K. B., Leone, V., & Chang, E. B. (2017). Microbial metabolites in health and disease: Navigating the unknown in search of function. Journal of Biological Chemistry, 292(21), 8553–8559. doi:10.1074/jbc.r116.752899
3\. Knights D, Lassen KG, Xavier RJ : Advances in inflammatory bowel disease pathogenesis: linking host genetics and the microbiome Gut 2013;62:1505-1510.
4\. Sarosiek I, Schicho R, Blandon P, Bashashati M. Urinary metabolites as noninvasive biomarkers of gastrointestinal diseases: A clinical review. World J Gastrointest Oncol. 2016;8(5):459-65.
5\. [Tereza Pavlova](https://www.sciencedirect.com/science/article/pii/S0003267017309595?via%3Dihub#!), [Veronika Vidova](https://www.sciencedirect.com/science/article/pii/S0003267017309595?via%3Dihub#!), [Julie Bienertova- Vasku](https://www.sciencedirect.com/science/article/pii/S0003267017309595?via%3Dihub#!), [Petr Janku](https://www.sciencedirect.com/science/article/pii/S0003267017309595?via%3Dihub#!), Martina Almasi, [Jana Klanova](https://www.sciencedirect.com/science/article/pii/S0003267017309595?via%3Dihub#!), [Zdenek Spacil](https://www.sciencedirect.com/science/article/pii/S0003267017309595?via%3Dihub#!), [Analytica Chimica Acta](https://www.sciencedirect.com/science/journal/00032670), [Volume 987](https://www.sciencedirect.com/science/journal/00032670/987/supp/C), 22 September 2017, Pages 72-80
6\. Beloborodova N, Bairamov I, Olenin A, Shubina V, Teplova V, Fedotcheva N. Effect of phenolic acids of microbial origin on production of reactive oxygen species in mitochondria and neutrophils. J Biomed Sci. 2012;19(1):89. Published 2012 Oct 12. doi:10.1186/1423-0127-19-89
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Depressed gut, Depressed mind
Author: BugSpeaks
Published: 2020-02-27
Category: Microbiome and Lifestyle
Meta Title: Depressed gut, Depressed mind
Meta Description: We all might have experienced bouts of depression some time in our life. Depression, a common psychiatric illness, presents a demo
URL: https://www.bugspeaks.com/blog/depressed-gut,-depressed-mind
We all might have experienced bouts of depression some time in our life. Depression, a common psychiatric illness, presents a demoralized state wherein both the mind frame and body get affected. It affects feelings and judgmental capabilities and compromises the [quality of life](https://www.guilford.com/books/Handbook-of-Depression/Gotlib-Hammen/9781462524167). Usually, depressed people are unable to concentrate and feel low all the time. Consequently, their performance in different areas such as academics or profession drops, causing additional stress. In extreme cases, it may lead to severe hopelessness and self-harm, with the thought of [self-destruction and death](https://www.who.int/topics/mental_health/factsheets/en/).
Depression, stress, and anxiety all are interrelated and characterized by alteration in food habits and sleeping patterns. Stress and depression influence the release of cortisol (stress hormone). [Cortisol](https://www.ncbi.nlm.nih.gov/pubmed/22972297) modifies gut immunity, ultimately altering the microbial population in gut. It causes a significant modification in the gut microbial diversity, in terms of intensification or decline of individual species. However, the introduction of different genera or the complete removal of specific bacterial genera is [not observed](https://www.ncbi.nlm.nih.gov/pubmed/27005587).

**Role of Gut Microbiome**
The gut flora of clinically depressed individuals differs in terms of the abundance and diversity of the common microbes. These include _Bacteroidetes_, _Firmicutes_, _Proteobacteria,_ and _Actinobacteria_. Further, a reduced abundance of bacteria metabolizing carbohydrates also occurs in individuals with depression as compared to healthy ones. These include [_Bifidobacterium_](https://www.ncbi.nlm.nih.gov/pubmed/24742328), [_Faecalibacterium_](https://www.researchgate.net/publication/320413695_Prebiotic_potential_of_pectin_and_pectic_oligosaccharides_to_promote_anti-inflammatory_commensal_bacteria_in_the_human_colon), _Bacteroides_ _Coprococcus_, _Dialister,_ and [_Ruminococcus_](https://www.ncbi.nlm.nih.gov/pubmed/20581222). While several other bacterial species, playing a pivotal role in protein and amino acid metabolism, remain over-populated in depressed subjects. Such bacteria involve [_Clostridium_](https://www.ncbi.nlm.nih.gov/pubmed/24499426), [_Klebsiella_](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5682003/), [_Parabacteroides_](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4850288/), [_Streptococcus_](https://www.ncbi.nlm.nih.gov/pubmed/24499426), _Oscillibacter,_ and _[Alistipes](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5682003/)._
**Dysbiosis**
Depression and gut dysbiosis find a [deep connection](https://www.researchgate.net/publication/322937907_Gut_microbiome_and_depression_What_we_know_and_what_we_need_to_know) as the gut and brain share a two-way communication. The reduced richness and assortment of the vast array of the microbes in our gastrointestinal tract also influence our mind frame and [mood swings](https://www.ncbi.nlm.nih.gov/pubmed/27005587). Such correlation has been indicated by a small number of studies where dysbiosis found a relation with abnormalities in behaviour observed in [neurodevelopmental disorders](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6378305/#B25).
**Production of Neurotransmitters**
Apart from digesting and fermenting food and producing vitamins, gut flora also acts as a source of [neurotransmitters](https://www.ncbi.nlm.nih.gov/pubmed/22612585), like, serotonin, gamma-aminobutyric acid (GABA), glutamate, dopamine, and noradrenalin. These metabolites play an essential role in maintaining [gut immunity](https://www.ncbi.nlm.nih.gov/pubmed/25505251) and prevent the [colonization of the gut](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4083503/) by pathogens. Reduction in levels of serotonin, noradrenaline, dopamine and malfunction of the [glutamate-GABA system](https://www.ncbi.nlm.nih.gov/pubmed/23064760), find a close connection with depression and changed gut biology. Moreover, GABA producing, _[Bifidobacterium](https://www.ncbi.nlm.nih.gov/pubmed/27512962),_ also occur in a low number in depression.
On the other hand, environmental factors, the well-being of gut and relative diversity of gut microbes actively modify microbes which regulate [neurotransmitter](https://www.ncbi.nlm.nih.gov/pubmed/29286051) levels. Another neurotransmitter serotonin and tryptophan metabolites if unused, enter the [circulation](https://www.nature.com/articles/mp201277) and influence [behavioural responses](https://www.ncbi.nlm.nih.gov/pubmed/27794467). The [gut microbiota](https://www.ncbi.nlm.nih.gov/pubmed/28053341) also modifies brain function indirectly through changes in inflammatory states and immune status of the gut.
**Carbohydrate metabolism**
Edible fibers found in cereals, fruits, vegetables, nuts, lentils and grains are indigestible. Our gut bacteria convert the long edible fibers to short chain fatty acids (SCFAs). These [SCFAs](https://www.ncbi.nlm.nih.gov/pubmed/24997036) play an essential role in gut biology by acting as an energy source for intestinal cells and the microbes in gut. Interestingly the SCFAs inhibit enzymes which function as immunosuppressive, [anti-inflammatory](https://www.ncbi.nlm.nih.gov/pubmed/25170769), and [anti-depressive agents](https://www.ncbi.nlm.nih.gov/pubmed/26488817). Resultantly, [low amounts of SCFA](https://www.ncbi.nlm.nih.gov/pubmed/27259147) contributes to poorer energy and altered neurotransmission, resulting in depression. The SCFAs produced by the gut-friendly bacteria often pass in the systemic circulation and cross the blood-brain barrier through specific transport and enter into the brain. These SCFAs generate neurotransmitters and offers [neuroprotection](https://www.ncbi.nlm.nih.gov/pubmed/24997036) and acting as natural [antidepressants](https://www.ncbi.nlm.nih.gov/pubmed/25818247).
**Protein and Vitamin metabolism**
Other than carbohydrate metabolism, gut flora also take part in protein and amino acid metabolism through fermentation or putrefaction. However, excessive intake of proteins can result in excessive production of toxic products such as ammonia, putrescine, and phenol, which contribute to [stress and depression](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5682003/). Also, dysbiosis, causing low carbohydrate metabolism and high protein metabolism, find an association with intestinal inflammation leading to [anxiety and depression](https://www.ncbi.nlm.nih.gov/pubmed/30184533).
Gut bacteria like Bifidobacterium, which synthesize vitamins like riboflavin, niacin, and folate, occur in a low number in [depressed individuals](https://www.ncbi.nlm.nih.gov/pubmed/28890155). Low folate and thiamine find a strong association with depression and an inverse association with [symptom severity](https://www.ncbi.nlm.nih.gov/pubmed/20841559).
**Depression and Dysbiosis**
On the other hand, it is reasonable to believe that depression leads to digestive problems and dysbiosis due to unhealthy eating habits or sleep-timings. Decreased ability to digest proteins causes accumulation of residual protein in the colon, aiding the growth of microbes feeding on them. This ultimately leads to elevated putrefaction and gut inflammation.
[Several disorders](https://www.ncbi.nlm.nih.gov/pubmed/25964226) linked with dysbiosis, like, irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), also find a relation with depression. Overpopulation of pathogenic bacteria causes gut inflammation, damages the gut epithelium and compromises intestinal wall permeability. In severe cases, the bacteria or the intestinal epithelial cells cross the blood-brain barrier, ultimately resulting in anxiety, depression, and related [disorders](https://www.ncbi.nlm.nih.gov/pubmed/21237166).
Often activation of the Vagus nerve, controlling digestion, takes responsibility for regulating both dysbiosis and our [mental health](https://www.nature.com/articles/mp201277). Pathogenic microbes in gut flora influence vagal nerve endings distributed in the intestine, causing subsequent pathologic changes in CNS, and propagate depression-like disease symptoms. Additionally, serotonin activates both the vagus nerve and gut motility.
**Management**
As the gut influences the mind, it is rational to emphasize the role of diet in the management of depression. One way of maintaining the integrity of the gut environment involves the intelligent consumption of Polyunsaturated fatty acids (PUFAs). PUFAs share a relationship with both the gut microbiome and our mood. Interestingly, PUFA balance plays an essential role in the sticking of bacteria on [gut mucus membrane](https://www.ncbi.nlm.nih.gov/pubmed/25498862). Omega-6 PUFA produces molecules that maintain gut mucosal permeability and protect against damage from [inflammation](https://www.ncbi.nlm.nih.gov/pubmed/24473752).
_Bifidobacterium,_ capable of degrading PUFAs, are less in number in clinically depressed individuals. Fascinatingly, probiotics containing gut-friendly live microbes, show [antidepressant effects](https://www.ncbi.nlm.nih.gov/pubmed/28483500). _Bifidobacterium longum,_ _Lactobacillus casei,_ and _Lactobacillus rhamnosus_ in probiotics help mend the gut microbial diversity and also elevate anxiety and [depression-like behaviours](https://www.pnas.org/content/108/38/16050). [Faecal microbiota transplant](https://www.ncbi.nlm.nih.gov/pubmed/24762631) is indeed another promising option in correcting depression.
**Taken together**
Depression and gut flora are entwined. So, to have an idea of the real situation of your gut, it is crucial to have a ‘Gut Microbiome Test’ done. Latest ‘DNA sequencing technology’ and algorithm-based data acquisition aids in identifying the individualized microbiome signature in the stool samples. Based on the results, a treatment plan consisting of a blend of diet, lifestyle habits, and probiotics are articulated. The resultant lifestyle can help you lead a depression free life.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Ulcerative colitis: Multiple issues of Dysbiosis
Author: BugSpeaks
Published: 2020-02-20
Category: Microbiome and Disease
Meta Title: Ulcerative colitis: Multiple issues of Dysbiosis
Meta Description: A common understanding of probable disease-causing mechanisms places diseases as genetic, autoimmune, or/and acquired due to the environment. But w
URL: https://www.bugspeaks.com/blog/ulcerative-colitis:-multiple-issues-of-dysbiosis
A common understanding of probable disease-causing mechanisms places diseases as genetic, autoimmune, or/and acquired due to the environment. But what if a condition can affect you through any of the three mechanisms. Well, that would be threatening. Such is the story plot of Ulcerative colitis (UC). A life-long illness, which affects the innermost lining of the lower part of the digestive tract, colon, and rectum. Think of ulcerative colitis as a three-headed dragon. It can exhale upon you the fire of ulcerative colitis through heredity, an autoimmune disorder, or [environmental factor](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6167487/).
You don't get reduced to ashes but indeed suffer from symptoms like diarrhoea, often accompanied by blood or pus, bowel pain and cramping, rectal bleeding, bloody stool, the urgency to defecate. Consequently, you can lose considerable weight. Generally, the symptoms develop over time, and they differ depending on the location of the inflammation. The type and severity of the symptoms vary from person to person. Though rare, at extreme conditions, UC can be debilitating and can sometimes lead to life-threatening [complications](https://www.ncbi.nlm.nih.gov/pubmed/24060951).

**Gut microbiome: your secret power**
Which head of UC dragon gets you depends on how you conduct your life. UC finds close association with the complex ecological system of different candidate microbes residing in our gut. The dominant anaerobic bacteria in the gut form a sort of biofilm over deep-lying tissues. Consequently, they play critical roles in nutrition, permeability, and immune regulation. This group of bacteria, e.g., Bifidobacterium, Bacteroides, and Peptococcus, exist in [symbiotic](https://www.ncbi.nlm.nih.gov/pubmed/23435359) relation with us. Other members of our gut are conditional pathogens, e.g., Enterococcus and Enterobacter.
They maintain harmonious relations with the gut of a healthy individual who has ecologically balanced gut flora. But, if you don't take care, this small group of bacteria can become quite detrimental and can damage the gut. The third category contains mostly pathogens, e.g., Proteus and Pseudomonas. In an ecologically balanced gut, these harmful bacteria stay under check. However, hampering of balance due to any external or internal disturbances accentuates the harmful pathogens. They overpopulate and replace the physiological bacteria and resulting in [disease development](https://www.ncbi.nlm.nih.gov/pubmed/23991417).
**Keep the Ulcerative colitis dragon tamed**
A symbiosis between us and gut microbes promote stability in intestine by inhibiting the colonization of pathogens. Our gut flora under normal circumstances maintains the inner layer of our intestinal tract to keep a relatively low level of inflammation. It helps our gut immune system to continuously screen and eradicate the [detrimental](https://www.ncbi.nlm.nih.gov/pubmed/23435359) microbes.
Moreover, the short-chain fatty acids (SCFAs) released by certain healthy gut bacteria, Faecalibacterium prausnitzii, and Roseburia, have several beneficial effects. They are the primary energy source of intestinal cells and also acts as potent modulators of [immune response](https://www.ncbi.nlm.nih.gov/pubmed/23991417). Additionally, they also suppress tumours and act as neuroendocrine modulators of the intestine. However, the most significant function of SCFAs comprises of control over anti-inflammatory genes involved in [gut immunity](https://www.ncbi.nlm.nih.gov/pubmed/24518984).
Thus, gut dysbiosis, which results in a loss of diversity of intestinal microbiota, influences UC. In UC, dysbiosis, by approximately 25% as compared with that of healthy controls, results in an invasion of pathogenic bacteria in the intestinal mucosa. Some common gut bacteria, Akkermansia muciniphila, and Roseburia lessen whereas detrimental ones, [Mycobacterium avium paratuberculosis](https://www.ncbi.nlm.nih.gov/pubmed/15984976), adherent-invasive [Escherichia coli](https://www.sciencedirect.com/science/article/abs/pii/S0016508506002757), [Clostridium difficile](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3837256/), [Helicobacter species](https://www.ncbi.nlm.nih.gov/pubmed/19438427)11, [Salmonella species](https://www.ncbi.nlm.nih.gov/pubmed/21383845), [Yersinia](https://www.ncbi.nlm.nih.gov/pubmed/15967332), [Fusobacterium](https://www.ncbi.nlm.nih.gov/pubmed/12477765), [Norovirus](https://www.ncbi.nlm.nih.gov/pubmed/24351661), and [Listeria](https://www.ncbi.nlm.nih.gov/pubmed/12737451)16 increase in UC.
These gut damaging bacteria secrete certain toxic elements, which makes the intestinal wall more porous. Furthermore, the enhanced synthesis and secretion of immunosuppressive factors lead to the degeneration of intestinal physiology. All of these ultimately results in immune dysfunction and damaged [intestinal epithelial cells](https://annals.org/aim/article-abstract/718938/new-concepts-pathophysiology-inflammatory-bowel-disease). All this amplifies gut inflammation. So, care for autoimmune reactions and environmental factors by keeping stress at bay and eating healthy fibres is necessary.
**Probiotics: Your knight in shining armour?**
Gut friendly bacteria in the live state are known as probiotics. Relevant candidates of this group are Lactobacillus, Bifidobacterium, and [Enterococcus](https://www.ncbi.nlm.nih.gov/pubmed/19783474). They play several essential functions in gut physiology. Firstly, they compete with pathogens for adhesion sites and nutrients and replaces the harmful ones, while, they correct the intestinal microbial imbalance. Additionally, they maintain and restore complete intestinal mucosal barrier function. Fourthly, they promote immune tolerance by secretion of anti-inflammatory factors of the intestinal mucosa and thus inhibit pathogenesis. Fifthly, they help to mend the intestinal and total immunity of the body. And lastly, they prevent cell death of intestinal mucosa. Thus, probiotics can effectively induce and maintain at-least temporary remission in [UC patients](https://www.ncbi.nlm.nih.gov/pubmed/16819502).
However, some studies associated with probiotics and UC present contradictory results. They have demonstrated that treatment with probiotics amplified bowel movement, resulting in frequent defaecation and diarrhoea like [symptoms](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3901512/#!po=1.21951). Other studies show that probiotics such as Lactobacillus acidophilus LA-5 and Bifidobacterium showed no difference between UC remission patients and the [control group](https://www.ncbi.nlm.nih.gov/pubmed/21453880)20\. Better study design with bigger study group based clinical trials, therefore, remains desirable to establish the exact role of probiotics in UC management.
**Faecal Microbiota Transplant: Helping the damsel in distress**
Faecal microbiota transplantation (FMT) remains another healthy option for slaying UC. The transplant of faecal bacteria from a healthy donor to a recipient, suffering from UC, can quickly resume the normal function and composition of intestinal microbiota. Don't worry. No one will make you eat the poop. In this process, freshly collected faeces are dissolved and homogenized in saline or water, filtered and transplanted within 6 to 8 hours of collection. After FMT, the microbial diversity in the receivers' gut became very much similar to that of [the donors](https://www.ncbi.nlm.nih.gov/pubmed/21871249).
FMT is a simple procedure and can be done with ease. This is also a rather safe mode of treatment. Interestingly, according to some reports, repeated FMT might be better than single transplantation. As, after several years of treatment, there is a chance of microbial reversal. Thus, it is vital to undergo FMT again after several years. However, the number of procedure and gap between two transplantations need to be evaluated depending on the personal diversity in [every case](https://www.ncbi.nlm.nih.gov/pubmed/24060759).
The proportion of beneficial bacteria and diversity increases and thus amending dysbiosis. This blend of beneficial bacteria often contains SFCA producing ones, which improve intestinal ecology and permeability of the mucosal wall — thereby lowering the disease intensity. The proportion of Lachnospiraceae, a butyrate-producing bacterium, gets amplified by FMT. It suggests Lachnospiraceae as the vital bacteria in the success of [FMT](https://www.ncbi.nlm.nih.gov/pubmed/24681177).
**Adverse reactions of gut flora**
Complications of FMT are rare but can be dangerous and life-threatening. The complications include abnormally [low blood pressure](https://www.ncbi.nlm.nih.gov/pubmed/24759832), [norovirus infection](https://www.ncbi.nlm.nih.gov/pubmed/23912408), perforation of the colon, [pneumonia](https://www.ncbi.nlm.nih.gov/pubmed/25982290), [cytomegalovirus](https://www.ncbi.nlm.nih.gov/pubmed/25119613) infection, [bacteraemia](https://www.ncbi.nlm.nih.gov/pubmed/24184170), and others, are just a few among many others. For these reasons, to be on the safe side, long-term follow-up remains essential for patients undergoing FMT.
**Gut microbiome test: A personalized armour against UC**
UC and gut flora are interlinked. To have an idea of the harbouring microbes in our digestive tract, a 'Gut Microbiome Test' is essential. Cutting-edge' DNA sequencing technology' helps to detect the exclusive range of micro-organisms in patient stool samples. Based on this result, a tailor-made, therapy, and diet are prescribed for every individual. Although a complete treatment of UC remains elusive, highly effective management remains a possibility. With proper treatment, the severity of the disease can be managed up to a large extent. Most people with UC attain long-lasting remission and a hassle-free life with appropriate therapy and lifestyle.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Gluten riddles the gut-land
Author: BugSpeaks
Published: 2020-02-13
Category: Nutrition
Meta Title: Gluten riddles the gut-land
Meta Description: You may eat your delicious grains, but should you or should you not, will be revealed how your gut reacts to something k
URL: https://www.bugspeaks.com/blog/gluten-riddles-the-gut-land
You may eat your delicious grains, but should you or should you not, will be revealed how your gut reacts to something key within those grains. What is this key thing you must wonder? Well, it is something that remains common in various food grains and constitutes a major food allergen in the western world. If you have guessed it as gluten, then you are right. But yes, the food you eat and drink, affects your health. Such is the curious case of gluten. For some it’s just an everyday family of proteins that nourish the body, while others suffer from bane of gluten intolerance.
But gluten isn't a single thing, rather it includes a group of hundreds of diverse proteins. So, yes, it is a party of many proteins primarily comprising of, prolamins and glutelins which may or may not cause you trouble. Gluten occurs with starch and accounts for 75–85% of the total wheat protein. It also occurs in many other grains like barley, rye, spelt, oats and triticale and the products derived from them, like, bread and malts. Gluten acts like glue which makes the food chewy, give it elasticity, and helps food-grains to retain their shape.

#### **Gluten in gut-land**
Despite all the delicious gooiness which gluten imparts to food, it can cause some adverse immunological and autoimmune reactions in certain susceptible people. Such a reaction makes up gluten intolerance, which refers to three types of conditions. These [include](https://www.ncbi.nlm.nih.gov/pubmed/27763541) gluten allergy, Celiac disease, and Non-celiac gluten sensitivity. Differentiating between these can rattle one's brains as symptoms may resemble each other. It is essentially a matter of how your gut behaves to the glutens which have dropped down the oesophagus.
A gluten allergy caused due to the consumption of gluten, wheat, or related food products may cause symptoms like mild skin rash, respiratory or gastrointestinal reactions or severe life-threatening symptoms like breathing disability followed by anaphylactic shock. Yet, the actual mechanism causing gluten allergies remains a riddle to be solved.
Different from an allergic reaction, when gluten triggers an autoimmune reaction outcome the [Celiac disease](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6221985/). In this case, antibodies raised against gluten damages the small finger-like projections in the small intestine called villi. Symptoms include abdominal pain, constipation, stomach pain, vomiting, bloating, nausea, and chronic diarrhoea.
Another potential form of gluten intolerance, [Non-celiac gluten sensitivity](https://celiac.org/about-celiac-disease/related-conditions/non-celiac-wheat-gluten-sensitivity/) (NCGS) is neither true gluten allergy nor exact Celiac Disease. It finds association with symptoms in the gut, brain, muscles, skin or elsewhere in response to the consumption of gluten or gluten-containing food products. However, the villi in small intestines remain healthy. The symptoms generally disappear on a gluten-free diet.
It is interesting to note that all the conditions find association with gut microbial dysbiosis.
**What makes gluten confuse the gut?**
Digestion of gluten with our own enzymes in the intestine produces immunogenic polypeptides, which stimulate immunologic cells in our body. These immune-toxic peptides are resistant to further breakdown. Bacteria present in small intestine participate in degradation and metabolism of gluten, which is otherwise is tough to [digest](https://www.ncbi.nlm.nih.gov/pubmed/23244345). The friendly gut bacteria like, Lactobacillus helveticus, Firmicutes and Actinobacteria reduce gluten’s immunogenicity. They produce enzymes that effectively cleave immunogenic polypeptides produced during the initial degradation of [gluten](https://www.ncbi.nlm.nih.gov/pubmed/28526528). But when such friendly microbes are not present as required the immunogenic polypeptides go mad and cause the conditions of gluten intolerance.
Experts think dysbiosis as the root cause of gluten intolerance, characterized by an imbalance between beneficial and detrimental bacteria. It has been observed that gluten allergy is associated with reduced numbers of Firmicutes and Bifidobacterium, leading to low levels of [butyrate](https://ngmedicine.com/scfas-part-2-the-benefits-of-butyrate/) in the gut and increased intestinal permeability. Proteolytic enzymes produced by harmful bacteria like Pseudomonas aeruginosa, Bacteroides fragilis and Proteobacteria generate longer peptide products from gluten. These peptides activate [immunologic-cells](https://www.ncbi.nlm.nih.gov/pubmed/23244345). Whereas digestive enzymes produced by beneficial bacteria, Lactobacillus produced non-immunogenic smaller peptides from gluten. Several factors like early-life antibiotic treatment can lead to dysbiosis with an expansion of unfriendly [Proteobacteria.](https://www.ncbi.nlm.nih.gov/pubmed/23244345)
**Should the gut-land be devoid of gluten?**
As gluten is the culprit behind allergies and other conditions, having a gluten-free diet is the easiest solution to reduce the pro-inflammatory signals via the renewal of gut-microbiome. This offers great relief with improvement in digestion, reduction in digestive tract associated problems, and other symptoms related to [gluten allergy](https://www.ncbi.nlm.nih.gov/pubmed/27373514). Ultimately, this gluten-free lifestyle has an additional benefit, it can help to lose weight. In a gluten-free diet, the healthier options, like quinoa, which doesn't contain gluten replace starches and polysaccharides.
However, the gains come at the cost of losses, as the members of gut-land may suffer when starch intake reduces. As in natural conditions, gluten occurs with starch, a gluten deficient diet significantly brings down the starch intake. It is notable to find that, a switch to a diet that is low or free from gluten, also [depletes](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3023594/) the comparative abundance of beneficial butyrate-producing, Clostridiales and [Lachnospiraceae.](https://www.ncbi.nlm.nih.gov/pubmed/25403367) Why? Because one simply takes away what these bacteria feed on. Moreover, this may also provide the opportunistic pathogens such as [Escherichia coli](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3023594/) to proliferate and cause further problems.
**Answer lies in Pre- and Pro-biotics**
As gluten allergy finds close association with dysbiosis, it is of worth to use pre-biotics or pro-biotics to overpopulate and diversify the gut microbiome. It helps reshape the bacterial community in a more beneficial mode. Such transformed gut flora would help improve gluten tolerance and reduce associated inflammation. Certain healthy gut bacteria like Bifidobacterium longum, Bifidobacterium animalis, and Lactobacillus rhamnosus have been found to diminish the destructive effects of gluten. Prebiotics, like, food loaded with fibre and green leafy vegetables can efficiently replace the pathogens with beneficial ones. Probiotics also help to diminish inflammation and address the fundamental reasons for [gluten allergy](https://www.ncbi.nlm.nih.gov/pubmed/25403367).
**Coming out of gluten allergy**
A jump out of the dysbiosis of the gut, back to the health is what your gut-land needs. Currently, a complete cure for gluten allergy is not possible and one might have to endure it for long. But there is nothing to lose hope. As gluten allergy and gut microbiome are interconnected, so, to keep it under control, it is crucial to regulate the gut microbiome. Before regulation in an effective manner, testing proves essential, hence the first step should entail a ‘Gut Microbiome Test’.
The test uses cutting-edge ‘DNA sequencing technology’ to identify the unique collection of gut flora harbouring in patient stool samples. Then a person-specific tailor-made diet plan is formulated based on the unique gut microbial signature. The signs and symptoms can be kept under control by religiously following a custom well-designed therapeutic plan. Like all other diet-based therapies, it often takes quite some time to get everything resolved. So, patience is the key.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Irritable gut - Irritable you
Author: BugSpeaks
Published: 2020-02-06
Category: Microbiome and Disease
Meta Title: Irritable gut - Irritable you
Meta Description: Like us, our bowel can get irritated too! In one-word, i
URL: https://www.bugspeaks.com/blog/irritable-gut---irritable-you
**Like us, our bowel can get irritated too!**
In one-word, irritation in our gut may indicate ‘Irritable bowel syndrome’ (IBS). Often associated with chronic, recurrent abdominal discomfort and pain, IBS gets worse after meals and resolves upon defecation. Symptoms include changes in bowel habits, diarrhoea, constipation, or alternating constipation and digestive symptoms, such as dyspepsia, dysphagia, and [nausea](https://www.ncbi.nlm.nih.gov/pubmed/27144627).
Sometimes, IBS finds a link with non-gastrointestinal disorders such as chronic pelvic pain, fibromyalgia, chronic fatigue syndrome, anxiety, and even depression. If unaddressed, these comorbidities may hamper your [day-to-day life](https://jamanetwork.com/journals/jama/article-abstract/2174034). A flare-up may last for several days, and then symptoms either improve or resolve completely. Don’t worry. Severe complications are rare in reality. Also, many such comorbidities often find an association with gut dysbiosis.
**Why??**
A complex assemblage of trillions of micro-organisms including fungi, eukaryotes, bacteria, viruses, and archaea live in our gut. These dynamic and vibrant microbial communities comprise of pro- and anti-inflammatory bacteria and maintain gastrointestinal [homeostasis](https://www.ncbi.nlm.nih.gov/pubmed/24665829). This homeostasis enables microbes to colonize the intestine and perform symbiotic functions persistently. Thus, gut microbes are the critical players in IBS [pathogenesis](https://www.nature.com/articles/nature25973).
**How??**
Dysbiosis, typically associated with IBS, occurs at a frequency of [73%](https://www.ncbi.nlm.nih.gov/pubmed/27444134). Pre-dominant bacteria contributing to dysbiosis include Bacillus and Ruminococcus gnavus, Shigella or Escherichia, and [Actinobacteria](https://www.ncbi.nlm.nih.gov/pubmed/27444134). Frequently, in IBS, commensals turn pathogenic through the gain of virulence. For example, Enterococcus faecalis turns to non-invasive E. coli, evading the immune system in disease pathogenesis. All this evidence indicates a strong relation between dysbiosis and the severity of [IBS](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4645401/).

**A little off-balance, a lot of imbalance**
Certain gut bacteria ferment polysaccharides to produce SCFAs and gasses such as hydrogen (H2) and methane (CH4). Thus, they influence bowel movement and permeability of the intestinal barrier. However, a breach may occur in the intestinal wall due to the influx of inflammatory mediators or pathogens. Consequently, severe inflammation occurs, affecting the gut milieu with alteration of [gut flora](https://www.ncbi.nlm.nih.gov/pubmed/18541210). As expected, a lower abundance of SCFA producing bacteria occurs in IBS, which also maintains lower counts of [methanogens](https://www.ncbi.nlm.nih.gov/pubmed/24751910). Accordingly, microbial metabolism disrupts, resulting in a high production of hydrogen gas, causing flatulence in the [abdomen](https://www.ncbi.nlm.nih.gov/pubmed/20680169).
Moreover, IBS subjects relative to healthy controls present overpopulation of Proteobacteria, Veillonella, Firmicutes, Lactobacillus, and [Ruminococcus](https://www.ncbi.nlm.nih.gov/pubmed/22986438). They show a decreased quantity of Bifidobacterium, Faecalibacterium, Erysipelotrichaceae, and [Methanogens](https://www.nature.com/articles/srep12693). Moreover, the severity of IBS finds a positive correlation with a specific intestinal microbiota signature, characterized by low microbial richness, Methanobacteriales absence, and enriched [Bacteroides](https://www.ncbi.nlm.nih.gov/pubmed/24751910).
**Psychological Association**
A stressful mind stresses your gut, so remains the case with IBS. Considered a stress-sensitive disorder, psychological factors like any abuse, adverse life event, and post-traumatic stress disorder can elicit IBS. How? These conditions are likely to be involved in gut-brain interaction and IBS [pathogenesis](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4202343/). IBS and gut dysbiosis find a link with the psychological wellbeing of an individual. A prominent example relates to depression resulting from the activation of colon immunity and the release of certain inflammatory [factors](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5825947/). Another common issue of high stress leads to altered metabolism of serotonin, a neurotransmitter that is responsible for both variations of gut motility and pathogenesis of [IBS](https://theromefoundation.org/wp-content/uploads/functional-gastrointestinal-disorders-history-pathophysiology-clinical-features-and-rome-iv.pdf).
**Genetic predisposition**
Specific genetic aberrations like a mutation in the [SC5NA gene](https://www.ncbi.nlm.nih.gov/pubmed/24613995) responsible for immune regulation and epithelial barrier function relate to IBS development. Additionally, epigenetic factors also influence [IBS occurrence](https://www.ncbi.nlm.nih.gov/pubmed/26670691).
**Therapeutic Options**
Probiotics are one of the essential therapeutic options. They play a role in modulating gut inflammation and produce antimicrobial peptides. These products help eliminate pathogenic bacteria and improve the mucosal barrier function easing out the symptoms of IBS. Consistent use of probiotics, the Biofidobacterium or [Lactobacillus](https://www.ncbi.nlm.nih.gov/pubmed/22730468), or a blend of bacteria consisting of Bifidobacterium, Lactobacillus, and Streptococcus help improve the symptoms of [IBS](https://www.ncbi.nlm.nih.gov/pubmed/24979556). However, due to a short lifespan, repeated doses of prebiotics are required. Therefore, prebiotics may serve as a better alternative treatment as they provide substrates that can help in the growth of specific bacteria and hence can alter the [microbiota](https://www.ncbi.nlm.nih.gov/pubmed/18685504).
Prebiotics present multipronged benefits. For example, the prebiotic lactulose promotes gut bacteria, water retention in stool, exhibiting laxative effects. Other synthetic prebiotics includes various kinds of oligosaccharides like fructo-oligosaccharides, soybean oligosaccharides, galacto-oligosaccharides, isomalto-oligosaccharides, xylo-oligosaccharides, and others. Accordingly, cereals, fruits, and vegetables are rich in prebiotics. Commensal bacteria in the colon ferment prebiotics to produce SCFAs, which help regulate inflammatory responses, and influence gut homeostasis. Resultantly, dysbiosis gets corrected through the promotion of positive alterations in the gut [microflora](https://www.ncbi.nlm.nih.gov/pubmed/29500265). A step ahead of probiotics and prebiotics, synbiotics, a combination of the two, show higher potency compared to either of [the two](https://www.ncbi.nlm.nih.gov/pubmed/28936910).
**A step ahead pro- and prebiotics**
Another advancement over probiotics includes paraprobiotic and postbiotic. Paraprobiotics are non-viable or inactivated probiotics, while postbiotics are non-viable soluble factors secreted by live bacteria or released upon [cell lysis](https://www.ncbi.nlm.nih.gov/pubmed/22301383). They benefit the host through their [biological activity](https://www.ncbi.nlm.nih.gov/pubmed/23990841). The advantage of postbiotics and paraprobiotic lies in their safety profile and longer shelf-life while conferring health benefits comparable to those of [probiotics](https://www.ncbi.nlm.nih.gov/pubmed/24336217). Moreover, some poorly/non-absorbed antibiotics with low toxicities, such as rifaximin and neomycin, also prove useful for the treatment of [IBS](https://www.ncbi.nlm.nih.gov/pubmed/23556126). These antibiotics cause remarkable improvements in some instances of IBS known as small intestinal bacterial [overgrowth](https://www.ncbi.nlm.nih.gov/pubmed/27402085).
**Dietary Influence**
Dietary interventions are helpful can help IBS patients as, in most cases, the consumption of certain foods causes aggravation of [symptoms](https://www.ncbi.nlm.nih.gov/pubmed/26109797). In terms of dietary influences, the main culprits are gluten and short-chain carbohydrates that trigger harmful alterations. A diet that corrects the microbiota dysbiosis could thus help manage IBS [symptoms](https://www.ncbi.nlm.nih.gov/pubmed/26311042). However, meagre consumption of short-chain carbohydrates may reduce the abundance of healthy gut commensal bacteria. Therefore, careful modification or restriction of dietary intake in IBS patients only proves helpful. But, to achieve an effective diet, an individual may need to go through a process of trial and error as IBS has no [universal diet](https://www.sciencedirect.com/science/article/abs/pii/S0924224417302765).
**Faecal Microbiota Transplant (FMT)**
Interestingly, some other person's excreta can also provide relief from IBS. Although it sounds a bit wicked, the poo treatment, known as Faecal Microbiota Transplant (FMT), can help. FMT involves applying a solution of faecal material from a healthy donor into the gut of a receiver, intending to restore the aberrant microbial [composition](https://www.ncbi.nlm.nih.gov/pubmed/23659729). The technique aims to restore the dysbiosis in the gut.
**Finally,**
As science continues to disentangle the mysteries of the gut microbiome, the mechanisms to manipulate the gut microbiome for better health continue to widen. With the help of a ‘Gut Microbiome Test,’ it is very easy to obtain an individualized microbiome profile. Through DNA sequencing technologies, experts can now devise personalized tailor-made treatment diet plans, probiotic plans as per the need of the individual patients. There is no cure for IBS. However, as IBS is a chronic condition, so you need to manage it on a long-term basis. It can be kept under control by following the doctor’s advice religiously, maintaining diet, lifestyle, and stress level.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Inflammatory bowel disease: Battle of microbes
Author: BugSpeaks
Published: 2020-01-30
Category: Microbiome and Disease
Meta Title: Inflammatory bowel disease: Battle of microbes
Meta Description: Here comes the superhero to save the day!! Wait, it seems the superhero is suffering from abdominal pain and cramping. Yet the sup
URL: https://www.bugspeaks.com/blog/inflammatory-bowel-disease:-battle-of-microbes
Here comes the superhero to save the day!! Wait, it seems the superhero is suffering from abdominal pain and cramping. Yet the superhero while pushing through the day, gets the work done. Did you guess the superhero?? No, it is not superman nor spiderman; it is anyone who suffers from Inflammatory bowel disease (IBD). IBD involves chronic inflammation of gut mediated by the immune system and includes Crohn's disease (CD) and ulcerative colitis (UC). A person who suffers from IBD is no less than a superhero pushing through the pain to get the life going.
IBD involves the transformation of cells of the inner lining of the gut causing simultaneous destruction and repair of the adjacent tissue. It operates in a person-specific manner in terms of location and behaviour of the disease. So just every superhero has their own super abilities, an individual with IBD presents an alternating pattern of intensification and reduction of the symptoms. Symptoms include abdominal pain, cramping, mild to severe diarrhoea, fever, low appetite, fatigue, and unintended [weight loss](https://www.ncbi.nlm.nih.gov/pubmed/22001864). A severe form of the disease can even lead to life-threatening complications.
Though the exact cause remains undiscovered, both genetic predisposition and immune system malfunction may influence IBD. Environmental factors such as diet, lifestyle, antibiotic use, and socioeconomic development exaggerate the [disease](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6167487/).

**Looking for the IBD villain**
As IBD involves inflammation of the gut mucosa, it is logical to associate the disease with the dynamic and diverse range of microbiome harbouring in there. The helpful gut bacteria inhibit mucosal inflammation and thus act as a shield against [IBD](https://www.ncbi.nlm.nih.gov/pubmed/23013615). The diversity of beneficial gut microbes declines during the diseased conditions and the equilibrium between commensal and potential pathogen shifts. This villainous takeover of harmful microbes in the gut thus relates to dysbiosis. Moreover, due to the person-specific diversity of gut microbiomes, significant differences occur in equilibrium shifts between [different individuals](https://www.ncbi.nlm.nih.gov/pubmed/28191884). Beneficial bacteria such as _Firmicutes_ and _Bacteroides_ decline_._ Pathogenic _Gammaproteobacteria, Veillonellaceae, Pasteurellacaea, Enterobacteriaceae_, _Fusobacteriaceae_, and adherent-invasive strains of _E. coli_ populate the gut.
**Villainous scheme of things in IBD**
Amongst the array of microbes affected in IBD comes a set of specific bacteria that ferment indigestible fibres to produce short-chain fatty acids (SCFA). The SCFA producing bacteria, _Faecalibacterium_, _Leuconostocaceae_, _Phascolarctobacterium,_ and _Roseburia_ suffer depletion in the gut. As epithelial cells of colon feed on the energy produced by the [SFCA](https://www.ncbi.nlm.nih.gov/pubmed/23828891), such decline further disrupts the gut energy [metabolism](https://www.ncbi.nlm.nih.gov/pubmed/23013615).
Other important function of gut microbiota includes synthesis of some vitamins, enzyme secretion, retention of intestinal mucosal integrity, and educating the gut immune system to prevent the pathogenic attack. Also, they take care of the host by a phenomenon ‘called colonization resistance,’ where they occupy host niches leaving no empty spaces for [invading pathogens](https://www.ncbi.nlm.nih.gov/pubmed/22869189).
The gut of healthy individuals also harbours certain beneficial fungus-like, _Saccharomyces, Candida_, and [_Cladosporium_](https://www.ncbi.nlm.nih.gov/pubmed/23799070) and helpful viruses like bacteriophages, _Caudovirales_, and [_Microviridae_](https://www.ncbi.nlm.nih.gov/pubmed/24621522). However, the abundance of some harmful fungi like _Basidiomycota, and Ascomycota_ elevates significantly in [IBD patients](https://www.ncbi.nlm.nih.gov/pubmed/18936492). Another candidate belonging to the healthy gut microbiome is helminths, playing an immune-regulatory role in the gut. Their absence also finds a link with [IBD development](https://www.ncbi.nlm.nih.gov/pubmed/27080105).
**What can help the superhero fight IBD villains?**
Usual therapies for IBD are [corticosteroids](https://www.ncbi.nlm.nih.gov/pubmed/16236742), [amino-salicylates](https://www.ncbi.nlm.nih.gov/pubmed/15909110), and [immunosuppressive agents](https://www.ncbi.nlm.nih.gov/pubmed/21337668). This symptomatic treatment acts as a quick fix and helps to get rid of complications. Such treatment also comes with adverse side effects, including loss of immune tolerance and [drug resistance](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3084301/). Thus, an alternative effective therapeutic strategy remains the need for the hour. The possibility that gut microbes drive inflammation in IBD inspires several clinical approaches aimed at correcting dysbiosis. These include dietary or microbial interventions like the use of probiotics, antibiotics, defined enteral nutritional therapy (ENT), and faecal microbiota transplantation (FMT).
**Probiotics:**
Probiotics are a combination of bacteria or yeasts with apparent beneficial health effects that serve to restore [gut microbial balance](https://www.ncbi.nlm.nih.gov/pubmed/23474420). A cocktail of eight live freeze-dried bacterial species has shown to reduce active inflammation and sustain remission in UC, [but not CD](https://www.ncbi.nlm.nih.gov/pubmed/18240278). Friendly bacteria, _Faecalibacterium,_ has shown a protective effect on the intestine by producing SFCA, responsible for barrier-enhancement and immunosuppression. Specific probiotic interventions have shown promise; however, the quest to link clinical improvements of IBD to probiotic-induced microbiota changes continues. Also, fundamental questions regarding the optimal composition of probiotics, timing of administration and durability of the response remain [unanswered](https://www.ncbi.nlm.nih.gov/pubmed/23474420).
**Synbiotics:**
Synbiotics, a combination of pro- and prebiotics, exert a beneficial effect on host health through working together of two formulations. Pro- and prebiotics in combination, present a novel approach with promising opportunities to evaluate their efficacy and potential [use in IBD](https://www.ncbi.nlm.nih.gov/pubmed/23040451).
**Diet:**
The diet of an individual exerts a significant effect on the composition of their gut microbiota by altering gut functionality and metabolism at the [genomic level](https://www.ncbi.nlm.nih.gov/pubmed/24503132). One treatment called ENT utilizes the potential of dietary modification. When food is replaced with a nutritionally complete formula in ENT, it can act as a first and foremost line of therapy IBD remission. This treatment results in both clinical improvement and mucosal healing. The idea of providing a fibre-rich diet that can selectively increase the abundance of SCFA-producing microbes remains attractive. Yet, satisfactory outcomes have not been [confirmed](https://www.ncbi.nlm.nih.gov/pubmed/27769810).
**Faecal microbiota transplantation:**
Antibiotics can ease inflammation, and people often pop them frequently. However, non-specific targeting of gut microbiota with broad-spectrum antibiotics could deplete both beneficial and pathogenic microorganisms. Well, punishing the innocent microbes often comes at its cost, often seriously compromising health. In such a situation, Faecal microbiota transplantation (FMT), or a stool transplant can help. Also helpful with dysbiosis sans antibiotic intake FMT includes transplantation of faecal bacteria from a healthy individual into a recipient. It helps restore the gut microbial homeostasis posing a potential strategy to correct dysbiosis in [IBD](https://www.ncbi.nlm.nih.gov/pubmed/27329806).
Overall, data regarding FMT remains scarce. A few points like the safety and durability of this approach, the most effective mode of administration, particularly in immunosuppressed patients, and how to select appropriate donors and recipients should be taken care of. More extensive randomized controlled trials are necessary for a better understanding of the process and to better define the role of FMT in the treatment of IBD. In the future, FMT will be probably be substituted by the use of a defined array of [micro-organisms](https://www.ncbi.nlm.nih.gov/pubmed/23842501).
**Winning the battle against IBD**
IBD and gut microbiome are closely associated with each other. So, to know the actual scenario of your gut, it is essential to take a ‘Gut Microbiome Test.’ Modern age ‘DNA sequencing technology’ and algorithm-based data acquisition helps to identify the person-specific array of micro-organisms in patient stool samples are. A therapeutic plan consisting of combinations of prebiotics and probiotics are formulated based on the unique microbial population residing in the gut. IBD is not curable. But there is nothing to lose confidence. The one and only way are to manage the signs and symptoms intelligently. Long-term remission is quite possible, with proper guidance and expert advice.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Crohn's disease: No reason to be Hopeless
Author: BugSpeaks
Published: 2020-01-23
Category: Microbiome and Disease
Meta Title: Crohn's disease: No reason to be Hopeless
Meta Description: Our gut harbours diverse flora working like an organ system made up of bacteria, viruses, fungi, and several other microbes. They
URL: https://www.bugspeaks.com/blog/crohn's-disease:-no-reason-to-be-hopeless
Our gut harbours diverse flora working like an organ system made up of bacteria, viruses, fungi, and several other microbes. They perform a variety of functions, such as training our [immune system](https://www.ncbi.nlm.nih.gov/pubmed/19343057) and suppressing harmful [microorganisms](https://www.ncbi.nlm.nih.gov/pubmed/23748339). But when the balance in the delicate microbial world tips off, dysbiosis occurs, paving the way to an array of health conditions. Crohn’s disease is one such condition wherein the impairment of [gut microbiota](https://www.ncbi.nlm.nih.gov/pubmed/28191884) causes a whole lot of pain to an individual. Down goes the number of healthy bacteria, like, _Firmicutes_ and _Bacteroides_, and pathogenic bacteria, _Gammaproteobacteria, Bacteroidales, Erysipelotrichales_ overpopulate the [gut](https://www.ncbi.nlm.nih.gov/pubmed/23013615).
Crohn’s disease includes long term inflammation in bowel with intermittent aggravation and remission. This illness affects different segments of the digestive tract in different people depending on the severity and activity of the disease. Patients suffer from pain in the abdomen, severe diarrhoea, mouth sores, tiredness, malnourishment, and excessive weight loss. Crohn’s disease may either be a fault of inherited genes or the immune system. The exact causation remains unknown. Diet rich in spices, processed food, and stressful lifestyle often worsens the [situation](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6167487/). Quite often, a deeper layer of the gut tissue gets involved, which can pose a [threat to life](https://www.ncbi.nlm.nih.gov/pubmed/26627550).

**The relation between Crohn's and gut microbiome**
Some friendly members of gut flora like _Faecalibacterium_, _Phascolarctobacterium,_ and _Roseburia_ ferment dietary fibre to produce short-chain fatty acids (SCFAs). SCFAs like butyrate, propionate, and acetate provide energy and immunity to the [colon](https://www.ncbi.nlm.nih.gov/pubmed/18839424) [epithelium](https://www.ncbi.nlm.nih.gov/pubmed/23842501). Sadly, SCFA producing bacteria decline in the [diseased gut](https://www.ncbi.nlm.nih.gov/pubmed/23842501). Moreover, _Lactobacillus_ and _Bifidobacterium_ populations, which promote protection against gut mucosal inflammation, [also decline](https://www.ncbi.nlm.nih.gov/pubmed/23013615). Like bacteria, numbers of beneficial fungi also fluctuate.
For example, _Dioszegia_ and _Candida glabrata_ assume predominance, whereas _Trichosporon_ and _Leptosphaeria_ become a [minority](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4957473/). Once imbalanced, the disturbed microbial diversity further alters the gut environment even during the remission period. For example, the neutral and acidic mucin - a key component of mucus in the gut membrane - secretion remains high during the [remission period](https://www.nature.com/articles/s41598-019-49893-5). It is due to a reduction in bacteria of genera _Oscollospira_ and _Akkermansia_, sulphate-reducing bacteria and _Saccharomyces cerevisiae_ and does not allow relief from inflammation.
**Pain with no treatment?**
Treating Crohn's diseases is as easy as soft-landing a rover on the moon's lunar surface. No one has done it, yet the efforts and research continue. Antibiotics, your mate in fighting most of the diseases, proves inadequate in Crohn’s disease. The broad-spectrum nontargeted microbiome-destroying approaches do not serve the [purpose](https://www.ncbi.nlm.nih.gov/pubmed/27769810). Increasing knowledge into the promising role of gut microbiota in the plethora of conditions now poses a hopeful avenue for Crohn's patients. The intriguing role of gut microbiota in host immune response ways may hold the key to their function in controlling gut immunity and thus health restoration.
**Manipulation of gut microbes: A hope for pain**
Aimed at manipulation of the gut microbial population efforts to harness prebiotics, probiotics, and postbiotics to harmonize the gut habitat in Crohn’s disease continues. Prebiotics promote the growth and colonization of gut-friendly organisms. fibre remains as one of the oldest and most frequently used prebiotics. Some beneficial organisms have the unique ability to ferment indigestible fibre to produce SCFA critical to a healthy colon. So, logically, a fibre-rich diet would lower the risk of Crohn’s disease and would have been a line of management in affected people. However, enough evidence lacks this support. Further studies for a better understanding and utilization of fibres in Crohn's disease management [remain warranted](https://www.ncbi.nlm.nih.gov/pubmed/21468064).
Knowing the association of Crohn’s disease with dysbiosis, the administration of probiotic strains to help stabilize the gut microbiome seems rational. It may also help redesign the gut habitat in a more advantageous way. This holds the reasonable potential to improve the disease condition. Thus, therapies to restructure microbial communities could be used for improved management of Crohn’s disease. However, more data can help establish the role of probiotic administration as a [therapy](https://www.ncbi.nlm.nih.gov/pubmed/28838410). Also, the negligible clinical achievement with probiotics shows that single or multiple non-target probiotic strain administration may not be helpful. To sustain this treatment, the cocktail of targeted micro-organisms should [be used](https://www.ncbi.nlm.nih.gov/pubmed/28005973).
**One man's waste another man's treasure**
Another strategy includes the use of postbiotics, the bioactive molecules produced by a microbial community, which maintain the health of the gastrointestinal tract. With the latest cutting-edge technology, identification of vital metabolic pathways and bioactive postbiotics is possible. Administration of postbiotics to patients with Crohn’s disease may help re-establish the full functioning [gut microbial community](https://www.ncbi.nlm.nih.gov/pubmed/28005973). Faecal microbiota transplantation (FMT) is another possible choice of therapy in Crohn’s disease and other diseases with altered microbial compositions.
Crohn’s disease has complicated pathogenesis involving host genetics and dysregulation of the host-microbiome crosstalk. The aim of FMT in Crohn’s disease is to cure dysbiosis and re-establish healthy communication between the host immune system and the microbiota. However, due to the complication of the underlying genetic predisposition of the host, success remains limited [in this case](https://www.ncbi.nlm.nih.gov/pubmed/26627550). However, the long-term consequence of FMT as a strategy to treat Crohn’s disease is still not clear. In few years down the lane, as a better approach, FMT will inevitably be replaced with a well-defined blend of [micro-organisms](https://www.ncbi.nlm.nih.gov/pubmed/23842501).
**Gut microbiome testing: A personal plan to eliminate Chron's disease**
The perturbation or the disturbance of gut flora in Crohn’s disease presents complexity and the mechanisms not yet fully understood. Moreover, it has not been possible to dissect out the dysbiotic changes that, as a consequence, may lead to mucosal inflammation. Severe and chronic inflammation of Crohn’s disease alters intestinal flora. This modification, in turn, leads to atypical host-microbiome interactions, ultimately exacerbating the primary disease. Possibly, through regulation of inflammation or control over the host-microbiome interaction may help disrupt the vicious circle of inflammation and [dysbiosis](https://www.ncbi.nlm.nih.gov/pubmed/26627550).
As Crohn’s disease and gut microbiome are entangled, so, to get resolved, it is a prerequisite to taking a ‘Gut Microbiome Test.’ With the help of the latest ‘DNA sequencing technology,’ the exclusive array of micro-organisms in patient stool samples are identified. Then the diet/ therapeutic plan is processed based on the unique microbial population harbouring in the gut. The complete cure of Crohn's disease is so far, not known. But there is hope. Like all chronic illnesses, the only way is to reduce the signs and symptoms significantly. Even long-term remission may be attained through the help of one single therapy or multiple therapies simultaneously or separately. With proper treatment, many people with Crohn's disease can function well and lead an active life.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## C. difficile infection: Managed through Gut Microflora Manipulation
Author: BugSpeaks
Published: 2020-01-16
Category: Microbiome and Disease
Meta Title: C. difficile infection: Managed through Gut Microflora Manipulation
Meta Description: Science now proves the existence of a diverse array of micro-organisms in our gastrointestinal (GI) tract. This variation of gut f
URL: https://www.bugspeaks.com/blog/c.-difficile-infection:-managed-through-gut-microflora-manipulation
Science now proves the existence of a diverse array of micro-organisms in our gastrointestinal (GI) tract. This variation of gut flora increases through the GI tract from the small intestine to end of the colon. Some bacteria like _Firmicutes_ are predominant in the terminal ileum along with _Streptococcus, Veilonella,_ and _Clostridium_. The colon is highly populated with _Bacteroidetes_ (genus _Bacteroides_) and _Firmicutes_ (genus _Clostridium,_ _Faecalibacterium, Lactobacillus, Ruminococcus_). While only a few _Actinobacteria_ and _Proteobacteria_ reside in the colon. Additionally, some notorious pathogens, like, _Salmonella enterica,_ also comprise 0.1% of total [gut microbiota](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4181834/).
_Clostridium difficile_ also belongs to the class of notorious enteric pathogens and resides in the gut of 1-3% of the [healthy population](https://www.ncbi.nlm.nih.gov/pubmed/26038491)_. C._ _difficile_ a spore-forming, gram-positive, and rod-shaped bacterium belongs to the class of enteric pathogens. Through inflammation of the colon, it can cause mild diarrhoea to possibly fatal pseudomembranous colitis and toxic megacolon [disease](https://www.ncbi.nlm.nih.gov/pubmed/24118601). _C._ _difficile_ infection is [prevalent](https://www.ncbi.nlm.nih.gov/pubmed/21762504) in hospitals but may be acquired from the [community](https://www.ncbi.nlm.nih.gov/pubmed/22536816) too. Being an anaerobe, the vegetative form of bacteria hates environments with oxygen. However, bacterial spores survive in the presence of oxygen, harsh temperatures, acidic environment in the [stomach](https://books.google.co.in/books?id=8pKqDwAAQBAJ&printsec=frontcover&dq=Mandell,+Douglas,+and+Bennett%27s+principles+and+practice+of+infectious+diseases.&hl=en&sa=X&ved=0ahUKEwiWrOXAl4bnAhVOWysKHQYhAJQQ6AEIQTAD#v=snippet&q=Clostridium%20difficile%20infection&f=true), and alcohol-containing hand [sanitizers](https://www.ncbi.nlm.nih.gov/pubmed/19715426) as well!

**Journey of** _**C. difficile in the gut**_
These robust _C. difficile_ spores found in soil, faeces, sewage, and other sources thus facilitate disease transmission. _C. difficile_ follows the faecal-oral route of transmission. It spreads either through contact with patients, contaminated objects, or due to [poor hygiene](https://www.ncbi.nlm.nih.gov/pubmed/24814671). Once the spores enter through the mouth, they survive and pass through the stomach to ultimately reach the small intestine. Bile acids and well-known amino acid, glycine, in the small intestine trigger the germination process, leading to [vegetative form](https://www.ncbi.nlm.nih.gov/pubmed/18245298). The vegetative form upon reaching the large intestine flourishes in an oxygen-deficient [environment](https://www.ncbi.nlm.nih.gov/pubmed/25483328). _C difficile_ colonizes on the epithelium of colon and secrete [toxins A and B](https://www.ncbi.nlm.nih.gov/pubmed/17693508). These toxins induce inflammation of the mucosal layer, destroy the epithelial layer and the [intestinal barrier](https://www.ncbi.nlm.nih.gov/pubmed/22237401).
**Curious case of** _**C. difficile**_ **and antibiotics**
The use of antibiotics also increases the risk of _C. difficile_ [infection](https://www.ncbi.nlm.nih.gov/pubmed/23620467), varying with the type of antibiotic. _C. difficile_ [infection](https://www.ncbi.nlm.nih.gov/pubmed/23478961) finds no association with tetracyclines, while penicillin poses intermediate risk. The fluoroquinolones, third generation cephalosporins, and clindamycin, on the other hand, pose an elevated risk. Using clindamycin poses the maximum risk of C. difficile [infection](https://www.ncbi.nlm.nih.gov/pubmed/24324224). Taking antibiotics even for a few days disturbs the gut flora. While frequent uptake can lead to a profound and [long-lasting damage](https://www.ncbi.nlm.nih.gov/pubmed/18043614). Known as “post-antibiotic dysbiosis”, such impairment includes loss of diversity and change in the metabolism of the microbiota.
This altered microbiota exhibits reduced colonization resistance against invading [pathogens](https://www.karger.com/Article/PDF/443360). The number and variety of dominant _Bacteroides_ and _Firmicutes_ drop in association with an overgrowth of [_Enterobacteriaceae_](https://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.0060280). Antibiotic consumption reduces the ability of _Bifidobacteriaceae_ to produce acetate_._ Acetate helps protect the epithelial layer of the colon from [infection](https://www.nature.com/articles/nature09646). As the micro-organisms in our gut work hand in hand, the antibiotic usage effects translate far beyond the immediate targets. The loss of one class of bacteria might indirectly hinder the growth and survival of others through mechanisms yet unknown. Consequently, the cumulative outcome of post-antibiotic dysbiosis causes the interruption of [colonization](https://www.ncbi.nlm.nih.gov/pubmed/22595318) resistance against _C. difficile_.
**When the acid reduces** _**C. difficile**_ **increase**
Another medication which relieves you of heartburn, ulcers and acid problem in stomach favours _C. difficile_ unintentionally. Known as Proton pump inhibitors (PPIs), these medications work by lowering the gastric acid production in the stomach. As the acid reduces, the environment of gut changes, and so does the variety of gut microbiota. Chronic use of PPIs shows a reduction in commensal bacteria, _Bacteroidetes_ and an increase in _Firmicutes_, enhancing the risk of _C. difficile_ [infection](https://www.ncbi.nlm.nih.gov/pubmed/26923470). Other exogenous factors, such as the use of histamine receptors, two blockers, prior hospitalization, and nasogastric tube placement, also find an association with increased risk of _C. difficile_ infection.
Some host-related risk factors like the severity of underlying illness, any recent abdominal surgery, aging, and others also influence _C. difficile_ infection. Our gut flora gradually alters with age. Evidence shows that the chance of _C. difficile_ infection increases with approximately 2% for every additional year of age after the [age of 18](https://www.nejm.org/doi/full/10.1056/NEJMoa1012413). In the elderly, the gut microbiota shows the high population of _Bacteroides_ spp. and [_Clostridium_ groups](https://www.ncbi.nlm.nih.gov/pubmed/20571116). However, a noticeable drop in microbial diversity occurs in _C. difficile_ infected patients when compared with a healthy individual of the [same age group](https://www.ncbi.nlm.nih.gov/pubmed/22162545).
**Restoring the gut microbiome**
Normal and healthy gut microbiota plays a vital role in preventing _C. difficile_ infection by offering colonization resistance against the [pathogen](https://www.ncbi.nlm.nih.gov/pubmed/22595318). Interruption in gut flora composition is profound in patients with recurrent _C. difficile_ infection, particularly those who have received numerous courses of [antibiotics](https://www.ncbi.nlm.nih.gov/pubmed/18199029). Thus, to prevent _C. difficile_ infection, the first and foremost target should be to restore and renew the healthy and beneficial intestinal flora. Another option is to eliminate or at least try to reduce the associated risk factors through the management and manipulation of gut flora back to normal.
Thus, to assess the risk of _C. difficile_ infection, all you need to do is to take a ‘Gut Microbiome Test.’ This test uses the cutting-edge DNA sequencing technology and advanced bioinformatics algorithms in a patient faecal sample to map the wide variety of micro-organisms present in the gut. The individual micro-organisms are identified by analysing the massive database of information. Prebiotics, probiotics and custom dietary recommendation prescribed on the basis of results help restore the gut flora back to health.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Gut Microbes: You grow - They Grow
Author: BugSpeaks
Published: 2020-01-03
Category: Microbiome
Meta Title: Gut Microbes: You grow - They Grow
Meta Description: We change with age, our habits, our body, our preferences, and a lot of other changes as we grow. So, when everything in our body
URL: https://www.bugspeaks.com/blog/gut-microbes:-you-grow---they-grow
We change with age, our habits, our body, our preferences, and a lot of other changes as we grow. So, when everything in our body changes as we age, how can the gut microbes not evolve? Yes, the millions of microbes have been growing and changing with you continuously, helping in healthy existence at every stage of life. The story of gut microbes begins since our inception in the mother’s womb. It is the embryo stage where the microbial colonization of the gut starts. Different ways of in-utero exposure to microbes involve [placenta](https://stm.sciencemag.org/content/6/237/237ra65), [amniotic fluid](https://www.ncbi.nlm.nih.gov/pubmed/22776980), [blood from the umbilical cord](https://www.ncbi.nlm.nih.gov/pubmed/16187156), [semen](https://www.cell.com/servlet/linkout?suffix=e_1_5_1_2_47_2&dbid=4&doi=10.1016/j.cell.2017.11.024&key=10.1016%2Fj.fertnstert.2013.07.1991&cf=pdf&site=fns-site), and [maternal gut microflora](https://www.nature.com/articles/srep23129). The colonization is indicated through similarity in microbial signatures between the amniotic fluid, placenta, and first stool of infant (meconium).

#### **Gut microbiome during birth**
As we complete 9-months of gestation in the mother’s womb, the brain releases oxytocin, and childbirth begins. Childbirth marks the second event of microbial transmission. The [nature of delivery](https://www.ncbi.nlm.nih.gov/pubmed/20566857), whether vaginal birth or a caesarean section, determines the type of microflora encountered by the infant. Vaginal delivery exposes the newborn to a range of microbes, such as _Enterobacteriaceae, Lactobacillus, Prevotella,_ which are present throughout the birth canal to the vaginal opening. A caesarean section, on the other hand, limits this interaction for the child and diminishes the diversity of microbial ecology. The microbes encountered during the caesarean section include _Bifidobacteria, Bacteroides, Staphylococcus, Propionibacterium_, and others. They are unique to the maternal epithelial cells’ representative of the interaction with maternal skin alone.
#### **Gut microbiome at infancy**
Further evolution of colonizing microbes depends on the interactions governed by early life events such as health status and childcare techniques employed by the parents. [Breastfeeding](https://www.ncbi.nlm.nih.gov/pubmed/21300508) is an essential part of ensuring immunity to the baby through antibodies present in breast milk. Besides immunity, it also serves as a critical source of microflora, such as _Bifidobacterium_. Moreover, it has also been found to provide nutrients for the microflora population in the infant’s gut through human milk oligosaccharides (HMOs). New-borns have a gut microflora composed mainly of _Enterobacteriaceae_ and _Staphylococcus_, which later attains _Bifidobacterium_ and _Lactobacillus_. Feeding formula to the infant, on the other hand, can alter the gut microflora at the initial stage of development, resulting in a microbial community with low biomass.
#### **Gut microbiome and toddlers**
As the infant grows further in age, the shift to [solid food](https://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.0050177), dietary supplements, and weaning from breast milk brings in a new wave of microbes such as _Bacteroidetes, Prevotella, Ruminococcus, Clostridium_, and _Veillonell._ The microflora ecology keeps diversifying with age as the number of food sources intensifies, and environmental interactions increase. Notably, the dominance of _Bifidobacteria_ diminishes as solid food is introduced, as the bacteria majorly processes the milk oligosaccharides. Such radical shifts of the microbial taxonomies continue up [to 3 years](https://www.ncbi.nlm.nih.gov/pubmed/20668239) of age as significant alterations in diet occur.
#### **Gut microbiome at adolescence**
The next stage of radical change in growth and development occurs at puberty. Significant hormonal changes drive the adolescent towards adulthood and sexual maturity with gender-specific modifications. Consequently, the microbial shifts also occur in a gender-specific manner, with women’s gut microflora [maturing earlier](https://www.sciencedaily.com/releases/2019/05/190515093613.htm) than men. Also, with an increase in age, the population of anaerobes increases where that of aerobes and facultative anaerobes decreases. The adolescent microbiome presents higher levels of _Clostridia_ and _Bifidobacteria_ compared to adults and expresses genes related to growth and development.
#### **Gut microbiome during adulthood**
Following growth and development in adolescence, the adult microbiome transforms to express genes associated with obesity and inflammation. In adulthood, a permanent, stable core community of [colonizers](https://www.ncbi.nlm.nih.gov/pubmed/24956966) exists. It works towards maintaining balance in the body and stable microbial configuration under conditions of duress. As the surface area of gut reaches a peak in the adult, the complexity of gut microbiome increases, which gradually expands to distinct niches in the gut. During pregnancy, the gut microflora also gets altered with an increase in abundance of [_Actinobacteria and Proteobacteria_](https://www.ncbi.nlm.nih.gov/pubmed/22863002)_._ The adult microbiome is susceptible to environmental distresses such as seasonal variation, temperature changes, or exposure to antibiotics.
Antibiotics affect the microflora composition; they promote the growth of proteobacteria and reduce the growth of actinobacteria and influence antibiotic resistance. For the later year of life, food and lifestyle have a significant impact on gut microflora[. Adult gut microflora](https://www.ncbi.nlm.nih.gov/pubmed/20571116) consists of _Bacteroides, Firmicutes, Clostridium, Ruminococcus, Eubacterium, Methanobrevibacter_ populations. With advanced age, the microflora ecology consists of _Faecalibacterium_, _Ruminococcus,_ and _Roseburia_, which are mostly obligate and facultative anaerobes.
#### **Gut microbiome during older age**
Ultimately, in the elder age, the significant variations in the gut microbiome constitution also appear. For example, _Bacteroides, Alistipes,_ and _Parabacteroides_ comprise almost 50% of the core microbiome in the elderly. Also, the maximum fluctuations occur between _Faecalibacterium, Ruminococcus_, and _Clostridium_ IV & XIVa clusters. Even the [living conditions](https://www.nature.com/articles/nature11319) of the elderly have also been indicated to influence the gut microbiome. For example, elderly living in long-term residential care presents a high proportion of Bacteriodetes such as _Coprococcus_ and _Roseburia_. While those living in community dwellings show a high proportion of Firmicutes such as _Coprobacillus, Anaerotruncus, Parabacteroides, Lactonifactor,_ and _Eubacterium._
#### **What does your gut speak to you?**
Thus, microflora composition plays a role in the predisposition of an individual to conditions like immune disorders, obesity, allergies, diabetes, neurological disorders, infections, metabolic diseases, and colorectal cancer at different stages of life. In light of the critical role the gut microflora plays in the healthy existence, scientists continue to harbour this knowledge to study the role of the microflora ecology and composition in finding ways to ensure a healthy body. The advancements in DNA sequencing technology allowed the generation of the catalogue of the unique microflora that inherits a host and deduces its role in the health status.
And, that’s why we developed [BugSpeaks](https://bugspeaks.com)[®](https://bugspeaks.com). To guide you in healthy ways customized as per your life stage and microflora Bug Speaks test with the help of cutting edge, ‘DNA sequencing technology’ identifies the array of your unique gut microbiome mapped from your faeces sample. The resulting customized results and dietary recommendations allow us to help you propagate healthy gut flora in accordance with your life stage and longevity.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Natural Gut microbiota as an alternative to Vitamin supplements??
Author: BugSpeaks
Published: 2019-12-19
Category: Diet and Supplements
Meta Title: Natural Gut microbiota as an alternative to Vitamin supplements??
Meta Description: Our gut serves as a natural habitat for trillions of micro-organisms. To keep us healthy the delicate balance of gut microbiota ne
URL: https://www.bugspeaks.com/blog/natural-gut-microbiota-as-an-alternative-to-vitamin-supplements
Our gut serves as a natural habitat for trillions of micro-organisms. To keep us healthy the delicate balance of gut microbiota needs harmonious balancing. Gut microbiota, managed by the mucosal barrier, draws a sort of [boundary](https://www.ncbi.nlm.nih.gov/pubmed/24129758) between these tiny tots and us. One of the countless ways these tiny tots help us includes providing several enzymes usually not encoded by our genome. These include the enzymes involved in the breakdown of polysaccharides, polyphenols, and synthesis of most of the [vitamins](https://www.ncbi.nlm.nih.gov/pubmed/28393285). Vitamins or micro-nutrients are biomolecules that act typically as cofactors / co-enzymes for several different enzymatic reactions associated with metabolism and other functions of the [body](https://www.ncbi.nlm.nih.gov/pubmed/27195115). As these little metabolic machineries share unique relation with our micronutrient requirements, an insight into their working take on a high degree of curiosity.
#### **Gut microbiome and Vitamin A**
Vitamin A or retinoic acid is essential for healthy skin, teeth, skeletal muscle, soft tissue, mucous membrane, central nervous system, and others. Trillion of gut microbes residing in intestinal mucosal lining metabolize Vitamin A obtained through food. One such relationship exists with commensal bacteria of _Clostridia_ class control vitamin A concentration in the gut. In turn, Vitamin A offers higher resistance to colonize the potential [pathogens](https://www.ncbi.nlm.nih.gov/pubmed/30566883) like _Salmonella_ _Typhimurium_. Vitamin A thus plays an important role in homeostatic interactions between the host and the microbiome.
#### **Gut microbiome and Vitamin B**
Most of the essential vitamins produced by our gut flora typically belong to the B group of vitamins, like, thiamine (B1), riboflavin (B2), niacin/ nicotinic acid (B3), pantothenic acid (B5), pyridoxine (B6), biotin (B7), folic acid (B9) and cobalamin (B12).
Folic acid produced by _Lactobacillus_ and [Escherichia coli](https://bmcbiol.biomedcentral.com/articles/10.1186/s12915-018-0534-3) is the most crucial member of the B vitamin family. It is vital for our genetic machinery as it takes a vital role in the biosynthesis of the purines and pyrimidines, the basic building blocks of DNA and RNA.
Riboflavin or vitamin B2, yet another vital vitamin, acts as co-enzymes in various vital reactions in cells. While biotin also belonging to B vitamin family helps convert food into energy in mitochondria. A group of useful microbes, _Bacteroidetes_, _Fusobacteria,_ and _Proteobacteria_, harbouring in our intestine own the complete pathway essential for the production of riboflavin and biotin [biosynthesis](https://www.ncbi.nlm.nih.gov/pubmed/31058161).
Another member of the group is cobalamin or Vitamin B12. It is a vital micro-nutrient that is responsible for healthy nerve and blood cells. There exists symbiosis or interdependence between us and the cobalamin producing gut micro-flora. For example, _Fusobacteria_, _Pseudomonas,_ and _Klebsiella sp_, present in the gut produce [vitamin B12](https://www.ncbi.nlm.nih.gov/pubmed/25941533). Thus, the microbes aid us by producing cobalamin in the gut. On the other hand, cobalamin protects, preserves, and revives the mucous membrane, the home of the whole [microbial community](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4260394/).

#### **Gut microbiome and vitamin C**
Vitamin C has numerous health benefits. So, we find ways to incorporate it into our diet, either by taking supplements or having foods loaded with vitamin C. It boosts our immune system and acts as a critical co-enzyme dealing with oxidative stress and scavenging free [radicals](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5472873/). _Burkholderia_, _Pseudomonas,_ and _Erwinia_ reside in the gut of normal healthy individuals. In the case of people suffering from Crohn’s disease, an **inflammatory bowel disease, these bacteria gain the capability of producing vitamin C, helping reduce inflammation.** However, these bacteria in normal conditions are incapable of producing [vitamin](https://www.nature.com/articles/s41575-018-0044-3).
#### **Gut microbiome and vitamin D**
Everyone knows about the relationship between sunlight and Vitamin D., But a recent Canadian study sheds light on the plausible association between microbes, vitamin D, and sunlight. [Findings](https://www.frontiersin.org/articles/10.3389/fmicb.2019.02410/full) suggest that exposure to sunlight, more precisely, UVB light can change the human gut microbiome. This alteration is more evident in vitamin D deficient people. Sunlight/ UVB light through the synthesis of vitamin D helps maintain healthy bones and teeth.
This vitamin also alters gut flora and helps to overpopulate the number of healthy bacteria present in our digestive tract. Enrichment of the good bacterial family of _Lachnospiracea_e and _Firmicutes_ and the decline of bad _Bacteroidetes_ in the gut following exposure to sunlight has been observed. Thus, _Lachnospiracea_es and _Firmicutes_ group of bacteria often present themselves in the gut of healthy individuals. Whereas, the people suffering from various inflammatory diseases lack the previously mentioned microbes in their gut.
#### **Gut microbiome and vitamin E**
Lack of Vitamin D renders the body prone to inflammatory diseases, while a lack of Vitamin E or ∞-tocopherol can impair antioxidant activities in our body that helps us get rid of the free radicals. These free radicals are otherwise detrimental to our health. However, the role of such a critical vitamin on gut microbiota composition and vice versa is still under [characterized](https://www.nature.com/articles/s41598-019-46193-w). Korean researchers showed an association between an altered gut microbiome composition and a low intake of vitamin E in [mice](https://www.ncbi.nlm.nih.gov/pubmed/31298050). Hopefully, more studies in the future will help delineate the effect of vitamin E on the intestinal flora.
#### **Gut microbiome and vitamin K**
To help you control blood loss upon injury Vitamin K comes to rescue. It is an essential factor for blood coagulation and also helps in the calcification of [bones](https://lpi.oregonstate.edu/mic/vitamins/vitamin-K). Vitamin K comprises of two structurally similar isoforms: vitamin K1 and vitamin K2. We get vitamin K1 through the green leafy vegetables, which we eat. However, for vitamin K2, we are dependent on our gut bacterial community. Our intestinal flora converts vitamin [K1 to K2](https://www.ncbi.nlm.nih.gov/pubmed/22940212). [Escherichia coli](https://www.ncbi.nlm.nih.gov/pubmed/22502809) and [Lactobacilli](https://www.ncbi.nlm.nih.gov/pubmed/22502809) present in our gut can also bio-synthesizes vitamin K2. However, vitamin K1 is not produced in our gut.
All the evidentiary material glorifies the intricate relationship between microbes and vitamins. The composition of the human gut microbiota, as established during the early years after birth, gets altered over time. However, under the influence of lifestyle factors and usage of drugs and antibiotics, the composition of gut bacterial species could go down the wrong road. Such changes may alter biosynthesis of vitamins, which are essential for our metabolism, [adversely](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4315779/).
**To conclude,**
So, for an ample supply of the essential vitamins vital for a healthy life, we need healthy gut flora. If a vitamin deficiency is caused by various reasons, to deal with it in a better way, a ‘Gut Microbiome Test’ can always help. In this process, with the help of cutting edge ‘DNA sequencing technology,’ the array of your unique gut microbiome will be identified and mapped from your faeces sample. Based on this, a tailor-made diet plan will be recommended to you. As it takes a while to change your gut microbiome, you must be patient to get good results.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Sound gut for Sound Sleep
Author: BugSpeaks
Published: 2019-12-05
Category: Microbiome and Lifestyle
Meta Title: Sound gut for Sound Sleep
Meta Description: Almost all of us have experienced sleeplessness some time in our life. Occasional sleep disruption is quite reasonable. However, i
URL: https://www.bugspeaks.com/blog/sound-gut-for-sound-sleep
Almost all of us have experienced sleeplessness some time in our life. Occasional sleep disruption is quite reasonable. However, it’s reasonably problematic to have a sleep problem regularly. Lack of good night sleep daily can restrict your everyday activities. It may seriously affect your quality of life through an adverse impact on mood, work efficiency, stress handling, and others.
According to [Mayo Clinic](https://www.mayoclinic.org/diseases-conditions/insomnia/symptoms-causes/syc-20355167), ‘A sleep disorder or insomnia is a condition that frequently impacts your ability to get enough good quality sleep’. It can arise from all sorts of stresses in your life, disrupted sleep-wake cycles, including jet lag, frequent change in work shifts, and poor lifestyle, like having a heavy meal or alcohol in the late evening or watching television till night. Insomnia often finds an association with medical conditions like **mental health disorders,** or [use of medications](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4634348/) like antidepressants, asthma or blood pressure, or products containing caffeine.
You would be quite surprised to know that disrupted sleep is often associated with poor gut microbial health!
**Sleeping for a healthy gut microbiome**
Trillions of micro-organisms inhabit your gut remains a known fact. They [outnumber](https://www.ncbi.nlm.nih.gov/pubmed/16497592/) the total of the cells in your body by at-least ten times. These micro-organisms form an ecosystem of their own and known as the microbiome. The various kinds of intestinal micro-organisms display a dynamic equilibrium in a healthy individual. This subtle balance is vital for health, and a slight imbalance increases the [host’s](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3027896/) [vulnerability](https://www.nature.com/articles/nature08821) to [disease](https://www.physiology.org/doi/full/10.1152/physrev.00045.2009).
It's quite apparent that diet would have the most significant influence on your gastro-intestinal tract and the micro-organisms residing in there. However, a recent review conducted by [Dr. James F. Cheeseman](https://unidirectory.auckland.ac.nz/profile/j-cheeseman) from the University of Auckland, New Zealand, established that not only the food we consume but also the sleep patterns are also connected to our [gut microbiome](https://www.ncbi.nlm.nih.gov/pubmed/30709031).
The researchers have observed that [gut microbiome](https://www.nature.com/articles/nature06244) plays a significant role in [circadian clock](https://www.ncbi.nlm.nih.gov/pubmed/25891358) function along with other phenotypic characteristics, like, immunity, [metabolism](https://www.ncbi.nlm.nih.gov/pubmed/20040631), and others. The circadian rhythm is our inner clock, which controls our body’s energy disbursement, hunger, and snooze. We usually get about seven hours of sound sleep every night. In the morning, when we wake up, our body warms up to carry out daily chores. To run our body, we need energy, and energy comes from the food we eat during the day. At night, our body needs rest to rewind, so we fast and go to [sleep](https://www.ncbi.nlm.nih.gov/pubmed/30568608).
**Gut microbiome resonates with the bodily rhythm**
The scientific world now accepts the robust connection between sleep and intestinal wellbeing. A good quality night sleep allows more flourishing and better functioning gut microbiome and [vice versa](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1395357/).
Researchers report the reflection of our body’s underlying rhythmicity in the intestinal flora too. For example, _Firmicutes sp_. remains overpopulated during the daytime when we have frequent meals. While, the number of some gut flora such as _Bacteroidetes_ _sp._, _Proteobacteria sp.,_ and _Verrucomicrobia sp_. increases during the night when we sleep and fast. Generally, the microbes which grow on food derived fibres grow at night, as what we consume during the day reaches the colon by [night time](https://www.ncbi.nlm.nih.gov/pubmed/24882755).
Gut flora follows the rhythm by secreting specific molecules at certain times of the day. At night, secretion of factors responsible for energy metabolism, DNA repair, and proliferation occurs. During day-time, flora [harbouring in the gut](https://www.ncbi.nlm.nih.gov/pubmed/31123355) releases molecules essential for their colonization.
As in circadian rhythm, we fast and sleep during the night. It is advantageous to maintain a healthy gut microbiome. During fasting the _Bifidobacteria sp._, _Ruminococcus bromii_ and _Clostridium chartatabidum_, produce [short-chain fatty acids](https://www.gutmicrobiotaforhealth.com/en/short-chain-fatty-acids/), including, butyrate, and propionate. Butyrate protects our gut and regulates glucose levels by a complex network of metabolites and various [hormones](https://www.ncbi.nlm.nih.gov/pubmed/29903615). Propionate plays a crucial role in fat metabolism in [liver](https://www.ncbi.nlm.nih.gov/pubmed/29903615). The scenario is entirely different at mealtimes, where the release of factors helpful for lipid and protein adsorption occurs.

**Gut microbiome, sleep, and hormones**
Neurotransmitters like serotonin and GABA secreted by brain control our [sleep-wake cycle](https://www.ncbi.nlm.nih.gov/pubmed/22025877). Astoundingly, certain intestinal bacteria including, _Turicibacter sanguinis_ and _Clostridia_ _sp.,_ release specific signalling molecules that trigger the production of [serotonin](https://www.nature.com/articles/s41564-019-0540-4). By modulating serotonin levels, the gut microbiome can interfere or improve our sleep pattern. Melatonin, the sleep hormone generated from serotonin, helps to improve our [sleep](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6445894/).
Interesting enough, studies have shown that ingesting probiotics may help increase melatonin secretion and sleeping quality, opening an avenue for the future [use of probiotics for sleep regulation](https://www.gutmicrobiotaforhealth.com/en/new-data-link-gut-bacteria-melatonin-circadian-clock/), gut health, and overall well-being16. Alteration in the gut flora often can induce inflammation [in the body](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4056765/), which may lead to [sleep deprivation](https://www.ncbi.nlm.nih.gov/pubmed/24409051) as well. It leads to a vicious cycle of sleep deprivation, which can exert an adverse influence on the well-being of gut microbiome and _vice-versa_.
Thus, ignoring sleep problems and disorders can end up badly for your physical health. Stress and anxiety due to sleep deprivation can lead to accidents, poor performance at work, and memory-related issues. Poor gut health due to lack of sleep can also lead to excessive weight gain, which plays a central role in many life-threatening diseases.
**Testing little gut microbes for healthy sleep**
By now, you may have a clear picture in your mind showing the entangled relation of the health of your gut microbiome and sleep. So, to resolve sleep disorders, common sleeping pills or CPAP machines will not help. To cure it of its root, all you need is to get a ‘Gut Microbiome Test’ done. ‘Gut Microbiome Test’ uses sophisticated ‘DNA sequencing technology’ inpatient stool samples to identify the exclusive collection of micro-organisms existing in your gut. Then this data is used to provide a diet plan exclusively for you, comprising of gut-friendly, plant-based, and fibre-rich food with no added sugar. You will also be advised to have a lifestyle supporting your body clock, like regularly having a meal at least a couple of hours before going to bed, and strictly restricting snacks/ light meals before going to bed.
Therefore, this plan aimed to rectify your sleep issues by improving your gut microbiome.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## From Gut to Joints: Microbiome & Arthritis
Author: BugSpeaks
Published: 2019-12-02
Category: Microbiome and Disease
Meta Title: From Gut to Joints: Microbiome & Arthritis
Meta Description: Arthritis is swelling and tenderness of one or more joints in your body. This disease is widespread, yet, not well understood. Fro
URL: https://www.bugspeaks.com/blog/from-gut-to-joints:-microbiome-&-arthritis
Arthritis is swelling and tenderness of one or more joints in your body. This disease is widespread, yet, not well understood. From the 100 different types of arthritis, osteoarthritis and rheumatoid arthritis represent the most common ones. Regardless of the type, symptoms are similar in all the cases, which include joint pain and stiffness. The symptoms typically worsen with age. However, people of all ages, sexes, and races have arthritis, with more prevalence in women. Being the leading cause of disability in the world, arthritis, often compromises quality of life.
Moreover, upon affecting weight-bearing joints, arthritis restricts one from walking comfortably or even sitting up straight. Per the treatment a doctor often advises medications, physical therapy, weight loss and exercise. But a diet alteration to transform your gut microbiome forms a rare recommendation. However, experts at world-renowned mainstream institutions nowadays acknowledge the role of the gut microbiome in certain types of arthritis, autoimmune arthritis. Autoimmune arthritis further includes rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis.
**Few pieces of evidence:**
Scientists have observed the digestive tracts of people with rheumatoid arthritis to be overpopulated with potentially disease-causing bacteria. Microbes like [Prevotella copri](https://ard.bmj.com/content/78/5/590), [Collinsella sp.](https://genomemedicine.biomedcentral.com/articles/10.1186/s13073-016-0299-7), and [Eggerthella lenta](https://genomemedicine.biomedcentral.com/articles/10.1186/s13073-016-0299-7) occur in high numbers while the number of healthy bacteria, like, [Bifidobacterium](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4973033/), the [Porphyromonas-Prevotella](https://www.ncbi.nlm.nih.gov/pubmed/18528968) group of bacteria, [Bacteroides fragilis](https://www.ncbi.nlm.nih.gov/pubmed/18528968) subgroup, [Eubacterium rectale](https://www.ncbi.nlm.nih.gov/pubmed/18528968), and [Clostridium coccoides](https://www.ncbi.nlm.nih.gov/pubmed/18528968) group is relatively low.
Also, people carrying the [HLA B27 gene](https://www.ncbi.nlm.nih.gov/pubmed/29287307) show a higher risk of developing autoimmune arthritis. This gene presents an association with changes in the gut microbiome. Thus, it presents a link between gut flora and rheumatoid arthritis and other forms of autoimmune arthritis.
**Gut microbiome and autoimmune arthritis**
Our gastrointestinal tract is home to a large and complex community of [micro-organisms](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3728647/). This gut microbiota keeps our body fit in many ways. These include healthy digestion, production of nutrients, detoxification, fat metabolism and storage, angiogenesis regulation, protection against pathogens, and regulation of the [immune system](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3728647/). However, overtime, under the influence of altered food habits, lifestyle, and medication (antibiotics and steroids) intake it undergoes modification. These alterations frequently lead to the imbalance of the intestinal microbiome. An imbalance in the gut microbiome can lead to chronic inflammatory and autoimmune diseases, such as arthritis.

**Leaky Gut Syndrome and Arthritis**
As per researchers and clinicians ‘Leaky Gut Syndrome,’ characterized with increased intestinal permeability also plays a role in arthritis. The syndrome results from the damage to the small intestine wall. When healthy, the small intestine only allows useful nutrients to pass across and enter the bloodstream. However, upon damage, the intestine permeability stands compromised and larger molecules like half-digested fats, proteins, starches, and even bacteria reach the blood. These microbes are often gram-negative gut bacteria whose outer membranes contain molecules called lipopolysaccharides. The body recognizes lipopolysaccharides and partially digested food products as foreign substances, and they trigger an immune response. The antibodies thus produced, travel through the bloodstream and create an inflammatory response in other parts of the body, including joints, leading to [arthritic pain](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5610356/) and [swelling](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5440529/). Though the matter is controversial, yet there is evidence that shows that healing a leaky gut with a strict diet and nutritional supplements can help to control arthritis.
**Inflammation and Arthritis**
Inflammation is a common feature in almost all types of arthritis such as rheumatoid arthritis, gout, ankylosing spondylitis, psoriatic arthritis, and pseudogout. Even osteoarthritis—which, lacks an autoimmune response— may present with inflammation. A healthy gut microbiota find implication in inflammation control by influencing the differentiation of inflammatory cell types, and production of regulatory molecules, [like cytokines](https://www.ahajournals.org/doi/10.1161/CIRCRESAHA.118.313234).
**A healthy gut means a healthy body**
These days correlation between intestinal health and the gut microbiome remains well-understood. So, the best way to deal with arthritis is to maintain a healthy diet to have a healthy gut flora. Diet alteration stands as a great way to achieve this but offers temporary and quick shift in the gut microbiome. To instil a permanent shift toward healthy gut flora a long-term commitment to a healthy diet full of leafy vegetables and plant-based whole foods remains essential. A fibre-rich diet helps beneficial gut flora to flourish. They produce short-chain fatty acids from fibre present in foods.
These short-chain fatty acids contribute to the wellbeing of the intestinal lining and may prevent [disease](https://www.ncbi.nlm.nih.gov/pubmed/16633129). Moreover, short-chain fatty acids help curb glycaemic response and satiety, promote weight loss, enhance mineral absorption, and reduce systemic [inflammation.](https://www.ncbi.nlm.nih.gov/pubmed/21840809) All of these are important for the management of arthritis. These changes can lead to a better functioning immune system, reduce inflammation, and lower arthritis-related [pain](https://www.ncbi.nlm.nih.gov/pubmed/23880135). On the other hand, the bacteria thriving on a high-protein, low-fibre diet produces ammonia and other compounds, having potentially harmful effects. Therefore, a healthy, balanced gut microbiome fosters healthy intestines, and vice versa, both of which play a crucial role in a person’s overall health.
**Other factors**
In addition to diet, other factors, including heredity, stress, and the environment also influence the gut microbiome. For this reason, regular exercise, meditation, and reduction in exposure to toxins must accompany gut rehabilitation through a healthy diet. Physical exercise also alters the composition and functional capacity of the gut microbiota, independent of diet. However, exercise-induced alterations of gut microbiota may depend on obesity status, exercise modality, and [exercise intensity](https://www.ncbi.nlm.nih.gov/pubmed/30883471).
**Get ‘Gut Microbiome Test’**
As we all have a unique set of gut-microbiota, the dietary recommendations must be tailor-made. So, if you want to fix your arthritis before it gets worse, take a ‘Gut Microbiome Test.’ This test uses cutting edge DNA sequencing technology in patient stool samples to map and identify the unique array of micro-organisms present in the gut. Then this data is used to provide a customized dietary recommendation consisting of probiotics and prebiotics. This diet is targeted at the improvement of arthritis. As it may take quite a few months to get your intestinal flora changed, you are advised to follow the diet plan religiously for several months before deciding whether it works or not. Patience is the key to succeed through gradual progress.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Resolving constipation at microbial level
Author: BugSpeaks
Published: 2019-11-21
Category: Microbiome and Disease
Meta Title: Resolving constipation at microbial level
Meta Description: You will surely be surprised to know that your gut, a big long muscle, more than six meters long, is home to trillions o
URL: https://www.bugspeaks.com/blog/resolving-constipation-at-microbial-level
You will surely be surprised to know that your gut, a big long muscle, more than six meters long, is home to trillions of microorganisms such as bacteria, archaea, viruses, fungi, metazoa, and protozoa. Each of the microbes has more than [1000 different species](https://www.ncbi.nlm.nih.gov/pubmed/27478747) in there. These communities of gut microbes work in harmony with the human body and evolve continually. The microbes come together to work in cooperation and competition and maintain balance in [microbiome](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4727996/). Nevertheless, some of them can be harmful too.
This vast array of gut-harbouring micro-organisms is exposed to the food you eat on a day-to-day basis. Thus, the food essentially has a far-reaching influence on the gut microbiome harmony and functioning4. Increased use of antibiotics, drugs, probiotics, and dietary changes disrupts the delicate balance of the gut microbiome. This imbalance further enhances vulnerability to infections, gastrointestinal tract [inflammation](https://www.ncbi.nlm.nih.gov/pubmed/26460205) and chronic illnesses ranging from metabolic diseases to [gastrointestinal disorders](https://www.ncbi.nlm.nih.gov/pubmed/31315227).

#### **Microbial issues of constipated gut**
The discussion on gastrointestinal disorders evokes the most common issue of constipation. What you eat reaches the large intestine in a couple of hours and exits body the next morning. However, the process can, [at times, take up to a week,](http://www.aboutconstipation.org/site/what-is-constipation/normal-function) and the result is a constipated stomach. It’s a sort of little awkward topic that no one wants to discuss openly, yet many suffer. A gut health survey conducted by Abbott Healthcare in 2018 showed that 22% of the country's adults suffer from constipation. Interestingly constipation shows higher prevalence in metro cities believed to be mostly due to urban diet and [lifestyle](https://www.ncbi.nlm.nih.gov/pubmed/27478747). Additionally, one-fourth of constipated individuals suffer from severe pain.
Constipation is clinically defined by difficult, infrequent, or inadequate [bowel movements](https://www.gastrojournal.org/article/S0016-5085\(16\)00222-5/abstract). It is one of the most common gastrointestinal diseases, which may result from a variety of reasons. Continued research shows altered gut flora as one of the primary causes of [constipation](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6379309/). For example, the number of certain bacterial species like Bifidobacteria, Lactobacilli, and Bacteroides decrease in constipated adults. Lower levels of Bifidobacteria and high level of Bacteroidetes cause both functional constipation and constipation type irritable bowel syndrome. Also, the numbers of potentially pathogenic microorganisms like Pseudomonas aeruginosa and Campylobacter jejuni increase in people suffering from [constipation](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3145058/). Such alteration in microbiome populations disrupts several metabolic processes causing constipation.
#### **Altered gut flora and constipation**
**_Metabolism_**
Studies show that gut microbiota helps metabolize substances using special enzymes which are not available in humans. Thereby, they help metabolize cholesterol, bile acid, and hormones. Moreover, gut microbes also produce short-chain fatty acids that prove beneficial in numerous ways. For example, bacteria produce short-chain fatty acids like butyrate, acetate, and propionate to carbohydrates in the absence of [oxygen](https://www.hindawi.com/journals/ecam/2019/9372563/). A high concentration of butyrate inhibits mucin secretion and stimulates water and electrolyte absorption in the large intestine. The absence of mucin and water retention reduces the stool volume and increases hardness leading to [constipation](https://www.ncbi.nlm.nih.gov/pubmed/24967088). Correspondingly, research findings support an increase in butyrate-producing genera in constipated [patients](https://www.ncbi.nlm.nih.gov/pubmed/28393285).
**_Immunity_**
Gut microbiome also concerns immunity as a huge proportion of the immune system finds an association with gut health. For example, Clostridial species, Bacteroidetes species, and Candidatus svagella are found to disrupt colonic immunity. Colonic immunity ultimately causes the development of colonic motility disorder and chronic constipation. They act through the secretion of anti-inflammatory molecules and through function modulation of immune cells. These changes further impact the motility and secretory functions of intestine by altering the amount of physiologically active substances and gut’s metabolic [environment](https://www.ncbi.nlm.nih.gov/pubmed/27478747).
Thus, intestinal flora modulates gut mobility by altering bacterial metabolites (such as SCFAs) or bacterial cell components (such as lipopolysaccharide) or through interactions between bacterial cells and the host immune system. Consequently, showing a crucial role in the pathology of constipation.
#### **Gut microbes and gastrointestinal movements**
Another microbe, Methanobrevibacter smithii, in concert with gut bacteria produces methane from dietary carbohydrates. The methane produced slows down gastric transit. Research supports its role in constipation as these micro-organisms remain abundant in constipated [individuals](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4769647/). Some indirect evidence also shows that gut microbiota regulates endocrine cells to secrete hormones like somatostatin and Neuropeptide Y, which finally modulate the gut motility and constipation1.
Further, saturated long-chain fatty acid (SLCFA) generated by gut bacteria enhances colonic contraction and increases stool frequency in [rat models](https://www.ncbi.nlm.nih.gov/pubmed/11171619). Thus, low abundance or absence of bacteria producing SLCFA may play role constipation. Moreover, the bacterial endotoxin lipopolysaccharide influences intestinal motility by delaying gastric emptying and inducing sphincteric [dysfunction](https://www.ncbi.nlm.nih.gov/pubmed/29903041). Hence over-population of bacteria producing lipopolysaccharide in the gut can be a reason for developing constipation.
For decades, people with constipation receive advise to change their diets, consume a fibre-rich diet with plenty of probiotics, and indulge in regular physical exercise. However, one size does not fit all, nor do these recommendations. The bacterial community within the stomach and intestines in each and every person remains unique. Thus, a tailor-made plan is needed for every constipated gut.
So, if you really want to build a diet that promotes your gut health, the ‘Gut Microbiome Test’ may be your saviour. Using DNA sequencing technology and advance bio-informatics algorithm, the test maps the array of micro-organisms present in your gut. With the help of an advanced artificial intelligence algorithm based on the nutrient database and disease susceptibility index, the BugSpeaks test provides a customized dietary recommendation. This diet targets the root cause of constipation and helps the bowels move freely.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Best Podcasts on Microbiome
Author: BugSpeaks
Published: 2019-09-23
Category: Microbiome
Meta Title: Best Podcasts on Microbiome
Meta Description: We are living in the "golden-age-of-podcasts”, with an
URL: https://www.bugspeaks.com/blog/best-podcasts-on-microbiome
We are living in the "[golden-age-of-podcasts](https://chartable.com/blog/golden-age-of-podcasts)”, with an increasing of people listening to podcasts daily or at least once a week. There has been a huge content explosion in this audio consumption format, with ~600 podcasts being started every day. Podcasts have opened different ways of consuming news, articles and even whole books, along with building habits, around walking, jogging and even daily commute to your workplace.
Another field that has witnessed a relatively similar rise in its adoption, is the gut microbiome testing in context of one's health and wellness. Leucine Rich Bio's BugSpeaks®, is India's first and only gut microbiome-based test, which is catering to a wide range of consumers committed to improve their overall health and wellness.
Merging the best of both worlds, here's a list of Podcasts that will give u a head start on both ends.
If you are already into podcasts, and you are one of those who have or who are willing to adopt a healthy lifestyle, then you can start understanding what and how does one's gut microbiome can aid you in achieving your health goals, through these podcasts.
On the other hand, you are already into a healthy lifestyle, and wants to keep constant tab on the most updated information about being healthy, especially through gut microbiome, then you can build a habit of listening these podcasts, which will have all the information you need at our finger tips.
So, here are some of the most consumed podcasts involving gut health, microbiome and overall wellness.

The Microbiome Report
Bio
The microbiome. It dictates so much of how we move through the world – from how we digest our food to the mates we choose as we spin around the globe. On this show, we're investigating how the things we do everyday impacts the bugs of our bodies. The Microbiome Report is powered by BIOHM Health.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/the-microbiome-report/id1443154886) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly90aGVtaWNyb2Jpb21lcmVwb3J0LmxpYnN5bi5jb20vcnNz) [PlayerFM](https://player.fm/series/the-microbiome-report)
The Perfect Stool Understanding and Healing the Gut Microbiome Lindsey Parsons
Bio
Hear host Lindsey Parsons interview functional and integrative medicine professionals, patients and scientists about the gut microbiome, the current state of research and how you can apply it to your life. Hear patients talk about their experiences with fecal microbiota transplantation (FMT) both in clinics and do-it-yourself and how it impacted their gut issues. Learn about the gut microbiome's influence not just on digestive health, but on cardiovascular health, mental health, autoimmune diseases, skin conditions and overweight/obesity.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/perfect-stool-understanding-healing-gut-microbiome/id1444156616) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly90aGVwZXJmZWN0c3Rvb2wubGlic3luLmNvbS9yc3M) [PlayerFM](https://player.fm/series/the-perfect-stool-understanding-and-healing-the-gut-microbiome)


BacterioFiles
Bio
The podcast for microbe lovers: reporting on exciting news about bacteria, archaea, and sometimes even eukaryotic microbes and viruses.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/bacteriofiles/id352470437) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cDovL2JhY3RlcmlvZmlsZXMubWljcm9iZXdvcmxkLmxpYnN5bnByby5jb20vcnNz) [PlayerFM](https://player.fm/series/series-1567470)
Metagenics Clinical Podcast
Bio
Welcome to the Metagenics Clinical Podcast where Natural Healthcare Practitioners can hear innovative, cutting-edge information from leading experts from around the world. Join us while we explore the latest evidence in Natural Health, challenge and debunk industry myths and offer practical, tangible, clinical tools which will transform your practice
[Apple Podcasts](https://podcasts.apple.com/au/podcast/metagenics-clinical-podcast/id1177865679) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly9mZWVkcy5maXJlc2lkZS5mbS9tZXRhZ2VuaWNzY2xpbmljYWxwb2RjYXN0L3Jzcw) [PlayerFM](https://player.fm/series/metagenics-clinical-podcast)


Microbe Talk
Bio
Microbe Talk is a podcast from the Microbiology Society, interviewing researchers about bacteria, viruses and parasites. We are the largest microbiology society in Europe, covering all aspects of microbial science.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/microbe-talk/id505999112) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly9taWNyb2JldGFsay5wb2RvbWF0aWMuY29tL3JzczIueG1s) [PlayerFM](https://player.fm/series/microbe-talk)
The Doctor's Kitchen Podcast
Bio
Welcome to The Doctor's Kitchen Podcast with Dr Rupy Aujla. Covering a range of topics from the principles of healthy eating to how to prevent and treat illness, Dr Rupy and his panel of experts draw on the latest research to give you actionable tips to help supercharge your health.What you choose to put on your plate is one of the most important health interventions anyone can make. Dr Rupy's cooksbook, The Doctor's Kitchen and his latest, Eat To Beat Illness, are out now all good bookshops and ebook.
[Apple Podcasts](https://podcasts.apple.com/gb/podcast/the-doctors-kitchen-podcast/id1316938642) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly9hdWRpb2Jvb20uY29tL2NoYW5uZWxzLzQ5MzUxNjYucnNz) [PlayerFM](https://player.fm/series/the-doctors-kitchen-podcast)


The Gutology Podcast
Bio
The Gutology project is on a mission to help you optimise your gut. Join our gut health expert Julia Davies and award winning podcaster Ollie Gallant for a fascinating journey into your digestive system. We bring the science behind gut health and deliver it in 'digestible' chunks. This will change the way you think about your health forever.
Julia Davies is a leading Nutritionist, SCENAR practitioner and lecturer in biomedicine and biochemistry. Ollie's digestive health started deteriorating in his early twenties and all of that changed when he met met Julia. They're now on a mission to help people do the same regardless of income, resource or location.
[Apple Podcasts](https://podcasts.apple.com/gb/podcast/the-gutology-podcast/id1462264594) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly9ndXRvbG9neS5saWJzeW4uY29tL3Jzcw) [Spotify](https://open.spotify.com/show/5mMSRQRPJH4FECwrfIgZnz)
Love & Guts
Bio
This platform is designed to speak to those who value living a full and magnificent life. Your host Lynda Griparic is a naturopath, nutritionist and yoga teacher with an avid and sometimes awkward interest in all things digestive health, especially bowel movements and the intelligent communication systems that exist in the body.
Love and Guts explores all things mind-set, brain health, digestive health, pooping patterns, connection, communication and so much more. It's content may challenge you and at times it may get a bit whacky but never too bio-hacky as I like to keep things practical and accessible to all.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/love-guts/id1345246309) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cDovL2x5bmRhZ3JpcGFyaWMubGlic3luLmNvbS9yc3M) [PlayerFM](https://player.fm/series/love-guts)


Gut Health Gurus Podcast
Bio
A big welcome to The Gut Health Gurus podcast brought to you by Food Scientist, Kriben Govender and Psychology graduate, James Schadrach on a journey to investigate the gut brain connection. If you're fascinated by all things gut health, the microbiome, fermented foods, biohacking, optimal health & wellness and longevity then this podcast is for you. We bring you, various experts from around the world and explore topics from a scientific perspective.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/gut-health-gurus-podcast/id1433882512) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cDovL2d1dGhlYWx0aGd1cnVzLmxpYnN5bi5jb20vcnNz) [PlayerFM](https://player.fm/series/2431066)
The Human Microbiome: Guts And Glory
Bio
A special series podcasts from NPR, concering the human microbiome, where trillions of bacteria, virus, fungi and other microbes hiding out in the human body shape health.
[Podcast Link](https://www.npr.org/series/218987212/microbiome)


Feel Better, Live More with Dr Rangan Chatterjee
Bio
In this podcast, we hear stories from leading health experts and exciting personalities who offer easy health life-hacks, expert advice and debunk common health myths giving you the tools to revolutionise how you eat, sleep, move and relax. Hosted by Dr Chatterjee – a GP with over 16 years experience, star of BBC 1's Doctor in the House and author of The 4 Pillar Plan – Feel Better, Live More aims to inspire, empower and transform the way we feel. When we are healthier we are happier because when we feel better we live more.
[Apple Podcasts](https://podcasts.apple.com/podcast/feel-better-live-more-with-dr-rangan-chatterjee/id1333552422?mt=2) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cHM6Ly9yc3MuYWNhc3QuY29tL2ZlZWxiZXR0ZXJsaXZlbW9yZQ) [PlayerFM](https://player.fm/series/feel-better-live-more-with-dr-rangan-chatterjee)
The Gut Loving Podcast: All about IBS and the low FODMAP diet
Bio
The Gut Loving Podcast is all about irritable bowel syndrome (IBS) and the low FODMAP diet! The low FODMAP diet is a relatively new approach to effectively treat IBS symptoms and is scientifically backed up by a vast number of medical studies.
The Gut Loving Podcast is hosted by Laura Tilt (an experienced Dietitian in the UK specialising in IBS and the low FODMAP diet) and Huelya Akyuez, patient and creator of project sezamee - gut loving food (video recipes and low FODMAP events) after living with IBS for over 20 years. Together, Laura and Huelya started The Gut Loving Podcast to help others with IBS get clear on the facts - and learn more about how to take control of their condition.
[Apple Podcasts](https://podcasts.apple.com/us/podcast/the-gut-loving-podcast-all-about-ibs-and-the-low-fodmap-diet/id1278767162) [Google Podcasts](https://podcasts.google.com/?feed=aHR0cDovL3RoZWd1dGxvdmluZ3BvZGNhc3QubGlic3luLmNvbS9yc3M) [PlayerFM](https://player.fm/series/the-gut-loving-podcast-all-about-ibs-and-the-low-fodmap-diet)

---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Ask the Doctor - Gut
Author: BugSpeaks
Published: 2019-03-29
Category: Others
Meta Title: Ask the Doctor - Gut
Meta Description: In our previous blog on “commen
URL: https://www.bugspeaks.com/blog/commensals-and-cancer-therapy
In our previous blog on “[commensals and cancers](https://www.bugspeaks.com/blog-details?id=25&&title=Commensals and Cancers)” we tried to compile the updated information on relationship between gut microbiome and its effect on cancer (in general) and colorectal cancer (CRC) (in specific). Dysbiosis of gut microbiome is one the best studied associations for colorectal cancer, apart from its genetic and lifestyle related triggers. The scientific community has already deemed that the next decade of research in the field of oncology will be dominated by greater integration of microbiome and its impact, which will drive the invention and discovery of novel microbiome derived therapeutics of cancer.
Given this context, cancer therapy has also gained momentum in the recent times, with _reports of microbial communities within the gut modifying (both potentiating and weakening) the cancer drugs._ This has started a dialogue on the need for in-depth knowledge of the impact of the microbiome on cancer therapeutic strategies.
**Microbiome and Modulation of Cancer Therapy**
Human microbiome, specifically the gut microbiome has been extensively reported to modulate antitumor efficacy of chemotherapies and immunotherapeutic agents, in many pre-clinical models. It achieves it so largely in two ways; _“Modulate the immune system and Modify the drug or aid the process of such modifications”._
Through such modification the microbiome is said to either potentiate (increase) or weaken (decrease) the efficacy of immunotherapy and hence deemed essential for optimal drug efficacy of cancer drugs.

**Modification of CRC Drug (5-FU)**
Standard systemic treatment for CRC (metastatic and/or irresectable stage) is mostly based on fluoropyrimidine, such as 5-fluorouracil (5-FU). It is orally administered as capecitabine, or TAS-102. Capecitabine is a precursor of 5-FU, which must get converted to its active form to be effective against CRC. Many _in vitro_ studies have indicated a significant role of gut microbiota in this conversion. An increased response of CRC cell lines to 5-FU in presence of Lactobacillus _plantarum_ supernatant has been reported. 5-fluorocytosine (5-FC) samples, incubated with viable Escherichia _coli_ showed a higher increase in active 5-FU concentration. 5-FC, incubated with human faecal samples, also displayed a significant degradation reaction, indicating a significant increase in its potentiation. Hence associated with efficacy of drug and response.

On the other end of the spectrum, Colorectal cancers enriched in Fusobacterium _nucleatum_ have demonstrated worse prognosis. Because of the bacteria conveyed resistance to oxaliplatin and 5-fluorouracil by inducing [autophagy](https://www.nature.com/articles/nrd.2017.22). Oral administration of 5-Fu has been reported to alter the bacterial diversity and community composition of the gut. Several such instances have been reported involving both chemotherapeutic efficacy and chemotherapeutic cytotoxicity.

**Microbiome and Modulation of the Immune System**
Effector T lymphocytes represent a critical branch of the adaptive immune response to antigens. But, controlling the length and strength of this activation is very critical, which if unchecked can go damage even self-cells. Hence, a series of coinhibitory molecules, called immune checkpoints, are expressed by antigen presenting cells for switching off T cell activation.
Tumour cells inactivate cytotoxic CD8+ T cells, by expressing the same immune checkpoint molecules, and evade the anti-tumour immune response. Hence, targeting these immune checkpoints has emerged as a promising approach for cancer therapy. The most prominent one has been the class of drugs that act against programmed cell death protein 1 (PD-1) and PD-1 ligand 1 (PD L1).
However, patient responses to immune checkpoint therapies have been heterogeneous, varying with every individual and been reported to be transient. A cohort of 61 patients evaluated that PD-L1 expression in colorectal cancer defines three subsets of tumour immune microenvironments. Of which, third group, with low TANs, high IICs, both IICs and NCs PD-L1 expressing, would be the ideal candidate for this type of therapy. However, heterogeneous and transient responses to anti-PD1 therapy was still a concern.
In this context, studies have reported that the gut microbiota, possessing a pronounced modulatory effect on the immune system, may enhance responses to immune checkpoint therapies. Several Bifidobacteria species seem to stimulate dendritic cells (a kind of antigen presenting cells) in tumours, boosting the number of CTLs. This together have been reported to boost the response of anti-PD-1 and PDL1 drugs. This not only cleared the air about the previously observed heterogeneous and transient responses, but also established the impact of gut microbiome on cancer therapy.

**Faecal Microbiota Transplantation (FMT) and CRC Immunotherapy**
By definition, "FMT involves administration of faecal material (containing intestinal microbiota) from a healthy individual (donor) to treat dysbiosis and restore beneficial intestinal flora and phylogenic diversity.” FMT has proven to be beneficial in the treatment of recurrent Clostridium _difficile_ infection (CDI) and few other gastroenteric disorders.
Given that gut microbiome has such a significant on cancer therapy, it has recently found traction in cancer chemotherapeutic assistance. The transfer of highly complex whole communities of microorganisms from responders has been shown to result in durable changes in the recipient. FMT from colorectal patients into germ-free mice can elicit dysplasia and polyp formation. FMT from cancer patients responding to PD-1 based immunotherapies into germ free mice have shown increased efficacy. Non-responders to immune therapies were characterized by low abundance of Akkermansia _muciniphila_. Response to tumour therapy was improved by oral supplementation of the same bacterium.

**Conclusion**
The learning from these emerging studies, linking tumour development, progression, therapeutics and microbiome composition, is the fact that it is the microbiome which is key for developing personalized therapeutic approaches, since the microbiome composition of every individual is unique. At the same time, the shear complexity of the microbiome also adds several layers of complications to the precision medicine paradigm, but will no doubt drive the development of future therapeutics in oncology and other medical disciplines. The vision of utilizing a panel of bacterial marker species, that would designate an individual to be a responder or non-responder, is not too far from implementation, and will be checked before by clinicians to decide on personalized therapeutic approach. Overall, it is evident that the composition and diversity of the intestinal microbiota has significant implications in cancer treatments but will have to be thoroughly evaluated before implementation.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Gut Microbiome and Healthy Aging
Author: BugSpeaks
Published: 2019-03-08
Category: Microbiome and Lifestyle
Meta Title: Gut Microbiome and Healthy Aging
Meta Description: Ageing is an inescapable process. The health of an individual will inevitably deteriorate due to ageing and other physiological pr
URL: https://www.bugspeaks.com/blog/gut-microbiome-and-healthy-aging
Ageing is an inescapable process. The health of an individual will inevitably deteriorate due to ageing and other physiological processes. We cannot stop ageing, however in all probability, maintaining a healthy life and ensuring a “healthy ageing” is definitely in one’s hands.
World Health Organisation (WHO) defines healthy ageing “[_as the process of developing and maintaining the functional ability that enables wellbeing in older age_](https://www.who.int/ageing/healthy-ageing/en/)”. Functional ability is about having an optimal intrinsic capacity (mental and physical) and suitable environment (home, community and society). It is this intrinsic capacity that gets deteriorated over time with various internal and external factors, with immunological reasons being the most dominant one. Adding on to the abundant data that is already present, new reports have established strong connection between gut microbiome and human ageing and wellbeing.
**Microbiota and Immunosenescence**
As mentioned above, immunity and immunological reasons are the most dominant ones that hinder the process of healthy ageing. “Immunosenescence refers to the gradual deterioration of the immune system brought on by natural ageing process”. The ageing process is generally accompanied by a chronic (slow developing) background of inflammation, which is termed as “inflammageing”. A noteworthy negative outcome of [immunosenescence and inflammageing is characterized by a significant decrease in the functionality of immune system](https://www.ncbi.nlm.nih.gov/pubmed/28035338). This results in development of most age-related disorders, deteriorating intrinsic capability of individuals and significantly increasing the morbidity and mortality risk in the elderly.
_The human gut microbiota has a two-way interaction with processes of Immunosenescence._
On one hand, the microbial community in the gut (or the microbial burden) causing chronic, age-related inflammation is known to influence immunosenescence. While it is not conclusively proven that our gut microbiome is the root cause of inflammageing, there is a growing body of evidence to support this notion. The basic idea is that the gut microbiome is essential for normal immune function and development, and the immune system gets impaired by perturbations in the gut microbial ecosystem.
On the other hand, the process of immunosenescence has a significant impact on composition of gut microbiome, once it has reached a certain stage. This can cause troublesome changes in the composition of the gut microbiota, specifically in aged individuals. This usually results in the loss of bacterial diversity and a marked decline of beneficial microbial species during ageing, in turn leading to a compromised gut barrier, dysbiosis, increased bacterial infiltration into the body and eventually a vicious cycle of chronic inflammation.
Several studies on maturing gut microbiota have uncovered that the diversity of microbiota in elder individuals is significantly lower as compared to young individuals. It has been observed that a varied abundance of phylum Firmicutes and an overall low diversity in elderly subjects, is associated with increased health risks. [A distinct study](https://www.ncbi.nlm.nih.gov/pubmed/28035338) has reported significantly higher abundance of Bacteroidetes, and lower abundance of Clostridium cluster IV in elderly compared to younger subjects.
**Healthy Ageing through Gut Microbiome**
Since, the worldwide population is progressing towards a less dynamic, sedentary culture, it has become critical to prevent or delay age-related complications and maintain a good health. Many studies have explained that human wellbeing needs a valuable gut microbiota for physical and mental improvement at every age and alteration in physiological functions during the ageing process can affect the composition and functions of gut microbiota.
Hence, investigations have proposed that keeping up a decent gut microbiota through any means is critical for healthy ageing. But, the microbial community in the gut varies between individuals to a great extent and is dependent on eating habits, topography, host genetics, early microbial introduction and many other factors. Of all these, diet and nutrition are the most accessible factors that can be moderated by us, hence giving us control over our ageing.
**Nutrition, Microbiota and Ageing**
[Nutrition and diet play a significant role in shaping up of the host's gut microbiota](https://www.ncbi.nlm.nih.gov/pubmed/30025401). Nutrition is a key environmental factor that interacts with host genes, particularly in nutrient signalling pathways, and as recently reported, via gut microbiome. Hence, nutrition is a critical factor that can interlink gut microbiome with host genome.
Consuming high fibre or Microbiota Available Carbohydrates (MACs) containing diets or taking optimal mix and quantity of prebiotics species may help in [aversion and treatment of age-related pathophysiological conditions, which can aid healthy ageing](https://www.ncbi.nlm.nih.gov/pubmed/28035338) and advance life span. A high-fibre rich diet stimulates the gut microbiota towards performing different functions, as protection from inflammation, obesity, diabetes, heart disease, and high blood pressure. It has been reported that eating high-fibre content enhance the abundance of Bacteroidetes and decrease the abundance of Firmicutes. Studies also postulated that fibre-containing diets can help in developing the gut microbiota towards protecting the host from inflammation and non-infectious colonic diseases, due to the high abundance of _Faecalibacterium prausnitzii_ along with other Short Chain Fatty Acids (SCFAs) producing bacteria.
Various studies have suggested that butyrate, one of the products of SCFAs-producing bacteria, play several beneficial roles to improve the health in elderly. It has been hypothesized that the level of SCFAs, especially acetate, butyrate, and propionate, in the [gut of aged people is reduced compared to young subjects](https://www.ncbi.nlm.nih.gov/pubmed/27808595), and hence a diet that can aid production of SCFAs is critical. Nutrition and microbiome, together, also influences the host's epigenome; for instance, folate and choline, as dietary methyl-givers, can influence DNA methylation. A detailed account of various fibre rich foods and much more, that can aid in the process of building a healthy gut microbiome has been detailed [here](https://www.bugspeaks.com/blog-details?id=13&&title=Vegetables_and_Microbiome) and [here](https://www.bugspeaks.com/blog-details?id=14&&title=Fermented_Foods_&_Microbiome).

**Probiotics and Prebiotics in Ageing**
Consumption of [prebiotics](https://www.bugspeaks.com/blog-details?id=12&&title=On_Prebiotics) and [probiotics](https://www.bugspeaks.com/blog-details?id=15&&title=Kefir_-_Bridging_traditional_dietary_and_current_nutritional_practices) can augment the benefits of nutritious diet by multiple fold. Plant based diets, can elevate the abundance of beneficial species which is already present in your gut, or taken as probiotic supplement along with food. Prebiotics, which are also dietary substances that selectively promote proliferation and activity of beneficial bacteria indigenous to our gut, will also have similar effect on improving the composition and abundance of beneficial microbes. Other than these, numerous biomolecules, like micro-nutrients, fermented products, gut-related hormones and other compounds, are produced by these endogenous commensal organisms that are important in neurological and mental health of adults. [Together, they can promote growth, stimulate the immune system,](http://europepmc.org/abstract/med/28035338)[prevent gut-related and other diseases, which together is of critical importance for elder individuals](http://europepmc.org/abstract/med/28035338).
**Conclusion**
Overall, the strength of our immune system is supported by a healthy gut, which includes a complex interplay of symbiotic relationships between the gut microbiome and our body. Hence, developing, maintaining and sustaining a solid gut microbiome is the key factor to maintain a sound and healthy ageing, and this process is directly influenced by food and diet, probiotics and prebiotics. With [BugSpeaks®](https://bugspeaks.com), our endeavour is to provide you with a personalized, actionable and easily adoptable dietary, probiotic and prebiotic recommendation, which can aid you a long way through your ageing process.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Commensals and Cancers
Author: BugSpeaks
Published: 2019-02-28
Category: Microbiome and Disease
Meta Title: Commensals and Cancers
Meta Description: The last decade has witnessed a major surge in interest in the field of microbiology, which in turn has instilled a huge perce
URL: https://www.bugspeaks.com/blog/commensals-and-cancers
_The last decade has witnessed a major surge in interest in the field of microbiology, which in turn has instilled a huge perceptional change into the field of oncology (study of cancers)._
The surge in interest in the field of microbiology is due to the gut "commensals", the symbiotic microorganisms living with us. They have gained traction in the past decade for both their beneficial and detrimental influence on us, and significant impact on our health and wellbeing. This in turn has influenced a change in perception about cancers, both in its understanding and how to treat them. Even though, years of adopting molecular biology techniques, next-generation sequencing (NGS) technologies, whole genome sequencing and then multi-omics approach have aided us to understand cancer better than ever, the human microbiome and its influence on cancers have introduced a new paradigm to this understanding.
The link between microbiome and 'trigger, development & progress of cancer', and more recently even its treatment, has been well established. These established correlations indicate that the microbiome and cancer affect each other in a _“bidirectional and cyclical manner”_. On one end the cancer development changes microbiome composition while such changes also contribute to cancer progression in return, and this process continues in a cyclical manner. Furthermore, it’s also been established that the effect of microbiomes on cancers can both be detrimental (increase the pathogenesis of cancer and more) or beneficial (being protective against the triggers of cancer and more).
**Microbiome and Cancer trigger and progression**
Factors like gene mutations, family's genetic history, lifestyle habits (like smoking and alcohol consumption) etc., have long been studies and addressed with respect to cancers. Recently, there has been a rise in evidences associating involvement of microbiome (more specifically gut microbiome) in cancer. Initial studies revealed significant differences in the relative abundance of specific microbes in cancer cases compared with control subjects, which triggered a tirade of conversations and debates. Later, well-known associations between gut microbiota and inflammation & metabolism also got converged with its influence on cancer, expanding our understanding of contribution of microbiome in human cancer.

_Carcinogenesis is a process where tightly regulated systems of our body, such as immune activity and cellular metabolism, go off the track._ Either some perturbations in the microbiome or presence/absence of some specific microbial species/component or both can lead to such changes in cellular control.
These perturbations have been demonstrated to destabilize DNA integrity, deregulate signal transduction, modulate cell proliferation and even promote pro-tumorigenic (cancer inducing) immune responses. Some bacterial toxins can trigger senescent cells (cells that have stopped dividing) into proliferating cells by secreting growth factors. Specific microbial changes and its components have also been demonstrated to block tumour-killing capabilities of immune cells, enabling tumour progression and persistence even further.

Even though such activities have associated the role of microbiome with tumour development and have underlined several possible functional links, it is _still under debate if microbiome alterations can cause cancer or if they are merely associated with the disease_. But it has become critical to combine knowledge from _tumour-microbe-immune interactions_ with that of genomic, nutritional, pharmacological, social and behavioural effects of these interactions, for discovering and inventing next class of cancer treatments.
**Gut Microbiome and Colorectal Cancer**
Of all the cancers, human colorectal cancer (CRC) has been the one studied most, with maximum number of clinical studies and demonstrated associations between gut microbiota dysbiosis and the cancer. Specific bacterial species and strains have been associated with CRC, indicating inter-related links between gut microbiota, nutrition, immunity, tumour evolution and even tumour immunotherapy.

Fusobacterium _nucleatum_, Bacteroides _fragilis_ and Escherichia _coli_ were more abundant in worse prognosis groups, whereas the presence of Faecalibacterium _prausnitzii_ was positively associated with an improved survival. Of these, the most intriguing link between the gut microbiota and CRC has been associated with Fusobacterium _nucleatum_.
F. _nucleatum_ represents the best studied bacterium with a clear association with CRC. F. _nucleatum_ has been consistently found more abundant in faces from CRC patients, while its lower abundance has been associated with overall longer survival time. Even patients with simple adenomas exhibit an increased prevalence of F. _nucleatum_ in colonic tissue, while its persistence in metastatic colorectal tumour has been recorded.
Immunologically speaking, its abundance is inversely associated with CD3+ cell density in tumour, associated with serrated neoplasia pathway, increased expression both in high-grade dysplasia and established CRC. It may suppress the adaptive T cell response and may function in a way like the immune checkpoint in CRC. Quantitative PCR abundance of two gene markers (butyryl-CoA dehydrogenase from F. _nucleatum_) clearly separated the CRC microbiome form healthy controls. It is also been abundantly present with recurrence after chemotherapy and is known to promote resistance to chemotherapy.

Escherichia _Coli_ has the potential to cause intestinal inflammation via toxins such as colibactin, which also has oncogenic potential. This has been supported by various metagenomic studies in large CRC patient populations. Experiments in mice have demonstrated that host inflammation is essential for the cancer-promoting potential of E. _coli_. Mucosa-associated E. _coli_ is significantly more prevalent in CRC tissue, correlates with tumour stage and prognosis. Colibactin-expressing E. _coli_ is known to enhance tumour, accompanied by production of hepatocyte growth factor, overrepresented polyketide synthase pathogenicity island. Western diet affects enhances susceptibility towards the pathogenic potential of adherent invasive E. _coli_.
Bacteroides _fragilis_ is yet another human commensal found in the intestine of most humans. Bacteroides _fragilis_\-derived toxin (BFT) causes inflammatory diarrhoea and inflammation-related cancer. B. _fragilis_ toxin stimulates intestinal epithelial cell shedding and gammasecretase-dependent E-cadherin cleavage. Enterotoxigenic Bacteroides _fragilis_ (ETBF) induces colitis and colonic tumours in mice by secreting BFT. Dominated by inflammatory T cells and neutralization of IL-17 and IL-23 reduced inflammation and tumour formation. B. _fragilis_ colonization was paralleled by increased expression of TNF-gamma, ß-catenin, NF-kB, COX-2 and MMP9. Together with F. _nucleatum_ and E. _Coli_, B. _fragilis_ was more abundant in worse prognosis groups of CRC.
Klebsiella _pneumoniae_ may play a role in development of colorectal cancer. High prevalence in isolates from patients suffering from gastrointestinal diseases and CRC. Action through colibactin toxin and DNA strand breaks. Continuous insult of the colibactin toxin supplemented with the influx of pro-inflammatory cytokines. Colonic microenvironment leads to chronic inflammation. Repeated infection and insult of the colonic mucosa causes alteration in the structure and function. Stimulation of epithelial cell proliferation and thus colorectal cancer.

Other than these bacterial markers and associations, some metabolites can also be and have been used as diagnostic and prognostic markers, for its association with CRC.

**Microbiome and protection against cancer**
_On the opposite spectrum, the human microbiome has also been known to suppress tumour development and disease progression._
Like that of cancer trigger and progression, either some specific perturbations or presence/absence of specific microbial species/component suppress cancer and its progression. The mechanisms of suppression have been reported to range from augmentation dendritic cell function to increasing tumour-killing capabilities of cytotoxic T cells. But, more than these cellular mechanisms, it’s the metabolites generated by the microbiome which is known to play a major role in tumour suppression. For example, short-chain fatty acids (SCFAs) such as butyrate, generated by microbial fermentation of dietary fibres and resistant starches are extensively used as energy source by intestinal cells. However, tumour cells, which prefer glucose over butyrate, starve and suppress its growth or at least its progression at such low glucose conditions. Many such metabolites like butyrate (in gut) and others in skin has also been studied to reduce the incidence and multiplicity of tumours at various locations of the body.

Diet and nutritional habits have been strongly linked to CRC. This diet related risk is influenced by the balance between production of health-promoting microbial metabolites (ex. butyrate) and potentially carcinogenic metabolites (ex. secondary bile acids). Butyrate, a chemoprotective agent, acts as a histone deacetylase (HDAC) inhibitor capable of decreasing proliferation and increasing apoptosis in CRC cells. Butyrate could also diminish tumorigenesis by attenuating inflammation and colonocyte permeability and maintaining barrier function of the epithelium.
This relationship between the gut microbiota and CRC opens new approaches for cancer prevention. There are high quality experimental studies that yield the scientific evidences for the clinical use of probiotic in the prevention of CRC. Probiotic and prebiotic supplement can modify gut microbiota structure by reducing pathogenic gut microbiome and increasing probiotics.

A significantly decreased polyp number was also seen in mice given Lactobacillus rhamnosus GG (LGG). Consumption of LGG helped maintain the overall functional potential and taxonomic profile in the resident microbes. A 25% decrease of total polyp counts were observed. Further, LGG enriched those microbes or microbial activities related to short-chain fatty acid production (e.g. Roseburia and Coprococcus). Furthermore, suppressed the ones that can lead to inflammation (e.g. Bilophila _wadsworthia_).
Numerous studies have shown that iron significantly influences the intestinal microbiota, its composition and abundance. Of these, Bifidobacteriaceae are capable of binding large amounts of iron in the large intestine., thereby limiting the formation of free radicals synthesized in the presence of iron. This is known to reduce the risk of colorectal cancer significantly.

**To summarize**
It been irrevocably established that the human microbiome has a tremendous effect on the human illness and wellness. While the relationship between gut microbiome and its effect on various gastroenteric diseases were well known, its significant impact on cancer and its therapy has recently gained momentum. Dysbiosis of gut microbiome is one the best studied associations for the colorectal cancer, apart from its genetic and lifestyle related triggers.
Even though these associations are well established with various animal and human studies, our knowledge is still limited. This limitation comes from the fact that the impact of the microbiome on cancer is multifactorial, with a major uncertainty that if the dysbiosis of the microbiome is cause or an effect.

Even with these uncertainties, scientific community is optimistic that the next decade of research in the field of oncology will be dominated by an even greater integration of microbiome and its impact, which will drive the invention and discovery of novel microbiome derived therapeutics of cancer.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## A Letter from Liver
Author: BugSpeaks
Published: 2019-02-22
Category: Microbiome and Disease
Meta Title: A Letter from Liver
Meta Description: Hi, I am your happy happy liver. I am the filter of your body. I filter and detox your body f
URL: https://www.bugspeaks.com/blog/a-letter-from-liver
Hi, I am your happy happy liver.
I am the filter of your body. I filter and detox your body from harmful substances that come in and allow only those which are needed. So, I am pretty important to you.
Specifically, I am a major filter of nutrients absorbed by the intestines and I am also the filter of microbiota generated by-products, filtering in the beneficial ones and filtering out the deleterious ones. Me, I am exposed (read bombarded) with these bacterial components and metabolites on a regular basis. One of the perks (or perils) of being so close to the intestine.
_**I have a two-way or cyclical connection with the gut, and in context, specifically with its microbiota.**_
Me and the intestine communicate extensively through the biliary tract, portal vein and systemic mediators. So, as I said, this begins with intestine passing on the digested metabolites and toxins to me and I filter in or filter out. For instance, some components of gut microbiota called Pathogen Associated Molecular Patterns ([PAMPs](https://www.ncbi.nlm.nih.gov/pubmed/25492994)) and Microbial Associated Molecular Patterns (MAMPs) needs to constantly filtered in or out. Some of these are endotoxins (lipopolysaccharide or LPS) that are deleterious and hence filtered out, and some are bacterial metabolites (examples) that are beneficial, hence filtered in. On the other hand, many intestinal factors regulating the bile acid production, glucose and lipid metabolism in ‘yours truly’ (liver). And… the cycle continues with my (liver) products, such as, influencing the gut microbiota composition, intestinal integrity and much more.
Things get messy when the [gut microbiome dysbiosis](https://www.ncbi.nlm.nih.gov/pubmed/27802157) happens (as in variation in gut microbiome composition and abundance), along with multiple interactions with human’s immune system and other cell types. This dysbiosis can cause imbalance in our otherwise harmonious communications, which in turn can have some deleterious effects on me (Well… just like any other relationship).
_**So what does the relationship counsellor say..?**_
They say there is some research that have already reported and much research going on linking my disorders and conditions to gut microbiota. It is now widely accepted that the damage caused to me (liver) can result from [extensive interplay](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6319369/) between the gut microbiota produced specialized molecules (such as TMA, acetaldehyde and LPS) and the host immune system, via Kupffer-cell-mediated liver inflammation.
I know, I know
This is too technical
But, let me explain what I have understood, taking an example.
Incidence of Non-alcoholic fatty liver (NAFLD) is globally rising and it is a common multi factorial liver disease. This is the condition where, I become inflamed and huge, scarred and scaled, and I don’t like myself like this.
_**How is this caused?**_
Remember the endotoxins? the deleterious things I filter out?
Some initial disruption of the intestinal integrity increases the permeability of its walls that leads to increased bacterial translocation, which in turn increased that load of bacterial endotoxins that penetrate the portal vein (more than I can handle). These endotoxins are recognized by toll-like receptors (TLRs) on hepatocytes (my cells). When bacterial LPS signals through TLR4, signalling ultimately activates NF-κB and proinflammatory cytokines, and initiate a barrage of immunologic responses. Together, this increases the risk of [NAFLD](https://www.ncbi.nlm.nih.gov/pubmed/26449892) development through the activation of hepatic inflammatory cells (that’s me!).

_**So, intestine is the cause**_
_**Why are u still in this relationship then?**_
Well… as it almost always happens in a relationship, when one door closes, another opens. What seems to be cause for my demise, the gut microbiota, is in itself the cure.
Firstly, increasing reports of links between NAFLD and gut microbiome, both at observational and at mechanistic levels, have indicated them as an attractive source of biomarkers for early diagnosis of NAFLD.
So, the microbiota that cause my disease, are also used to identify if I will get the disease.
That’s a good thing.
In fact, serval gut microbiome-based tests are already available, which can understand what your [BugSpeaks®](https://bugspeaks.com/) about your health and provide you with risk estimations of diseases that you may be predisposed to. This can be used later to confirm the findings with physicians and clinicians, and even get personalized recommendations.
_**Now to the cure or alleviation or protection of my disease**_
A major beneficial role of the gut microbiota in liver disorders is also supported by [accumulating evidences](https://www.ncbi.nlm.nih.gov/pubmed/30227645) that several complications of severe liver disease, such as hepatic encephalopathy, are efficiently treated by various prebiotics and probiotics. Gut [microbiota manipulation](https://www.ncbi.nlm.nih.gov/pubmed/28298269) through nutritional factors, with [prebiotic potential](https://www.bugspeaks.com/blog-details?id=12&&title=On_Prebiotics), with additive [probiotics](https://www.bugspeaks.com/blog-details?id=15&&title=Kefir_-_Bridging_traditional_dietary_and_current_nutritional_practices), and supplementary polyphenols can alleviate the characteristics of NAFLD.
Of these, dietary factors are strong predictors of the gut microbiota composition. In fact, it has been projected that dietary factors play a more important role in shaping the gut microbiota composition than do genetic factors. Nutritional factors and the gut microbiota have bilateral interactions that contribute to the development and/or protection from NAFLD.
Most importantly, gut microbiota using prebiotic and fermentable carbohydrates as energy sources, have major beneficial impact on NFALD. Short Chain Fatty Acids (SCFAs), such as propionate, produced through this fermentation, cross the gut barrier and reach the liver through the portal vein blood. Propionate inhibits lipogenesis by acting on the transcription of several rate-limiting enzymes involved in _de novo_ lipogenesis. Consequently, these fibres and their metabolites are putative tools to reduce steatosis and inflammation.
_**Hence, I am saved. Phew!**__**!**_
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## How changing a body's microbes leads to illness
Author: BugSpeaks
Published: 2019-02-11
Category: Microbiome and Disease
Meta Title: How changing a body's microbes leads to illness
Meta Description: Claire M. Fraser, PhD, in this TED talk explains how microbes affect our lives. We are more obsessed with cleanliness than ever: we use antibacterial cleanse
URL: https://www.bugspeaks.com/blog/how-changing-a-body's-microbes-leads-to-illness
Claire M. Fraser, PhD, in this TED talk explains how microbes affect our lives. We are more obsessed with cleanliness than ever: we use antibacterial cleansers, we keep our children away from dirt, and we give out antibiotics with little regard for long-term effects. But in the process, we are altering our microbiota, the microbes that live on and inside us. And it's causing us to be more sick.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Diet, Digestion and Microbiome
Author: BugSpeaks
Published: 2019-02-11
Category: Diet and Supplements
Meta Title: Diet, Digestion and Microbiome
Meta Description: Professor Kevin Whelan talks about diet and the microbiome at Core's Exploring the Science of Digestion event at the Kia Oval, London. The human body comprises 10-100 trillions of microbial cells, including archaea, bacteria, viruses, eukaryotes. The micr
URL: https://www.bugspeaks.com/blog/the-human-gut-microbiome-and-microbiomics:-progress-towards-personalized-healthcare
The human body comprises 10-100 trillions of microbial cells, including archaea, bacteria, viruses, eukaryotes. The microbiome includes the combined genetic material of all microbes in a specific environment i.e., microbiota. It plays a significant role in improving human health and preventing diseases in many ways, though their mechanisms are yet to be probed in detail. The occurrence and function of microbiota differ depending on several factors, such as ages, sexes, races, diets, and change in the locations of the host. The microbiome profoundly restores normal phenotypes in the host. The past few years have investigated the microbiome in detail, and the results have tremendously reshaped our knowledge of human biology. Some of the innovative insights range from the understanding of how microbial cells intervene digestion activities, disease initiation, and progressions (e.g., inflammatory bowel disease) to unforeseen links with autism, depression, anxiety/mood disorders, Alzheimer's, Parkinson's disease, etc.
So far, studies have discovered around 10,000 microbial species (mainly bacteria) in the human body and mostly, their occurrence is predominant in the gut. Interestingly, the gut microbiota contains about 150 times more number of genes than that are observed in the whole human genome. Thus, the gut microbiome is sometimes recognized as an ‘essential organ’ because it produces products, it responds to the environment, and interacts with other systems. It is believed that the impact of these bacterial genomic DNA on our health might be more than our own genomic DNA. Likewise, it has been realized that the microbiota of gut and brain are intricately associated. There is an exchange of messengers/signals between the brain and gut microbiota. The entire body cells are influenced by the microbiome in one or the other way. In specific, it affects our mood, metabolism, libido, immune system, sensitivity, and even the clarity of your thoughts. Scientific evidences have shown that human health and diseases are dictated by what goes on in the microbiome of the gut. Diseases such as inflammatory bowel disease (IBD), obesity, diabetes, cardiovascular diseases, atherosclerosis, nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), cirrhosis, and colorectal and liver cancer have been connected to the activities of the human microbiota (Figure 1).

**Figure 1. Human microbial symbiosis has a close relationship with diseases of different systems (Adopted from Wang et al. 2017; [https://doi.org/10.1016/J.ENG.2017.01.008](https://doi.org/10.1016/J.ENG.2017.01.008)).**
Overall, the diversity in microbiome can lead to both health and illness. Commensal microbes colonize the host soon after their birth. This microbiota community gradually develops with the host growth depending on varying ecosystems. Over time, these host-microbe interactions lead to a beneficial association, and symbiotically offer many advantages. For example, improvements in digestion, supply of essential nutrients, metabolizing indigestible compounds, and to shield against the invading pathogens. Knowing about the microbiota can help in preventing possible diseases in humans. However, their complex nature makes it difficult to study them. The standard microbial culture methods may not be sufficient enough to isolate and explore all microbes present, because some may not grow or require special growth conditions that are yet to be standardized. Later, culture independent methods for evaluating microbes was developed based on the targeted sequencing of 16S and 5S ribosomal RNA (rRNA) genes which showed differences for each species and effective in identifying organisms. Over the last decade, human microbiome researches have been transformed by using next-generation sequencing (NGS) technologies. These high-throughput sequencing allow us to study complex bacterial systems without the requirement of cloning individual genes. The targeted sequencing of 16S rRNA, 18S rRNA, 28S rRNA genes, and internal transcribed spacer (ITS) regions, and whole microbial genome of microbial communities all at once (metagenomics) is very useful in the identification of microbes and to track the functional activity of the entire community. The increasing number of such metagenomic studies of microbiomes in humans is a new hope of its potential benefits in the clinical practice.
Recent studies have witnessed the occurrence of human genetic variations as influenced by interpersonal dissimilarities in microbiomes. Thus, human genes might influence health by promoting beneficial microbiomes. The heritability study of the gut microbiota has revealed that subsets of microorganism’s abundance are genetically determined by the host. For example, a genetic change enables lactase persistence in adulthood. Interestingly, the genome-wide association studies (GWASs) of the microbiome has revealed that there is a relationship between the host genotype, consumption of milk, and gut beneficial bacteria (Bifidobacteria). It has been noticed that a single nucleotide polymorphism (SNPs) near the LCT gene on chromosome 2 and Bifidobacterium in the fecal microbiota has a close association. Bifidobacteria utilizes the lactose of milk as an energy source. This depends on the genotype of the host and diet interaction (Figure 2).

**Figure 2: Interaction between the human lactase nonpersister genotype, Bifidobacterium abundance, and lactose in the host’s diet. Genetic variants near the LCT gene are associated with lactase persistence and in several microbiome GWASs were recently associated with Bifidobacterium relative abundance in fecal samples. The association may function according to the following scenario: (a) If an individual is a lactase persister and consumes lactose, the lactose is typically broken down into glucose and galactose in the small intestine by host lactase. (b) However, if the individual is a lactase nonpersister and consumes lactose, it travels to the large intestine, where it is fermented by lactose-utilizing bacteria, which includes Bifidobacterium. If Bifidobacteria are present, then the presence of lactose promotes their abundance. In individuals who do not consume lactose, Bifidobacterium abundance remains unaffected by their lactase persistence status (not shown). (Adopted from Goodrich et al. 2016; [https://doi.org/10.1016/j.cell.2014.06.037](https://doi.org/10.1016/j.cell.2014.06.037)).**
Likewise, GWASs has revealed that immunity related genes are influenced by the gut microbiome. Evidences suggest that genetic variations might act on digestive tract tissues to disturb the composition of the gut microbiome. Further, genetic variants in the genes, PLD1 and LINGO2 are associated with obesity. Based on the analysis of 16S transcripts, the phylogeny of microbial community has been revealed. The most active microbiota included Prevotellaceae, Lachnospiraceae, Bacteroidaceae, Ruminococcaceae, and Rickenellaceae. RNAseq analysis of bacteriophage populations in the periodontal microbiota showed the abundance of oral phages highly expressed in persons having good periodontal health. More recently, 16S rRNA gene-based approach has confirmed the predominant occurrence of Corynebacterium spp., Staphylococcus spp. and Propionibacterium spp. on healthy human skin.
Thus, knowledge on the microbiomics help to improve human health conditions. A defined diet can influence on the microbiome and restore health. Likewise, a proper use of chemicals/drugs in disease management is another factor to be considered. For example, both foods and drugs are used in treating type 2 diabetes. It is generally advised to avoid consuming simple carbohydrates to control blood sugar. Nevertheless, there exist inter-individual glycemic variations in response to diet, and some of which can be described on the basis of the individual’s microbiome. Similarly, the human microbiome plays a significant role in genetic diversity, modify disease conditions, impart immunity, provide nutrients, influences assimilation, and modulates drug interactions. Thus, intake of probiotics or beneficial bacteria (Lactobacillus, Bifidobacterium, Faecalibacterium prausnitzii, Akkermansia muciniphila, Pediococcus pentosaceus, etc.) may be useful in preventing or treating certain diseases, though most of them cannot be cultured presently. As a diagnostic tool, microbiomics is employed for detecting and measuring the levels of biomarkers i.e., cholesterol, cortisol, glucose, and creatine kinease occurring in blood which are known to indicate about diseases or infections. These novel diagnostic methods will allow clinicians to prescribe right drugs or diet to restore patient’s health.
Future microbiome investigations should involve many more new techniques to predict the functions of microbiotas, their crucial role in human development, and the mechanisms of various diseases, such as metabolic diseases, liver diseases, cancers, psychiatric diseases, etc. In this regard, many new-startups and biopharmaceutical companies, including Luecine Rich Bio Pvt. Ltd are exploring the field of microbiomics to develop novel therapeutics, diagnostic platforms and to provide clinical genomic services. A detailed data on this metagenomics might be very useful in the microbiome-based diagnosis of diseases and for the development of novel treatment strategies in the forthcoming personalized medicine era.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## BugSpeaks through Cladogram
Author: BugSpeaks
Published: 2019-02-04
Category: On Diversity Debate
Meta Title: BugSpeaks through Cladogram
Meta Description: Charles Darwin, with this famous entry in his journal, begins with "I think" on the top left-hand corner of his notebook
URL: https://www.bugspeaks.com/blog/bugspeaks-through-cladogram
Charles Darwin, with this famous entry in his journal, begins with "I think" on the top left-hand corner of his notebook and then sketched out the first “tree of life”, which was his perception of how all living things on earth are connected through evolutionary history. Through this he proposed the theory of "the origin of species through natural selection" and divergence of all species from an ancestral population.

In short, he meant we all are related!!
That doesn't mean only us humans. But every living being on earth can trace its descendance back to a common ancestor.
To understand this, let's begin with basic taxonomic classification, which we all studied during our biology 101 classes. By definition, _“Taxonomy is the science of defining and naming groups of biological organisms based on shared characteristics.”_ Organisms are grouped together into groups termed as ‘taxa’, and these groups are given a taxonomic rank, which looks something like this
**Kingdom => Phylum => Class => Order => Family => Genus => Species**
Now, when you want to show these relationships among organisms in a simplified way, the phylogenetic tree comes into picture, through which relationships are illustrated by a phylogenetic tree/dendrogram.
**Phylogenetic trees**
represent hypotheses about the evolutionary relationships (the descendance) among a group of organisms, originating from a common ancestor and can be built using morphological (body shape), biochemical, behavioural, or molecular characteristics of species or other groups. Dendrograms are often used to illustrate such evolutionary relationships, along with information like _evolutionary time line_ separating the taxa (like, how many million year ago did this species come into being), _genetic distance_ (between the gene sequences being classified), and so on.
**A cladogram** (from Greek, clados "branch" and gramma "drawing") is a type of phylogenetic tree that only shows tree topology. Basically, a cladogram uses lines that branch off in different directions, representing a group of organisms with a last common ancestor, ending at a clade. The branching pattern is called the tree’s topology.

There are many shapes and forms of cladograms, through which the phylogenies are represented, but they all have lines that branch off from other, and the lines can be traced back to where they branched off.
By topology, it also means showing only the basic relationships between the taxa, without any additional details. For example, say we want to represent human evolution through a cladogram, then we represent chimpanzees to be more closely related to us than gorillas. But this is represented just as a tree, without the time line (unlike the pictorial representation besides).

Now let's take a closer look at the cladogram, where we will zoom-in on one clade within the cladogram. Always, the species (taxa) are found at the tips (represented by B, F and C), extended by lines referred to as the tree's “**branches”.** The pattern in which the branches connect represents our understanding of how the species in the tree evolved from a series of common ancestors. Each node represents a **divergence** event or splitting of one group into two descendant groups.

Take a look at this amazing [candy bars](https://wildlifesnpits.wordpress.com/2014/03/22/understanding-phylogenies-terminology/) phylogeny that shows the relatedness between candy bars represented as a cladogram. This draws an accurate analogy to understand the basic evolutionary relatedness of species (and has the added advantage of candy bars being easily accessible (unlike animals) and easy to understand the phylogenetic tree structure even by a layman).

These candy bars are related to each other based on their content and shape. In a similar way species can also be compared and represented based on their genetic content (DNA/RNA) and their morphological characteristics. If we dig deep, the peanut clade has three candy bars or taxas (representative of species) with peanut as its common ingredient (representative of common ancestor). With such analogies we can understand how phylogenies are connected and important to understand the evolutionary insights.
**Cladograms and Microbiota**
Mainly phylogenies are used to represent species evolution, as in of higher organisms like humans, fishes and tress and so on. But there are also other applications like, plotting microbial taxonomic compositions of metagenomic data, which is the base of our [BugSpeaks](https://bugspeaks.com/)® analysis. In the specific context of microbial genomics and metagenomics, next-generation sequencing data, of say gut microbiome, produces datasets of unprecedented size and tremendous genetic diversity of microorganisms. And cladogram seems to be the simplest way of displaying these diverse phylogenies with thousands of microbial taxa within the sample.
Since cladograms are simple topology-based representations, they are simpler to understand by the end users and does not complicate the data with too much scientific information. However, we have also made specific modifications to these representations, to not just represent microbial composition, but also to display it’s metadata like species abundances, host or environmental phenotypes etc. With the end user in mind, just this simple cladogram can present the overall microbial diversity and other relevant information, yet also provide specific scientific data (like alpha diversity) of the sample, if the same representation is viewed by a researcher or a clinician with microbiome background.
Let’s try to read one such annotated cladogram representing microbial composition of a human gut sample.

This is a circular cladogram with basic visualization of the tree’s hierarchical structure. It has all the same representations as nodes, branches and taxas that were discussed above. It has four major clades (kingdoms) starting from centre, including Archaea, Bacteria, Eukaryota and Viruses presented in four different colours. Each has branches which represents next levels (Linnaean ranks), including Phylum, Class, Order, Family, Genus and finally the tip representing species (this cladogram image shows family level phylogeny). The size of circles at nodes and tips represents the abundance of rank and outer ring represents the species abundance in the form of bar plot (grey bars on the outermost circle). Highly abundant taxas are annotated as legends at top left corner.
By providing such a cladogram, we give the overall picture of an individual’s microbial diversity in his/her gut. Supplementing this with other plots and graphs, we provide a thorough summary of one’s [BugSpeaks](https://bugspeaks.com/)® profile, all represented through pictures.
Coming a full circle, what Darwin sketched as the simplest idea of tree of life, is still relevant and applicable even after a century, which is only a testament to his genius.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Diabetes and Dysbiosis: A tale of two Diabetics
Author: BugSpeaks
Published: 2019-01-29
Category: Microbiome and Lifestyle
Meta Title: Diabetes and Dysbiosis: A tale of two Diabetics
Meta Description: Below are some ‘Keyword’ searches for ‘diabetes’ being googled daily by millions of people across the glob
URL: https://www.bugspeaks.com/blog/diabetes-and-dysbiosis:-a-tale-of-two-diabetics
Below are some ‘Keyword’ searches for ‘diabetes’ being googled daily by millions of people across the globe.

And it is for a reason…
Diabetes Mellitus Type 1 & 2 or largely known just as ‘Diabetes’, is a rampant chronic (slow developing) disease around the globe. It’s also one of the most searched because of increasing number of risk factors for diabetes and diabetes being the risk factor for many other diseases. It has significant effect on body and mind of individuals and greatly reduces their quality of life.
Coming back to searches, since we all believe Dr Google so much, and almost always end up searching (and believing) anything and everything about the disease we are diagnosed with. But we also know that most of us won’t succeed in even understanding the disease, let alone managing it well.
And there begins a tale of two diabetics….
**Meet Mr A**
An everyday common man, taking his car to work all the way across the city. The to and fro daily commute nearly leaves him drained. His 8 to 8 job makes him too tired to get up early and hit the gym. His desk job is convenient and is just an exercise of the mind. His weekends usually pass by doing house chores and shopping, a movie, may be a resort spa outing, some relaxation and back to Monday blues. His life is great and the biggest problem he currently faces is slow moving traffic and being conscious about his stomach paunch at 32.
As life goes by, he begins to feel tired all the time, fatigue becomes a daily thing, hungry in between meals, heightened craving for sweets, chips and snacks, too drowsy usually an hour before his lunch, forced to have early lunches and so on. This goes on for few months to about a year.
On a seemingly normal day, a routine check-up and blood test at the company (because he is too busy being busy for these check-ups to be a regular part of his life), it’s revealed that his blood sugar is off the charts, and he is diabetic.
This rings a bell in his head, and he thinks, now is the time to change and turn my life forever. And the first thing he does… search (not read) anything and everything about diabetes on the internet.
A series of statistics pops up, stating diabetes is so rampant, with close to 422 million people being affected worldwide. With the first few searches on how to fix it, he gets that his lifestyle needs some serious changes and that too immediately. No junk food, balanced diet and vigorous exercise; the whole package. He thinks, may be walking and working out at a gym everyday should become part of his routine.

Then he digs a bit further, and starts to read about glycaemic index, and ...
what if the disease was diagnosed late?
what if it’s progressed far?
what if, if I can’t get my sugar under control, even with all the new diet and exercise routine I am planning.
Oh my god, my insulin production will get depleted at an alarming rate
I might have to take insulin intravenously to supplement my glucose metabolism
my quality of life will dangerously be deteriorated, and my family & friends will be concerned too much
Oh, this search also says I might get other disorders, liver problems and heart diseases, my kidney function could get affected forever, with increased risk for glaucoma
Oh damn, here it says if I get a cut on my leg, the wound will never get healed, it will get infected, and the doctors will have to amputate my leg
Oh god… Oh god… what will I do now
And… this downward whirl of “what-ifs” goes on for a while, and he gets so worried… so worried… that…
….he goes to sleep.
Next day morning, he goes back to his “so called regular life”.
**Now, meet Mr B**
Mr B on the other hand, having a similar lifestyle was also diagnosed with diabetes a while ago. He too, like most others, searched the heck out of google for all possible causes of and cures for diabetes. But, being a sensible man he is, without self-instigating much of panic or pressure, he starts to gather relevant information from credible sources, like doctors, clinicians and nutritionists. He too gets to know about glycaemic index and glaucoma, lifestyle and dietary changes that he needs to adopt, and most of the other information that Mr A got, but with actionable recommendations that are largely easy to implement in his lifestyle.
He also understands that, the long-standing notion that diabetes is only caused due to genetic factors or is largely lifestyle related (unhealthy diet, overweight, physical inactivity etc.), has been recently questioned. With the new and rapidly developing field of microbiome research, several new links have been established between [gut microbiota](https://www.gutmicrobiotaforhealth.com/en/digging-into-the-hidden-world-of-the-microbiome/) and diabetes, not only to its causation but also to the drug efficacy, alternate forms of treatments and many more.
And suddenly an interesting phrase catches his attention
_Local communities, Global Impact_
He reads again,
_Local communities, Global Impact_
He realizes, for the first time, that there are trillions of tiny tenants living within his gut, forming several local communities, have a global impact on our body and mind, and have been known to play significant role in disease and health.
Now he is intrigued, and his interest is peaked…
And he continues to understand more.
The gut microbiota affects numerous biological functions throughout the body and its characterisation has become a major research area in biomedicine. Studying the relationship between microbial alterations within the gut and functional repercussions linked to disease(s) remains challenging, and even more complex when it comes to their interactions with therapies and drugs. Animal studies and other human trials have associated microbial patterns in individuals with diseases and traits, thereby implicating gastrointestinal microbial communities in a range of human disorders.
Oh… all this can happen from my gut? Inside out?
The complex relationship between [obesity](https://www.ncbi.nlm.nih.gov/pubmed/20876708), diabetes, and gut microbiota is being uncovered, with consistent growing body of evidence. Presence/absence, relative and/or absolute abundance of specific microbes within the gut has been shown to have a direct impact on insulin sensitivity and glucose metabolism, adding an important dimension to understanding of diabetes.

Now this is way too technical for me…
He quips!!
But he continues…
Then came a bunch of biological names like Collinsella, Oscillospira, Akkermansia (most of which he could not pronounce), followed by some more bio jargon about pathways and stuff. All he could get from the picture besides was the burger and high-fat diet. He also gets fascinated by realizing that [metformin](https://www.ncbi.nlm.nih.gov/pubmed/26633628), which is one of the most common medication given to manage diabetes (which was also prescribed to him), not only helps to reduce the amount of sugar the liver releases into the blood but could also aid by improving the gut microbiome.
The most relatable thing that he grasps, by these searches and readings, was the extensive information about probiotics. It seems like [probiotics](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4402713/) can help a whole host of gut and other issues. Even though it is not proven to have absolute beneficial effect on diabetics, efficacy of probiotics in diabetes has been proven by their ability to lower fasting glucose and insulin levels in various. The link between gut dysbiosis and diabetes has been convincing and promising, particularly since probiotics have been shown to normalize the disturbed metabolism in diabetes patients.
he feels a sense of optimism...
but with a disclaimer that,
It’s hard to rule out the effect of other factors that may have affected glycaemic control in these [studies](https://www.nature.com/articles/nm.4345), yet, this link between probiotics and alleviated diabetes is a promising new supplementary to metformin treatment, in lowering fasting blood sugar.
…and regarding probiotics, his take home message was that, probiotics undoubtedly have an overall positive outcome on diabetics, and its can greatly supplement the main treatment options to manage diabetes well for a long time.
Overall, he got the gist of it.
That,
His gut microbiome and gut health can greatly influence diabetes and by understanding and managing his gut well, he can manage his diabetes too.
Now, he took Google’s help (the right way this time around) to find a [microbiome-based company](https://bugspeaks.com/) providing testing services and evidence-based interpretation of gut microbiome, which can help him better understand and guide him to make optimal effective changes to his health and lifestyle. He sent his stool sample to the testing facility, followed up well, and received an extensive report, including his disease susceptibilities (diabetes and others), as set of customized dietary, probiotic and dietary recommendations, follow up with nutritionists and clinicians for further advice, overall a complete package.

Having diagnosed early and his sensibility to understand and rely on credible sources, and to explore new avenues of diagnosis and treatment, aided Mr B with effective management strategies for diabetes. He inculcated all the necessary exercise and diet much early into his diagnosis, he took the help of nutritionists and turned around his life for the better, supplementing it with exercise and improved lifestyle. Now, that’s what we call a happy ending!

---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Obesity is Contagious
Author: BugSpeaks
Published: 2019-01-25
Category: Microbiome and Lifestyle
Meta Title: Obesity is Contagious
Meta Description: Let’s begin with this 140 So, it’s been two months past the new year. We all are coming back to our normal selves, we are eating (binging) whatever we
URL: https://www.bugspeaks.com/blog/what-happens-in-vagus,-does-not-stay-in-vagus
So, it’s been two months past the new year. We all are coming back to our normal selves, we are eating (binging) whatever we want to, not so keen on working out or even waking up early...for that matter.
Now that we are enlightened about new year diets, workout regimes & resolutions, and have understood these to be just myths, we can actually focus on doing what we do best
"**sit and ponder about everything**"
Everything, from menial questions about career, marriage, relationships ... to ... more serious questions like - should I cook today or order pizza?... then to ... potentially life changing questions like... Why am I fat ?!!
We always talk about stuff that we eat going directly to our hips (especially for us girls, right?!)
Here is an interesting observation:
What if, the food we eat directly goes to (or affects) our head?
hmmm... Now that's a thing to ponder upon,
I mean how's that even possible?
Well, you might have heard or even felt that, when you are in stress you eat more, or when you are tensed you have gastric problems, or when you are hungry for long duration you get a headache. So, you know these things are somehow connected, and mostly seem to be related to the food that you eat.
But, in fact, this relationship, between your behavior and your food (more importantly the other way around), is surviving (read hanging in there) because of the tiny tenants in your body.
_**And this is where the story begins...**_
When you are in your mother's womb there are no microorganisms in your body, and you are virtually sterile, inside out!!
As a new-born, you are exposed, for the first time, to a wide array of microbes from a variety of sources, including maternal bacteria. The process of birth exposes you to the first microbes from your mother's vaginal tract or stomach, depending on the mode of birth. (fascinating? or you feeling eeeww?)
Later, your mother's milk gives you the major share of microbes, that actually help you build your immunity. In fact, [a recent study](https://www.ncbi.nlm.nih.gov/pubmed/23401408) talks about how infants born by cesarean section are at increased risk of asthma, obesity and Type 1 diabetes, as compared to babies born through normal labor and exposed to the maternal vaginal microbes.
As and when you start eating different kinds of foods, drinking beverages, use a specific kind of bathing soap etc., the microbiome (total microbes on and) in your body gets enriched. More specifically, your gut microbiota gets developed with every exposure to different environments. In fact, what you eat, drink, smoke, or gets exposed to, all defines the microbiota of your gut.

I still remember the day, when I had my first beer... and my gut microbes were like... bring it on girl!!
Another interesting fact is that, since diet and food habits are largely an extension of your culture, your gut microbiome differs from American's or Chinese, given that we all are culturally world apart, and so is our lifestyle and dietary habits.
_**The other side of the story…**_
On one end, the food you eat and drink affects the composition of your gut microbiota. However, over time, the enriched and developed gut microbiota influence the digestion of specific food you eat, indirectly influencing your diet. So, lot of these gut bacteria influence and more importantly help you digest food in an efficient way.
Another interesting fact is that, whenever these good bacteria goes low in number, it gives way for over growth of bad microbes like Clostridium, E. coli etc., that causes diseases like diarrhea, colitis, etc.
And why would these good bacteria go low in number?
Well... it can be due to any reason from imbalanced or untimely diet to exposure to antibiotics or smoking. [One study found](https://www.ncbi.nlm.nih.gov/pubmed/3846592) that after a single treatment of intravenous antibiotics, fecal bacteria demonstrated a significant change in the variety and abundance of bacterial strains.
Overall, the food that we eat influence the gut microbiota and the microbiota influence the what we eat - requiring a fine balance between the two.
Okay, now we are drifting
_**Let's come back to the point where food and head are connected**_
Studies, involving college kids (the junk eating generation), eating burgers and fries, guzzling cokes and sodas, observed that these students felt lethargic, largely unwell, depressed, with general fatigue, disturbed / unfocused mind, just after a few weeks of continuous ingestion of the junk diet. Testing their gut revealed that, and I quote “gut microbes (the microbiome) had been devastated”, with potential disease causing firmicutes replacing good Bacteroidetes, as the dominant type, while the friendly Bifidobacteria, that suppress various kinds of inflammation, halved in its abundance being the clearest marker of an unhealthy gut.
The question is why? and How does what you eat .... affecting your gut microbes... affect your state of mind?
The human gut microbiome is so dynamic and complex, that it consists of approximately 1 kg of bacteria in an average adult, and is approximately the weight of the human brain. This microbiota produce a myriad of neuroactive compounds, regulating everything from your behavior to cognitive function, social interaction to stress management. It regulates these brain functions by steering the "Vagus Nerve".
This thing called 'Vagus nerve' (Not "Vegas and Casinos") within your nervous system, is a part of cranial nerves unit, that forms an extensive network, linking your gut and brain. Creatively enough, termed as [the gut brain axis](https://fivethirtyeight.com/features/gut-week-gut-brain-axis-can-fixing-my-stomach-fix-me/), it is the sole reason for all the food and head related things. This system, largely involving the Vagus nerve, controls the body’s unconscious actions, such as digestion, excretion, and sexual arousal.

In the absence of certain (good) microbes, due to imbalanced diet (read junk eating), the neuroactive compounds, that regulate the above said unconscious actions, are profoundly altered. It turns out, what you eat changes the gut ecosystem, in turn resulting in altered vagal activity.
_**But, what happens in Vagus does not stay in Vagus!!**_
The altered vagal activity leads to a lot of intermittent chain reactions, including decreased pancreatic enzyme secretion, poor gallbladder function, overall disturbed gut function. This suppresses the intestinal immune system, decreases intestinal blood flow, increases intestinal permeability, eventually leading to leaky gut, causing digestive problems, inflammation and infection, further aggravating the issues.
The cytokines produced during this imbalance, cross the blood-brain barrier, leading to decreased activity in brain, decreased activation of the vagal motor nuclei, inflammation and decrease in nerve conductance. The effects being anything from mild headaches to aberrant stress to long term depression. A causal association, between gut microbes and cognitive decline and disorders of cognitive function such as Alzheimer's disease and multi-infarct dementia (high prevalence after the age of 65 years), is already been studied.
Wait a minute, before, you thought, when you are in stress you eat more, or when you are tensed you have gastric problems, or when you are hungry for long duration you get a headache. Now you understand that these things are connected, but, possibly in the opposite order, where the food you eat cause the stress, gastric issues and headache.

So, coming back the full circle, to where the story began...
The gut microbes (or the whole microbiome) play a very important role in your growth, wellbeing and longevity, from your birth to senility. In fact, it has been established that post-birth, the gut microbial colonization occurs in parallel with cognitive development. There is [increasing evidence](https://www.ncbi.nlm.nih.gov/pubmed/20966022) to support the view that the evolving cognitive activity is critically dependent on the microbiota and its metabolic activity.

_**Now that’s a lot of pondering!!**_
(as I said, that’s what we do best, right?)
So, the next time you think about a diet plan (especially the new year ones), or any resolution for that matter, [listen to your gut](https://bugspeaks.com/) and gradually change and adapt, without hitting the Vagus.
Or else, come and read this blog, again, next year!!
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Anxiety, Depression & Gut Microbiome
Author: BugSpeaks
Published: 2019-01-21
Category: Microbiome and Lifestyle
Meta Title: Anxiety, Depression & Gut Microbiome
Meta Description: Have you ever felt butterflies in your stomach, trembling hands and voice, sweaty palms, racing heart beat and heavy breathing&hel
URL: https://www.bugspeaks.com/blog/anxiety,-depression-&-gut-microbiome
Have you ever felt butterflies in your stomach, trembling hands and voice, sweaty palms, racing heart beat and heavy breathing… and you feel just fine briefly after? sounds familiar?
At some point in our lives, everyone has experienced this; when teacher asked us to solve a math problem in front of the whole class, may be while performing on stage or while talking in front of a large crowd. These manifestations mostly arise out of dreaded situations and these outcomes you feel is cumulatively called as “Anxiety” and specifically these are temporary in nature and hence termed as “Occasional Anxiety”.
But, if you scan through scientific literature and articles, anxiety is almost always coupled with depression, and commonly used together.
Anxiety is a type of emotion or feeling wherein [hypervigilance, attentional bias](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4211931/) and nervousness conquer the person. Whereas, depression is an observable trait, significantly affecting daily routines and moods of the individual, how they feel and think and even have some long-term effects. Like occasional anxiety, temporary episodes or moderate intensity of depression are termed as “Temporary Depression”. But care should be taken not to confuse this with ‘Anxiety and Depressive Disorders’, whose symptoms can persist for longer time with greater tendency to become a full-blown disorder.
Until recently, all these mental health and neurological disorders, specifically Anxiety and Depressive Disorders, have largely been understood in a unidirectional way as a sole function of brain and its nervous system. But imagine something sneaky at play here, something subtler than brain, yet having a greater impact than brain.
_Enter “Gut Microbiota”_
**The old friends and the Gut Brain Axis**
Rook et. al. proposed the ["old friends hypothesis](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5102282/)”, as an extension of the famous [hygiene hypothesis,](https://en.wikipedia.org/wiki/Hygiene_hypothesis) which states that we humans have evolved with a bunch of friendly and non-friendly microorganisms, since thousands of years (as species) and from our birth (as individuals). This is specifically true with respect to our guts, where these microorganisms have thrived as an entire ecosystem with continuous availability of nutrition. Current estimates indicate that the _human gut is occupied with approximately 100 trillion cells_ of different microorganisms, happily living within us.
The same _GI tract is also intertwined with ~600 million nerves of ENS_ (Enteric Nervous System), which works along with CNS (Central Nervous System) bidirectionally, controlling various digestive functions. Until recently, this bidirectional crosstalk, _known as "Gut Brain Axis" (GBA)_, was thought to maintain only the gastrointestinal homeostasis, some immune activation, enteric reflux and enteroendocrine signalling. A detailed account of this complex nervous system has been described in one of our other blogs “[The Brain in Your Gut](https://www.bugspeaks.com/blog-details?id=44&&title=The_brain_in_your_gut)”
However, peaked interest in the gut microbiota and its sheer number in the gut (I mean, a 100 trillion is a lot!!), forced us to view the function of GBA under a different light. Which indeed led to our understanding that GBA is indulged in monitoring and connecting the emotional and cognitive parts of human brain to peripheral gut. Which in simpler terms says that “_there is a new meaning for gut feeling_”. (Know more [here](https://www.bugspeaks.com/blog-details?id=46&&title=Microbiome_and_the_Brain_Interactions) and [here](https://www.bugspeaks.com/blog-details?id=19&&title=What_happens_in_Vagus,_does_not_stay_in_Vagus!!))
_So, What’s the take home message here?_
_A 100 trillion microbial cells are constantly interacting with 600 million neurons, communicating and controlling everything from when you feel hungry to why you feel angry!!_
It is critical to note that this is a two-way interface, and the interplay can happen from “_microbiome-to-brain_” or “_from brain-to-microbiome_”, which was a key aspect that was ignored before.
[**Brain to Microbiome – HPA Axis**](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5987167/)
In its simplest form, it is _a stress response_.
[Interleukin-6 (IL-6) is a component of immune system that gets released due to stress](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2978751/), which reduces the expression of glucocorticoid receptors and over activates the hypothalamic pituitary adrenal (HPA) axis (a kind of brain and hormonal interaction). This ends up in over activating the inflammatory reactions leading to increased pro-inflammatory cytokines and stress-induced cortisone. Increased level of cortisone is a risk factor or is strongly associated with anxiety.
On the other hand, same stress can change your gut microbiota (composition and abundance).
Several intestinal microbes are capable of reducing anxiety and are largely considered beneficial to us. Predominantly, [_Lactobacillus rhamnosus_ and few more from the genus _Lactobacillus and Bifidobacterium,_](https://www.progressnp.com/article/psychobiotics-bacterial-hope-depression/) are specifically capable of this function, through their anti-inflammatory and anti-oxidant actions. These beneficial bacteria can negate the inflammatory effects caused by IL6 and in turn reduce anxiety. However, the same stress can reduce the abundance of such beneficial bacteria, in turn increasing the risk for anxiety.

**Microbiome to Brain – The [Leaky Gut](https://www.ncbi.nlm.nih.gov/pubmed/18283240)**
Stress can also change your microbiota (composition and abundance), activating other components of this complex system, “_inducing_” anxiety and/or depression.
Some intestinal bacteria produce toxins, known as [Lipopolysaccharide (LPS)](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5987167/), which disturb the intestinal environment and its permeability. The disruption of the intestinal barrier can lead to entry of these toxins in the blood, a phenomenon popularly known as “_leaky gut_”. Later, LPS can disrupt the blood-brain barrier (which is very hard to do), enter brain and modulate mood and behaviour by increasing the activity of amygdala in the brain.
If unchecked, this also allows for entry of other environmental toxins and harmful agents, which increases the pro-inflammatory cytokines and neurotransmitter norepinephrine, and many other factors, leading to development of depression.
Several such behavioural changes, with direct or indirect involvement of gut microbiota, has been reported and studied till date, some of which are detailed [here](https://www.bugspeaks.com/blog-details?id=20&&title=The_gut_microbiome_influence_the_function_of_brain_and_behaviors_in_humans) and [here](https://www.bugspeaks.com/blog-details?id=54&&title=Mind_altering_microbes).
**Psychobiotics**
Now that we know that this is a two-way streak and changes in gut microbiome can affect brain and its functions, several combinations of probiotics and prebiotics, either as solitary or as combined supplementations, has been tested, under the collective name of “[Psychobiotics](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5102282/)”. The ultimate goal is either to manipulate or to naturally have a gut microbiota, enriched with commensals that can maintain healthy neural, behavioural, cognitive and emotional functions.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Kefir - Bridging traditional dietary and current nutritional practices
Author: BugSpeaks
Published: 2019-01-18
Category: Diet and Supplements
Meta Title: Kefir - Bridging traditional dietary and current nutritional practices
Meta Description: Kefir is a friendly probiotic drink with slightly fizzy and tangy taste, a FANTASTIC fermented milk beverage originating from East
URL: https://www.bugspeaks.com/blog/kefir---bridging-traditional-dietary-and-current-nutritional-practices
Kefir is a friendly probiotic drink with slightly fizzy and tangy taste, a FANTASTIC fermented milk beverage originating from Eastern Europe and Southwest Asia. It looks like and tastes like thin yogurt, but far superior than others, as a probiotic. It is incredibly beneficial for digestion and gut health maintenance when consumed regularly. It is loaded with valuable vitamins and minerals and contains easily digestible complete proteins. People with lactose intolerance can eat kefir without problems, adding to its list of benefits.

Kefir can be prepared from any type of milk, cow, goat or sheep, coconut, rice or soy. It is made by adding kefir “grains” to milk. These are not grains in the conventional sense, but cultures of yeast and lactic acid bacteria. Over a period of 24 hours’ time, the microorganisms in the kefir grains multiply and ferment the sugars in the milk, turning it into kefir.
Even though Kefir has been widely recognized as a health drink with benefits, recent development in the field of probiotics, have raised enormous interest in Kefir, with researchers reporting a wide spectrum of progressive elements present in it benefits the person who is consuming it, as it is known for its anticarcinogenic, anti-inflammatory, and antipathogenic effects.
Unfortunately, misuse of the term probiotic has also become a common place and a major issue, with many products exploiting the term without meeting the requisite criteria. Consequently, there is a certain level of apprehension about these effects of Kefir, bringing it under the scrutiny. Can Kefir really show these effects in human, as most of the studies are done on mice and cell lines? If so then, what is the mechanism? What is the uniqueness of Kefir in terms of its bacterial composition?
Initially, some nutritional benchmarking has clearly identified the presence of beneficial components within Kefir (see picture below). “[Antimicrobial and healing activity of kefir and kefiran extract](https://www.ncbi.nlm.nih.gov/pubmed/15848295)” has also be studied early on, demonstrating highest activity against Streptococcus _pyogenes._ Further, Lactobacilli have been recognized as a major component of Kefir, which have GRAS (Generally Regarded As Safe) status. Of these, Lactobacillus _kefiri_ has been identified as the key player in Kefir that has already assessed many functional properties, [safety characterization and antimicrobial properties](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4052788/). However, parameters regarding safety of Kefir was imperative, before calling them probiotics.

Paul Cotter, of Teagasc Food Research Centre in Cork, Ireland, approached the issue to answer such questions associated with kefir, [employing tools of high-throughput sequencing to analyse the microbial composition of Kefir](http://msystems.asm.org/content/1/5/e00052-16). His team of researchers performed Shotgun metagenomic sequencing of the microbial population of Kefir. Along with identifying the composition, they also determined the systematic progression of the microbial communities during fermentation. They observed that, as the fermentation progressed, the bacterial diversity diminished, and species that dominate the population at the beginning are replaced by others, just hours after. For instance, _Lactobacillus_ _kefiranofaciens_ was the dominant bacterial species recorded to be in kefir during early stages of fermentations, but _Leuconostoc_ _mesenteroides_ became more prevalent during later stages. They could further establish the link between individual species present in kefir, flavour generating compounds, several genes and associated pathways. They identified genes that are responsible for some of the intestinal benefits seen in kefir-drinkers. Through metabolomic approach they could map the acidic taste of kefir to the presence of Acetobacter _pasteurianus_ and cheesy flavours to Lactobacillus _kefiranofaciens_. Additionally, they were able to change the flavour and taste of kefir by altering the ratio of these identified microbes.
Metagenomics, again, have proven to be a new way to look at bacteria, not only within hosts, but within commodities that have traditionally proven to possess health benefits. This whole new approach of analysis could in fact provide scientific insights and widen the consumer audience by imparting necessary knowledge to bridge the gap between traditional dietary & current nutritional practices.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Fermented Foods & Microbiome
Author: BugSpeaks
Published: 2019-01-17
Category: Microbiome
Meta Title: Fermented Foods & Microbiome
Meta Description: Since early history fermented foods have been consumed and have been an integral part of our diet. Fermented food gives The Broccoli conundrum – to eat or not to eat? Starting with, how to m
URL: https://www.bugspeaks.com/blog/vegetables-and-gut-microbiome
**The Broccoli conundrum – to eat or not to eat?**
Starting with, how to make children eat broccoli and then how to eat them correctly - raw or cooked or moderately cooked, then moving on to [studying all the health benefits](https://www.ncbi.nlm.nih.gov/pubmed/29513123), so much so that claims, such as “[broccoli is the only vegetable you need to eat to boost your health](https://www.independent.co.uk/life-style/food-and-drink/broccoli-vegetable-healthy-gut-immune-system-doctor-chatterjee-a8347261.html)”, have been made constantly.
On the other hand “[broccoli and other cruciferous vegetables having a dark side](https://www.kevinstock.io/health/health-dangers-of-cruciferous-vegetables/)”, with specific examples of how it can be [bad for your thyroid](https://healthyeating.sfgate.com/broccoli-bad-thyroid-9761.html) and other health risks of (at least) having (only) broccoli, have also come up.
Perhaps _“to eat broccoli or not to eat broccoli”_ has been the biggest conflict of the decade!!
To complicate things a bit more, recent peak in interest about our gut microbiota has added another dimension to this conflict.
May be to solve this conflict for once and for all?
Let’s figure it out, shall we?
**Diet play an important role in establishing and maintain a healthy gut**
We get exposed to microbes right at birth and they start to establish as our microbiome, more often as a unique fingerprint. In our gut, there are trillions of microbes – both beneficial and not so beneficial ones - together having a major impact on our health. There is a huge diversity within the gut microbiome, yet it is cumulatively unique to that individual.
In context of the blog, apart from various other factors, our [food, nutrition and diet play a very critical tole in establishment of our gut microbiota](https://www.bustle.com/p/11-foods-to-eat-for-a-healthy-gut-microbiome-7951955).
**Generally speaking,**
[Whole grains, beans, fruits and vegetables packed with nutrients, seed our gut with healthy bacteria and helps in maintaining gut diversity](https://foodrevolution.org/blog/best-foods-for-gut-health/). Eating a lot of green leafy vegetables and low-sugar fruits establish our gut with healthy and diverse microbes. Fibre-rich foods are essential for gut health. Beneficial microbes absorb vitamins to enhance immunity and decrease inflammation.

To understand this better, here are some specific classes of vegetables and how they help to establish and sustain a healthy microbiota of the gut.
**Resistant starch vegetables for gut microbiome**
High starch content of some foods has been known to have adverse effects, and consuming excessive starch can indeed lead to health problems.

[_However, all starch is not bad and its misconception that high starch is detrimental to health_](https://www.thehealthyhomeeconomist.com/best-resistant-starch-for-gut-health/).
A sub-group of starches, called resistant starch, has lately gained importance for its beneficial impact. Specifically, individuals with digestive problems, by slowly adding such foods in one’s diet, have responded with alleviated symptoms like reduced gas and discomfort, along with an overall improvement in their digestive capabilities. Largely, such foods are deemed helpful in IBS, colitis, some allergies cases and in autoimmune diseases.
The primary reason why resistant starches are important is because of their ability of enhance friendly bacteria in our colon. Resistant starches cannot be broken down by our gut (they ‘resist” digestion). Unlike other starches, instead of getting digested and being absorbed as glucose, it passes through the small intestine to the colon, where it gets converted into favourable, energy boosting, inflammation supressing “short-chain fatty acids (SCFAs)” by the intestinal bacteria. In turn, the process gives the beneficial bacteria an abundance of nutrients for their growth and sustenance.
**Foods rich in fibres and gut microbiome**
Similar two-way streak has been observed between fibre rich foods and gut microbiome. Fibre rich foods boost the growth of beneficial gut bacteria (mostly, _Bifidobacteria_). Largely, there are two types of fibres, viz., soluble and insoluble fibres. Soluble fibres help in lowering blood glucose levels and LDL cholesterol, whereas, insoluble fibres concentrate more on cleansing our digestive system. Radishes, leeks, Jerusalem artichokes, asparagus, jicama, carrots and many others belong to the class of fibre rich foods.
**Cruciferous vegetables for gut health**
**

**
This category of vegetables has been [estimated to possess high fibre content, and hence has similar mode actions and beneficial effects](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2728691/) as fibre rich foods, described above.
The most interesting aspect of cruciferous vegetables, that has set itself apart from rest of the veggie categories, is the presence of distinctive phytochemicals, such as glucosinolates.
Further, [Isothiocyanates (ITCs) are a group of bioactive compounds that are generated by the break-down of glucosinolates and are deemed to have many health benefits, including anticarcinogenic properties](https://www.ncbi.nlm.nih.gov/pubmed/?term=9865427). Various researchers have reported that, ITCs are potential chemoprotectors, they fight cancer and supress the neoplastic effects of various carcinogens at different organ sites. A detailed analysis of ITCs and glucosinolates, post consumption of dietary crucifers, have also yielded similar observations, adding support to the published reports.
_The most fascinating aspect here is the connection between these phytochemicals and our gut microbiome._
For dietary crucifers to be effective as anticarcinogenic, glucosinolates must get converted to ITCs. In plants, myrosinase enzyme is the enzyme responsible for this conversion. However, these myrosinases gets deactivated after cooking. And, here enters the gut microbiota!!
Certain groups of bacteria populating the human gut have myrosinase-like activity, which metabolizes glucosinolates to ITCs and hence re-instating the anticarcinogenic property of cruciferous veggies. Hence, when we consume cooked cruciferous vegetables, we depend on gut microbiota to convert glucosinolates to ITCs.
**Diversity in vegetables, not quantity or specific type, is key to good gut health**
The same studies that established this fascinating connection, also identified diverse bacterial species that can degrade glucosinolates. Similar observations have been made with digestion of fibre rich foods, resistant starches and many others. _These strongly indicates, that any health benefits that is conferred by these vegetables are not only dependent on the quantity and type of vegetables we consume but also depends on bacterial composition of our gut._
Further, it is a two-way streak, where eating more vegetables alters the bacterial composition of the gut, which in turn aids the digestion of these foods. A diet including many different types of vegetables (consisting of 30 or more different plant types each week) has been correlated with a much higher diversity in their gut microbiota. This pattern was noted irrespective of the type of diet consumed by the participant, greater bacterial diversity was recorded in both meat-eaters and vegans, so long as they consumed a large variety of vegetables and plant matter.
**So, should we eat broccoli or not?**
Let’s make a simple check list and see if broccoli fits the bill.

So, yes!!
You can eat broccoli
But remember
[“_Diversity in vegetables, not quantity or specific type, is key to good gut health”_](http://www.thegoodgut.org/eating-for-gut-health-variety-not-quantity-of-vegetables-is-key/)
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## On Prebiotics
Author: BugSpeaks
Published: 2019-01-14
Category: Diet and Supplements
Meta Title: On Prebiotics
Meta Description: Let’s begin with our birth... Mother’s breast milk contains rela
URL: https://www.bugspeaks.com/blog/on-prebiotics
**Let’s begin with our birth...**
Mother’s breast milk contains relatively high concentrations of small oligosaccharides (short chain of sugars) that are undigestible by new-borns. The role of these oligosaccharides in breast milk was not understood at first and in fact researchers were puzzled as to why breast milk would contain components that are unusable by the new-born. Only later did they found out that these [oligosaccharides were fuelling growth of beneficial bifidobacteria](https://books.google.co.in/books/about/Gut_Insight.html?id=EQcXAgAAQBAJ&redir_esc=y) found in new-born’s gut. This also explained why bifidobacteria is the most predominant in the guts of breast-fed babies.
**

**
_These oligosaccharides, along with many other components of food, now constitute a class of food and supplementation called “Prebiotics”_
**Technically speaking…**
Prebiotics are dietary substances that selectively promote proliferation and activity of beneficial bacteria indigenous to the colon. The concept, first developed by Gibson and Roberfoid in 1995, has been refined and redefined on several occasions. Prebiotics are currently defined as “[_selectively fermented ingredients that result in specific changes in the composition and activity of the GI microbiota thus conferring benefits upon host health_](https://www.ncbi.nlm.nih.gov/pubmed/?term=25545101)”.
Prebiotics alone can offer protection against a range of chronic diseases or conditions common in humans, like IBD, Type 2 Diabetes, Obesity, CRC etc., by preventing colonization of enteric pathogens. However, the most ideal and effective prebiotic would also be able to improve bowel function, metabolic potential, mineral bioavailability, immune modulation and most importantly support the growth of a diverse set of beneficial microbes in the gut.

**How does it work?**
In context, the most important effect of these [prebiotics and their fermented products is to selectively alter the taxonomic composition, abundance and activity of the colonic microflora](https://books.google.co.in/books?id=H6HAAgAAQBAJ&pg=PP1&lpg=PP1&dq=Probiotics+and+Prebiotics+in+Food,+Nutrition+and+Health+edited+by+Semih+Otle&source=bl&ots=-0PfnlUMtG&sig=ACfU3U0HcPETlrc4XGQW3uumJwGbUGZsbw&hl=en&sa=X&ved=2ahUKEwinh_L9q4PgAhVISX0KHa0-D2kQ6AEwAXoECAUQAQ#v=onepage&q=Probiotics%20and%20Prebiotics%20in%20Food%2C%20Nutrition%20and%20Health%20edited%20by%20Semih%20Otle&f=false), eventually conferring demonstrated health benefits. For example, prebiotic supplementation can beneficially enhance Bifidobacterium spp., the most dominant and beneficial flora of the gut. The beneficial effects of the these bifidobacteria is critically dependent on their viability and metabolic activity, which in turn is dependent on the presence of complex carbohydrates such as oligosaccharides and their metabolic products. Therefore, prebiotics are largely considered as bifidogenic factors and has been effectively utilized in diet applications.
Further, prebiotic food ingredients generally escape assimilation in the small intestine (because of their chemical structure are resistant to digestive enzymes) and upon reaching the colon gets fermented by the microbiota. There are two basic types of fermentations that take place in the gut viz., saccharolytic fermentation (of sugar/saccharide-based prebiotics) and proteolytic fermentation (or amino acid/protein/fibre-based prebiotics).
Either way, [the end products of such fermentation are largely short chain fatty acids, including acetate, propionate and butyrate](https://www.ncbi.nlm.nih.gov/pubmed/?term=27224877). These may further be metabolized systemically or locally to generate energy for us. However, they also play several other key roles that can promote heath, as depicted in the figure below.

**Great!!**
**So… where do I find these prebiotics?**
[Oligosaccharides have been proposed as the quintessential prebiotics that includes a whole set of short-chain carbohydrates](https://www.ncbi.nlm.nih.gov/pubmed/?term=19087441), mostly nondigestible by humans, but are great food sources for the gut microbes.
Most important and studied prebiotics includes [fructo-oligosaccharides (FOS), mannose-oligosaccharides (MOS), galacto-oligosaccharides (GOS) and inulin-type fructans](https://www.ncbi.nlm.nih.gov/pubmed/?term=25545101). The target bacterial groups are typically Bifidobacterium and Lactobacillus. The prebiotic potential of this oligos stems from their selective fermentation by Bifidobacterium spp. and to a lesser extent by Lactobacillus spp. in the colonic microflora.
[Fructan prebiotics such as inulin and FOS occur naturally in various foods including cereals, fruits and vegetables and so are ubiquitous in most diets](https://books.google.co.in/books?id=H6HAAgAAQBAJ&pg=PP1&lpg=PP1&dq=Probiotics+and+Prebiotics+in+Food,+Nutrition+and+Health+edited+by+Semih+Otle&source=bl&ots=-0PfnlUMtG&sig=ACfU3U0HcPETlrc4XGQW3uumJwGbUGZsbw&hl=en&sa=X&ved=2ahUKEwinh_L9q4PgAhVISX0KHa0-D2kQ6AEwAXoECAUQAQ#v=onepage&q=Probiotics%20and%20Prebiotics%20in%20Food%2C%20Nutrition%20and%20Health%20edited%20by%20Semih%20Otle&f=false). The genera that uses these oligos and which contribute the most to our health include _Ruminococcus bromii, Roseburia intestinalis, Eubacterium rectale_ and _Faecalibacterium prausnitzi,_ but there are many others that may benefit from these prebiotics. Much of the interest in these oligos are also because of the fact the more than 36000 plants worldwide contain FOS. And inulin and other fructans are commonly found in onions (2-6%), garlic (9-16%), leek (3-10%), banana (0.3-07%), asparagus (10-15%), Jerusalem artichokes (15-20%), chicory (13-20%) and even wheat (1-4%).

Dietary constituents other than carbohydrates conceivably could function as prebiotics. For instance, cocoa flavonols can increase the relative abundance of Bifidobacterium and Lactobacillus, by selectively suppressing potentially pathogenic bacteria of C. histolyticum group.
Also, it is important to note that because these prebiotics reach colon and only then gets utilized, depending on the type of prebiotics they might get concentrated and utilized in various sections of colon, as shown in the infographic below.

**Prospects of prebiotics**
Other than supplementation of prebiotics, either naturally as food or as processed supplementation, they are being utilized as “[Synbiotics](Link%20to%20BK;s%20various%20biotics%20blog)” in combination with probiotics (live cultured beneficial bacteria). Further, there are many other desirable attributes that can be encompassed into the approach of enhanced prebiotics. [Properties like highly selective fermentation, increased persistence through the colon, anti-adhesive properties, attenuation of virulence of pathogens and reduction of gas production](https://www.ncbi.nlm.nih.gov/pubmed/?term=19087441) etc., have been studied exclusively. Prebiotics are increasingly used in development of new food products, eg. drinks, yogurts, biscuits and table spreads. Even though many of the positive effects of prebiotic consumption are evident, many of these health claims require further research.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Research Services - A Crown of Thorns
Author: BugSpeaks
Published: 2019-01-04
Category: Pivot and Progress
Meta Title: Research Services - A Crown of Thorns
Meta Description: On completion of our first year as a start-up, our CEO Kumar wrote The collaborative spirit cannot be taught. It just happens. In the last blog you would have r
URL: https://www.bugspeaks.com/blog/the-collaborative-team-spirit
The collaborative spirit cannot be taught. It just happens.
In the last blog you would have read about our pivot towards the microbiome. This would not be possible without all members of our team coming together and developing an awesome product. How data and information flows from one team to another is vital for meaningful science. It is also very essential for a seamless experience. I wanted to introduce the dynamic team behind it.
**Computational Biology**
**DNA: The RAM guzzlers!!**
**RNA: Probably the most vociferous team in the office.**
No matter how many terabytes of RAM is provided they manage to consume it. RNA, DNA, short read, long read, no matter what the sample or technology they have a solution for it.
For the microbiome program they have developed a platform that can take any type of sequencing data, 16s, metagenomic or metatransciptomic data and process them into actionable outcomes.They have probably processed close to a petabyte of microbiome data to develop this robust system. Every month they bring down the computational run time and reduce the size of the data footprint. They have also developed a robust systems biology engine to discover the unknown.

**Microbiome & Nutrition**
**DNA: The biome experts!!**
**RNA: Probably the most combative team in the office.**
They claim to know what every living organism on this planet feeds on. They know how bacteria, virus & fungus thrive.

For the microbiome program they have developed proprietary databases on nutrition, microbiome and human diseases. The team has spent countless hours in validating the correlation as well as causation between the microbiome and specific traits or diseases.
**Reporting & Content**
**DNA: The translation specialists.**
**RNA: They are the nimblest team in the organisation.**
Given any complex data, they will simplify it to a language understood by all.
For the microbiome program they have meticulously refined data delivery. The sliced the reports into multiple formats to better serve the consumers, the clinicians, the nutritionist and lifestyle coaches. Each report is handcrafted and personalized.

**Product Design**
**DNA: Wood is the new cardboard!!**
**RNA: They are the pickiest team.**
Their moon shot is handsfree faecal collection.
For the microbiome program they have hand crafted a beautiful kit that signifies how precious faecal matter is. With every iteration a new feature is introduced to make the collection to report experience seamless.

**The X Factor**
**DNA: The secret sauce**
**RNA: They are very silent, yet salient group.**
They make Bug Speaks talk. Even sing.
For the microbiome program they have taken raw data from.....

**AI**
**DNA: The horse whisperers.**
**RNA: They are the most secretive team.**
They talk to the data and some say the data talks back to them.
For the microbiome program they have implemented machine learning over knowledge graphs. They have also used blockchains to maintain transparency.

**Accounts & Logistics**
**DNA: The poop importers!!**
**RNA: They are busiest team in the office.**
“_Hello FedEx, we would like to import poop”._
_

_
For the microbiome program they have amalgamated multiple logistics partners to ensure there are no difficulty in sample transportation, no matter which part of the world the sample is coming from. They have also ensured that EMIs are available for all tests.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The Microbiome Pivot
Author: BugSpeaks
Published: 2019-01-01
Category: Pivot and Progress
Meta Title: The Microbiome Pivot
Meta Description: What drives Leucine Rich Bio? What is it that we really want to do?
Were the questions that kept coming back to me…<
URL: https://www.bugspeaks.com/blog/the-microbiome-pivot
What drives Leucine Rich Bio? What is it that we really want to do?
Were the questions that kept coming back to me…
And the answer always boiled down to healthcare.
_“Health care” and not “Sick care”_.
When we started the company, we wanted to provide the necessary answers to solve healthcare problems. We developed databases and algorithms. We crunched data and simplified complex interactions to meaningful actionable points. All this led to our expanded capabilities of looking at complex problems in biology and we did not limit ourselves to healthcare alone. Over 4 years we processed, analysed and reported over 100 terabytes of data for customers from 10 different countries.
And then came the pivot!!

I was nervous. I had to go up to my colleagues and tell them about the pivot. How we were going to stop all other projects and concentrate on “_one wellness product_”.
Why?
Because deep down all of us cared about human health. We cared about staying healthy and when it came to human health our passion produced creativity. But I was not sure whether my colleagues sensed my inner passion. I was nervous.
I gathered everyone.
I knew we would fail if everyone did not participate enthusiastically.
“_Let’s pivot around the microbiome_”, I said.
The room was silent with few glances at each other.
Team Leucine Rich Bio is known for many things, silence is not one of them. Raucous laughter with vociferous discussions is how team meetings usually go. Ask the people in the next building. This time was different though!!
“What’s pivot?”, asked one.
That’s when it dawned to me that I was talking in ‘_start-up lingo_’ to a bunch of ‘_scientists and techies_’. My 1-minute monologue on pivot was met with bombardment of ideas and solutions. We were going to pivot around the microbiome and all of us were backing it. All 22 of us. (More about this collaborative spirit in my next blog).
This was a great moment for us. We were not going to just churn data, we were going to have positive impact on everyone’s health.
Our health is impacted greatly by the microbiome. The human microbiome is defined as the collective genomes of the microbes (bacteria, bacteriophage, fungi, protozoa and viruses) that live inside our body. In simple words it’s the different kind of microbes living inside and on our body. Some estimates suggest that there are more microbes in our body than human cells. Given the sheer volume of the microbiome, it must have a significant influence on our body.
Of these, 90% of the microbes reside in our gut. The gut microflora is influenced by nutrition and lifestyle. So, in essence, what we eat and how we live shapes our microbiome, which in turn influences our health.
Below is a complex graph showing the different species of microbes found in different parts our body.

We had started working on the microbiome in 2015. The very first sample we analyzed looked at a few hundred microbes. We now look at around 1,40,000 microbes. We have identified close to 10,000 different species of microbes. Usually we see anywhere from 800 to 2000 different types of microbes in an individual.
I’ll let you read that sentence again.
“800 to 2000 different types of microbes are present in an individual’s gut”.
Each of them have millions of copies. Just fathom the impact that it might have on us. This impact has been shown by different academic labs around the world. As we delved into the science, we realized the impact is even greater.
_To top it off, the solution is simple. Nutrition._
Our mission from the beginning was to bring about a paradigm shift to _Health Care from Sick Care_. We strongly believe the microbiome is the way forward to achieve our mission and the technology that we have developed is on par if not better than the rest.
We pivoted around the microbiome so that we could leverage our expertise and passion to provide everyone with better health. Better health is not a moonshot, it is as simple as including an extra food item in your diet.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Studies Suggest the Relationship Between the Gut Microbiome and Allergic Diseases
Author: BugSpeaks
Published: 2018-07-27
Category: Microbiome and Disease
Meta Title: Studies Suggest the Relationship Between the Gut Microbiome and Allergic Diseases
Meta Description: The past few years have witnessed the increased cases of patients suffering from allergies and inflammatory diseases. Scientific explorations have sug
URL: https://www.bugspeaks.com/blog/studies-suggest-the-relationship-between-the-gut-microbiome-and-allergic-diseases
The past few years have witnessed the increased cases of patients suffering from allergies and inflammatory diseases. Scientific explorations have suggested the existence of the relationship between the gut microbiome and allergic diseases. The gut microbiome produces both beneficial and damaging metabolites from the diet. Along with bacterial components, these metabolites modulate the host immunity, and significant influence on the occurrence of allergic illnesses.
The human gut harbors several trillions of commensal microorganisms, mainly the bacterial population with more than thousands of species. This collection of microbes is generally represented as the gut microbiome. Due to obvious reasons, culture-based analysis of microbiome diversity is replaced by the recent advanced high-throughput DNA sequencing approaches, such as the bacterial 16S ribosomal RNA sequencing and microbiomics. These approaches enable the correct documentation of commensal bacteria directly without requiring to culture. Such studies have evidenced that alteration in the gut microflora composition or declined diversity, also known as dysbiosis state may lead to the development of intestinal inflammations and subsequently cause allergic conditions. For example, in a study comprising patients having food allergies in the United States has revealed the occurrence of reduced microbial diversity in the gut, and found an increased number of bacterial species belonging to Bacteroidales, while Clostridiales were found in low numbers. Further, some gut bacteria produce useful metabolites using the host's diet, and they directly take part in the mediation of host’s immune responses. Researchers have highlighted the role of commensal gut bacteria and their metabolites in regulating host immune responses and allergic diseases.

Long-chain fatty acids (LCFAs), such as ω3 and ω6 fatty acids function as energy sources and take part in the immune responses. However, mammals cannot produce LCFAs, and interestingly they are produced by the gut microbes using dietary components. Further, it is suggested that ω3 fatty acids have anti-inflammatory and anti-allergic properties (Miyata and Arita 2015). The gut commensal bacteria, mainly lactic acid bacteria (e.g., Lactobacillus plantarum) are reported to produce ω6 fatty acid-derived lipid metabolites (linoleic acids, oxo and hydroxy fatty acids). Likewise, 17,18-epoxy-eicosatetraenoic acid, an anti-allergic lipid metabolite is speculated to be produced in the colon via cytochrome P450 function. However, evidences have suggested the role of commensal bacteria in LCFAs metabolism. In a study, germ-free animals were shown to produce high levels of colonic lipid metabolites, such as 17-hydroxy docosahexaenoic acid, 14-hydroxy docosahexaenoic acid, resolvin D1, and protectin D1 when compared to conventional mice. Resolvin D1 is known to down-regulate the expression of interleukin-1β (IL-1β) during microbial infections. Further, IL-1β intensifies allergic reactions to cause asthma, atopic eczema, and dermatitis. Also, it is suggested that microbe-dependent inhibition of resolvin D1 production is linked to allergic inflammations (Hirata and Kunisawa 2017). Experimental evidences suggest that 10-hydroxy-cis-12-octadecenoic acid produced by Lactobacillus spp. may prevent the progress of food allergies, which is mediated by maintaining intestinal epithelial barrier function. Figure 1 illustrates the role of bacterial metabolites in the gut towards regulating immunity for the allergic diseases.
[](/media/uploads/blog/163a.jpg)
[

](/media/uploads/blog/163a.jpg)
**Figure-1: Bacterial metabolites in the gut regulate immune responses for the allergic diseases. Bacterial metabolites regulate versatile biologic and immunologic functions related to allergic diseases. Epithelial barrier function is enhanced by 10-hydroxy-cis-12-octadecenoic acid, a linoleic acid-derived metabolite, and SCFAs such acetate and butylate, fermentation products by some bacteria. In addition, SCFAs (e.g., acetate, butyrate, and propionate) as well as D-type tryptophan have a potential to enhance the induction of Treg cells. Vitamin B9 plays a key role in the maintenance of Treg cells and its metabolite prevents the activation of MAIT cells by competing with vitamin B2 metabolite, MAIT cell activating ligand, in their binding to MR1. PPAR ligands such as LCFAs decrease the inflammatory cytokines such as IL-8 and induce the production of anti-inflammatory cytokines such as IL-10. (Source: Hirata and Kunisawa 2017; [https://doi.org/10.1016/j.alit.2017.06.008](https://doi.org/10.1016/j.alit.2017.06.008))**
In a recent cohort study, the association amongst the gut microbiome, their lipids, and allergic diseases in humans was established. It was observed that the composition of the gut microbiota plays a significant role in the development of atopy and asthma in childhood. 16S rRNA sequencing analysis evidenced that high risk for the development of allergy in neonates is due to the reduced abundance of gut bacterial species, such as Bifidobacterium, Akkermansia and Faecalibacterium and increased abundance of fungi, such as Candida and Rhodotorula. Interestingly, the low-risk subjects were observed to have increased levels of anti-inflammatory lipid metabolites (docosapentaenoate and dihomo-γ-linolenate). On the other hand, high-risk groups were shown with enhanced levels of pro-inflammatory metabolites, such as 12,13-dihydroxy-9Z-octadecenoic acid, stigmasterol and sitosterol, and 8-hydoxyoctanoate (Fujimura et al. 2016). LPSAs are reported to modulate immune reactions via the peroxisome proliferator-activated receptor (PPAR) family, i.e., the most common negative regulators for allergic conditions. As stated by Hirata and Kunisawa (2017), Bacteroides thetaiotaomicron stimulates PPAR-γ-dependent export of NF-κB from the nucleus, and subsequently it decreases NF-κB-dependent IL-8 production to curb the IL-8-intermediated infiltration of granulocytes in bronchial allergy.
Moreover, immune cells, including iNKT (invariant natural killer T) cells are activated by microbial glycolipids. For example, Sphingomonas spp. producing glycosphingolipids, Sphingomonas spp., and Borrelia burgdorferi producing glycodiacylglycerols, Helicobacter pylori producing cholesteryl-α-glucoside. Further, D-type amino acids (e.g., D-alanine, D-glutamic acid, D-asparagine, and D-prorine) secreted from the gut bacteria are believed to control host allergic reactions. Likewise, D-tryptophan from the probiotic bacteria, such as Lactobacillus rhamnosus GG, Lactobacillus casei W56 is shown to ameliorate allergic airway inflammation and hyperresponsiveness in the gut and lung. Similarly, microbial vitamins, including B-family members and K, serve as an important supplementary source of vitamins that are take part in the regulation of the immune responses and controlling allergic diseases. Likewise, Short-chain fatty acids, such as propionate (produced from Bacteroidetes and some Firmicutes), acetate (produced from many genera of gut microflora, including Bifidobacterium spp.), butyrate (produced from Clostridium cluster XIVa species and Bacteroides thetaiotaomicron) exert diverse anti-allergic properties.
Overall, commensal gut bacteria produce many useful metabolites that are involved in regulating allergic responses mediated via various ways, such as the induction of Treg cells, up-regulation of IL-10 expression, suppression of the Th2-type phenotype and maintenance of the gut barrier function. Also, they are known to produce pro-inflammatory metabolites. There are many factors that are involved in the regulation of immune response and allergic diseases. Hence, having a right balance between diet and the gut microbiome may certainly benefit in overcoming allergic diseases. However, more research is highly recommended in this regard to clearly understand about the mechanisms involved and to disclose various cross-talk between commensal microbes in the gut and host immune system.
**Keywords:** The gut microbes, metabolites, microbiome, short-chain fatty acids, vitamins, allergic diseases, immunity
**References:**
* Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, Panzer AR, LaMere B, Rackaityte E, Lukacs NW, Wegienka G. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nature medicine. 2016 Oct;22(10):1187.
* Hirata SI, Kunisawa J. Gut microbiome, metabolome, and allergic diseases. Allergology International. 2017;66(4):523-8.
* Miyata J, Arita M. Role of omega-3 fatty acids and their metabolites in asthma and allergic diseases. Allergology International. 2015;64(1):27-34.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The Gut Microbiome Alterations is related to Autism Spectrum Disorders
Author: BugSpeaks
Published: 2018-07-17
Category: Gestational and Neonatal
Meta Title: The Gut Microbiome Alterations is related to Autism Spectrum Disorders
Meta Description: Autism spectrum disorder (ASD) is a hereditary linked disorder of brain function with symptoms, such as restricted forms of behaviors and activities,
URL: https://www.bugspeaks.com/blog/the-gut-microbiome-alterations-is-related-to-autism-spectrum-disorders
Autism spectrum disorder (ASD) is a hereditary linked disorder of brain function with symptoms, such as restricted forms of behaviors and activities, or persistent discrepancies in societal interaction and communication. ASD has a significant impact on the growth and development of children and on the general public. The assessed incidence of ASD in 2012 was about 14.6 per 1,000 children (aged 8 years), and its occurrence was relatively higher in boys (23.6 per 1,000) when compared to girls (5.3 per 1,000). As per the estimate, in the United States alone, the cost of caring for a child with ASD was about $1.4 million, while in the United Kingdom, it was reported to be £0.92 million (Li et al. 2017). The main costs associated with the care of children with ASD are special education services and a loss of parental productivity. Thus, the financial effects of ASD have driven scientists to hunt for effective medications (Buescher et al. 2014; Li et al. 2017).
ASD is described by numerous clinical endophenotypes, and is hypothetically connected with certain comorbidities. As per the current recommendations, gastrointestinal symptoms are the reasons for the comorbidity in patients with ASD, however underlying mechanisms are unfamiliar. Several investigations have revealed that patients with ASD have alterations in the fecal flora composition, and the gut microbiome metabolic products. The dietary constituents along with metabolic activities of the microbiome are now assumed to be linked to alterations in behavior and cognition, mainly in patients having neurodegenerative diseases. The gut microbiome have a significant role in the development of the brain and controls the behavior via the neuroimmune, neuroendocrine, and autonomic nervous systems. There is a bidirectional communication between the central nervous system and the gastrointestinal tract (brain-gut axis) and the gut microbiome functions. As stated by most recent studies, collective pathogenetic elements and pathophysiological mechanisms probably associating to gastrointestinal disturbances and ASD include intestinal inflammation with or without autoimmunity, gluten-related disorders (celiac disease, wheat allergy, non-celiac gluten sensitivity), immunoglobulin E-mediated and/or cell-mediated gastrointestinal food allergies, abdominal pain, dysautonomia connected with gastroesophageal reflux and gastrointestinal dysmotility. In addition, dysregulation of the gut microbiome has the capability to upset intestinal permeability, motility and sensitivity, and mucosal immune function.
The human gut contains about 1000 grams of bacteria, and approximately about 9.9 million numbers of bacterial genes exist in the gut i.e., the ratio of microbiome DNA and host DNA is 10:1. Gastrointestinal indications are noticeable in ASD patients. Researchers have observed more gastrointestinal syndromes, including diarrhea and constipation in children having ASD compared with their unaffected siblings. ASD patients with gastrointestinal syndromes display substantial interactive or behavioral manifestations, including self-injury, aggression and anxiety. Many studies have demonstrated that the gut microbiota composition and function is directly or indirectly linked with ASD signs, partly by prompting the immune system and metabolic rate. Patients with ASD are observed to have a higher percentage of abnormal intestinal permeability leading to greater antigenic load from the gastrointestinal tract. ASD-associated cytokines, such as interferon-γ (IFN-γ), interleukin-6 (IL-6), IL-1β and tumor necrosis factor-α (TNF-α) in addition to lymphocytes present in the circulation cross the blood-brain barrier (BBB). Successively, IL-6, IL-1β and TNF-α bind to brain endothelial cells and encourage immune responses in the brain. Variations in the gut microbiota composition and their metabolic products secretion are generally noticed in ASD patients and in animal models of ASD. In a mouse model study featuring ASD, researchers have found that gastrointestinal barrier defects caused microbiota alterations. They observed more abundant bacteria, belonging to Prevotellaceae, Porphyromonadaceae, unclassified Bacteroidales and Lachnospiraceae in offspring of mothers with maternal immune activation, while in control offspring, Erysipelotrichaceae, Ruminococcaceae and Alcaligenaceae were found to occur more abundantly. The gut microbiota diversity in children with ASD is observed to be very less as compared to that of children without ASD. In ASD patients, higher levels of Desulfovibrio, Lactobacillus, Bacteroidetes, Clostridium, Caloramator and Sarcina, and lower levels of Bifidobacterium and Firmicutes are observed. Further, children with ASD and gastrointestinal syndromes possess inferior loads of unclassified Veillonellaceae and the genera, Coprococcus, and Prevotella than that found in gastrointestinal syndrome-free neurotypical children. Clostridium histolyticum, found in the fecal samples of children with ASD was at a higher level when compared with healthy children without ASD. Moreover, altered levels of Prevotella, Bifidobacterium, and Sutterella are found in children with ASD. Researchers also have observed that Candida species (Candida krusei and C. glabrata) were 2-times higher in individuals affected with autism than in normal individuals. Also, it is observed that Candida species can release ammonia and toxins that can encourage autistic behaviors.
The gut–brain axis is viewed as a way of interaction between the gut microbiome and the brain, and it is a two-way communication structure. The outcome of several studies suggests that the gut-brain axis contributes to the origin and development of ASD. In a comprehensive review by Li et al. (2017) stated that the gut flora affects functions of the brain via the neuroendocrine, neuroimmune and autonomic nervous systems and through toxin production by microbiota (Figure 1). The presence of millions of neurons in the mucosa of the gastrointestinal tract constitutes the enteric nervous system and modulate various gastrointestinal functions. Hence, the gut is recognized as ‘the second brain’. ?The potential interactions between the gut microbiota and the pathogenesis of ASD is outlined in Figure 1.

[](/media/uploads/blog/162a.jpg)
[

](/media/uploads/blog/162a.jpg)
**Figure-1: Potential relationships between the microbiota and ASD (the gut-brain axis). The production of metabolites, such as SCFAs and toxin metabolites, by certain microbiota (e.g., Lactobacillus) can cross the “leaky gut” to affect brain function. Some microbiota can produce neuroactive compounds (e.g., 5-HT and GABA) that cross the “leaky gut” and influence brain function and induce abnormal behaviors. These neuroactive compounds can directly influence the HPA axis and increase circulating levels of cortisol. Metabolites, certain microbiota and neuroactive compounds can activate enteric neurons and affect brain function through the vagus nerve. Some microbiota and metabolites can activate gut immune cells, which can release cytokines into circulation. 4-EPS, 4-ethylphenyl sulfate; 5-HT, serotonin; HPA, hypothalamic–pituitary–adrenal; SCFAs, short-chain fatty acids; BBB, blood-brain barrier; 5-HT, 5-hydroxytryptamine; ENS, enteric nervous system; GABA, γ-aminobutyric acid; DA, dopamine. (Adapted from Li et al. (2017); doi: [10.3389/fncel.2017.00120](https://dx.doi.org/10.3389%2Ffncel.2017.00120))**
The gut microbiota secretes certain metabolites, such as phenolics, short-chain fatty acids (acetic acid, proprionic acid, isobutyric acid, butyrate, valeric acid and isovaleric acid), and free amino acids that affect ASD-like behaviors via the vagal pathways mediating the microbiome-brain-gut axis communication. Likewise, the gut microbiota releases neuroactive compounds (dopamine, 5-HT, γ-aminobutyric acid, and histamine), which trigger or impede central neurons via the vagus nerve.
Currently, there are no available effective therapies for ASD. Parents often take their children to obtain mediations that are personalized to their specific needs. Recent evidences have indicated that the gut microbiota modulation can be a potential therapy in children having ASD. In this regard, prebiotics, probiotics, fecal microbiota transplantation and a suitable personalized diet have gained substantial attention in recent days. Moreover, microbiome-facilitated therapies are relatively safe and effective. Recent clinical studies have reported that ASD patient’s symptoms can be improved by treatments that normalize the gut microbiota. Nevertheless, more research studies involving more participants are required to know about the treatments that supports the gut microbiome in overcoming ASD symptoms.
**Keywords:**
The gut microbiota, microbiome, Autism spectrum disorder, Brain-gut axis, Probiotics, Fecal microbiota transplantation, Microbiome-mediated therapies
**References:**
* Li Q, Han Y, Dy AB, Hagerman RJ. The gut microbiota and autism spectrum disorders. Frontiers in cellular neuroscience. 2017 Apr 28;11:120.
* Tomova A, Husarova V, Lakatosova S, Bakos J, Vlkova B, Babinska K, Ostatnikova D. Gastrointestinal microbiota in children with autism in Slovakia. Physiology & behavior. 2015 Jan 1;138:179-87.
* Buescher AV, Cidav Z, Knapp M, Mandell DS. Costs of autism spectrum disorders in the United Kingdom and the United States. JAMA pediatrics. 2014 Aug 1;168(8):721-8.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The gut microbiome have a role in Dementia
Author: BugSpeaks
Published: 2018-07-11
Category: Microbiome and Disease
Meta Title: The gut microbiome have a role in Dementia
Meta Description: Dementia, as a comprehensive type of brain disease is known to upset an individual’s daily activities. The disease is characterized by the deter
URL: https://www.bugspeaks.com/blog/the-gut-microbiome-have-a-role-in-dementia
Dementia, as a comprehensive type of brain disease is known to upset an individual’s daily activities. The disease is characterized by the deteriorated memory, thinking ability, behavior and the capability to carry out everyday functions. Globally, ~47.5 million individuals are affected and the prevalence of dementia is increasing continuously. Dementia leads to disability and dependency amongst the aged people, and thus has a colossal physical, mental, societal and economic effect on families, caregivers, and the general public. The common form of dementia is the Alzheimer’s disease, which account to about 60-70% of the cases followed by other forms, such as Lewy body dementia, vascular dementia, frontotemporal dementia, and Parkinson’s disease with dementia.
Recent research advancements have deciphered the role of gut microbiome and its composition associated with the gut permeability and inflammation in dementia patients. Also, it is hypothesized that dysbiosis (an impaired microbiota) may augment the gut permeability, microbial translocation and trigger an inflammatory-immune response, which may perhaps encourage pathogenesis and progression of dementia. In this regard, a novel approach is suggested for the management of these health disorders, and as an adjuvant for psychiatric treatment of neurological disorders, including dementia and other interrelated diseases via modulating the microbiota composition (e.g., with the use of probiotics).
The expansion of advanced high-throughput analytical techniques including next generation sequencing has disclosed the possible role of the commensal microbial population, particularly the gut microbiome in several human diseases. Lately, the theory of the gut brain-axis has been well-established. Accordingly, a communication exists between the gut and brain site to control and mediate the process of many biological pathways involving the autonomic and enteric nervous system, the neuroendocrine system and the immune system. However, any deviations in such communication due to dysbiosis may involve in disease development. For example, the enteric microbiota i.e., Bifidobacterium and Escherichia, which are considered as both commensal and pathogenic microbes are identified to have interactions between the brain and gut axis.
The gut microbiota supports numerous daily activities of the brain, comprising the regulation of the hypothalamic-pituitary-adrenal (HPA) axis activation state. The HPA axis activation releases cortisol, which in turn manage the triggering of microglia of the brain, and influence to release cytokines. This action rule the functions of the central nervous system through various communications, including vagal nerve and adrenergic nerve activation in addition to the secretion of many molecular candidates (endocrine hormones, neuropeptides, neurotransmitters, and immunomodulators). The stress hormones of hosts, for example noradrenaline might affect signalling between bacteria or their activities in the gut and lead to changes in the microbial diversity and overall functions of the gut microbiota. Nevertheless, these gut bacteria synthesize and release many neurotransmitters and neuromodulators, and induce the synthesis as well as the release of neuropeptides from enteroendocrine cells. For instance: the species of Bifidobacterium and Lactobacillus produce short-chain fatty acids; spore-forming bacteria produce 5-HT; Bacillus, Escherichia and Saccharomyces spp. secrete norepinephrine; Bacillus and Lactobacillus spp. produce dopamine and acetylcholine, respectively. In specific, recent findings have suggested that microglia maturation and function is governed by the short-chain fatty acids produced by the gut microbiota, and hence highlighted on the possible usefulness of the gut microbiome as an impending diagnostic target in dementia and its cure.
The gut microbiota regulates neuro-inflammations and the HPA axis activity, and might lead to Alzheimer’s disease. The neuropathological features of Alzheimer’s disease are the two pathologic protein aggregates, namely amyloid beta (Aβ) plaques and hyper-phosphorylated tangles of tau-protein, which cause neuro-inflammation, mainly facilitated by the innate immune system. This process mainly includes microglia cells that represent the resident macrophages of the brain. Though microglia cells can remove extracellular Aβ aggregates, in later phases of the disease, cells remain in a dystrophic state and fail to exert their positive actions. In addition, the inflammation and the pathogens (e.g., bacteria: Salmonella enterica; fungi: Candida albicans; viruses: herpes simplex virus; and parasites: Toxoplasma gondii) interactions can also lead to the development of Alzheimer’s disease. Thus, a strong gut microbiota may pay a significant role in preventing general infections by restricting pathogen growth, improving the microbial barriers, and enhancing the immune response. The representation of a few key factors in the Alzheimer’s disease pathogenesis is given in the figure-1.

[](/media/uploads/blog/161a.jpg)
[

](/media/uploads/blog/161a.jpg)
**Figure -1: Schematic of some key players in the pathogenesis of AD. The gut microbiota regulation of neuro-inflammation and the hypothalamic–pituitary–adrenal (HPA) axis activity and may lead to AD. The bacterial products that gain access to the brain through the bloodstream and the area postrema, via cytokine release from mucosal immune cells, through the release of gut hormones such as 5-HT from EEC cells, or via afferent neural pathways, including the vagal nerve. NP: Neuropeptide; NT: Neurotransmitter; 5-HT: 5-hydroxytryptamine; DC: Dendritic cell; EEC: Enteroendocrine cell; Aβ: amyloid beta protein; AD: Alzheimer’s disease.**
**(Adapted from Alkasir et al. 2017; doi: [10.1007/s13238-016-0338-6](https://dx.doi.org/10.1007%2Fs13238-016-0338-6)).**
In the course of ageing, the gut microbiome composition undergoes fluctuations. In aged people, decreased microbial diversity with a loss of helpful taxa and increased facultative pathogens have been observed. Further, aging is connected with inflammation, which is also represented as inflammaging, which is linked with the accelerated gut permeability, mucosal inflammation and bacterial translocation. As aging is one of the chief risk factors of dementia, it is expected that the gut-brain axis is disapprovingly involved in the development of dementia. Considering this, a precise diet and the surrounding environment play a vital role in shaping the good microbiome composition and help in controlling many metabolic disorders. Some of the animal investigations have suggested that dementia is associated with fluctuations in the gut microbiome composition.
Studies have clearly indicated the positive link between the gut microbiome composition and the neuropathology of dementia. However, more investigations on the gut microflora is required to enable additional novel vision into the biology of dementia. This will facilitate in the development of novel therapeutic strategies for dementia. As stated in a review by Alkasir et al. (2017), the modulation of gut microbiota (by probiotics or other dietary intervention) or direct targeting of gut microbiota enzymes (by pharmacological inhibitors or activators) may be a growing area for pharmaceutical and functional food industries, with the goal of decreasing the widespread growth of adiposity, insulin resistance, Alzheimer’s disease, and other metabolic diseases.
**Keywords:**
Gut microbiome, Dysbiosis, Dementia, Alzheimer’s disease, Neuro-inflammation, Probiotics, Prebiotics
**References:**
* Alkasir R, Li J, Li X, Jin M, Zhu B. Human gut microbiota: the links with dementia development. Protein & cell. 2017 Feb 1;8(2):90-102.
* Hu X, Wang T, Jin F. Alzheimer’s disease and gut microbiota. Sci China Life Sci. 2016. doi: 10.1007/s11427-016-5083-9.
* Pistollato F, Sumalla Cano S, Elio I, Masias Vergara M, Giampieri F, Battino M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutrition reviews. 2016 Oct 1;74(10):624-34.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The gut microbiome promotes immune homeostasis
Author: BugSpeaks
Published: 2018-07-06
Category: Microbiome and Disease
Meta Title: The gut microbiome promotes immune homeostasis
Meta Description: The gut microbiome is defined as the group of microorganisms and their genes existing inside the human gut. Recently, it has arisen as a primary facto
URL: https://www.bugspeaks.com/blog/the-gut-microbiome-promotes-immune-homeostasis
The gut microbiome is defined as the group of microorganisms and their genes existing inside the human gut. Recently, it has arisen as a primary factor in various human health conditions and diseases. A symbiotic relationship between humans and microbes is prevailing ever since from the birth, and any perturbations in this relationship could be the basis for immunological dysregulation, which might lead to the onset of many health complications and diseases including inflammatory bowel disease, autoimmunity, rheumatoid arthritis, cancer and multiple sclerosis. Symbiotic bacteria of the human gut provide a good environment and several advantages, such as providing essential nutrients, metabolizing indigestible food compounds, defense against the opportunistic pathogen colonization, and even contribute in the development of the intestinal architecture.
The immune system is responsible to recognize, retort and acclimatize to numerous foreign substances/molecules and also self-molecules. Thus, immune responses significantly modulate the development of health and disease. The human gut placidly co-exist with vast and diverse beneficial microbiota, and our immune system has evolved with gut bacteria to protect against various infectious microbial pathogens. In specific, the gastrointestinal tract is the main site at which both symbiotic and pathogenic microbes interact with the host immune system. More recent evidences have suggested a beneficial partnership has evolved amongst the symbiotic gut bacteria and the host immune system. The molecular communications/exchanges between them are directly linked to the growth of immune responses, and sequentially, the immune system of the host shapes the configuration of the microbiota.
Research evidences have related the role of the gut microbiota in shaping our immune system responses during diseases, however, the question still remains as to which specific bacteria are responsible in the mediation of these helpful responses and, more importantly, how this is accomplished. Some inspiring examples of the gut microorganisms having a pivotal role in the prevention of inflammatory bowel diseases, and beneficial immune reactions they stimulate during the defense are highlighted below.
At the start of 1900s, Ilya Mechnikov proposed for the first time about the use of live microbes to sustain bowel health and prolong life. Presently, the word “probiotic” is used for describing dietetic microbes with the ability of offering health benefits to the host. Numerous bacterial species either individually or in combination have been reported to ameliorate the symptoms of inflammatory bowel diseases in humans and mouse models. Some of them include Bifidobacteria lactis, B. infantis, E. coli Nissle 1917, Lactobacillus rhamnosus GG, L. salivarius, L. fermentum, Bacteriodes fragilis, B. thetaiotaomicron, etc. Mainly, these probiotic strains decrease toxic microbial metabolic events, however, more recent evidences have demonstrated their capability to modulate gut immune responses. The probiotic strains have a common feature of controlling the inflammation. The probiotic bacteria act on different cell types including epithelial cells, T cells and dendritic cells. Further, evidences have suggested that these probiotic strains induce regulatory T cells, which is the central to regulating inflammation and diseases. For example, treating colitic mice with the VSL#3 (a mixture of 8 lactic acid bacteria probiotic strains) increased the production of interleukin (IL-10) and the percentage of TGFβ-expressing T cells. Likewise, in another model of pathogen-induced inflammation study, the treatment of mice with B. infantis significantly down-regulated the intestinal inflammation and increased the number of CD4+CD25+ TReg cells. Moreover, the adaptive transmission of the CD4+CD25+ TReg-cells from mice served with B. infantis repressed inflammation-related activation of NF-κB (nuclear factor-κB) in the recipient mice. Interestingly, when the bone-marrow-derived dendritic cells incubated with L. rhamnosus were transferred into a colitic mice, a protection against inflammation and disease was observed. Further studies have shown that L. rhamnosus-treated dendritic cells can initiate TReg-cell activity. It has been shown that certain patients with Crohn's disease possess a reduced level of a noticeable gut bacteria, Faecalibacterium prausnitzii. As reviewed and explained by Round and Mazmanian (2009), this organism or its secreted substances are capable of inducing IL-10 expression (anti-inflammatory response), and ameliorate the induction of tumor necrosis factor-α (TNF-α) and bowel diseases, when administered orally to the experimental animals. This study further indicated that there exists a direct link between the reduced numbers of F. prausnitzii from the gut microbiota and the development of a bowel disease. This clearly indicates that symbiotic gut bacteria can intermediate in inflammatory bowel diseases and health. However, the explicit molecules/compounds secreted by these beneficial microbes of gut microbiota to guide in the immune responses still remains unclear. But, existing scientific data support the clue that symbiotic gut microbes actively interconnect with the host immune system to modulate anti-inflammatory processes.
A single molecule, polysaccharide A (PSA) secreted by the human commensal bacterium (Bacteroides fragilis) is predicted to shape favorable immune responses (Figure 1). When the germ-free mice were colonized with B. fragilis or treated with the purified PSA, the cellular and physical development of the immune system was improved with the increase of differentiated splenic CD4+ T cells. Experimental evidences have suggested that PSA protects by decreasing the levels of the pro-inflammatory cytokines (TNFα, IL-17 and IL-23). Also, it inhibits epithelial hyperplasia and neutrophil infiltration to the gut associated with disease induction in these models (Round and Mazmanian 2009). Likewise, the gut microbiota on T-cells immune responses are depicted in figure 1.

[](/media/uploads/blog/160a.jpg)
[

](/media/uploads/blog/160a.jpg)
**Figure-1: Impact of the gut microbiota on T-cells immune responses. Colonization with segmented filamentous bacteria (SFB) occurs by intimate attachment to the intestinal epithelium and promotes the development of T-helper 17 (Th17) cells via intestinal epithelial cell (IEC)-derived cytokines, serum amyloid A (SAA), as well as antigen presentation by dendritic cells (DCs). Adhesion of SFB to IEC can potentially generate a circuit, wherein DC-derived IL-23 stimulates IL-22 production by type 3 innate lymphoid cells (ILC3), which in turn induces SAA from IEC and can lead to Th17 cell differentiation. Conversely, colonization of beneficial commensal bacteria induces de novo generation of Tregs and downregulates Th17 immune responses. Commensal bacteria, including most Clostridia species, produce short-chain fatty acids, i.e., butyrate, which participates in the de novo generation of T-regulatory cells by suppressing proinflammatory cytokines, by promoting RA production from DCs, and by inducing Foxp3 transcription. Among different Bacteroides fragilis strains, those expressing polysaccharide A (PSA) mediate the generation of Tregs via TLR2, while those secreting B. fragilis toxin (BFT) alter the function of IEC tight junctions. Upon disruption of barrier function, dissemination of microbial products, recognized by microbe-associated molecular patterns (MAMPs), occurs and activates the IL-23 pathway, resulting in subsequent barrier repair and stimulation of Th17 immune responses. (Adapted from Omenetti et al. 2015; [https://doi.org/10.3389/fimmu.2015.00639](https://doi.org/10.3389/fimmu.2015.00639)).**
Nevertheless, it can be predicted that some beneficial symbiont microbial species have evolved, and produce molecules that can prompt defensive intestinal immune reactions. The presently available treatments for inflammatory bowel diseases are either ineffectual in most patients or result in severe side effects. Therefore, understanding of the beneficial gut microbial species and their secreted compounds can aid in preventing or curing such diseases, and help in designing new and effective natural therapeutics against several inflammatory bowel diseases in near future.
**References:**
* Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nature Reviews Immunology. 2009 May;9(5):313.
* Omenetti S, Pizarro TT. The Treg/Th17 axis: a dynamic balance regulated by the gut microbiome. Frontiers in immunology. 2015 Dec 17;6:639.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Antibiotics influence on the gut microbiome functions
Author: BugSpeaks
Published: 2018-06-27
Category: Others
Meta Title: Antibiotics influence on the gut microbiome functions
Meta Description: The current research findings have witnessed a revolution in the biological sciences and, unambiguously, in human health care. In this regard, ‘
URL: https://www.bugspeaks.com/blog/antibiotics-influence-on-the-gut-microbiome-functions
The current research findings have witnessed a revolution in the biological sciences and, unambiguously, in human health care. In this regard, ‘omics’ approaches, such as genomics, proteomics and metabolomics have helped us to unlock the paradigm of microbiome role in improving human health. A balanced maintenance of a good microbiota in our body significantly promotes our health conditions and keep away many metabolic diseases.
The microbiome usually exists in our body through an intimate symbiotic relationship, and the status of microbial flora can be an indicator of a good health. However, the microbiota is constantly being exposed to several factors that might impact on it vigorously. The microbial composition and its alterations vary from one individual to another, and this mainly depends on the microbiota stability of an individual and the nature of a disturbing factor. Under some circumstances, when the environmental or other disturbing factors are very strong, they can induce stress in our body sites, especially the gut location leading to imbalance in the taxonomic configuration of the microbiota, and their functions (a dysbiotic state). For instance, the composition of microbiome is rapidly altered when exposed to antibiotics, and may cause many possible direct or indirect health effects (Figure 1).
Antibiotics are known to kill pathogens. They are classified into natural, semisynthetic and synthetic based on their production mode and origin. For example, the fungus, Penicillium notatum producing penicillins is the base for most of the beta-lactam antibiotics. The treatment of diseases using antibiotics must be considered with a proper dosage. However, when antibiotics are consumed wrongly (not specific to the pathogen) or at high concentrations, i.e., they produce co-lateral effects in our microbiota. It has been reported that broad-spectrum antibiotics affect the richness of bacteria (up to 30%) in the gut microbial community, and cause a significant dysbiotic state. Indeed, the microbiota alterations due to antibiotics may remain for long periods, even up to a few years. Likewise, in infants, it has been evidenced that early exposure to antibiotics significantly reduces the microbial diversity of the infants’ microbiota, with weakening of Bifidobacterium and marked increases of Proteobacteria. Such effects of antibiotics on the gut microbiome have been investigated thoroughly in recent times with the availability of a variety of ‘omic’ approaches. The changes are mainly identified by 16S ribosomal RNA gene sequences analysis and shotgun sequence datasets evaluation. These researches have revealed that, in addition of altering the composition of microbiota, antibiotics upset the gene expression, biomolecules function and inclusive metabolic activities of the gut microbiota. Antibiotics swiftly modify the physiological state and functions of the gut microbiota.
The antibiotic-induced gut microbiome alter the immune and metabolic health of an individual. An immediate threat of an altered gut microbiome is the augmented vulnerability to intestinal infections, which could be because of the newly assimilated pathogens or sudden over-growth and pathogenicity by the already existing opportunistic microbes in the microbial flora. For example, antibiotic-associated diarrhea caused by the nosocomial pathogens, such as Staphylococcus aureus, Klebsiella pneumoniae, and Clostridium difficile is a common incidence. Studies have provided enough evidences to suggest that a chronic infection with C. difficile emerges with a loss of intestinal microbial diversity due to antibiotics. In a similar way, bloodstream infections in immunocompromised patients are increasing due to antibiotics treatments. Researchers have shown that bloodstream infections treated with vancomycin (an antibiotic) set a stage for the intestinal outgrowth of vancomycin-resistant Enterococcus. Studies have revealed that bacteria belonging to a limited set of phyla and genera are strongly affected by antibiotics. Fluoroquinolones, being the most widely prescribed antibiotics are reported to influence our microbiota to a greater extent with the modifications in about 32 major microbial groups. Hence, it has led to the increased resistance worldwide. The major phyla effected include Actinobacteria, Firmicutes, Bacteroidetes, and Proteobacteria. In the major genera, Bifidobacterium, Escherichia, Bacteroides, Ruminococcus, Enterobacter, Faecalibacterium, Bacteroides, Lactobacillus, Eubacterium and Faecalibacterium are highly influenced by antibiotics. The effects of usages of a single antibiotic or a cocktail of antibiotics significantly disturb the microbial community. For example, the treatment with ampicillin alter only the bacteria belonging to the genera Klebsiella, Streptococcus, Bacillus, and Salmonella. Some antibiotics reduce the richness of bacteria community, which is beneficial for human health. For instance, bacteria belonging to the genus, Faecalibacterium, Bifidobacterium, and Blautia to name a few.
Few studies have investigated the influence of antibiotics on our microbes, especially gut microbiome. They suggested the role of microbial diversity and activity, expression of genes, levels of proteins or metabolites produced by microbes in their community. Antibiotics damage and/or destroy bacterial cells and therefore decreases enzymatic activities. However, in the process of antibiotic medications, antibiotic-vulnerable bacteria might be changed into resistant bacteria.
During the treatment course of antibiotics, alterations in specific enzymatic activities have been witnessed, for example the hydrolysis of dietary polysaccharides. Largely, treatments with ampicillin, cefazolin, and sulbactam are reported to alter the activity level of glycoside hydrolases and favor the rapid or disturbed absorption of carbohydrates leading to type-2 diabetes and obesity. Likewise, antibiotics treatment can cause alterations in the microbial metabolite production. In addition, antibiotics bring changes in the fluxes of genes and metabolites and the activities they mediate. A study has detected over 87% of all fecal metabolites in abundance due to streptomycin treatment. This suggests that antibiotics impacts the metabolite fluxes to a different extent. As stated by Ferrer et al. (2017), a comparative ‘omic’ investigation of microbial communities in fecal samples taken at multiple time points from an individual (with a bacterial infection) subjected to β-lactam therapy of ampicillin, sulbactam and cefazolin has revealed antibiotic-associated imbalances in long linear and branched, saturated and unsaturated fatty acids, branched chain amino acids, cholesterol derivatives, vitamins, polyols, sugars, short peptides and polyamines. Likewise, antibiotic intervention during pregnancy causes variations or copiousness of short-chain fatty acids, mostly, propionate, acetate, and butyrate, which are linked to the delivery of premature babies.
Recent years have been noticed with a rapid rise in the cases of multidrug-resistant microorganisms worldwide. This has become a great threat to the use of antibiotics for treating several microbial diseases. The resistance catastrophe is mainly attributed to the misuse and overuse of antibiotics/drugs. The effects of over-use of antibiotics are not only observed in pathogenic gut bacteria, but also in symbiotic healthy microbiota. In this regard, a preventive measures are very necessary to antibiotic-based treatment tactics that do not disturb or unbalance our inhabitant microbial flora, especially good gut bacteria having a dynamic role in health promotion.

[](/media/uploads/blog/159a.jpg)
[

](/media/uploads/blog/159a.jpg)
**Figure-1: Antibiotic effects on the gut microbiota and associated health problems. The main biological consequences of antibiotic-induced dysbioses and the potential diseases that can ensue from them are shown (only diseases with published evidence of association with antibiotic exposure are included). Involved mechanisms are shown in pink-shaded boxes.**
**(Source: Francino, 2016; [https://doi.org/10.3389/fmicb.2015.01543](https://doi.org/10.3389/fmicb.2015.01543)).**
**References:**
* Francino MP. Antibiotics and the human gut microbiome: dysbioses and accumulation of resistances. Frontiers in microbiology. 2016 Jan 12;6:1543.
* Ferrer M, Méndez-García C, Rojo D, Barbas C, Moya A. Antibiotic use and microbiome function. Biochemical pharmacology. 2017 Jun 15;134:114-26.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The Gut Microbiome Influence the Function of Brain and Behaviors in Humans
Author: BugSpeaks
Published: 2018-06-19
Category: Microbiome and Lifestyle
Meta Title: The Gut Microbiome Influence the Function of Brain and Behaviors in Humans
Meta Description: In recent years, the role of microbiome has been increasingly acknowledged for its capability to affect the development and function of the nervous sy
URL: https://www.bugspeaks.com/blog/the-gut-microbiome-influence-the-function-of-brain-and-behaviors-in-humans
In recent years, the role of microbiome has been increasingly acknowledged for its capability to affect the development and function of the nervous system and numerous intricate host behaviors. The gut microbiome modulate the anxiety-like behavior, anxiety, social and communicative behavior, and influence the learning ability and memory. Several animal studies combined with the advanced sequencing, bioinformatics, mass spectrometry, and gnotobiotics approaches have witnessed that various societal behaviors and psychological disorders are associated with the interactions of gut-brain microbiota. However, till now, due to dearth of human studies, the relationship between gut microbiota and different behaviors is yet to be fully established. Interestingly, researchers have documented a definite role of the gut microbial composition influencing on human neurophysiology i.e., the development of brain, gene expression, and neural signaling across brain regions, such as the hippocampus, amygdala, frontal cortex, and hypothalamus.
The gut microbiome represents the collective genomes of all microbes, including bacteria, fungi, protozoa, bacteriophage, and viruses that live in our gut ecosystem. This microbial community includes both beneficial (good bacteria) and possibly harmful bacteria (bad bacteria). In the human gut, over 100 trillion microbes may exist, which is nearly 10 times the total number of cells found in the entire human body. In general, microbes harbor within the gastrointestinal tract soon after the birth and play a vital role in the human health, immunity, and various important biological functions, including the neurophysiology.
As said previously, several researchers have used rodent, chimpanzee, and other wild animal models to understand the gut-brain-microbiota interaction influencing the social and communicative behaviors, and cognitive behaviors. For example, microbiome-related modifications occur in animals due to a variety of volatile chemicals secreted by microbes, which function as olfactory signals. An animal's microbiome could influence on the communication. Likewise, odorant profiles control social isolation against attraction in insects. A high level of bacterial colonization on harvester ants increases the chances of an ant being attacked and driven out of the colony. Related effects were observed in the lower termite (Reticulitermes speratus), signifying that colonization with external microbes produces different scents indicating the entry of invaders into the colony.
In fruit fly, both larvae and adult flies preferred nourishment that was used by other larvae in comparison with new or fresh food. One more study revealed that fruit fly’s mating preference is controlled by the gut microbiota. In hyenas, the social versus lonely grown animals showed varied adjustments in specific microbial taxa. Also, there was a variation in certain volatile fatty acids produced in the social and lonely raised hyenas, which reveals a correlation between microbiome arrangement, odorant profiles, and social behaviors in mammals. Regardless of olfactory message being less prevailing in primates as compared to other mammals, evidences suggest that any deviations in the human skin microflora is linked to changes in odorant profiles. Nonetheless, the microbiome influencing on the secretion of pheromones leading to human behavior changes is yet to be understood in detail.

[](/media/uploads/blog/158a.jpg)
[

](/media/uploads/blog/158a.jpg)
**Figure -1: A schematic representation of the microbiota-gut-brain axis. (Source: Quigley 2018, doi:[10.3390/jcm7010006](http://dx.doi.org/10.3390/jcm7010006))**
In a social approach study, germ-free rats when raised without microbial colonization showed diminished social behavior with microbiome-complemented rats. Juvenile offsprings of antibiotic-treated rats demonstrated different microbiomes with decreased social behaviors. Similarly, another study in mice has revealed that altered gut microbiota profiles is linked to poor social behavior. When mice were subjected to initial life stress (induced neonatal stress) by splitting newborn babies from their mother for 3 hrs per day, there was an increase in the gut abnormality as noticeable from the deviation in microbiota, which was correlated to the changed brain function as part of a bidirectional feedback loop. In Chimpanzee, having repeated social interactions certainly stimulated the microbial biodiversity required to maintain a good gut health.
Studies have witnessed that probiotics positively effect on controlling many stress-related behaviors. For example, chronically stressed rats, when treated with Lactobacillus helveticus NS8 significantly enhanced exploratory behavior in the open field and reduced adrenocorticotropic and corticosterone hormone levels. Matching human trials are deficient, but in a randomized placebo-controlled study, healthy humans treated with Bifidobacterium longum R0175 and L. helveticus R0052 showed lessened anxiety-related scores. Several findings have suggested that specific probiotic treatments may modulate learning and memory behaviors in animals. Thus, these emerging studies are encouraging towards finding a solution to treat various neurodevelopmental diseases characterized by stereotyped behavior, compromised social communication, and related to several medical comorbidities, including immune dysfunction and gastrointestinal problems. However, the exact role of gut microbiota influencing the brain and behavior or how fluctuations in the brain stimulate the colonization of microbial flora in the gut is poorly understood.
Overall, these research findings present exciting promises in solving many neurological disorders of humans, including anxiety and depression. Though many neurological phenotypes have been characterized in response to microbiota depletion/alterations, gnotobiotic interventions, and other microbiome-related stimulus, prime queries concerning how changes in microbiome modulate the host behaviors remain unanswered. In this regard, future clinical studies directed towards understanding the mechanisms underlying the gut-microbiota-brain axis using novel approaches are highly necessary.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Weight loss for a better health through gut microbiota analysis
Author: BugSpeaks
Published: 2018-06-13
Category: Microbiome and Lifestyle
Meta Title: Weight loss for a better health through gut microbiota analysis
Meta Description: Overweight and obesity are defined as disproportionate body fat accumulation, and considered as a global epidemic. As per the World Health Organizatio
URL: https://www.bugspeaks.com/blog/weight-loss-for-a-better-health-through-gut-microbiota-analysis
Overweight and obesity are defined as disproportionate body fat accumulation, and considered as a global epidemic. As per the World Health Organization report, over 1.9 billion adults remained as overweight in 2014, and among them, in excess of 600 million were found to be obese. The prevailing obesity is correlated to a number of health-threatening illnesses, such as arterial hypertension, coronary heart disease, stroke, type 2 diabetes mellitus, arthritis, and asthma to name a few. Besides, this condition has significant economic effects, i.e., increase of medical expenses for its treatment or related conditions, and secondarily it causes a reduced efficiency, incapacities and early mortality in patients. Typically, obesity is due to over consumption of high calorie food in addition to a sedentary daily life. Factors, such as including genetic predisposition, increased parent age, sleep deficiency, endocrine disorders, drug iatrogenesis and epigenetic changes also contribute to the onset of obesity.
In recent times, the gut microbiome constituting the overall collection of the genetic material of microorganisms inhabiting in the gut region has been observed to have implications in triggering obesity. Interestingly, the gut bacteria constitute nearly 250-800 times more number of genes than human genes. These bacterial genes produce substances/constituents that get into the human bloodstream, and upset the body’s biochemistry. These gut bacteria are known to play a vital role in many biological processes, such as digestion, metabolism, and absorption. Thus, they involve and contribute to the development of obesity and other metabolic disorders, which is attributed to the fact that they have the ability to produce more energy from the diet, regulate and influence the fatty acid composition, encourage inflammation, and also play a part in regulating appetite via the gut–brain axis.
Various pre-clinical and clinical investigations have correlated the gut microbiome to obesity. For example, from the data of animal studies, it is evidently proved that mice which are germfree can resist obesity, however, upon introducing gut microbes into their body, the animals increased their uptake of calories from the diet and advanced towards fat deposition, and became insulin resistant. Likewise, when the gut microbiota of obese mice were colonized into normal mice, they were able to harvest energy more proficiently and accumulated body fat rapidly. The analysis of 16S rRNA gene sequenced data of the gut microbiota obtained from the distal intestine in mice indicated that a distinctive pattern occurs between obese and lean animals with respect to the most important bacterial phyla, Bacteroidetes and Firmicutes. Similarly, there exists a declined population of Bacteroidetes in the obese population. However, in the same animals, an increased number of bacterial population belonging to the phylum, Firmicutes is observed. Also, clinical findings have supported the existence of a close relationship between the gut microbiome and obesity. But, the existence of dissimilarities in the gut microbiota at the phylum level between obese and lean persons still remains as contradictory, and yet to be clarified through investigations. Hence, the gut microbiome have a critical role in causing obesity, but the causative versus substantial relationship, and the degree of its influence in humans, still remains ambiguous.
At present, obesity can be addressed through the therapeutic preferences, such as lifestyle modifications (diet, physical exercises and behavioral therapies), pharmacological treatments, and weight loss surgery in the case of severity. Lifestyle modifications may help to lose the original bodyweight up to 5–10% on a long-term basis, though there is a possibility of regaining 30-50% of the lost weight within a few years. Pharmacological treatments endorse uncertain weight loss, nevertheless lead to side effects. In severe cases, Bariatric surgery is the preferred option, which promotes a significant weight loss and reduce mortality. Yet, a large number of patients experience reduced weight-loss effects, and after the treatment, the patients may regain weight up to 75% over a period of time. Thus, there is a need to use alternative approaches for overcoming these issues and benefit patients to reduce and accomplish a healthy weight. In this regard, the manipulation of gut microbiota is a very useful approach.

[](/media/uploads/blog/157a.jpg)
[

](/media/uploads/blog/157a.jpg)
Imbalances in the gut microbiota are central to the pathogenesis of obesity. Hence, an optimal gut microbiota balance could be helpful in fixing this problem through a precise diet choice. Including lots of fiber, healthy proteins and fats in a diet improves the gut microbiome. Good fatty acids, such as omega−3 fatty acids and monounsaturated fatty acids, i.e., extra-virgin olive oil, avocados oil or almond oil improves good gut microflora, while inflammatory omega-6 fatty acids, i.e., vegetable oils (corn, safflower, cottonseed, sunflower, and soybean oils), promote the growth of bad microbes leading to diseases and weight gain. These bad microbes secrete toxins called lipopolysacchardies that trigger inflammation, pre-diabetes (insulin resistance) and consequently, stimulate weight gain. Even chronic stress and sleep deprivation can contribute to gut microbial imbalance. Hence, practicing a healthy life schedule will favor the growth of good microbes in the gut. Overall, the percentage of good bacteria in the gut decides on how much weight a person may lose or gain, and under what conditions. The best way to improve the gut microbiota with good bacteria is to eat a healthy diet having increased fiber, good fats, avoiding unprocessed and unrefined foods, probiotic supplementation, and reducing bad bugs with medication.
Already, microbiomics has become a new frontier in medicine and providing many health solutions. Therefore, the genomic analysis of gut microbiota can precisely suggest on the intake of food, and modifications required in the daily life for an overweight or obese persons in order to lose weight, and remain fit and healthy. The clinical application of weight modifications through the gut microbiota analysis still remains unclear. Hence, additional investigations are warranted to recommend its use as a therapeutic tool in the current clinical practice of treating obese and overweight individuals.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The role of Gut Microbiome in the Wellness World
Author: BugSpeaks
Published: 2018-06-05
Category: Microbiome and Lifestyle
Meta Title: The role of Gut Microbiome in the Wellness World
Meta Description: The indigenous microbial communities are represented as microbiota, while the collection of microorganisms in the host environment where they inhabit
URL: https://www.bugspeaks.com/blog/the-role-of-gut-microbiome-in-the-wellness-world
The indigenous microbial communities are represented as microbiota, while the collection of microorganisms in the host environment where they inhabit and their genomic contents is known as microbiome. Recent advancements have transformed clinicians’ ideas about the role of microorganisms in human health and diseases. Interestingly, it has been realized that the microbiota inhabiting in our body provides a necessary ecosystem for mutual benefits, such as improving the digestion process, protection against pathogens, bioconversion of nutrients, etc. It has been reported that several trillions of microbial cells occupy different parts of the human body. Our body constitutes more bacterial cells than human cells, and they considerably affect our day-to-day lives. Therefore, the microbiome influence on human diseases and vice versa can be wide-ranging. For instance, the gut microbiota can modulate the functions of the gastrointestinal tract, produce chemicals, improve immune functions and even control the behavior. Likewise, a chronic lung disease may modify the composition of lung microbiome, and successively impact on immunity and host defense, leading to additional exacerbations of diseases. Also, bacteria, such as _Lactobacillus_, _Bacteroides_ _and_ _Bifidobacterium_ synthesize secondary bile acids, which are crucial for the lipid transport. As a beneficial bacteria, _Bifidobacterium_ generates Vitamins (B12, K, biotin, thiamine). The microbial diversity is very high in the gut region as compared to any other body parts (the skin, gums, and vaginal sites). Hence, most of the current investigations are focused on the gut microbiome and its significant role in establishing good health. The gut microbiome varies from one person to another, though they have some common illnesses like Obesity, Type 2 Diabetes or Inflammatory Bowel Disease.
A disease can be anticipated with the loss of helpful functions by the gut microbiome or by invading microbes in the gut. In other words, a disturbed gut microbiome leads to several diseases, particularly which are allied with our lifestyle aspects. Investigations have proved the connection between these microbes in our gut and many chronic disorders, such as Obesity, Diabetes, Coronary Artery Disease, Parkinson’s, Alzheimer’s, Lupus, Psoriasis, Autism, etc. Therefore, a proper nourishment of our gut microbiota can restore health conditions and make us feel happy. The consumption of healthy diet included with prebiotics and probiotics, avoiding antibiotics intake unless very obligatory, and a regular exercise may be helpful in improving our gut microbiota. In humans, the gut microbiome has co-evolved with its host since from ages and, hence, it performs a variety of crucial activities in the host, including digestion, nutrition, detoxification, body defense, improving immunity, and disease mediation. Accordingly, the human gut inhabits a wide range of microbes with high species diversity, with most of them being _Bacteroidetes_ and _Firmicutes._
The occurrence of harmful bacteria in our gut may cause a leaky gut syndrome, which happens due to the high permeability of the intestinal walls, causing leakage of undigested food particles, bacteria, and many other substances into the nearby tissues. The leaky gut syndrome is directly connected with several health problems, such as chronic fatigue, Stomach aches, Insomnia, Inflammatory Bowel Syndrome, Constipation, Diarrhea, Headaches, Depression, Cardiac problems, Pancreatic illness, etc. Our brain is directly connected to the gut microbiome, which is known to have a role in controlling or regulating our body functions. It may act autonomously (as another brain), or in combination with the brain. Hence, an unbalanced gut microbiota may trigger variations in our mood and behavior. Some of the neurological problems, such as Depression, Anxiety, Autism, and anger are correlated with health conditions of gut microbiome. A healthy microbiome can defend disease causing microbes in the body because, the beneficial microbes secrete substances or create the detrimental environment that can resist the growth of pathogens or even destroy them. For instance, volatile fatty acids, and its other byproducts produced by healthy gut microbes prevent the survival of fungi and yeast. Likewise, the pH in the digestive tract is also balanced by the microbiome so that many toxic bacteria like _Shigella, E. coli,_ _and_ _Salmonella_ _cannot survive in that condition. These bacteria are known to cause_ intestinal diseases, Diarrhea, and Food poisoning.

[](/media/uploads/blog/156a.jpg)
[

](/media/uploads/blog/156a.jpg)
Following good practices and lifestyle habits can keep our gut microbiota healthy, which in turn can impact on a healthy life. The conservation of a varied and prospering beneficial gut microbial population helps in keeping away harmful bacteria due to higher competition for the same nutrients and colonization sites. A diet, particularly rich in fibers is highly advised to maintain a healthy gut microbiota population. In addition, certain Prebiotics and Probiotic foods, such as Yogurt, Kefir, Cottage Cheese, Natto, Tempeh, etc., will certainly enhance our gut microbiota population and health. However, some of the bad lifestyle habits need to be avoided as they destroy both good microbiome and their diversity. It is suggested to avoid these bad habits such as, overuse of antibiotics, antacids, laxatives, and painkillers; drinking chlorinated water, eating sterilized foods or foods with altered fats; high intake of carbohydrates to name a few.
Molecular technologies, such as 16S ribosomal RNA sequencing, Microbiomics, Metatranscriptomics, Metaproteomics, and Metabolomics analysis have impressively improved our knowledge of the diversity, complexity, and functions of the gut microbiome within and between individuals. These high throughput technologies have delivered us with the capability to gain insights into the genes being expressed vigorously in the complex microbial communities. It has enabled us to elucidate the functional changes in directing the microbiome functions at specific environments, its interactions with the host, and functional changes involved in translating a _healthy_ microbiome in the direction of a disease-driving configuration. Thus, the gut microbiome genomic analysis can provide a clue to the role of the gut microbiota and its impact on our health.
The analysis of different genes expressed by microbes in the gut facilitates us to identify the types of metabolites that they will produce. It helps in determining the microbiome role in our body. However, just getting a data may not be useful. We have to understand about the type of genes expressed, the levels of metabolites produced and their functions, and how to improve them through a defined diet, supplementations, or modifying lifestyle activities to improve the gut microbiota of an individual. Overall, this is a personalized medicine approach to help patients to improve their gut microbiome health to overcome many diseases. It will help us to precisely decide what, how and when to take right food in order to develop wellness in a short time. Based on the microbiomics data, a patient can be accurately treated with a proper diet and lifestyle modifications. In specific, this Gut microbiome test will help us to know about the absence of beneficial bacteria and good metabolites in our gut, and the need of probiotics, carbs, proteins, and fats or nutritional endorsements to restore good health in a personalized way.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The Human Gut Microbiome and Microbiomics: Progress Towards Personalized Healthcare
Author: BugSpeaks
Published: 2018-05-28
Category: General
Meta Title: The Human Gut Microbiome and Microbiomics: Progress Towards Personalized Healthcare
Meta Description: The human body comprises 10-100 trillions of microbial cells, including archaea, bacteria, viruses, eukaryotes. The microbiome includes the combined g
URL: https://www.bugspeaks.com/blog/the-human-gut-microbiome-and-microbiomics-progress-towards-personalized-healthcare
The human body comprises 10-100 trillions of microbial cells, including archaea, bacteria, viruses, eukaryotes. The microbiome includes the combined genetic material of all microbes in a specific environment i.e., microbiota. It plays a significant role in improving human health and preventing diseases in many ways, though their mechanisms are yet to be probed in detail. The occurrence and function of microbiota differ depending on several factors, such as ages, sexes, races, diets, and change in the locations of the host. The microbiome profoundly restores normal phenotypes in the host. The past few years have investigated the microbiome in detail, and the results have tremendously reshaped our knowledge of human biology. Some of the innovative insights range from the understanding of how microbial cells intervene digestion activities, disease initiation, and progressions (e.g., inflammatory bowel disease) to unforeseen links with autism, depression, anxiety/mood disorders, Alzheimer's, Parkinson's disease, etc.
So far, studies have discovered around 10,000 microbial species (mainly bacteria) in the human body and mostly, their occurrence is predominant in the gut. Interestingly, the gut microbiota contains about 150 times more number of genes than that are observed in the whole human genome. Thus, the gut microbiome is sometimes recognized as an ‘essential organ’ because it produces products, it responds to the environment, and interacts with other systems. It is believed that the impact of these bacterial genomic DNA on our health might be more than our own genomic DNA. Likewise, it has been realized that the microbiota of gut and brain are intricately associated. There is an exchange of messengers/signals between the brain and gut microbiota. The entire body cells are influenced by the microbiome in one or the other way. In specific, it affects our mood, metabolism, libido, immune system, sensitivity, and even the clarity of your thoughts. Scientific evidences have shown that human health and diseases are dictated by what goes on in the microbiome of the gut. Diseases such as inflammatory bowel disease (IBD), obesity, diabetes, cardiovascular diseases, atherosclerosis, nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), cirrhosis, and colorectal and liver cancer have been connected to the activities of the human microbiota (Figure 1).

[](/media/uploads/blog/155a.jpg)
[

](/media/uploads/blog/155a.jpg)
**Figure 1. Human microbial symbiosis has a close relationship with diseases of different systems (Adopted from Wang et al. 2017; [https://doi.org/10.1016/J.ENG.2017.01.008](https://doi.org/10.1016/J.ENG.2017.01.008)).**
Overall, the diversity in microbiome can lead to both health and illness. Commensal microbes colonize the host soon after their birth. This microbiota community gradually develops with the host growth depending on varying ecosystems. Over time, these host-microbe interactions lead to a beneficial association, and symbiotically offer many advantages. For example, improvements in digestion, supply of essential nutrients, metabolizing indigestible compounds, and to shield against the invading pathogens. Knowing about the microbiota can help in preventing possible diseases in humans. However, their complex nature makes it difficult to study them. The standard microbial culture methods may not be sufficient enough to isolate and explore all microbes present, because some may not grow or require special growth conditions that are yet to be standardized. Later, culture independent methods for evaluating microbes was developed based on the targeted sequencing of 16S and 5S ribosomal RNA (rRNA) genes which showed differences for each species and effective in identifying organisms. Over the last decade, human microbiome researches have been transformed by using next-generation sequencing (NGS) technologies. These high-throughput sequencing allow us to study complex bacterial systems without the requirement of cloning individual genes. The targeted sequencing of 16S rRNA, 18S rRNA, 28S rRNA genes, and internal transcribed spacer (ITS) regions, and whole microbial genome of microbial communities all at once (metagenomics) is very useful in the identification of microbes and to track the functional activity of the entire community. The increasing number of such metagenomic studies of microbiomes in humans is a new hope of its potential benefits in the clinical practice.
Recent studies have witnessed the occurrence of human genetic variations as influenced by interpersonal dissimilarities in microbiomes. Thus, human genes might influence health by promoting beneficial microbiomes. The heritability study of the gut microbiota has revealed that subsets of microorganism’s abundance are genetically determined by the host. For example, a genetic change enables lactase persistence in adulthood. Interestingly, the genome-wide association studies (GWASs) of the microbiome has revealed that there is a relationship between the host genotype, consumption of milk, and gut beneficial bacteria (Bifidobacteria). It has been noticed that a single nucleotide polymorphism (SNPs) near the LCT gene on chromosome 2 and Bifidobacterium in the fecal microbiota has a close association. Bifidobacteria utilizes the lactose of milk as an energy source. This depends on the genotype of the host and diet interaction (Figure 2).

[](/media/uploads/blog/155b.jpg)
[

](/media/uploads/blog/155b.jpg)
**Figure 2: Interaction between the human lactase nonpersister genotype, Bifidobacterium abundance, and lactose in the host’s diet. Genetic variants near the LCT gene are associated with lactase persistence and in several microbiome GWASs were recently associated with Bifidobacterium relative abundance in fecal samples. The association may function according to the following scenario: (a) If an individual is a lactase persister and consumes lactose, the lactose is typically broken down into glucose and galactose in the small intestine by host lactase. (b) However, if the individual is a lactase nonpersister and consumes lactose, it travels to the large intestine, where it is fermented by lactose-utilizing bacteria, which includes Bifidobacterium. If Bifidobacteria are present, then the presence of lactose promotes their abundance. In individuals who do not consume lactose, Bifidobacterium abundance remains unaffected by their lactase persistence status (not shown). (Adopted from Goodrich et al. 2016; [https://doi.org/10.1016/j.cell.2014.06.037](https://doi.org/10.1016/j.cell.2014.06.037)).**
Likewise, GWASs has revealed that immunity related genes are influenced by the gut microbiome. Evidences suggest that genetic variations might act on digestive tract tissues to disturb the composition of the gut microbiome. Further, genetic variants in the genes, PLD1 and LINGO2 are associated with obesity. Based on the analysis of 16S transcripts, the phylogeny of microbial community has been revealed. The most active microbiota included Prevotellaceae, Lachnospiraceae, Bacteroidaceae, Ruminococcaceae, and Rickenellaceae. RNAseq analysis of bacteriophage populations in the periodontal microbiota showed the abundance of oral phages highly expressed in persons having good periodontal health. More recently, 16S rRNA gene-based approach has confirmed the predominant occurrence of Corynebacterium spp., Staphylococcus spp. and Propionibacterium spp. on healthy human skin.
Thus, knowledge on the microbiomics help to improve human health conditions. A defined diet can influence on the microbiome and restore health. Likewise, a proper use of chemicals/drugs in disease management is another factor to be considered. For example, both foods and drugs are used in treating type 2 diabetes. It is generally advised to avoid consuming simple carbohydrates to control blood sugar. Nevertheless, there exist inter-individual glycemic variations in response to diet, and some of which can be described on the basis of the individual’s microbiome. Similarly, the human microbiome plays a significant role in genetic diversity, modify disease conditions, impart immunity, provide nutrients, influences assimilation, and modulates drug interactions. Thus, intake of probiotics or beneficial bacteria (Lactobacillus, Bifidobacterium, Faecalibacterium prausnitzii, Akkermansia muciniphila, Pediococcus pentosaceus, etc.) may be useful in preventing or treating certain diseases, though most of them cannot be cultured presently. As a diagnostic tool, microbiomics is employed for detecting and measuring the levels of biomarkers i.e., cholesterol, cortisol, glucose, and creatine kinease occurring in blood which are known to indicate about diseases or infections. These novel diagnostic methods will allow clinicians to prescribe right drugs or diet to restore patient’s health.
Future microbiome investigations should involve many more new techniques to predict the functions of microbiotas, their crucial role in human development, and the mechanisms of various diseases, such as metabolic diseases, liver diseases, cancers, psychiatric diseases, etc. In this regard, many new-startups and biopharmaceutical companies, including Luecine Rich Bio Pvt. Ltd are exploring the field of microbiomics to develop novel therapeutics, diagnostic platforms and to provide clinical genomic services. A detailed data on this metagenomics might be very useful in the microbiome-based diagnosis of diseases and for the development of novel treatment strategies in the forthcoming personalized medicine era.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The microbiome role in human health and diseases
Author: BugSpeaks
Published: 2018-03-14
Category: Microbiome
Meta Title: The microbiome role in human health and diseases
Meta Description: In recent years, studies on the native microflora (microbiota) of the host environment has profoundly renewed our knowledge of human biology, health a
URL: https://www.bugspeaks.com/blog/the-microbiome-role-in-human-health-and-diseases
In recent years, studies on the native microflora (microbiota) of the host environment has profoundly renewed our knowledge of human biology, health and diseases. The term, microbiome is generally referred to the pool of genes present within a community of microbes including bacteria, fungi, archaea, protozoa, and viruses found over and inside the body. While, studies aiming to describe the occurrence and function of uncultured microbiota are frequently stated collectively as metagenomics. The life style of the host including diet habits, medical interventions, environment, and disease conditions can lead to variations in the microbiome. The structural and functional diversity of microbes allow them to survive symbiotically for a long-term in hosts. The microbiome contributes to the vital functions of the body such as metabolism, immunologic activity, energy homeostasis, gut epithelial health and neurodevelopment of the gut brain axis. They influence or mediate the process of digestion in gut, regulate immune system, protect against other pathogenic bacteria and secrete vitamins including Thiamine, Vitamins B12, Vitamin K, etc.. Overall, microbiota improves human health and helps to combat diseases. For instance, gut microbes make drugs to work better. On the other hand, they play a significant role in causing several disease including inflammatory bowel disease, autism, Parkinson's disease, type 2 diabetes, cancer, and depression. In addition, they can affect therapeutic approaches by reducing drug’s effectiveness.
The advent of next generation sequencing (NGS) technologies, metagenomics, transcriptomics, metabolomics, and analytical methods combined with the latest analytical software and the unified databases on experimental results have revolutionized the emergence of a new field, the microbiomics. It aims to identify the microbiota, genome DNA analysis, elucidate the microbiome and host interactions, and determine its role in disease pathogenicity. Basically, these approaches are used to amplify and sequence the specific microbial DNA segments followed by integrating computational analysis to identify and diversify microbes on the basis of sequence similarity and comparing with reference genomic databases of microbes. In specific, 16S rDNA (ribosome DNA) is used as the target for sequencing bacterial genome as this region is highly conserved with hypervariability and vary amongst species. In recent times, whole-genome shotgun sequencing (WGS) is the choice of analysis. This is because, WGS permits taxonomic scrutiny, investigations on the microbial gene inventories, and documents non-bacterial microorganisms that are omitted from 16S rDNA sequencing (Figure 1). Initiatives of various projects such as the Metagenomics of the Human Intestinal Tract (MetaHIT), Human Microbiome Project (HMP) have quickly enriched the microbial genomic data. This mammoth and wealthy data rapidly led to overcome computational bottlenecks and driven to develop tools such as Quantitative Insights into Microbial Ecology (QIIME) and Visualization and Analysis of Microbial Population Structures (VAMPS) which can handle big volumes of genomic data and can function in different settings ranging from supercomputers to laptops. Estimates suggest that the human body is associated with approximately 2-20 million number of microbial genes. The studies have reported the ratio of microbial cells to human cells to be 10:1. This suggest the profound impact of microbes and their genomic functions on human biology. Thus, exploration of their genomic DNA can offer exciting prospects for developing unique therapies against many chronic diseases.

[](/media/uploads/blog/148a.jpg)
[

](/media/uploads/blog/148a.jpg)
**Figure 1. Bioinformatic methods for functional metagenomics.**
(Source: Adopted from Morgan and Huttenhower (2012); [https://doi.org/10.1371/journal.pcbi.1002808](https://doi.org/10.1371/journal.pcbi.1002808))
The microbes living within us are not invaders, but helpful colonizers and play an essential role in human development, nutrition and immunity. Several autoimmune diseases including diabetes, muscular dystrophy, rheumatoid arthritis, and multiple sclerosis, and fibromyalgia are linked with dysfunctioning of the microbiome. The higher number of Lactobacillus counts in pregnancy is linked to protect the developing infant from allergic reactions. Also, studies have suggested that _Lactobacillus_\-dominated populations are protected from bacterial vaginosis. A failure in the vaginal microbiome may increase the risk of pre-term birth. Thus, the mother’s microbiome may influence her children’s health. Likewise, disease-causing microorganisms add over time, and change the gene action and metabolic functions. This may lead to irregular immune responses against body’s own constituents and tissues and results in autoimmune diseases. Overall, varied microbiome of an individual can make them susceptible to infectious diseases and lead to Crohn’s disease, irritable bowel syndrome and other chronic disorders of the intestine. Some microbial communities regulate the response of a person treated with a drug. When, a broad-range antimicrobials are taken both good and bad bacteria are killed. This might lead to the antibiotic resistance crisis. Therefore, there is a need to accurately identify pathogenic microbes so as to know their role in contributing diseases and strategize treatments which can selectively kill only pathogenic bacteria. Therefore, mapping the human microbiome is necessary to discover more microbial species and their genes that are associated with definite human health conditions. The microbiome varies depending on the population, race, geography, and diet habits. The microbiomics can help clinicians to prescribe the right drugs and diet to manage diseases effectively. The addition of probiotics is one of the nutritional supplementation to improve the microbiota of the gut. However, the application of microbiome in therapies is challenging because microbiomes are distinctive to every single person and vary over time. Thus, it complicates the use of the microbiome for both diagnosis and to design a therapy. At present, research studies are aimed to investigate on the following aspects:
* Nutrition in diet affecting the microbiome.
* The microbiome affecting immune response and contributing to diseases.
* Antibiotics and the microbiome interactions to overcome drug-resistance.
* The microbial response to various drugs.
* Altering the microbiome to improve health conditions.
The microbiome is very diverse, and complex and many fundamental questions remain unanswered. Improved computational approaches for the metagenome assembly/analysis are the urgent need in this regard. Moreover, their relations to metabolism, epigenetics, host genetics, and immunology can facilitate to unravel their complex relationships with human health and diseases. This can be better exploited for curing several chronic diseases. Some of the ethical concerns of the microbiomic research include; data being obtained from a typical sample of the population, informing about the consent to samples collection, data sharing, protecting privacy, invasiveness of taking the samples, etc. In future, investigations should aim to explain the mechanisms of interactions between the microbiome and host, decipher the microbiome growth during host growth and development, its impact on health conditions, elucidate the role of its pathogenicity, and measure the feasibility of diagnostic examinations, and target the therapies against human diseases.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Let Food be thy Medicine
Author: BugSpeaks
Published: 2018-01-23
Category: Nutrition
Meta Title: Let Food be thy Medicine
Meta Description: About the Speaker:
Dr. Sripriya Venkiteswaran, is a nutritionist by profession, and an avid follower of all things that are nutritious for the past
URL: https://www.bugspeaks.com/blog/let-food-be-thy-medicine
**About the Speaker:**
Dr. Sripriya Venkiteswaran, is a nutritionist by profession, and an avid follower of all things that are nutritious for the past two decades. She has a bachelor’s degree in Nutrition, a PG Diploma in Clinical Nutrition and Dietetics, followed by a PhD in Nutritional Sciences.
Her interest for the molecular aspects of nutrition were ignited during her PhD from Rutgers University, which has turned in to her core strength and passion. In this TEDx\_IBA\_Bangalore talk she shares her views and knowledge about nutritional facts and how food itself can heal as well as shape the body.
**Highlights**
* How food, in extension nutrition, affects your body from within (at genome level)
* How most regulatory aspects or dietary recommendations have been focused around "the general" or concentrated on "the average human being".
* Why this approach has largely been unsuccessful
* Later, she proposes that, since everyone is genetically unique, the way we respond to various foods would also be unique, which in extension have largely been the root cause why "one size fits them all" approach does not work.
* She explains the two-way effect of gene-to-nutrition and nutrition-to-gene on our body, providing some compelling examples of Agouti and MTHFR genes.
* With expanding prospects of genomic profiling, she feels excited about the future, but she also urges both scientific community to gather comprehensive data before customizing the nutritional recommendations.
* She also touches upon several other topics pertinent like patient persuasion, status of nutritionists within the medical community and even reviving our (Indian) traditional nutritional practices advocated with genomic data.
* She concludes reiterating the point that 'your genotype is also as important as your appetite".
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Are we humans or are we bacteria
Author: BugSpeaks
Published: 2017-03-15
Category: Microbiome
Meta Title: Are we humans or are we bacteria
Meta Description: Josh Neufeld is an Associate Professor, University of Waterloo in this TED video explains the abundance of microbes in our body and its function.
URL: https://www.bugspeaks.com/blog/are-we-humans-or-are-we-bacteria
Josh Neufeld is an Associate Professor, University of Waterloo in this TED video explains the abundance of microbes in our body and its function.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Gut microbiome and inflammation
Author: BugSpeaks
Published: 2017-03-14
Category: Microbiome and Disease
Meta Title: Gut microbiome and inflammation
Meta Description: In this short video, Prof. Adiel Tel-Oren explains the inflamed gut and how to eliminate risks of intestinal diseases.
URL: https://www.bugspeaks.com/blog/gut-microbiome-and-inflammation
In this short video, Prof. Adiel Tel-Oren explains the inflamed gut and how to eliminate risks of intestinal diseases.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## How nutrition shapes gut microbiota
Author: BugSpeaks
Published: 2017-03-13
Category: Nutrition
Meta Title: How nutrition shapes gut microbiota
Meta Description: How Nutrition Can Shape Gut Microbiota and its Implications in the Autoimmunity Epidemics is explained here by Alesso Fasano, MD Massachusetts
Genera
URL: https://www.bugspeaks.com/blog/how-nutrition-shapes-gut-microbiota
How Nutrition Can Shape Gut Microbiota and its Implications in the Autoimmunity Epidemics is explained here by Alesso Fasano, MD Massachusetts
General Hospital for Children (MGHfC)
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The brain in your gut
Author: BugSpeaks
Published: 2017-03-10
Category: Microbiome and Lifestyle
Meta Title: The brain in your gut
Meta Description: Did you know you have functioning neurons in your intestines -- about a hundred millions of them? Food scientist Heribert Watzke tells us about the &q
URL: https://www.bugspeaks.com/blog/the-brain-in-your-gut
Did you know you have functioning neurons in your intestines -- about a hundred millions of them? Food scientist Heribert Watzke tells us about the "hidden brain" in our gut and the surprising things it makes us feel in this TED talk.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## All About Bacteria
Author: BugSpeaks
Published: 2017-03-09
Category: Microbiome
Meta Title: All About Bacteria
Meta Description: Ravi Mantha, in this TED video speaks about health disorders that occur to human being on the theme "All about bacteria" and human health sy
URL: https://www.bugspeaks.com/blog/all-about-bacteria
Ravi Mantha, in this TED video speaks about health disorders that occur to human being on the theme "All about bacteria" and human health system.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Microbiome and the Brain Interactions
Author: BugSpeaks
Published: 2017-03-08
Category: Microbiome and Lifestyle
Meta Title: Microbiome and the Brain Interactions
Meta Description: Dr. Emeran Mayer, David Geffen School of Medicine at UCLA in this video talks about the fascinating world of microbes and its interaction with brain.
URL: https://www.bugspeaks.com/blog/microbiome-and-the-brain-interactions
Dr. Emeran Mayer, David Geffen School of Medicine at UCLA in this video talks about the fascinating world of microbes and its interaction with brain. The neurobiological aspects of the gut-brain axis are very well explained in this video.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## What happens in Vagus, does not stay in Vagus!!
Author: BugSpeaks
Published: 2017-03-07
Category: Microbiome and Lifestyle
Meta Title: What happens in Vagus, does not stay in Vagus!!
Meta Description: So, it’s been two months past the new year. We all are coming back to our normal selves, we are eating (binging) whatever we want to, not so kee
URL: https://www.bugspeaks.com/blog/what-happens-in-vagus-does-not-stay-in-vagus
So, it’s been two months past the new year. We all are coming back to our normal selves, we are eating (binging) whatever we want to, not so keen on working out or even waking up early...for that matter.
Now that we are enlightened about new year diets, workout regimes & resolutions, and have understood these to be just myths, we can actually focus on doing what we do best
"**sit and ponder about everything**"
Everything, from menial questions about career, marriage, relationships ... to ... more serious questions like - should I cook today or order pizza?... then to ... potentially life changing questions like... Why am I fat ?!!
We always talk about stuff that we eat going directly to our hips (especially for us girls, right?!)
Here is an interesting observation:
What if, the food we eat directly goes to (or affects) our head?
hmmm... Now that's a thing to ponder upon,
I mean how's that even possible?
Well, you might have heard or even felt that, when you are in stress you eat more, or when you are tensed you have gastric problems, or when you are hungry for long duration you get a headache. So, you know these things are somehow connected, and mostly seem to be related to the food that you eat.
But, in fact, this relationship, between your behavior and your food (more importantly the other way around), is surviving (read hanging in there) because of the tiny tenants in your body.
_**And this is where the story begins...**_
When you are in your mother's womb there are no microorganisms in your body, and you are virtually sterile, inside out!!
As a new-born, you are exposed, for the first time, to a wide array of microbes from a variety of sources, including maternal bacteria. The process of birth exposes you to the first microbes from your mother's vaginal tract or stomach, depending on the mode of birth. (fascinating? or you feeling eeeww?)
Later, your mother's milk gives you the major share of microbes, that actually help you build your immunity. In fact, [a recent study](https://www.ncbi.nlm.nih.gov/pubmed/23401408) talks about how infants born by cesarean section are at increased risk of asthma, obesity and Type 1 diabetes, as compared to babies born through normal labor and exposed to the maternal vaginal microbes.
As and when you start eating different kinds of foods, drinking beverages, use a specific kind of bathing soap etc., the microbiome (total microbes on and) in your body gets enriched. More specifically, your gut microbiota gets developed with every exposure to different environments. In fact, what you eat, drink, smoke, or gets exposed to, all defines the microbiota of your gut.

[](/media/uploads/blog/100a.jpg)
[

](/media/uploads/blog/100a.jpg)
I still remember the day, when I had my first beer... and my gut microbes were like... bring it on girl!!
Another interesting fact is that, since diet and food habits are largely an extension of your culture, your gut microbiome differs from American's or Chinese, given that we all are culturally world apart, and so is our lifestyle and dietary habits.
_**The other side of the story…**_
On one end, the food you eat and drink affects the composition of your gut microbiota. However, over time, the enriched and developed gut microbiota influence the digestion of specific food you eat, indirectly influencing your diet. So, lot of these gut bacteria influence and more importantly help you digest food in an efficient way.
Another interesting fact is that, whenever these good bacteria goes low in number, it gives way for over growth of bad microbes like Clostridium, E. coli etc., that causes diseases like diarrhea, colitis, etc.
And why would these good bacteria go low in number?
Well... it can be due to any reason from imbalanced or untimely diet to exposure to antibiotics or smoking. [One study found](https://www.ncbi.nlm.nih.gov/pubmed/3846592) that after a single treatment of intravenous antibiotics, fecal bacteria demonstrated a significant change in the variety and abundance of bacterial strains.
Overall, the food that we eat influence the gut microbiota and the microbiota influence the what we eat - requiring a fine balance between the two.
Okay, now we are drifting
_**Let's come back to the point where food and head are connected**_
Studies, involving college kids (the junk eating generation), eating burgers and fries, guzzling cokes and sodas, observed that these students felt lethargic, largely unwell, depressed, with general fatigue, disturbed / unfocused mind, just after a few weeks of continuous ingestion of the junk diet. Testing their gut revealed that, and I quote “gut microbes (the microbiome) had been devastated”, with potential disease causing firmicutes replacing good Bacteroidetes, as the dominant type, while the friendly Bifidobacteria, that suppress various kinds of inflammation, halved in its abundance being the clearest marker of an unhealthy gut.
The question is why? and How does what you eat .... affecting your gut microbes... affect your state of mind?
The human gut microbiome is so dynamic and complex, that it consists of approximately 1 kg of bacteria in an average adult, and is approximately the weight of the human brain. This microbiota produce a myriad of neuroactive compounds, regulating everything from your behavior to cognitive function, social interaction to stress management. It regulates these brain functions by steering the "Vagus Nerve".
This thing called 'Vagus nerve' (Not "Vegas and Casinos") within your nervous system, is a part of cranial nerves unit, that forms an extensive network, linking your gut and brain. Creatively enough, termed as [the gut brain axis](https://fivethirtyeight.com/features/gut-week-gut-brain-axis-can-fixing-my-stomach-fix-me/), it is the sole reason for all the food and head related things. This system, largely involving the Vagus nerve, controls the body’s unconscious actions, such as digestion, excretion, and sexual arousal.

[](/media/uploads/blog/100b.jpg)
[

](/media/uploads/blog/100b.jpg)
In the absence of certain (good) microbes, due to imbalanced diet (read junk eating), the neuroactive compounds, that regulate the above said unconscious actions, are profoundly altered. It turns out, what you eat changes the gut ecosystem, in turn resulting in altered vagal activity.
_**But, what happens in Vagus does not stay in Vagus!!**_
The altered vagal activity leads to a lot of intermittent chain reactions, including decreased pancreatic enzyme secretion, poor gallbladder function, overall disturbed gut function. This suppresses the intestinal immune system, decreases intestinal blood flow, increases intestinal permeability, eventually leading to leaky gut, causing digestive problems, inflammation and infection, further aggravating the issues.
The cytokines produced during this imbalance, cross the blood-brain barrier, leading to decreased activity in brain, decreased activation of the vagal motor nuclei, inflammation and decrease in nerve conductance. The effects being anything from mild headaches to aberrant stress to long term depression. A causal association, between gut microbes and cognitive decline and disorders of cognitive function such as Alzheimer's disease and multi-infarct dementia (high prevalence after the age of 65 years), is already been studied.
Wait a minute, before, you thought, when you are in stress you eat more, or when you are tensed you have gastric problems, or when you are hungry for long duration you get a headache. Now you understand that these things are connected, but, possibly in the opposite order, where the food you eat cause the stress, gastric issues and headache.

[](/media/uploads/blog/100c.jpg)
[

](/media/uploads/blog/100c.jpg)
So, coming back the full circle, to where the story began...
the gut microbes (or the whole microbiome) play a very important role in your growth, wellbeing and longevity, from your birth to senility. In fact, it has been established that post-birth, the gut microbial colonization occurs in parallel with cognitive development. There is [increasing evidence](https://www.ncbi.nlm.nih.gov/pubmed/20966022) to support the view that the evolving cognitive activity is critically dependent on the microbiota and its metabolic activity.

[](/media/uploads/blog/100d.jpg)
[

](/media/uploads/blog/100d.jpg)
_**Now that’s a lot of pondering!!**_
(as I said, that’s what we do best, right?)
So, the next time you think about a diet plan (especially the new year ones), or any resolution for that matter, listen to your gut and gradually change and adapt, without hitting the Vagus.
Or else, come and read this blog, again, next year!!
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## How changing a body's microbes leads to illness
Author: BugSpeaks
Published: 2017-03-06
Category: General
Meta Title: How changing a body's microbes leads to illness
Meta Description: Claire M. Fraser, PhD, in this TED talk explains how microbes affect our lives. We are more obsessed with cleanliness than ever: we use antibacterial
URL: https://www.bugspeaks.com/blog/how-changing-a-body-s-microbes-leads-to-illness
Claire M. Fraser, PhD, in this TED talk explains how microbes affect our lives. We are more obsessed with cleanliness than ever: we use antibacterial cleansers, we keep our children away from dirt, and we give out antibiotics with little regard for long-term effects. But in the process, we are altering our microbiota, the microbes that live on and inside us. And it's causing us to be more sick.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Gut Microbiome - Its impact on our lives
Author: BugSpeaks
Published: 2017-03-03
Category: General
Meta Title: Gut Microbiome - Its impact on our lives
Meta Description: The human intestinal tract is home to a diverse and complex microbial community that is increasingly being recognized for its importance in overall hu
URL: https://www.bugspeaks.com/blog/gut-microbiome-its-impact-on-our-lives
The human intestinal tract is home to a diverse and complex microbial community that is increasingly being recognized for its importance in overall human health. This community is commonly referred to as a hidden metabolic 'organ' due to its pivotal impact on a vast array of functions including metabolism, physiology, nutrition and immunity. Thus, alterations of the microbiome can significantly impact health and thus contribute to multiple chronic disease states. In this presentation, Dr. Robert Rountree in this video discusses the role of the microbiome in health and disease - including practical ways to restore and maintain a healthy microbiome.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Psychology and Gut Bacteria
Author: BugSpeaks
Published: 2017-03-02
Category: Microbiome and Lifestyle
Meta Title: Psychology and Gut Bacteria
Meta Description: Marc David, founder of the Institute for the Psychology of Eating, explains in this fascinating new video, where he explains about the psychology and
URL: https://www.bugspeaks.com/blog/psychology-and-gut-bacteria
Marc David, founder of the Institute for the Psychology of Eating, explains in this fascinating new video, where he explains about the psychology and how we relate to “gut bacteria.” And that psychology will powerfully inform our health and our well-being. To that end, it’s a great idea to examine the psychic forces that drive us in relation to anything that’s important in our lives.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Diet, Digestion and Microbiome
Author: BugSpeaks
Published: 2017-02-28
Category: General
Meta Title: Diet, Digestion and Microbiome
Meta Description: Professor Kevin Whelan talks about diet and the microbiome at Core's Exploring the Science of Digestion event at the Kia Oval, London.
URL: https://www.bugspeaks.com/blog/diet-digestion-and-microbiome
Professor Kevin Whelan talks about diet and the microbiome at Core's Exploring the Science of Digestion event at the Kia Oval, London.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Microbiome and Autoimmunity
Author: BugSpeaks
Published: 2017-02-27
Category: Microbiome and Disease
Meta Title: Microbiome and Autoimmunity
Meta Description: At the 2012 International Congress on Autoimmunity, in Granada, Spain, Prof Trevor Marshall delivered this plenary lecture -- explaining how the Human
URL: https://www.bugspeaks.com/blog/microbiome-and-autoimmunity
At the 2012 International Congress on Autoimmunity, in Granada, Spain, Prof Trevor Marshall delivered this plenary lecture -- explaining how the Human Microbiome causes Human Immune dysfunction, including many autoimmune diseases.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The Microbiome Diet
Author: BugSpeaks
Published: 2017-02-24
Category: Microbiome
Meta Title: The Microbiome Diet
Meta Description: In The Microbiome Diet book, Dr. Kellman gives some delicious gut-health promoting foods; populating friendly bacteria for optimal health, good bacter
URL: https://www.bugspeaks.com/blog/the-microbiome-diet
In The Microbiome Diet book, Dr. Kellman gives some delicious gut-health promoting foods; populating friendly bacteria for optimal health, good bacteria foods for a healthy gut, the best way to restore your gut flora, how to recover gut health, how probiotics heal leaky gut, how do you heal a leaky gut, best micro biome diet, why your body needs probiotics, best probiotic supplement in the US, health benefits of biotic balance, why you should take biotic balance, health benefits of probiotics, health benefits of probiotics are highlighted in this video.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Microbes, Wellness and Mealtime
Author: BugSpeaks
Published: 2017-02-23
Category: General
Meta Title: Microbes, Wellness and Mealtime
Meta Description: Much of our population struggles with issues stemming from brain imbalances. New research is finding a strong connection between our gut bacteria and
URL: https://www.bugspeaks.com/blog/microbes-wellness-and-mealtime
Much of our population struggles with issues stemming from brain imbalances. New research is finding a strong connection between our gut bacteria and our mental health. In this video, Lisa Kilgour, R.H.N. is a Registered Holistic Nutritionist, discusses how food impacts our gut, and has the power to either exasperate imbalances like depression, or boost our overall wellbeing.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Mind altering microbes
Author: BugSpeaks
Published: 2017-02-22
Category: Microbiome and Lifestyle
Meta Title: Mind altering microbes
Meta Description: laine Hsiao. a postdoctoral fellow in chemistry and biology at Caltech, in this TEDx talks about various mechanisms by which microbes modulate host pr
URL: https://www.bugspeaks.com/blog/mind-altering-microbes
laine Hsiao. a postdoctoral fellow in chemistry and biology at Caltech, in this TEDx talks about various mechanisms by which microbes modulate host production of neuroactive molecules and aims to better understand how the human microbiota influences health and disease. Here she throws light on Neuroimmune mechanisms underlying the pathogenesis of neurodevelopmental disorders and uncovered a role for the commensal microbiota in regulating autism-related behaviours’, metabolism, and intestinal physiology.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Human Microbiome as a community
Author: BugSpeaks
Published: 2017-02-21
Category: Microbiome
Meta Title: Human Microbiome as a community
Meta Description: The recent explosion of research into our body's microbial universe will change the treatment of metabolic, immune and psycho-emotional disorders.
URL: https://www.bugspeaks.com/blog/human-microbiome-as-a-community
The recent explosion of research into our body's microbial universe will change the treatment of metabolic, immune and psycho-emotional disorders. What’s more, it will cast a whole new perspective on our metaphors, cultural assumptions, and the very identity of “self”, as described in this talk by cultural anthropologist Miriam Lueck Avery.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Our Microbiome and Health
Author: BugSpeaks
Published: 2017-02-20
Category: Microbiome and Lifestyle
Meta Title: Our Microbiome and Health
Meta Description: Did you know that you have up to ten times as many microbial cells on your body as you have human cells? What are these tiny microbes doing and how di
URL: https://www.bugspeaks.com/blog/our-microbiome-and-health
Did you know that you have up to ten times as many microbial cells on your body as you have human cells? What are these tiny microbes doing and how did they find their way to you? Rob Knight, PhD joins our host David Granet, MD to discuss how these cells that make up our microbiome can impact everything from mood, weight, sleep patterns, allergies and more.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The gut feeling
Author: BugSpeaks
Published: 2017-02-17
Category: Microbiome
Meta Title: The gut feeling
Meta Description: This video explains how people are venturing in to microbiome. Advances in DNA sequencing and big data allow us to analyse bacteria like never before.
URL: https://www.bugspeaks.com/blog/the-gut-feeling
This video explains how people are venturing in to microbiome. Advances in DNA sequencing and big data allow us to analyse bacteria like never before. Scientists and entrepreneurs have entered a race to develop tools and datasets that uncover the role microbes play in human health.
Bacteria are no longer the enemy. Will bacteria enable personalized medicine?
Yes, there are many companies that focus on the microbiome that could transform multi-billion dollar concerns like weight loss, probiotics, cosmetics, dental health, and even chronic diseases like diabetes. Will microbiome analysis change modern medicine? How are entrepreneurs involved? Is the money in sequencing, analytics, or drug development?
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The bugs inside your gut
Author: BugSpeaks
Published: 2017-02-16
Category: General
Meta Title: The bugs inside your gut
Meta Description: Join Warren Peters in his TEDx talks about the journey into understanding your gut microbiome and the new discoveries changing the way we understand d
URL: https://www.bugspeaks.com/blog/the-bugs-inside-your-gut
Join Warren Peters in his TEDx talks about the journey into understanding your gut microbiome and the new discoveries changing the way we understand diseases like diabetes, obesity, Alzheimer's disease, autism, and our everyday health and wellness.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The Microbiome
Author: BugSpeaks
Published: 2017-02-07
Category: General
Meta Title: The Microbiome
Meta Description: Dr. Daniel Beiting, Research Assistant Professor at UPenn in the School of Veterinary Medicine in tis TED talk explains about the microbiome in g
URL: https://www.bugspeaks.com/blog/the-microbiome
Dr. Daniel Beiting, Research Assistant Professor at UPenn in the School of Veterinary Medicine in tis TED talk explains about the microbiome in general.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## The gut flora
Author: BugSpeaks
Published: 2017-02-06
Category: Microbiome and Disease
Meta Title: The gut flora
Meta Description: Jeroen Raes, researcher on human microbiome, Flanders Institute of Biology explains his discovery in this TED talk, the major breakthrough not just in
URL: https://www.bugspeaks.com/blog/the-gut-flora
Jeroen Raes, researcher on human microbiome, Flanders Institute of Biology explains his discovery in this TED talk, the major breakthrough not just in gastro-intestinal medicine, but in our fundamental knowledge of the human biology. It turns out that there are only three different types of gut bacteria and, just like blood groups, the three types are totally independent of race, sex, age or diet. Such a baffling finding leads to more research of course and Raes is currently testing his idea on a larger group. The implications for Crohn's Disease or obesity could be dramatic.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Microbiome, the future of medicine?!
Author: BugSpeaks
Published: 2017-02-03
Category: General
Meta Title: Microbiome, the future of medicine?!
Meta Description: We've all heard about the microbiome, but what is it? Why should we care? And, most importantly, what should we do about it? Integrative gastroent
URL: https://www.bugspeaks.com/blog/microbiome-the-future-of-medicine
We've all heard about the microbiome, but what is it? Why should we care? And, most importantly, what should we do about it? Integrative gastroenterologist Dr. Robynne Chutkan, in this video breaks down what you need to know about the microbiome, the vast collection of microbes that live in and on your body, and how you can optimize your gut health to live a longer, better life.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## How our microbes make us who we are
Author: BugSpeaks
Published: 2017-02-02
Category: Microbiome
Meta Title: How our microbes make us who we are
Meta Description: Rob Knight, a pioneer in studying human microbes, the community of tiny single-cell organisms living inside our bodies that have a huge and largely un
URL: https://www.bugspeaks.com/blog/how-our-microbes-make-us-who-we-are
Rob Knight, a pioneer in studying human microbes, the community of tiny single-cell organisms living inside our bodies that have a huge and largely unexplored role in our health. In this video, he explains why “The three pounds of microbes that you carry around with you might be more important than every single gene you carry around in your genome,”.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Human Microbiome - The Invisible Universe
Author: BugSpeaks
Published: 2017-02-01
Category: General
Meta Title: Human Microbiome - The Invisible Universe
Meta Description: Human body consists of 10 times more cells from microorganisms like bacteria and fungi than there are human cells. These tiny compatriots are invisibl
URL: https://www.bugspeaks.com/blog/human-microbiome-the-invisible-universe
Human body consists of 10 times more cells from microorganisms like bacteria and fungi than there are human cells. These tiny compatriots are invisible to the naked eye but their contribution towards human health and immunity is enormous. Ben Arthur, artist, in this video gives us a guided tour of the rich universe of the human microbiome.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Bridging Traditional Dietary and Current Nutritional Practices
Author: BugSpeaks
Published: 2016-12-22
Category: General
Meta Title: Bridging Traditional Dietary and Current Nutritional Practices
Meta Description: Kefir is a friendly probiotic drink with slightly fizzy and tangy taste, a FANTASTIC fermented milk beverage originating from Eastern Europe and South
URL: https://www.bugspeaks.com/blog/bridging-traditional-dietary-and-current-nutritional-practices
Kefir is a friendly probiotic drink with slightly fizzy and tangy taste, a FANTASTIC fermented milk beverage originating from Eastern Europe and Southwest Asia. It looks like and tastes like thin yogurt, but far superior than others, as a probiotic. It is incredibly beneficial for digestion and gut health maintenance when consumed regularly. It is loaded with valuable vitamins and minerals and contains easily digestible complete proteins. People with lactose intolerance can eat kefir without problems, adding to its list of benefits.
[](/media/uploads/blog/59a.jpg)
[

](/media/uploads/blog/59a.jpg)
Kefir can be prepared from any type of milk, cow, goat or sheep, coconut, rice or soy. It is made by adding kefir “grains” to milk. These are not grains in the conventional sense, but cultures of yeast and lactic acid bacteria. Over a period of 24 hours’ time, the microorganisms in the kefir grains multiply and ferment the sugars in the milk, turning it into kefir.

Even though Kefir has been widely recognized as a health drink with benefits, recent development in the field of probiotics, have raised enormous interest in Kefir, with researchers reporting a wide spectrum of progressive elements present in it benefits the person who is consuming it, as it is known for its anticarcinogenic, anti-inflammatory, and antipathogenic effects.
Unfortunately, misuse of the term probiotic has also become a common place and a major issue, with many products exploiting the term without meeting the requisite criteria. Consequently, there is a certain level of apprehension about these effects of Kefir, bringing it under the scrutiny. Can Kefir really show these effects in human, as most of the studies are done on mice and cell lines? If so then, what is the mechanism? What is the uniqueness of Kefir in terms of its bacterial composition?
Initially, some nutritional benchmarking has clearly identified the presence of beneficial components within Kefir (see picture below). “[Antimicrobial and healing activity of kefir and kefiran extract](https://www.ncbi.nlm.nih.gov/pubmed/15848295)” has also be studied early on, demonstrating highest activity against Streptococcus _pyogenes._ Further, Lactobacilli have been recognized as a major component of Kefir, which have GRAS (Generally Regarded As Safe) status. Of these, Lactobacillus _kefiri_ has been identified as the key player in Kefir that has already assessed many functional properties, [safety characterization and antimicrobial properties](https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4052788/). However, parameters regarding safety of Kefir was imperative, before calling them probiotics.

[](/media/uploads/blog/59b.jpg)
[

](/media/uploads/blog/59b.jpg)
Paul Cotter, of Teagasc Food Research Centre in Cork, Ireland, approached the issue to answer such questions associated with kefir, [employing tools of high-throughput sequencing to analyse the microbial composition of Kefir](http://msystems.asm.org/content/1/5/e00052-16). His team of researchers performed Shotgun metagenomic sequencing of the microbial population of Kefir. Along with identifying the composition, they also determined the systematic progression of the microbial communities during fermentation. They observed that, as the fermentation progressed, the bacterial diversity diminished, and species that dominate the population at the beginning are replaced by others, just hours after. For instance, _Lactobacillus_ _kefiranofaciens_ was the dominant bacterial species recorded to be in kefir during early stages of fermentations, but _Leuconostoc_ _mesenteroides_ became more prevalent during later stages. They could further establish the link between individual species present in kefir, flavour generating compounds, several genes and associated pathways. They identified genes that are responsible for some of the intestinal benefits seen in kefir-drinkers. Through metabolomic approach they could map the acidic taste of kefir to the presence of Acetobacter _pasteurianus_ and cheesy flavours to Lactobacillus _kefiranofaciens_. Additionally, they were able to change the flavour and taste of kefir by altering the ratio of these identified microbes.
Metagenomics, again, have proven to be a new way to look at bacteria, not only within hosts, but within commodities that have traditionally proven to possess health benefits. This whole new approach of analysis could in fact provide scientific insights and widen the consumer audience by imparting necessary knowledge to bridge the gap between traditional dietary & current nutritional practices.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Are you dating a meat eater?!
Author: BugSpeaks
Published: 2016-12-19
Category: Diet and Supplements
Meta Title: Are you dating a meat eater?!
Meta Description: He is tall, handsome, tattooed, white collar college grad, financially secure, overall “fits the bill” (The bill?!!) and the perfect guy t
URL: https://www.bugspeaks.com/blog/are-you-dating-a-meat-eater
He is tall, handsome, tattooed, white collar college grad, financially secure, overall “fits the bill” ([The bill?!!](https://www.youtube.com/watch?v=Dw6VE8jjP-0)) and the perfect guy to spend the rest of my life with.
Sounds like a fairytale, right?
Ohhh, wait a minute, what does fairytales usually have? Monsters and witches who always twist and turn the story to eleven.
An ace in the hole for this fairytale, HE EATS MEAT!! And, I don’t.
I mean, I don’t now, but I used to. But, have been a vegan for a while.
So now, being with him means, I would return to meat eating?? Psychology says majority of the converted vegans and vegetarians do return to meat eating. But, I have achieved this with great difficulty and have become a no-meat eater (obviously, I will make one of my kind) and I don’t want to call it a quit now.
So, what should I do?
May be, just maybe, I make him go vegan, and I do have ample reasons to persuade him to go vegan, then me returning to meat eating.
In retrospect, my reasons for not eating meat began with general health, concern towards animal welfare, spanning across to high carbon footprint in meat and global wellbeing. Thus, beginning my vegan diet saga, saving animals and the world I live in, and by eating more of grains, legumes, beans, and of course, BROCOLLI !!

[](/media/uploads/blog/58a.jpg)
[

](/media/uploads/blog/58a.jpg)
Initially I wasn’t happy, had constant hunger and felt it was a daily chore to deal with. I was tired of being hungry and was always seen with low energy. However, with great difficulty I adhered to vegan diet and the rewards were worth the pain. I started feeling lighter, more energetic and improved my overall wellbeing.
So, I would start with this background, but I know this would not convince him (remember, he is intelligent too). So, I began imagining the arguments he would put in and try to come up with valid reasons that would convince him.
**“The ancient argument”**
That humans are naturally omnivorous and we are evolutionarily designed to eat both animal meat and plants.
The relationship between the microbiome, human health and diseaseFirst of all, enough evidence to prove this point isn’t available. Further, talking of evolution, we “EVOLVED” from meat eating wanderers to lake-side living ‘agriCULTURE’ driven social animals, a long time ago.
“**Meat tastes far better than plants”**
Well, not true. Don’t believe me? It actually assumes the taste of flavors and additives we put in, and, even ‘The Matrix’ has acknowledged this. [Chicken tastes like everything](https://www.youtube.com/watch?v=2oEnJfZ9joY)!! (Yes, I have seen ‘the matrix trilogy’ and have UNDERSTOOD it!!)
Moreover, there are lots of vegan and vegetarian dishes that are tastier, and I have quite a few recipes from my own R & D (I mean, my kitchen), which makes vegan diet deliciously healthy and gut friendly too.
**The iron in meat is easier for the body to process than the iron in plants. Meat has higher amount of protein compared to plants**
Oh, now we are getting technical, aren’t we!!
Sure, meat might have these advantages, but all this comes at a cost of unwanted fat. Meat has far more fat than plants and fat isn’t good neither for your adipose nor for your gut. In fact, some of the foods like tofu has higher amount of protein, vitamins, iron than meat and in addition contains 9 essential amino acids, as well as, some may even have cancer-protecting qualities. How about that for technical, eh ?!
“**Change in sudden diet might cause health issues”**
Yes initially, there could be some specific health-related symptoms, while moving from meat to no-meat diet, but then your body and your gut gets used to the new diet. Trust me, it will work charms for you and I am sure the bacteria in your gut would be happier.
**What’s with the gut and the bacteria?**
We have trillions of bacteria in our gut, and these bacteria (called the microbiome) has been known to determine everything from our digestion to mood, immunity to diseases. The vegans/vegetarians have been shown to contain lots of good bacteria in their guts, would fall less sick because of high immune power with other obvious advantages. Besides this they would assist in vitamin and essential nutrient synthesis that are required for your health. The diversified microbes in your gut would reduce the anxiety and depressions and make us feel good. The list doesn’t end here, it goes on.
……And I sat there imaging this conversation we could have in near future.
May be I would persuade him, maybe I’ll help him learn some cool vegan comebacks! Like, it saves animals, reduces global warming, or it’s the best way to be healthy and even make him talk about good bacteria - good mood...etc. Our world would be so perfect, with us chilling, taking pics with cute animals; would look amazing wearing those animal rights tees and of course, shopping for our vegan diet!!

[](/media/uploads/blog/58b.jpg)
[

](/media/uploads/blog/58b.jpg)
**Everything is fair in love isn’t it** **?**
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---
## Exasperation around discovery of new antibiotics
Author: BugSpeaks
Published: 2015-05-28
Category: Others
Meta Title: Exasperation around discovery of new antibiotics
Meta Description: It is exasperating to notice the continued discovery and use of antibiotics, even after the world has proclaimed “the golden age of antibiotics
URL: https://www.bugspeaks.com/blog/exasperation-around-discovery-of-new-antibiotics
It is exasperating to notice the continued discovery and use of antibiotics, even after the world has proclaimed “[the golden age of antibiotics to be over](http://s.telegraph.co.uk/graphics/projects/antibiotic-resistance/)” and that we have reached the brim of post-antibiotic era, after witnessing the unprecedented rise in the antimicrobial resistance. To add to the woes, the international agenda steering infectious disease control, intervention and elimination (is still) defined around “discovery of new antibiotics”, as if, the control and prevention of [already abundant count of resistant pathogens](http://www.cdc.gov/drugresistance/threat-report-2013/) is not a big ask.

[](/media/uploads/blog/6a.jpg)
[

](/media/uploads/blog/6a.jpg)
[Too much of a good thing, by Joe Shute](http://s.telegraph.co.uk/graphics/projects/antibiotic-resistance/), © Copyright of Telegraph Media Group Limited 2013
Current guidelines and strategies do recognize and support the attempts to decelerate the spread of existing resistant infections, emergence of new resistant pathogens through expansion & strengthening of surveillance efforts and development & deployment of rapid diagnostic tests. This shift away from just presumptive treatment towards properly diagnosed and directed treatments is welcome, but it does not justify the continued use of current antibiotics and certainly not the discovery of new ones.
On the contrary, over past few years, there has been a renewed interest in finding new antibiotics, with this particular research group claiming the discovery of “[a new game changing antibiotic](http://www.nature.com/nature/journal/v517/n7535/full/nature14098.html#affil-auth)”. Accordingly, “teixobactin”, mined from “[deep soil microbial dark matter](http://kemnovation.com/use-of-microbial-genomics-to-identify-soil-dark-matter/)”, is capable of combating MRSA (Methicillin-Resistant_Staphylococcus_ aureus) and multi-drug-resistant _Mycobacterium_ tuberculosis. The attribute that sets apart teixobactin from other antibiotics is “the indication that it will be difficult for pathogens to develop resistance against it”. “Unusually for an antibiotic, teixobactin is thought to attack microbes by binding to fatty lipids that make up the bacterial cell wall, and it is difficult for a bacterium to alter such fundamental building blocks of the cell. By comparison, most antibiotics target proteins and it can be relatively easy for a microbe to become resistant to those drugs by accumulating mutations that alter the target protein’s shape” [explains, Dr. Kim Lewis](http://www.nature.com/news/promising-antibiotic-discovered-in-microbial-dark-matter-1.16675#/b1). It is possible that certain antibiotics, based on their mode of action, might have a low frequency of resistance, however, it is improbable to perceive that microbes cannot develop resistance to it, if so, it will be (foolishly) similar to the original claim that “[the time has come to close the book on infectious diseases](http://www.who.int/bulletin/volumes/86/8/08-056242/en/)”.

[](/media/uploads/blog/6b.jpg)
[

](/media/uploads/blog/6b.jpg)
This progressively worsening issue of discovery, (over) use and resistance to drugs is expanding beyond bacterial pathogens, with the discovery of emerging parasite resistance to antimalarials and mosquito resistance to insecticides. Rise of multi drug resistant _Plasmodium_ falciparum has been globally reported, with [emerging parasite resistance to artemisinin](http://www.wellcome.ac.uk/News/Media-office/Press-releases/2015/WTP058719.htm), which is recognized to be an immediate threat to the recent progress and a major obstacle to the future control of malaria. Inanely, [the strategies employed for malaria intervention](http://blogs.plos.org/biologue/2015/04/23/stressed-to-death/) is no different than the contemporary case of bacterial pathogens, with approaches such as combinational therapy or extended therapy (extending the treatment from 3 to 6 days). Another approach that has caught some attention, is treating drug-resistant parasites with [artemisinins and a proteasome inhibitor](http://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.1002132), which in combination has the ability to stress the parasite and prevent its ability to protect itself from damaged proteins, hence synergistically killing the resistant parasites. The use of proteasome inhibitor has been around since early 2000, and has been [successfully tested for cancer therapy](http://www.nlm.nih.gov/medlineplus/druginfo/meds/a607007.html) before. However, its use in combination with artemisinin to combat drug resistant malaria seems fatuous since the intrinsic and acquired resistance to proteasome inhibitor drugs like “bortezomib” has already been reported, with several [mechanisms being studied extensively](http://www.ncbi.nlm.nih.gov/pubmed/22978849).
There might be some utility for conserved and appropriate use of current (and new) drugs, say in case of emergencies or until suitable vaccines are developed against, but it is should not be a sustainable model for the future nor it should hinder the pursuit for safer alternative approaches. It is imperative to learn from our (bad) experience and presume that most of the current tools against these pathogens and even newly discovered drugs could render ineffective in near (rather than far) future. It is absolutely necessary to reiterate the fact that we are dealing with highly adaptive life forms and the only way to tackle them is to develop equally adaptive combative therapies, keeping in mind their evolutionary projectile.
The actual shift has to be “a cultural evolution to create an environment where we do not use or need antibiotics.” The dramatic progress that France made through the “[Antibiotics Are Not Automatic](http://journals.plos.org/plosmedicine/article?id=10.1371/journal.pmed.1000080)” campaign, can be a notable example that demonstrates the positive cumulative impact of reducing the overuse of antibiotic.
Advancement in molecular methods and genomics have enabled deeper comprehension of pathogen-host interactions, which in turn have paved way for new[approaches to develop non-chemical antimicrobial strategies](http://blogs.cdc.gov/genomics/2014/10/30/outsmarting/). Utilization and translation of decades of molecular studies identifying genetic variations involving drug resistance, virulence, transmission and human immunity, followed by completion of the whole genome sequences of pathogens, vectors and the human genome, should lead us to relevant clinical tools for intervention and elimination of these pathogens. Any limitations on the way should force us to concentrate our resources in areas where we can identify and exploit significant susceptibilities of these complex pathogens, instead of relying on “Einstein’s definition of Insanity”.
---
This blog is powered by Superblog. Visit https://superblog.ai to know more.
---