fbpx
Wikipedia

Gut microbiota

Gut microbiota, gut microbiome, or gut flora, are the microorganisms, including bacteria, archaea, fungi, and viruses, that live in the digestive tracts of animals.[1][2] The gastrointestinal metagenome is the aggregate of all the genomes of the gut microbiota.[3][4] The gut is the main location of the human microbiome.[5] The gut microbiota has broad impacts, including effects on colonization, resistance to pathogens, maintaining the intestinal epithelium, metabolizing dietary and pharmaceutical compounds, controlling immune function, and even behavior through the gut–brain axis.

Escherichia coli, one of the many species of bacteria present in the human gut

The microbial composition of the gut microbiota varies across regions of the digestive tract. The colon contains the highest microbial density of any human-associated microbial community studied so far, representing between 300 and 1000 different species.[6] Bacteria are the largest and to date, best studied component and 99% of gut bacteria come from about 30 or 40 species.[7] Up to 60% of the dry mass of feces is bacteria.[8] Over 99% of the bacteria in the gut are anaerobes, but in the cecum, aerobic bacteria reach high densities.[5] It is estimated that the human gut microbiota have around a hundred times as many genes as there are in the human genome.

Overview edit

 
Composition and distribution of gut microbiota in human body.

In humans, the gut microbiota has the largest numbers and species of bacteria compared to other areas of the body.[9] The approximate number of bacteria composing the gut microbiota is about 1013-1014.[10] In humans, the gut flora is established at one to two years after birth, by which time the intestinal epithelium and the intestinal mucosal barrier that it secretes have co-developed in a way that is tolerant to, and even supportive of, the gut flora and that also provides a barrier to pathogenic organisms.[11][12]

The relationship between some gut microbiota and humans is not merely commensal (a non-harmful coexistence), but rather a mutualistic relationship.[5]: 700  Some human gut microorganisms benefit the host by fermenting dietary fiber into short-chain fatty acids (SCFAs), such as acetic acid and butyric acid, which are then absorbed by the host.[9][13] Intestinal bacteria also play a role in synthesizing vitamin B and vitamin K as well as metabolizing bile acids, sterols, and xenobiotics.[5][13] The systemic importance of the SCFAs and other compounds they produce are like hormones and the gut flora itself appears to function like an endocrine organ.[13] Dysregulation of the gut flora has been correlated with a host of inflammatory and autoimmune conditions.[9][14]

The composition of human gut microbiota changes over time, when the diet changes, and as overall health changes.[9][14] A systematic review from 2016 examined the preclinical and small human trials that have been conducted with certain commercially available strains of probiotic bacteria and identified those that had the most potential to be useful for certain central nervous system disorders.[15]

Classifications edit

The microbial composition of the gut microbiota varies across the digestive tract. In the stomach and small intestine, relatively few species of bacteria are generally present.[6][16] The colon, in contrast, contains the highest microbial density of any human-associated microbial community studied so far[17] with between 1010 and 1011 cells per gram of intestinal content.[18] These bacteria represent between 300 and 1000 different species.[6][16] However, 99% of the bacteria come from about 30 or 40 species.[7] As a consequence of their abundance in the intestine, bacteria also make up to 60% of the dry mass of feces.[8] Fungi, protists, archaea, and viruses are also present in the gut flora, but less is known about their activities.[19]

Over 99% of the bacteria in the gut are anaerobes, but in the cecum, aerobic bacteria reach high densities.[5] It is estimated that these gut flora have around a hundred times as many genes in total as there are in the human genome.[20]

 
Candida albicans, a dimorphic fungus that grows as a yeast in the gut

Many species in the gut have not been studied outside of their hosts because they cannot be cultured.[16][7][21] While there are a small number of core microbial species shared by most individuals, populations of microbes can vary widely.[22] Within an individual, their microbial populations stay fairly constant over time, with some alterations occurring due to changes in lifestyle, diet and age.[6][23] The Human Microbiome Project has set out to better describe the microbiota of the human gut and other body locations.[citation needed]

The four dominant bacterial phyla in the human gut are Bacillota (Firmicutes), Bacteroidota, Actinomycetota, and Pseudomonadota.[24] Most bacteria belong to the genera Bacteroides, Clostridium, Faecalibacterium,[6][7] Eubacterium, Ruminococcus, Peptococcus, Peptostreptococcus, and Bifidobacterium.[6][7] Other genera, such as Escherichia and Lactobacillus, are present to a lesser extent.[6] Species from the genus Bacteroides alone constitute about 30% of all bacteria in the gut, suggesting that this genus is especially important in the functioning of the host.[16]

Fungal genera that have been detected in the gut include Candida, Saccharomyces, Aspergillus, Penicillium, Rhodotorula, Trametes, Pleospora, Sclerotinia, Bullera, and Galactomyces, among others.[25][26] Rhodotorula is most frequently found in individuals with inflammatory bowel disease while Candida is most frequently found in individuals with hepatitis B cirrhosis and chronic hepatitis B.[25]

Archaea constitute another large class of gut flora which are important in the metabolism of the bacterial products of fermentation.

Industrialization is associated with changes in the microbiota and the reduction of diversity could drive certain species to extinction; in 2018, researchers proposed a biobank repository of human microbiota.[27]

Enterotype edit

An enterotype is a classification of living organisms based on its bacteriological ecosystem in the human gut microbiome not dictated by age, gender, body weight, or national divisions.[28] There are indications that long-term diet influences enterotype.[29] Three human enterotypes have been proposed,[28][30] but their value has been questioned.[31]

Composition edit

Bacteriome edit

Stomach edit

Due to the high acidity of the stomach, most microorganisms cannot survive there. The main bacterial inhabitants of the stomach include: Streptococcus, Staphylococcus, Lactobacillus, Peptostreptococcus.[5]: 720  Helicobacter pylori is a gram-negative spiral bacterium that establishes on gastric mucosa causing chronic gastritis, and peptic ulcer disease, and is a carcinogen for gastric cancer.[5]: 904 

Intestines edit

Bacteria commonly found in the human colon[32]
Bacterium Incidence (%)
Bacteroides fragilis 100
Bacteroides melaninogenicus 100
Bacteroides oralis 100
Enterococcus faecalis 100
Escherichia coli 100
Enterobacter sp. 40–80
Klebsiella sp. 40–80
Bifidobacterium bifidum 30–70
Staphylococcus aureus 30–50
Lactobacillus 20–60
Clostridium perfringens 25–35
Proteus mirabilis 5–55
Clostridium tetani 1–35
Clostridium septicum 5–25
Pseudomonas aeruginosa 3–11
Salmonella enterica 3–7
Faecalibacterium prausnitzii ?common
Peptostreptococcus sp. ?common
Peptococcus sp. ?common

The small intestine contains a trace amount of microorganisms due to the proximity and influence of the stomach. Gram-positive cocci and rod-shaped bacteria are the predominant microorganisms found in the small intestine.[5] However, in the distal portion of the small intestine alkaline conditions support gram-negative bacteria of the Enterobacteriaceae.[5] The bacterial flora of the small intestine aid in a wide range of intestinal functions. The bacterial flora provide regulatory signals that enable the development and utility of the gut. Overgrowth of bacteria in the small intestine can lead to intestinal failure.[33] In addition the large intestine contains the largest bacterial ecosystem in the human body.[5] About 99% of the large intestine and feces flora are made up of obligate anaerobes such as Bacteroides and Bifidobacterium.[34] Factors that disrupt the microorganism population of the large intestine include antibiotics, stress, and parasites.[5]

Bacteria make up most of the flora in the colon[35] and 60% of the dry mass of feces.[6] This fact makes feces an ideal source of gut flora for any tests and experiments by extracting the nucleic acid from fecal specimens, and bacterial 16S rRNA gene sequences are generated with bacterial primers. This form of testing is also often preferable to more invasive techniques, such as biopsies.

Five phyla dominate the intestinal microbiota: Bacteroidota, Bacillota (Firmicutes), Actinomycetota, Pseudomonadota, and Verrucomicrobiota—with Bacteroidota and Bacillota constituting 90% of the composition.[36] Somewhere between 300[6] and 1000 different species live in the gut,[16] with most estimates at about 500.[37][38] However, it is probable that 99% of the bacteria come from about 30 or 40 species, with Faecalibacterium prausnitzii (phylum firmicutes) being the most common species in healthy adults.[7][39]

Research suggests that the relationship between gut flora and humans is not merely commensal (a non-harmful coexistence), but rather is a mutualistic, symbiotic relationship.[16] Though people can survive with no gut flora,[37] the microorganisms perform a host of useful functions, such as fermenting unused energy substrates, training the immune system via end products of metabolism like propionate and acetate, preventing growth of harmful species, regulating the development of the gut, producing vitamins for the host (such as biotin and vitamin K), and producing hormones to direct the host to store fats.[5] Extensive modification and imbalances of the gut microbiota and its microbiome or gene collection are associated with obesity.[40] However, in certain conditions, some species are thought to be capable of causing disease by causing infection or increasing cancer risk for the host.[6][35]

Mycobiome edit

Fungi and protists also make up a part of the gut flora, but less is known about their activities.[41]

Virome edit

The human virome is mostly bacteriophages.[42]

Variation edit

Age edit

It has been demonstrated that there are common patterns of microbiome composition evolution during life.[43] In general, the diversity of microbiota composition of fecal samples is significantly higher in adults than in children, although interpersonal differences are higher in children than in adults.[44] Much of the maturation of microbiota into an adult-like configuration happens during the three first years of life.[44]

As the microbiome composition changes, so does the composition of bacterial proteins produced in the gut. In adult microbiomes, a high prevalence of enzymes involved in fermentation, methanogenesis and the metabolism of arginine, glutamate, aspartate and lysine have been found. In contrast, in infant microbiomes the dominant enzymes are involved in cysteine metabolism and fermentation pathways.[44]

Geography edit

Gut microbiome composition depends on the geographic origin of populations. Variations in a trade-off of Prevotella, the representation of the urease gene, and the representation of genes encoding glutamate synthase/degradation or other enzymes involved in amino acids degradation or vitamin biosynthesis show significant differences between populations from the US, Malawi, or Amerindian origin.[44]

The US population has a high representation of enzymes encoding the degradation of glutamine and enzymes involved in vitamin and lipoic acid biosynthesis; whereas Malawi and Amerindian populations have a high representation of enzymes encoding glutamate synthase and they also have an overrepresentation of α-amylase in their microbiomes. As the US population has a diet richer in fats than Amerindian or Malawian populations which have a corn-rich diet, the diet is probably the main determinant of the gut bacterial composition.[44]

Further studies have indicated a large difference in the composition of microbiota between European and rural African children. The fecal bacteria of children from Florence were compared to that of children from the small rural village of Boulpon in Burkina Faso. The diet of a typical child living in this village is largely lacking in fats and animal proteins and rich in polysaccharides and plant proteins. The fecal bacteria of European children were dominated by Firmicutes and showed a marked reduction in biodiversity, while the fecal bacteria of the Boulpon children was dominated by Bacteroidetes. The increased biodiversity and different composition of the gut microbiome in African populations may aid in the digestion of normally indigestible plant polysaccharides and also may result in a reduced incidence of non-infectious colonic diseases.[45]

On a smaller scale, it has been shown that sharing numerous common environmental exposures in a family is a strong determinant of individual microbiome composition. This effect has no genetic influence and it is consistently observed in culturally different populations.[44]

Malnourishment edit

Malnourished children have less mature and less diverse gut microbiota than healthy children, and changes in the microbiome associated with nutrient scarcity can in turn be a pathophysiological cause of malnutrition.[46][47] Malnourished children also typically have more potentially pathogenic gut flora, and more yeast in their mouths and throats.[48] Altering diet may lead to changes in gut microbiota composition and diversity.[49]

Race and ethnicity edit

Researchers with the American Gut Project and Human Microbiome Project found that twelve microbe families varied in abundance based on the race or ethnicity of the individual. The strength of these associations is limited by the small sample size: the American Gut Project collected data from 1,375 individuals, 90% of whom were white.[50] The Healthy Life in an Urban Setting (HELIUS) study in Amsterdam found that those of Dutch ancestry had the highest level of gut microbiota diversity, while those of South Asian and Surinamese descent had the lowest diversity. The study results suggested that individuals of the same race or ethnicity have more similar microbiomes than individuals of different racial backgrounds.[50]

Socioeconomic status edit

As of 2020, at least two studies have demonstrated a link between an individual's socioeconomic status (SES) and their gut microbiota. A study in Chicago found that individuals in higher SES neighborhoods had greater microbiota diversity. People from higher SES neighborhoods also had more abundant Bacteroides bacteria. Similarly, a study of twins in the United Kingdom found that higher SES was also linked with a greater gut diversity.[50]

Acquisition in human infants edit

The establishment of a gut flora is crucial to the health of an adult, as well as the functioning of the gastrointestinal tract.[51] In humans, a gut flora similar to an adult's is formed within one to two years of birth as microbiota are acquired through parent-to-child transmission and transfer from food, water, and other environmental sources.[52][11]

 
Illustration showing the developmental colonization of gut microbiota.

The traditional view of the gastrointestinal tract of a normal fetus is that it is sterile, although this view has been challenged in the past few years.[timeframe?][53] Multiple lines of evidence have begun to emerge that suggest there may be bacteria in the intrauterine environment. In humans, research has shown that microbial colonization may occur in the fetus[54] with one study showing Lactobacillus and Bifidobacterium species were present in placental biopsies.[55] Several rodent studies have demonstrated the presence of bacteria in the amniotic fluid and placenta, as well as in the meconium of babies born by sterile cesarean section.[56][57] In another study, researchers administered a culture of bacteria orally to pregnant mice, and detected the bacteria in the offspring, likely resulting from transmission between the digestive tract and amniotic fluid via the blood stream.[58] However, researchers caution that the source of these intrauterine bacteria, whether they are alive, and their role, is not yet understood.[59][55]

During birth and rapidly thereafter, bacteria from the mother and the surrounding environment colonize the infant's gut.[11] The exact sources of bacteria are not fully understood, but may include the birth canal, other people (parents, siblings, hospital workers), breastmilk, food, and the general environment with which the infant interacts.[60] Research has shown that the microbiome of babies born vaginally differs significantly from that of babies delivered by caesarean section and that vaginally born babies got most of their gut bacteria from their mother, while the microbiota of babies born by caesarean section had more bacteria associated with hospital environments.[61]

During the first year of life, the composition of the gut flora is generally simple and changes a great deal with time and is not the same across individuals.[11] The initial bacterial population are generally facultative anaerobic organisms; investigators believe that these initial colonizers decrease the oxygen concentration in the gut, which in turn allows obligately anaerobic bacteria like Bacteroidota, Actinomycetota, and Bacillota to become established and thrive.[11] Breast-fed babies become dominated by bifidobacteria, possibly due to the contents of bifidobacterial growth factors in breast milk, and by the fact that breast milk carries prebiotic components, allowing for healthy bacterial growth.[55][62] Breast milk also contains higher levels of Immunoglobulin A (IgA) to help with the tolerance and regulation of the baby's immune system.[63] In contrast, the microbiota of formula-fed infants is more diverse, with high numbers of Enterobacteriaceae, enterococci, bifidobacteria, Bacteroides, and clostridia.[64]

Caesarean section, antibiotics, and formula feeding may alter the gut microbiome composition.[55] Children treated with antibiotics have less stable, and less diverse floral communities.[65] Caesarean sections have been shown to be disruptive to mother-offspring transmission of bacteria, which impacts the overall health of the offspring by raising risks of disease such as celiac disease, asthma, and type 1 diabetes.[55] This further evidences the importance of a healthy gut microbiome. Various methods of microbiome restoration are being explored, typically involving exposing the infant to maternal vaginal contents, and oral probiotics.[55]

Functions edit

When the study of gut flora began in 1995,[66] it was thought to have three key roles: direct defense against pathogens, fortification of host defense by its role in developing and maintaining the intestinal epithelium and inducing antibody production there, and metabolizing otherwise indigestible compounds in food. Subsequent work discovered its role in training the developing immune system, and yet further work focused on its role in the gut-brain axis.[67]

Direct inhibition of pathogens edit

The gut flora community plays a direct role in defending against pathogens by fully colonising the space, making use of all available nutrients, and by secreting compounds known as cytokines that kill or inhibit unwelcome organisms that would compete for nutrients with it.[68] Different strains of gut bacteria cause the production of different cytokines. Cytokines are chemical compounds produced by our immune system for initiating the inflammatory response against infections. Disruption of the gut flora allows competing organisms like Clostridium difficile to become established that otherwise are kept in abeyance.[68]

Development of enteric protection and immune system edit

 
Microfold cells transfer antigens (Ag) from the lumen of the gut to gut-associated lymphoid tissue (GALT) via transcytosis and present them to different innate and adaptive immune cells.

In humans, a gut flora similar to an adult's is formed within one to two years of birth.[11] As the gut flora gets established, the lining of the intestines – the intestinal epithelium and the intestinal mucosal barrier that it secretes – develop as well, in a way that is tolerant to, and even supportive of, commensalistic microorganisms to a certain extent and also provides a barrier to pathogenic ones.[11] Specifically, goblet cells that produce the mucosa proliferate, and the mucosa layer thickens, providing an outside mucosal layer in which "friendly" microorganisms can anchor and feed, and an inner layer that even these organisms cannot penetrate.[11][12] Additionally, the development of gut-associated lymphoid tissue (GALT), which forms part of the intestinal epithelium and which detects and reacts to pathogens, appears and develops during the time that the gut flora develops and established.[11] The GALT that develops is tolerant to gut flora species, but not to other microorganisms.[11] GALT also normally becomes tolerant to food to which the infant is exposed, as well as digestive products of food, and gut flora's metabolites (molecules formed from metabolism) produced from food.[11]

The human immune system creates cytokines that can drive the immune system to produce inflammation in order to protect itself, and that can tamp down the immune response to maintain homeostasis and allow healing after insult or injury.[11] Different bacterial species that appear in gut flora have been shown to be able to drive the immune system to create cytokines selectively; for example Bacteroides fragilis and some Clostridia species appear to drive an anti-inflammatory response, while some segmented filamentous bacteria drive the production of inflammatory cytokines.[11][69] Gut flora can also regulate the production of antibodies by the immune system.[11][70] One function of this regulation is to cause B cells to class switch to IgA. In most cases B cells need activation from T helper cells to induce class switching; however, in another pathway, gut flora cause NF-kB signaling by intestinal epithelial cells which results in further signaling molecules being secreted.[71] These signaling molecules interact with B cells to induce class switching to IgA.[71] IgA is an important type of antibody that is used in mucosal environments like the gut. It has been shown that IgA can help diversify the gut community and helps in getting rid of bacteria that cause inflammatory responses.[72] Ultimately, IgA maintains a healthy environment between the host and gut bacteria.[72] These cytokines and antibodies can have effects outside the gut, in the lungs and other tissues.[11]

The immune system can also be altered due to the gut bacteria's ability to produce metabolites that can affect cells in the immune system. For example short-chain fatty acids (SCFA) can be produced by some gut bacteria through fermentation.[73] SCFAs stimulate a rapid increase in the production of innate immune cells like neutrophils, basophils and eosinophils.[73] These cells are part of the innate immune system that try to limit the spread of infection.

Metabolism edit

Tryptophan metabolism by human gastrointestinal microbiota ()
 
This diagram shows the biosynthesis of bioactive compounds (indole and certain other derivatives) from tryptophan by bacteria in the gut.[74] Indole is produced from tryptophan by bacteria that express tryptophanase.[74] Clostridium sporogenes metabolizes tryptophan into indole and subsequently 3-indolepropionic acid (IPA),[75] a highly potent neuroprotective antioxidant that scavenges hydroxyl radicals.[74][76][77] IPA binds to the pregnane X receptor (PXR) in intestinal cells, thereby facilitating mucosal homeostasis and barrier function.[74] Following absorption from the intestine and distribution to the brain, IPA confers a neuroprotective effect against cerebral ischemia and Alzheimer's disease.[74] Lactobacillaceae (Lactobacillus s.l.) species metabolize tryptophan into indole-3-aldehyde (I3A) which acts on the aryl hydrocarbon receptor (AhR) in intestinal immune cells, in turn increasing interleukin-22 (IL-22) production.[74] Indole itself triggers the secretion of glucagon-like peptide-1 (GLP-1) in intestinal L cells and acts as a ligand for AhR.[74] Indole can also be metabolized by the liver into indoxyl sulfate, a compound that is toxic in high concentrations and associated with vascular disease and renal dysfunction.[74] AST-120 (activated charcoal), an intestinal sorbent that is taken by mouth, adsorbs indole, in turn decreasing the concentration of indoxyl sulfate in blood plasma.[74]

Without gut flora, the human body would be unable to utilize some of the undigested carbohydrates it consumes, because some types of gut flora have enzymes that human cells lack for breaking down certain polysaccharides.[13] Rodents raised in a sterile environment and lacking in gut flora need to eat 30% more calories just to remain the same weight as their normal counterparts.[13] Carbohydrates that humans cannot digest without bacterial help include certain starches, fiber, oligosaccharides, and sugars that the body failed to digest and absorb like lactose in the case of lactose intolerance and sugar alcohols, mucus produced by the gut, and proteins.[9][13]

Bacteria turn carbohydrates they ferment into short-chain fatty acids by a form of fermentation called saccharolytic fermentation.[38] Products include acetic acid, propionic acid and butyric acid.[7][38] These materials can be used by host cells, providing a major source of energy and nutrients.[38] Gases (which are involved in signaling[78] and may cause flatulence) and organic acids, such as lactic acid, are also produced by fermentation.[7] Acetic acid is used by muscle, propionic acid facilitates liver production of ATP, and butyric acid provides energy to gut cells.[38]

Gut flora also synthesize vitamins like biotin and folate, and facilitate absorption of dietary minerals, including magnesium, calcium, and iron.[6][23] Methanobrevibacter smithii is unique because it is not a species of bacteria, but rather a member of domain Archaea, and is the most abundant methane-producing archaeal species in the human gastrointestinal microbiota.[79]

Gut microbiota also serve as a source of Vitamins K and B12 that are not produced by the body or produced in little amount.[80][81]

Pharmacomicrobiomics edit

The human metagenome (i.e., the genetic composition of an individual and all microorganisms that reside on or within the individual's body) varies considerably between individuals.[82][83] Since the total number of microbial cells in the human body (over 100 trillion) greatly outnumbers Homo sapiens cells (tens of trillions),[note 1][82][84] there is considerable potential for interactions between drugs and an individual's microbiome, including: drugs altering the composition of the human microbiome, drug metabolism by microbial enzymes modifying the drug's pharmacokinetic profile, and microbial drug metabolism affecting a drug's clinical efficacy and toxicity profile.[82][83][85]

Apart from carbohydrates, gut microbiota can also metabolize other xenobiotics such as drugs, phytochemicals, and food toxicants. More than 30 drugs have been shown to be metabolized by gut microbiota.[86] The microbial metabolism of drugs can sometimes inactivate the drug.[87]

Contribution to drug metabolism edit

The gut microbiota is an enriched community that contains diverse genes with huge biochemical capabilities to modify drugs, especially those taken by mouth.[88] Gut microbiota can affect drug metabolism via direct and indirect mechanisms.[89] The direct mechanism is mediated by the microbial enzymes that can modify the chemical structure of the administered drugs.[90] Conversely, the indirect pathway is mediated by the microbial metabolites which affect the expression of host metabolizing enzymes such as cytochrome P450.[91][89] The effects of the gut microbiota on the pharmacokinetics and bioavailability of the drug have been investigated a few decades ago.[92][93][94] These effects can be varied; it could activate the inactive drugs such as lovastatin,[95] inactivate the active drug such as digoxin[96] or induce drug toxicity as in irinotecan.[97] Since then, the impacts of the gut microbiota on the pharmacokinetics of many drugs were heavily studied.[98][88]

The human gut microbiota plays a crucial role in modulating the effect of the administered drugs on the human. Directly, gut microbiota can synthesize and release a series of enzymes with the capability to metabolize drugs such as microbial biotransformation of L-dopa by decarboxylase and dehydroxylase enzymes.[90] On the contrary, gut microbiota may also alter the metabolism of the drugs by modulating the host drug metabolism. This mechanism can be mediated by microbial metabolites or by modifying host metabolites which in turn change the expression of host metabolizing enzymes.[91]

A large number of studies have demonstrated the metabolism of over 50 drugs by the gut microbiota.[98][89] For example, lovastatin (a cholesterol-lowering agent) which is a lactone prodrug is partially activated by the human gut microbiota forming active acid hydroxylated metabolites.[95] Conversely, digoxin (a drug used to treat Congestive Heart Failure) is inactivated by a member of the gut microbiota (i.e. Eggerthella lanta).[99] Eggerthella lanta has a cytochrome-encoding operon up-regulated by digoxin and associated with digoxin-inactivation.[99] Gut microbiota can also modulate the efficacy and toxicity of chemotherapeutic agents such as irinotecan.[100] This effect is derived from the microbiome-encoded β-glucuronidase enzymes which recover the active form of the irinotecan causing gastrointestinal toxicity.[101]

Secondary metabolites edit

This microbial community in the gut has a huge biochemical capability to produce distinct secondary metabolites that are sometimes produced from the metabolic conversion of dietary foods such as fibers, endogenous biological compounds such as Indole or Bile acids.[102][103][104] Microbial metabolites especially short chain fatty acids (SCFAs) and secondary bile acids (BAs) play important roles for the human in health and disease states.[105][106][107]

One of the most important bacterial metabolites produced by the gut microbiota is secondary bile acids (BAs).[104] These metabolites are produced by the bacterial biotransformation of the primary bile acids such as cholic acid (CA) and chenodeoxycholic acid (CDCA) into secondary bile acids (BAs) lithocholic acid (LCA) and deoxy cholic acid (DCA) respectively.[108] Primary bile acids which are synthesized by hepatocytes and stored in the gall bladder possess hydrophobic characters. These metabolites are subsequently metabolized by the gut microbiota into secondary metabolites with increased hydrophobicity.[108] Bile salt hydrolases (BSH) which are conserved across gut microbiota phyla such as Bacteroides, Firmicutes, and Actinobacteria responsible for the first step of secondary bile acids metabolism.[108] Secondary bile acids (BAs) such as DCA and LCA have been demonstrated to inhibit both Clostridium difficile germination and outgrowth.[107]

Dysbiosis edit

The gut microbiota is important for maintaining homeostasis in the intestine. Development of intestinal cancer is associated with an imbalance in the natural microflora (dysbiosis).[109] The secondary bile acid deoxycholic acid is associated with alterations of the microbial community that lead to increased intestinal carcinogenesis.[109] Increased exposure of the colon to secondary bile acids resulting from dysbiosis can cause DNA damage, and such damage can produce carcinogenic mutations in cells of the colon.[110] The high density of bacteria in the colon (about 1012 per ml.) that are subject to dysbiosis compared to the relatively low density in the small intestine (about 102 per ml.) may account for the greater than 10-fold higher incidence of cancer in the colon compared to the small intestine.[110]

Gut-brain axis edit

The gut-brain axis is the biochemical signaling that takes place between the gastrointestinal tract and the central nervous system.[67] That term has been expanded to include the role of the gut flora in the interplay; the term "microbiome-gut-brain axis" is sometimes used to describe paradigms explicitly including the gut flora.[67][111][112] Broadly defined, the gut-brain axis includes the central nervous system, neuroendocrine and neuroimmune systems including the hypothalamic–pituitary–adrenal axis (HPA axis), sympathetic and parasympathetic arms of the autonomic nervous system including the enteric nervous system, the vagus nerve, and the gut microbiota.[67][112]

A systematic review from 2016 examined the preclinical and small human trials that have been conducted with certain commercially available strains of probiotic bacteria and found that among those tested, Bifidobacterium and Lactobacillus genera (B. longum, B. breve, B. infantis, L. helveticus, L. rhamnosus, L. plantarum, and L. casei), had the most potential to be useful for certain central nervous system disorders.[15]

Alterations in microbiota balance edit

Effects of antibiotic use edit

Altering the numbers of gut bacteria, for example by taking broad-spectrum antibiotics, may affect the host's health and ability to digest food.[113] Antibiotics can cause antibiotic-associated diarrhea by irritating the bowel directly, changing the levels of microbiota, or allowing pathogenic bacteria to grow.[7] Another harmful effect of antibiotics is the increase in numbers of antibiotic-resistant bacteria found after their use, which, when they invade the host, cause illnesses that are difficult to treat with antibiotics.[113]

Changing the numbers and species of gut microbiota can reduce the body's ability to ferment carbohydrates and metabolize bile acids and may cause diarrhea. Carbohydrates that are not broken down may absorb too much water and cause runny stools, or lack of SCFAs produced by gut microbiota could cause diarrhea.[7]

A reduction in levels of native bacterial species also disrupts their ability to inhibit the growth of harmful species such as C. difficile and Salmonella kedougou, and these species can get out of hand, though their overgrowth may be incidental and not be the true cause of diarrhea.[6][7][113] Emerging treatment protocols for C. difficile infections involve fecal microbiota transplantation of donor feces (see Fecal transplant).[114] Initial reports of treatment describe success rates of 90%, with few side effects. Efficacy is speculated to result from restoring bacterial balances of bacteroides and firmicutes classes of bacteria.[115]

The composition of the gut microbiome also changes in severe illnesses, due not only to antibiotic use but also to such factors as ischemia of the gut, failure to eat, and immune compromise. Negative effects from this have led to interest in selective digestive tract decontamination, a treatment to kill only pathogenic bacteria and allow the re-establishment of healthy ones.[116]

Antibiotics alter the population of the microbiota in the gastrointestinal tract, and this may change the intra-community metabolic interactions, modify caloric intake by using carbohydrates, and globally affects host metabolic, hormonal and immune homeostasis.[117]

There is reasonable evidence that taking probiotics containing Lactobacillus species may help prevent antibiotic-associated diarrhea and that taking probiotics with Saccharomyces (e.g., Saccharomyces boulardii ) may help to prevent Clostridium difficile infection following systemic antibiotic treatment.[118]

Pregnancy edit

The gut microbiota of a woman changes as pregnancy advances, with the changes similar to those seen in metabolic syndromes such as diabetes. The change in gut microbiota causes no ill effects. The newborn's gut microbiota resemble the mother's first-trimester samples. The diversity of the microbiome decreases from the first to third trimester, as the numbers of certain species go up.[55][119]

Probiotics, prebiotics, synbiotics, and pharmabiotics edit

Probiotics are microorganisms that are believed to provide health benefits when consumed.[120][121] With regard to gut microbiota, prebiotics are typically non-digestible, fiber compounds that pass undigested through the upper part of the gastrointestinal tract and stimulate the growth or activity of advantageous gut flora by acting as substrate for them.[38][122]

Synbiotics refers to food ingredients or dietary supplements combining probiotics and prebiotics in a form of synergism.[123]

The term "pharmabiotics" is used in various ways, to mean: pharmaceutical formulations (standardized manufacturing that can obtain regulatory approval as a drug) of probiotics, prebiotics, or synbiotics;[124] probiotics that have been genetically engineered or otherwise optimized for best performance (shelf life, survival in the digestive tract, etc.);[125] and the natural products of gut flora metabolism (vitamins, etc.).[126]

There is some evidence that treatment with some probiotic strains of bacteria may be effective in irritable bowel syndrome[127] and chronic idiopathic constipation. Those organisms most likely to result in a decrease of symptoms have included:

Fecal floatation edit

Feces of about 10-15% of people consistently floats in toilet water ('floaters'), while the rest produce feces that sinks ('sinkers') and production of gas causes feces to float.[131] While conventional mice often produce 'floaters', gnotobiotic germfree mice no gut microbiota (bred in germfree isolator) produce 'sinkers', and gut microbiota colonization in germfree mice leads to food transformation to microbial biomass and enrichment of multiple gasogenic bacterial species that turns the 'sinkers' into 'floaters'.[132]

Research edit

Tests for whether non-antibiotic drugs may impact human gut-associated bacteria were performed by in vitro analysis on more than 1000 marketed drugs against 40 gut bacterial strains, demonstrating that 24% of the drugs inhibited the growth of at least one of the bacterial strains.[133]

Effects of exercise edit

Gut microbiota and exercise have recently been shown to be interconnected. Both moderate and intense exercise are typically part of the training regimen of endurance athletes, but they exert different effects on health. The interconnection between gut microbiota and endurance sports depends upon exercise intensity and training status.[134]

Role in disease edit

Bacteria in the digestive tract can contribute to and be affected by disease in various ways. The presence or overabundance of some kinds of bacteria may contribute to inflammatory disorders such as inflammatory bowel disease.[6] Additionally, metabolites from certain members of the gut flora may influence host signalling pathways, contributing to disorders such as obesity and colon cancer.[6] Alternatively, in the event of a breakdown of the gut epithelium, the intrusion of gut flora components into other host compartments can lead to sepsis.[6]

Ulcers edit

Helicobacter pylori infection can initiate formation of stomach ulcers when the bacteria penetrate the stomach epithelial lining, then causing an inflammatory phagocytotic response.[135] In turn, the inflammation damages parietal cells which release excessive hydrochloric acid into the stomach and produce less of the protective mucus.[136] Injury to the stomach lining, leading to ulcers, develops when gastric acid overwhelms the defensive properties of cells and inhibits endogenous prostaglandin synthesis, reduces mucus and bicarbonate secretion, reduces mucosal blood flow, and lowers resistance to injury.[136] Reduced protective properties of the stomach lining increase vulnerability to further injury and ulcer formation by stomach acid, pepsin, and bile salts.[135][136]

Bowel perforation edit

Normally-commensal bacteria can harm the host if they extrude from the intestinal tract.[11][12] Translocation, which occurs when bacteria leave the gut through its mucosal lining, can occur in a number of different diseases.[12] If the gut is perforated, bacteria invade the interstitium, causing a potentially fatal infection.[5]: 715 

Inflammatory bowel diseases edit

The two main types of inflammatory bowel diseases, Crohn's disease and ulcerative colitis, are chronic inflammatory disorders of the gut; the causes of these diseases are unknown and issues with the gut flora and its relationship with the host have been implicated in these conditions.[14][137][138][139] Additionally, it appears that interactions of gut flora with the gut-brain axis have a role in IBD, with physiological stress mediated through the hypothalamic–pituitary–adrenal axis driving changes to intestinal epithelium and the gut flora in turn releasing factors and metabolites that trigger signaling in the enteric nervous system and the vagus nerve.[4]

The diversity of gut flora appears to be significantly diminished in people with inflammatory bowel diseases compared to healthy people; additionally, in people with ulcerative colitis, Proteobacteria and Actinobacteria appear to dominate; in people with Crohn's, Enterococcus faecium and several Proteobacteria appear to be over-represented.[4]

There is reasonable evidence that correcting gut flora imbalances by taking probiotics with Lactobacilli and Bifidobacteria can reduce visceral pain and gut inflammation in IBD.[118]

Irritable bowel syndrome edit

Irritable bowel syndrome is a result of stress and chronic activation of the HPA axis; its symptoms include abdominal pain, changes in bowel movements, and an increase in proinflammatory cytokines. Overall, studies have found that the luminal and mucosal microbiota are changed in irritable bowel syndrome individuals, and these changes can relate to the type of irritation such as diarrhea or constipation. Also, there is a decrease in the diversity of the microbiome with low levels of fecal Lactobacilli and Bifidobacteria, high levels of facultative anaerobic bacteria such as Escherichia coli, and increased ratios of Firmicutes: Bacteroidetes.[112]

Asthma edit

With asthma, two hypotheses have been posed to explain its rising prevalence in the developed world. The hygiene hypothesis posits that children in the developed world are not exposed to enough microbes and thus may contain lower prevalence of specific bacterial taxa that play protective roles.[140] The second hypothesis focuses on the Western pattern diet, which lacks whole grains and fiber and has an overabundance of simple sugars.[14] Both hypotheses converge on the role of short-chain fatty acids (SCFAs) in immunomodulation. These bacterial fermentation metabolites are involved in immune signalling that prevents the triggering of asthma and lower SCFA levels are associated with the disease.[140][141] Lacking protective genera such as Lachnospira, Veillonella, Rothia and Faecalibacterium has been linked to reduced SCFA levels.[140] Further, SCFAs are the product of bacterial fermentation of fiber, which is low in the Western pattern diet.[14][141] SCFAs offer a link between gut flora and immune disorders, and as of 2016, this was an active area of research.[14] Similar hypotheses have also been posited for the rise of food and other allergies.[142]

Diabetes mellitus type 1 edit

The connection between the gut microbiota and diabetes mellitus type 1 has also been linked to SCFAs, such as butyrate and acetate. Diets yielding butyrate and acetate from bacterial fermentation show increased Treg expression.[143] Treg cells downregulate effector T cells, which in turn reduces the inflammatory response in the gut.[144] Butyrate is an energy source for colon cells. butyrate-yielding diets thus decrease gut permeability by providing sufficient energy for the formation of tight junctions.[145] Additionally, butyrate has also been shown to decrease insulin resistance, suggesting gut communities low in butyrate-producing microbes may increase chances of acquiring diabetes mellitus type 2.[146] Butyrate-yielding diets may also have potential colorectal cancer suppression effects.[145]

Obesity and metabolic syndrome edit

The gut flora have been implicated in obesity and metabolic syndrome due to a key role in the digestive process; the Western pattern diet appears to drive and maintain changes in the gut flora that in turn change how much energy is derived from food and how that energy is used.[139][147] One aspect of a healthy diet that is often lacking in the Western-pattern diet is fiber and other complex carbohydrates that a healthy gut flora require flourishing; changes to gut flora in response to a Western-pattern diet appear to increase the amount of energy generated by the gut flora which may contribute to obesity and metabolic syndrome.[118] There is also evidence that microbiota influence eating behaviours based on the preferences of the microbiota, which can lead to the host consuming more food eventually resulting in obesity. It has generally been observed that with higher gut microbiome diversity, the microbiota will spend energy and resources on competing with other microbiota and less on manipulating the host. The opposite is seen with lower gut microbiome diversity, and these microbiotas may work together to create host food cravings.[49]

Additionally, the liver plays a dominant role in blood glucose homeostasis by maintaining a balance between the uptake and storage of glucose through the metabolic pathways of glycogenesis and gluconeogenesis. Intestinal lipids regulate glucose homeostasis involving a gut-brain-liver axis. The direct administration of lipids into the upper intestine increases the long chain fatty acyl-coenzyme A (LCFA-CoA) levels in the upper intestines and suppresses glucose production even under subdiaphragmatic vagotomy or gut vagal deafferentation. This interrupts the neural connection between the brain and the gut and blocks the upper intestinal lipids' ability to inhibit glucose production. The gut-brain-liver axis and gut microbiota composition can regulate the glucose homeostasis in the liver and provide potential therapeutic methods to treat obesity and diabetes.[148]

Just as gut flora can function in a feedback loop that can drive the development of obesity, there is evidence that restricting intake of calories (i.e., dieting) can drive changes to the composition of the gut flora.[139]

Other animals edit

The composition of the human gut microbiome is similar to that of the other great apes. However, humans' gut biota has decreased in diversity and changed in composition since our evolutionary split from Pan.[149] Humans display increases in Bacteroidetes, a bacterial phylum associated with diets high in animal protein and fat, and decreases in Methanobrevibacter and Fibrobacter, groups that ferment complex plant polysaccharides.[149] These changes are the result of the combined dietary, genetic, and cultural changes humans have undergone since evolutionary divergence from Pan.[citation needed]

In addition to humans and vertebrates, some insects also have complex and diverse gut microbiota that play key nutritional roles.[2] Microbial communities associated with termites can constitute a majority of the weight of the individuals and perform important roles in the digestion of lignocellulose and nitrogen fixation.[150] These communities are host-specific, and closely related insect species share comparable similarities in gut microbiota composition.[151][152] In cockroaches, gut microbiota have been shown to assemble in a deterministic fashion, irrespective of the inoculum;[153] the reason for this host-specific assembly remains unclear. Bacterial communities associated with insects like termites and cockroaches are determined by a combination of forces, primarily diet, but there is some indication that host phylogeny may also be playing a role in the selection of lineages.[151][152]

For more than 51 years it has been known that the administration of low doses of antibacterial agents promotes the growth of farm animals to increase weight gain.[117]

In a study carried out on mice the ratio of Firmicutes and Lachnospiraceae was significantly elevated in animals treated with subtherapeutic doses of different antibiotics. By analyzing the caloric content of faeces and the concentration of small chain fatty acids (SCFAs) in the GI tract, it was concluded that the changes in the composition of microbiota lead to an increased capacity to extract calories from otherwise indigestible constituents, and to an increased production of SCFAs. These findings provide evidence that antibiotics perturb not only the composition of the GI microbiome but also its metabolic capabilities, specifically with respect to SCFAs.[117]

See also edit

Notes edit

  1. ^ There is substantial variation in microbiome composition and microbial concentrations by anatomical site.[82][83] Fluid from the human colon – which contains the highest concentration of microbes of any anatomical site – contains approximately one trillion (10^12) bacterial cells/ml.[82]

References edit

  1. ^ Moszak, M; Szulińska, M; Bogdański, P (15 April 2020). "You Are What You Eat-The Relationship between Diet, Microbiota, and Metabolic Disorders-A Review". Nutrients. 12 (4): 1096. doi:10.3390/nu12041096. PMC 7230850. PMID 32326604. S2CID 216108564.
  2. ^ a b Engel, P.; Moran, N. (2013). "The gut microbiota of insects–diversity in structure and function". FEMS Microbiology Reviews. 37 (5): 699–735. doi:10.1111/1574-6976.12025. PMID 23692388.
  3. ^ Segata, N; Boernigen, D; Tickle, TL; Morgan, XC; Garrett, WS; Huttenhower, C (14 May 2013). "Computational meta'omics for microbial community studies". Molecular Systems Biology. 9: 666. doi:10.1038/msb.2013.22. PMC 4039370. PMID 23670539.
  4. ^ a b c Saxena, R.; Sharma, V.K (2016). "A Metagenomic Insight Into the Human Microbiome: Its Implications in Health and Disease". In Kumar, D.; S. Antonarakis (eds.). Medical and Health Genomics. Elsevier Science. p. 117. doi:10.1016/B978-0-12-420196-5.00009-5. ISBN 978-0-12-799922-7.
  5. ^ a b c d e f g h i j k l m Sherwood, Linda; Willey, Joanne; Woolverton, Christopher (2013). Prescott's Microbiology (9th ed.). New York: McGraw Hill. pp. 713–21. ISBN 9780073402406. OCLC 886600661.
  6. ^ a b c d e f g h i j k l m n o Guarner, F; Malagelada, J (2003). "Gut flora in health and disease". The Lancet. 361 (9356): 512–19. doi:10.1016/S0140-6736(03)12489-0. PMID 12583961. S2CID 38767655.
  7. ^ a b c d e f g h i j k Beaugerie, Laurent; Petit, Jean-Claude (2004). "Antibiotic-associated diarrhoea". Best Practice & Research Clinical Gastroenterology. 18 (2): 337–52. doi:10.1016/j.bpg.2003.10.002. PMID 15123074.
  8. ^ a b Stephen, A. M.; Cummings, J. H. (1980). "The Microbial Contribution to Human Faecal Mass". Journal of Medical Microbiology. 13 (1): 45–56. doi:10.1099/00222615-13-1-45. PMID 7359576.
  9. ^ a b c d e Quigley, E. M (2013). "Gut bacteria in health and disease". Gastroenterology & Hepatology. 9 (9): 560–9. PMC 3983973. PMID 24729765.
  10. ^ Turnbaugh, Peter J.; Ley, Ruth E.; Hamady, Micah; Fraser-Liggett, Claire M.; Knight, Rob; Gordon, Jeffrey I. (October 2007). "The Human Microbiome Project". Nature. 449 (7164): 804–810. Bibcode:2007Natur.449..804T. doi:10.1038/nature06244. ISSN 0028-0836. PMC 3709439. PMID 17943116.
  11. ^ a b c d e f g h i j k l m n o p Sommer, Felix; Bäckhed, Fredrik (2013). "The gut microbiota — masters of host development and physiology". Nature Reviews Microbiology. 11 (4): 227–38. doi:10.1038/nrmicro2974. PMID 23435359. S2CID 22798964.
  12. ^ a b c d Faderl, Martin; Noti, Mario; Corazza, Nadia; Mueller, Christoph (2015). "Keeping bugs in check: The mucus layer as a critical component in maintaining intestinal homeostasis". IUBMB Life. 67 (4): 275–85. doi:10.1002/iub.1374. PMID 25914114. S2CID 25878594.
  13. ^ a b c d e f Clarke, Gerard; Stilling, Roman M; Kennedy, Paul J; Stanton, Catherine; Cryan, John F; Dinan, Timothy G (2014). "Minireview: Gut Microbiota: The Neglected Endocrine Organ". Molecular Endocrinology. 28 (8): 1221–38. doi:10.1210/me.2014-1108. PMC 5414803. PMID 24892638.
  14. ^ a b c d e f Shen, Sj; Wong, Connie HY (2016). "Bugging inflammation: Role of the gut microbiota". Clinical & Translational Immunology. 5 (4): e72. doi:10.1038/cti.2016.12. PMC 4855262. PMID 27195115.
  15. ^ a b Wang, Huiying; Lee, In-Seon; Braun, Christoph; Enck, Paul (2016). "Effect of Probiotics on Central Nervous System Functions in Animals and Humans: A Systematic Review". Journal of Neurogastroenterology and Motility. 22 (4): 589–605. doi:10.5056/jnm16018. PMC 5056568. PMID 27413138.
  16. ^ a b c d e f Sears, Cynthia L. (2005). "A dynamic partnership: Celebrating our gut flora". Anaerobe. 11 (5): 247–51. doi:10.1016/j.anaerobe.2005.05.001. PMID 16701579.
  17. ^ Shapira, Michael (2016-07-01). "Gut Microbiotas and Host Evolution: Scaling Up Symbiosis". Trends in Ecology & Evolution. 31 (7): 539–549. doi:10.1016/j.tree.2016.03.006. ISSN 0169-5347. PMID 27039196.
  18. ^ Walker, Alan W.; Hoyles, Lesley (August 2023). "Human microbiome myths and misconceptions". Nature Microbiology. 8 (8): 1392–1396. doi:10.1038/s41564-023-01426-7. ISSN 2058-5276. PMID 37524974.
  19. ^ Lozupone, Catherine A.; Stombaugh, Jesse I.; Gordon, Jeffrey I.; Jansson, Janet K.; Knight, Rob (2012). "Diversity, stability and resilience of the human gut microbiota". Nature. 489 (7415): 220–30. Bibcode:2012Natur.489..220L. doi:10.1038/nature11550. PMC 3577372. PMID 22972295.
  20. ^ Qin, Junjie; Li, Ruiqiang; Raes, Jeroen; Arumugam, Manimozhiyan; Burgdorf, Kristoffer Solvsten; Manichanh, Chaysavanh; Nielsen, Trine; Pons, Nicolas; Levenez, Florence; Yamada, Takuji; Mende, Daniel R.; Li, Junhua; Xu, Junming; Li, Shaochuan; Li, Dongfang; Cao, Jianjun; Wang, Bo; Liang, Huiqing; Zheng, Huisong; Xie, Yinlong; Tap, Julien; Lepage, Patricia; Bertalan, Marcelo; Batto, Jean-Michel; Hansen, Torben; Le Paslier, Denis; Linneberg, Allan; Nielsen, H. Bjørn; Pelletier, Eric; Renault, Pierre (2010). "A human gut microbial gene catalogue established by metagenomic sequencing". Nature. 464 (7285): 59–65. Bibcode:2010Natur.464...59.. doi:10.1038/nature08821. PMC 3779803. PMID 20203603.
  21. ^ Shanahan, Fergus (2002). "The host–microbe interface within the gut". Best Practice & Research Clinical Gastroenterology. 16 (6): 915–31. doi:10.1053/bega.2002.0342. PMID 12473298.
  22. ^ Tap, Julien; Mondot, Stanislas; Levenez, Florence; Pelletier, Eric; Caron, Christophe; Furet, Jean-Pierre; Ugarte, Edgardo; Muñoz-Tamayo, Rafael; Paslier, Denis L. E.; Nalin, Renaud; Dore, Joel; Leclerc, Marion (2009). "Towards the human intestinal microbiota phylogenetic core". Environmental Microbiology. 11 (10): 2574–84. Bibcode:2009EnvMi..11.2574T. doi:10.1111/j.1462-2920.2009.01982.x. PMID 19601958.
  23. ^ a b O'Hara, Ann M; Shanahan, Fergus (2006). "The gut flora as a forgotten organ". EMBO Reports. 7 (7): 688–93. doi:10.1038/sj.embor.7400731. PMC 1500832. PMID 16819463.
  24. ^ Khanna, Sahil; Tosh, Pritish K (2014). "A Clinician's Primer on the Role of the Microbiome in Human Health and Disease". Mayo Clinic Proceedings. 89 (1): 107–14. doi:10.1016/j.mayocp.2013.10.011. PMID 24388028.
  25. ^ a b Cui, Lijia; Morris, Alison; Ghedin, Elodie (2013). "The human mycobiome in health and disease". Genome Medicine. 5 (7): 63. doi:10.1186/gm467. PMC 3978422. PMID 23899327.
  26. ^ Erdogan, Askin; Rao, Satish S. C (2015). "Small Intestinal Fungal Overgrowth". Current Gastroenterology Reports. 17 (4): 16. doi:10.1007/s11894-015-0436-2. PMID 25786900. S2CID 3098136.
  27. ^ Bello, Maria G. Dominguez; Knight, Rob; Gilbert, Jack A.; Blaser, Martin J. (4 October 2018). "Preserving microbial diversity". Science. 362 (6410): 33–34. Bibcode:2018Sci...362...33B. doi:10.1126/science.aau8816. PMID 30287652. S2CID 52919917.
  28. ^ a b Arumugam, Manimozhiyan; Raes, Jeroen; Pelletier, Eric; Le Paslier, Denis; Yamada, Takuji; Mende, Daniel R.; Fernandes, Gabriel R.; Tap, Julien; Bruls, Thomas; Batto, Jean-Michel; Bertalan, Marcelo; Borruel, Natalia; Casellas, Francesc; Fernandez, Leyden; Gautier, Laurent; Hansen, Torben; Hattori, Masahira; Hayashi, Tetsuya; Kleerebezem, Michiel; Kurokawa, Ken; Leclerc, Marion; Levenez, Florence; Manichanh, Chaysavanh; Nielsen, H. Bjørn; Nielsen, Trine; Pons, Nicolas; Poulain, Julie; Qin, Junjie; Sicheritz-Ponten, Thomas; Tims, Sebastian (2011). "Enterotypes of the human gut microbiome". Nature. 473 (7346): 174–80. Bibcode:2011Natur.473..174.. doi:10.1038/nature09944. PMC 3728647. PMID 21508958.
  29. ^ Wu, G. D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.-Y.; Keilbaugh, S. A.; Bewtra, M.; Knights, D.; Walters, W. A.; Knight, R.; Sinha, R.; Gilroy, E.; Gupta, K.; Baldassano, R.; Nessel, L.; Li, H.; Bushman, F. D.; Lewis, J. D. (2011). "Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes". Science. 334 (6052): 105–08. Bibcode:2011Sci...334..105W. doi:10.1126/science.1208344. PMC 3368382. PMID 21885731.
  30. ^ Zimmer, Carl (April 20, 2011). "Bacteria Divide People Into 3 Types, Scientists Say". The New York Times. Retrieved April 21, 2011. a group of scientists now report just three distinct ecosystems in the guts of people they have studied.
  31. ^ Knights, Dan; Ward, Tonya; McKinlay, Christopher; Miller, Hannah; Gonzalez, Antonio; McDonald, Daniel; Knight, Rob (8 October 2014). "Rethinking "Enterotypes"". Cell Host & Microbe. 16 (4): 433–37. doi:10.1016/j.chom.2014.09.013. PMC 5558460. PMID 25299329.
  32. ^ Todar, Kenneth (2012). "The Normal Bacterial Flora of Humans". Todar's Online Textbook of Bacteriology. Retrieved June 25, 2016.
  33. ^ Quigley, Eamonn M.M; Quera, Rodrigo (2006). "Small Intestinal Bacterial Overgrowth: Roles of Antibiotics, Prebiotics, and Probiotics". Gastroenterology. 130 (2): S78–90. doi:10.1053/j.gastro.2005.11.046. PMID 16473077. S2CID 16904501.
  34. ^ Adams, M. R.; Moss, M. O. (2007). Food Microbiology. doi:10.1039/9781847557940. ISBN 978-0-85404-284-5. S2CID 241261974.
  35. ^ a b (PDF) (slideshow). 2004. Archived from the original (PDF) on 2004-05-26. Retrieved 2023-01-02 – via University of Glasgow.
  36. ^ Braune A, Blaut M (2016). "Bacterial species involved in the conversion of dietary flavonoids in the human gut". Gut Microbes. 7 (3): 216–234. doi:10.1080/19490976.2016.1158395. PMC 4939924. PMID 26963713.
  37. ^ a b Steinhoff, U (2005). "Who controls the crowd? New findings and old questions about the intestinal microflora". Immunology Letters. 99 (1): 12–16. doi:10.1016/j.imlet.2004.12.013. PMID 15894105.
  38. ^ a b c d e f Gibson, Glenn R (2004). "Fibre and effects on probiotics (the prebiotic concept)". Clinical Nutrition Supplements. 1 (2): 25–31. doi:10.1016/j.clnu.2004.09.005.
  39. ^ Miquel, S; Martín, R; Rossi, O; Bermúdez-Humarán, LG; Chatel, JM; Sokol, H; Thomas, M; Wells, JM; Langella, P (2013). "Faecalibacterium prausnitzii and human intestinal health". Current Opinion in Microbiology. 16 (3): 255–61. doi:10.1016/j.mib.2013.06.003. PMID 23831042.
  40. ^ Ley, Ruth E (2010). "Obesity and the human microbiome". Current Opinion in Gastroenterology. 26 (1): 5–11. doi:10.1097/MOG.0b013e328333d751. PMID 19901833. S2CID 23329156.
  41. ^ Nash, Andrea K; Auchtung, Thomas A; Wong, Matthew C; Smith, Daniel P; Gesell, Jonathan R; Ross, Matthew C; Stewart, Christopher J; Metcalf, Ginger A; Muzny, Donna M; Gibbs, Richard A; Ajami, Nadim J; Petrosino, Joseph F (2017). "The gut mycobiome of the Human Microbiome Project healthy cohort". Microbiome. 5 (1): 153. doi:10.1186/s40168-017-0373-4. PMC 5702186. PMID 29178920.
  42. ^ Scarpellini, Emidio; Ianiro, Gianluca; Attili, Fabia; Bassanelli, Chiara; De Santis, Adriano; Gasbarrini, Antonio (2015). "The human gut microbiota and virome: Potential therapeutic implications". Digestive and Liver Disease. 47 (12): 1007–12. doi:10.1016/j.dld.2015.07.008. PMC 7185617. PMID 26257129.
  43. ^ Gerritsen, Jacoline; Smidt, Hauke; Rijkers, Ger; de Vos, Willem (27 May 2011). "Intestinal microbiota in human health and disease: the impact of probiotics". Genes & Nutrition. 6 (3): 209–40. doi:10.1007/s12263-011-0229-7. PMC 3145058. PMID 21617937.
  44. ^ a b c d e f Yatsunenko, T.; Rey, F. E.; Manary, M. J.; Trehan, I.; Dominguez-Bello, M. G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R. N.; Anokhin, A. P.; Heath, A. C.; Warner, B.; Reeder, J.; Kuczynski, J.; Caporaso, J. G.; Lozupone, C. A.; Lauber, C.; Clemente, J. C.; Knights, D.; Knight, R.; Gordon, J. I. (2012). "Human gut microbiome viewed across age and geography". Nature. 486 (7402): 222–27. Bibcode:2012Natur.486..222Y. doi:10.1038/nature11053. PMC 3376388. PMID 22699611.
  45. ^ De Filippo, C; Cavalieri, D; Di Paola, M; Ramazzotti, M; Poullet, J. B; Massart, S; Collini, S; Pieraccini, G; Lionetti, P (2010). "Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa". Proceedings of the National Academy of Sciences. 107 (33): 14691–6. Bibcode:2010PNAS..10714691D. doi:10.1073/pnas.1005963107. PMC 2930426. PMID 20679230.
  46. ^ Jonkers, Daisy M.A.E. (2016). "Microbial perturbations and modulation in conditions associated with malnutrition and malabsorption". Best Practice & Research Clinical Gastroenterology. 30 (2): 161–72. doi:10.1016/j.bpg.2016.02.006. PMID 27086883.
  47. ^ Million, Matthieu; Diallo, Aldiouma; Raoult, Didier (May 2017). "Gut microbiota and malnutrition" (PDF). Microbial Pathogenesis. 106: 127–138. doi:10.1016/j.micpath.2016.02.003. PMID 26853753. S2CID 20381329.
  48. ^ Rytter, Maren Johanne Heilskov; Kolte, Lilian; Briend, André; Friis, Henrik; Christensen, Vibeke Brix (2014). "The Immune System in Children with Malnutrition—A Systematic Review". PLOS ONE. 9 (8): e105017. Bibcode:2014PLoSO...9j5017R. doi:10.1371/journal.pone.0105017. PMC 4143239. PMID 25153531.
  49. ^ a b Alcock, Joe; Maley, Carlo C; Aktipis, C. Athena (2014). "Is eating behavior manipulated by the gastrointestinal microbiota? Evolutionary pressures and potential mechanisms". BioEssays. 36 (10): 940–9. doi:10.1002/bies.201400071. PMC 4270213. PMID 25103109.
  50. ^ a b c Renson, Audrey; Herd, Pamela; Dowd, Jennifer B. (2020). "Sick Individuals and Sick (Microbial) Populations: Challenges in Epidemiology and the Microbiome". Annual Review of Public Health. 41: 63–80. doi:10.1146/annurev-publhealth-040119-094423. PMC 9713946. PMID 31635533.
  51. ^ Turroni, Francesca; Peano, Clelia; Pass, Daniel A; Foroni, Elena; Severgnini, Marco; Claesson, Marcus J; Kerr, Colm; Hourihane, Jonathan; Murray, Deirdre; Fuligni, Fabio; Gueimonde, Miguel; Margolles, Abelardo; De Bellis, Gianluca; o'Toole, Paul W; Van Sinderen, Douwe; Marchesi, Julian R; Ventura, Marco (2012). "Diversity of Bifidobacteria within the Infant Gut Microbiota". PLOS ONE. 7 (5): e36957. Bibcode:2012PLoSO...736957T. doi:10.1371/journal.pone.0036957. PMC 3350489. PMID 22606315.
  52. ^ Davenport, Emily R.; Sanders, Jon G.; Song, Se Jin; Amato, Katherine R.; Clark, Andrew G.; Knight, Rob (2017-12-27). "The human microbiome in evolution". BMC Biology. 15 (1): 127. doi:10.1186/s12915-017-0454-7. ISSN 1741-7007. PMC 5744394. PMID 29282061.
  53. ^ Perez-Muñoz, Maria Elisa; Arrieta, Marie-Claire; Ramer-Tait, Amanda E; Walter, Jens (2017). "A critical assessment of the 'sterile womb' and 'in utero colonization' hypotheses: Implications for research on the pioneer infant microbiome". Microbiome. 5 (1): 48. doi:10.1186/s40168-017-0268-4. PMC 5410102. PMID 28454555.
  54. ^ Matamoros, Sebastien; Gras-Leguen, Christele; Le Vacon, Françoise; Potel, Gilles; de la Cochetiere, Marie-France (2013). "Development of intestinal microbiota in infants and its impact on health". Trends in Microbiology. 21 (4): 167–73. doi:10.1016/j.tim.2012.12.001. PMID 23332725.
  55. ^ a b c d e f g Mueller, Noel T.; Bakacs, Elizabeth; Combellick, Joan; Grigoryan, Zoya; Dominguez-Bello, Maria G. (2015). "The infant microbiome development: mom matters". Trends in Molecular Medicine. 21 (2): 109–17. doi:10.1016/j.molmed.2014.12.002. PMC 4464665. PMID 25578246.
  56. ^ Jiménez, Esther; Fernández, Leonides; Marín, María L; Martín, Rocío; Odriozola, Juan M; Nueno-Palop, Carmen; Narbad, Arjan; Olivares, Mónica; Xaus, Jordi; Rodríguez, Juan M (2005). "Isolation of Commensal Bacteria from Umbilical Cord Blood of Healthy Neonates Born by Cesarean Section". Current Microbiology. 51 (4): 270–4. doi:10.1007/s00284-005-0020-3. PMID 16187156. S2CID 43438656.
  57. ^ Collado, Maria Carmen; Rautava, Samuli; Aakko, Juhani; Isolauri, Erika; Salminen, Seppo (2016). "Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid". Scientific Reports. 6: 23129. Bibcode:2016NatSR...623129C. doi:10.1038/srep23129. PMC 4802384. PMID 27001291.
  58. ^ Jiménez, Esther; Marín, María L.; Martín, Rocío; Odriozola, Juan M.; Olivares, Mónica; Xaus, Jordi; Fernández, Leonides; Rodríguez, Juan M. (2008). "Is meconium from healthy newborns actually sterile?". Research in Microbiology. 159 (3): 187–93. doi:10.1016/j.resmic.2007.12.007. PMID 18281199.
  59. ^ Perez-Muñoz, Maria Elisa; Arrieta, Marie-Claire; Ramer-Tait, Amanda E; Walter, Jens (2017). "A critical assessment of the "sterile womb" and "in utero colonization" hypotheses: Implications for research on the pioneer infant microbiome". Microbiome. 5 (1): 48. doi:10.1186/s40168-017-0268-4. PMC 5410102. PMID 28454555.
  60. ^ Adlerberth, I; Wold, AE (2009). "Establishment of the gut microbiota in Western infants". Acta Paediatrica. 98 (2): 229–38. doi:10.1111/j.1651-2227.2008.01060.x. PMID 19143664. S2CID 205859933.
  61. ^ "Babies' gut bacteria affected by delivery method: Vaginal delivery promotes mother's gut bacteria in babies' gut". ScienceDaily. 18 September 2019. from the original on 24 November 2021. Retrieved 31 May 2022.
  62. ^ Coppa, G. V.; Zampini, L.; Galeazzi, T.; Gabrielli, O. (2006). "Prebiotics in human milk: A review". Digestive and Liver Disease. 38: S291–4. doi:10.1016/S1590-8658(07)60013-9. PMID 17259094.
  63. ^ Mady, Eman A.; Doghish, Ahmed S.; El-Dakroury, Walaa A.; Elkhawaga, Samy Y.; Ismail, Ahmed; El-Mahdy, Hesham A.; Elsakka, Elsayed G. E.; El-Husseiny, Hussein M. (2023-07-01). "Impact of the mother's gut microbiota on infant microbiome and brain development". Neuroscience & Biobehavioral Reviews. 150: 105195. doi:10.1016/j.neubiorev.2023.105195. ISSN 0149-7634. PMID 37100161. S2CID 258302702.
  64. ^ Fanaro, S; Chierici, R; Guerrini, P; Vigi, V (2007). "Intestinal microflora in early infancy: Composition and development". Acta Paediatrica. 92 (441): 48–55. doi:10.1111/j.1651-2227.2003.tb00646.x. PMID 14599042. S2CID 10316311.
  65. ^ Yassour, Moran; Vatanen, Tommi; Siljander, Heli; Hämäläinen, Anu-Maaria; Härkönen, Taina; Ryhänen, Samppa J; Franzosa, Eric A; Vlamakis, Hera; Huttenhower, Curtis; Gevers, Dirk; Lander, Eric S; Knip, Mikael; Xavier, Ramnik J (2016). "Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability". Science Translational Medicine. 8 (343): 343ra81. doi:10.1126/scitranslmed.aad0917. PMC 5032909. PMID 27306663.
  66. ^ Gibson, G. R.; Roberfroid, M. B. (1995). "Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics". The Journal of Nutrition. 125 (6): 1401–1412. doi:10.1093/jn/125.6.1401. PMID 7782892.
  67. ^ a b c d Wang, Yan; Kasper, Lloyd H (2014). "The role of microbiome in central nervous system disorders". Brain, Behavior, and Immunity. 38: 1–12. doi:10.1016/j.bbi.2013.12.015. PMC 4062078. PMID 24370461.
  68. ^ a b Yoon, My Young; Lee, Keehoon; Yoon, Sang Sun (2014). "Protective role of gut commensal microbes against intestinal infections". Journal of Microbiology. 52 (12): 983–9. doi:10.1007/s12275-014-4655-2. PMID 25467115. S2CID 54622675.
  69. ^ Reinoso Webb, Cynthia; Koboziev, Iurii; Furr, Kathryn L; Grisham, Matthew B (2016). "Protective and pro-inflammatory roles of intestinal bacteria". Pathophysiology. 23 (2): 67–80. doi:10.1016/j.pathophys.2016.02.002. PMC 4867289. PMID 26947707.
  70. ^ Mantis, N J; Rol, N; Corthésy, B (2011). "Secretory IgA's complex roles in immunity and mucosal homeostasis in the gut". Mucosal Immunology. 4 (6): 603–11. doi:10.1038/mi.2011.41. PMC 3774538. PMID 21975936.
  71. ^ a b Peterson, Lance W; Artis, David (2014). "Intestinal epithelial cells: Regulators of barrier function and immune homeostasis". Nature Reviews Immunology. 14 (3): 141–53. doi:10.1038/nri3608. PMID 24566914. S2CID 3351351.
  72. ^ a b Honda, Kenya; Littman, Dan R (2016). "The microbiota in adaptive immune homeostasis and disease". Nature. 535 (7610): 75–84. Bibcode:2016Natur.535...75H. doi:10.1038/nature18848. PMID 27383982. S2CID 4461492.
  73. ^ a b Levy, M.; Thaiss, C.A.; Elinav, E. (2016). "Metabolites: messengers between the microbiota and the immune system". Genes & Development. 30 (14): 1589–97. doi:10.1101/gad.284091.116. PMC 4973288. PMID 27474437.
  74. ^ a b c d e f g h i Zhang LS, Davies SS (April 2016). "Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions". Genome Med. 8 (1): 46. doi:10.1186/s13073-016-0296-x. PMC 4840492. PMID 27102537. Lactobacillus spp. convert tryptophan to indole-3-aldehyde (I3A) through unidentified enzymes [125]. Clostridium sporogenes convert tryptophan to IPA [6], likely via a tryptophan deaminase. ... IPA also potently scavenges hydroxyl radicals
    Table 2: Microbial metabolites: their synthesis, mechanisms of action, and effects on health and disease
    Figure 1: Molecular mechanisms of action of indole and its metabolites on host physiology and disease
  75. ^ Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G (March 2009). "Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites". Proc. Natl. Acad. Sci. U.S.A. 106 (10): 3698–3703. Bibcode:2009PNAS..106.3698W. doi:10.1073/pnas.0812874106. PMC 2656143. PMID 19234110. Production of IPA was shown to be completely dependent on the presence of gut microflora and could be established by colonization with the bacterium Clostridium sporogenes.
    IPA metabolism diagram
  76. ^ "3-Indolepropionic acid". Human Metabolome Database. University of Alberta. Retrieved 12 June 2018.
  77. ^ Chyan YJ, Poeggeler B, Omar RA, Chain DG, Frangione B, Ghiso J, Pappolla MA (July 1999). "Potent neuroprotective properties against the Alzheimer beta-amyloid by an endogenous melatonin-related indole structure, indole-3-propionic acid". J. Biol. Chem. 274 (31): 21937–21942. doi:10.1074/jbc.274.31.21937. PMID 10419516. S2CID 6630247. [Indole-3-propionic acid (IPA)] has previously been identified in the plasma and cerebrospinal fluid of humans, but its functions are not known. ... In kinetic competition experiments using free radical-trapping agents, the capacity of IPA to scavenge hydroxyl radicals exceeded that of melatonin, an indoleamine considered to be the most potent naturally occurring scavenger of free radicals. In contrast with other antioxidants, IPA was not converted to reactive intermediates with pro-oxidant activity.
  78. ^ Hopper, Christopher P.; De La Cruz, Ladie Kimberly; Lyles, Kristin V.; Wareham, Lauren K.; Gilbert, Jack A.; Eichenbaum, Zehava; Magierowski, Marcin; Poole, Robert K.; Wollborn, Jakob; Wang, Binghe (2020-12-23). "Role of Carbon Monoxide in Host–Gut Microbiome Communication". Chemical Reviews. 120 (24): 13273–13311. doi:10.1021/acs.chemrev.0c00586. ISSN 0009-2665. PMID 33089988. S2CID 224824871.
  79. ^ Rajilić-Stojanović, Mirjana; De Vos, Willem M (2014). "The first 1000 cultured species of the human gastrointestinal microbiota". FEMS Microbiology Reviews. 38 (5): 996–1047. doi:10.1111/1574-6976.12075. PMC 4262072. PMID 24861948.
  80. ^ Hill, M. J. (March 1997). "Intestinal flora and endogenous vitamin synthesis". European Journal of Cancer Prevention. 6 (Suppl 1): S43–45. doi:10.1097/00008469-199703001-00009. ISSN 0959-8278. PMID 9167138. S2CID 8740364.
  81. ^ "The Microbiome". Tufts Now. 2013-09-17. Retrieved 2020-12-09.
  82. ^ a b c d e ElRakaiby M, Dutilh BE, Rizkallah MR, Boleij A, Cole JN, Aziz RK (July 2014). "Pharmacomicrobiomics: the impact of human microbiome variations on systems pharmacology and personalized therapeutics". Omics. 18 (7): 402–414. doi:10.1089/omi.2014.0018. PMC 4086029. PMID 24785449.
  83. ^ a b c Cho I, Blaser MJ (March 2012). "The human microbiome: at the interface of health and disease". Nature Reviews. Genetics. 13 (4): 260–270. doi:10.1038/nrg3182. PMC 3418802. PMID 22411464. The composition of the microbiome varies by anatomical site (Figure 1). The primary determinant of community composition is anatomical location: interpersonal variation is substantial23,24 and is higher than the temporal variability seen at most sites in a single individual25.
  84. ^ Hutter T, Gimbert C, Bouchard F, Lapointe FJ (2015). "Being human is a gut feeling". Microbiome. 3: 9. doi:10.1186/s40168-015-0076-7. PMC 4359430. PMID 25774294.
  85. ^ Kumar K, Dhoke GV, Sharma AK, Jaiswal SK, Sharma VK (January 2019). "Mechanistic elucidation of amphetamine metabolism by tyramine oxidase from human gut microbiota using molecular dynamics simulations". Journal of Cellular Biochemistry. 120 (7): 11206–15. doi:10.1002/jcb.28396. PMID 30701587. S2CID 73413138.
  86. ^ Sousa, Tiago; Paterson, Ronnie; Moore, Vanessa; Carlsson, Anders; Abrahamsson, Bertil; Basit, Abdul W (2008). "The gastrointestinal microbiota as a site for the biotransformation of drugs". International Journal of Pharmaceutics. 363 (1–2): 1–25. doi:10.1016/j.ijpharm.2008.07.009. PMID 18682282.
  87. ^ Haiser, H. J; Gootenberg, D. B; Chatman, K; Sirasani, G; Balskus, E. P; Turnbaugh, P. J (2013). "Predicting and Manipulating Cardiac Drug Inactivation by the Human Gut Bacterium Eggerthella lenta". Science. 341 (6143): 295–8. Bibcode:2013Sci...341..295H. doi:10.1126/science.1235872. PMC 3736355. PMID 23869020.
  88. ^ a b Koppel, Nitzan; Maini Rekdal, Vayu; Balskus, Emily P. (2017-06-23). "Chemical transformation of xenobiotics by the human gut microbiota". Science. 356 (6344). doi:10.1126/science.aag2770. ISSN 0036-8075. PMC 5534341. PMID 28642381.
  89. ^ a b c Spanogiannopoulos, Peter; Bess, Elizabeth N.; Carmody, Rachel N.; Turnbaugh, Peter J. (2016-03-14). "The microbial pharmacists within us: a metagenomic view of xenobiotic metabolism". Nature Reviews Microbiology. 14 (5): 273–287. doi:10.1038/nrmicro.2016.17. ISSN 1740-1526. PMC 5243131. PMID 26972811.
  90. ^ a b Maini Rekdal, Vayu; Bess, Elizabeth N.; Bisanz, Jordan E.; Turnbaugh, Peter J.; Balskus, Emily P. (2019-06-14). "Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism". Science. 364 (6445). doi:10.1126/science.aau6323. ISSN 0036-8075. PMC 7745125. PMID 31196984.
  91. ^ a b Dempsey, Joseph L.; Cui, Julia Yue (2019-10-19). "Microbiome Is a Functional Modifier of P450 Drug Metabolism". Current Pharmacology Reports. 5 (6): 481–490. doi:10.1007/s40495-019-00200-w. ISSN 2198-641X. PMC 7731899. PMID 33312848.
  92. ^ Boerner, Udo; Abbott, Seth; Roe, Robert L. (January 1975). "The Metabolism of Morphine and Heroin in Man". Drug Metabolism Reviews. 4 (1): 39–73. doi:10.3109/03602537508993748. ISSN 0360-2532. PMID 1204496.
  93. ^ Dobkin, Jay F.; Saha, Jnan R.; Butler, Vincent P.; Neu, Harold C.; Lindenbaum, John (1983-04-15). "Digoxin-Inactivating Bacteria: Identification in Human Gut Flora". Science. 220 (4594): 325–327. doi:10.1126/science.6836275. ISSN 0036-8075. PMID 6836275.
  94. ^ Sahota, S. S.; Bramley, P. M.; Menzies, I. S. (1982-02-01). "The Fermentation of Lactulose by Colonic Bacteria". Microbiology. 128 (2): 319–325. doi:10.1099/00221287-128-2-319. ISSN 1350-0872. PMID 6804597.
  95. ^ a b Yoo, Dae-Hyoung; Kim, In Sook; Van Le, Thi Kim; Jung, Il-Hoon; Yoo, Hye Hyun; Kim, Dong-Hyun (2014-06-19). "Gut Microbiota-Mediated Drug Interactions between Lovastatin and Antibiotics". Drug Metabolism and Disposition. 42 (9): 1508–1513. doi:10.1124/dmd.114.058354. ISSN 0090-9556. PMID 24947972. S2CID 7524335.
  96. ^ Haiser, Henry J.; Gootenberg, David B.; Chatman, Kelly; Sirasani, Gopal; Balskus, Emily P.; Turnbaugh, Peter J. (2013-07-19). "Predicting and Manipulating Cardiac Drug Inactivation by the Human Gut Bacterium Eggerthella lenta". Science. 341 (6143): 295–298. Bibcode:2013Sci...341..295H. doi:10.1126/science.1235872. ISSN 0036-8075. PMC 3736355. PMID 23869020.
  97. ^ Parvez, Md Masud; Basit, Abdul; Jariwala, Parth B.; Gáborik, Zsuzsanna; Kis, Emese; Heyward, Scott; Redinbo, Matthew R.; Prasad, Bhagwat (2021-06-01). "Quantitative Investigation of Irinotecan Metabolism, Transport, and Gut Microbiome Activation". Drug Metabolism and Disposition. 49 (8): 683–693. doi:10.1124/dmd.121.000476. ISSN 0090-9556. PMC 8407663. PMID 34074730.
  98. ^ a b Sousa, Tiago; Paterson, Ronnie; Moore, Vanessa; Carlsson, Anders; Abrahamsson, Bertil; Basit, Abdul W. (November 2008). "The gastrointestinal microbiota as a site for the biotransformation of drugs". International Journal of Pharmaceutics. 363 (1–2): 1–25. doi:10.1016/j.ijpharm.2008.07.009. ISSN 0378-5173. PMID 18682282.
  99. ^ a b Koppel, Nitzan; Bisanz, Jordan E; Pandelia, Maria-Eirini; Turnbaugh, Peter J; Balskus, Emily P (2018-05-15). Ley, Ruth Emily (ed.). "Discovery and characterization of a prevalent human gut bacterial enzyme sufficient for the inactivation of a family of plant toxins". eLife. 7: e33953. doi:10.7554/eLife.33953. ISSN 2050-084X. PMC 5953540. PMID 29761785.
  100. ^ Alexander, James L.; Wilson, Ian D.; Teare, Julian; Marchesi, Julian R.; Nicholson, Jeremy K.; Kinross, James M. (2017-03-08). "Gut microbiota modulation of chemotherapy efficacy and toxicity". Nature Reviews Gastroenterology & Hepatology. 14 (6): 356–365. doi:10.1038/nrgastro.2017.20. ISSN 1759-5045. PMID 28270698. S2CID 9654170.
  101. ^ Brandi, Giovanni; Dabard, Jean; Raibaud, Pierre; Di Battista, Monica; Bridonneau, Chantal; Pisi, Anna Maria; Morselli Labate, Antonio Maria; Pantaleo, Maria Abbondanza; De Vivo, Antonello; Biasco, Guido (2006-02-15). "Intestinal microflora and digestive toxicity of irinotecan in mice". Clinical Cancer Research. 12 (4): 1299–1307. doi:10.1158/1078-0432.ccr-05-0750. ISSN 1078-0432. PMID 16489087. S2CID 26655779.
  102. ^ Koh, Ara; De Vadder, Filipe; Kovatcheva-Datchary, Petia; Bäckhed, Fredrik (June 2016). "From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites". Cell. 165 (6): 1332–1345. doi:10.1016/j.cell.2016.05.041. ISSN 0092-8674. PMID 27259147. S2CID 8562345.
  103. ^ Konopelski, Piotr; Ufnal, Marcin (2018-09-14). "Indoles - Gut Bacteria Metabolites of Tryptophan with Pharmacotherapeutic Potential". Current Drug Metabolism. 19 (10): 883–890. doi:10.2174/1389200219666180427164731. ISSN 1389-2002. PMID 29708069. S2CID 13979774.
  104. ^ a b Collins, Stephanie L.; Stine, Jonathan G.; Bisanz, Jordan E.; Okafor, C. Denise; Patterson, Andrew D. (2022-10-17). "Bile acids and the gut microbiota: metabolic interactions and impacts on disease". Nature Reviews Microbiology. 21 (4): 236–247. doi:10.1038/s41579-022-00805-x. ISSN 1740-1526. PMID 36253479. S2CID 252970168.
  105. ^ Yang, Wenjing; Yu, Tianming; Huang, Xiangsheng; Bilotta, Anthony J.; Xu, Leiqi; Lu, Yao; Sun, Jiaren; Pan, Fan; Zhou, Jia; Zhang, Wenbo; Yao, Suxia; Maynard, Craig L.; Singh, Nagendra; Dann, Sara M.; Liu, Zhanju (2020-09-08). "Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity". Nature Communications. 11 (1): 4457. Bibcode:2020NatCo..11.4457Y. doi:10.1038/s41467-020-18262-6. ISSN 2041-1723. PMC 7478978. PMID 32901017.
  106. ^ Murugesan, Selvasankar; Nirmalkar, Khemlal; Hoyo-Vadillo, Carlos; García-Espitia, Matilde; Ramírez-Sánchez, Daniela; García-Mena, Jaime (2017-12-02). "Gut microbiome production of short-chain fatty acids and obesity in children". European Journal of Clinical Microbiology & Infectious Diseases. 37 (4): 621–625. doi:10.1007/s10096-017-3143-0. ISSN 0934-9723. PMID 29196878. S2CID 254132108.
  107. ^ a b Thanissery, Rajani; Winston, Jenessa A.; Theriot, Casey M. (June 2017). "Inhibition of spore germination, growth, and toxin activity of clinically relevant C. difficile strains by gut microbiota derived secondary bile acids". Anaerobe. 45: 86–100. doi:10.1016/j.anaerobe.2017.03.004. ISSN 1075-9964. PMC 5466893. PMID 28279860.
  108. ^ a b c Jones, Brian V.; Begley, Máire; Hill, Colin; Gahan, Cormac G. M.; Marchesi, Julian R. (2008-09-09). "Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome". Proceedings of the National Academy of Sciences. 105 (36): 13580–13585. Bibcode:2008PNAS..10513580J. doi:10.1073/pnas.0804437105. ISSN 0027-8424. PMC 2533232. PMID 18757757.
  109. ^ a b Cao H, Xu M, Dong W, Deng B, Wang S, Zhang Y, Wang S, Luo S, Wang W, Qi Y, Gao J, Cao X, Yan F, Wang B (June 2017). "Secondary bile acid-induced dysbiosis promotes intestinal carcinogenesis". International Journal of Cancer. 140 (11): 2545–2556. doi:10.1002/ijc.30643. PMID 28187526.
  110. ^ a b Bernstein H, Bernstein C (January 2023). "Bile acids as carcinogens in the colon and at other sites in the gastrointestinal system". Experimental Biology and Medicine (Maywood, N.J.). 248 (1): 79–89. doi:10.1177/15353702221131858. PMC 9989147. PMID 36408538.
  111. ^ 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): 15490–6. doi:10.1523/JNEUROSCI.3299-14.2014. PMC 4228144. PMID 25392516.
  112. ^ a b c Dinan, Timothy G; Cryan, John F (2015). "The impact of gut microbiota on brain and behaviour". Current Opinion in Clinical Nutrition and Metabolic Care. 18 (6): 552–8. doi:10.1097/MCO.0000000000000221. PMID 26372511. S2CID 21424690.
  113. ^ a b c Carman, Robert J.; Simon, Mary Alice; Fernández, Haydée; Miller, Margaret A.; Bartholomew, Mary J. (2004). "Ciprofloxacin at low levels disrupts colonization resistance of human fecal microflora growing in chemostats". Regulatory Toxicology and Pharmacology. 40 (3): 319–26. doi:10.1016/j.yrtph.2004.08.005. PMID 15546686.
  114. ^ Hvas, Christian Lodberg; Baunwall, Simon Mark Dahl; Erikstrup, Christian (2020-07-01). "Faecal microbiota transplantation: A life-saving therapy challenged by commercial claims for exclusivity". eClinicalMedicine. 24: 100436. doi:10.1016/j.eclinm.2020.100436. ISSN 2589-5370. PMC 7334803. PMID 32642633.
  115. ^ Brandt, Lawrence J.; Borody, Thomas Julius; Campbell, Jordana (2011). "Endoscopic Fecal Microbiota Transplantation". Journal of Clinical Gastroenterology. 45 (8): 655–57. doi:10.1097/MCG.0b013e3182257d4f. PMID 21716124. S2CID 2508836.
  116. ^ Knight, DJW; Girling, KJ (2003). "Gut flora in health and disease". The Lancet. 361 (9371): 512–19. doi:10.1016/S0140-6736(03)13438-1. PMID 12781578. S2CID 40683723.
  117. ^ a b c Cho, I.; Yamanishi, S.; Cox, L.; Methé, B. A.; Zavadil, J.; Li, K.; Gao, Z.; Mahana, D.; Raju, K.; Teitler, I.; Li, H.; Alekseyenko, A. V.; Blaser, M. J. (2012). "Antibiotics in early life alter the murine colonic microbiome and adiposity". Nature. 488 (7413): 621–26. Bibcode:2012Natur.488..621C. doi:10.1038/nature11400. PMC 3553221. PMID 22914093.
  118. ^ a b c Schneiderhan, J; Master-Hunter, T; Locke, A (2016). "Targeting gut flora to treat and prevent disease". The Journal of Family Practice. 65 (1): 34–8. PMID 26845162.
  119. ^ Baker, Monya (2012). "Pregnancy alters resident gut microbes". Nature. doi:10.1038/nature.2012.11118. S2CID 87078157.
  120. ^ Hill, Colin; Guarner, Francisco; Reid, Gregor; Gibson, Glenn R; Merenstein, Daniel J; Pot, Bruno; Morelli, Lorenzo; Canani, Roberto Berni; Flint, Harry J; Salminen, Seppo; Calder, Philip C; Sanders, Mary Ellen (2014). "The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic". Nature Reviews Gastroenterology & Hepatology. 11 (8): 506–14. doi:10.1038/nrgastro.2014.66. PMID 24912386.
  121. ^ Rijkers, Ger T; De Vos, Willem M; Brummer, Robert-Jan; Morelli, Lorenzo; Corthier, Gerard; Marteau, Philippe (2011). "Health benefits and health claims of probiotics: Bridging science and marketing". British Journal of Nutrition. 106 (9): 1291–6. doi:10.1017/S000711451100287X. PMID 21861940.
  122. ^ Hutkins, Robert W; Krumbeck, Janina A; Bindels, Laure B; Cani, Patrice D; Fahey, George; Goh, Yong Jun; Hamaker, Bruce; Martens, Eric C; Mills, David A; Rastal, Robert A; Vaughan, Elaine; Sanders, Mary Ellen (2016). "Prebiotics: Why definitions matter". Current Opinion in Biotechnology. 37: 1–7. doi:10.1016/j.copbio.2015.09.001. PMC 4744122. PMID 26431716.
  123. ^ Pandey, Kavita. R; Naik, Suresh. R; Vakil, Babu. V (2015). "Probiotics, prebiotics and synbiotics- a review". Journal of Food Science and Technology. 52 (12): 7577–87. doi:10.1007/s13197-015-1921-1. PMC 4648921. PMID 26604335.
  124. ^ Broeckx, Géraldine; Vandenheuvel, Dieter; Claes, Ingmar J.J; Lebeer, Sarah; Kiekens, Filip (2016). "Drying techniques of probiotic bacteria as an important step towards the development of novel pharmabiotics" (PDF). International Journal of Pharmaceutics. 505 (1–2): 303–18. doi:10.1016/j.ijpharm.2016.04.002. hdl:10067/1328840151162165141. PMID 27050865.
  125. ^ Sleator, Roy D; Hill, Colin (2009). "Rational Design of Improved Pharmabiotics". Journal of Biomedicine and Biotechnology. 2009: 275287. doi:10.1155/2009/275287. PMC 2742647. PMID 19753318.
  126. ^ Patterson, Elaine; Cryan, John F; Fitzgerald, Gerald F; Ross, R. Paul; Dinan, Timothy G; Stanton, Catherine (2014). "Gut microbiota, the pharmabiotics they produce and host health". Proceedings of the Nutrition Society. 73 (4): 477–89. doi:10.1017/S0029665114001426. PMID 25196939.
  127. ^ Guandalini, Stefano; Magazzù, Giuseppe; Chiaro, Andrea; La Balestra, Valeria; Di Nardo, Giovanni; Gopalan, Sarath; Sibal, A.; Romano, Claudio; Canani, Roberto Berni; Lionetti, Paolo; Setty, Mala (July 2010). "VSL#3 Improves Symptoms in Children with Irritable Bowel Syndrome: A Multicenter, Randomized, Placebo-Controlled, Double-Blind, Crossover Study". Journal of Pediatric Gastroenterology and Nutrition. 51 (1): 24–30. doi:10.1097/MPG.0b013e3181ca4d95. PMID 20453678. S2CID 33659736.
  128. ^ Ford, Alexander C; Quigley, Eamonn M M; Lacy, Brian E; Lembo, Anthony J; Saito, Yuri A; Schiller, Lawrence R; Soffer, Edy E; Spiegel, Brennan M R; Moayyedi, Paul (2014). "Efficacy of Prebiotics, Probiotics and Synbiotics in Irritable Bowel Syndrome and Chronic Idiopathic Constipation: Systematic Review and Meta-analysis". The American Journal of Gastroenterology. 109 (10): 1547–61, quiz 1546, 1562. doi:10.1038/ajg.2014.202. PMID 25070051. S2CID 205100508.
  129. ^ Dupont, Andrew; Richards; Jelinek, Katherine A; Krill, Joseph; Rahimi, Erik; Ghouri, Yezaz (2014). "Systematic review of randomized controlled trials of probiotics, prebiotics, and synbiotics in inflammatory bowel disease". Clinical and Experimental Gastroenterology. 7: 473–87. doi:10.2147/CEG.S27530. PMC 4266241. PMID 25525379.
  130. ^ Yu, Cheng Gong; Huang, Qin (2013). "Recent progress on the role of gut microbiota in the pathogenesis of inflammatory bowel disease". Journal of Digestive Diseases. 14 (10): 513–7. doi:10.1111/1751-2980.12087. PMID 23848393. S2CID 26982085.
  131. ^ Levitt, Michael D.; Duane, William C. (1972-05-04). "Floating Stools — Flatus versus Fat". New England Journal of Medicine. 286 (18): 973–975. doi:10.1056/NEJM197205042861804. ISSN 0028-4793. PMID 5015442.
  132. ^ Aalam, Syed Mohammed Musheer; Crasta, Daphne Norma; Roy, Pooja; Miller, A. Lee; Gamb, Scott I.; Johnson, Stephen; Till, Lisa M.; Chen, Jun; Kashyap, Purna; Kannan, Nagarajan (2022-10-27). "Genesis of fecal floatation is causally linked to gut microbial colonization in mice". Scientific Reports. 12 (1): 18109. Bibcode:2022NatSR..1218109A. doi:10.1038/s41598-022-22626-x. ISSN 2045-2322. PMC 9613883. PMID 36302811.
  133. ^ Maier, Lisa; Pruteanu, Mihaela; Kuhn, Michael; Zeller, Georg; Telzerow, Anja; Anderson, Exene Erin; Brochado, Ana Rita; Fernandez, Keith Conrad; Dose, Hitomi; Mori, Hirotada; Patil, Kiran Raosaheb; Bork, Peer; Typas, Athanasios (2018). "Extensive impact of non-antibiotic drugs on human gut bacteria". Nature. 555 (7698): 623–628. Bibcode:2018Natur.555..623M. doi:10.1038/nature25979. PMC 6108420. PMID 29555994.
  134. ^ Clauss, Matthieu; Gérard, Philippe; Mosca, Alexis; Leclerc, Marion (2021). "Interplay Between Exercise and Gut Microbiome in the Context of Human Health and Performance". Frontiers in Nutrition. 8: 637010. doi:10.3389/fnut.2021.637010. PMC 8222532. PMID 34179053.
  135. ^ a b Kamboj, AK; Cotter, TG; Oxentenko, AS (2017). "Helicobacter pylori: The Past, Present, and Future in Management". Mayo Clinic Proceedings. 92 (4): 599–604. doi:10.1016/j.mayocp.2016.11.017. ISSN 0025-6196. PMID 28209367.
  136. ^ a b c "Peptic ulcer disease" (PDF). The Johns Hopkins University School of Medicine. 2013. Retrieved 21 October 2020.
  137. ^ Burisch, Johan; Jess, Tine; Martinato, Matteo; Lakatos, Peter L (2013). "The burden of inflammatory bowel disease in Europe". Journal of Crohn's and Colitis. 7 (4): 322–37. doi:10.1016/j.crohns.2013.01.010. PMID 23395397.
  138. ^ Blandino, G; Inturri, R; Lazzara, F; Di Rosa, M; Malaguarnera, L (2016). "Impact of gut microbiota on diabetes mellitus". Diabetes & Metabolism. 42 (5): 303–315. doi:10.1016/j.diabet.2016.04.004. PMID 27179626.
  139. ^ a b c Boulangé, Claire L; Neves, Ana Luisa; Chilloux, Julien; Nicholson, Jeremy K; Dumas, Marc-Emmanuel (2016). "Impact of the gut microbiota on inflammation, obesity, and metabolic disease". Genome Medicine. 8 (1): 42. doi:10.1186/s13073-016-0303-2. PMC 4839080. PMID 27098727.
  140. ^ a b c Arrieta, Marie-Claire; Stiemsma, Leah T; Dimitriu, Pedro A; Thorson, Lisa; Russell, Shannon; Yurist-Doutsch, Sophie; Kuzeljevic, Boris; Gold, Matthew J; Britton, Heidi M; Lefebvre, Diana L; Subbarao, Padmaja; Mandhane, Piush; Becker, Allan; McNagny, Kelly M; Sears, Malcolm R; Kollmann, Tobias; Mohn, William W; Turvey, Stuart E; Brett Finlay, B (2015). "Early infancy microbial and metabolic alterations affect risk of childhood asthma". Science Translational Medicine. 7 (307): 307ra152. doi:10.1126/scitranslmed.aab2271. PMID 26424567. S2CID 206687974.
  141. ^ a b Stiemsma, Leah T; Turvey, Stuart E (2017). "Asthma and the microbiome: Defining the critical window in early life". Allergy, Asthma & Clinical Immunology. 13: 3. doi:10.1186/s13223-016-0173-6. PMC 5217603. PMID 28077947.
  142. ^ Ipci, Kagan; Altıntoprak, Niyazi; Muluk, Nuray Bayar; Senturk, Mehmet; Cingi, Cemal (2016). "The possible mechanisms of the human microbiome in allergic diseases". European Archives of Oto-Rhino-Laryngology. 274 (2): 617–626. doi:10.1007/s00405-016-4058-6. PMID 27115907. S2CID 27328940.
  143. ^ Mariño, Eliana; Richards, James L.; McLeod, Keiran H.; et al. (May 2017). "Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes". Nature Immunology. 18 (5): 552–562. doi:10.1038/ni.3713. ISSN 1529-2916. PMID 28346408. S2CID 30078908.
  144. ^ Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK (May 2006). "Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells". Nature. 441 (7090): 235–8. Bibcode:2006Natur.441..235B. doi:10.1038/nature04753. PMID 16648838.
  145. ^ a b Säemann, Marcus D.; Böhmig, Georg A.; Österreicher, Christoph H.; et al. (December 2000). "Anti‐inflammatory effects of sodium butyrate on human monocytes: potent inhibition of IL‐12 and up‐regulation of IL‐10 production". The FASEB Journal. 14 (15): 2380–2382. doi:10.1096/fj.00-0359fje. ISSN 0892-6638. PMID 11024006. S2CID 41553220.
  146. ^ Gao, Z; Yin, J; Zhang, J; Ward, R. E; Martin, R. J; Lefevre, M; Cefalu, W. T; Ye, J (2009). "Butyrate Improves Insulin Sensitivity and Increases Energy Expenditure in Mice". Diabetes. 58 (7): 1509–17. doi:10.2337/db08-1637. PMC 2699871. PMID 19366864.
  147. ^ Mazidi, Mohsen; Rezaie, Peyman; Kengne, Andre Pascal; Mobarhan, Majid Ghayour; Ferns, Gordon A (2016). "Gut microbiome and metabolic syndrome". Diabetes & Metabolic Syndrome: Clinical Research & Reviews. 10 (2): S150–7. doi:10.1016/j.dsx.2016.01.024. PMID 26916014.
  148. ^ Chen, Xiao; d'Souza, Roshan; Hong, Seong-Tshool (2013). "The role of gut microbiota in the gut-brain axis: Current challenges and perspectives". Protein & Cell. 4 (6): 403–14. doi:10.1007/s13238-013-3017-x. PMC 4875553. PMID 23686721.
  149. ^ a b Moeller, Andrew H.; Li, Yingying; Ngole, Eitel Mpoudi; Ahuka-Mundeke, Steve; Lonsdorf, Elizabeth V.; Pusey, Anne E.; Peeters, Martine; Hahn, Beatrice H.; Ochman, Howard (2014-11-18). "Rapid changes in the gut microbiome during human evolution". Proceedings of the National Academy of Sciences. 111 (46): 16431–16435. Bibcode:2014PNAS..11116431M. doi:10.1073/pnas.1419136111. ISSN 0027-8424. PMC 4246287. PMID 25368157.
  150. ^ Brune, A. (2014). "Symbiotic digestion of lignocellulose in termite guts". Nature Reviews Microbiology. 12 (3): 168–80. doi:10.1038/nrmicro3182. PMID 24487819. S2CID 5220210.
  151. ^ a b Dietrich, C.; Köhler, T.; Brune, A. (2014). "The cockroach origin of the termite gut microbiota: patterns in bacterial community structure reflect major evolutionary events". Applied and Environmental Microbiology. 80 (7): 2261–69. Bibcode:2014ApEnM..80.2261D. doi:10.1128/AEM.04206-13. PMC 3993134. PMID 24487532.
  152. ^ a b Mikaelyan, A.; Dietrich, C.; Köhler, T.; Poulsen, M.; Sillam-Dussès, D.; Brune, A. (2015). "Diet is the primary determinant of bacterial community structure in the guts of higher termites". Molecular Ecology. 24 (20): 5824–95. Bibcode:2015MolEc..24.5284M. doi:10.1111/mec.13376. PMID 26348261. S2CID 206182668.
  153. ^ Mikaelyan, A.; Thompson, C.; Hofer, M.; Brune, A. (2016). "The deterministic assembly of complex bacterial communities in germ-free cockroach guts". Applied and Environmental Microbiology. 82 (4): 1256–63. doi:10.1128/AEM.03700-15. PMC 4751828. PMID 26655763.

Further reading edit

Review articles
  • De Preter, Vicky; Hamer, Henrike M; Windey, Karen; Verbeke, Kristin (2011). "The impact of pre- and/or probiotics on human colonic metabolism: Does it affect human health?". Molecular Nutrition & Food Research. 55 (1): 46–57. doi:10.1002/mnfr.201000451. PMID 21207512.
  • Maranduba, Carlos Magno da Costa; De Castro, Sandra Bertelli Ribeiro; Souza, Gustavo Torres de; Rossato, Cristiano; Da Guia, Francisco Carlos; Valente, Maria Anete Santana; Rettore, João Vitor Paes; Maranduba, Claudinéia Pereira; Souza, Camila Maurmann de; Carmo, Antônio Márcio Resende do; MacEdo, Gilson Costa; Silva, Fernando de Sá (2015). "Intestinal Microbiota as Modulators of the Immune System and Neuroimmune System: Impact on the Host Health and Homeostasis". Journal of Immunology Research. 2015: 931574. doi:10.1155/2015/931574. PMC 4352473. PMID 25759850.
  • Prakash, Satya; Rodes, Laetitia; Coussa-Charley, Michael; Tomaro-Duchesneau, Catherine; Tomaro-Duchesneau, Catherine; Coussa-Charley; Rodes (2011). "Gut microbiota: Next frontier in understanding human health and development of biotherapeutics". Biologics: Targets and Therapy. 5: 71–86. doi:10.2147/BTT.S19099. PMC 3156250. PMID 21847343.
  • Wu, G. D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.-Y.; Keilbaugh, S. A.; Bewtra, M.; Knights, D.; Walters, W. A.; Knight, R.; Sinha, R.; Gilroy, E.; Gupta, K.; Baldassano, R.; Nessel, L.; Li, H.; Bushman, F. D.; Lewis, J.D. (2011). "Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes". Science. 334 (6052): 105–08. Bibcode:2011Sci...334..105W. doi:10.1126/science.1208344. PMC 3368382. PMID 21885731.

microbiota, enteric, bacteria, redirects, here, other, uses, enteric, bacteria, disambiguation, microbiome, flora, microorganisms, including, bacteria, archaea, fungi, viruses, that, live, digestive, tracts, animals, gastrointestinal, metagenome, aggregate, ge. Enteric bacteria redirects here For other uses see Enteric bacteria disambiguation Gut microbiota gut microbiome or gut flora are the microorganisms including bacteria archaea fungi and viruses that live in the digestive tracts of animals 1 2 The gastrointestinal metagenome is the aggregate of all the genomes of the gut microbiota 3 4 The gut is the main location of the human microbiome 5 The gut microbiota has broad impacts including effects on colonization resistance to pathogens maintaining the intestinal epithelium metabolizing dietary and pharmaceutical compounds controlling immune function and even behavior through the gut brain axis Escherichia coli one of the many species of bacteria present in the human gutThe microbial composition of the gut microbiota varies across regions of the digestive tract The colon contains the highest microbial density of any human associated microbial community studied so far representing between 300 and 1000 different species 6 Bacteria are the largest and to date best studied component and 99 of gut bacteria come from about 30 or 40 species 7 Up to 60 of the dry mass of feces is bacteria 8 Over 99 of the bacteria in the gut are anaerobes but in the cecum aerobic bacteria reach high densities 5 It is estimated that the human gut microbiota have around a hundred times as many genes as there are in the human genome Contents 1 Overview 2 Classifications 2 1 Enterotype 3 Composition 3 1 Bacteriome 3 1 1 Stomach 3 1 2 Intestines 3 2 Mycobiome 3 3 Virome 4 Variation 4 1 Age 4 2 Geography 4 2 1 Malnourishment 4 3 Race and ethnicity 4 4 Socioeconomic status 5 Acquisition in human infants 6 Functions 6 1 Direct inhibition of pathogens 6 2 Development of enteric protection and immune system 6 3 Metabolism 6 3 1 Pharmacomicrobiomics 6 3 1 1 Contribution to drug metabolism 6 3 1 2 Secondary metabolites 6 3 2 Dysbiosis 6 4 Gut brain axis 7 Alterations in microbiota balance 7 1 Effects of antibiotic use 7 2 Pregnancy 7 3 Probiotics prebiotics synbiotics and pharmabiotics 7 4 Fecal floatation 7 5 Research 7 6 Effects of exercise 8 Role in disease 8 1 Ulcers 8 2 Bowel perforation 8 3 Inflammatory bowel diseases 8 4 Irritable bowel syndrome 8 4 1 Asthma 8 4 2 Diabetes mellitus type 1 8 5 Obesity and metabolic syndrome 9 Other animals 10 See also 11 Notes 12 References 13 Further readingOverview edit nbsp Composition and distribution of gut microbiota in human body In humans the gut microbiota has the largest numbers and species of bacteria compared to other areas of the body 9 The approximate number of bacteria composing the gut microbiota is about 1013 1014 10 In humans the gut flora is established at one to two years after birth by which time the intestinal epithelium and the intestinal mucosal barrier that it secretes have co developed in a way that is tolerant to and even supportive of the gut flora and that also provides a barrier to pathogenic organisms 11 12 The relationship between some gut microbiota and humans is not merely commensal a non harmful coexistence but rather a mutualistic relationship 5 700 Some human gut microorganisms benefit the host by fermenting dietary fiber into short chain fatty acids SCFAs such as acetic acid and butyric acid which are then absorbed by the host 9 13 Intestinal bacteria also play a role in synthesizing vitamin B and vitamin K as well as metabolizing bile acids sterols and xenobiotics 5 13 The systemic importance of the SCFAs and other compounds they produce are like hormones and the gut flora itself appears to function like an endocrine organ 13 Dysregulation of the gut flora has been correlated with a host of inflammatory and autoimmune conditions 9 14 The composition of human gut microbiota changes over time when the diet changes and as overall health changes 9 14 A systematic review from 2016 examined the preclinical and small human trials that have been conducted with certain commercially available strains of probiotic bacteria and identified those that had the most potential to be useful for certain central nervous system disorders 15 Classifications editThe microbial composition of the gut microbiota varies across the digestive tract In the stomach and small intestine relatively few species of bacteria are generally present 6 16 The colon in contrast contains the highest microbial density of any human associated microbial community studied so far 17 with between 1010 and 1011 cells per gram of intestinal content 18 These bacteria represent between 300 and 1000 different species 6 16 However 99 of the bacteria come from about 30 or 40 species 7 As a consequence of their abundance in the intestine bacteria also make up to 60 of the dry mass of feces 8 Fungi protists archaea and viruses are also present in the gut flora but less is known about their activities 19 Over 99 of the bacteria in the gut are anaerobes but in the cecum aerobic bacteria reach high densities 5 It is estimated that these gut flora have around a hundred times as many genes in total as there are in the human genome 20 nbsp Candida albicans a dimorphic fungus that grows as a yeast in the gutMany species in the gut have not been studied outside of their hosts because they cannot be cultured 16 7 21 While there are a small number of core microbial species shared by most individuals populations of microbes can vary widely 22 Within an individual their microbial populations stay fairly constant over time with some alterations occurring due to changes in lifestyle diet and age 6 23 The Human Microbiome Project has set out to better describe the microbiota of the human gut and other body locations citation needed The four dominant bacterial phyla in the human gut are Bacillota Firmicutes Bacteroidota Actinomycetota and Pseudomonadota 24 Most bacteria belong to the genera Bacteroides Clostridium Faecalibacterium 6 7 Eubacterium Ruminococcus Peptococcus Peptostreptococcus and Bifidobacterium 6 7 Other genera such as Escherichia and Lactobacillus are present to a lesser extent 6 Species from the genus Bacteroides alone constitute about 30 of all bacteria in the gut suggesting that this genus is especially important in the functioning of the host 16 Fungal genera that have been detected in the gut include Candida Saccharomyces Aspergillus Penicillium Rhodotorula Trametes Pleospora Sclerotinia Bullera and Galactomyces among others 25 26 Rhodotorula is most frequently found in individuals with inflammatory bowel disease while Candida is most frequently found in individuals with hepatitis B cirrhosis and chronic hepatitis B 25 Archaea constitute another large class of gut flora which are important in the metabolism of the bacterial products of fermentation Industrialization is associated with changes in the microbiota and the reduction of diversity could drive certain species to extinction in 2018 researchers proposed a biobank repository of human microbiota 27 Enterotype edit An enterotype is a classification of living organisms based on its bacteriological ecosystem in the human gut microbiome not dictated by age gender body weight or national divisions 28 There are indications that long term diet influences enterotype 29 Three human enterotypes have been proposed 28 30 but their value has been questioned 31 Composition editSee also Human microbiome Gastrointestinal tract Bacteriome edit Stomach edit Due to the high acidity of the stomach most microorganisms cannot survive there The main bacterial inhabitants of the stomach include Streptococcus Staphylococcus Lactobacillus Peptostreptococcus 5 720 Helicobacter pylori is a gram negative spiral bacterium that establishes on gastric mucosa causing chronic gastritis and peptic ulcer disease and is a carcinogen for gastric cancer 5 904 Intestines edit Bacteria commonly found in the human colon 32 Bacterium Incidence Bacteroides fragilis 100Bacteroides melaninogenicus 100Bacteroides oralis 100Enterococcus faecalis 100Escherichia coli 100Enterobacter sp 40 80Klebsiella sp 40 80Bifidobacterium bifidum 30 70Staphylococcus aureus 30 50Lactobacillus 20 60Clostridium perfringens 25 35Proteus mirabilis 5 55Clostridium tetani 1 35Clostridium septicum 5 25Pseudomonas aeruginosa 3 11Salmonella enterica 3 7Faecalibacterium prausnitzii commonPeptostreptococcus sp commonPeptococcus sp commonThe small intestine contains a trace amount of microorganisms due to the proximity and influence of the stomach Gram positive cocci and rod shaped bacteria are the predominant microorganisms found in the small intestine 5 However in the distal portion of the small intestine alkaline conditions support gram negative bacteria of the Enterobacteriaceae 5 The bacterial flora of the small intestine aid in a wide range of intestinal functions The bacterial flora provide regulatory signals that enable the development and utility of the gut Overgrowth of bacteria in the small intestine can lead to intestinal failure 33 In addition the large intestine contains the largest bacterial ecosystem in the human body 5 About 99 of the large intestine and feces flora are made up of obligate anaerobes such as Bacteroides and Bifidobacterium 34 Factors that disrupt the microorganism population of the large intestine include antibiotics stress and parasites 5 Bacteria make up most of the flora in the colon 35 and 60 of the dry mass of feces 6 This fact makes feces an ideal source of gut flora for any tests and experiments by extracting the nucleic acid from fecal specimens and bacterial 16S rRNA gene sequences are generated with bacterial primers This form of testing is also often preferable to more invasive techniques such as biopsies Five phyla dominate the intestinal microbiota Bacteroidota Bacillota Firmicutes Actinomycetota Pseudomonadota and Verrucomicrobiota with Bacteroidota and Bacillota constituting 90 of the composition 36 Somewhere between 300 6 and 1000 different species live in the gut 16 with most estimates at about 500 37 38 However it is probable that 99 of the bacteria come from about 30 or 40 species with Faecalibacterium prausnitzii phylum firmicutes being the most common species in healthy adults 7 39 Research suggests that the relationship between gut flora and humans is not merely commensal a non harmful coexistence but rather is a mutualistic symbiotic relationship 16 Though people can survive with no gut flora 37 the microorganisms perform a host of useful functions such as fermenting unused energy substrates training the immune system via end products of metabolism like propionate and acetate preventing growth of harmful species regulating the development of the gut producing vitamins for the host such as biotin and vitamin K and producing hormones to direct the host to store fats 5 Extensive modification and imbalances of the gut microbiota and its microbiome or gene collection are associated with obesity 40 However in certain conditions some species are thought to be capable of causing disease by causing infection or increasing cancer risk for the host 6 35 Mycobiome edit Further information Mycobiome Fungi and protists also make up a part of the gut flora but less is known about their activities 41 Virome edit Further information Virome The human virome is mostly bacteriophages 42 Variation editAge edit It has been demonstrated that there are common patterns of microbiome composition evolution during life 43 In general the diversity of microbiota composition of fecal samples is significantly higher in adults than in children although interpersonal differences are higher in children than in adults 44 Much of the maturation of microbiota into an adult like configuration happens during the three first years of life 44 As the microbiome composition changes so does the composition of bacterial proteins produced in the gut In adult microbiomes a high prevalence of enzymes involved in fermentation methanogenesis and the metabolism of arginine glutamate aspartate and lysine have been found In contrast in infant microbiomes the dominant enzymes are involved in cysteine metabolism and fermentation pathways 44 Geography edit Gut microbiome composition depends on the geographic origin of populations Variations in a trade off of Prevotella the representation of the urease gene and the representation of genes encoding glutamate synthase degradation or other enzymes involved in amino acids degradation or vitamin biosynthesis show significant differences between populations from the US Malawi or Amerindian origin 44 The US population has a high representation of enzymes encoding the degradation of glutamine and enzymes involved in vitamin and lipoic acid biosynthesis whereas Malawi and Amerindian populations have a high representation of enzymes encoding glutamate synthase and they also have an overrepresentation of a amylase in their microbiomes As the US population has a diet richer in fats than Amerindian or Malawian populations which have a corn rich diet the diet is probably the main determinant of the gut bacterial composition 44 Further studies have indicated a large difference in the composition of microbiota between European and rural African children The fecal bacteria of children from Florence were compared to that of children from the small rural village of Boulpon in Burkina Faso The diet of a typical child living in this village is largely lacking in fats and animal proteins and rich in polysaccharides and plant proteins The fecal bacteria of European children were dominated by Firmicutes and showed a marked reduction in biodiversity while the fecal bacteria of the Boulpon children was dominated by Bacteroidetes The increased biodiversity and different composition of the gut microbiome in African populations may aid in the digestion of normally indigestible plant polysaccharides and also may result in a reduced incidence of non infectious colonic diseases 45 On a smaller scale it has been shown that sharing numerous common environmental exposures in a family is a strong determinant of individual microbiome composition This effect has no genetic influence and it is consistently observed in culturally different populations 44 Malnourishment edit Malnourished children have less mature and less diverse gut microbiota than healthy children and changes in the microbiome associated with nutrient scarcity can in turn be a pathophysiological cause of malnutrition 46 47 Malnourished children also typically have more potentially pathogenic gut flora and more yeast in their mouths and throats 48 Altering diet may lead to changes in gut microbiota composition and diversity 49 Race and ethnicity edit Researchers with the American Gut Project and Human Microbiome Project found that twelve microbe families varied in abundance based on the race or ethnicity of the individual The strength of these associations is limited by the small sample size the American Gut Project collected data from 1 375 individuals 90 of whom were white 50 The Healthy Life in an Urban Setting HELIUS study in Amsterdam found that those of Dutch ancestry had the highest level of gut microbiota diversity while those of South Asian and Surinamese descent had the lowest diversity The study results suggested that individuals of the same race or ethnicity have more similar microbiomes than individuals of different racial backgrounds 50 Socioeconomic status edit As of 2020 at least two studies have demonstrated a link between an individual s socioeconomic status SES and their gut microbiota A study in Chicago found that individuals in higher SES neighborhoods had greater microbiota diversity People from higher SES neighborhoods also had more abundant Bacteroides bacteria Similarly a study of twins in the United Kingdom found that higher SES was also linked with a greater gut diversity 50 Acquisition in human infants editThe establishment of a gut flora is crucial to the health of an adult as well as the functioning of the gastrointestinal tract 51 In humans a gut flora similar to an adult s is formed within one to two years of birth as microbiota are acquired through parent to child transmission and transfer from food water and other environmental sources 52 11 nbsp Illustration showing the developmental colonization of gut microbiota The traditional view of the gastrointestinal tract of a normal fetus is that it is sterile although this view has been challenged in the past few years timeframe 53 Multiple lines of evidence have begun to emerge that suggest there may be bacteria in the intrauterine environment In humans research has shown that microbial colonization may occur in the fetus 54 with one study showing Lactobacillus and Bifidobacterium species were present in placental biopsies 55 Several rodent studies have demonstrated the presence of bacteria in the amniotic fluid and placenta as well as in the meconium of babies born by sterile cesarean section 56 57 In another study researchers administered a culture of bacteria orally to pregnant mice and detected the bacteria in the offspring likely resulting from transmission between the digestive tract and amniotic fluid via the blood stream 58 However researchers caution that the source of these intrauterine bacteria whether they are alive and their role is not yet understood 59 55 During birth and rapidly thereafter bacteria from the mother and the surrounding environment colonize the infant s gut 11 The exact sources of bacteria are not fully understood but may include the birth canal other people parents siblings hospital workers breastmilk food and the general environment with which the infant interacts 60 Research has shown that the microbiome of babies born vaginally differs significantly from that of babies delivered by caesarean section and that vaginally born babies got most of their gut bacteria from their mother while the microbiota of babies born by caesarean section had more bacteria associated with hospital environments 61 During the first year of life the composition of the gut flora is generally simple and changes a great deal with time and is not the same across individuals 11 The initial bacterial population are generally facultative anaerobic organisms investigators believe that these initial colonizers decrease the oxygen concentration in the gut which in turn allows obligately anaerobic bacteria like Bacteroidota Actinomycetota and Bacillota to become established and thrive 11 Breast fed babies become dominated by bifidobacteria possibly due to the contents of bifidobacterial growth factors in breast milk and by the fact that breast milk carries prebiotic components allowing for healthy bacterial growth 55 62 Breast milk also contains higher levels of Immunoglobulin A IgA to help with the tolerance and regulation of the baby s immune system 63 In contrast the microbiota of formula fed infants is more diverse with high numbers of Enterobacteriaceae enterococci bifidobacteria Bacteroides and clostridia 64 Caesarean section antibiotics and formula feeding may alter the gut microbiome composition 55 Children treated with antibiotics have less stable and less diverse floral communities 65 Caesarean sections have been shown to be disruptive to mother offspring transmission of bacteria which impacts the overall health of the offspring by raising risks of disease such as celiac disease asthma and type 1 diabetes 55 This further evidences the importance of a healthy gut microbiome Various methods of microbiome restoration are being explored typically involving exposing the infant to maternal vaginal contents and oral probiotics 55 Functions editWhen the study of gut flora began in 1995 66 it was thought to have three key roles direct defense against pathogens fortification of host defense by its role in developing and maintaining the intestinal epithelium and inducing antibody production there and metabolizing otherwise indigestible compounds in food Subsequent work discovered its role in training the developing immune system and yet further work focused on its role in the gut brain axis 67 Direct inhibition of pathogens edit The gut flora community plays a direct role in defending against pathogens by fully colonising the space making use of all available nutrients and by secreting compounds known as cytokines that kill or inhibit unwelcome organisms that would compete for nutrients with it 68 Different strains of gut bacteria cause the production of different cytokines Cytokines are chemical compounds produced by our immune system for initiating the inflammatory response against infections Disruption of the gut flora allows competing organisms like Clostridium difficile to become established that otherwise are kept in abeyance 68 Development of enteric protection and immune system edit nbsp Microfold cells transfer antigens Ag from the lumen of the gut to gut associated lymphoid tissue GALT via transcytosis and present them to different innate and adaptive immune cells In humans a gut flora similar to an adult s is formed within one to two years of birth 11 As the gut flora gets established the lining of the intestines the intestinal epithelium and the intestinal mucosal barrier that it secretes develop as well in a way that is tolerant to and even supportive of commensalistic microorganisms to a certain extent and also provides a barrier to pathogenic ones 11 Specifically goblet cells that produce the mucosa proliferate and the mucosa layer thickens providing an outside mucosal layer in which friendly microorganisms can anchor and feed and an inner layer that even these organisms cannot penetrate 11 12 Additionally the development of gut associated lymphoid tissue GALT which forms part of the intestinal epithelium and which detects and reacts to pathogens appears and develops during the time that the gut flora develops and established 11 The GALT that develops is tolerant to gut flora species but not to other microorganisms 11 GALT also normally becomes tolerant to food to which the infant is exposed as well as digestive products of food and gut flora s metabolites molecules formed from metabolism produced from food 11 The human immune system creates cytokines that can drive the immune system to produce inflammation in order to protect itself and that can tamp down the immune response to maintain homeostasis and allow healing after insult or injury 11 Different bacterial species that appear in gut flora have been shown to be able to drive the immune system to create cytokines selectively for example Bacteroides fragilis and some Clostridia species appear to drive an anti inflammatory response while some segmented filamentous bacteria drive the production of inflammatory cytokines 11 69 Gut flora can also regulate the production of antibodies by the immune system 11 70 One function of this regulation is to cause B cells to class switch to IgA In most cases B cells need activation from T helper cells to induce class switching however in another pathway gut flora cause NF kB signaling by intestinal epithelial cells which results in further signaling molecules being secreted 71 These signaling molecules interact with B cells to induce class switching to IgA 71 IgA is an important type of antibody that is used in mucosal environments like the gut It has been shown that IgA can help diversify the gut community and helps in getting rid of bacteria that cause inflammatory responses 72 Ultimately IgA maintains a healthy environment between the host and gut bacteria 72 These cytokines and antibodies can have effects outside the gut in the lungs and other tissues 11 The immune system can also be altered due to the gut bacteria s ability to produce metabolites that can affect cells in the immune system For example short chain fatty acids SCFA can be produced by some gut bacteria through fermentation 73 SCFAs stimulate a rapid increase in the production of innate immune cells like neutrophils basophils and eosinophils 73 These cells are part of the innate immune system that try to limit the spread of infection Metabolism edit Tryptophan metabolism by human gastrointestinal microbiota vte nbsp Tryptophan Clostridiumsporogenes Lacto bacilli Tryptophanase expressingbacteria IPA I3A Indole Liver Brain IPA I3A Indole Indoxylsulfate AST 120 AhR Intestinalimmunecells Intestinalepithelium PXR Mucosal homeostasis TNF a Junction protein coding mRNAs L cell GLP 1 T J Neuroprotectant Activation of glial cells and astrocytes 4 Hydroxy 2 nonenal levels DNA damage Antioxidant Inhibits b amyloid fibril formation Maintains mucosal reactivity IL 22 production Associated with vascular disease Oxidative stress Smooth muscle cell proliferation Aortic wall thickness and calcification Associated with chronic kidney disease Renal dysfunction Uremic toxin Kidneys nbsp This diagram shows the biosynthesis of bioactive compounds indole and certain other derivatives from tryptophan by bacteria in the gut 74 Indole is produced from tryptophan by bacteria that express tryptophanase 74 Clostridium sporogenes metabolizes tryptophan into indole and subsequently 3 indolepropionic acid IPA 75 a highly potent neuroprotective antioxidant that scavenges hydroxyl radicals 74 76 77 IPA binds to the pregnane X receptor PXR in intestinal cells thereby facilitating mucosal homeostasis and barrier function 74 Following absorption from the intestine and distribution to the brain IPA confers a neuroprotective effect against cerebral ischemia and Alzheimer s disease 74 Lactobacillaceae Lactobacillus s l species metabolize tryptophan into indole 3 aldehyde I3A which acts on the aryl hydrocarbon receptor AhR in intestinal immune cells in turn increasing interleukin 22 IL 22 production 74 Indole itself triggers the secretion of glucagon like peptide 1 GLP 1 in intestinal L cells and acts as a ligand for AhR 74 Indole can also be metabolized by the liver into indoxyl sulfate a compound that is toxic in high concentrations and associated with vascular disease and renal dysfunction 74 AST 120 activated charcoal an intestinal sorbent that is taken by mouth adsorbs indole in turn decreasing the concentration of indoxyl sulfate in blood plasma 74 Without gut flora the human body would be unable to utilize some of the undigested carbohydrates it consumes because some types of gut flora have enzymes that human cells lack for breaking down certain polysaccharides 13 Rodents raised in a sterile environment and lacking in gut flora need to eat 30 more calories just to remain the same weight as their normal counterparts 13 Carbohydrates that humans cannot digest without bacterial help include certain starches fiber oligosaccharides and sugars that the body failed to digest and absorb like lactose in the case of lactose intolerance and sugar alcohols mucus produced by the gut and proteins 9 13 Bacteria turn carbohydrates they ferment into short chain fatty acids by a form of fermentation called saccharolytic fermentation 38 Products include acetic acid propionic acid and butyric acid 7 38 These materials can be used by host cells providing a major source of energy and nutrients 38 Gases which are involved in signaling 78 and may cause flatulence and organic acids such as lactic acid are also produced by fermentation 7 Acetic acid is used by muscle propionic acid facilitates liver production of ATP and butyric acid provides energy to gut cells 38 Gut flora also synthesize vitamins like biotin and folate and facilitate absorption of dietary minerals including magnesium calcium and iron 6 23 Methanobrevibacter smithii is unique because it is not a species of bacteria but rather a member of domain Archaea and is the most abundant methane producing archaeal species in the human gastrointestinal microbiota 79 Gut microbiota also serve as a source of Vitamins K and B12 that are not produced by the body or produced in little amount 80 81 Pharmacomicrobiomics edit The human metagenome i e the genetic composition of an individual and all microorganisms that reside on or within the individual s body varies considerably between individuals 82 83 Since the total number of microbial cells in the human body over 100 trillion greatly outnumbers Homo sapiens cells tens of trillions note 1 82 84 there is considerable potential for interactions between drugs and an individual s microbiome including drugs altering the composition of the human microbiome drug metabolism by microbial enzymes modifying the drug s pharmacokinetic profile and microbial drug metabolism affecting a drug s clinical efficacy and toxicity profile 82 83 85 Apart from carbohydrates gut microbiota can also metabolize other xenobiotics such as drugs phytochemicals and food toxicants More than 30 drugs have been shown to be metabolized by gut microbiota 86 The microbial metabolism of drugs can sometimes inactivate the drug 87 Contribution to drug metabolism edit The gut microbiota is an enriched community that contains diverse genes with huge biochemical capabilities to modify drugs especially those taken by mouth 88 Gut microbiota can affect drug metabolism via direct and indirect mechanisms 89 The direct mechanism is mediated by the microbial enzymes that can modify the chemical structure of the administered drugs 90 Conversely the indirect pathway is mediated by the microbial metabolites which affect the expression of host metabolizing enzymes such as cytochrome P450 91 89 The effects of the gut microbiota on the pharmacokinetics and bioavailability of the drug have been investigated a few decades ago 92 93 94 These effects can be varied it could activate the inactive drugs such as lovastatin 95 inactivate the active drug such as digoxin 96 or induce drug toxicity as in irinotecan 97 Since then the impacts of the gut microbiota on the pharmacokinetics of many drugs were heavily studied 98 88 The human gut microbiota plays a crucial role in modulating the effect of the administered drugs on the human Directly gut microbiota can synthesize and release a series of enzymes with the capability to metabolize drugs such as microbial biotransformation of L dopa by decarboxylase and dehydroxylase enzymes 90 On the contrary gut microbiota may also alter the metabolism of the drugs by modulating the host drug metabolism This mechanism can be mediated by microbial metabolites or by modifying host metabolites which in turn change the expression of host metabolizing enzymes 91 A large number of studies have demonstrated the metabolism of over 50 drugs by the gut microbiota 98 89 For example lovastatin a cholesterol lowering agent which is a lactone prodrug is partially activated by the human gut microbiota forming active acid hydroxylated metabolites 95 Conversely digoxin a drug used to treat Congestive Heart Failure is inactivated by a member of the gut microbiota i e Eggerthella lanta 99 Eggerthella lanta has a cytochrome encoding operon up regulated by digoxin and associated with digoxin inactivation 99 Gut microbiota can also modulate the efficacy and toxicity of chemotherapeutic agents such as irinotecan 100 This effect is derived from the microbiome encoded b glucuronidase enzymes which recover the active form of the irinotecan causing gastrointestinal toxicity 101 Secondary metabolites edit This microbial community in the gut has a huge biochemical capability to produce distinct secondary metabolites that are sometimes produced from the metabolic conversion of dietary foods such as fibers endogenous biological compounds such as Indole or Bile acids 102 103 104 Microbial metabolites especially short chain fatty acids SCFAs and secondary bile acids BAs play important roles for the human in health and disease states 105 106 107 One of the most important bacterial metabolites produced by the gut microbiota is secondary bile acids BAs 104 These metabolites are produced by the bacterial biotransformation of the primary bile acids such as cholic acid CA and chenodeoxycholic acid CDCA into secondary bile acids BAs lithocholic acid LCA and deoxy cholic acid DCA respectively 108 Primary bile acids which are synthesized by hepatocytes and stored in the gall bladder possess hydrophobic characters These metabolites are subsequently metabolized by the gut microbiota into secondary metabolites with increased hydrophobicity 108 Bile salt hydrolases BSH which are conserved across gut microbiota phyla such as Bacteroides Firmicutes and Actinobacteria responsible for the first step of secondary bile acids metabolism 108 Secondary bile acids BAs such as DCA and LCA have been demonstrated to inhibit both Clostridium difficile germination and outgrowth 107 Dysbiosis edit The gut microbiota is important for maintaining homeostasis in the intestine Development of intestinal cancer is associated with an imbalance in the natural microflora dysbiosis 109 The secondary bile acid deoxycholic acid is associated with alterations of the microbial community that lead to increased intestinal carcinogenesis 109 Increased exposure of the colon to secondary bile acids resulting from dysbiosis can cause DNA damage and such damage can produce carcinogenic mutations in cells of the colon 110 The high density of bacteria in the colon about 1012 per ml that are subject to dysbiosis compared to the relatively low density in the small intestine about 102 per ml may account for the greater than 10 fold higher incidence of cancer in the colon compared to the small intestine 110 Gut brain axis edit Main article Gut brain axis The gut brain axis is the biochemical signaling that takes place between the gastrointestinal tract and the central nervous system 67 That term has been expanded to include the role of the gut flora in the interplay the term microbiome gut brain axis is sometimes used to describe paradigms explicitly including the gut flora 67 111 112 Broadly defined the gut brain axis includes the central nervous system neuroendocrine and neuroimmune systems including the hypothalamic pituitary adrenal axis HPA axis sympathetic and parasympathetic arms of the autonomic nervous system including the enteric nervous system the vagus nerve and the gut microbiota 67 112 A systematic review from 2016 examined the preclinical and small human trials that have been conducted with certain commercially available strains of probiotic bacteria and found that among those tested Bifidobacterium and Lactobacillus genera B longum B breve B infantis L helveticus L rhamnosus L plantarum and L casei had the most potential to be useful for certain central nervous system disorders 15 Alterations in microbiota balance editEffects of antibiotic use edit Altering the numbers of gut bacteria for example by taking broad spectrum antibiotics may affect the host s health and ability to digest food 113 Antibiotics can cause antibiotic associated diarrhea by irritating the bowel directly changing the levels of microbiota or allowing pathogenic bacteria to grow 7 Another harmful effect of antibiotics is the increase in numbers of antibiotic resistant bacteria found after their use which when they invade the host cause illnesses that are difficult to treat with antibiotics 113 Changing the numbers and species of gut microbiota can reduce the body s ability to ferment carbohydrates and metabolize bile acids and may cause diarrhea Carbohydrates that are not broken down may absorb too much water and cause runny stools or lack of SCFAs produced by gut microbiota could cause diarrhea 7 A reduction in levels of native bacterial species also disrupts their ability to inhibit the growth of harmful species such as C difficile and Salmonella kedougou and these species can get out of hand though their overgrowth may be incidental and not be the true cause of diarrhea 6 7 113 Emerging treatment protocols for C difficile infections involve fecal microbiota transplantation of donor feces see Fecal transplant 114 Initial reports of treatment describe success rates of 90 with few side effects Efficacy is speculated to result from restoring bacterial balances of bacteroides and firmicutes classes of bacteria 115 The composition of the gut microbiome also changes in severe illnesses due not only to antibiotic use but also to such factors as ischemia of the gut failure to eat and immune compromise Negative effects from this have led to interest in selective digestive tract decontamination a treatment to kill only pathogenic bacteria and allow the re establishment of healthy ones 116 Antibiotics alter the population of the microbiota in the gastrointestinal tract and this may change the intra community metabolic interactions modify caloric intake by using carbohydrates and globally affects host metabolic hormonal and immune homeostasis 117 There is reasonable evidence that taking probiotics containing Lactobacillus species may help prevent antibiotic associated diarrhea and that taking probiotics with Saccharomyces e g Saccharomyces boulardii may help to prevent Clostridium difficile infection following systemic antibiotic treatment 118 Pregnancy edit The gut microbiota of a woman changes as pregnancy advances with the changes similar to those seen in metabolic syndromes such as diabetes The change in gut microbiota causes no ill effects The newborn s gut microbiota resemble the mother s first trimester samples The diversity of the microbiome decreases from the first to third trimester as the numbers of certain species go up 55 119 Probiotics prebiotics synbiotics and pharmabiotics edit Probiotics are microorganisms that are believed to provide health benefits when consumed 120 121 With regard to gut microbiota prebiotics are typically non digestible fiber compounds that pass undigested through the upper part of the gastrointestinal tract and stimulate the growth or activity of advantageous gut flora by acting as substrate for them 38 122 Synbiotics refers to food ingredients or dietary supplements combining probiotics and prebiotics in a form of synergism 123 The term pharmabiotics is used in various ways to mean pharmaceutical formulations standardized manufacturing that can obtain regulatory approval as a drug of probiotics prebiotics or synbiotics 124 probiotics that have been genetically engineered or otherwise optimized for best performance shelf life survival in the digestive tract etc 125 and the natural products of gut flora metabolism vitamins etc 126 There is some evidence that treatment with some probiotic strains of bacteria may be effective in irritable bowel syndrome 127 and chronic idiopathic constipation Those organisms most likely to result in a decrease of symptoms have included Bifidobacterium breve Bifidobacterium infantis Enterococcus faecium Lactobacillus plantarum Lactobacillus reuteri Lactobacillus rhamnosus Lactobacillus salivarius Propionibacterium freudenreichii Streptococcus thermophilus 128 129 130 Fecal floatation edit Feces of about 10 15 of people consistently floats in toilet water floaters while the rest produce feces that sinks sinkers and production of gas causes feces to float 131 While conventional mice often produce floaters gnotobiotic germfree mice no gut microbiota bred in germfree isolator produce sinkers and gut microbiota colonization in germfree mice leads to food transformation to microbial biomass and enrichment of multiple gasogenic bacterial species that turns the sinkers into floaters 132 Research edit Tests for whether non antibiotic drugs may impact human gut associated bacteria were performed by in vitro analysis on more than 1000 marketed drugs against 40 gut bacterial strains demonstrating that 24 of the drugs inhibited the growth of at least one of the bacterial strains 133 Effects of exercise edit Gut microbiota and exercise have recently been shown to be interconnected Both moderate and intense exercise are typically part of the training regimen of endurance athletes but they exert different effects on health The interconnection between gut microbiota and endurance sports depends upon exercise intensity and training status 134 Role in disease editBacteria in the digestive tract can contribute to and be affected by disease in various ways The presence or overabundance of some kinds of bacteria may contribute to inflammatory disorders such as inflammatory bowel disease 6 Additionally metabolites from certain members of the gut flora may influence host signalling pathways contributing to disorders such as obesity and colon cancer 6 Alternatively in the event of a breakdown of the gut epithelium the intrusion of gut flora components into other host compartments can lead to sepsis 6 Ulcers edit Helicobacter pylori infection can initiate formation of stomach ulcers when the bacteria penetrate the stomach epithelial lining then causing an inflammatory phagocytotic response 135 In turn the inflammation damages parietal cells which release excessive hydrochloric acid into the stomach and produce less of the protective mucus 136 Injury to the stomach lining leading to ulcers develops when gastric acid overwhelms the defensive properties of cells and inhibits endogenous prostaglandin synthesis reduces mucus and bicarbonate secretion reduces mucosal blood flow and lowers resistance to injury 136 Reduced protective properties of the stomach lining increase vulnerability to further injury and ulcer formation by stomach acid pepsin and bile salts 135 136 Bowel perforation edit Normally commensal bacteria can harm the host if they extrude from the intestinal tract 11 12 Translocation which occurs when bacteria leave the gut through its mucosal lining can occur in a number of different diseases 12 If the gut is perforated bacteria invade the interstitium causing a potentially fatal infection 5 715 Inflammatory bowel diseases edit The two main types of inflammatory bowel diseases Crohn s disease and ulcerative colitis are chronic inflammatory disorders of the gut the causes of these diseases are unknown and issues with the gut flora and its relationship with the host have been implicated in these conditions 14 137 138 139 Additionally it appears that interactions of gut flora with the gut brain axis have a role in IBD with physiological stress mediated through the hypothalamic pituitary adrenal axis driving changes to intestinal epithelium and the gut flora in turn releasing factors and metabolites that trigger signaling in the enteric nervous system and the vagus nerve 4 The diversity of gut flora appears to be significantly diminished in people with inflammatory bowel diseases compared to healthy people additionally in people with ulcerative colitis Proteobacteria and Actinobacteria appear to dominate in people with Crohn s Enterococcus faecium and several Proteobacteria appear to be over represented 4 There is reasonable evidence that correcting gut flora imbalances by taking probiotics with Lactobacilli and Bifidobacteria can reduce visceral pain and gut inflammation in IBD 118 Irritable bowel syndrome edit Irritable bowel syndrome is a result of stress and chronic activation of the HPA axis its symptoms include abdominal pain changes in bowel movements and an increase in proinflammatory cytokines Overall studies have found that the luminal and mucosal microbiota are changed in irritable bowel syndrome individuals and these changes can relate to the type of irritation such as diarrhea or constipation Also there is a decrease in the diversity of the microbiome with low levels of fecal Lactobacilli and Bifidobacteria high levels of facultative anaerobic bacteria such as Escherichia coli and increased ratios of Firmicutes Bacteroidetes 112 Asthma edit With asthma two hypotheses have been posed to explain its rising prevalence in the developed world The hygiene hypothesis posits that children in the developed world are not exposed to enough microbes and thus may contain lower prevalence of specific bacterial taxa that play protective roles 140 The second hypothesis focuses on the Western pattern diet which lacks whole grains and fiber and has an overabundance of simple sugars 14 Both hypotheses converge on the role of short chain fatty acids SCFAs in immunomodulation These bacterial fermentation metabolites are involved in immune signalling that prevents the triggering of asthma and lower SCFA levels are associated with the disease 140 141 Lacking protective genera such as Lachnospira Veillonella Rothia and Faecalibacterium has been linked to reduced SCFA levels 140 Further SCFAs are the product of bacterial fermentation of fiber which is low in the Western pattern diet 14 141 SCFAs offer a link between gut flora and immune disorders and as of 2016 this was an active area of research 14 Similar hypotheses have also been posited for the rise of food and other allergies 142 Diabetes mellitus type 1 edit The connection between the gut microbiota and diabetes mellitus type 1 has also been linked to SCFAs such as butyrate and acetate Diets yielding butyrate and acetate from bacterial fermentation show increased Treg expression 143 Treg cells downregulate effector T cells which in turn reduces the inflammatory response in the gut 144 Butyrate is an energy source for colon cells butyrate yielding diets thus decrease gut permeability by providing sufficient energy for the formation of tight junctions 145 Additionally butyrate has also been shown to decrease insulin resistance suggesting gut communities low in butyrate producing microbes may increase chances of acquiring diabetes mellitus type 2 146 Butyrate yielding diets may also have potential colorectal cancer suppression effects 145 Obesity and metabolic syndrome edit The gut flora have been implicated in obesity and metabolic syndrome due to a key role in the digestive process the Western pattern diet appears to drive and maintain changes in the gut flora that in turn change how much energy is derived from food and how that energy is used 139 147 One aspect of a healthy diet that is often lacking in the Western pattern diet is fiber and other complex carbohydrates that a healthy gut flora require flourishing changes to gut flora in response to a Western pattern diet appear to increase the amount of energy generated by the gut flora which may contribute to obesity and metabolic syndrome 118 There is also evidence that microbiota influence eating behaviours based on the preferences of the microbiota which can lead to the host consuming more food eventually resulting in obesity It has generally been observed that with higher gut microbiome diversity the microbiota will spend energy and resources on competing with other microbiota and less on manipulating the host The opposite is seen with lower gut microbiome diversity and these microbiotas may work together to create host food cravings 49 Additionally the liver plays a dominant role in blood glucose homeostasis by maintaining a balance between the uptake and storage of glucose through the metabolic pathways of glycogenesis and gluconeogenesis Intestinal lipids regulate glucose homeostasis involving a gut brain liver axis The direct administration of lipids into the upper intestine increases the long chain fatty acyl coenzyme A LCFA CoA levels in the upper intestines and suppresses glucose production even under subdiaphragmatic vagotomy or gut vagal deafferentation This interrupts the neural connection between the brain and the gut and blocks the upper intestinal lipids ability to inhibit glucose production The gut brain liver axis and gut microbiota composition can regulate the glucose homeostasis in the liver and provide potential therapeutic methods to treat obesity and diabetes 148 Just as gut flora can function in a feedback loop that can drive the development of obesity there is evidence that restricting intake of calories i e dieting can drive changes to the composition of the gut flora 139 Other animals editThe composition of the human gut microbiome is similar to that of the other great apes However humans gut biota has decreased in diversity and changed in composition since our evolutionary split from Pan 149 Humans display increases in Bacteroidetes a bacterial phylum associated with diets high in animal protein and fat and decreases in Methanobrevibacter and Fibrobacter groups that ferment complex plant polysaccharides 149 These changes are the result of the combined dietary genetic and cultural changes humans have undergone since evolutionary divergence from Pan citation needed In addition to humans and vertebrates some insects also have complex and diverse gut microbiota that play key nutritional roles 2 Microbial communities associated with termites can constitute a majority of the weight of the individuals and perform important roles in the digestion of lignocellulose and nitrogen fixation 150 These communities are host specific and closely related insect species share comparable similarities in gut microbiota composition 151 152 In cockroaches gut microbiota have been shown to assemble in a deterministic fashion irrespective of the inoculum 153 the reason for this host specific assembly remains unclear Bacterial communities associated with insects like termites and cockroaches are determined by a combination of forces primarily diet but there is some indication that host phylogeny may also be playing a role in the selection of lineages 151 152 For more than 51 years it has been known that the administration of low doses of antibacterial agents promotes the growth of farm animals to increase weight gain 117 In a study carried out on mice the ratio of Firmicutes and Lachnospiraceae was significantly elevated in animals treated with subtherapeutic doses of different antibiotics By analyzing the caloric content of faeces and the concentration of small chain fatty acids SCFAs in the GI tract it was concluded that the changes in the composition of microbiota lead to an increased capacity to extract calories from otherwise indigestible constituents and to an increased production of SCFAs These findings provide evidence that antibiotics perturb not only the composition of the GI microbiome but also its metabolic capabilities specifically with respect to SCFAs 117 See also edit nbsp Biology portal nbsp Medicine portalColonisation resistance List of human flora List of microbiota species of the lower reproductive tract of women Skin flora Verotoxin producing Escherichia coliNotes edit There is substantial variation in microbiome composition and microbial concentrations by anatomical site 82 83 Fluid from the human colon which contains the highest concentration of microbes of any anatomical site contains approximately one trillion 10 12 bacterial cells ml 82 References edit Moszak M Szulinska M Bogdanski P 15 April 2020 You Are What You Eat The Relationship between Diet Microbiota and Metabolic Disorders A Review Nutrients 12 4 1096 doi 10 3390 nu12041096 PMC 7230850 PMID 32326604 S2CID 216108564 a b Engel P Moran N 2013 The gut microbiota of insects diversity in structure and function FEMS Microbiology Reviews 37 5 699 735 doi 10 1111 1574 6976 12025 PMID 23692388 Segata N Boernigen D Tickle TL Morgan XC Garrett WS Huttenhower C 14 May 2013 Computational meta omics for microbial community studies Molecular Systems Biology 9 666 doi 10 1038 msb 2013 22 PMC 4039370 PMID 23670539 a b c Saxena R Sharma V K 2016 A Metagenomic Insight Into the Human Microbiome Its Implications in Health and Disease In Kumar D S Antonarakis eds Medical and Health Genomics Elsevier Science p 117 doi 10 1016 B978 0 12 420196 5 00009 5 ISBN 978 0 12 799922 7 a b c d e f g h i j k l m Sherwood Linda Willey Joanne Woolverton Christopher 2013 Prescott s Microbiology 9th ed New York McGraw Hill pp 713 21 ISBN 9780073402406 OCLC 886600661 a b c d e f g h i j k l m n o Guarner F Malagelada J 2003 Gut flora in health and disease The Lancet 361 9356 512 19 doi 10 1016 S0140 6736 03 12489 0 PMID 12583961 S2CID 38767655 a b c d e f g h i j k Beaugerie Laurent Petit Jean Claude 2004 Antibiotic associated diarrhoea Best Practice amp Research Clinical Gastroenterology 18 2 337 52 doi 10 1016 j bpg 2003 10 002 PMID 15123074 a b Stephen A M Cummings J H 1980 The Microbial Contribution to Human Faecal Mass Journal of Medical Microbiology 13 1 45 56 doi 10 1099 00222615 13 1 45 PMID 7359576 a b c d e Quigley E M 2013 Gut bacteria in health and disease Gastroenterology amp Hepatology 9 9 560 9 PMC 3983973 PMID 24729765 Turnbaugh Peter J Ley Ruth E Hamady Micah Fraser Liggett Claire M Knight Rob Gordon Jeffrey I October 2007 The Human Microbiome Project Nature 449 7164 804 810 Bibcode 2007Natur 449 804T doi 10 1038 nature06244 ISSN 0028 0836 PMC 3709439 PMID 17943116 a b c d e f g h i j k l m n o p Sommer Felix Backhed Fredrik 2013 The gut microbiota masters of host development and physiology Nature Reviews Microbiology 11 4 227 38 doi 10 1038 nrmicro2974 PMID 23435359 S2CID 22798964 a b c d Faderl Martin Noti Mario Corazza Nadia Mueller Christoph 2015 Keeping bugs in check The mucus layer as a critical component in maintaining intestinal homeostasis IUBMB Life 67 4 275 85 doi 10 1002 iub 1374 PMID 25914114 S2CID 25878594 a b c d e f Clarke Gerard Stilling Roman M Kennedy Paul J Stanton Catherine Cryan John F Dinan Timothy G 2014 Minireview Gut Microbiota The Neglected Endocrine Organ Molecular Endocrinology 28 8 1221 38 doi 10 1210 me 2014 1108 PMC 5414803 PMID 24892638 a b c d e f Shen Sj Wong Connie HY 2016 Bugging inflammation Role of the gut microbiota Clinical amp Translational Immunology 5 4 e72 doi 10 1038 cti 2016 12 PMC 4855262 PMID 27195115 a b Wang Huiying Lee In Seon Braun Christoph Enck Paul 2016 Effect of Probiotics on Central Nervous System Functions in Animals and Humans A Systematic Review Journal of Neurogastroenterology and Motility 22 4 589 605 doi 10 5056 jnm16018 PMC 5056568 PMID 27413138 a b c d e f Sears Cynthia L 2005 A dynamic partnership Celebrating our gut flora Anaerobe 11 5 247 51 doi 10 1016 j anaerobe 2005 05 001 PMID 16701579 Shapira Michael 2016 07 01 Gut Microbiotas and Host Evolution Scaling Up Symbiosis Trends in Ecology amp Evolution 31 7 539 549 doi 10 1016 j tree 2016 03 006 ISSN 0169 5347 PMID 27039196 Walker Alan W Hoyles Lesley August 2023 Human microbiome myths and misconceptions Nature Microbiology 8 8 1392 1396 doi 10 1038 s41564 023 01426 7 ISSN 2058 5276 PMID 37524974 Lozupone Catherine A Stombaugh Jesse I Gordon Jeffrey I Jansson Janet K Knight Rob 2012 Diversity stability and resilience of the human gut microbiota Nature 489 7415 220 30 Bibcode 2012Natur 489 220L doi 10 1038 nature11550 PMC 3577372 PMID 22972295 Qin Junjie Li Ruiqiang Raes Jeroen Arumugam Manimozhiyan Burgdorf Kristoffer Solvsten Manichanh Chaysavanh Nielsen Trine Pons Nicolas Levenez Florence Yamada Takuji Mende Daniel R Li Junhua Xu Junming Li Shaochuan Li Dongfang Cao Jianjun Wang Bo Liang Huiqing Zheng Huisong Xie Yinlong Tap Julien Lepage Patricia Bertalan Marcelo Batto Jean Michel Hansen Torben Le Paslier Denis Linneberg Allan Nielsen H Bjorn Pelletier Eric Renault Pierre 2010 A human gut microbial gene catalogue established by metagenomic sequencing Nature 464 7285 59 65 Bibcode 2010Natur 464 59 doi 10 1038 nature08821 PMC 3779803 PMID 20203603 Shanahan Fergus 2002 The host microbe interface within the gut Best Practice amp Research Clinical Gastroenterology 16 6 915 31 doi 10 1053 bega 2002 0342 PMID 12473298 Tap Julien Mondot Stanislas Levenez Florence Pelletier Eric Caron Christophe Furet Jean Pierre Ugarte Edgardo Munoz Tamayo Rafael Paslier Denis L E Nalin Renaud Dore Joel Leclerc Marion 2009 Towards the human intestinal microbiota phylogenetic core Environmental Microbiology 11 10 2574 84 Bibcode 2009EnvMi 11 2574T doi 10 1111 j 1462 2920 2009 01982 x PMID 19601958 a b O Hara Ann M Shanahan Fergus 2006 The gut flora as a forgotten organ EMBO Reports 7 7 688 93 doi 10 1038 sj embor 7400731 PMC 1500832 PMID 16819463 Khanna Sahil Tosh Pritish K 2014 A Clinician s Primer on the Role of the Microbiome in Human Health and Disease Mayo Clinic Proceedings 89 1 107 14 doi 10 1016 j mayocp 2013 10 011 PMID 24388028 a b Cui Lijia Morris Alison Ghedin Elodie 2013 The human mycobiome in health and disease Genome Medicine 5 7 63 doi 10 1186 gm467 PMC 3978422 PMID 23899327 Erdogan Askin Rao Satish S C 2015 Small Intestinal Fungal Overgrowth Current Gastroenterology Reports 17 4 16 doi 10 1007 s11894 015 0436 2 PMID 25786900 S2CID 3098136 Bello Maria G Dominguez Knight Rob Gilbert Jack A Blaser Martin J 4 October 2018 Preserving microbial diversity Science 362 6410 33 34 Bibcode 2018Sci 362 33B doi 10 1126 science aau8816 PMID 30287652 S2CID 52919917 a b Arumugam Manimozhiyan Raes Jeroen Pelletier Eric Le Paslier Denis Yamada Takuji Mende Daniel R Fernandes Gabriel R Tap Julien Bruls Thomas Batto Jean Michel Bertalan Marcelo Borruel Natalia Casellas Francesc Fernandez Leyden Gautier Laurent Hansen Torben Hattori Masahira Hayashi Tetsuya Kleerebezem Michiel Kurokawa Ken Leclerc Marion Levenez Florence Manichanh Chaysavanh Nielsen H Bjorn Nielsen Trine Pons Nicolas Poulain Julie Qin Junjie Sicheritz Ponten Thomas Tims Sebastian 2011 Enterotypes of the human gut microbiome Nature 473 7346 174 80 Bibcode 2011Natur 473 174 doi 10 1038 nature09944 PMC 3728647 PMID 21508958 Wu G D Chen J Hoffmann C Bittinger K Chen Y Y Keilbaugh S A Bewtra M Knights D Walters W A Knight R Sinha R Gilroy E Gupta K Baldassano R Nessel L Li H Bushman F D Lewis J D 2011 Linking Long Term Dietary Patterns with Gut Microbial Enterotypes Science 334 6052 105 08 Bibcode 2011Sci 334 105W doi 10 1126 science 1208344 PMC 3368382 PMID 21885731 Zimmer Carl April 20 2011 Bacteria Divide People Into 3 Types Scientists Say The New York Times Retrieved April 21 2011 a group of scientists now report just three distinct ecosystems in the guts of people they have studied Knights Dan Ward Tonya McKinlay Christopher Miller Hannah Gonzalez Antonio McDonald Daniel Knight Rob 8 October 2014 Rethinking Enterotypes Cell Host amp Microbe 16 4 433 37 doi 10 1016 j chom 2014 09 013 PMC 5558460 PMID 25299329 Todar Kenneth 2012 The Normal Bacterial Flora of Humans Todar s Online Textbook of Bacteriology Retrieved June 25 2016 Quigley Eamonn M M Quera Rodrigo 2006 Small Intestinal Bacterial Overgrowth Roles of Antibiotics Prebiotics and Probiotics Gastroenterology 130 2 S78 90 doi 10 1053 j gastro 2005 11 046 PMID 16473077 S2CID 16904501 Adams M R Moss M O 2007 Food Microbiology doi 10 1039 9781847557940 ISBN 978 0 85404 284 5 S2CID 241261974 a b The normal gut flora PDF slideshow 2004 Archived from the original PDF on 2004 05 26 Retrieved 2023 01 02 via University of Glasgow Braune A Blaut M 2016 Bacterial species involved in the conversion of dietary flavonoids in the human gut Gut Microbes 7 3 216 234 doi 10 1080 19490976 2016 1158395 PMC 4939924 PMID 26963713 a b Steinhoff U 2005 Who controls the crowd New findings and old questions about the intestinal microflora Immunology Letters 99 1 12 16 doi 10 1016 j imlet 2004 12 013 PMID 15894105 a b c d e f Gibson Glenn R 2004 Fibre and effects on probiotics the prebiotic concept Clinical Nutrition Supplements 1 2 25 31 doi 10 1016 j clnu 2004 09 005 Miquel S Martin R Rossi O Bermudez Humaran LG Chatel JM Sokol H Thomas M Wells JM Langella P 2013 Faecalibacterium prausnitzii and human intestinal health Current Opinion in Microbiology 16 3 255 61 doi 10 1016 j mib 2013 06 003 PMID 23831042 Ley Ruth E 2010 Obesity and the human microbiome Current Opinion in Gastroenterology 26 1 5 11 doi 10 1097 MOG 0b013e328333d751 PMID 19901833 S2CID 23329156 Nash Andrea K Auchtung Thomas A Wong Matthew C Smith Daniel P Gesell Jonathan R Ross Matthew C Stewart Christopher J Metcalf Ginger A Muzny Donna M Gibbs Richard A Ajami Nadim J Petrosino Joseph F 2017 The gut mycobiome of the Human Microbiome Project healthy cohort Microbiome 5 1 153 doi 10 1186 s40168 017 0373 4 PMC 5702186 PMID 29178920 Scarpellini Emidio Ianiro Gianluca Attili Fabia Bassanelli Chiara De Santis Adriano Gasbarrini Antonio 2015 The human gut microbiota and virome Potential therapeutic implications Digestive and Liver Disease 47 12 1007 12 doi 10 1016 j dld 2015 07 008 PMC 7185617 PMID 26257129 Gerritsen Jacoline Smidt Hauke Rijkers Ger de Vos Willem 27 May 2011 Intestinal microbiota in human health and disease the impact of probiotics Genes amp Nutrition 6 3 209 40 doi 10 1007 s12263 011 0229 7 PMC 3145058 PMID 21617937 a b c d e f Yatsunenko T Rey F E Manary M J Trehan I Dominguez Bello M G Contreras M Magris M Hidalgo G Baldassano R N Anokhin A P Heath A C Warner B Reeder J Kuczynski J Caporaso J G Lozupone C A Lauber C Clemente J C Knights D Knight R Gordon J I 2012 Human gut microbiome viewed across age and geography Nature 486 7402 222 27 Bibcode 2012Natur 486 222Y doi 10 1038 nature11053 PMC 3376388 PMID 22699611 De Filippo C Cavalieri D Di Paola M Ramazzotti M Poullet J B Massart S Collini S Pieraccini G Lionetti P 2010 Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa Proceedings of the National Academy of Sciences 107 33 14691 6 Bibcode 2010PNAS 10714691D doi 10 1073 pnas 1005963107 PMC 2930426 PMID 20679230 Jonkers Daisy M A E 2016 Microbial perturbations and modulation in conditions associated with malnutrition and malabsorption Best Practice amp Research Clinical Gastroenterology 30 2 161 72 doi 10 1016 j bpg 2016 02 006 PMID 27086883 Million Matthieu Diallo Aldiouma Raoult Didier May 2017 Gut microbiota and malnutrition PDF Microbial Pathogenesis 106 127 138 doi 10 1016 j micpath 2016 02 003 PMID 26853753 S2CID 20381329 Rytter Maren Johanne Heilskov Kolte Lilian Briend Andre Friis Henrik Christensen Vibeke Brix 2014 The Immune System in Children with Malnutrition A Systematic Review PLOS ONE 9 8 e105017 Bibcode 2014PLoSO 9j5017R doi 10 1371 journal pone 0105017 PMC 4143239 PMID 25153531 a b Alcock Joe Maley Carlo C Aktipis C Athena 2014 Is eating behavior manipulated by the gastrointestinal microbiota Evolutionary pressures and potential mechanisms BioEssays 36 10 940 9 doi 10 1002 bies 201400071 PMC 4270213 PMID 25103109 a b c Renson Audrey Herd Pamela Dowd Jennifer B 2020 Sick Individuals and Sick Microbial Populations Challenges in Epidemiology and the Microbiome Annual Review of Public Health 41 63 80 doi 10 1146 annurev publhealth 040119 094423 PMC 9713946 PMID 31635533 Turroni Francesca Peano Clelia Pass Daniel A Foroni Elena Severgnini Marco Claesson Marcus J Kerr Colm Hourihane Jonathan Murray Deirdre Fuligni Fabio Gueimonde Miguel Margolles Abelardo De Bellis Gianluca o Toole Paul W Van Sinderen Douwe Marchesi Julian R Ventura Marco 2012 Diversity of Bifidobacteria within the Infant Gut Microbiota PLOS ONE 7 5 e36957 Bibcode 2012PLoSO 736957T doi 10 1371 journal pone 0036957 PMC 3350489 PMID 22606315 Davenport Emily R Sanders Jon G Song Se Jin Amato Katherine R Clark Andrew G Knight Rob 2017 12 27 The human microbiome in evolution BMC Biology 15 1 127 doi 10 1186 s12915 017 0454 7 ISSN 1741 7007 PMC 5744394 PMID 29282061 Perez Munoz Maria Elisa Arrieta Marie Claire Ramer Tait Amanda E Walter Jens 2017 A critical assessment of the sterile womb and in utero colonization hypotheses Implications for research on the pioneer infant microbiome Microbiome 5 1 48 doi 10 1186 s40168 017 0268 4 PMC 5410102 PMID 28454555 Matamoros Sebastien Gras Leguen Christele Le Vacon Francoise Potel Gilles de la Cochetiere Marie France 2013 Development of intestinal microbiota in infants and its impact on health Trends in Microbiology 21 4 167 73 doi 10 1016 j tim 2012 12 001 PMID 23332725 a b c d e f g Mueller Noel T Bakacs Elizabeth Combellick Joan Grigoryan Zoya Dominguez Bello Maria G 2015 The infant microbiome development mom matters Trends in Molecular Medicine 21 2 109 17 doi 10 1016 j molmed 2014 12 002 PMC 4464665 PMID 25578246 Jimenez Esther Fernandez Leonides Marin Maria L Martin Rocio Odriozola Juan M Nueno Palop Carmen Narbad Arjan Olivares Monica Xaus Jordi Rodriguez Juan M 2005 Isolation of Commensal Bacteria from Umbilical Cord Blood of Healthy Neonates Born by Cesarean Section Current Microbiology 51 4 270 4 doi 10 1007 s00284 005 0020 3 PMID 16187156 S2CID 43438656 Collado Maria Carmen Rautava Samuli Aakko Juhani Isolauri Erika Salminen Seppo 2016 Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid Scientific Reports 6 23129 Bibcode 2016NatSR 623129C doi 10 1038 srep23129 PMC 4802384 PMID 27001291 Jimenez Esther Marin Maria L Martin Rocio Odriozola Juan M Olivares Monica Xaus Jordi Fernandez Leonides Rodriguez Juan M 2008 Is meconium from healthy newborns actually sterile Research in Microbiology 159 3 187 93 doi 10 1016 j resmic 2007 12 007 PMID 18281199 Perez Munoz Maria Elisa Arrieta Marie Claire Ramer Tait Amanda E Walter Jens 2017 A critical assessment of the sterile womb and in utero colonization hypotheses Implications for research on the pioneer infant microbiome Microbiome 5 1 48 doi 10 1186 s40168 017 0268 4 PMC 5410102 PMID 28454555 Adlerberth I Wold AE 2009 Establishment of the gut microbiota in Western infants Acta Paediatrica 98 2 229 38 doi 10 1111 j 1651 2227 2008 01060 x PMID 19143664 S2CID 205859933 Babies gut bacteria affected by delivery method Vaginal delivery promotes mother s gut bacteria in babies gut ScienceDaily 18 September 2019 Archived from the original on 24 November 2021 Retrieved 31 May 2022 Coppa G V Zampini L Galeazzi T Gabrielli O 2006 Prebiotics in human milk A review Digestive and Liver Disease 38 S291 4 doi 10 1016 S1590 8658 07 60013 9 PMID 17259094 Mady Eman A Doghish Ahmed S El Dakroury Walaa A Elkhawaga Samy Y Ismail Ahmed El Mahdy Hesham A Elsakka Elsayed G E El Husseiny Hussein M 2023 07 01 Impact of the mother s gut microbiota on infant microbiome and brain development Neuroscience amp Biobehavioral Reviews 150 105195 doi 10 1016 j neubiorev 2023 105195 ISSN 0149 7634 PMID 37100161 S2CID 258302702 Fanaro S Chierici R Guerrini P Vigi V 2007 Intestinal microflora in early infancy Composition and development Acta Paediatrica 92 441 48 55 doi 10 1111 j 1651 2227 2003 tb00646 x PMID 14599042 S2CID 10316311 Yassour Moran Vatanen Tommi Siljander Heli Hamalainen Anu Maaria Harkonen Taina Ryhanen Samppa J Franzosa Eric A Vlamakis Hera Huttenhower Curtis Gevers Dirk Lander Eric S Knip Mikael Xavier Ramnik J 2016 Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability Science Translational Medicine 8 343 343ra81 doi 10 1126 scitranslmed aad0917 PMC 5032909 PMID 27306663 Gibson G R Roberfroid M B 1995 Dietary modulation of the human colonic microbiota Introducing the concept of prebiotics The Journal of Nutrition 125 6 1401 1412 doi 10 1093 jn 125 6 1401 PMID 7782892 a b c d Wang Yan Kasper Lloyd H 2014 The role of microbiome in central nervous system disorders Brain Behavior and Immunity 38 1 12 doi 10 1016 j bbi 2013 12 015 PMC 4062078 PMID 24370461 a b Yoon My Young Lee Keehoon Yoon Sang Sun 2014 Protective role of gut commensal microbes against intestinal infections Journal of Microbiology 52 12 983 9 doi 10 1007 s12275 014 4655 2 PMID 25467115 S2CID 54622675 Reinoso Webb Cynthia Koboziev Iurii Furr Kathryn L Grisham Matthew B 2016 Protective and pro inflammatory roles of intestinal bacteria Pathophysiology 23 2 67 80 doi 10 1016 j pathophys 2016 02 002 PMC 4867289 PMID 26947707 Mantis N J Rol N Corthesy B 2011 Secretory IgA s complex roles in immunity and mucosal homeostasis in the gut Mucosal Immunology 4 6 603 11 doi 10 1038 mi 2011 41 PMC 3774538 PMID 21975936 a b Peterson Lance W Artis David 2014 Intestinal epithelial cells Regulators of barrier function and immune homeostasis Nature Reviews Immunology 14 3 141 53 doi 10 1038 nri3608 PMID 24566914 S2CID 3351351 a b Honda Kenya Littman Dan R 2016 The microbiota in adaptive immune homeostasis and disease Nature 535 7610 75 84 Bibcode 2016Natur 535 75H doi 10 1038 nature18848 PMID 27383982 S2CID 4461492 a b Levy M Thaiss C A Elinav E 2016 Metabolites messengers between the microbiota and the immune system Genes amp Development 30 14 1589 97 doi 10 1101 gad 284091 116 PMC 4973288 PMID 27474437 a b c d e f g h i Zhang LS Davies SS April 2016 Microbial metabolism of dietary components to bioactive metabolites opportunities for new therapeutic interventions Genome Med 8 1 46 doi 10 1186 s13073 016 0296 x PMC 4840492 PMID 27102537 Lactobacillus spp convert tryptophan to indole 3 aldehyde I3A through unidentified enzymes 125 Clostridium sporogenes convert tryptophan to IPA 6 likely via a tryptophan deaminase IPA also potently scavenges hydroxyl radicals Table 2 Microbial metabolites their synthesis mechanisms of action and effects on health and diseaseFigure 1 Molecular mechanisms of action of indole and its metabolites on host physiology and disease Wikoff WR Anfora AT Liu J Schultz PG Lesley SA Peters EC Siuzdak G March 2009 Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites Proc Natl Acad Sci U S A 106 10 3698 3703 Bibcode 2009PNAS 106 3698W doi 10 1073 pnas 0812874106 PMC 2656143 PMID 19234110 Production of IPA was shown to be completely dependent on the presence of gut microflora and could be established by colonization with the bacterium Clostridium sporogenes IPA metabolism diagram 3 Indolepropionic acid Human Metabolome Database University of Alberta Retrieved 12 June 2018 Chyan YJ Poeggeler B Omar RA Chain DG Frangione B Ghiso J Pappolla MA July 1999 Potent neuroprotective properties against the Alzheimer beta amyloid by an endogenous melatonin related indole structure indole 3 propionic acid J Biol Chem 274 31 21937 21942 doi 10 1074 jbc 274 31 21937 PMID 10419516 S2CID 6630247 Indole 3 propionic acid IPA has previously been identified in the plasma and cerebrospinal fluid of humans but its functions are not known In kinetic competition experiments using free radical trapping agents the capacity of IPA to scavenge hydroxyl radicals exceeded that of melatonin an indoleamine considered to be the most potent naturally occurring scavenger of free radicals In contrast with other antioxidants IPA was not converted to reactive intermediates with pro oxidant activity Hopper Christopher P De La Cruz Ladie Kimberly Lyles Kristin V Wareham Lauren K Gilbert Jack A Eichenbaum Zehava Magierowski Marcin Poole Robert K Wollborn Jakob Wang Binghe 2020 12 23 Role of Carbon Monoxide in Host Gut Microbiome Communication Chemical Reviews 120 24 13273 13311 doi 10 1021 acs chemrev 0c00586 ISSN 0009 2665 PMID 33089988 S2CID 224824871 Rajilic Stojanovic Mirjana De Vos Willem M 2014 The first 1000 cultured species of the human gastrointestinal microbiota FEMS Microbiology Reviews 38 5 996 1047 doi 10 1111 1574 6976 12075 PMC 4262072 PMID 24861948 Hill M J March 1997 Intestinal flora and endogenous vitamin synthesis European Journal of Cancer Prevention 6 Suppl 1 S43 45 doi 10 1097 00008469 199703001 00009 ISSN 0959 8278 PMID 9167138 S2CID 8740364 The Microbiome Tufts Now 2013 09 17 Retrieved 2020 12 09 a b c d e ElRakaiby M Dutilh BE Rizkallah MR Boleij A Cole JN Aziz RK July 2014 Pharmacomicrobiomics the impact of human microbiome variations on systems pharmacology and personalized therapeutics Omics 18 7 402 414 doi 10 1089 omi 2014 0018 PMC 4086029 PMID 24785449 a b c Cho I Blaser MJ March 2012 The human microbiome at the interface of health and disease Nature Reviews Genetics 13 4 260 270 doi 10 1038 nrg3182 PMC 3418802 PMID 22411464 The composition of the microbiome varies by anatomical site Figure 1 The primary determinant of community composition is anatomical location interpersonal variation is substantial23 24 and is higher than the temporal variability seen at most sites in a single individual25 Hutter T Gimbert C Bouchard F Lapointe FJ 2015 Being human is a gut feeling Microbiome 3 9 doi 10 1186 s40168 015 0076 7 PMC 4359430 PMID 25774294 Kumar K Dhoke GV Sharma AK Jaiswal SK Sharma VK January 2019 Mechanistic elucidation of amphetamine metabolism by tyramine oxidase from human gut microbiota using molecular dynamics simulations Journal of Cellular Biochemistry 120 7 11206 15 doi 10 1002 jcb 28396 PMID 30701587 S2CID 73413138 Sousa Tiago Paterson Ronnie Moore Vanessa Carlsson Anders Abrahamsson Bertil Basit Abdul W 2008 The gastrointestinal microbiota as a site for the biotransformation of drugs International Journal of Pharmaceutics 363 1 2 1 25 doi 10 1016 j ijpharm 2008 07 009 PMID 18682282 Haiser H J Gootenberg D B Chatman K Sirasani G Balskus E P Turnbaugh P J 2013 Predicting and Manipulating Cardiac Drug Inactivation by the Human Gut Bacterium Eggerthella lenta Science 341 6143 295 8 Bibcode 2013Sci 341 295H doi 10 1126 science 1235872 PMC 3736355 PMID 23869020 a b Koppel Nitzan Maini Rekdal Vayu Balskus Emily P 2017 06 23 Chemical transformation of xenobiotics by the human gut microbiota Science 356 6344 doi 10 1126 science aag2770 ISSN 0036 8075 PMC 5534341 PMID 28642381 a b c Spanogiannopoulos Peter Bess Elizabeth N Carmody Rachel N Turnbaugh Peter J 2016 03 14 The microbial pharmacists within us a metagenomic view of xenobiotic metabolism Nature Reviews Microbiology 14 5 273 287 doi 10 1038 nrmicro 2016 17 ISSN 1740 1526 PMC 5243131 PMID 26972811 a b Maini Rekdal Vayu Bess Elizabeth N Bisanz Jordan E Turnbaugh Peter J Balskus Emily P 2019 06 14 Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism Science 364 6445 doi 10 1126 science aau6323 ISSN 0036 8075 PMC 7745125 PMID 31196984 a b Dempsey Joseph L Cui Julia Yue 2019 10 19 Microbiome Is a Functional Modifier of P450 Drug Metabolism Current Pharmacology Reports 5 6 481 490 doi 10 1007 s40495 019 00200 w ISSN 2198 641X PMC 7731899 PMID 33312848 Boerner Udo Abbott Seth Roe Robert L January 1975 The Metabolism of Morphine and Heroin in Man Drug Metabolism Reviews 4 1 39 73 doi 10 3109 03602537508993748 ISSN 0360 2532 PMID 1204496 Dobkin Jay F Saha Jnan R Butler Vincent P Neu Harold C Lindenbaum John 1983 04 15 Digoxin Inactivating Bacteria Identification in Human Gut Flora Science 220 4594 325 327 doi 10 1126 science 6836275 ISSN 0036 8075 PMID 6836275 Sahota S S Bramley P M Menzies I S 1982 02 01 The Fermentation of Lactulose by Colonic Bacteria Microbiology 128 2 319 325 doi 10 1099 00221287 128 2 319 ISSN 1350 0872 PMID 6804597 a b Yoo Dae Hyoung Kim In Sook Van Le Thi Kim Jung Il Hoon Yoo Hye Hyun Kim Dong Hyun 2014 06 19 Gut Microbiota Mediated Drug Interactions between Lovastatin and Antibiotics Drug Metabolism and Disposition 42 9 1508 1513 doi 10 1124 dmd 114 058354 ISSN 0090 9556 PMID 24947972 S2CID 7524335 Haiser Henry J Gootenberg David B Chatman Kelly Sirasani Gopal Balskus Emily P Turnbaugh Peter J 2013 07 19 Predicting and Manipulating Cardiac Drug Inactivation by the Human Gut Bacterium Eggerthella lenta Science 341 6143 295 298 Bibcode 2013Sci 341 295H doi 10 1126 science 1235872 ISSN 0036 8075 PMC 3736355 PMID 23869020 Parvez Md Masud Basit Abdul Jariwala Parth B Gaborik Zsuzsanna Kis Emese Heyward Scott Redinbo Matthew R Prasad Bhagwat 2021 06 01 Quantitative Investigation of Irinotecan Metabolism Transport and Gut Microbiome Activation Drug Metabolism and Disposition 49 8 683 693 doi 10 1124 dmd 121 000476 ISSN 0090 9556 PMC 8407663 PMID 34074730 a b Sousa Tiago Paterson Ronnie Moore Vanessa Carlsson Anders Abrahamsson Bertil Basit Abdul W November 2008 The gastrointestinal microbiota as a site for the biotransformation of drugs International Journal of Pharmaceutics 363 1 2 1 25 doi 10 1016 j ijpharm 2008 07 009 ISSN 0378 5173 PMID 18682282 a b Koppel Nitzan Bisanz Jordan E Pandelia Maria Eirini Turnbaugh Peter J Balskus Emily P 2018 05 15 Ley Ruth Emily ed Discovery and characterization of a prevalent human gut bacterial enzyme sufficient for the inactivation of a family of plant toxins eLife 7 e33953 doi 10 7554 eLife 33953 ISSN 2050 084X PMC 5953540 PMID 29761785 Alexander James L Wilson Ian D Teare Julian Marchesi Julian R Nicholson Jeremy K Kinross James M 2017 03 08 Gut microbiota modulation of chemotherapy efficacy and toxicity Nature Reviews Gastroenterology amp Hepatology 14 6 356 365 doi 10 1038 nrgastro 2017 20 ISSN 1759 5045 PMID 28270698 S2CID 9654170 Brandi Giovanni Dabard Jean Raibaud Pierre Di Battista Monica Bridonneau Chantal Pisi Anna Maria Morselli Labate Antonio Maria Pantaleo Maria Abbondanza De Vivo Antonello Biasco Guido 2006 02 15 Intestinal microflora and digestive toxicity of irinotecan in mice Clinical Cancer Research 12 4 1299 1307 doi 10 1158 1078 0432 ccr 05 0750 ISSN 1078 0432 PMID 16489087 S2CID 26655779 Koh Ara De Vadder Filipe Kovatcheva Datchary Petia Backhed Fredrik June 2016 From Dietary Fiber to Host Physiology Short Chain Fatty Acids as Key Bacterial Metabolites Cell 165 6 1332 1345 doi 10 1016 j cell 2016 05 041 ISSN 0092 8674 PMID 27259147 S2CID 8562345 Konopelski Piotr Ufnal Marcin 2018 09 14 Indoles Gut Bacteria Metabolites of Tryptophan with Pharmacotherapeutic Potential Current Drug Metabolism 19 10 883 890 doi 10 2174 1389200219666180427164731 ISSN 1389 2002 PMID 29708069 S2CID 13979774 a b Collins Stephanie L Stine Jonathan G Bisanz Jordan E Okafor C Denise Patterson Andrew D 2022 10 17 Bile acids and the gut microbiota metabolic interactions and impacts on disease Nature Reviews Microbiology 21 4 236 247 doi 10 1038 s41579 022 00805 x ISSN 1740 1526 PMID 36253479 S2CID 252970168 Yang Wenjing Yu Tianming Huang Xiangsheng Bilotta Anthony J Xu Leiqi Lu Yao Sun Jiaren Pan Fan Zhou Jia Zhang Wenbo Yao Suxia Maynard Craig L Singh Nagendra Dann Sara M Liu Zhanju 2020 09 08 Intestinal microbiota derived short chain fatty acids regulation of immune cell IL 22 production and gut immunity Nature Communications 11 1 4457 Bibcode 2020NatCo 11 4457Y doi 10 1038 s41467 020 18262 6 ISSN 2041 1723 PMC 7478978 PMID 32901017 Murugesan Selvasankar Nirmalkar Khemlal Hoyo Vadillo Carlos Garcia Espitia Matilde Ramirez Sanchez Daniela Garcia Mena Jaime 2017 12 02 Gut microbiome production of short chain fatty acids and obesity in children European Journal of Clinical Microbiology amp Infectious Diseases 37 4 621 625 doi 10 1007 s10096 017 3143 0 ISSN 0934 9723 PMID 29196878 S2CID 254132108 a b Thanissery Rajani Winston Jenessa A Theriot Casey M June 2017 Inhibition of spore germination growth and toxin activity of clinically relevant C difficile strains by gut microbiota derived secondary bile acids Anaerobe 45 86 100 doi 10 1016 j anaerobe 2017 03 004 ISSN 1075 9964 PMC 5466893 PMID 28279860 a b c Jones Brian V Begley Maire Hill Colin Gahan Cormac G M Marchesi Julian R 2008 09 09 Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome Proceedings of the National Academy of Sciences 105 36 13580 13585 Bibcode 2008PNAS 10513580J doi 10 1073 pnas 0804437105 ISSN 0027 8424 PMC 2533232 PMID 18757757 a b Cao H Xu M Dong W Deng B Wang S Zhang Y Wang S Luo S Wang W Qi Y Gao J Cao X Yan F Wang B June 2017 Secondary bile acid induced dysbiosis promotes intestinal carcinogenesis International Journal of Cancer 140 11 2545 2556 doi 10 1002 ijc 30643 PMID 28187526 a b Bernstein H Bernstein C January 2023 Bile acids as carcinogens in the colon and at other sites in the gastrointestinal system Experimental Biology and Medicine Maywood N J 248 1 79 89 doi 10 1177 15353702221131858 PMC 9989147 PMID 36408538 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 6 doi 10 1523 JNEUROSCI 3299 14 2014 PMC 4228144 PMID 25392516 a b c Dinan Timothy G Cryan John F 2015 The impact of gut microbiota on brain and behaviour Current Opinion in Clinical Nutrition and Metabolic Care 18 6 552 8 doi 10 1097 MCO 0000000000000221 PMID 26372511 S2CID 21424690 a b c Carman Robert J Simon Mary Alice Fernandez Haydee Miller Margaret A Bartholomew Mary J 2004 Ciprofloxacin at low levels disrupts colonization resistance of human fecal microflora growing in chemostats Regulatory Toxicology and Pharmacology 40 3 319 26 doi 10 1016 j yrtph 2004 08 005 PMID 15546686 Hvas Christian Lodberg Baunwall Simon Mark Dahl Erikstrup Christian 2020 07 01 Faecal microbiota transplantation A life saving therapy challenged by commercial claims for exclusivity eClinicalMedicine 24 100436 doi 10 1016 j eclinm 2020 100436 ISSN 2589 5370 PMC 7334803 PMID 32642633 Brandt Lawrence J Borody Thomas Julius Campbell Jordana 2011 Endoscopic Fecal Microbiota Transplantation Journal of Clinical Gastroenterology 45 8 655 57 doi 10 1097 MCG 0b013e3182257d4f PMID 21716124 S2CID 2508836 Knight DJW Girling KJ 2003 Gut flora in health and disease The Lancet 361 9371 512 19 doi 10 1016 S0140 6736 03 13438 1 PMID 12781578 S2CID 40683723 a b c Cho I Yamanishi S Cox L Methe B A Zavadil J Li K Gao Z Mahana D Raju K Teitler I Li H Alekseyenko A V Blaser M J 2012 Antibiotics in early life alter the murine colonic microbiome and adiposity Nature 488 7413 621 26 Bibcode 2012Natur 488 621C doi 10 1038 nature11400 PMC 3553221 PMID 22914093 a b c Schneiderhan J Master Hunter T Locke A 2016 Targeting gut flora to treat and prevent disease The Journal of Family Practice 65 1 34 8 PMID 26845162 Baker Monya 2012 Pregnancy alters resident gut microbes Nature doi 10 1038 nature 2012 11118 S2CID 87078157 Hill Colin Guarner Francisco Reid Gregor Gibson Glenn R Merenstein Daniel J Pot Bruno Morelli Lorenzo Canani Roberto Berni Flint Harry J Salminen Seppo Calder Philip C Sanders Mary Ellen 2014 The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic Nature Reviews Gastroenterology amp Hepatology 11 8 506 14 doi 10 1038 nrgastro 2014 66 PMID 24912386 Rijkers Ger T De Vos Willem M Brummer Robert Jan Morelli Lorenzo Corthier Gerard Marteau Philippe 2011 Health benefits and health claims of probiotics Bridging science and marketing British Journal of Nutrition 106 9 1291 6 doi 10 1017 S000711451100287X PMID 21861940 Hutkins Robert W Krumbeck Janina A Bindels Laure B Cani Patrice D Fahey George Goh Yong Jun Hamaker Bruce Martens Eric C Mills David A Rastal Robert A Vaughan Elaine Sanders Mary Ellen 2016 Prebiotics Why definitions matter Current Opinion in Biotechnology 37 1 7 doi 10 1016 j copbio 2015 09 001 PMC 4744122 PMID 26431716 Pandey Kavita R Naik Suresh R Vakil Babu V 2015 Probiotics prebiotics and synbiotics a review Journal of Food Science and Technology 52 12 7577 87 doi 10 1007 s13197 015 1921 1 PMC 4648921 PMID 26604335 Broeckx Geraldine Vandenheuvel Dieter Claes Ingmar J J Lebeer Sarah Kiekens Filip 2016 Drying techniques of probiotic bacteria as an important step towards the development of novel pharmabiotics PDF International Journal of Pharmaceutics 505 1 2 303 18 doi 10 1016 j ijpharm 2016 04 002 hdl 10067 1328840151162165141 PMID 27050865 Sleator Roy D Hill Colin 2009 Rational Design of Improved Pharmabiotics Journal of Biomedicine and Biotechnology 2009 275287 doi 10 1155 2009 275287 PMC 2742647 PMID 19753318 Patterson Elaine Cryan John F Fitzgerald Gerald F Ross R Paul Dinan Timothy G Stanton Catherine 2014 Gut microbiota the pharmabiotics they produce and host health Proceedings of the Nutrition Society 73 4 477 89 doi 10 1017 S0029665114001426 PMID 25196939 Guandalini Stefano Magazzu Giuseppe Chiaro Andrea La Balestra Valeria Di Nardo Giovanni Gopalan Sarath Sibal A Romano Claudio Canani Roberto Berni Lionetti Paolo Setty Mala July 2010 VSL 3 Improves Symptoms in Children with Irritable Bowel Syndrome A Multicenter Randomized Placebo Controlled Double Blind Crossover Study Journal of Pediatric Gastroenterology and Nutrition 51 1 24 30 doi 10 1097 MPG 0b013e3181ca4d95 PMID 20453678 S2CID 33659736 Ford Alexander C Quigley Eamonn M M Lacy Brian E Lembo Anthony J Saito Yuri A Schiller Lawrence R Soffer Edy E Spiegel Brennan M R Moayyedi Paul 2014 Efficacy of Prebiotics Probiotics and Synbiotics in Irritable Bowel Syndrome and Chronic Idiopathic Constipation Systematic Review and Meta analysis The American Journal of Gastroenterology 109 10 1547 61 quiz 1546 1562 doi 10 1038 ajg 2014 202 PMID 25070051 S2CID 205100508 Dupont Andrew Richards Jelinek Katherine A Krill Joseph Rahimi Erik Ghouri Yezaz 2014 Systematic review of randomized controlled trials of probiotics prebiotics and synbiotics in inflammatory bowel disease Clinical and Experimental Gastroenterology 7 473 87 doi 10 2147 CEG S27530 PMC 4266241 PMID 25525379 Yu Cheng Gong Huang Qin 2013 Recent progress on the role of gut microbiota in the pathogenesis of inflammatory bowel disease Journal of Digestive Diseases 14 10 513 7 doi 10 1111 1751 2980 12087 PMID 23848393 S2CID 26982085 Levitt Michael D Duane William C 1972 05 04 Floating Stools Flatus versus Fat New England Journal of Medicine 286 18 973 975 doi 10 1056 NEJM197205042861804 ISSN 0028 4793 PMID 5015442 Aalam Syed Mohammed Musheer Crasta Daphne Norma Roy Pooja Miller A Lee Gamb Scott I Johnson Stephen Till Lisa M Chen Jun Kashyap Purna Kannan Nagarajan 2022 10 27 Genesis of fecal floatation is causally linked to gut microbial colonization in mice Scientific Reports 12 1 18109 Bibcode 2022NatSR 1218109A doi 10 1038 s41598 022 22626 x ISSN 2045 2322 PMC 9613883 PMID 36302811 Maier Lisa Pruteanu Mihaela Kuhn Michael Zeller Georg Telzerow Anja Anderson Exene Erin Brochado Ana Rita Fernandez Keith Conrad Dose Hitomi Mori Hirotada Patil Kiran Raosaheb Bork Peer Typas Athanasios 2018 Extensive impact of non antibiotic drugs on human gut bacteria Nature 555 7698 623 628 Bibcode 2018Natur 555 623M doi 10 1038 nature25979 PMC 6108420 PMID 29555994 Clauss Matthieu Gerard Philippe Mosca Alexis Leclerc Marion 2021 Interplay Between Exercise and Gut Microbiome in the Context of Human Health and Performance Frontiers in Nutrition 8 637010 doi 10 3389 fnut 2021 637010 PMC 8222532 PMID 34179053 a b Kamboj AK Cotter TG Oxentenko AS 2017 Helicobacter pylori The Past Present and Future in Management Mayo Clinic Proceedings 92 4 599 604 doi 10 1016 j mayocp 2016 11 017 ISSN 0025 6196 PMID 28209367 a b c Peptic ulcer disease PDF The Johns Hopkins University School of Medicine 2013 Retrieved 21 October 2020 Burisch Johan Jess Tine Martinato Matteo Lakatos Peter L 2013 The burden of inflammatory bowel disease in Europe Journal of Crohn s and Colitis 7 4 322 37 doi 10 1016 j crohns 2013 01 010 PMID 23395397 Blandino G Inturri R Lazzara F Di Rosa M Malaguarnera L 2016 Impact of gut microbiota on diabetes mellitus Diabetes amp Metabolism 42 5 303 315 doi 10 1016 j diabet 2016 04 004 PMID 27179626 a b c Boulange Claire L Neves Ana Luisa Chilloux Julien Nicholson Jeremy K Dumas Marc Emmanuel 2016 Impact of the gut microbiota on inflammation obesity and metabolic disease Genome Medicine 8 1 42 doi 10 1186 s13073 016 0303 2 PMC 4839080 PMID 27098727 a b c Arrieta Marie Claire Stiemsma Leah T Dimitriu Pedro A Thorson Lisa Russell Shannon Yurist Doutsch Sophie Kuzeljevic Boris Gold Matthew J Britton Heidi M Lefebvre Diana L Subbarao Padmaja Mandhane Piush Becker Allan McNagny Kelly M Sears Malcolm R Kollmann Tobias Mohn William W Turvey Stuart E Brett Finlay B 2015 Early infancy microbial and metabolic alterations affect risk of childhood asthma Science Translational Medicine 7 307 307ra152 doi 10 1126 scitranslmed aab2271 PMID 26424567 S2CID 206687974 a b Stiemsma Leah T Turvey Stuart E 2017 Asthma and the microbiome Defining the critical window in early life Allergy Asthma amp Clinical Immunology 13 3 doi 10 1186 s13223 016 0173 6 PMC 5217603 PMID 28077947 Ipci Kagan Altintoprak Niyazi Muluk Nuray Bayar Senturk Mehmet Cingi Cemal 2016 The possible mechanisms of the human microbiome in allergic diseases European Archives of Oto Rhino Laryngology 274 2 617 626 doi 10 1007 s00405 016 4058 6 PMID 27115907 S2CID 27328940 Marino Eliana Richards James L McLeod Keiran H et al May 2017 Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes Nature Immunology 18 5 552 562 doi 10 1038 ni 3713 ISSN 1529 2916 PMID 28346408 S2CID 30078908 Bettelli E Carrier Y Gao W Korn T Strom TB Oukka M Weiner HL Kuchroo VK May 2006 Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells Nature 441 7090 235 8 Bibcode 2006Natur 441 235B doi 10 1038 nature04753 PMID 16648838 a b Saemann Marcus D Bohmig Georg A Osterreicher Christoph H et al December 2000 Anti inflammatory effects of sodium butyrate on human monocytes potent inhibition of IL 12 and up regulation of IL 10 production The FASEB Journal 14 15 2380 2382 doi 10 1096 fj 00 0359fje ISSN 0892 6638 PMID 11024006 S2CID 41553220 Gao Z Yin J Zhang J Ward R E Martin R J Lefevre M Cefalu W T Ye J 2009 Butyrate Improves Insulin Sensitivity and Increases Energy Expenditure in Mice Diabetes 58 7 1509 17 doi 10 2337 db08 1637 PMC 2699871 PMID 19366864 Mazidi Mohsen Rezaie Peyman Kengne Andre Pascal Mobarhan Majid Ghayour Ferns Gordon A 2016 Gut microbiome and metabolic syndrome Diabetes amp Metabolic Syndrome Clinical Research amp Reviews 10 2 S150 7 doi 10 1016 j dsx 2016 01 024 PMID 26916014 Chen Xiao d Souza Roshan Hong Seong Tshool 2013 The role of gut microbiota in the gut brain axis Current challenges and perspectives Protein amp Cell 4 6 403 14 doi 10 1007 s13238 013 3017 x PMC 4875553 PMID 23686721 a b Moeller Andrew H Li Yingying Ngole Eitel Mpoudi Ahuka Mundeke Steve Lonsdorf Elizabeth V Pusey Anne E Peeters Martine Hahn Beatrice H Ochman Howard 2014 11 18 Rapid changes in the gut microbiome during human evolution Proceedings of the National Academy of Sciences 111 46 16431 16435 Bibcode 2014PNAS 11116431M doi 10 1073 pnas 1419136111 ISSN 0027 8424 PMC 4246287 PMID 25368157 Brune A 2014 Symbiotic digestion of lignocellulose in termite guts Nature Reviews Microbiology 12 3 168 80 doi 10 1038 nrmicro3182 PMID 24487819 S2CID 5220210 a b Dietrich C Kohler T Brune A 2014 The cockroach origin of the termite gut microbiota patterns in bacterial community structure reflect major evolutionary events Applied and Environmental Microbiology 80 7 2261 69 Bibcode 2014ApEnM 80 2261D doi 10 1128 AEM 04206 13 PMC 3993134 PMID 24487532 a b Mikaelyan A Dietrich C Kohler T Poulsen M Sillam Dusses D Brune A 2015 Diet is the primary determinant of bacterial community structure in the guts of higher termites Molecular Ecology 24 20 5824 95 Bibcode 2015MolEc 24 5284M doi 10 1111 mec 13376 PMID 26348261 S2CID 206182668 Mikaelyan A Thompson C Hofer M Brune A 2016 The deterministic assembly of complex bacterial communities in germ free cockroach guts Applied and Environmental Microbiology 82 4 1256 63 doi 10 1128 AEM 03700 15 PMC 4751828 PMID 26655763 Further reading editReview articlesDe Preter Vicky Hamer Henrike M Windey Karen Verbeke Kristin 2011 The impact of pre and or probiotics on human colonic metabolism Does it affect human health Molecular Nutrition amp Food Research 55 1 46 57 doi 10 1002 mnfr 201000451 PMID 21207512 Maranduba Carlos Magno da Costa De Castro Sandra Bertelli Ribeiro Souza Gustavo Torres de Rossato Cristiano Da Guia Francisco Carlos Valente Maria Anete Santana Rettore Joao Vitor Paes Maranduba Claudineia Pereira Souza Camila Maurmann de Carmo Antonio Marcio Resende do MacEdo Gilson Costa Silva Fernando de Sa 2015 Intestinal Microbiota as Modulators of the Immune System and Neuroimmune System Impact on the Host Health and Homeostasis Journal of Immunology Research 2015 931574 doi 10 1155 2015 931574 PMC 4352473 PMID 25759850 Prakash Satya Rodes Laetitia Coussa Charley Michael Tomaro Duchesneau Catherine Tomaro Duchesneau Catherine Coussa Charley Rodes 2011 Gut microbiota Next frontier in understanding human health and development of biotherapeutics Biologics Targets and Therapy 5 71 86 doi 10 2147 BTT S19099 PMC 3156250 PMID 21847343 Wu G D Chen J Hoffmann C Bittinger K Chen Y Y Keilbaugh S A Bewtra M Knights D Walters W A Knight R Sinha R Gilroy E Gupta K Baldassano R Nessel L Li H Bushman F D Lewis J D 2011 Linking Long Term Dietary Patterns with Gut Microbial Enterotypes Science 334 6052 105 08 Bibcode 2011Sci 334 105W doi 10 1126 science 1208344 PMC 3368382 PMID 21885731 Retrieved from https en wikipedia org w index php title Gut microbiota amp oldid 1189274841, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.