35 Basic Questions about Gut Microbiome and Our Health.

January 19, 2024 77 min read

35 Basic Questions about Gut Microbiome and Our Health.

Table of Content (Click on each topic to navigate) 

·        How does the gut microbiome influence overall health?

The gut microbiome refers to the trillions of microorganisms, including bacteria, fungi, and viruses, that live in the human gastrointestinal tract. Research has shown that the gut microbiome plays an important role in various aspects of human health, including digestion, immune system function, metabolism, and mental health.

One way that the gut microbiome influences overall health is through its role in the immune system. The gut microbiome helps to train and regulate the immune system, protecting against harmful pathogens while avoiding harmful immune responses to harmless substances. This is partly accomplished through the production of short-chain fatty acids (SCFAs) and other metabolites that influence the development and function of immune cells. A disrupted gut microbiome, known as dysbiosis, has been linked to various autoimmune and inflammatory diseases, including inflammatory bowel disease (IBD), multiple sclerosis, and type 1 diabetes .

The gut microbiome also plays a crucial role in digestion and nutrient absorption. It helps to break down complex carbohydrates and fibers that are otherwise indigestible by the human body, producing SCFAs and other metabolites that provide energy to the gut lining and other tissues. Additionally, the gut microbiome produces enzymes and other molecules that aid in the breakdown and absorption of nutrients, such as vitamins and minerals. A disrupted gut microbiome can lead to digestive disorders, such as irritable bowel syndrome (IBS) and malabsorption disorders . 

Furthermore, the gut microbiome has been linked to various metabolic processes, including glucose and lipid metabolism. Studies have shown that a disrupted gut microbiome can lead to insulin resistance, obesity, and other metabolic disorders .

Finally, emerging evidence suggests that the gut microbiome may influence mental health and behavior. The gut-brain axis refers to the communication pathway between the gut and the brain, and the gut microbiome is thought to play a role in this pathway. Research has shown that the gut microbiome can influence brain function and behavior through various mechanisms, including the production of neurotransmitters and the regulation of inflammation. Dysbiosis has been linked to various mental health disorders, including depression and anxiety .

Overall, the gut microbiome plays an essential role in various aspects of human health, and disruptions to its composition and function have been linked to various diseases and disorders. Further research is needed to fully understand the complex interactions between the gut microbiome and overall health. 

·        What is leaky gut syndrome?

"Leaky gut syndrome," also known as increased intestinal permeability, is a condition in which the lining of the intestine becomes more porous, allowing toxins, bacteria, and other substances to pass into the bloodstream. The concept of leaky gut syndrome is controversial, and some experts argue that it is not a recognized medical condition. However, there is some evidence to suggest that increased intestinal permeability may be associated with certain health conditions.

Some potential causes of increased intestinal permeability include:

  • chronic stress,
  • poor diet,
  • overuse of antibiotics,
  • certain medications (such as nonsteroidal anti-inflammatory drugs).

Research has shown that certain conditions, such as inflammatory bowel disease, celiac disease, and type 1 diabetes, are associated with increased intestinal permeability .

Symptoms of leaky gut syndrome may include bloating, gas, cramps, fatigue, joint pain, skin rashes, and food sensitivities. However, these symptoms are non-specific and can be caused by a wide range of conditions.

There is some preliminary evidence to suggest that certain dietary and lifestyle changes may help to improve intestinal permeability. For example, reducing intake of inflammatory foods (such as sugar and processed foods), increasing intake of fiber and prebiotics, and taking probiotics may help to promote a healthy gut microbiome and reduce inflammation in the gut .

However, more research is needed to fully understand the relationship between increased intestinal permeability and overall health, as well as the most effective treatments for this condition. 

·        How can I improve my gut health?

There are several ways you can improve your gut health. Here are some evidence-based tips:

  1. Eat a diverse diet rich in fiber: A diet rich in fiber can help promote the growth of beneficial gut bacteria, which can improve gut health. Aim for at least 25-30 grams of fiber per day from a variety of sources, such as fruits, vegetables, whole grains, nuts, and seeds .
  2. Incorporate probiotic-rich foods: Probiotic-rich foods, such as yogurt, kefir, sauerkraut, and kimchi, contain live beneficial bacteria that can help improve gut health .
  3. Avoid excessive use of antibiotics: Antibiotics can disrupt the balance of gut bacteria, so try to avoid using them unless absolutely necessary .
  4. Manage stress: Chronic stress can negatively affect gut health, so finding ways to manage stress, such as through meditation or yoga, can be beneficial .
  5. Get enough sleep: Sleep is important for gut health, as it helps regulate the immune system and promote the growth of beneficial gut bacteria .
  6. Stay hydrated: Drinking plenty of water can help keep the digestive system functioning properly and prevent constipation .
  7. Avoid processed foods and added sugars: Processed foods and added sugars can negatively affect gut health and promote the growth of harmful bacteria .
  8. Use prebiotic supplement to help feed the key bacteria in your gut microbiome .

It's important to note that individual responses to dietary changes may vary, so it may be helpful to work with a healthcare professional to determine the best approach for you. 

·        What are the best foods for gut health?

Yogurt: Yogurt contains live cultures of beneficial bacteria, such as Lactobacillus and Bifidobacterium, which can help improve gut health .

  1. Kefir: Kefir is a fermented milk drink that contains a variety of beneficial bacteria and yeasts, making it a good source of probiotics .
  2. Sauerkraut: Sauerkraut is a fermented cabbage dish that is high in beneficial bacteria and can help improve gut health .
  3. Kimchi: Kimchi is a Korean dish made from fermented vegetables and spices, which can provide a good source of beneficial bacteria for the gut .
  4. Whole grains: Whole grains, such as oats, barley, and quinoa, are high in fiber, which can help promote the growth of beneficial gut bacteria .
  5. Fruits and vegetables: Fruits and vegetables are also high in fiber and contain a variety of other nutrients that can help support gut health .
  6. Nuts and seeds: Nuts and seeds, such as almonds, chia seeds, and flaxseeds, are high in fiber and can also provide a source of beneficial fats for the gut .
  7. Fermented vegetables: Fermented vegetables, such as pickles, beets, and carrots, are a good source of probiotics and prebiotics, which can help support the growth of beneficial gut bacteria .
  8. Garlic and onions: Garlic and onions contain compounds that can help promote the growth of beneficial gut bacteria and reduce the growth of harmful bacteria .
  9. Bone broth: Bone broth is a rich source of collagen, gelatin, and amino acids, which can help support gut health by promoting the growth of beneficial gut bacteria and reducing inflammation .
  10. Tempeh: Tempeh is a fermented soybean product that is high in protein and a good source of probiotics, which can help improve gut health .
  11. Apple cider vinegar: Apple cider vinegar contains acetic acid, which can help improve digestion and promote the growth of beneficial gut bacteria .

It's important to note that while these foods can be beneficial for gut health, they should be consumed as part of a well-rounded, balanced diet. It's also important to listen to your body and make adjustments as needed to determine what works best for you.

·        Can gut health affect my mental health? 

Yes, gut health can affect mental health. There is a growing body of research suggesting that the gut microbiome plays an important role in brain function and behavior, and that imbalances in the gut microbiome can contribute to mental health issues such as anxiety, depression, and even autism .

One way that the gut microbiome can influence mental health is through the gut-brain axis, which is a bidirectional communication system between the gut and the brain. The gut microbiome communicates with the brain via the vagus nerve, as well as through the production of neurotransmitters and other signaling molecules that can influence brain function and behavior .

Several studies have found that probiotics, which are beneficial bacteria that can be found in certain foods and supplements, can improve mood and reduce symptoms of anxiety and depression in both healthy individuals and those with mental health disorders (3,4).

Other studies have found that diets high in fiber, which can help promote the growth of beneficial gut bacteria, are associated with a lower risk of depression .

It's worth noting that the relationship between gut health and mental health is complex, and more research is needed to fully understand the mechanisms involved. However, the evidence to date suggests that maintaining a healthy gut microbiome is an important aspect of overall mental health and well-being. 

·        How does the gut-brain axis work? 

The gut-brain axis is a bidirectional communication system between the gut and the brain. It involves the nervous system, the immune system, and the endocrine system, and is influenced by the gut microbiome, which plays a critical role in regulating gut-brain communication .

One way that the gut communicates with the brain is through the vagus nerve, which runs from the brainstem to the gut and carries signals in both directions. The gut microbiome can influence the activity of the vagus nerve, which in turn can affect brain function and behavior .

Another way that the gut communicates with the brain is through the production of neurotransmitters and other signaling molecules that can affect brain function. For example, the gut produces a number of neurotransmitters, including serotonin, which plays a role in regulating mood and is also involved in the regulation of gastrointestinal function .

The gut microbiome can also affect the immune system, which plays an important role in regulating inflammation in the body. Chronic inflammation has been linked to a number of diseases, including depression and anxiety, and the gut microbiome can influence the level of inflammation in the body .

The gut-brain axis is a complex system, and the mechanisms involved are still being studied. However, the evidence to date suggests that maintaining a healthy gut microbiome is an important aspect of overall brain function and mental health.

·        What is dysbiosis? Can dysbiosis cause health problems? 

Dysbiosis refers to an imbalance in the composition and/or function of the gut microbiome, which is the collection of microorganisms that inhabit the human gastrointestinal tract. This imbalance can lead to changes in the gut environment that may cause or contribute to a variety of health problems.

There are many factors that can contribute to dysbiosis, including changes in diet, use of antibiotics or other medications, stress, and certain medical conditions. For example, a diet that is high in fat and low in fiber can promote the growth of bacteria that produce harmful byproducts, while antibiotics can disrupt the balance of bacteria in the gut, leading to an overgrowth of potentially harmful species. Similarly, chronic stress can cause changes in gut motility and secretion that can alter the gut environment and promote dysbiosis.

Research has shown that dysbiosis may be associated with a number of health problems. Gastrointestinal disorders such as inflammatory bowel disease, irritable bowel syndrome, and colorectal cancer have all been linked to dysbiosis. Inflammatory bowel disease, for example, has been associated with alterations in the gut microbiome, including a decrease in beneficial bacteria and an increase in potentially harmful species. Similarly, some studies have suggested that dysbiosis may be a contributing factor in the development of colorectal cancer.

In addition to gastrointestinal disorders, dysbiosis has also been linked to metabolic disorders such as obesity and type 2 diabetes. This is thought to occur because dysbiosis can lead to changes in the gut environment that promote inflammation and insulin resistance, both of which are associated with these conditions.

There is also growing evidence to suggest that dysbiosis may be involved in the development of neurological disorders such as anxiety and depression. The gut and the brain are connected through a complex network of nerves, hormones, and other signaling molecules, known as the gut-brain axis. Dysbiosis can disrupt this communication, leading to alterations in brain function and behavior. Some studies have even suggested that interventions aimed at restoring a healthy gut microbiome, such as probiotics, may be effective in treating these conditions.

Overall, dysbiosis is a complex condition that can be caused by a variety of factors and may contribute to a wide range of health problems. Restoring a healthy gut microbiome through interventions such as probiotics, prebiotics, and fecal microbiota transplantation may be an effective strategy for preventing or treating these conditions.

·        What are the symptoms of an unhealthy gut? 

An unhealthy gut can manifest in a variety of symptoms, which can range from mild discomfort to chronic and debilitating conditions. Here are some of the most common symptoms associated with an unhealthy gut:

  1. Bloating: Bloating is a common symptom of an unhealthy gut. It occurs when there is an imbalance of bacteria in the gut, leading to excess gas production.
  2. Constipation or diarrhea: When the gut microbiome is imbalanced, it can lead to either constipation or diarrhea. Constipation occurs when stool moves slowly through the digestive tract, while diarrhea occurs when stool moves too quickly.
  3. Abdominal pain or discomfort: Pain or discomfort in the abdomen can be a sign of an unhealthy gut. This can be caused by inflammation, infection, or a digestive disorder.
  4. Gas and flatulence: Excess gas and flatulence are common symptoms of an unhealthy gut. This can be caused by an imbalance of bacteria in the gut or a food intolerance.
  5. Heartburn or acid reflux: Heartburn or acid reflux occurs when stomach acid backs up into the esophagus. This can be caused by a weakened lower esophageal sphincter, which can be linked to an unhealthy gut.
  6. Nausea or vomiting: Nausea and vomiting can be caused by an unhealthy gut, particularly if it is accompanied by other symptoms such as abdominal pain, bloating, or diarrhea.
  7. Unintentional weight changes: Changes in weight, either gain or loss, can be a sign of an unhealthy gut. This is often due to changes in the gut microbiome or changes in nutrient absorption.
  8. Fatigue or lack of energy: An unhealthy gut can lead to fatigue or a lack of energy due to poor nutrient absorption and inflammation in the body.
  9. Mood changes: The gut-brain axis connects the gut and the brain, and an unhealthy gut can lead to mood changes such as anxiety, depression, or irritability.
  10. Skin conditions: The health of the gut microbiome can also affect the skin. Conditions such as acne or eczema can be linked to an unhealthy gut.

It's important to note that these symptoms can also be caused by other health conditions, and a healthcare professional should be consulted for proper diagnosis and treatment.

  • Can gut health affect weight? Can prebiotic help with weight management 

Yes, gut health can affect weight management. The gut microbiota, which refers to the community of microorganisms that inhabit the human gastrointestinal tract, plays a crucial role in regulating many aspects of human health, including weight management. The gut microbiota can influence weight management by regulating energy homeostasis, appetite control, and inflammation, among other mechanisms. 

One of the key ways that the gut microbiota can influence weight management is through its role in regulating energy homeostasis, which is the balance between energy intake (food consumption) and energy expenditure (metabolic processes and physical activity). The gut microbiota can affect energy homeostasis by influencing the production of hormones that control appetite and food intake, the breakdown and absorption of nutrients from food, and the metabolism of dietary fats and carbohydrates. 

Several studies have shown that the gut microbiota can influence body weight in both humans and animals. For example, a study by Turnbaugh et al. (2006) found that mice that were colonized with gut microbiota from obese humans gained more weight and had a higher body fat content than mice that were colonized with gut microbiota from lean humans, even when they were fed the same diet. This suggests that the gut microbiota from obese individuals may have a greater capacity to extract energy from food, leading to increased calorie absorption and weight gain. 

On the other hand, some types of gut bacteria may promote weight loss or prevent weight gain. For example, a study by Cani et al. (2007) found that mice treated with prebiotics (non-digestible dietary fibers that promote the growth of beneficial gut bacteria) had a reduced body weight and improved glucose tolerance, compared to control mice. This was attributed to the prebiotics' ability to increase the abundance of Bifidobacterium and Lactobacillus in the gut, which are associated with improved metabolic health. 

In addition to regulating energy homeostasis, the gut microbiota can also influence weight management by modulating inflammation. Chronic low-grade inflammation is associated with many metabolic disorders, including obesity, insulin resistance, and type 2 diabetes. The gut microbiota can influence inflammation by producing metabolites that interact with immune cells and by modulating the intestinal barrier function, which can impact the translocation of bacteria and their metabolites into the bloodstream and other tissues. 

A study by Everard et al. (2013) found that mice treated with antibiotics to disrupt the gut microbiota had a higher body weight and increased insulin resistance, compared to control mice. This was attributed to the antibiotics' ability to alter the gut microbiota composition and reduce the abundance of beneficial gut bacteria, such as Bifidobacterium and Lactobacillus. These bacteria are known to produce short-chain fatty acids (SCFAs), which have anti-inflammatory properties and can improve glucose metabolism.

Moreover, a study by Cani et al. (2008) found that mice treated with SCFAs had a reduced body weight and improved glucose tolerance, compared to control mice. This was attributed to the SCFAs' ability to reduce inflammation and improve insulin sensitivity. 

In addition to these mechanisms, the gut microbiota can also affect weight management through its influence on the gut-brain axis. The gut-brain axis refers to the bidirectional communication between the gut and the central nervous system, which is mediated by various neural, hormonal, and immune signaling pathways. The gut microbiota can affect the gut-brain axis by producing neurotransmitters and other signaling molecules that can influence mood, behavior, and appetite.

·        How can I maintain a healthy gut while taking antibiotics? 

Antibiotics are an essential tool for fighting bacterial infections, but they can also disrupt the balance of the gut microbiota, leading to unwanted side effects such as diarrhea, bloating, and other gastrointestinal symptoms. Here are some tips for maintaining a healthy gut while taking antibiotics:

  1. Several studies have shown that prebiotics can help maintain a healthy gut microbiota and reduce the risk of gastrointestinal symptoms associated with antibiotics. For example, one study found that supplementing with a prebiotic blend during antibiotic treatment reduced the incidence of diarrhea and improved stool consistency compared to placebo.
  2. Take probiotics: Probiotics are live microorganisms that can help replenish the beneficial bacteria in the gut that may be destroyed by antibiotics. Several studies have shown that taking probiotics during and after antibiotic treatment can reduce the risk of antibiotic-associated diarrhea and other gastrointestinal symptoms (1,2). 
  3. Eat a balanced diet: A diet rich in fiber, fruits, and vegetables can help support the growth of beneficial gut bacteria and reduce inflammation in the gut. On the other hand, a diet high in processed foods and sugar can promote the growth of harmful bacteria and worsen gastrointestinal symptoms .
  4. Stay hydrated: Drinking plenty of water and other fluids can help flush out toxins and waste products from the body and prevent dehydration, which can worsen gastrointestinal symptoms.
  5. Avoid certain foods: Some foods, such as dairy products, spicy foods, and high-fat foods, can worsen gastrointestinal symptoms and should be avoided while taking antibiotics. It's also a good idea to limit your intake of alcohol and caffeine, which can irritate the gut lining.
  6. Take antibiotics as prescribed: It's important to take antibiotics exactly as prescribed by your healthcare provider, even if you start to feel better before the course of treatment is complete. Skipping doses or stopping antibiotics early can increase the risk of antibiotic resistance and prolong the disruption of the gut microbiota.
  7. Talk to your healthcare provider: If you experience severe or persistent gastrointestinal symptoms while taking antibiotics, it's important to talk to your healthcare provider. They may be able to adjust your dose or switch to a different antibiotic to minimize side effects.

In summary, maintaining a healthy gut while taking antibiotics requires a balanced approach that includes taking probiotics, eating a balanced diet, staying hydrated, avoiding certain foods, taking antibiotics as prescribed, and seeking medical attention if necessary. By following these tips, you can minimize the disruption to your gut microbiota and promote optimal health.

 

·        How do I know if I have IBS and how is IBS treated?

Irritable bowel syndrome (IBS) is a common gastrointestinal disorder characterized by a group of symptoms, including abdominal pain or discomfort, bloating, and changes in bowel habits such as diarrhea, constipation, or a combination of both. The exact cause of IBS is unknown, but it is believed to be related to a combination of factors, including abnormal gut motility, visceral hypersensitivity, and alterations in the gut microbiota.

To diagnose IBS, your healthcare provider will typically perform a physical exam and review your medical history and symptoms. They may also order additional tests, such as stool analysis, blood tests, or imaging studies, to rule out other underlying conditions that may be causing your symptoms.

Treatment for IBS typically focuses on managing symptoms through lifestyle changes, dietary modifications, and medications. Here are some common approaches to treating IBS:

  1. Lifestyle changes: Regular exercise, stress reduction techniques such as meditation or yoga, and getting enough sleep can help improve symptoms of IBS.
  2. Dietary modifications: Some people with IBS may benefit from avoiding certain foods that can trigger symptoms, such as fatty foods, spicy foods, caffeine, and alcohol. Increasing fiber intake can also help alleviate constipation, but it's important to do so gradually to avoid worsening symptoms.
  3. Medications: Several medications may be used to treat specific symptoms of IBS, such as antispasmodics to relieve abdominal pain and cramping, laxatives to treat constipation, and antidiarrheals to treat diarrhea. In some cases, antidepressants may be prescribed to help manage both gastrointestinal symptoms and psychological distress associated with IBS.
  4. Probiotics: Some evidence suggests that certain probiotics may be beneficial in reducing symptoms of IBS, particularly those containing Bifidobacterium and Lactobacillus strains .

It's important to note that there is no one-size-fits-all approach to treating IBS, as symptoms can vary widely between individuals. It may take some trial and error to find the right combination of treatments that work best for you.

In summary, if you are experiencing symptoms of IBS, it's important to speak with your healthcare provider for an accurate diagnosis and appropriate treatment plan. With the right approach, many people with IBS are able to manage their symptoms and improve their quality of life. 

·        Can stress affect gut health? How can I reduce stress to improve gut health? 

Yes, stress can affect gut health in several ways. When the body is under stress, the nervous system releases hormones such as cortisol and adrenaline, which can affect digestion, increase inflammation in the gut, and alter the composition of the gut microbiome . Stress has also been linked to a number of gastrointestinal disorders, including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and gastroesophageal reflux disease (GERD) .

Reducing stress can be beneficial for gut health. Here are some strategies that may help:

  1. Mindfulness meditation: Mindfulness meditation involves focusing on the present moment and cultivating an attitude of non-judgmental awareness. Studies have shown that mindfulness meditation can help reduce symptoms of IBS and improve quality of life .
  2. Exercise: Regular exercise has been shown to reduce stress and anxiety and improve gut health by increasing gut motility and reducing inflammation.
  3. Yoga: Yoga is a mind-body practice that can help reduce stress and improve gut health by increasing relaxation, improving gut motility, and reducing inflammation .
  4. Breathing exercises: Deep breathing exercises can help reduce stress and promote relaxation. One technique, called the 4-7-8 breathing technique, involves inhaling for 4 seconds, holding the breath for 7 seconds, and exhaling for 8 seconds .
  5. Cognitive-behavioral therapy (CBT): CBT is a form of psychotherapy that can help people identify and change negative thought patterns and behaviors that contribute to stress and anxiety. Studies have shown that CBT can be effective in reducing symptoms of IBS and improving quality of life .
  6. Social support: Having a supportive social network can help reduce stress and improve overall well-being. Seeking out supportive relationships with family and friends, or joining a support group for people with gastrointestinal disorders, may be helpful. 

·        Can gut health affect skin health? 

The gut and the skin are connected through the gut-skin axis, which is a bidirectional communication system that involves the immune system, hormones, and the gut microbiome . Studies have shown that changes in the gut microbiome can lead to alterations in the immune system, which can affect the skin .

Here are some ways in which gut health can affect skin health:

  1. Inflammation: An imbalance in the gut microbiome can lead to increased inflammation in the body, which can affect the skin. Chronic inflammation has been linked to a number of skin conditions, including acne, eczema, and psoriasis .
  2. Nutrient absorption: The gut is responsible for absorbing nutrients from food, and a healthy gut is essential for proper nutrient absorption. Nutrient deficiencies, such as vitamin D deficiency, have been linked to skin conditions like psoriasis .
  3. Hormonal imbalances: The gut microbiome can affect hormone levels in the body, including sex hormones like estrogen and testosterone. Hormonal imbalances have been linked to acne and other skin conditions .
  4. Immune system dysfunction: The gut is home to a large portion of the body's immune system, and an imbalance in the gut microbiome can lead to immune system dysfunction. This can lead to autoimmune skin conditions like psoriasis .
  5. Stress: As mentioned earlier, stress can affect gut health, and it can also affect skin health. Stress has been linked to a number of skin conditions, including acne, eczema, and psoriasis .

Improving gut health can be beneficial for skin health. Here are some strategies that may help:

  1. Probiotics: Probiotics are beneficial bacteria that can help improve gut health by restoring the balance of the gut microbiome. Studies have shown that probiotics can be beneficial for skin health by reducing inflammation and improving skin barrier function .
  2. Prebiotics: Prebiotics are non-digestible fibers that feed the beneficial bacteria in the gut. By promoting the growth of beneficial bacteria, prebiotics can help improve gut health and, in turn, skin health .
  3. Diet: A healthy diet that is rich in fruits, vegetables, whole grains, and lean protein can help improve gut health and, in turn, skin health. Processed foods, sugar, and unhealthy fats should be avoided.
  4. Stress reduction: stress can affect gut health and skin health. Strategies for stress reduction, such as meditation, yoga, and exercise, may be helpful for both gut and skin health.

In conclusion, gut health can affect skin health in several ways, including inflammation, nutrient absorption, hormonal imbalances, immune system dysfunction, and stress. Improving gut health through probiotics, prebiotics, a healthy diet, and stress reduction may be beneficial for skin health. 

·        How can I tell if my gut is healthy?

There are several ways to determine if your gut is healthy. Here are some common indicators:

  1. Regular bowel movements: A healthy gut should produce regular bowel movements that are well-formed and easy to pass. Typically, this means one to three bowel movements per day.
  2. Absence of gastrointestinal symptoms: If you don't experience any gastrointestinal symptoms like bloating, gas, diarrhea, constipation, or abdominal pain, it is likely that your gut is healthy.
  3. Good digestion and nutrient absorption: If your gut is healthy, you should be able to digest your food easily and absorb nutrients efficiently.
  4. Strong immune system: Your gut is responsible for about 70% of your immune system, so if your immune system is strong and you don't get sick easily, it may be a sign of good gut health.
  5. Healthy weight: There is a strong link between gut health and weight management. If you maintain a healthy weight and have no trouble losing or gaining weight as needed, it may indicate good gut health.
  6. Positive mood: Research has found a strong connection between gut health and mental health. If you have a positive mood, good energy, and good mental clarity, it may indicate good gut health. 

·        What is Bifidobacterium? 

Bifidobacterium is a genus of gram-positive bacteria that naturally inhabit the human gut. These bacteria are considered beneficial for their ability to ferment dietary fibers and produce short-chain fatty acids, which can have numerous health benefits. Some of the most commonly studied Bifidobacterium strains include:

  • Bifidobacterium bifidum
  • Bifidobacterium breve
  • Bifidobacterium lactis
  • Bifidobacterium longum
  • Bifidobacterium longum. Sub infantis
  • Bifidobacterium animalis

Here are some potential benefits of Bifidobacterium:

  • Improved digestive health: Bifidobacterium can help improve gut motility, reduce inflammation, and prevent gastrointestinal infections.
  • Enhanced immune function: Some strains of Bifidobacterium can help regulate the immune system by increasing the production of certain antibodies and reducing inflammation.
  • Reduced risk of allergies: Bifidobacterium supplementation during pregnancy and early infancy has been associated with a reduced risk of eczema and other allergic conditions.
  • Improved mental health: Some studies have found that certain strains of Bifidobacterium can improve mood and reduce symptoms of anxiety and depression.
  • Lowered risk of chronic disease: Bifidobacterium may help reduce the risk of chronic diseases such as cardiovascular disease, type 2 diabetes, and colorectal cancer. 

·        What is lactobacillus?

Lactobacillus is a genus of Gram-positive, lactic acid bacteria that are found in various habitats including the human gut, mouth, and vaginal tract. It is also found in a variety of fermented foods and dairy products, such as yogurt, kefir, and cheese. Additionally, Lactobacillus can be found in some dietary supplements, including probiotics and some prebiotics. Here are some species, functions, and benefits of lactobacillus:

  1. Lactobacillus acidophilus: This species is commonly found in the human gut and is known to improve digestion, boost the immune system, and support vaginal health in women .
  2. Lactobacillus rhamnosus: This strain has been shown to improve gut health by reducing inflammation, promoting healthy gut bacteria, and preventing infections caused by harmful bacteria .
  3. Lactobacillus casei: This species has been shown to improve the immune system and reduce the risk of certain infections, such as respiratory infections and diarrhea .
  4. Lactobacillus plantarum: This strain has been found to help with digestive issues, such as irritable bowel syndrome (IBS), and may also have anti-inflammatory properties .
  5. Lactobacillus fermentum: This species has been shown to improve gut health by reducing inflammation and preventing the growth of harmful bacteria in the gut .
  6. Lactobacillus reuteri: This strain is known to help reduce inflammation in the gut, promote healthy gut bacteria, and may also have anti-anxiety and anti-depressant effects .

Overall, lactobacillus strains have been found to have many potential benefits for gut health and overall well-being.

·        What is Faecalibacterium prausnitzii?

Faecalibacterium prausnitzii is a Gram-positive, anaerobic, commensal bacterium that belongs to the Firmicutes phylum. It is considered to be a beneficial bacterium for human health due to its various functions and benefits. Here are five key points summarizing what Faecalibacterium prausnitzii is and its benefits:

  • Faecalibacterium prausnitzii is one of the most abundant bacteria in the human gut microbiota, comprising up to 15% of the total gut microbiota. It is commonly found in the large intestine, where it plays an important role in maintaining gut health.
  • Faecalibacterium prausnitzii has anti-inflammatory properties and is known to produce short-chain fatty acids (SCFAs) such as butyrate, which has been shown to have anti-inflammatory effects on the gut. This makes it an important bacterium in regulating the immune system and preventing chronic inflammatory diseases.
  • Studies have shown that the presence of Faecalibacterium prausnitzii in the gut is associated with lower incidence of various diseases, including inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and colorectal cancer. It is believed that this is due to its anti-inflammatory properties and its ability to maintain gut barrier integrity.
  • Faecalibacterium prausnitzii has been shown to have a positive impact on mental health. Studies have found that individuals with depression and anxiety have lower levels of Faecalibacterium prausnitzii in their gut microbiota, suggesting a possible link between gut microbiota and mental health.
  • Faecalibacterium prausnitzii is sensitive to various antibiotics and can be depleted by their use, which can lead to dysbiosis and gut health issues. Therefore, it is important to avoid unnecessary use of antibiotics and to consider probiotics or prebiotics as a way to restore and maintain healthy levels of Faecalibacterium prausnitzii in the gut.

Overall, Faecalibacterium prausnitzii is an important bacterium in the human gut microbiota, with various functions and benefits for human health. Its anti-inflammatory properties, ability to produce SCFAs, and positive impact on mental health make it a beneficial bacterium to have in the gut. However, its sensitivity to antibiotics highlights the importance of maintaining a healthy gut microbiota through proper use of antibiotics and use of probiotics or prebiotics to support beneficial bacteria like Faecalibacterium prausnitzii. 

·        What is Roseburia? 

Roseburia is a genus of bacteria belonging to the family Lachnospiraceae, which is known for its beneficial effects on gut health. Here are some key points about Roseburia:

  • Roseburia is a Gram-positive, anaerobic, butyrate-producing bacteria that is commonly found in the human gut. It is one of the most abundant species of bacteria in the human colon, where it plays a crucial role in maintaining gut health.
  • Roseburia is known to have a positive impact on the immune system by producing short-chain fatty acids (SCFAs) such as butyrate, which serve as a source of energy for colon cells and are involved in regulating inflammation in the gut. Butyrate also has anti-inflammatory properties and is important for the maintenance of gut barrier function.
  • Studies have shown that a reduction in Roseburia abundance in the gut is associated with several inflammatory bowel diseases (IBD), including Crohn's disease and ulcerative colitis. In addition, a decrease in Roseburia levels has been observed in individuals with obesity and type 2 diabetes.
  • Roseburia is also important for the breakdown of complex carbohydrates, such as dietary fiber, which cannot be digested by the human host. The bacteria uses enzymes to break down these complex carbohydrates into simple sugars, which can then be used for energy production by the host. This process is important for maintaining a healthy gut microbiome.
  • Furthermore, Roseburia has been shown to produce metabolites that have potential therapeutic effects, such as bacteriocins, which are antimicrobial peptides that can help to prevent the growth of harmful bacteria in the gut.

·        What is Streptococcus thermophilus?

Streptococcus thermophilus is a lactic acid bacteria that is widely used in the food industry as a starter culture in the production of various dairy products such as yogurt and cheese. It is a gram-positive, non-motile, and non-sporulating bacterium that can grow under aerobic and anaerobic conditions. Here are some key points about Streptococcus thermophilus:

  • Benefits: Streptococcus thermophilus has been shown to have a number of potential health benefits. For example, it can help improve lactose digestion in individuals who are lactose intolerant. It has also been found to have anti-inflammatory properties and may help reduce the risk of inflammatory bowel disease. Additionally, some studies have suggested that Streptococcus thermophilus may help boost the immune system and have a positive effect on cholesterol levels.
  • Probiotic: Streptococcus thermophilus is considered a probiotic bacteria due to its ability to survive passage through the gastrointestinal tract and reach the colon, where it can have a positive impact on gut health. Studies have shown that it can help improve the balance of gut bacteria and enhance the production of short-chain fatty acids, which are important for maintaining a healthy gut environment.
  • Food industry: Streptococcus thermophilus is widely used in the food industry as a starter culture in the production of various dairy products such as yogurt, cheese, and sour cream. It is particularly useful in yogurt production due to its ability to ferment lactose and produce lactic acid, which helps to create the characteristic tangy flavor and texture of yogurt.
  • Safety: Streptococcus thermophilus is generally considered safe for consumption and has a long history of use in the food industry. However, individuals who are immunocompromised or have other health conditions should consult with a healthcare professional before consuming products containing Streptococcus thermophilus. 

·        What is Escherichia coli in your gut?

Escherichia coli (E. coli) is a type of bacteria that can be found in the gut of humans and animals. It is a natural part of the gut microbiota, where it plays an important role in maintaining gut health.

Here are some key points about E. coli in the gut:

  • E. coli strains in the gut are generally considered to be harmless or even beneficial to human health, although there are some strains that can cause illness when they are present in other parts of the body or when they are consumed through contaminated food or water .
  • E. coli in the gut can help to prevent colonization by pathogenic bacteria, produce essential vitamins and nutrients, and regulate the immune system (2, 3).
  • E. coli strains in the gut are highly diverse and can vary depending on factors such as age, diet, and health status .
  • Disruptions to the gut microbiota, such as those caused by antibiotics or poor diet, can lead to overgrowth of harmful E. coli strains and increase the risk of infection and disease .
  • Some probiotic supplements contain strains of E. coli that have been shown to have beneficial effects on gut health, although more research is needed to fully understand their mechanisms of action .

Overall, E. coli is a natural and important part of the gut microbiota that can play a beneficial role in gut health when present in appropriate amounts and strains. 

·        What is Bacteroides fragilis & Is it good or bad?

Bacteroides fragilis is generally considered to be a beneficial bacterium in the gut, as it is involved in many important functions such as digestion, immune system regulation, and protection against harmful microorganisms. In certain situations, B. fragilis can also become pathogenic and cause infections such as sepsis and abdominal abscesses, particularly in individuals with compromised immune systems or underlying health conditions.

Some of the functions and benefits of Bacteroides fragilis include:

  • Immune system modulation: Bacteroides fragilis has been shown to help regulate the immune system by promoting the development of regulatory T cells, which help to prevent excessive inflammation and autoimmune responses.
  • Protection against pathogens: Bacteroides fragilis produces various antimicrobial peptides that can help to protect against pathogenic bacteria in the gut.
  • Production of short-chain fatty acids: Bacteroides fragilis is capable of producing short-chain fatty acids (SCFAs), which have been shown to have various health benefits, such as improving gut barrier function and reducing inflammation.
  • Breakdown of complex carbohydrates: Bacteroides fragilis is able to break down complex carbohydrates that cannot be digested by the host, allowing for the release of nutrients that can be utilized by other members of the gut microbiota.

However, Bacteroides fragilis has been implicated in various diseases such as inflammatory bowel disease (IBD), colorectal cancer, and sepsis. Studies have shown that increased abundance of Bacteroides fragilis in the gut is associated with inflammation and the development of IBD. Furthermore, certain strains of Bacteroides fragilis have been found to carry virulence factors such as metalloproteases, lipases, and endotoxins, which can cause tissue damage and inflammation. In sepsis, Bacteroides fragilis is a common cause of infection and can lead to life-threatening complications.

Bacteroides fragilis is a complex bacterial species with both beneficial and harmful effects on the host. While it plays an important role in maintaining gut homeostasis, excessive or pathogenic strains can lead to inflammatory and disease states. Therefore, it is important to maintain a balance of Bacteroides fragilis and other gut bacteria to promote overall gut health. 

·        What is Clostridium butyricum?

Clostridium butyricum is a gram-positive, anaerobic bacterium that is commonly found in soil and the gastrointestinal tract of humans and animals. It is one of the most extensively studied species of the Clostridium genus due to its numerous health benefits.

One of the most significant health benefits of Clostridium butyricum is its ability to produce butyric acid,a short-chain fatty acid that has been shown to have numerous positive effects on the body. Butyric acid is an important energy source for colonocytes, the cells that line the colon, and is also involved in regulating inflammation and improving gut barrier function. Studies have suggested that butyric acid may also have potential therapeutic benefits for inflammatory bowel disease, colon cancer, and metabolic disorders such as type 2 diabetes.

In addition to its role in butyric acid production, Clostridium butyricum has also been shown to have other health-promoting effects, including:

  1. Regulation of the immune system: Research has suggested that Clostridium butyricum may help regulate the immune system by increasing the production of anti-inflammatory cytokines and reducing the production of pro-inflammatory cytokines. This effect may have potential therapeutic benefits for a range of conditions associated with inflammation, such as autoimmune diseases and allergies.
  2. Antibacterial activity: Clostridium butyricum has been shown to have antibacterial activity against a range of pathogenic bacteria, including Staphylococcus aureus and Helicobacter pylori. This antibacterial activity may help protect against infections and reduce the risk of antibiotic resistance.
  3. Promotion of healthy gut microbiota: Clostridium butyricum has been shown to promote the growth of other beneficial bacteria in the gut, such as Lactobacillus and Bifidobacterium. This may help maintain a healthy balance of gut microbiota and reduce the risk of dysbiosis, which has been linked to a range of health problems.
  4. Prevention of antibiotic-associated diarrhea: Clostridium butyricum has been shown to be effective in preventing antibiotic-associated diarrhea, a common side effect of antibiotic therapy. This effect may be due to its ability to restore the balance of gut microbiota disrupted by antibiotics.
  5. Prevention of respiratory tract infections: Some studies have suggested that Clostridium butyricum may help prevent respiratory tract infections, particularly in children. This effect may be due to its ability to modulate the immune system and enhance the production of antibodies.

While Clostridium butyricum has numerous health benefits, it should be noted that it can also have negative effects in certain circumstances. For example, in individuals with compromised immune systems, Clostridium butyricum can cause infections such as sepsis and necrotizing enterocolitis. Additionally, excessive growth of Clostridium butyricum in the gut can lead to the production of toxic metabolites that can cause inflammation and damage to the gut lining.

In conclusion, Clostridium butyricum is a beneficial bacterium that plays an important role in the gut microbiota and has numerous health-promoting effects. Its ability to produce butyric acid and regulate the immune system, as well as its antibacterial activity and ability to promote the growth of other beneficial bacteria, make it a valuable addition to the gut microbiome. However, as with all bacteria, it is important to maintain a healthy balance and avoid excessive growth, particularly in individuals with compromised immune systems. 

·        What is Ruminococcus?

Ruminococcus is a type of gram-positive anaerobic bacteria that is commonly found in the human gut microbiota. It is a member of the family Ruminococcaceae, which is one of the most abundant bacterial families in the human gut.

Ruminococcus bacteria are known for their ability to ferment complex carbohydrates, which is an important process for the production of short-chain fatty acids (SCFAs) such as butyrate. Butyrate is a key energy source for colonocytes and has been linked to several health benefits, including improved gut barrier function and reduced inflammation. Ruminococcus bacteria have also been shown to play a role in the metabolism of amino acids and bile acids, as well as the production of neurotransmitters such as serotonin.

Research has identified several species of Ruminococcus that are particularly abundant in the human gut and have been linked to specific health outcomes. Some of these species include:

  1. Ruminococcus bromii: This species is known for its ability to break down resistant starch, a type of dietary fiber that is not digested in the small intestine. Resistant starch is fermented by R. bromii in the large intestine, leading to the production of butyrate and other beneficial SCFAs.
  2. Ruminococcus champanellensis: This species has been found to be more abundant in the gut microbiota of individuals with healthy glucose metabolism, suggesting a potential role in the prevention of type 2 diabetes.
  3. Ruminococcus gnavus: This species has been linked to the production of toxic metabolites that may contribute to the development of inflammatory bowel disease (IBD). However, other studies have suggested that R. gnavus may also have anti-inflammatory properties and could play a beneficial role in the gut microbiota.
  4. Ruminococcus obeum: This species has been shown to produce a variety of SCFAs, including butyrate and propionate, which have been linked to improved gut health and reduced risk of disease.

While Ruminococcus bacteria are generally considered to be beneficial for human health, there are some studies that have suggested a potential link between certain species of Ruminococcus and the development of gut dysbiosis and disease. For example, a 2018 study found that an overgrowth of Ruminococcus gnavus was associated with an increased risk of developing colorectal cancer.

Overall, the research suggests that Ruminococcus bacteria are an important component of the human gut microbiota and play a key role in the fermentation of complex carbohydrates and the production of beneficial metabolites such as butyrate. While some species of Ruminococcus have been linked to disease, further research is needed to fully understand their potential roles in the gut microbiota and their impact on human health.

·        What is Lactobacillus acidophilus?

Lactobacillus acidophilus is a species of bacteria that belongs to the Lactobacillus genus, which is a group of lactic acid bacteria commonly found in the human gut and other environments. It is a gram-positive, anaerobic, and rod-shaped bacteria that produces lactic acid as a byproduct of fermentation. It can also be found in some fermented foods, such as yogurt, kefir, and sauerkraut.

  1. acidophilus is considered a probiotic, which means it is a beneficial bacteria that can have a positive impact on human health when consumed in adequate amounts. It has been studied for its potential health benefits in a number of areas, including digestive health, immune function, and vaginal health.

Here are some potential benefits of L. acidophilus, supported by research:

  1. Digestive health: L. acidophilus has been shown to have a positive impact on digestive health by helping to break down lactose, reducing the symptoms of lactose intolerance. It has also been studied for its potential to alleviate symptoms of irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD) by reducing inflammation in the gut and improving gut barrier function.
  2. Immune function: L. acidophilus may have a positive impact on immune function by stimulating the production of antibodies and enhancing the activity of natural killer cells, which are important components of the immune system.
  3. Vaginal health: L. acidophilus is a naturally occurring bacteria in the vaginal microbiome and is important for maintaining a healthy vaginal environment. It can help prevent and treat bacterial vaginosis, yeast infections, and urinary tract infections by producing lactic acid, which lowers the pH of the vagina and inhibits the growth of harmful bacteria.
  4. Cholesterol management: Some research suggests that L. acidophilus may help lower cholesterol levels by breaking down bile in the gut, which is necessary for cholesterol metabolism.
  5. Allergy prevention: L. acidophilus may also play a role in preventing allergies by modulating the immune system and reducing inflammation.
  6. L. acidophilus can be found in a variety of food sources, including yogurt, kefir, sauerkraut, kimchi, and other fermented foods. It is also available in supplement form, either alone or in combination with other probiotic strains. Overall, L. acidophilus is considered a safe and beneficial probiotic for most people. 

·        What is Lactobacillus reuteri?

Lactobacillus reuteri is a Gram-positive, rod-shaped bacterium that is part of the Lactobacillus genus. It is found in the human gastrointestinal tract, oral cavity, and breast milk. Lactobacillus reuteri is considered a probiotic bacterium due to its ability to promote health in the host. It has been studied extensively for its potential therapeutic applications, including the treatment of gastrointestinal disorders, immune modulation, and prevention of infections.

One of the most well-known strains of Lactobacillus reuteri is DSM 17938, which has been extensively studied in clinical trials. This strain has been shown to have a number of beneficial effects on the gastrointestinal tract, including reducing symptoms of functional gastrointestinal disorders such as colic and irritable bowel syndrome. It has also been shown to have immunomodulatory effects, promoting the production of anti-inflammatory cytokines and enhancing the activity of natural killer cells.

Another strain of Lactobacillus reuteri, ATCC PTA 6475, has been studied for its potential effects on oral health. This strain has been shown to reduce the levels of Streptococcus mutans, a bacteria associated with dental caries, in saliva and dental plaque. It has also been shown to reduce the levels of periodontal pathogens in the oral cavity, leading to a reduction in inflammation and improved periodontal health.

In addition to its potential effects on gastrointestinal and oral health, Lactobacillus reuteri has been studied for its potential immunomodulatory effects. It has been shown to have anti-inflammatory effects in animal models of inflammatory bowel disease and to enhance the production of anti-inflammatory cytokines in human cells. These effects may make Lactobacillus reuteri a promising candidate for the prevention and treatment of inflammatory disorders such as arthritis and psoriasis.

The benefits of L. reuteri can be summarized as: 

  • L. reuteri has been shown to promote bone health by increasing bone mineral density and improving bone microstructure in animal studies.
  • Some studies suggest that L. reuteri may have potential as a therapeutic agent for treating acne, as it has been shown to reduce inflammation and improve skin barrier function.
  • L. reuteri has also been studied for its potential to improve oral health by reducing plaque formation and promoting the growth of beneficial bacteria in the mouth.
  • In infants, L. reuteri has been shown to have a protective effect against colic and regurgitation.
  • Preliminary studies have also suggested that L. reuteri may have potential as a therapeutic agent for treating gastrointestinal disorders such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS).

Overall, Lactobacillus reuteri is a promising probiotic bacterium with a wide range of potential health benefits. While more research is needed to fully understand its mechanisms of action and potential therapeutic applications, current evidence suggests that it may be useful in the prevention and treatment of a variety of health conditions.

There are several ways to increase the amount of L. reuteri in your gut, including:

  1. Consuming fermented foods: Foods like kefir, yogurt, sauerkraut, and kimchi contain live cultures of L. reuteri that can help increase its levels in your gut.
  2. Taking probiotic supplements: L. reuteri is available in probiotic supplements, which can be found in health food stores or online. 
  3. Breastfeeding: L. reuteri is present in breast milk, and breastfeeding has been shown to help establish a healthy gut microbiome in infants, including L. reuteri.
  4. Prebiotic foods: Consuming prebiotic-rich foods like asparagus, garlic, onions, and green bananas can also support the growth of L. reuteri in the gut.
  5. Avoiding antibiotics: Antibiotics can disrupt the natural balance of bacteria in the gut and reduce the levels of L. reuteri. If you need to take antibiotics, talk to your healthcare provider about strategies to support your gut health during and after treatment.
  6. Managing stress: Chronic stress can negatively affect the gut microbiome and reduce the levels of beneficial bacteria like L. reuteri. Practicing stress-management techniques like meditation, deep breathing, and yoga may help support the growth of L. reuteri in the gut. 

·        What is Streptococcus salivarius?

Streptococcus salivarius is a Gram-positive, facultative anaerobic bacterium that is commonly found in the oral cavity, upper respiratory tract, and gastrointestinal tract of humans. It is considered as a commensal organism that plays an important role in maintaining oral and overall health.

Here are some key points about Streptococcus salivarius:

  • Streptococcus salivarius is a member of the genus Streptococcus and is one of the most prevalent bacteria in the human oral cavity. It has a spherical or ovoid shape, appears in chains or pairs, and is non-motile.
  • One of the most significant features of Streptococcus salivarius is its ability to produce bacteriocins, which are antimicrobial peptides that can inhibit the growth of other bacteria in the oral cavity, including Streptococcus mutans, a major cause of dental caries.
  • Streptococcus salivarius has also been shown to have immunomodulatory properties. It can stimulate the production of anti-inflammatory cytokines, such as IL-10 and TGF-β, and reduce the production of pro-inflammatory cytokines, such as IL-1β and TNF-α, thereby regulating the immune response and promoting oral health.
  • In addition to its role in oral health, Streptococcus salivarius has been studied for its potential therapeutic applications in other areas. For example, some strains have been found to produce bacteriocins that can inhibit the growth of Helicobacter pylori, a bacterium associated with gastric ulcers and stomach cancer.
  • Streptococcus salivarius is generally considered safe and is widely used as a probiotic in various products, including oral hygiene products, dietary supplements, and functional foods.

Overall, Streptococcus salivarius is a commensal bacterium that plays a vital role in maintaining oral health and has potential therapeutic applications in other areas. Its ability to produce bacteriocins and modulate the immune response makes it an attractive candidate for further research and development of probiotic products. 

·        What is Streptococcus mutans? 

Streptococcus mutans is a gram-positive bacterium that is commonly found in the human oral cavity. It is one of the main bacteria responsible for tooth decay and dental caries. S. mutans is part of the normal oral microbiota, but its overgrowth can cause significant harm to the teeth and gums. In this article, we will discuss the characteristics of S. mutans, its role in tooth decay, and ways to prevent its overgrowth.

  1. mutans is a non-motile, non-sporulating, and facultatively anaerobic bacterium that belongs to the Streptococcaceae family. It is an oval-shaped bacterium that forms chains or pairs. S. mutans is known for its ability to produce sticky, extracellular polysaccharides called glucans, which allow it to adhere to the tooth surface and form biofilms. These biofilms protect the bacteria from host defenses and antimicrobial agents and enable it to ferment dietary carbohydrates and produce lactic acid, which can cause tooth demineralization and eventually lead to cavities.

The presence of S. mutans in the oral cavity is a significant risk factor for dental caries. Caries is a multifactorial disease that results from the interplay of several factors, including the composition of the oral microbiota, host factors, and dietary habits. The caries process starts when acid-producing bacteria such as S. mutans ferment carbohydrates, producing organic acids that lower the pH of the dental plaque. The acidic environment can then dissolve the tooth enamel, leading to the formation of a cavity.

Prevention of S. mutans overgrowth is essential in maintaining good oral health. The primary approach to preventing S. mutans overgrowth is through good oral hygiene practices such as brushing twice a day with fluoride toothpaste, flossing daily, and using mouthwash. A healthy diet low in added sugars and high in fiber can also reduce the risk of S. mutans overgrowth. Moreover, the use of probiotics containing beneficial bacteria such as Lactobacillus and Bifidobacterium can help in reducing the colonization of S. mutans in the oral cavity. The characteristics of Streptococcus mutans can be summarized:

  • Streptococcus mutans is a type of bacteria that is commonly found in the human mouth and is known to cause dental caries or tooth decay.
  • It is an acidogenic and aciduric bacteria, which means it can produce acids and survive in an acidic environment, leading to demineralization of tooth enamel and the formation of dental caries.
  • S. mutans is also able to form biofilms on the tooth surface, allowing it to adhere and colonize more effectively, making it difficult to remove by normal oral hygiene practices such as brushing and flossing.
  • Risk factors for colonization and growth of S. mutans include poor oral hygiene, a diet high in sugars and carbohydrates, decreased saliva production, and a compromised immune system.
  • The use of fluoride and other remineralizing agents, along with improved oral hygiene practices, can help prevent the growth and colonization of S. mutans and the formation of dental caries.
  • In addition to dental caries, S. mutans has been implicated in the development of other systemic diseases, such as infective endocarditis, pneumonia, and septicemia.
  • Treatment for S. mutans infections may include the use of antibiotics, although antibiotic resistance is a concern with this approach.
  • Strategies to prevent the growth and colonization of S. mutans include the use of probiotics, such as Streptococcus salivarius, which can compete with S. mutans for colonization sites and nutrients in the oral cavity.
  • Other strategies for preventing S. mutans growth include reducing the consumption of sugars and carbohydrates, increasing saliva production through chewing gum or other methods, and maintaining good oral hygiene practices.
  • Regular dental check-ups and cleanings are also important for preventing the formation and progression of dental caries caused by S. mutans.

In conclusion, S. mutans is a gram-positive bacterium that is commonly found in the oral cavity and is a significant risk factor for dental caries. Its ability to produce glucans and form biofilms enables it to adhere to the tooth surface and ferment dietary carbohydrates, producing lactic acid that can cause tooth demineralization. Preventing its overgrowth through good oral hygiene practices, a healthy diet, and the use of probiotics can help in maintaining good oral health.

·        What is the magic yeast - Saccharomyces boulardii?

Saccharomyces boulardii, commonly known as the "magic yeast," is a probiotic yeast strain that has been extensively studied for its beneficial effects on gut health. This unique yeast strain is not a part of the normal gut microbiota, but it has been found to colonize the gut and exert several positive effects on the host. In this response, we will provide an overview of Saccharomyces boulardii, its potential health benefits, and the scientific evidence supporting its use.

Overview:

  • Saccharomyces boulardii is a non-pathogenic yeast strain that is resistant to stomach acid and bile salts, allowing it to survive passage through the gastrointestinal tract and colonize the gut.
  • It was first isolated from lychee and mangosteen fruit in 1923 by French scientist Henri Boulard, and has since been extensively studied for its probiotic properties.
  • Unlike other probiotics, which are bacteria, Saccharomyces boulardii is a yeast and belongs to the same family as baker's yeast (Saccharomyces cerevisiae).
  • It is available as a dietary supplement and is often used to support digestive health and treat various gastrointestinal disorders.

Potential Health Benefits:

  • Supports gastrointestinal health: Saccharomyces boulardii has been found to help maintain the balance of beneficial bacteria in the gut, support the intestinal barrier function, and reduce inflammation in the gut.
  • Helps prevent and treat diarrhea: Numerous clinical trials have shown that Saccharomyces boulardii can effectively prevent and treat various types of diarrhea, including antibiotic-associated diarrhea, traveler's diarrhea, and Clostridium difficile infection.
  • Reduces symptoms of irritable bowel syndrome (IBS): Some studies have suggested that Saccharomyces boulardii may help reduce abdominal pain, bloating, and other symptoms of IBS.
  • Supports immune function: Saccharomyces boulardii has been found to stimulate the production of secretory immunoglobulin A (sIgA), an important component of the immune system that helps protect the body against pathogens.
  • Helps protect against gut dysbiosis: Saccharomyces boulardii has been found to help protect against gut dysbiosis, which is an imbalance of the gut microbiota that has been linked to various health problems.

Scientific Evidence:

  • A meta-analysis of 23 randomized controlled trials found that Saccharomyces boulardii significantly reduced the risk of antibiotic-associated diarrhea.
  • Another meta-analysis of 31 clinical trials found that Saccharomyces boulardii was effective in preventing and treating Clostridium difficile infection.
  • A double-blind, placebo-controlled study of 57 patients with IBS found that Saccharomyces boulardii significantly reduced abdominal pain, bloating, and other symptoms.
  • Animal studies have shown that Saccharomyces boulardii can reduce gut inflammation and improve gut barrier function.
  • A study in healthy volunteers found that Saccharomyces boulardii increased the production of sIgA, suggesting that it may have immune-boosting effects.

In summary, Saccharomyces boulardii is a unique probiotic yeast strain that has been found to have several potential health benefits, particularly for digestive health. The scientific evidence supporting its use is robust, and it is considered safe and well-tolerated for most people. 

·        What is Enterococcus faecium? 

Enterococcus faecium is a Gram-positive bacterium that is commonly found in the intestinal tracts of humans and animals, as well as in soil and water. It is a facultative anaerobe, meaning that it can grow in the presence or absence of oxygen. While Enterococcus faecium is typically considered a commensal microbe, it has been associated with various infections, especially in immunocompromised individuals. In this article, we will discuss the characteristics of Enterococcus faecium, its potential benefits, and the possible health concerns associated with its presence in the gut.

Characteristics of Enterococcus faecium

Enterococcus faecium is a spherical bacterium that is approximately 0.5-1.0 μm in diameter. It is a member of the Enterococcaceae family and is closely related to other Enterococcus species, including Enterococcus faecalis, Enterococcus hirae, and Enterococcus durans. Enterococcus faecium is capable of surviving in a wide range of environmental conditions, including high salt concentrations and low pH levels. It is also resistant to several antibiotics, which has led to concerns about its potential role in antibiotic resistance.

Potential benefits of Enterococcus faecium

Despite its association with infections, Enterococcus faecium has been found to have some potential benefits when present in the gut. Here are a few potential benefits of Enterococcus faecium:

  1. Probiotic properties: Enterococcus faecium has been identified as a potential probiotic, which means that it may provide health benefits when consumed in adequate amounts. Some studies have suggested that Enterococcus faecium may help to regulate the immune system and improve the barrier function of the gut.
  2. Production of antimicrobial substances: Enterococcus faecium is capable of producing antimicrobial substances, including bacteriocins and organic acids. These substances can help to inhibit the growth of harmful bacteria in the gut, potentially reducing the risk of infections.
  3. Digestive health: Enterococcus faecium may help to promote digestive health by aiding in the breakdown of food and the absorption of nutrients. It may also help to prevent diarrhea by reducing the growth of pathogenic bacteria in the gut.

Health concerns associated with Enterococcus faecium

While Enterococcus faecium has potential benefits, it has also been associated with various infections. Here are some of the health concerns associated with Enterococcus faecium:

  1. Antibiotic resistance: Enterococcus faecium is naturally resistant to several antibiotics, and it has been identified as a significant contributor to the spread of antibiotic resistance. This is particularly concerning in hospital settings, where Enterococcus faecium infections can be difficult to treat.
  2. Infections: Enterococcus faecium has been associated with various infections, including urinary tract infections, endocarditis, and bacteremia. These infections are often more severe in individuals who are immunocompromised or have underlying medical conditions.
  3. Virulence factors: Enterococcus faecium is capable of producing various virulence factors that can contribute to its pathogenicity. These factors include adhesins, biofilm formation, and capsule production.

Overall, the presence of Enterococcus faecium in the gut can have both potential benefits and health concerns. Further research is needed to better understand the role of Enterococcus faecium in gut health and the risks associated with its presence. 

·        What is Enterococcus durans?

Enterococcus durans is a lactic acid bacteria that is commonly found in the human gastrointestinal tract, as well as in various foods and the environment. It is a gram-positive, non-motile, facultatively anaerobic bacterium that is able to produce lactic acid through the fermentation of carbohydrates. Here are some key facts about Enterococcus durans:

  1. Taxonomy and Characteristics: Enterococcus durans is a member of the genus Enterococcus, which is a diverse group of lactic acid bacteria that are commonly found in various environments including the human gut, soil, water, and food products. The species Enterococcus durans is characterized by its ability to grow in the presence of high salt concentrations and its resistance to vancomycin, a common antibiotic.
  2. Role in the gut microbiome: Like other members of the genus Enterococcus, Enterococcus durans is a commensal bacterium that is typically found in the human gastrointestinal tract. It has been shown to play a role in maintaining gut homeostasis, modulating the immune system, and preventing colonization by pathogenic bacteria.
  3. Probiotic potential: Given its role in the gut microbiome and its potential health benefits, Enterococcus durans has been investigated as a potential probiotic. Some studies have shown that the administration of Enterococcus durans can improve gut health, reduce inflammation, and enhance the immune response.
  4. Food applications: Enterococcus durans is commonly found in various fermented foods, including dairy products, sausages, and vegetables. It is also commonly used as a starter culture in the production of fermented foods such as cheese and sourdough bread.
  5. Safety concerns: While Enterococcus durans is generally considered safe and is commonly found in various foods, there have been some concerns about its potential pathogenicity. Some strains of Enterococcus durans have been shown to possess virulence factors and antibiotic resistance genes, which could pose a risk to human health.

In conclusion, Enterococcus durans is a common member of the human gut microbiome and has been shown to have potential health benefits as a probiotic. It is also commonly found in various foods and is used as a starter culture in the production of fermented foods. However, safety concerns do exist regarding its potential pathogenicity and further research is needed to fully understand its role in human health. 

·        What is Bacteroides thetaiotaomicron?

Bacteroides thetaiotaomicron, commonly known as B. theta, is a Gram-negative, anaerobic bacterium that inhabits the human gut. It is a member of the Bacteroidetes phylum and is one of the most abundant bacterial species found in the human intestinal microbiota. B. theta plays an important role in maintaining gut health by breaking down complex dietary polysaccharides and producing short-chain fatty acids (SCFAs). Additionally, B. theta has been associated with various health benefits such as promoting anti-inflammatory responses, regulating the immune system, and protecting against certain diseases.

Here are some key points about B. theta:

  • Dietary polysaccharide degradation: B. theta is known for its ability to break down complex dietary polysaccharides, such as inulin and pectin, that cannot be digested by human enzymes. These polysaccharides serve as a source of energy for the bacteria and help to maintain the health of the gut microbiota.
  • Short-chain fatty acid production: As B. theta degrades dietary polysaccharides, it produces short-chain fatty acids (SCFAs) as a byproduct. SCFAs are important energy sources for colonocytes and can also regulate immune function, promote anti-inflammatory responses, and improve gut barrier function.
  • Anti-inflammatory responses: B. theta has been shown to promote anti-inflammatory responses in the gut by inducing regulatory T cells and promoting the production of anti-inflammatory cytokines. This may help to protect against inflammatory bowel diseases such as ulcerative colitis and Crohn's disease.
  • Immune system regulation: B. theta has been shown to modulate the immune system by promoting the development of immune cells and regulating the production of immune mediators. This may help to protect against autoimmune diseases and infections.
  • Protection against certain diseases: Studies have suggested that B. theta may play a role in protecting against certain diseases such as colon cancer, metabolic disorders, and cardiovascular diseases.

However, in certain cases, an overgrowth of B. thetaiotaomicron can be associated with certain health conditions. One such condition is inflammatory bowel disease (IBD). Studies have shown that individuals with IBD have higher levels of B. thetaiotaomicron in their gut microbiome than healthy individuals. Additionally, B. thetaiotaomicron has been shown to increase the production of pro-inflammatory cytokines in the gut, which can exacerbate inflammation in individuals with IBD.

Another potential negative effect of B. thetaiotaomicron overgrowth is its ability to produce a toxin called fragilysin. Fragilysin has been shown to damage the intestinal barrier and increase gut permeability, leading to a variety of health issues including inflammation, metabolic disorders, and even neurological disorders.

Research on B. theta is still ongoing, and scientists continue to explore the mechanisms by which this bacterium contributes to gut health. Understanding the role of B. theta in the gut microbiota may lead to the development of new therapeutic strategies for gut-related diseases.

·        What is the controversy Clostridium difficile?

Clostridium difficile, commonly known as C. diff, is a bacterium that can cause severe diarrhea and other intestinal problems. The bacterium can be found in the gut of healthy individuals, but when the balance of the gut microbiome is disrupted, C. diff can overgrow and cause disease.

Clostridium difficile is considered a harmful bacterium because it produces toxins that can cause severe diarrhea and inflammation of the colon. The toxins, known as toxins A and B, damage the cells lining the colon, leading to inflammation and fluid accumulation in the colon. This results in diarrhea, abdominal pain, fever, and in severe cases, colitis or pseudomembranous colitis, which can be life-threatening.

  1. difficile infections typically occur in individuals who have recently taken antibiotics, which can disrupt the normal balance of bacteria in the gut and create an environment favorable to C. difficile growth. The elderly, hospitalized patients, and those with weakened immune systems are at higher risk for C. difficile infections.

The symptoms of C. difficile infection include:

  • Diarrhea (often watery and frequent)
  • Abdominal pain and cramping
  • Nausea and vomiting
  • Fever
  • Loss of appetite
  • Dehydration

In severe cases, C. difficile infection can lead to colitis or pseudomembranous colitis, which can cause complications such as:

  • Bowel perforation
  • Toxic megacolon (severe colon inflammation)
  • Septicemia (blood infection)
  • Kidney failure
  • Death

Prompt diagnosis and treatment are essential to prevent complications and reduce the risk of transmission to others.

Cause of C. difficile: Antibiotics have been found to be a significant risk factor for C. diff infection. Antibiotics can disrupt the natural balance of the gut microbiome, leading to the overgrowth of C. diff. This overgrowth can cause diarrhea, inflammation, and other intestinal problems. Studies have shown that up to 20% of individuals who receive antibiotics may develop C. diff infections, and the risk is even higher for those who receive broad-spectrum antibiotics or multiple courses of antibiotics.

Treatment: While antibiotics are often used to treat C. diff infections, there has been concern about the development of antibiotic-resistant strains of C. diff. Antibiotic use can also further disrupt the gut microbiome, which can prolong C. diff infections or lead to recurrence of the infection. As a result, there has been growing interest in the use of FMT as an alternative treatment for C. diff infections.

Fecal microbiota transplantation (FMT): FMT involves the transfer of fecal material from a healthy donor into the gut of a patient with C. diff infection. The goal of FMT is to restore the balance of the gut microbiome and eliminate the overgrowth of C. diff. While FMT has been found to be the most effective method for treating C. diff infections, there has been controversy surrounding its use.

One concern about FMT is the potential risk of transmitting infectious diseases from the donor to the recipient. Donors must be carefully screened for infectious diseases, and the fecal material must be prepared and administered under strict conditions to minimize the risk of infection. Despite these precautions, there have been reports of infections and adverse events associated with FMT.

Another concern about FMT is the lack of standardized protocols for preparation and administration. There is currently no consensus on the optimal method for preparing and administering fecal material, which can lead to variability in effectiveness and safety. Additionally, FMT is not widely available and can be expensive, which can limit its use as a treatment option.

While there is controversy surrounding the use of FMT for C. diff infections, there is growing interest in the potential of FMT for other conditions that are thought to be related to the gut microbiome, such as inflammatory bowel disease and metabolic disorders. As research in this area continues, it is likely that the controversy surrounding FMT will continue as well.

In summary, C. diff is a bacterium that can cause severe intestinal problems, and antibiotics are a significant risk factor for infection. While antibiotics are often used to treat C. diff infections, there has been growing interest in the use of FMT as an alternative treatment. However, there are concerns about the potential risk of infectious disease transmission and the lack of standardized protocols for FMT. As research in this area continues, it is likely that the controversy surrounding FMT will continue as well. 

·        What are the top reasons for constipation and how do you address it?

Constipation is a common gastrointestinal issue that can be caused by a variety of factors. According to the National Institute of Diabetes and Digestive and Kidney Diseases, constipation is defined as having fewer than three bowel movements per week, passing hard or dry stools, experiencing difficulty passing stools, or feeling incomplete evacuation after bowel movements.

Here are the top reasons for constipation and ways to address them:

  1. Low fiber diet: A low intake of dietary fiber can lead to constipation because fiber adds bulk to stool and helps it move through the digestive system. To address this, increasing fiber intake by incorporating fruits, vegetables, whole grains, nuts, and seeds into the diet can be helpful.
  2. Dehydration: Dehydration can cause the body to absorb more water from the colon, making stools harder and more difficult to pass. Drinking more water and fluids can help address dehydration-related constipation.
  3. Lack of physical activity: A sedentary lifestyle can lead to constipation because physical activity helps stimulate the muscles in the intestines, promoting regular bowel movements. Increasing physical activity, such as walking or jogging, can help alleviate constipation.
  4. Medications: Certain medications, such as opioid pain relievers, antacids containing calcium or aluminum, and antidepressants, can cause constipation as a side effect. It is important to talk to a healthcare provider about any medication-related constipation and potential alternatives.
  5. Medical conditions: Some medical conditions, such as irritable bowel syndrome, hypothyroidism, and diabetes, can cause constipation. Treating and managing these underlying conditions can help alleviate constipation.
  6. Mental health conditions: Mental health conditions, such as anxiety and depression, can also contribute to constipation. Managing mental health through therapy, medication, or lifestyle changes can help address constipation caused by these conditions.
  7. Ignoring the urge to have a bowel movement: Ignoring the urge to have a bowel movement can lead to constipation because the longer stool remains in the colon, the harder and drier it becomes. It is important to listen to and respond to the body’s signals and have a bowel movement when the urge arises.
  8. Pregnancy: Hormonal changes during pregnancy can cause constipation. Maintaining a healthy diet, staying hydrated, and engaging in physical activity can help address constipation during pregnancy.
  9. Aging: As people age, the muscles in the intestines may weaken, leading to constipation. Engaging in physical activity and consuming a high-fiber diet can help address constipation in older adults.
  10. Imbalanced Gut Microbiome: Studies have shown that alterations in the gut microbiota, such as a reduction in the abundance of beneficial bacteria like Bifidobacterium and Lactobacillus, and an increase in potentially harmful bacteria like Enterobacteriaceae, are associated with an increased risk of constipation. Additionally, certain types of fiber, known as prebiotics, can stimulate the growth of beneficial bacteria in the gut and improve bowel function, potentially reducing the risk of constipation.

In conclusion, constipation can be caused by a variety of factors, including low fiber diet, dehydration, lack of physical activity, medications, medical and mental health conditions, ignoring the urge to have a bowel movement, pregnancy, and aging. Addressing these factors through dietary changes, physical activity, hydration, and management of underlying medical and mental health conditions can help alleviate constipation and promote regular bowel movements.

·        How do you strengthen gut linings?

The gut lining is an important barrier that protects the body from harmful substances and pathogens. It also plays a crucial role in nutrient absorption and immune function. A healthy gut lining is essential for overall health and wellbeing. However, many factors can damage the gut lining, such as poor diet, stress, medications, and environmental toxins. Fortunately, there are several ways to strengthen the gut lining and support optimal gut health.

  1. Consume gut-supportive foods:

Consuming a diet rich in whole, unprocessed foods can support gut health and strengthen the gut lining. Foods that are particularly beneficial for the gut include:

  • Fermented foods: Fermented foods like kefir, sauerkraut, kimchi, and kombucha are rich in beneficial bacteria that can help to balance the gut microbiome and support gut health.
  • Prebiotic foods: Prebiotic foods like garlic, onions, leeks, asparagus, and bananas contain fiber that feeds beneficial gut bacteria and supports the growth of a healthy gut microbiome.
  • Bone broth: Bone broth is rich in collagen, a protein that can help to strengthen the gut lining and reduce inflammation in the gut.
  • Omega-3 fatty acids: Omega-3 fatty acids, found in fatty fish like salmon and sardines, can help to reduce inflammation in the gut and support gut health.
  • Prebiotic supplement: like human milk oligosaccharides (HMO) prebiotics
  1. Supplement with gut-supportive nutrients:

In addition to consuming gut-supportive foods, supplementing with certain nutrients can help to strengthen the gut lining and support gut health. These include:

  • L-glutamine: L-glutamine is an amino acid that can help to repair and strengthen the gut lining. It is also essential for immune function and can help to reduce inflammation in the gut.
  • Zinc: Zinc is a mineral that is essential for gut health. It can help to repair and strengthen the gut lining and support immune function.
  • Vitamin D: Vitamin D is important for immune function and can help to reduce inflammation in the gut.
  1. Reduce stress:

Chronic stress can contribute to gut lining damage and disrupt the gut microbiome. Reducing stress through practices like meditation, yoga, or deep breathing exercises can help to support gut health and strengthen the gut lining.

  1. Avoid gut-damaging substances:

Certain substances can damage the gut lining and disrupt the gut microbiome. These include:

  • Antibiotics: Antibiotics can kill off beneficial gut bacteria and disrupt the balance of the gut microbiome.
  • Nonsteroidal anti-inflammatory drugs (NSAIDs): NSAIDs like aspirin and ibuprofen can damage the gut lining and contribute to leaky gut syndrome.
  • Artificial sweeteners: Artificial sweeteners like saccharin, aspartame, and sucralose can disrupt the gut microbiome and contribute to gut inflammation.
  1. Support detoxification:

Toxins and environmental pollutants can contribute to gut lining damage and disrupt the gut microbiome. Supporting the body's natural detoxification pathways through practices like sauna use or regular exercise can help to support gut health and strengthen the gut lining.

In summary, there are several ways to strengthen the gut lining and support optimal gut health. Consuming a diet rich in gut-supportive foods, supplementing with gut-supportive nutrients, reducing stress, avoiding gut-damaging substances, and supporting detoxification can all contribute to a healthy gut lining and overall gut health.

·         What are the top gut irritants?

  1. Alcohol: Excessive alcohol consumption can irritate the lining of the gut and disrupt the gut microbiome. It can also increase the risk of inflammation and damage to the intestinal lining, leading to leaky gut syndrome.
  2. Spicy foods: Spicy foods can irritate the lining of the gut and trigger inflammation, leading to digestive discomfort and symptoms like bloating, gas, and diarrhea.
  3. Caffeine: Caffeine can irritate the gut lining and increase stomach acid production, leading to heartburn, acid reflux, and other digestive issues.
  4. Gluten: Gluten is a protein found in wheat, barley, and rye that can trigger an immune response in some people, leading to inflammation and damage to the gut lining. This can result in conditions like celiac disease and non-celiac gluten sensitivity.
  5. Artificial sweeteners: Artificial sweeteners like aspartame and sucralose can disrupt the gut microbiome and trigger inflammation in the gut, leading to digestive discomfort and symptoms like bloating and diarrhea.
  6. Processed foods: Processed foods are often high in sugar, salt, and unhealthy fats, and low in fiber and nutrients. They can disrupt the gut microbiome and lead to inflammation and damage to the gut lining, increasing the risk of digestive issues and chronic diseases.
  7. Non-steroidal anti-inflammatory drugs (NSAIDs): NSAIDs like ibuprofen and aspirin can irritate the lining of the gut and increase the risk of stomach ulcers and bleeding.
  8. Pesticides and herbicides: Exposure to pesticides and herbicides in food and the environment can disrupt the gut microbiome and trigger inflammation in the gut, leading to digestive issues and chronic diseases.
  9. Stress: Chronic stress can disrupt the gut microbiome and increase the risk of inflammation and damage to the gut lining, leading to digestive discomfort and symptoms like bloating, constipation, and diarrhea.

·        What foods contain gut irritants?

There are several foods that may act as gut irritants for some individuals. However, it is important to note that not all individuals will have the same response to these foods, and some people may be able to tolerate them without any issues. Some examples of gut irritant foods include:

  1. Gluten-containing grains: Wheat, barley, and rye contain gluten, a protein that can trigger inflammation in the gut and exacerbate symptoms in people with celiac disease or non-celiac gluten sensitivity.
  2. Dairy products: Many people have trouble digesting lactose, a sugar found in milk and dairy products, leading to bloating, gas, and other digestive symptoms. Additionally, some people may be sensitive to the proteins in dairy products, such as casein and whey.
  3. Processed and high-fat foods: Foods that are high in fat or heavily processed can be difficult to digest, leading to bloating and other digestive discomfort.
  4. Spicy foods: Spicy foods, particularly those containing capsaicin, can irritate the lining of the stomach and cause digestive distress.
  5. FODMAPs: Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are a group of carbohydrates that are poorly absorbed in the small intestine and can cause bloating, gas, and other digestive symptoms. FODMAPs are found in a variety of foods, including wheat, onions, garlic, beans, and some fruits.
  6. Alcohol: Alcohol can irritate the lining of the stomach and intestines, leading to inflammation and digestive symptoms.
  7. Coffee: Coffee, particularly when consumed in large amounts or on an empty stomach, can stimulate the production of stomach acid and irritate the lining of the digestive tract.
  8. Artificial sweeteners: Some artificial sweeteners, particularly those containing sugar alcohols like sorbitol, can be difficult to digest and cause bloating and other digestive discomfort.
  9. Raw cruciferous vegetables: Raw cruciferous vegetables, such as broccoli, cauliflower, and cabbage, contain compounds that can be difficult to digest and can cause bloating and gas in some people.
  10. Nightshade vegetables: Some people may be sensitive to nightshade vegetables, such as tomatoes, eggplant, and peppers, which can irritate the gut and exacerbate digestive symptoms.

·        What preservatives are bad for gut microbiome? And what food contains those preservatives?

Preservatives are used in food to extend its shelf life and prevent spoilage. While they are generally considered safe for consumption, some preservatives can have negative effects on the gut microbiome. Here are some common preservatives that can be harmful to the gut microbiome:

  1. Sodium nitrite and sodium nitrate: These preservatives are commonly used in processed meats like bacon, hot dogs, and deli meats. They have been linked to an increased risk of colorectal cancer and can disrupt the gut microbiome.
  2. Propionate: This preservative is commonly used in baked goods, dairy products, and processed meats. It has been shown to disrupt the gut microbiome and increase inflammation.
  3. Butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT): These preservatives are commonly used in processed foods, snack foods, and cereals. They have been shown to disrupt the gut microbiome and increase inflammation.
  4. Sulfites: Sulfites are commonly used in wine, dried fruits, and other preserved foods. They can cause digestive issues and disrupt the gut microbiome.
  5. Benzoates: Benzoates are commonly used in soft drinks, fruit juices, and other processed foods. They have been linked to an increased risk of inflammatory bowel disease and can disrupt the gut microbiome.

It's important to note that not all preservatives are bad for the gut microbiome, and some can even have beneficial effects. For example, fermented foods like sauerkraut and kimchi contain naturally occurring preservatives that can promote the growth of beneficial bacteria in the gut.

To avoid harmful preservatives, it's best to stick to whole, unprocessed foods as much as possible. When buying packaged foods, check the ingredients list for preservatives and try to choose products with fewer additives.

·        What is Veillonella and how does this bacteria increase athletic performance?

Veillonella is a genus of anaerobic, gram-negative bacteria that inhabit various parts of the human body, including the oral cavity, gastrointestinal tract, respiratory tract, and vagina. This genus is among the most prevalent and abundant bacterial groups found in the human gut microbiota, and recent research has revealed their potential roles in human health and disease.

Basics: Veillonella bacteria have unique metabolic capabilities that allow them to metabolize lactate, a byproduct of anaerobic glycolysis, as their primary energy source. This feature makes them a key player in the gut microbial ecosystem by enabling them to scavenge lactate produced by other gut bacteria, thereby reducing lactate accumulation, which can cause inflammation and gut dysbiosis. Veillonella bacteria also produce short-chain fatty acids (SCFAs), including propionate, which is known to have anti-inflammatory properties and regulate host metabolism.

Functions: Recent research has suggested that Veillonella bacteria have a number of important functions in human health, including:

  1. Athletic Performance: A study published in the journal Nature Medicine found that athletes with high levels of Veillonella bacteria in their gut microbiota had enhanced athletic performance. This was attributed to their ability to metabolize lactate produced during exercise, which can improve endurance.
  2. Immune Regulation: Veillonella bacteria have been shown to promote the production of anti-inflammatory cytokines, such as interleukin-10 (IL-10), which help to regulate the immune response and prevent excessive inflammation.
  3. Gut Health: Veillonella bacteria are important members of the gut microbiota and have been associated with reduced risk of inflammatory bowel disease (IBD) and other gut-related conditions.
  4. Oral Health: Veillonella bacteria are commonly found in the oral cavity, where they help to maintain oral health by producing SCFAs and preventing the growth of harmful bacteria.

Research: Recent studies have demonstrated the importance of Veillonella bacteria in human health and disease. In a study published in the journal Nature Medicine, researchers found that Veillonella bacteria were present in higher levels in the gut microbiota of elite athletes compared to sedentary individuals. Another study published in the journal Cell Host & Microbe found that Veillonella bacteria were associated with reduced risk of inflammatory bowel disease (IBD) in a mouse model.

How to boost Veillonella: There are several ways to boost the levels of Veillonella bacteria in the gut microbiota:

  1. Diet: Foods rich in lactate, such as yogurt, kefir, and sourdough bread, can provide a source of energy for Veillonella bacteria.
  2. Exercise: Regular exercise has been shown to increase the levels of Veillonella bacteria in the gut microbiota, which may improve athletic performance.
  3. Probiotics and Prebiotics: Probiotic and prebiotic supplements can help to increase the levels of Veillonella bacteria in the gut microbiota. HMO prebiotic (2’-FL) is helpful to boost the level of Veillonella based on a study conducted on human and a pending-patent.

References:

  1. Belkaid, Y. & Harrison, O.J. (2017). Homeostatic Immunity and the Microbiota. Immunity, 46, 562-576.
  2. O'Hara, A. & Shanahan, F. (2006). The gut flora as a forgotten organ. EMBO reports, 7, 688-693.
  3. Tremaroli, V. & Bäckhed, F. (2012). Functional interactions between the gut microbiota and host metabolism. Nature, 489(7415), 242-249.
  4. Mayer, E.A., Knight, R., Mazmanian, S.K., Cryan, J.F., & Tillisch, K. (2014). Gut microbes and the brain: paradigm shift in neuroscience. Journal of Neuroscience, 34(46), 15490-15496.
  5. Fasano, A. (2012). Leaky gut and autoimmune diseases. Clinical reviews in allergy & immunology, 42, 71-78.
  6. Zmora, N., Zilberman-Schapira, G., & Segal, E. (2019). The gut microbiome and its potential therapeutic application. Nature Reviews Gastroenterology & Hepatology, 16, 332-343.
  7. Slavin, J. (2013). Fiber and prebiotics: mechanisms and health benefits. Nutrients, 5, 1417-1435.
  8. Marco, M. L., Heeney, D., Binda, S., Cifelli, C. J., Cotter, P. D., Foligné, B., ... & Ventura, M. (2021). Health benefits of fermented foods: microbiota and beyond. Current Opinion in Biotechnology, 70, 1-8.
  9. Buffie, C. G., & Pamer, E. G. (2013). Microbiota-mediated colonization resistance against intestinal pathogens. Nature Reviews Immunology, 13, 790-801.
  10. Kelly, J. R., Borre, Y., O'Brien, C., Patterson, E., El Aidy, S., Deane, J., ... & Dinan, T. G. (2015). Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. Journal of Psychiatric Research, 63, 9-17.
  11. Kaczmarczyk, M. M., Miller, M. J., & Freund, G. G. (2017). The health benefits of dietary fiber: beyond the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer. Metabolism, 68, 1-5.
  12. Rao, S. S. C., & Rehman, A. (2018). Effects of coffee and caffeine on constipation and bowel movements in healthy adults. Digestive Diseases and Sciences, 63, 315-321.
  13. Singh, R. K., Chang, H. W., Yan, D., Lee, K. M., Ucmak, D., Wong, K., ... & Liao, W. (2017). Influence of diet on the gut microbiome and implications for human health. Journal of Translational Medicine, 15, 73.
  14. Marco, M. L., Heeney, D., Binda, S., Cifelli, C. J., Cotter, P. D., Foligné, B., ... & Ventura, M. (2021). Health benefits of fermented foods: microbiota and beyond. Current Opinion in Biotechnology, 70, 1-8.
  15. Parnell, J. A., & Reimer, R. A. (2012). Prebiotic fiber modulation of the gut microbiota improves risk factors for obesity and the metabolic syndrome. Gut Microbes, 3, 29-34.
  16. Marco, M. L., Heeney, D., Binda, S., Cifelli, C. J., Cotter, P. D., Foligné, B., ... & Ventura, M. (2021). Health benefits of fermented foods: microbiota and beyond. Current Opinion in Biotechnology, 70, 1-8.
  17. Kim, E. K., An, S. Y., Lee, M. S., Kim, T. H., & Rhee, Y. (2018). Effects of probiotics on body weight, body mass index, fat mass and fat percentage in subjects with overweight or obesity: a systematic review and meta-analysis of randomized controlled trials. Nutrients, 10, 1-14.
  18. Kaczmarczyk, M. M., Miller, M. J., & Freund, G. G. (2017). The health benefits of dietary fiber: beyond the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer. Metabolism, 68, 1-5.
  19. Singh, R. K., Chang, H. W., Yan, D., Lee, K. M., Ucmak, D., Wong, K., ... & Liao, W. (2017). Influence of diet on the gut microbiome and implications for human health. Journal of Translational Medicine, 15, 73.
  20. Fuentes‐Zaragoza, Evangélica, et al. "Resistant starch as prebiotic: A review." Starch‐Stärke7 (2011): 406-415.
  21. Marco, M. L., Heeney, D., Binda, S., Cifelli, C. J., Cotter, P. D., Foligné, B., ... & Ventura, M. (2021). Health benefits of fermented foods: microbiota and beyond. Current Opinion in Biotechnology, 70, 1-8.
  22. Liu, Y., Wang, Y., Ni, Y., Cheung, C. K., Lam, K. S., & Wang, Y. (2017). Gut microbiome fermentation determines the efficacy of exercise for diabetes prevention. Cell Metabolism, 27, 391-404.
  23. Zhou, A. L., Hergert, N., Rompato, G., & Miller, M. J. (2017). Bone broth supplementation increases blood amino acid concentrations and inhibits markers of muscle damage during eccentric training. Journal of the International Society of Sports Nutrition, 14, 1-9.
  24. Zarena, A. S., & Sankar, K. U. (2018). Health benefits of soybean and its constituents: a review. Journal of Nutrition and Food Science, 8, 1-10.
  25. Petsiou, E. I., Mitrou, P. I., Raptis, S. A., & Dimitriadis, G. D. (2014). Effect and mechanisms of action of vinegar on glucose metabolism, lipid profile, and body weight. Nutrition Reviews, 72, 651-661.
  26. Dinan, T. G., & Cryan, J. F. (2017). Gut instincts: microbiota as a key regulator of brain development, ageing and neurodegeneration. Journal of Physiology, 595, 489-503.
  27. Mayer, E. A., Knight, R., Mazmanian, S. K., Cryan, J. F., & Tillisch, K. (2014). Gut microbes and the brain: paradigm shift in neuroscience. Journal of Neuroscience, 34(46), 15490-15496.
  28. Wallace, C. J., Milev, R., & Theoharides, T. C. (2017). Effects of probiotics on the symptoms of anxiety: A systematic review and meta-analysis. Journal of Psychiatric Research, 90, 1-13.
  29. Slykerman, R. F., Hood, F., Wickens, K., Thompson, J. M., Barthow, C., Murphy, R., ... & Crane, J. (2017). Effect of Lactobacillus rhamnosus HN001 in pregnancy on postpartum symptoms of depression and anxiety: a randomised double-blind placebo-controlled trial. EBioMedicine, 24, 159-165.
  30. Li, Y., Lv, M. R., Wei, Y. J., Sun, L., Zhang, J. X., Zhang, H. G., & Li, B. (2019). Dietary fiber intake and risk of depressive symptoms: a systematic review and meta-analysis of epidemiological studies. Nutrition Journal, 18, 1-13.
  31. Mayer, E. A., Knight, R., Mazmanian, S. K., Cryan, J. F., & Tillisch, K. (2014). Gut microbes and the brain: paradigm shift in neuroscience. Journal of Neuroscience, 34(46), 15490-15496.
  32. Foster, J. A., & McVey Neufeld, K. A. (2013). Gut-brain axis: how the microbiome influences anxiety and depression. Trends in Neurosciences, 36, 305-312.
  33. Yano, J. M., & Hsiao, E. Y. (2017). The gut-brain axis in health and disease. Annual Review of Pathology: Mechanisms of Disease, 12, 341-364.
  34. Dinan, T. G., & Cryan, J. F. (2017). Gut instincts: microbiota as a key regulator of brain development, ageing and neurodegeneration. Journal of Physiology, 595, 489-503.
  35. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012 Mar 16;148:1258-70.
  36. Belizário JE, Napolitano M. Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approaches. Front Microbiol. 2015;6:1050.
  37. Marchesi JR, Ravel J. The vocabulary of microbiome research: a proposal. Microbiome. 2015;3:31.
  38. Sanders ME, Merenstein DJ, Ouwehand AC, Reid G, Salminen S, Cabana MD, Paraskevakos G, Leyer G. Probiotic use in at-risk populations. J Am Pharm Assoc (2003). 2016;56:680-6.
  39. Cryan JF, O'Mahony SM. The microbiome-gut-brain axis: from bowel to behavior. Neurogastroenterol Motil. 2011 Mar;23:187-92.
  40. Tang WH, Kitai T, Hazen SL. Gut microbiota in cardiovascular health and disease. Circ Res. 2017 Apr 28;120:1183-1196. doi: 10.1161/CIRCRESAHA.117.309715. PMID: 28450350; PMCID: PMC5466933.
  41. Harvard Health Publishing. The gut-brain connection. Available at: https://www.health.harvard.edu/diseases-and-conditions/the-gut-brain-connection. Accessed May 3, 2023.
  42. Rinninella E, Cintoni M, Raoul P, et al. Food components and dietary habits: Keys for a healthy gut microbiota composition. Nutrients. 2019;11:2393. doi:10.3390/nu11102393.
  43. Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027-1031.
  44. Cani PD, Delzenne NM. The role of the gut microbiota in energy metabolism and metabolic disease. Curr Pharm Des. 2009;15:1546-1558.
  45. Cani PD, Dewever C, Delzenne NM. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. Br J Nutr. 2004;92:521-526.
  46. Everard A, Lazarevic V, Derrien M, et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes. 2011;60:2775-2786.
  47. Cani PD, Possemiers S, Van de Wiele T, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58:1091-1103.
  48. Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761-1772.
  49. Cani PD, Bibiloni R, Knauf C, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470-1481.
  50. Tillisch K, Labus J, Kilpatrick L, et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology. 2013;144:1394-1401.
  51. Zhao L. The gut microbiota and obesity: from correlation to causality. Nat Rev Microbiol. 2013;11:639-647.
  52. McFarland LV. Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol. 2010;16:2202-2222.
  53. Hempel S, Newberry SJ, Maher AR, et al. Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. JAMA. 2012;307:1959-1969.
  54. Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. 2017;8:172-184.
  55. Moayyedi P, Ford AC, Talley NJ, et al. The efficacy of probiotics in the treatment of irritable bowel syndrome: a systematic review. Gut. 2010;59:325-332.
  56. Cognitive-behavioral therapy (CBT): CBT is a form of psychotherapy that can help people with IBS learn coping strategies to manage symptoms and reduce psychological distress. It involves identifying negative thoughts and behaviors that may be contributing to symptoms and replacing them with more positive and adaptive ones .
  57. Hypnotherapy: Hypnotherapy has been shown to be effective in reducing symptoms of IBS, particularly in reducing abdominal pain and improving quality of life. It involves inducing a state of deep relaxation and focused attention to help alleviate psychological and physical symptoms .
  58. Herbal supplements: Some herbal supplements, such as peppermint oil and ginger, may be helpful in reducing symptoms of IBS. However, it's important to talk to your healthcare provider before taking any supplements, as they may interact with other medications or have potential side effects.
  59. Konturek PC, Brzozowski T, Konturek SJ. Stress and the gut: pathophysiology, clinical consequences, diagnostic approach and treatment options. J Physiol Pharmacol. 2011;62:591-599.
  60. Mayer EA. Gut feelings: the emerging biology of gut-brain communication. Nat Rev Neurosci. 2011;12:453-466.
  61. Gaylord SA, Palsson OS, Garland EL, et al. Mindfulness training reduces the severity of irritable bowel syndrome in women: results of a randomized controlled trial. Am J Gastroenterol. 2011;106:1678-1688.
  62. Monda V, Villano I, Messina A, et al. Exercise modifies the gut microbiota with positive health effects. Oxid Med Cell Longev. 2017;2017:3831972.
  63. Cramer H, Haller H, Lauche R, et al. A systematic review and meta-analysis of yoga for digestive disorders. Eur J Gastroenterol Hepatol. 2016;28:1161-1171.
  64. Weil A. The 4-7-8 breath: how it works, how to do it, and its benefits. Dr. Weil's Wellness Therapies. https://www.drweil.com/health-wellness/body-mind-s
  65. Salem I, Ramser A, Isham N, Ghannoum MA. The gut microbiome as a major regulator of the gut-skin axis. Front Microbiol. 2018;9:1459.
  66. Chien AL, Tsai J, Leung S, et al. Review of the role of the gut microbiome in the management of skin conditions in dermatology. J Am Acad Dermatol. 2019;80:930-938.
  67. Ganceviciene R, Liakou AI, Theodoridis A, et al. The role of the skin microbiome in acne pathogenesis. Clin Dermatol. 2018;36:136-142.
  68. Jager N, Szegedi A, Gonda A, et al. Altered gut microbiota in major depressive disorder: A systematic review and meta-analysis. J Affect Disord. 2021;281:887-899.
  69. Jafari M, Javadzadeh Y, Jamebozorgi N, et al. Sex hormones and immune dimorphism in aging and longevity. Expert Rev Clin Immunol. 2018;14:747-756.
  70. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396(10247):345-360.
  71. Arck P, Handjiski B, Hagen E, et al. Is there a 'gut-brain-skin axis'? Exp Dermatol. 2010;19:401-405.
  72. Jung GW, Tse JE, Guiha I, et al. Prospective, randomized, open-label trial comparing the safety, efficacy, and tolerability of an acne treatment regimen with and without a probiotic supplement and minocycline in subjects with mild to moderate acne. J Cutan Med Surg. 2013;17:114-122.
  73. Gao X, Liu X, Xu J, et al. Dietary fiber intake is positively associated with the risk of moderate-to-severe acne vulgaris in Chinese adolescents. J Am Acad Dermatol. 2020;82:415-422.
  74. Guarner F, Malagelada J. Gut flora in health and disease. Lancet. 2003;361(9356):512-519.
  75. Singh RK, Chang HW, Yan D, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. 2017;15:73.
  76. McBurney MI, Davis C, Fraser CM, et al. Establishing what constitutes a healthy human gut microbiome: state of the science, regulatory considerations, and future directions. J Nutr. 2019;149:1882-1895.
  77. Liu RT. The microbiome as a novel paradigm in studying stress and mental health. Am Psychol. 2017;72:655-667.
  78. Plaza-Diaz, J., et al. (2017). "Beneficial effects of probiotics on the maturation of immune response and intestinal permeability in preterm infants." Neonatology, 112, 237-244.
  79. Martín, R., et al. (2010). "Functional characterization of Bifidobacterium strains isolated from breast milk." Journal of Dairy Research, 77, 335-343.
  80. Kato, K., et al. (2014). "Oral administration of Bifidobacterium bifidum G9-1 suppresses total and antigen-specific immunoglobulin E production in mice." Biological and Pharmaceutical Bulletin, 37, 1610-1615.
  81. Li, M., et al. (2021). "Bifidobacterium adolescentis Exerts Strain-Specific Effects on Immune Responses in Mice." Frontiers in Nutrition, 8, 632335.
  82. Azad, M. B., et al. (2013). "Probiotic supplementation during pregnancy or infancy for the prevention of asthma and wheeze: systematic review and meta-analysis." BMJ, 347, f6471.
  83. Kalliomaki, M., et al. (2001). "Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial." Lancet, 357(9262), 1076-1079.
  84. Allen, A. P., et al. (2016). "Bifidobacterium longum 1714 as a translational psychobiotic: modulation of stress, electrophysiology and neurocognition in healthy volunteers." Translational Psychiatry, 6, e939.
  85. Schmidt, K., et al. (2015). "Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers." Psychopharmacology, 232, 1793-1801.
  86. Delzenne, N. M., et al. (2011). "Functional foods and gut microbiota." Current Opinion in Clinical Nutrition and Metabolic Care, 14, 4.
  87. O’Keefe, S. J. D., et al. (2015). "Fat, fibre and cancer
  88. Lee YK, Salminen S. Handbook of probiotics and prebiotics. John Wiley & Sons; 2009.
  89. Kechagia M, Basoulis D, Konstantopoulou S, et al. Health benefits of probiotics: a review. ISRN Nutr. 2013;2013:481651.
  90. Ouwehand AC, Salminen S, Isolauri E. Probiotics: an overview of beneficial effects. Antonie Van Leeuwenhoek. 2002;82(1-4):279-289.
  91. Sanders ME. Probiotics: definition, sources, selection, and uses. Clin Infect Dis. 2008;46 Suppl 2:S58-61; discussion S144-151.
  92. Wadhwani N, Shedbalkar UU, Nair R, et al. Biogenic synthesis of antibacterial silver chloride nanoparticles using culture supernatant of Lactobacillus fermentum. Microb Pathog. 2017;104:365-370.
  93. Bruce-Keller AJ, Salbaum JM, Luo M, Blanchard E, Taylor CM, Welsh DA, et al. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol Psychiatry. 2015;77:607-15.
  94. Miquel, S., Martín, R., Rossi, O., Bermúdez-Humarán, L. G., Chatel, J. M., Sokol, H., ... & Langella, P. (2013). Faecalibacterium prausnitzii and human intestinal health. Current opinion in microbiology, 16, 255-261.
  95. Balamurugan, R., George, G., Kabeerdoss, J., Hepsiba, J., Chandragunasekaran, A. M., & Ramakrishna, B. S. (2010). Quantitative differences in intestinal Faecalibacterium prausnitzii in obese Indian children. British Journal of Nutrition, 103, 335-338.
  96. Quévrain, E., Maubert, M. A., Michon, C., Chain, F., Marquant, R., Tailhades, J., ... & Langella, P. (2015). Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn's disease. Gut, 65, 415-425.
  97. Sánchez, B., González-Rodríguez, I., Arboleya, S., López, P., Suárez, A., & de los Reyes-Gavilán, C. G. (2017). Probiotic properties of 20 common foodborne isolates of faecalibacterium androsii. International journal of food microbiology, 241, 217-225.
  98. Khan, M. T., Duncan, S. H., Stams, A. J., van Dijl, J. M., Flint, H. J., & Harmsen, H. J. (2012). The gut anaerobe Faecalibacterium prausnitzii uses an extracellular electron shuttle to grow at oxic–anoxic interphases. The ISME journal, 6, 1578-1585.
  99. Belenguer A, Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, Flint HJ. Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol. 2006 Dec;72:3593-9. doi: 10.1128/AEM.72.5.3593-3599.2006. PMID: 16751530; PMCID: PMC1489366.
  100. Lopez-Siles M, Enrich-Capó N, Aldeguer X, Sabat-Mir M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Alterations in the Abundance and Co-occurrence of Akkermansia muciniphila and Faecalibacterium prausnitzii in the Colonic Mucosa of Inflammatory Bowel Disease Subjects. Front Cell Infect Microbiol. 2018 Mar 13;8:281. doi: 10.3389/fcimb.2018.00281. PMID: 29594161; PMCID: PMC5853734.
  101. Louis P, Flint HJ. Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol. 2017 Jan;19:29-41. doi: 10.1111/1462-2920.13589. Epub 2016 Dec 20. PMID: 27813210.
  102. Vital M, Howe AC, Tiedje JM. Revealing the bacterial butyrate synthesis pathways by analyzing (meta)genomic data. MBio. 2014 Dec 9;5:e00889. doi: 10.1128/mBio.00889-14. PMID: 25491348; PMCID: PMC4278896.
  103. Rossi M, Amaretti A, Raimondi S. Folate Production by Probiotic Bacteria. Nutrients. 2011;3:118-134. doi:10.3390/nu3010118
  104. Drouault-Holowacz S, Bieuvelet S, Burckel A, et al. Streptococcus thermophilus fermentation of milk increases the levels of the neuroactive amino acids tyrosine, phenylethylamine, and tryptophan. Int J Food Microbiol. 2007;113:28-34. doi:10.1016/j.ijfoodmicro.2006.06.017
  105. Smug LN, Salminen S, Sanders ME, et al. The functional food concept and probiotics. Curr Opin Biotechnol. 2011;22:184-190. doi:10.1016/j.copbio.2010.10.014
  106. EFSA Panel on Biological Hazards (BIOHAZ). Scientific Opinion on the maintenance of the list of QPS biological agents intentionally added to food and feed (2013 update). EFSA Journal. 2013;11:3449. doi:10.2903/j.efsa.2013.3449.
  107. Kaper JB, Nataro JP, Mobley HL. Pathogenic Escherichia coli. Nature Reviews Microbiology. 2004;2:123-140.
  108. O'Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO Reports. 2006;7:688-693.
  109. Chassaing B, Gewirtz AT. Gut microbiota, low-grade inflammation, and metabolic syndrome. Toxicologic Pathology. 2014;42:49-53.
  110. Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59-65.
  111. Patel RM, Denning PW. Therapeutic use of prebiotics, probiotics, and postbiotics to prevent necrotizing enterocolitis: what is the current evidence?. Clinics in Perinatology. 2013;40:11-25.
  112. Alberda C, Gramlich L, Meddings J, et al. Effects of probiotic therapy in critically ill patients: a randomized, double-blind, placebo-controlled trial. American Journal of Clinical Nutrition. 2007;85:816-823.
  113. Wu, H. J., & Wu, E. (2012). The role of gut microbiota in immune homeostasis and autoimmunity. Gut microbes, 3, 4-14.
  114. Tursi, S. A., Tükel, Ç., & Raffatellu, M. (2017). The gut microbiome in infectious diarrhea and inflammation. Current opinion in gastroenterology, 33, 471-477.
  115. He, Y., Wu, W., Wu, S., Zheng, H. M., Li, P., Sheng, H. F., ... & Zhou, H. W. (2018). Linking gut microbiota, metabolic syndrome and economic status based on a population-level analysis. Microbiome, 6, 172.
  116. Round, J. L., & Mazmanian, S. K. (2009). The gut microbiota shapes intestinal immune responses during health and disease. Nature reviews immunology, 9, 313-323.
  117. Gevers D, Kugathasan S, Denson LA, et al. The Treatment-Naive Microbiome in New-Onset Crohn's Disease. Cell Host Microbe. 2014;15:382-392. doi:10.1016/j.chom.2014.02.005
  118. Wexler HM. Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev. 2007;20:593
  119. Wang W, Lv H, Zhang J, et al. The safety evaluation of a new strain of Clostridium butyricum. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association. 2013;57:1-6. doi:10.1016/j.fct.2013.02.014
  120. Hwang NR, Yim SH, Kim YM, et al. Transcriptome-Based Identification of the Gene(s) Responsible for the High-Quality Meat Phenotype of the Korean Native Black Pig, Jeju Black Pig. PloS one. 2015;10:e0133432. doi:10.1371/journal.pone.0133432
  121. Liang X, Li Y, Zhang W, et al. Clostridium butyricum alleviates experimental colitis by suppressing IFN-γ-induced predominant lipid metabolism. Journal of gastroenterology and hepatology. 2020;35:477-488. doi:10.1111/jgh.14847
  122. Xiao H, Ge C, Feng G, et al. Improvement of growth performance and gut microbiota by supplementing piglets with Clostridium butyricum. PloS one. 2019;14:e0215680. doi:10.1371/journal.pone.0215680
  123. Zhuo Z, Zhang L, Fan D, et al. Clostridium butyricum regulates intestinal barrier function in experimental colitis via the IL-10/STAT3 signaling pathway. International immunopharmacology. 2020;78:105974. doi:10.1016/j.intimp.2019.105974
  124. Flint HJ, Duncan SH, Scott KP, Louis P. Interactions and competition within the microbial community of the human colon: links between diet and health. Environ Microbiol. 2007 Nov;9:1101-11. doi: 10.1111/j.1462-2920.2007.01281.x. PMID: 17956537.
  125. Lopez-Siles M, Khan TM, Duncan SH, Harmsen HJM, Garcia-Gil LJ, Flint HJ. Cultured representatives of two major phylogroups of human colonic Faecalibacterium prausnitzii can utilize pectin, uronic acids, and host-derived substrates for growth. Appl Environ Microbiol. 2012 Mar;78:420-8. doi: 10.1128/AEM.06858-11. Epub 2011 Dec
  126. Ouwehand AC, Salminen S, Isolauri E. Probiotics: an overview of beneficial effects. Antonie Van Leeuwenhoek. 2002 Nov;82(1-4):279-89.
  127. Elmer GW, Surawicz CM, McFarland LV. Biotherapeutic agents. A neglected modality for the treatment and prevention of selected intestinal and vaginal infections. JAMA. 1996 Dec 25;276(24):1975-80.
  128. Lebeer S, Vanderleyden J, De Keersmaecker SC. Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol. 2010 Apr;8:171-84.
  129. Boyle RJ, Robins-Browne RM, Tang ML. Probiotic use in clinical practice: what are the risks? Am J Clin Nutr. 2006 Jun;83:1256-64.
  130. Yoon HS, Ju JH, Kim HN, Park HJ, Ji Y, Park KY. Effects of Lactobacillus acidophilus on gut microbiota composition in broiler chickens challenged with Clostr
  131. Plaza-Díaz, J., Ruiz-Ojeda, F. J., Vilchez-Padial, L. M., Gil, A., & Perez-Navero, J. L. (2018). Evidence of the Anti-Inflammatory Effects of Probiotics and Synbiotics in Intestinal Chronic Diseases. Nutrients, 10, 1539. doi: 10.3390/nu10101539
  132. Burton, J. P., Westerik, N., Harris, C. L., van den Beld, M. J. C., Christensen, E. G., & Schuren, F. H. J. (2017). Probiotic applications: 7. Improving oral health. Research in Microbiology, 168(9-10), 934-950. doi: 10.1016/j.resmic.2017.04.001
  133. Vandenplas, Y., Abkari, A., Bellaiche, M., Benninga, M., Chouraqui, J.-P., Çokura, F., . . . Szajewska, H. (2015). Prebiotics, probiotics, and synbiotics: Impact on the gut immune system and allergic reactions. Journal of Clinical Gastroenterology, 49(Suppl 1), S20-S26. doi: 10.1097/MCG.0000000000000428
  134. Jones ML, Martoni CJ, Prakash S. Cholesterol lowering and inhibition of sterol absorption by Lactobacillus reuteri NCIMB 30242: a randomized controlled trial. Eur J Clin Nutr. 2012;66:1234-1241. doi:10.1038/ejcn.2012.126
  135. Ohlsson C, Englund-Ögge L, Näsman P, et al. Lactobacillus reuteri for the prevention of colic in infants: a randomized controlled trial. Pediatrics. 2020;145:e20191235. doi:10.1542/peds.2019-1235
  136. Di Marzio L, Cinque B, Cuppone R, et al. Effects of probiotic bacteria (Lactobacillus reuteri) on immune system responses and intestinal flora in middle-aged rats. J Microbiol Immunol Infect. 2010;43:351-359. doi:10.1016/j.jmii.2009.10.009
  137. Rosander A, Connolly E, Roos S. Removal of antibiotic resistance gene-carrying plasmids from Lactobacillus reuteri ATCC 55730 and characterization of the resulting daughter strain, L. reuteri DSM 17938. Appl Environ Microbiol. 2008;74:6032-6040. doi:10.1128/AEM.00488-08
  138. Scholz-Ahrens KE, Ade P, Marten B, et al. Prebiotics, probiotics, and synbiotics affect mineral absorption, bone mineral content, and bone structure. J Nutr. 2007;137(3 Suppl 2):838S-846S. doi:10.1093/jn/137.3.838S
  139. Wescombe PA, Heng NC, Burton JP, et al. The diversity of bacteriocins in Gram-positive bacteria. In: Riley MA, Chavan MA, eds. Bacteriocins: Ecology and Evolution. Springer International Publishing; 2016:45-92. doi:10.1007/978-3-319-33449-4_3
  140. Tagg JR, Dierksen KP. Bacterial replacement therapy: adapting 'germ warfare' to infection prevention. Trends Biotechnol. 2003;21:217-223. doi:10.1016/S0167-7799(03)00084-6
  141. Kumar PS, Mason MR, Brooker MR, O'Brien K. Pyrosequencing reveals unique microbial signatures associated with healthy and failing dental implants. J Clin Periodontol. 2012;39:425-433. doi:10.1111/j.1600-051X.2012.01852.x
  142. Burton JP, Wescombe PA, Cadieux PA, et al. Beneficial microbes for oral health: time to harness the oral streptococci? Benef Microbes. 2013;4:89-97. doi:10.3920/BM2012.0047
  143. Loesche WJ. Role of Streptococcus mutans in human dental decay. Microbiol Rev. 1986;50:353-380. doi:10.1128/MMBR.50.4.353-380.1986
  144. Marsh PD. Dental plaque as a biofilm and a microbial community - implications for health and disease. BMC Oral Health. 2006;6 Suppl 1(Suppl 1):S14. doi:10.1186/1472-6831-6-S1-S14
  145. Featherstone JD. The caries balance: contributing factors and early detection. J Calif Dent Assoc. 2003;31:129-133.
  146. Sánchez GA, Miozza VA, Delgado A, Busch L, Salanitro JP, Barrio DA. The role of probiotics in the prevention of oral diseases. Microorganisms. 2021;9:772. doi:10.3390/microorganisms9040772
  147. Bowen, W. H., & Koo, H. (2011). Biology of Streptococcus mutans-derived glucosyltransferases: role in extracellular matrix formation of cariogenic biofilms. Caries research, 45, 69-86.
  148. Marsh, P. D. (2003). Dental plaque as a biofilm and a microbial community-implications for treatment. Journal of oral biosciences, 45, 19-26.
  149. Featherstone, J. D. (2009). Dental caries: a dynamic disease process. Australian dental journal, 54(1_suppl), S10-S17.
  150. Bradshaw, D. J., & Marsh, P. D. (1998). Analysis of pH-driven disruption of oral microbial communities in vitro. Caries research, 32, 456-462.
  151. Simón-Soro, Á., & Mira, A. (2015). Solving the etiology of dental caries. Trends in microbiology, 23, 76-82.
  152. Marsh, P. D. (2006). Dental plaque as a microbial biofilm. Caries research, 38, 204-211.
  153. Lee, S. H., & Kim, Y. J. (2017). The role of Streptococcus mutans in the formation of dental caries. Journal of dental hygiene science, 17, 228-235.
  154. Cardoso, F. F., Rocha, M. C., & Nascimento, M. M. (2014). Microbiological and immunological aspects of dental caries. Brazilian oral research, 28(spe), 94-101.
  155. Dige, I., Raarup, M. K., Nyengaard, J. R., Kilian, M., & Nyvad, B. (2019). Actinomyces and S. mutans biofilm formation in children with oral malformations. Scientific reports, 9, 1-12.
  156. McFarland LV. Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol. 2010;16:2202-2222. doi: 10.3748/wjg.v16.i18.2202
  157. Dinleyici EC, Eren M, Ozen M, Yargic ZA, Vandenplas Y. Effectiveness and safety of Saccharomyces boulardii for acute infectious diarrhea. Expert Opin Biol Ther. 2012;12:395-410. doi: 10.1517/14712598.2012.668013
  158. McFarland LV. Evidence-based review of probiotics for antibiotic-associated diarrhea and Clostridium difficile infections. Anaerobe. 2009;15:274-280. doi: 10.1016/j.anaerobe.2009.09.002
  159. McFarland LV. Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol. 2010;16:2202-2222. doi: 10.3748/wjg.v16.i18.2202
  160. Kabbani TA, Pallav K, Dowd SE, et al. Prospective randomized controlled study on the effects of Saccharomyces boulardii CNCM I-745 and amoxicillin-clavulanate or the combination on the gut microbiota of healthy volunteers. Gut Microbes. 2017;8:17-32. doi: 10.1080/19490976.2016.1267896
  161. Dinleyici EC, Eren M, Ozen M, Yargic ZA, Vandenplas Y. Saccharomyces boulardii for prevention of necrotizing enterocolitis in preterm infants: a randomized, controlled study. Acta Paediatr. 2013;102:e560-e565. doi: 10.1111/apa.12414
  162. McFarland LV. Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol. 2010;16:2202-2222. doi: 10.3748/wjg.v16.i18.2202
  163. Ihekweazu FD, Versalovic J. Development of the Pediatric Gut Microbiome: Impact on Health and Disease. Am J Med Sci. 2018;356:413-423. doi: 10.1016/j.amjms.2018.06.011
  164. Wohlgemuth S, Haller D, Blaut M, Loh G. Reduced microbial diversity and high numbers of one single Escherichia coli strain in the intestine of colitic mice. Environ Microbiol. 2009;11:1562-1571. doi: 10.1111/j.1462-2920.2009.01893.x
  165. Zhou X, Brown CJ, Abdo Z, Davis CC, Hansmann MA, Joyce P, Foster JA, Forney LJ. Differences in the composition of vaginal microbial communities found in healthy Caucasian and black women. ISME J. 2007 Aug;1:121-33. doi: 10.1038/ismej.2007.12. PMID: 18043614.
  166. Franz, C. M. A. P., Muscholl-Silberhorn, A., Yousif, N. M. K., Vancanneyt, M., Swings, J., & Holzapfel, W. H. (2001). Incidence of virulence factors and antibiotic resistance among enterococci isolated from food. Applied and environmental microbiology, 67, 4385-4389. https://doi.org/10.1128/AEM.67.9.4385-4389.2001
  167. Kühn, I., Iversen, A., Burman, L. G., Olsson-Liljequist, B., Franklin, A., Finn, M., & Aarestrup, F. M. (2000). Epidemiology and ecology of enterococci, with special emphasis on antibiotic resistant strains, in animals, humans and the environment—Example of an ongoing project within the European research programme. International journal of antimicrobial agents, 14, 337-342. https://doi.org/10.1016/S0924-8579(00)00145-1
  168. Eaton, T. J., Gasson, M. J., & Molecular Biology of Gram-positive Bacteria Symposium. (2001). Molecular screening of Enterococcus virulence determinants and potential for genetic exchange between food and medical isolates. Journal of food protection, 64, 849-859. https://doi.org/10.4315/0362-028X-64.6.849
  169. Pessione, E., & Cirrincione, S. (2016). Bioactive molecules released in food by lactic acid bacteria: encrypted peptides and biogenic amines. Frontiers in microbiology, 7, 876. https://doi.org/10.3389/fmicb.2016.00876
  170. Chajęcka-Wierzchowska, W., Zadernowska, A., Nalepa, B., Sierpińska, M., & Laniewska-Trokenheim, Ł. (2012). Enterococcus spp.–from commensals to leading causes of drug resistant infections–are they still in the shadow of staphylococci?. Journal of microbiology and biotechnology, 22, 677-690. https://doi.org/10.4014/jmb.1110.10005
  171. Lebreton, F., Willems, R. J. L., & Gilmore, M. S. (2014). Enterococcus diversity, origins in nature, and gut colonization.
  172. Franz, C. M. A. P., Holzapfel, W. H., & Stiles, M. E. (Eds.). (2014). Enterococci in Foods. Springer.
  173. Lee, S., Lee, H., Lee, S., & Kim, J. (2019). Enterococcus durans HDL-2 inhibits the epithelial-to-mesenchymal transition via modulation of Snail expression and suppression of TGF-β1 signaling pathway. Journal of Functional Foods, 57, 1-8.
  174. Liu, Y., & Li, J. (2021). Probiotic Enterococcus durans NCIMB 30168 modulates gut microbiota and intestinal inflammation in weaned piglets. BMC Microbiology, 21, 1-11.
  175. Mundy, L. M., Sahm, D. F., & Gilmore, M. S. (2000). Relationship between Enterococcal Virulence and Antibiotic Resistance. Clinical Microbiology Reviews, 13, 513–522.
  176. Zommiti, M., Feuilloley, M. G. J., & Connil, N. (2019). Enterococcus durans: Biological and technological aspects. Journal of
  177. Belenguer A, Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, Flint HJ. Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol. 2006 May;72:3593-9. doi: 10.1128/AEM.72.5.3593-3599.2006. PMID: 16672523; PMCID: PMC1472386.
  178. Cao Y, Shen J, Ran ZH. Association between Faecalibacterium prausnitzii reduction and inflammatory bowel disease: a meta-analysis and systematic review of the literature. Gastroenterol Res Pract. 2014;2014:872725. doi: 10.1155/2014/872725. Epub 2014 Aug 28. PMID: 25247191; PMCID: PMC4167942.
  179. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012 Jun 8;336(6086):1268-73. doi: 10.1126/science.1223490. PMID: 22674334; PMCID: PMC3601187.
  180. Martens EC, Lowe EC, Chiang H, Pudlo NA, Wu M, McNulty
  181. Machiels, K., Joossens, M., Sabino, J., De Preter, V., Arijs, I., Eeckhaut, V., ... & Vermeire, S. (2014). A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut, 63, 1275-1283.
  182. Wu, H. J., Wu, E., & Chen, L. (2017). Apigenin attenuates colonic inflammation and tumorigenesis by inhibiting the IRAK1/TAK1/NF-κB signaling pathway in IBD. Molecular carcinogenesis, 56, 197-207.
  183. Duboc, H., & Rainteau, D. (2014). Role of the gut microbiota in the pathogenesis of inflammatory bowel disease. World journal of gastroenterology, 20(44), 17293-17308.
  184. Zhang, Y., Limaye, P. B., Renaud, H. J., Klaassen, C. D., & Desai, D. H. (2017). Ontogeny of hepatic energy metabolism genes in mice as revealed by RNA-sequencing. PloS one, 12, e0174541.
  185. Centers for Disease Control and Prevention. (2019). Clostridioides difficile (C. diff). Retrieved from https://www.cdc.gov/cdiff/index.html
  186. Leffler, D. A., & Lamont, J. T. (2015). Clostridium difficile infection. New England Journal of Medicine, 372, 1539-1548. doi: 10.1056/NEJMra1403772
  187. Surawicz, C. M. (2013). Clostridium difficile infection: Risk factors, diagnosis and management. Nature Reviews Gastroenterology & Hepatology, 11, 504-515. doi: 10.1038/nrgastro.2013.105
  188. Kelly, C. R., Kahn, S., Kashyap, P., Laine, L., Rubin, D., Atreja, A., . . . Fischer, M. (2015). Update on Fecal Microbiota Transplantation 2015: Indications, Methodologies, Mechanisms, and Outlook. Gastroenterology, 149, 223-237.e9. doi:10.1053/j.gastro.2015.05
  189. National Institute of Diabetes and Digestive and Kidney Diseases. (2017). Definition and Facts for Constipation. Retrieved from https://www.niddk.nih.gov/health-information/digestive-diseases/constipation/definition-facts
  190. Mayo Clinic. (2019). Constipation. Retrieved from https://www.mayoclinic.org/diseases-conditions/constipation/symptoms-causes/syc-20354253
  191. Cleveland Clinic. (2018). Constipation. Retrieved from https://my.clevelandclinic.org/health/diseases/4059-constipation
  192. Harvard Health Publishing. (2018). The gut-brain connection. Retrieved from https://www.health.harvard.edu/diseases-and-conditions/the-gut-brain-connection
  193. Wald, A. (2019). Constipation. New England Journal of Medicine, 380, 1281-1288. doi: 10.1056/NEJMcp1809649
  194. Hoda, K. M., & Jafri, W. (2021). Constipation. In StatPearls [Internet]. StatPearls Publishing. Available from: https://www.ncbi.nlm.nih.gov/books/NBK459312/
  195. Rao, S. S. C., Rattanakovit, K., & Patcharatrakul, T. (2016). Diagnosis and management of chronic constipation in adults. Nature Reviews Gastroenterology & Hepatology, 13, 295-305. doi: 10.1038/nrgastro.2016.53
  196. Bharucha, A. E., Pemberton, J. H., & Locke III, G. R. (2013). American Gastroenterological Association technical review on constipation. Gastroenterology, 144, 218-238. doi: 10.1053/j.gastro.2012.10.028
  197. Parthasarathy G, Chen J, Chen X, Chia N, O'Connor HM, Gaskins HR, Bharucha AE. Relationship Between Microbiota of the Colonic Mucosa vs Feces and Symptoms, Colonic Transit, and Methane Production in Female Patients With Chronic Constipation. Gastroenterology. 2016 Mar;150:367-79.e1. doi: 10.1053/j.gastro.2015.10.005. Epub 2015 Oct 17. PMID: 26477914; PMCID: PMC4763108.
  198. Zhong W, Lu X, Shi H, Zhao C, Zhang Q, Liang S, Xia Y, Wang Y, Zhang W, Wang C. Distinct microbial populations exist in the mucosa-associated microbiota of diarrhea predominant irritable bowel syndrome and ulcerative colitis. J Clin Gastroenterol. 2017 Apr;51:312-321. doi: 10.1097/MCG.0000000000000635. PMID: 27607443.
  199. Vandeputte D, Falony G, Vieira-Silva S, Tito RY, Joossens M, Raes J. Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut. 2016 Mar;65:57-62. doi: 10.1136/gutjnl-2015-309618. Epub 2015 Aug 12. PMID: 26265223.
  200. Kim M, Kim CH, Hong SN, Ahn YT. Independent and Interdependent Effects of the Intestinal Microbiota and SC
  201. Brown, K., DeCoffe, D., Molcan, E., & Gibson, D. L. (2012). Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients, 4, 1095-1119.
  202. Ye, Y., Wang, R., Feng, J., & Cui, H. (2021). Dietary fiber-mediated gut barrier function. International journal of molecular sciences, 22, 1055. doi: 10.3390/ijms22031055
  203. König, J., Wells, J., Cani, P. D., García-Ródenas, C. L., MacDonald, T., Mercenier, A., ... & Wolvers, D. (2016). Human intestinal barrier function in health and disease. Clinical and translational gastroenterology, 7, e196. doi: 10.1038/ctg.2016.54
  204. Bischoff, S. C., Barbara, G., Buurman, W., Ockhuizen, T., Schulzke, J. D., Serino, M., ... & Wells, J. M. (2014). Intestinal permeability–a new target for disease prevention and therapy. BMC gastroenterology, 14, 189. doi: 10.1186/s12876-014-0189-7
  205. Leclercq, S., Matamoros, S., Cani, P. D., Neyrinck, A. M., Jamar, F., Stärkel, P., ... & Delzenne, N. M. (2014). Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proceedings of the National Academy of Sciences, 111(42), E4485-E4493. doi: 10.1073/pnas.1415174111
  206. Wang, Y., Gao, X., Gao, R., Zhu, C., Li, L., Zhang, H., ... & Ji, K. (2020). The effect of fermented milk prepared with Lactobacillus fermentum on intestinal barrier function in a rat model of irritable bowel syndrome. Journal of Dairy Science, 103, 1492-1502. doi: 10.3168/jds.2019-17032
  207. Shah, E., Pimentel, M. & Chey, W. D. (2014). Irritable bowel syndrome: update on epidemiology, diagnosis, and treatment. American Journal of Gastroenterology, 109, 537-548.
  208. Vojdani, A. (2013). The effects of gluten on the gut microbiome and immunity. Integrative Medicine, 12, 26-33.
  209. Mayo Clinic. (2021). Lactose intolerance. Retrieved from https://www.mayoclinic.org/diseases-conditions/lactose-intolerance/symptoms-causes/syc-20374232
  210. Wölnerhanssen, B. K., & Meyer-Gerspach, A. C. (2019). Dietary factors in irritable bowel syndrome. Current Opinion in Gastroenterology, 35, 98-104.
  211. National Institute of Diabetes and Digestive and Kidney Diseases. (2018). Treatment for heartburn and gastroesophageal reflux disease (GERD). Retrieved from https://www.niddk.nih.gov/health-information/digestive-diseases/acid-reflux-ger-gerd-adults/treatment
  212. Wu, G. D., Compher, C., Chen, E. Z., Smith, S. A., Shah, R. D., Bittinger, K., ... & Lewis, J. D. (2014). Comparative metabolomics in veg
  213. Elli L et al. Diagnosis of gluten related disorders: Celiac disease, wheat allergy and non-celiac gluten sensitivity. World J Gastroenterol. 2015 Sep 21;21(35):10523-31.
  214. Misselwitz B et al. Lactose malabsorption and intolerance: pathogenesis, diagnosis and treatment. United European Gastroenterol J. 2013 Oct;1:151-9.
  215. Simrén M et al. Food-related gastrointestinal symptoms in the irritable bowel syndrome. Digestion. 2001;63:108-15.
  216. DiBaise JK et al. Diet and Irritable Bowel Syndrome. Am J Gastroenterol Suppl. 2012 Mar;1:25-31.
  217. Halmos EP et al. The FODMAP diet: FODMAPs, food, and functional gastrointestinal disorders. J Gastroenterol Hepatol. 2014 Feb;29 Suppl 1:136-46.
  218. Lu CL et al. Effects of alcohol on gastric emptying and gastrointestinal transit. Alcohol Clin Exp Res. 2010 Jan;34:162-8.
  219. Boekema, P. J., Samsom, M., & van Berge Henegouwen, G. P. (1999). Coffee and gastrointestinal function: facts and fiction. A review. Scandinavian Journal of Gastroenterology. Supplement, 230, 35-39. doi: 10.1080/003655299750025487
  220. Haruma, K., & Kamada, T. (2015). Review article: epidemiology and management of constipation in the Asia-Pacific region. Journal of Gastroenterology and Hepatology, 30, 109-114. doi: 10.1111/jgh.12695
  221. Rao, S. S., & Yu, S. (2014). Fedorak RN. Systematic review: dietary fibre and FODMAP-restricted diet in the management of constipation and irritable bowel syndrome. Alimentary Pharmacology & Therapeutics, 41, 1256-1270. doi: 10.1111/apt.12710
  222. Singh, R. K., Chang, H. W., Yan, D., Lee, K. M., Ucmak, D., Wong, K., … & Liao, W. (2017). Influence of diet on the gut microbiome and implications for human health. Journal of Translational Medicine, 15, 73. doi: 10.1186/s12967-017-1175-y
  223. Rhee, S. (2018). Effects of diet on gut microbiota: a review of the literature. Journal of Gastroenterology and Hepatology Research, 7, 1731-1738.
  224. Pizzorno, J. (Ed.). (2017). Textbook of natural medicine. Elsevier.
  225. Krajmalnik-Brown, R., Ilhan, Z. E., Kang, D. W., & DiBaise, J. K. (2012). Effects of gut microbes on nutrient absorption and energy regulation. Nutrition in Clinical Practice, 27, 201-214.
  226. Scheiman, J. et al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat. Med. 25, 1104–1109 (2019).
  227. Suez, J. et al. Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT. Cell 174, 1406-1423.e16 (2018).
  228. Rivera-Chávez, F. et al. Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella. Cell Host & Microbe 19, 443–454 (2016). 

Edited and Reviewed by: Layer Origin Content Team


Leave a comment


Also in GUT HEALTH KNOWLEDGE CENTER

How Does Akkermansia Interact with Your Body, Dietary Fibers, and Its Microbial Allies to Benefit Your Health
How Does Akkermansia Interact with Your Body, Dietary Fibers, and Its Microbial Allies to Benefit Your Health

April 28, 2024 8 min read

In the vast ecosystem of the human gut, Akkermansia muciniphila is emerging as a pivotal player and taking center stage as a next-generation probiotic. It's unique interactions with the human gut not only bolster its own population but also promote synergies with other beneficial gut bacteria, leading to a more resilient and healthy microbiome. Understanding the intricate and complex interplay between dietary fibers, host-derived substances, other probiotic bacteria and Akkermansia muciniphila, offers new opportunities in nurturing the human gut and optimizing wellbeing.
Read More
What Gut Bacteria Reduce Your Risk of Heart Disease, and How? Discoveries from a Study Involving 1400 People on Oscillibacter and Cholesterol
What Gut Bacteria Reduce Your Risk of Heart Disease, and How? Discoveries from a Study Involving 1400 People on Oscillibacter and Cholesterol

April 21, 2024 8 min read

Scientists have recently found that certain gut microbes may have protective benefits against cardiovascular disease. One particular genus, Oscillibacter, is equipped with the tools needed to metabolize cholesterol in its surrounding environment, helping to rid it from the body.
Read More
8 Ways To Modulate Your Gut For Better Metabolism and Weight Management Cover Image
8 Ways To Modulate Your Gut For Better Metabolism and Weight Management

April 14, 2024 7 min read

Discover eight effective ways to fine-tune your gut health for enhanced metabolism and weight management in our latest blog. From incorporating prebiotics and probiotics to exploring exercise, learn how simple dietary and lifestyle changes can lead to significant health benefits.
Read More