Category: Health

Metabolism and gut health

Metabolism and gut health

GF mice present Mindful eating for weight loss decreased colonic neuron density and intestinal Herbal remedies for eczema compared with Metaboliwm in specific Metabolism and gut health Yut mice. EMtabolism microbiota within the znd Metabolism and gut health signal to enteroendocrine EE cells Metabolis multiple pathways. Article PubMed PubMed Central CAS Google Scholar Sun, H. Provided by the Springer Nature SharedIt content-sharing initiative. The other authors declare that they have no competing interests. Veillonellaceae and Oscillospiraceae Firmicutes are negatively associated with 25 OH -D levels and vitamin D supplementation Bellerba et al. Whether exploring the potential of GLP-1 medications or embracing a holistic approach, the symbiotic connection between our gut and metabolism is an exciting frontier in the pursuit of optimal well-being. Metabolism and gut health

Metabolism and gut health -

Cole LK, Vance JE, Vance DE. Phosphatidylcholine biosynthesis and lipoprotein metabolism. Biochim Biophys Acta. Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A, et al.

Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI.

Obesity alters gut microbial ecology. Murphy EF, Cotter PD, Healy S, Marques TM, O'Sullivan O, Fouhy F, et al. Composition and energy harvesting capacity of the gut microbiota: relationship to diet, obesity and time in mouse models.

Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. De Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, et al.

Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Duncan SH, Lobley GE, Holtrop G, Ince J, Johnstone AM, Louis P, et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes Lond. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS.

TLR4 links innate immunity and fatty acid-induced insulin resistance. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al.

Richness of human gut microbiome correlates with metabolic markers. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome.

Chassaing B, Gewirtz AT. Has provoking microbiota aggression driven the obesity epidemic? Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, et al.

Dietary intervention impact on gut microbial gene richness. Vreugdenhil AC, Rousseau CH, Hartung T, Greve JW, van't Veer C, Buurman WA. Lipopolysaccharide LPS -binding protein mediates LPS detoxification by chylomicrons.

J Immunol. Neal MD, Leaphart C, Levy R, Prince J, Billiar TR, Watkins S, et al. Enterocyte TLR4 mediates phagocytosis and translocation of bacteria across the intestinal barrier.

Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Ghoshal S, Witta J, Zhong J, de Villiers W, Eckhardt E. Chylomicrons promote intestinal absorption of lipopolysaccharides. J Lipid Res.

Verges B, Duvillard L, Lagrost L, Vachoux C, Garret C, Bouyer K, et al. Changes in lipoprotein kinetics associated with type 2 diabetes affect the distribution of lipopolysaccharides among lipoproteins. J Clin Endocrinol Metab. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al.

Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Harte AL, Varma MC, Tripathi G, McGee KC, Al-Daghri NM, Al-Attas OS, et al. High fat intake leads to acute postprandial exposure to circulating endotoxin in type 2 diabetic subjects.

Diabetes Care. Ghanim H, Abuaysheh S, Sia CL, Korzeniewski K, Chaudhuri A, Fernandez-Real JM, et al. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implications for insulin resistance.

Russell WR, Hoyles L, Flint HJ, Dumas ME. Colonic bacterial metabolites and human health. Curr Opin Microbiol. DeMoss RD, Moser K. Tryptophanase in diverse bacterial species. J Bacteriol. CAS PubMed PubMed Central Google Scholar. Russell WR, Duncan SH, Scobbie L, Duncan G, Cantlay L, Calder AG, et al.

Major phenylpropanoid-derived metabolites in the human gut can arise from microbial fermentation of protein. Mol Nutr Food Res. Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4.

Ramadoss P, Marcus C, Perdew GH. Role of the aryl hydrocarbon receptor in drug metabolism. Expert Opin Drug Metab Toxicol. Cani PD, Osto M, Geurts L, Everard A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity.

Gut Microbes. Li YT, Cai HF, Wang ZH, Xu J, Fang JY. Systematic review with meta-analysis: long-term outcomes of faecal microbiota transplantation for Clostridium difficile infection.

Aliment Pharmacol Ther. Colman RJ, Rubin DT. Fecal microbiota transplantation as therapy for inflammatory bowel disease: a systematic review and meta-analysis. J Crohns Colitis. Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS, Bartelsman JF, et al.

Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Kobyliak N, Conte C, Cammarota G, Haley AP, Styriak I, Gaspar L, et al. Probiotics in prevention and treatment of obesity: a critical view.

Nutr Metab Lond. Article Google Scholar. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al.

Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM, Burcelin R. Improvement of glucose tolerance and hepatic insulin sensitivity by oligofructose requires a functional glucagon-like peptide 1 receptor.

Delzenne NM, Kok N. Effects of fructans-type prebiotics on lipid metabolism. Am J Clin Nutr. van Hoffen E, Ruiter B, Faber J, M'Rabet L, Knol EF, Stahl B, et al. A specific mixture of short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides induces a beneficial immunoglobulin profile in infants at high risk for allergy.

Daddaoua A, Puerta V, Requena P, Martinez-Ferez A, Guadix E, de Medina FS, et al. Goat milk oligosaccharides are anti-inflammatory in rats with hapten-induced colitis. J Nutr. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia.

Effect of non-digestible fermentable carbohydrates on hepatic fatty acid metabolism. Biochem Soc Trans. Tazoe H, Otomo Y, Karaki S, Kato I, Fukami Y, Terasaki M, et al. Expression of short-chain fatty acid receptor GPR41 in the human colon.

Biomed Res. Karaki S, Tazoe H, Hayashi H, Kashiwabara H, Tooyama K, Suzuki Y, et al. Expression of the short-chain fatty acid receptor, GPR43, in the human colon.

J Mol Histol. Zhou J, Hegsted M, McCutcheon KL, Keenan MJ, Xi X, Raggio AM, et al. Peptide YY and proglucagon mRNA expression patterns and regulation in the gut. Obesity Silver Spring. Zhou J, Martin RJ, Tulley RT, Raggio AM, McCutcheon KL, Shen L, et al. Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents.

Am J Physiol Endocrinol Metab. Delzenne NM, Cani PD, Neyrinck AM. Modulation of glucagon-like peptide 1 and energy metabolism by inulin and oligofructose: experimental data.

Garidou L, Pomie C, Klopp P, Waget A, Charpentier J, Aloulou M, et al. The gut microbiota regulates intestinal CD4 T cells expressing RORgammat and controls metabolic disease. Cell Metab. Ivey KL, Hodgson JM, Kerr DA, Lewis JR, Thompson PL, Prince RL. The effects of probiotic bacteria on glycaemic control in overweight men and women: a randomised controlled trial.

Eur J Clin Nutr. Rajkumar H, Mahmood N, Kumar M, Varikuti SR, Challa HR, Myakala SP. Effect of probiotic VSL 3 and omega-3 on lipid profile, insulin sensitivity, inflammatory markers, and gut colonization in overweight adults: a randomized, controlled trial.

Mediators Inflamm. Sanchez M, Darimont C, Drapeau V, Emady-Azar S, Lepage M, Rezzonico E, et al. Effect of Lactobacillus rhamnosus CGMCC1. Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults.

Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal.

Vulevic J, Juric A, Tzortzis G, Gibson GR. A mixture of trans-galactooligosaccharides reduces markers of metabolic syndrome and modulates the fecal microbiota and immune function of overweight adults. Vulevic J, Drakoularakou A, Yaqoob P, Tzortzis G, Gibson GR.

Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture B-GOS in healthy elderly volunteers. Zhang C, Yin A, Li H, Wang R, Wu G, Shen J, et al. Dietary modulation of gut microbiota contributes to alleviation of both genetic and simple obesity in children.

Ussar S, Griffin NW, Bezy O, Fujisaka S, Vienberg S, Softic S, et al. Interactions between gut microbiota, host genetics and diet modulate the predisposition to obesity and metabolic syndrome. Download references. CLB had a PhD scholarship funded by Nestlé RDLS and is currently funded by Metabometrix Ltd.

Metabometrix Ltd, Bio-incubator, Prince Consort Road, South Kensington, London, SW7 2BP, UK. Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2PH, UK.

Ana Luisa Neves, Julien Chilloux, Jeremy K. You can also search for this author in PubMed Google Scholar. Correspondence to Jeremy K. Nicholson or Marc-Emmanuel Dumas. CLB, ALN, JC, and M-ED were involved in drafting the manuscript. CLB, ALN, JC, M-ED, and JKN were involved in critically revising the manuscript for the intellectual content.

All the authors read and approved the final manuscript. Open Access This article is distributed under the terms of the Creative Commons Attribution 4. Reprints and permissions.

Boulangé, C. et al. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med 8 , 42 Download citation. Published : 20 April Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative. Skip to main content. Search all BMC articles Search. Download PDF. Review Open access Published: 20 April Impact of the gut microbiota on inflammation, obesity, and metabolic disease Claire L.

Boulangé 1 , Ana Luisa Neves 2 , Julien Chilloux 2 , Jeremy K. Abstract The human gut harbors more than trillion microbial cells, which have an essential role in human metabolic regulation via their symbiotic interactions with the host.

Full size image. Obesity and the metabolic syndrome Obesity is characterized by an excess of adipose tissue and occurs when an imbalance exists between energy intake and energy expenditure [ 14 ]. Link between impaired insulin action, low-grade inflammation, and obesity In healthy individuals, insulin triggers glucose uptake in peripheral organs and the secretion of this hormone is activated by the rise in postprandial plasma glucose concentration.

Interactions between gut microbes and host metabolism in the physiopathology of obesity and the metabolic syndrome Although genetic variants have been associated with susceptibility to developing obesity and type 2 diabetes, the heritability of these variants is fairly modest.

The gut microbiota affects calorie harvest and energy homeostasis A body of evidence shows that the gut microbiota helps to harvest energy and increase host fat storage [ 33 , 34 ]. Therapeutic potential of manipulating the gut microbial ecology The study of the metabolic, signaling, and immune interactions between gut microbes and the host, and how these interactions modulate host brain, muscle, liver and gut functions, has raised the concept of therapeutic microbial manipulation to combat or prevent diseases [ 4 , 10 ].

Conclusions and future directions The evidence for a strong contribution of the gut microbiota to the onset of obesity and metabolic diseases is growing. References Bruzzese E, Volpicelli M, Squaglia M, Tartaglione A, Guarino A.

PubMed Google Scholar Costello EK, Stagaman K, Dethlefsen L, Bohannan BJ, Relman DA. Article CAS PubMed PubMed Central Google Scholar Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, et al.

Article CAS PubMed Google Scholar Hooper LV, Littman DR, Macpherson AJ. Article CAS PubMed PubMed Central Google Scholar Abrams GD, Bishop JE. Article CAS PubMed Google Scholar Rajilic-Stojanovic M, de Vos WM.

Article CAS PubMed PubMed Central Google Scholar Nicholson JK, Wilson ID. Article CAS PubMed Google Scholar Neves AL, Chilloux J, Sarafian MH, Rahim MB, Boulange CL, Dumas ME. Article CAS PubMed Google Scholar Nicholson JK, Holmes E, Wilson ID.

Article CAS PubMed Google Scholar Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. Article PubMed PubMed Central Google Scholar Nazli A, Yang PC, Jury J, Howe K, Watson JL, Soderholm JD, et al. Article PubMed PubMed Central Google Scholar Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, et al.

Article PubMed PubMed Central Google Scholar World Health Organization WHO. Accessed 2 April Alberti KG, Zimmet P, Shaw J. Article PubMed Google Scholar Despres JP, Lemieux I, Bergeron J, Pibarot P, Mathieu P, Larose E, et al.

Article CAS PubMed Google Scholar Saito I. Article CAS PubMed Google Scholar Kahn SE, Hull RL, Utzschneider KM. Article CAS PubMed Google Scholar Perry RJ, Samuel VT, Petersen KF, Shulman GI.

Article CAS PubMed PubMed Central Google Scholar Saltiel AR, Kahn CR. Article CAS PubMed Google Scholar Delarue J, Magnan C. Article CAS Google Scholar Gregor MF, Hotamisligil GS.

Article CAS PubMed Google Scholar Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Article CAS PubMed Google Scholar Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, et al.

Article CAS PubMed PubMed Central Google Scholar Sell H, Habich C, Eckel J. Article CAS PubMed Google Scholar Lumeng CN, Saltiel AR. Article CAS PubMed PubMed Central Google Scholar Tanti JF, Ceppo F, Jager J, Berthou F.

Google Scholar Piya MK, McTernan PG, Kumar S. The utilization of these substrates is mainly the result of cross-feeding between gut microbiota members, rather than host absorption. Three main strategies for this activity exist in the human gut: 1 acetogens, for example, Blautia spp.

A higher abundance of these cross-feeders may improve the overall efficiency of metabolism in the gut; for example, an increase in methanogens is observed in the GI tract of anorexia nervosa patients, which may be a coping strategy by the gut microbiota in response to a lack of food sources [ 78 , 79 ].

Sulfate-reducing bacteria are the most efficient of the hydrogenotrophs, but require a source of sulfate; in the gut, the most prominent source of sulfate is sulfated glycans [ 80 ]. Although some of these glycans may be obtained from the diet, the most accessible source is mucin produced by the host [ 38 ].

Sulfate-reducing bacteria obtain sulfate from these substrates via cross-feeding with microbes such as Bacteroides , which produce sulfatases [ 80 , 81 ].

Hydrogen sulfide is both directly toxic to IECs through inhibition of mitochondrial cytochrome C oxidase, and pro-inflammatory via activation of T helper 17 cells [ 82 , 83 ]. Hydrogen sulfide can additionally directly act on disulfide bonds in mucin to further facilitate mucin degradation [ 84 ].

Elevated hydrogen sulfide concentrations and increased proportions of sulfate-reducing bacteria are reported in IBD [ 85 ]. The digestibility of proteins by the host is more variable than that of carbohydrates and fats, and is influenced by the previously mentioned factors of food processing, macronutrient ratios, and transit time [ 14 , 18 ], in addition to its source e.

The extra steps of interconversion required for amino acid fermentation yield a large diversity of by-products. However, it is important to note that not all amino acids are fermented to toxic products as a result of gut microbial activity; in fact, the most abundant end products are SCFAs [ 13 , 14 ].

Therefore, it may not be protein catabolism per se that negatively impacts the host, but instead specific metabolisms or overall increased protein fermentation activity.

It is thus important to examine these subtleties. A microbe can exhibit one of two strategies for the initial step of amino acid catabolism, either deamination to produce a carboxylic acid plus ammonia or decarboxylation to produce an amine plus carbon dioxide [ 12 ].

Ammonia can inhibit mitochondrial oxygen consumption and decrease SCFA catabolism by IECs, which has led to the assumption that excess ammonia production can negatively impact the host [ 87 , 88 , 89 ].

However, the gut microbiota also rapidly assimilates ammonia into microbial amino acid biosynthetic processes [ 13 ], and host IECs can additionally control ammonia concentration through conversion to citrulline and glutamine, or through slow release into the bloodstream [ 90 , 91 ].

It is thus unclear how much protein catabolism is necessary to achieve toxic ammonia concentrations, and this may vary between hosts. This uncertainty, coupled with the multiple negative impacts amines can have on the host discussed below , have led to speculation that deamination would improve host outcomes.

Fortunately, deamination appears to be the more common strategy of amino acid catabolism by the gut microbiota, because high concentrations of SCFAs are produced from amino acid degradation via this pathway [ 12 , 13 ].

The next steps depend on the class of amino acid starting substrate, with most eventually resulting in tricarboxylic acid cycle intermediates, pyruvate, or coenzyme A-linked SCFA precursors [ 39 , 75 ]. An exception would be the series of Stickland reactions exhibited by certain Clostridia , in which a coupled oxidation and reduction of two amino acids occurs as an alternative to using hydrogen ions as the electron acceptor [ 40 , 41 ].

Phosphate is simultaneously added to the reduced amino acid in this case, and thus oxidative phosphorylation for the production of ATP can occur directly from the resultant acyl phosphate.

In turn, branched-chain fatty acids BCFAs , such as isovalerate and isobutyrate, can be produced as end-products. Additionally, some gut microbial species, mainly from the class Bacilli, also possess a specialized branched-chain keto acid dehydrogenase complex to yield energy from the oxidized forms of the branched-chain amino acids directly, which also leads to BCFA production [ 13 , 75 ].

The major SCFA and BCFA products generated from degradation of each amino acid are presented in Table 2.

BCFAs are often used as a biomarker of protein catabolism, with the promoted goal to reduce their concentration in order to improve health outcomes [ 14 ].

However, little is actually known about the impact of BCFAs on host health. In fact, preliminary work has shown that BCFAs are able to modulate glucose and lipid metabolism in the liver similarly to SCFAs [ 93 ], and isobutyrate can be used as a fuel source by IECs when butyrate is scarce [ 94 ].

What is undisputed, however, are the negative consequences of the pro-inflammatory, cytotoxic, and neuroactive compounds yielded from the sulfur-containing, basic and aromatic amino acids.

Catabolism of the sulfur-containing amino acids, cysteine and methionine, results in the production of hydrogen sulfide and methanethiol, respectively [ 13 , 14 ], and a large number of taxonomically diverse bacterial species contain the requisite degradative enzymes within their genomes, including members of the Proteobacteria phylum, the Bacilli class, and the Clostridium and Bifidobacterium genera [ 13 , 75 ].

Hydrogen sulfide can be methylated to methanethiol, which can be further methylated to dimethyl sulfide, and this methylation is thought to be part of the detoxification process due to the progressively less toxic nature of these compounds [ 95 ].

However, methanethiol may also be converted to hydrogen sulfide, then oxidized to sulfate, for detoxification; this sulfate can then be utilized by sulfate-reducing bacteria [ 80 , 81 , 95 ].

Indeed, this latter reaction has been observed in cecal tissue, and is part of the sulfur cycle of the gut [ 96 ]. A wide diversity of bacterial species within the gut microbiota can decarboxylate the basic amino acids, thus resulting in the formation of amine by-products shown in Additional file 1 , including bifidobacteria, clostridia , lactobacilli, enterococci, streptococci, and members of the Enterobacteriaceae family [ 97 ].

Agmatine inhibits the proliferation of IECs, which is thought to stem from its ability to reduce the synthesis and promote the degradation of other polyamines [ 98 ]. This effect may not be negative depending on the context; for example, the resultant decrease of fatty acid metabolism in tissues reduced both weight gain and the hormonal derangements associated with obesity in rats fed a high fat chow [ 99 ].

Agmatine also may be anti-inflammatory through inhibition of nitric oxide synthase [ ], and is a candidate neurotransmitter, with agonism for α 2 -adenoceptors and imidazoline binding sites, while simultaneously blocking ligand-gated cation channels NMDA class [ ].

The latter activity has therapeutic potential for remediating some forms of hyperalgesia and for its neuroprotectivity. Putrescine, on the other hand, is essential for the proliferation of IECs [ ]. All three polyamines improve the integrity of the gut by increasing expression of tight junction proteins [ ], promoting intestinal restitution [ ] and increasing mucus secretion [ , ].

Finally, both putrescine and spermine are able to inhibit the production of pro-inflammatory cytokines, such as IL-1, IL-6, and TNF-α [ , ]. Therefore, any benefits of agmatine must be weighed against its consequent reduction of these polyamines; it may be effective in the treatment of certain conditions such as metabolic syndrome but could be detrimental in excess under normal conditions.

Arginine can additionally be converted to glutamate, which can be deaminated to produce 4-aminobutryate GABA. GABA is the major inhibitory neurotransmitter of the central nervous system, and alterations in the expression of its receptor have been linked to the pathogenesis of depression and anxiety [ ].

Administration of lactobacilli and bifidobacteria that produce GABA to mice and rats has resulted in a decrease of depressive behaviors, a reduction of corticosterone induced stress and anxiety, and lessened visceral pain sensation [ , , ].

GABA can additionally regulate the proliferation of T cells and thus has immunomodulatory properties [ ]. Interestingly, chronic GI inflammation not only induces anxiety in mice, but depression and anxiety often present comorbidity with GI disorders, including irritable bowel syndrome IBS [ , ].

The catabolism of histidine can produce histamine Additional file 1. Histamine may be synonymous with its exertion of inflammation in allergic responses, but bacterially produced histamine has actually been shown to inhibit the production of the pro-inflammatory cytokines TNF-α in vivo [ ], and IL-1, and IL in vitro [ ], while simultaneously preventing intestinal bacterial translocation.

Histamine is also a neurotransmitter, modulating several processes such as wakefulness, motor control, dendritic cell activity, pain perception, and learning and memory [ ]. The catabolism of lysine can produce cadaverine Additional file 1.

Cadaverine is a poorly studied metabolite; it can be toxic, but only in high amounts [ 13 , 97 ]. Cadaverine has, however, been shown to potentiate histamine toxicity [ ] and higher concentrations of cadaverine are associated with ulcerative colitis UC [ ].

Aromatic amino acid degradation can yield a wide diversity of indolic and phenolic compounds that can act as toxins or neurotransmitters as shown in Additional file 2.

The catabolism of tryptophan can produce tryptamine and indoles Additional file 2. Tryptamine is a neurotransmitter that plays a role in regulating intestinal motility and immune function [ ]. Particularly, it is able to interact with both indoleamine 2,3-dioxygenase and the aryl hydrocarbon receptor to heighten immune surveillance, and dampen the expression of pro-inflammatory cytokines, respectively [ , ].

A lack of these activities has therefore been implicated in the pathology of IBD; although, it should be noted that most tryptophan metabolites can interact with these receptors, thus it is not tryptamine-specific [ 13 , , ].

Tryptamine can also both potentiate the inhibitory response of cells to serotonin and induce its release from enteroendocrine cells [ , ]. Serotonin is a neurotransmitter involved in many processes including mood, appetite, hemostasis, immunity, and bone development [ 13 , ].

Its dysregulation is thus reported in many disorders, including IBD [ ], IBS [ ], cardiovascular disease [ ], and osteoporosis [ ]. Tryptophan decarboxylation is a rare activity among species of the gut microbiota, but certain Firmicutes have been found to be capable of it, including the IBD-associated species, Ruminococcus gnavus [ , ].

Indole, on the other hand, is a major bacterial metabolite of tryptophan, produced by many species of Bacteroides and Enterobacteriaceae [ ].

It plays an important role in host defense, by interacting with the pregnane X receptor and the aryl hydrocarbon receptor [ ]. This activity fortifies the intestinal barrier by increasing tight junction protein expression and downregulates the expression of pro-inflammatory cytokines [ , ].

It also induces glucagon like peptide-1 an incretin secretion by enteroendocrine cells, inhibiting gastric secretion and motility, to promote satiety [ , ]. Indole is additionally a signaling molecule for bacteria, influencing motility, biofilm formation, antibiotic resistance, and virulence, and shown to inhibit the colonization capabilities of pathogens such as Salmonella enterica [ ].

However, indole overproduction can increase its export to the liver, where it is sulfated to indoxyl sulfate, a uremic toxin associated with chronic kidney disease [ ].

Further, its effects as a signaling molecule for both enteroendocrine cells and bacteria are dose dependent, with high concentrations rendering it ineffective [ , , ]. The catabolism of tyrosine can produce tyramine, phenols, and p-coumarate Additional file 2.

Tyramine is a neurotransmitter that can be produced by certain gut bacteria via decarboxylation, including Enterococcus and Enterobacteriaceae [ 97 ]. Tyramine facilitates the release of norepinephrine that induces peripheral vasoconstriction, elevates blood glucose levels, and increases cardiac output and respiration [ ].

It has also been shown to increase the synthesis of serotonin by enteroendocrine cells in the gut, elevating its release into circulation [ ]. Phenol and p-cresol are phenolic metabolites that have been shown to both decrease the integrity of the gut epithelium and the viability of IECs [ , ], and can be produced by many gut bacterial species, such as members of the Enterobacteriaceae and Clostridium clusters I, XI, and XIVa [ ].

P-cresol in particular is genotoxic, elevates the production of superoxide, and inhibits proliferation of IECs [ ]. P-cresol may additionally be sulfated to cresyl sulfate in the gut or liver, which has been found to suppress the T helper 1-mediated immune response in mice [ ], and, interestingly, phenolic sulfation was found to be impaired in the gut mucosa of UC patients [ ].

Indeed, the colonic damage induced by unconjugated phenols is similar to that observed in IBD [ ]. Cresyl sulfate is also associated with chronic kidney disease, however, as it can damage renal tubular cells through induction of oxidative stress [ ].

This compound is also particularly elevated in the urine of autistic patients, but a causative link in this case has not been elucidated [ ].

The catabolism of phenylalanine can produce phenylethylamine and trans-cinnamic acid Additional file 2. Unlike tyrosine and tryptophan, little is known about these phenylalanine-derived metabolites.

Through facilitating the release of catecholamine and serotonin, phenylethylamine in turn elevates mood, energy, and attention [ ]. However, it has been reported that ingesting phenylethylamine can induce headache, dizziness, and discomfort in individuals with a reduced ability to convert it to phenylacetate, suggesting excessive amounts have negative consequences [ ].

These metabolic pathways were found to so far specifically occur within species of Clostridium and Peptostreptococcus , respectively [ , ]. The chlorogenic acid phenotype is associated with both autism and schizophrenia, suggesting a role of altered aromatic amino acid metabolism in these disorders [ , , ].

However, further research is still needed, as there remains no mechanistic explanation of these metabolites toward disease development. Further, both trans-cinnamic acid and p-coumaric acid are negatively associated with cardiovascular disease [ , ]. P-coumaric acid, in particular, is a common phenolic compound derived from plant matter that has anti-inflammatory properties, and has been demonstrated to prevent platelet aggregation [ ].

Thus, these metabolites may simply be an indicator of altered microbial metabolism in general, when found in excess. Microorganisms in the gut are known to possess lipases, which can degrade triglycerides and phospholipids into their polar head groups and free lipids [ 16 , ].

Certain bacteria inhabiting the GI tract, including species of lactobacilli, enterococci, clostridia, and Proteobacteria, can utilize the backbone of triglycerides as an electron sink, reducing glycerol to 1,3-propanediol [ ].

Reuterin has antimicrobial properties acting against pathogens and commensals alike [ ], but it can also be spontaneously dehydrated to acrolein [ 71 ].

Acrolein is a highly reactive genotoxin, with an equivalent mutagenic potency to formaldehyde, raising concerns about this metabolic process [ 71 , ].

Meanwhile, choline can additionally be metabolized to trimethylamine by species of the gut microbiota, particularly Clostridia especially members of Clostridium cluster XIVa and Eubacterium spp.

and Proteobacteria [ , ]. Trimethylamine is oxidized in the liver to trimethylamine N-oxide [ , ], which exacerbates atherosclerosis by promoting the formation of foam cells lipid-laden macrophages [ ] and altering cholesterol transport [ ]. High levels of serum trimethylamine N-oxide are thus associated with cardiovascular disease [ ] and atherosclerosis [ ].

However, it should be noted that active research in these areas is in its early stages, and thus the link between the gut microbiota-mediated lipid head group metabolism and health consequences is still unclear.

For example, a study on the metabolism of glycerol by fecal microbial communities found that only a subset could reduce it to 1,3-propanediol, and the authors did not detect any reuterin [ ]. Further, some members of the gut microbiota e.

In contrast to the polar head groups, microorganisms are not thought to have the ability to catabolize free lipids in the anaerobic environment of the gut [ ]. However, free lipids have antimicrobial properties [ , ] and can directly interact with host pattern recognition receptors.

Particularly, saturated fatty acids are TLR4 agonists that promote inflammation [ ], whereas omega-3 unsaturated fatty acids are TLR4 antagonists that prevent inflammation [ ]. Interestingly, chronic inflammation co-occurring with obesity has been well described [ ], and could be a result of the aforementioned pro-inflammatory properties of free lipids, the lack of anti-inflammatory SCFAs produced from carbohydrate fermentation high-fat diets tend to be low in carbohydrates , or a combination of both.

High-fat diets do have a reported impact on the composition of the gut microbiota, yet it is unclear whether it is the increased fat content per se or the relative decrease in carbohydrates, which often accompanies these diets, that is the chief influencer [ 16 , ].

Indeed, Morales et al. observed that a high-fat diet including fiber supplementation induces inflammation without altering the composition of the gut microbiota [ 16 ]. Regardless, the gut microbiota is required for the development of obesity, as shown in GF mice experiments, because of the ability of SCFAs to alter energy balance as previously discussed [ ].

Metabolism of exogenous substrates greatly affects the use of endogenous substrates by the gut microbiota. Dietary fiber reduces the degradation of mucin, and the utilization of mucin is thought to cycle daily depending on the availability of food sources [ , ].

Mucin is a sulfated glycoprotein [ 38 ], thus the same concepts of carbohydrate and protein degradation from dietary sources discussed above apply. However, it should be noted that mucin turnover by the gut microbiota is a naturally occurring process, and only when it occurs in elevated amounts does it have negative connotations.

For example, Akkermansia muciniphila is a mucin-utilizing specialist that is depleted in the GI tract of IBD [ ] and metabolic syndrome [ ] patients. muciniphila has a demonstrated ability to cross-talk with host cells, promoting an increase in concentration of glucagon-like peptides, 2-arabinoglycerol, and antimicrobial peptides that improve barrier function, reduce inflammation, and induce proliferation of IECs [ ].

Through this communication, A. muciniphila also, paradoxically, restored the thickness of the mucin layer in obese mice. Dietary fat intake can also alter the profile of bile acids. Dairy-derived saturated lipids increase the relative amount of taurine-conjugation, and this sulfur-containing compound leads to the expansion of sulfate-reducing bacteria in the gut [ ].

Bile acid turnover is, however, a naturally occurring process, which modulates bile acid reabsorption, inflammation, triglyceride control, and glucose homeostasis from IEC signaling [ ]. The critical contributions of the gut microbiota toward human digestion have just begun to be elucidated.

Particularly, more recent research is revealing how the impacts of microbial metabolism extend beyond the GI tract, denoting the so-called gut-brain e. The primary focus to date has been on the SCFAs derived mainly from complex carbohydrates, and crucial knowledge gaps still remain in this area, specifically on how the SCFAs modulate glucose metabolism and fat deposition upon reaching the liver.

However, the degradation of proteins and fats are comparatively less well understood. Due to both the diversity of metabolites that can be yielded and the complexity of microbial pathways, which can act as a self-regulating system that removes toxic by-products, it is not merely a matter of such processes effecting health positively or negatively, but rather how they are balanced.

Further, the presentation of these substrates to the gut microbiota, as influenced by the relatively understudied host digestive processes occurring in the small intestine, is equally important. Future work could therefore aim to determine which of these pathways are upregulated and downregulated in disease states, such as autism and depression gut-brain , NAFLD gut-liver , chronic kidney disease gut-kidney , and cardiovascular disease gut-heart.

Further, a combination of human- and culture- in vitro and in vivo based studies could resolve the spectrum of protein and fat degradation present among healthy individuals, in order to further our understanding of nutrient cycling in gut microbial ecosystems, and thus gain a necessary perspective for improving wellness.

Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. Article CAS PubMed Google Scholar. Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett C, Knight R, Gordon JI.

The human microbiome project: exploring the microbial part of ourselves in a changing world. Article CAS PubMed PubMed Central Google Scholar. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome.

Article CAS Google Scholar. Belkaid Y, Hand T. Role of the microbiota in immunity and inflammation. Spiljar M, Merkler D, Trajkovski M.

The immune system bridges the gut microbiota with systemic energy homeostasis: focus on TLRs, mucosal barrier, and SCFAs. Front Immunol. Article PubMed PubMed Central CAS Google Scholar. Hillman ET, Lu H, Yao T, Nakatsu CH. Microbial ecology along the gastrointestinal tract.

Microbes Environ. Article PubMed PubMed Central Google Scholar. Martinez-Guryn K, Hubert N, Frazier K, Urlass S, Musch MW, Ojeda P, et al. Small intestine microbiota regulate host digestive and absorptive adaptive responses to dietary lipids.

Cell Host Microbe. Sommer F, Anderson JM, Bharti R, Raes J, Rosenstiel P. The resilience of the intestinal microbiota influences health and disease.

Nat Rev Microbiol. Theriot CM, Young VB. Interactions between the gastrointestinal microbiome and Clostridium difficile. Annu Rev Microbiol. Stecher B, Hardt W-D. Mechanisms controlling pathogen colonization of the gut. Curr Opin Microbiol. Fan P, Li L, Rezaei A, Eslamfam S, Che D, Ma X.

Metabolites of dietary protein and peptides by intestinal microbes and their impacts on gut. Curr Protein Pept Sci. Portune KJ, Beaumont M, Davila A-M, Tomé D, Blachier F, Sanz Y.

Gut microbiota role in dietary protein metabolism and health-related outcomes: the two sides of the coin. Trends Food Sci Technol. Yao CK, Muir JG, Gibson PR.

Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther. Krajmalnik-Brown R, Ilhan Z-E, Kang D-W, DiBaise JK. Effects of gut microbes on nutrient absorption and energy regulation.

Nutr Clin Pract Off Publ Am Soc Parenter Enter Nutr. Article Google Scholar. Morales P, Fujio S, Navarrete P, Ugalde JA, Magne F, Carrasco-Pozo C, et al.

Impact of dietary lipids on colonic function and microbiota: an experimental approach involving orlistat-induced fat malabsorption in human volunteers. Clin Transl Gastroenterol. Wong JMW, Jenkins DJA.

Carbohydrate digestibility and metabolic effects. J Nutr. Roager HM, Hansen LBS, Bahl MI, Frandsen HL, Carvalho V, Gøbel RJ, et al. Colonic transit time is related to bacterial metabolism and mucosal turnover in the gut. Nat Microbiol.

Degen LP, Phillips SF. Variability of gastrointestinal transit in healthy women and men. Biesalski HK. Nutrition meets the microbiome: micronutrients and the microbiota. Ann N Y Acad Sci. Article PubMed Google Scholar. Ozdal T, Sela DA, Xiao J, Boyacioglu D, Chen F, Capanoglu E.

The reciprocal interactions between polyphenols and gut microbiota and effects on bioaccessibility. Bhattacharya T, Ghosh TS, Mande SS. Global profiling of carbohydrate active enzymes in human gut microbiome.

PLoS One. Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, Chiang HC, et al. A genomic view of the human- Bacteroides thetaiotaomicron symbiosis.

Singh RK, Chang H-W, Yan D, Lee KM, Ucmak D, Wong K, et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. Wolfe AJ. Glycolysis for the microbiome generation.

Microbiol Spectr. Vergnolle N. Protease inhibition as new therapeutic strategy for GI diseases. Lin R, Liu W, Piao M, Zhu H. A review of the relationship between the gut microbiota and amino acid metabolism. Amino Acids.

Smith EA, Macfarlane GT. Enumeration of amino acid fermenting bacteria in the human large intestine: effects of pH and starch on peptide metabolism and dissimilation of amino acids. FEMS Microbiol Ecol. Geboes KP, De Hertogh G, De Preter V, Luypaerts A, Bammens B, Evenepoel P, et al.

The influence of inulin on the absorption of nitrogen and the production of metabolites of protein fermentation in the colon. Br J Nutr. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon.

Cell Metab. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, et al. Population-level analysis of gut microbiome variation. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome.

Genome Med. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Macfarlane GT, Macfarlane S. Bacteria, colonic fermentation, and gastrointestinal health.

J AOAC Int. Mountfort DO, Grant WD, Clarke R, Asher RA. Eubacterium callanderi sp. that demethoxylates O-methoxylated aromatic acids to volatile fatty acids.

Int J Syst Evol Microbiol. CAS Google Scholar. McDonald JAK, Mullish BH, Pechlivanis A, Liu Z, Brignardello J, Kao D, et al. Inhibiting growth of Clostridioides difficile by restoring Valerate, produced by the intestinal microbiota.

Article PubMed CAS Google Scholar. Wolf PG, Biswas A, Morales SE, Greening C, Gaskins HR. H2 metabolism is widespread and diverse among human colonic microbes.

Gut Microbes. Tailford LE, Crost EH, Kavanaugh D, Juge N. Mucin glycan foraging in the human gut microbiome. Front Genet.

Louis P, Flint HJ. Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol. Fischbach MA, Sonnenburg JL. Eating for two: how metabolism establishes interspecies interactions in the gut. de Vladar HP. Amino acid fermentation at the origin of the genetic code.

Biol Direct. Lopetuso LR, Scaldaferri F, Petito V, Gasbarrini A. Commensal clostridia: leading players in the maintenance of gut homeostasis. Gut Pathog. Pokusaeva K, Fitzgerald GF, van Sinderen D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr. Jumas-Bilak E, Carlier J-P, Jean-Pierre H, Teyssier C, Gay B, Campos J, et al.

Veillonella montpellierensis sp. Paixão L, Oliveira J, Veríssimo A, Vinga S, Lourenço EC, Ventura MR, et al.

Host glycan sugar-specific pathways in Streptococcus pneumonia : galactose as a key sugar in colonisation and infection. Duncan SH, Hold GL, Harmsen HJM, Stewart CS, Flint HJ.

Growth requirements and fermentation products of Fusobacterium prausnitzii , and a proposal to reclassify it as Faecalibacterium prausnitzii gen. CAS PubMed Google Scholar. Charalampopoulos D, Pandiella SS, Webb C.

Growth studies of potentially probiotic lactic acid bacteria in cereal-based substrates. J Appl Microbiol. Taras D, Simmering R, Collins MD, Lawson PA, Blaut M.

Reclassification of Eubacterium formicigenerans Holdeman and Moore as Dorea formicigenerans gen. Holdeman LV, Moore WEC. New genus, Coprococcus , twelve new species, and emended descriptions of four previously described species of bacteria from human feces.

Google Scholar. Liu C, Finegold SM, Song Y, Lawson PA. Reclassification of Clostridium coccoides, Ruminococcus hansenii, Ruminococcus hydrogenotrophicus, Ruminococcus luti, Ruminococcus productus and Ruminococcus schinkii as Blautia coccoides gen. and description of Blautia wexlerae sp.

Roh H, Ko H-J, Kim D, Choi DG, Park S, Kim S, et al. Complete genome sequence of a carbon monoxide-utilizing Acetogen, Eubacterium limosum KIST J Bacteriol.

Polansky O, Sekelova Z, Faldynova M, Sebkova A, Sisak F, Rychlik I. Important metabolic pathways and biological processes expressed by chicken cecal microbiota. Appl Environ Microbiol. Article CAS PubMed Central Google Scholar.

Sakamoto M, Benno Y. Reclassification of Bacteroides distasonis , Bacteroides goldsteinii and Bacteroides merdae as Parabacteroides distasonis gen. and Parabacteroides merdae comb. Rautio M, Eerola E, Väisänen-Tunkelrott M-L, Molitoris D, Lawson P, Collins MD, et al. Reclassification of Bacteroides putredinis Weinberg et al.

Syst Appl Microbiol. Kaneuchi C, Miyazato T, Shinjo T, Mitsuoka T. Taxonomic study of helically coiled, Sporeforming anaerobes isolated from the intestines of humans and other animals: Clostridium cocleatum sp.

and Clostridium spiroforme sp. Yutin N, Galperin MY. A genomic update on clostridial phylogeny: gram-negative spore-formers and other misplaced clostridia. CAS PubMed PubMed Central Google Scholar. Liang K, Shen CR. Selection of an endogenous 2,3-butanediol pathway in Escherichia coli by fermentative redox balance.

Metab Eng. Chassard C, Delmas E, Robert C, Lawson PA. Bernalier-Donadille a. Ruminococcus champanellensis sp. Mashima I, Liao Y-C, Miyakawa H, Theodorea CF, Thawboon B, Thaweboon S, et al. Veillonella infantium sp. Elshaghabee FMF, Bockelmann W, Meske D, de Vrese M, Walte H-G, Schrezenmeir J, et al.

Ethanol production by selected intestinal microorganisms and lactic acid bacteria growing under different nutritional conditions. Front Microbiol. Kelly WJ, Henderson G, Pacheco DM, Li D, Reilly K, Naylor GE, et al.

The complete genome sequence of Eubacterium limosum SA11, a metabolically versatile rumen acetogen. Stand Genomic Sci. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism.

den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D-J, Bakker BM. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M.

In addition, one study suggests that bacterial fermentation of fiber increased GLP-1 by increasing the number of GLP-1 secreting L-cells. The effect can also go the other way: Some bacteria impair GLP-1 receptors, leading to GLP-1 resistance. Bile acids are another example. Produced by the liver and released into the small intestine, they are processed by gut bacteria, and can also stimulate GLP-1 secretion from the L-cells.

So changes in the gut microbiome that reduce SCFA-producing bacteria or bile acid-processing bacteria may have consequences in GLP-1 signaling, which can impact our ability to process glucose efficiently. Chronic inflammation underlies many metabolic disorders.

One pathway to that inflammation is when elements in the gut leak through the gut lining and enter our system—a condition termed metabolic endotoxemia.

What prevents that leaking is the so-called gut barrier , a multi-layer physical and functional system separating the gut from the rest of the body. The gut barrier is lined with a single, intact layer of epithelial cells similar to cells that cover the surface of the body, such as the skin that absorbs nutrients and protects from invading pathogens.

A layer of mucus covers the inside of that gut epithelium; on the other side are immune cells tasked with surveillance against microbes that may attempt to escape from the gut and enter the body. Studies in animal models show that the microbiome regulates different components of this gut barrier defense mechanism.

The mucus is created by specialized goblet cells in the gut lining to coat and protect the epithelial cells from microbes and digestive enzymes. This viscous layer is also infused with a diverse array of antimicrobial agents and antibodies as additional protection.

Without a gut microbiome, germ-free mice have a thinner mucosal layer and fewer antimicrobial agents—traits often associated with metabolic and immunological diseases.

Similarly, a fiber-deprived diet shifts the gut microbiome to consume rather than build up the mucus, resulting in a compromised gut barrier. On the other hand, probiotic supplementation and fiber-rich dietary interventions can restore a healthy mucus barrier.

Covered and protected by the mucosa is the selectively permeable barrier made of epithelial cells. The cells are bound together by specialized tight junction proteins that allow only desired nutrients to pass through.

In many inflammatory diseases , these tight junction proteins are reduced or not functional, allowing harmful microbes and toxins to enter. The gut microbiome also influences tight junctions , which can, at least in animals, be restored by probiotics and dietary fiber.

A blood biomarker, Lipopolysaccharide LPS , allows us to measure a dysfunctional gut barrier. Probiotics , dietary fiber , and other nutritional interventions have been shown to lower the level of blood LPS, as well as markers of inflammation, in people with obesity and diabetes. Inflammation helps heal your body, but chronic inflammation can cause serious damage.

Beyond the gut barrier, the gut microbiome can also shape the immune system to be more resilient to fight invaders while being less damaging to the body. Inflammation is a double-edged sword: it protects the body from infection and injury, but excessive inflammation can be damaging.

Therefore a balance, or immune homeostasis , is crucial. Unfortunately, the modern lifestyle puts the body under constant stress, and the result is a chronic low-grade inflammation that underlies many diseases.

The gut is home to the largest number of immune cells within the body, and the gut microbiome and its metabolites are known drivers of the immune repertoire. In addition to the antimicrobial function at the mucosal barrier, normal gut bacteria help develop immune cells that fight pathogenic microbes.

Other bacteria directly increase regulatory T cells , an important immune cell type that suppresses inflammation and keeps the body from attacking itself. Again, microbially produced SCFA are important players behind this mechanism , along with vitamin A-derived retinoic acids.

On the flip side, dietary and probiotic-mediated gut microbiome changes have been shown to blunt inflammation, presenting possible microbiome-based solutions for inflammatory diseases. Diet shapes the gut microbiome. The modern Western diet is low on fiber and high in unnatural additives—this combination reduces or removes certain groups of microbes from the gut.

When those conditions persist, the weakened gut microbiome damages our immune and metabolic functions in the ways shown above, which further harms the microbiome, creating a negative feedback loop.

While we need more controlled human-intervention studies, research thus far suggests that some simple interventions can reduce damages in microbiome-regulated metabolic and immune functions.

The benefits of fiber-rich food are well known— fiber slows your digestion so you stay fuller longer, and it promotes blood-sugar and cholesterol control. We have seen above that the gut microbiome has everything to do with these benefits. Fiber-rich foods— such as beans , legumes, fresh vegetables , nuts , seeds, fruit , and whole grains —are particularly good at supporting the growth of beneficial gut microbes that regulate metabolic hormones and reduce inflammation.

In addition, polyphenol-rich foods, including green tea and berries, appear to modulate the gut microbiome and increase beneficial groups of bacteria. Prebiotic supplements pack specific types of fiber known to feed selective groups of bacteria that confer health benefits.

Taking prebiotic supplements is an excellent way to ensure you foster the growth of these bacteria. Inulin, which occurs naturally in high concentrations in chicory root and Jerusalem artichokes, is a fiber source that supports the desirable microbes and is available in multiple prebiotic supplements.

If your gut microbiome has already suffered from a prolonged Western diet, just adding back fiber in the short-term will not be enough to completely restore beneficial bacteria.

While both may be beneficial, it is important to understand that you can maximize the benefit by understanding the needs of your very unique gut microbiome. These needs have been identified in studies that have profiled the microbiome changes associated with various diseases, and may sometimes be echoed in personal microbiome testing results.

Pendulum Therapeutics, Inc. has authored this review article, with editorial assistance from Levels.

Trouble losing weight continues to be a struggle, despite decades of advice to bealth calories. Metabolusm Type diabetes treatment advancements our standard American Metabolidm is Herbal remedies for eczema in saturated fats and very gjt in fiber, this creates a perfect storm for breeding bad bacteria and wiping out the good bacteria in your gut. Our digestive tract is host to an entire ecosystem of microorganisms that break down our food, create vitamins, and control our immune system. Healthy humans are likely to have a more diverse species of gut bacteria. The food you eat breaks down to its simplest form during digestion and also feeds your gut bacteria. The Metabolism and gut health community of the gut conveys significant benefits to host physiology. A ane relationship has now been hea,th between Metabolim bacteria and host Herbal remedies for eczema in which microbial-mediated gut hormone release Merabolism an important role. Within Skin rejuvenation gut lumen, Herbal remedies for eczema Antioxidant-rich foods for liver detoxification a number of metabolites and contain Heart-friendly choices components gkt act as signaling molecules to a number of cell types within the mucosa. Enteroendocrine cells within the mucosal lining of the gut synthesize and secrete a number of hormones including CCK, PYY, GLP-1, GIP, and 5-HT, which have regulatory roles in key metabolic processes such as insulin sensitivity, glucose tolerance, fat storage, and appetite. Release of these hormones can be influenced by the presence of bacteria and their metabolites within the gut and as such, microbial-mediated gut hormone release is an important component of microbial regulation of host metabolism. Dietary or pharmacological interventions which alter the gut microbiome therefore pose as potential therapeutics for the treatment of human metabolic disorders.

Video

Pierre DESTROYS Trudeau UNTIL Palestine Protesters BREAK IN

Author: Doushicage

0 thoughts on “Metabolism and gut health

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com