Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-05-01T07:09:32.448Z Has data issue: false hasContentIssue false

SCFA: mechanisms and functional importance in the gut

Published online by Cambridge University Press:  02 April 2020

Camille Martin-Gallausiaux
Affiliation:
Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
Ludovica Marinelli
Affiliation:
Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350Jouy-en-Josas, France
Hervé M. Blottière
Affiliation:
Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350Jouy-en-Josas, France Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, MetaGenoPoliS, 78350Jouy-en-Josas, France
Pierre Larraufie
Affiliation:
Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 75005Paris, France
Nicolas Lapaque*
Affiliation:
Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350Jouy-en-Josas, France
*
*Corresponding author: Nicolas Lapaque, email nicolas.lapaque@inrae.fr
Rights & Permissions [Opens in a new window]

Abstract

In recent years, the importance of the gut microbiota in human health has been revealed and many publications have highlighted its role as a key component of human physiology. Owing to the use of modern sequencing approaches, the characterisation of the microbiome in healthy individuals and in disease has demonstrated a disturbance of the microbiota, or dysbiosis, associated with pathological conditions. The microbiota establishes a symbiotic crosstalk with their host: commensal microbes benefit from the nutrient-rich environment provided by the gut and the microbiota produces hundreds of proteins and metabolites that modulate key functions of the host, including nutrient processing, maintenance of energy homoeostasis and immune system development. Many bacteria-derived metabolites originate from dietary sources. Among them, an important role has been attributed to the metabolites derived from the bacterial fermentation of dietary fibres, namely SCFA linking host nutrition to intestinal homoeostasis maintenance. SCFA are important fuels for intestinal epithelial cells (IEC) and regulate IEC functions through different mechanisms to modulate their proliferation, differentiation as well as functions of subpopulations such as enteroendocrine cells, to impact gut motility and to strengthen the gut barrier functions as well as host metabolism. Recent findings show that SCFA, and in particular butyrate, also have important intestinal and immuno-modulatory functions. In this review, we discuss the mechanisms and the impact of SCFA on gut functions and host immunity and consequently on human health.

Type
Conference on ‘Diet and Digestive Disease’
Copyright
Copyright © The Authors 2020

Human subjects are colonised, at birth, by bacteria, archaea, fungi and viruses, which are collectively called microbiota. Distinct microbiota inhabit all epithelial surfaces of the body: skin, oral cavity, respiratory, gastrointestinal and reproductive tracts(Reference Reid, Younes and Van der Mei1); with the largest and most diverse microbiota residing in the colon. The intestinal microbiota is composed of 100 trillions of bacteria which represent about 25 times as many genes as our own Homo sapiens genome. The diversity and complexity of the microbiota is influenced by the host genetic background, the diet and the environment. Reciprocally, this microbiota encodes thousands of genes absent in human genome that exert diverse functions often associated with beneficial physiological effects for its host(Reference Lepage, Leclerc and Joossens2Reference Rakoff-Nahoum, Kong and Kleinstein4). From this close symbiotic relationship emerged the notion that human subjects and their microbiota form a composite organism, namely a holobiont(Reference van de Guchte, Blottiere and Dore5). Advances in next-generation sequencing and bioinformatics tools have shown that this relationship is far more complex than anticipated. Indeed, over the past decade, studies highlighted that perturbation of the microbiota, referred to as dysbiosis, and loss of bacterial diversity affect different host systems, particularly metabolic and immuneo processes, that participate in host physiology and pathophysiologic conditions(Reference Lepage, Leclerc and Joossens2). Moreover, growing lines of evidence suggest that the dialogue between microbiota and the host systems has a homoeostatic role beyond the gut, and contributes directly to the global wellbeing of the host. In agreement with this, animal studies have demonstrated that microbiota is implicated in liver diseases, allergy, diabetes, airway hypersensitivity, autoimmune arthritis and even neurological disorders(Reference Atarashi and Honda6Reference Benakis, Brea and Caballero8).

The human body has evolved to functionally interact with thousands of naturally occurring or microbiota-derived metabolites. Thus, the intestinal microbiome provides an extended repertoire of molecules and metabolites that influence the host health. Amongst those molecules, SCFA, derived from bacteria-dependent hydrolysis of fibres, have attracted considerable attention because of their role in host health (Fig. 1a). Indeed, decreased abundance of SCFA-producing bacteria or decreased genomic potential for SCFA-production have been identified in many studies such as type-1 diabetes, type-2 diabetes, liver cirrhosis, inflammatory bowel diseases and atherosclerosis(Reference Qin, Li and Raes9Reference Vatanen, Franzosa and Schwager14). Here, we aim to provide an overview of bacterial SCFA production in the gut, their impact on intestinal cells and host functions, and their different mechanisms of action.

Fig. 1. (a) Functional impact of SCFA on the host. (b) Mechanisms: (1) G protein-coupled receptor (GPCR)-dependent signalling, (2) histone and transcription factor acetylation by SCFA and (3) role of butyrate as a ligand of transcription factors. AhR, aryl hydrocarbon receptor; ARNT, aryl hydrocarbon receptor nuclear translocator; HAT, histone acetyltransferase; K/HDAC, lysine/histone deacetylase; MAPK, mitogen-activated protein kinase; PLC, phospholipase C; TF, transcription factor; XRE, xenobiotic response element.

SCFA production and transport

Production of SCFA

Complex dietary carbohydrates are metabolised by intestinal microbiota through an extensive set of enzymes, mostly absent in mammals and belonging to the large family of carbohydrate-active enzymes (reviewed in(Reference Garron and Henrissat15)). The degradation of dietary fibres by gut microbiota produces organic acids, gases and a large amount of SCFA. Acetate (C2), propionate (C3) and butyrate (C4) are the main SCFA produced (60:20:20 mm/kg in human colon). SCFA can reach a combined concentration of 50–150 mm mainly in the colon where the microbial biomass is the highest(Reference Duncan, Holtrop and Lobley16Reference Macfarlane and Macfarlane19). Substrates for bacterial fermentation include non-digestible carbohydrates derived from dietary fibres such as polysaccharide plant cell walls, resistant starch, soluble oligosaccharide and endogenous products, such as mucin(Reference Dalile, Van Oudenhove and Vervliet20). Besides bacterial fermentation, SCFA can also be found in plant oil and animal fats. Butter contains 3–4 % of butyrate in the form of tributyrin(Reference Bourassa, Alim and Bultman21). However, when fermentable fibre supply decreases, some bacterial species can switch to amino acids and protein fermentation as an alternative energy source, also contributing to SCFA and branched chain fatty acid production(Reference Smith and Macfarlane22,Reference Davila, Blachier and Gotteland23) . The branched chain fatty acids, i.e. isovalerate, 2-methylbutyrate and isobutyrate, are present at lower concentrations compared to SCFA and originate only from protein breakdown. Acetate is a net fermentation product for most gut bacteria while butyrate and propionate are produced by more specific bacterial species. Butyrate is produced from acetate, lactate, amino acids and various carbohydrates via glycolysis from two different pathways, the butyryl-CoA:acetate CoA-transferase or the phosphotransbutyrylase and butyrate kinase pathway. Using Fluorescence In Situ Hybridization probes and PCR, Flint and colleagues have shown that specific families belonging to the Clostridiales order (Firmicutes) have the capabilities to produce butyrate: Lachnospiraceae (Coprococcus, Eubacterium, Anaerostipes and Roseburia), Ruminococcaceae (Faecalibacterium and Subdoligranulum) and Erysipelotrichaceae (Holdemanella)(Reference Flint24Reference Louis and Flint26). The butyrate-producing capability of Clostridiales has been confirmed using in vitro culture in other genera such as Clostridium, Butyrivibrio, Lachnoclostridium, Marvinbryantia, Oscillibacter, Flavonifractor, Erysipelatoclostridium, Anaerotruncus, Dorea, Blautia and Ruminiclostridium (Reference Martin-Gallausiaux, Larraufie and Jarry27,Reference Martin-Gallausiaux, Beguet-Crespel and Marinelli28) . Propionate is produced in the gut from various substrates, including amino acids, carbohydrates, lactate and 1,2-propanediol. Hence, most hexoses and pentoses enter the succinate pathway and result in succinate production, a precursor of propionate. The succinate pathway is present in Bacteroidetes and some Firmicutes, such as the Negativicutes (Veillonella and Phascolarctobacterium). Some other Firmicutes, belonging to Negativicutes (Megasphaera), Lachnospiraceae (Coprococcus) and Ruminococcaceae, use the acrylate pathway, in which lactate is the substrate to produce propionate. The propanediol pathway is present in Proteobacteria and Lachnospiraceae species and use deoxyhexose sugars (e.g. fucose) as substrates. The commensal bacterium Akkermansia muciniphila, member of the Verrucomicrobia phylum also produces propionate from this latter pathway(Reference Belzer, Chia and Aalvink29). Some bacteria, notably in the Lachnospiraceae family, can produce both propionate and butyrate but from different substrates, e.g. Roseburia inulivorans (Reference Reichardt, Duncan and Young30).

In vitro experiments have shown that Bacteroides growth is reduced relative to Firmicutes and Actinobacteria because SCFA negatively impact Bacteroides at mild acid pH(Reference Duncan, Louis and Thomson31). Thus, SCFA production by Firmicutes and Bacteroides may to be regulated by pH variations, with more Firmicutes fermentation in the proximal colon (pH about 5⋅6) and conditions favouring Bacteroides fermentation in the distal colon with a more neutral pH (pH about 6⋅3)(Reference Hamer, Jonkers and Venema32). This selective gradient is limiting the propionate production and promoting butyrate formation in the more proximal part of the colon(Reference Flint24). Intestinal pH is not the only factor that impact microbiota composition and consequently SCFA production. Indeed, intestinal gas production (e.g. oxygen and hydrogen) and diet composition and intake (e.g. types of fibres and iron) have been reported to influence the microbiota composition and the gut SCFA concentration(Reference Dostal, Lacroix and Bircher33,Reference Dostal, Lacroix and Pham34) .

Transport of SCFA

In the host, SCFA have distinct roles depending of their absorption and local physiologic concentrations(Reference Conn, Fell and Steele35,Reference Frost, Sleeth and Sahuri-Arisoylu36) . Acetate, propionate and butyrate are weak acids with pK a 4⋅8 for butyrate. Under physiological conditions the colonic pH range from 5⋅5 to 6⋅7, thus most SCFA are in the ionised form and require transporters for absorption(Reference Cummings, Pomare and Branch37,Reference Bergman38) . SCFA transporters are expressed at different levels: in the small intestine: monocarboxylate transporter (MCT)1 (SLC16A1), sodium-coupled MCT(SMCT)2 (SLC5A12) and SLC16A7 and in the colon: MCT1 (SLC16A1), SMCT2 (SLC5A12), SMCT1 (SL5CA8) and SLC26A3(Reference Dalile, Van Oudenhove and Vervliet20,Reference Sivaprakasam, Bhutia and Yang39) . The transporters MCT1, SMCT1 and SLC26A3 show affinities for all three major SCFA while the other ones are more selective, e.g. SMCT2 only transports butyrate. Butyrate is mainly absorbed via MCT1 that is expressed both at apical and basolateral membrane of colonic epithelial cells(Reference Sivaprakasam, Bhutia and Yang39,Reference Sepponen, Ruusunen and Pakkanen40) . From approximately 20 mm in gut lumen, butyrate concentration on portal vein reaches a range of 5–10 μm. The liver significantly uptakes butyrate as there is almost no splanchnic release(Reference Bloemen, Venema and van de Poll41,Reference van der Beek, Bloemen and van den Broek42) . Butyrate venous concentration ranges from 0⋅5 to 3⋅3 μm(Reference Hamer, Jonkers and Venema32). Similarly, a larger amount of propionate is found in portal vein, about 32 μm, but there is only a very small release from the liver. Venous concentration of propionate ranges from 3⋅8 to 5⋅4 μm. In contrast, acetate is weakly absorbed by epithelial cells and the liver. The portal vein concentration of acetate is 98–143 μm. Hence, the liver efficiently metabolises the butyrate and propionate released by the gut epithelium and avoids any acute increase even in the case of artificial enema(Reference Hamer, Jonkers and Venema32,Reference Bloemen, Venema and van de Poll41) .

Cellular uptake of SCFA in their anionic form is through H+- or Na+-coupled transporters. Thus, butyrate transport directly participates in electrolyte absorption with increases of Na+ and Cl absorption and release of bicarbonate (HCO3) in the lumen(Reference Sivaprakasam, Bhutia and Yang39,Reference McNeil, Cummings and James43,Reference Binder and Mehta44) . Interestingly, electrolyte absorption is region specific due to different transporter expression levels in each gut region(Reference Lutz and Scharrer45). Transport of butyrate is electroneutral through SMCT2 (Na+), resulting in the transport of one Na+ for each butyrate anion absorbed(Reference Srinivas, Gopal and Zhuang46). On the contrary, SMCT1 transport is electrogenic as two Na+ are transported with one butyrate anion. This results in electrolytes and water absorption(Reference Coady, Chang and Charron47,Reference Gupta, Martin and Prasad48) . MCT1 is a proton-coupled transporter and has no direct role in ion transport. However, MCT1 indirectly regulates bicarbonate secretion through Na+/H+ and Cl/HCO3 exchangers. Interestingly, butyrate modulates the expression of many transporters including MCT1 and SMCT1, therefore potentially increasing electrolyte exchanges as well as its own transport. Butyrate blocks Cl secretion by inhibiting Na-K-2Cl cotransporter expression and increases expression of the Na+/H+ transporter NHE3 through histone deacetylase (HDAC) inhibition and a specificity protein dependent pathway(Reference Matthews, Hassan and Meng49Reference Subramanya, Rajendran and Srinivasan52).

Mechanisms

SCFA receptors

The human genome encodes for six potential G protein-coupled receptors (GPCR) sensitive to SCFA: GPR41 (FFAR3), GPR42, GPR43 (FFAR2), GPR109a (HCAR2), GPR164 (OR51E1) and OR51E2. GPR41 and GPR109a are exclusively Gαi/o-coupled receptors whereas GPR43 can be coupled to either Gαβγq and Gαi/o and OR51E2 is αs coupled(Reference Gelis, Jovancevic and Veitinger53). GPR42 has recently been identified as a functional GPCR-modulating Ca2+ channel flux, but only the Gβγ pathway downstream this receptor was explored(Reference Puhl, Won and Lu54). GPR41, GPR43 and GPR109a are expressed in numerous organs including the small and large intestine by various cell types: immune cells, adipose tissues, heart, skeletal muscle or neurons(Reference Dalile, Van Oudenhove and Vervliet20). GPR43 (FFAR2) and GPR41 (FFAR3) recognise acetate, butyrate and propionate with affinities that differ between species, whereas only butyrate activates GPR109a (Fig. 1b)(Reference Le Poul, Loison and Struyf55Reference Hudson, Tikhonova and Pandey58). Schematically, GPR41 activation by propionate and butyrate and GPR109a activation by butyrate lead to the inhibition of cyclic adenosine monophosphate (cAMP) accumulation and protein kinase A and mitogen-activated protein kinases (ERK and p38) activation. Conversely, GPR43 is activated by the three main SCFA with approximately the same affinities. GPR43 engagement stimulates the phospholipase-Cβ, which releases intracellular Ca2+ and activates protein kinase C in addition to cAMP accumulation inhibition and protein kinase A and ERK activation(Reference Dorsam and Gutkind59). The highly polymorphic GPR42 is activated by propionate and modulates Ca2+ by a yet unknown mechanism that could be similar to GPR43 due to the very high homology between these two receptors. In human subjects, GPR42 is expressed in the colon and in sympathetic ganglia(Reference Puhl, Won and Lu54). Butyrate is the ligand of GPR164 (OR51E1) expressed all along the gastrointestinal tract and specifically by enteroendocrine cells (EEC)(Reference Priori, Colombo and Clavenzani60,Reference Han, Kang and Oh61) . The olfactory receptor OR51E2 (Olfr78 in mouse) is activated by propionate and acetate and result in cAMP and Ca2+ increase. Olfr78 is expressed at the gut mucosal level by peptide YY (PYY)-positive colonic EEC(Reference Fleischer, Bumbalo and Bautze62). It is also detected in various tissues, including kidney, blood vessels, lung and specific nerves in the heart and gut(Reference Pluznick63).

Transcriptional regulations and post-translational modifications

SCFA have a broad impact on the host: metabolism, differentiation, proliferation mainly due to their impact on gene regulation. Indeed, several studies revealed that butyrate regulates the expression of 5–20 % of human genes(Reference Basson, Liu and Hanly64Reference Donohoe, Collins and Wali66). Within the cells, butyrate and propionate exhibit strong inhibition capacity of lysine and histone deacetylase (K/HDAC) activity, with butyrate being more potent than propionate(Reference Candido, Reeves and Davie67,Reference Sealy and Chalkley68) . Moreover, butyrate is metabolised into acetyl-CoA which stimulates histone acetyltransferase by further enhancing histone acetylation (Fig. 1b)(Reference Donohoe, Collins and Wali66,Reference Donohoe and Bultman69) . By their HDAC inhibitor and histone acetyltransferase stimulatory properties, SCFA promote post-translational modification of histones through increasing their acetylation. Histone hyperacetylation leads to an increased accessibility of transcription factors to the promoter regions of targeted genes owing to the modulation of their transcription. HDAC inhibition by butyrate does not only up-regulate gene transcription, repression of several genes such as LHR, XIAP or IDO-1 has been reported(Reference Martin-Gallausiaux, Larraufie and Jarry27,Reference Bose, Dai and Grant70) . In a colonic cell line, 75 % of the upregulated genes are dependent of the ATP citrate lyase activity and 25 % are independent at 0⋅5 mm concentration but the proportion is reversed at high concentration (5 mm). This suggests that the gene regulation mechanisms are different, depending on the butyrate concentration. It has been shown that butyrate does not only tune the histone acetylation level but also acetylation of other proteins, including transcription factors such as SP1 and Foxp3(Reference Arpaia, Campbell and Fan71,Reference Thakur, Dasgupta and Ta72) . SCFA derived from the gut microbiota also promote crotonylation through their histone acetylase properties(Reference Fellows, Denizot and Stellato73). Histone crotonylation is abundant in the small and large bowel epithelium as well as in the brain. Crotonyl-CoA modification of histones is linked to the cell cycle regulation. Moreover, several studies have shown that butyrate also modifies DNA and protein methylation and phosphorylation levels(Reference Boffa, Gruss and Allfrey74Reference Parker, de Haan and Gevers76).

Novel role of butyrate as a ligand of transcription factors

Besides the extensive described effects of SCFA on host physiology through GPR and post-translational modifications, a novel role emerged for butyrate as a ligand of two transcription factors, expanding our knowledge on microbial–host crosstalk. By exploring the mechanisms involved in the microbial modulation of angiopoietin-like protein 4, Alex and co-workers demonstrated that SCFA induce angiopoietin-like protein 4 transcription and secretion through a novel role as the selective modulator of PPARγ in colonic cell lines(Reference Alex, Lange and Amolo77). In this study, Alex and co-workers showed that butyrate promotes, similarly to PPARγ ligands, the interactions between PPARγ and multiple coactivators and binds into PPARγ binding pocket with a conformation similar to the known PPARγ agonist, decanoic acid(Reference Alex, Lange and Amolo77). The evidence suggests, for the first time, an original function of butyrate as a ligand for a transcription factor. This original mechanism was also reported for another nuclear transcription factor, the aryl hydrocarbon receptor in human colonic cell lines(Reference Marinelli, Martin-Gallausiaux and Bourhis78). This latter study described a ligand-dependent activation of human aryl hydrocarbon receptor by butyrate in synergy with its role as a HDAC inhibitor. By using selective ligand antagonists and structural modelling, it emerges that butyrate activates human aryl hydrocarbon receptor by binding into its ligand binding pocket similarly to the aryl hydrocarbon receptor ligand FICZ(Reference Marinelli, Martin-Gallausiaux and Bourhis78). Together, these reports provide an expanded view of the possible mechanisms for butyrate to modulate human transcription factor activity that might apply to other transcription factors (Fig. 1b).

Functional impact of SCFA on the host

SCFA, regulators of the gut metabolism, proliferation and differentiation

SCFA are efficiently taken up from the gut lumen by the intestinal epithelial cells (IEC) with different fates (Fig. 1b). Butyrate is the primary energy source of IEC, being oxidised via β-oxidation in the mitochondria. This catabolic process represents from 73 to 75 % of oxygen consumption by human colonocytes, by which part of butyrate is converted into ketone bodies(Reference Fleming, Choi and Fitch79Reference Roediger81). The main substrates of colonocytes are by order of preference, butyrate > ketone bodies > amino acid > glucose. By using a high level of oxygen, the colonocyte metabolism maintains epithelial hypoxia with an oxygen partial pressure <1 % oxygen (7⋅6 mmHg), thus favouring anaerobic commensals(Reference Furuta, Turner and Taylor82). The capacity to produce ketone bodies and oxidise butyrate is a crucial difference between the small and large bowel. Epithelial cell butyrate oxidative capacity has been determined in vitro to be between 1 and 5 mm butyrate, therefore when a greater concentration is available, SCFA can affect cell functions such as K/HDAC inhibition(Reference Donohoe and Bultman69,Reference Andriamihaja, Chaumontet and Tome83) . Moreover, butyrate absorption increases the pyruvate dehydrogenase kinases which negatively regulates the pyruvate dehydrogenase complex. The pyruvate dehydrogenase decarboxylates pyruvate to produce acetyl-CoA and NADH, both necessary to tricarboxylic acid(Reference Blouin, Penot and Collinet84). This dual action pushes the colonocyte metabolism from glycolysis to β-oxidation. After transport into the cells, butyrate enhances oxidative phosphorylation, which consumes oxygen(Reference Andriamihaja, Chaumontet and Tome83). Similarly, it has been demonstrated that fatty acid oxidation is reduced in germ-free mice compared to conventional mice(Reference Donohoe, Garge and Zhang85). Butyrate is not the only fatty acid metabolised. Acetate is a substrate for cholesterol and fatty acid synthesis and is metabolised in muscles. Propionate is a precursor for the synthesis of glucose in the liver(Reference Dalile, Van Oudenhove and Vervliet20,Reference Louis, Hold and Flint25,Reference Donohoe, Garge and Zhang85) . Acetate and butyrate are also major substrates for lipogenesis in rat colonocytes(Reference Zambell, Fitch and Fleming86).

Through the production of SCFA, gut microbiota actively communicates with host cells and strongly modulates a variety of cellular mechanisms. Two of the main functions influenced by SCFA and thus gut microbiota are cell proliferation and differentiation. Indeed, the proliferative activity of crypt epithelial cells as well as the migration of mature epithelial cells along the crypt–villus axis are greatly attenuated in antibiotic-treated and germ-free mice(Reference Park, Kotani and Konno87). In the physiological state, butyrate favours cell differentiation and inhibits proliferation. First, evidences on IEC were demonstrated on cell lines(Reference Augeron and Laboisse88,Reference Barnard and Warwick89) . In these studies, long-term incubation of intestinal cancerous cell lines with SCFA resulted in differentiated phenotypes coupled to decreased cell proliferation. High concentration of butyrate is associated with inhibition of stem cells and proliferative cells in the crypts, through a HDAC inhibition-dependent binding of Foxo3 to promoters of key genes in the cell cycle(Reference Kaiko, Ryu and Koues90). Butyrate concentration near the crypt base is estimated to be 50–800 μm dose equivalent(Reference Donohoe, Garge and Zhang85,Reference Csordas91,Reference Sengupta, Muir and Gibson92) . These studies indicate that butyrate concentration is low in the deep crypts and increasing in a gradient along the lumen-to-crypt axis. Butyrate metabolisation by differentiated cells on the epithelium plateau may result in a protective depletion in the crypts that is protective for stem cell proliferation. Hence, the crypt structure is suggested to be an adaptive mechanism protecting the gut epithelial stem cells of butyrate high concentration found in the lumen(Reference Kaiko, Ryu and Koues90).

Interestingly, butyrate has a dual role in epithelial cellular metabolisms: it supports healthy cells as primary energy source for IEC and represses cancerous cell expansion. This is known as the butyrate paradox or Warburg effect(Reference Donohoe, Collins and Wali66). This is explained by a metabolic shift occurring in cancerous cells using preferentially glucose as the energy source. The inhibition of cell proliferation is generally characterised by an increase in reactive oxygen species production, DNA damages and cell cycle arrest, suggesting that SCFA initiate apoptosis signalling in cancer cells(Reference Arun, Madhavan and Reshmitha93Reference Verma, Agarwal and Das96). Indeed, through the activation of the pro-apoptotic protein BAX, the upregulation of apoptosis-inducing factor-mitochondria associated 1 isoform 6 and the reduction of mitochondrial membrane potential, SCFA stimulate the cytochrome c release which drives caspase 3 activation(Reference Arun, Madhavan and Reshmitha93). Coherently, the induction of the cyclin-dependent kinase inhibitors p21 and p27 and the downregulation of heat-shock cognate 71 kDa protein isoform is observed, leading to growth arrest(Reference Kim, Kwon and Ryu97,Reference Wang, Chiang and Chu98) .

Another mechanism for propionate to inhibit cell proliferation is suggested to involve its role as GPCR agonist. In human monocyte and lymphoblast cancer cell lines, Bindels and colleagues observe that the effect on cell proliferation is dependent on GPR43 activation(Reference Bindels, Porporato and Dewulf99). GPR43 displays a dual coupling through Gi and Gq protein families. While phospholipase C blockage does not influence cell proliferation, increase in cAMP, mediated by the inhibition of Gi subunit, slightly reduces the propionate anti-proliferative effect, suggesting a mechanism dependent on cAMP levels(Reference Bindels, Porporato and Dewulf99).

Considering the important metabolic shift occurring in cancer cells, the production and availability of a large variety of metabolites are modified among which acetyl-CoA. Acetyl-CoA is crucial in several metabolic pathways and a fundamental cofactor for histone acetyltransferases. Consequently, different cell metabolites are produced, such as a large amount of lactate, which in turn could stimulate the growth of commensal bacteria and partially explain the anti-tumorigenic effect of some probiotics(Reference Casanova, Azevedo-Silva and Rodrigues100).

Regulation of gut endocrine functions, importance on host physiology

Among IEC, EEC play an important role in host physiology by secreting hormones that regulate food intake, insulin secretion and gut functions in response to a variety of stimuli(Reference Gribble and Reimann101). Among these stimuli, fibre-rich diets or infusion with SCFA have been associated with increased circulating levels of gut hormones(Reference Cani, Amar and Iglesias102,Reference Samuel, Shaito and Motoike103) . Supporting these results, expression of butyrate receptors GPR43, GPR41 and GPR109a have been reported in EEC(Reference Karaki, Tazoe and Hayashi104Reference Nohr, Pedersen and Gille106). Acute stimulation of EEC by SCFA is shown to trigger hormone secretion such as glucagon-like peptide (GLP)-1 and PYY. The mechanism involves GPR43 activation leading to increased intracellular calcium, corresponding to the activation of a Gq-coupled receptor(Reference Tolhurst, Heffron and Lam107). Several studies have confirmed the role of GPR43 in the EEC response to SCFA using additional knockout models or agonists(Reference Bolognini, Tobin and Milligan108Reference Psichas, Sleeth and Murphy110). In particular EEC, the L-cells, GPR41 is also involved in the GLP-1 secretory response as suggested by the results in GPR41 knockout animals or GPR41 agonists(Reference Nohr, Pedersen and Gille106,Reference Tolhurst, Heffron and Lam107) . However, GPR41 stimulation also inhibits glucose insulinotropic polypeptide secretion from glucose insulinotropic polypeptide -producing EEC(Reference Lee, Zhang and Miyamoto111). This inhibition of glucose insulinotropic polypeptide-producing cells could correspond to the activation of Gi/o pathways which are mainly resulting in inhibitory responses. The exact role of GPR41 in GLP-1 secretion remains to be fully understood. The possibility of GPR41 hetero-dimerisation with GPR43 has been recently highlighted and could explain a role of GPR41 in GLP-1 stimulatory activity(Reference Ang, Xiong and Wu112). Additionally, species differences are described in response to the different SCFA. If propionate and acetate are strong stimuli for PYY and GLP-1 secretion in rodents at low concentrations, much higher concentrations are required to induce secretion in human subjects(Reference Psichas, Sleeth and Murphy110,Reference Chambers, Morrison and Frost113) . These divergences can be explained both by the variation of SCFA affinities to the receptor families as well as the different receptor expression levels. Indeed, GPR41 is expressed in fewer EEC in human subjects compared to rodents(Reference Nohr, Pedersen and Gille106,Reference Tazoe, Otomo and Karaki114) . The role of other SCFA receptors GPR109a, GPR42, OR51E1 and OR51E2, is still to be deciphered but some studies show that they are also enriched in some EEC subpopulations(Reference Fleischer, Bumbalo and Bautze62,Reference Roberts, Larraufie and Richards115) .

In addition to the SCFA-dependent acute stimulation of gut hormone secretion, it emerged that SCFA also tune EEC identity and consequently long-term hormonal production. Indeed, animals fed with fibre-rich diets have, in addition to a higher circulating gut hormone levels, an elevated number of EEC(Reference Cani, Amar and Iglesias102). Supporting this result, an increase in the differentiation of epithelial cells into L-cells by SCFA has been reported, with a higher GLP-1, PYY and serotonin production(Reference Samuel, Shaito and Motoike103,Reference Larraufie, Martin-Gallausiaux and Lapaque116Reference Zhou, Martin and Tulley120) . GPR43 and GPR41 play important but different roles in the differentiation of EEC. GPR43 stimulation increased the number of the PYY-producing cells and PYY expression but not the number of GLP-1-positive cells which is dependent on GPR41(Reference Larraufie, Martin-Gallausiaux and Lapaque116,Reference Brooks, Viardot and Tsakmaki117) .

Moreover, receptor-independent pathways are also involved in the expression regulation of gut hormone genes. Indeed, butyrate HDAC inhibitory activity highly increased PYY expression in human L-cells with a much stronger effect compared to GPR43 stimulation(Reference Larraufie, Martin-Gallausiaux and Lapaque116). The modulation of PYY gene expression is associated with increased production and secretion both under basal and stimulated conditions and could explain the long-term effects of SCFA on circulating gut hormone levels seen with fibre-enriched diets. Butyrate also impacts EEC responses to external stimuli by regulating the expression of receptors sensing exogenous molecules deriving from the microbiota. In particular, butyrate increases Toll-like receptor expressions in L-cells leading to an amplified stimulation by Toll-like receptor ligands and a consequent higher NF-κB activation and butyrate-dependent PYY expression(Reference Larraufie, Dore and Lapaque121).

Due to their important functions on host, gut hormones link SCFA and the modulation of other gut functions such as electrolyte absorption. Indeed, PYY is strongly associated with the modulation of electrolyte and water absorption functions due to the expression of neuropeptide Y receptors on epithelial cells and neuronal cells(Reference Cox122,Reference Pais, Rievaj and Larraufie123) . As SCFA stimulate PYY release, they impact electrolyte absorption(Reference Okuno, Nakanishi and Shinomura124). Similarly, serotonin is also important in water and electrolyte absorption. SCFA also increase serotonin production, and blockade of serotonin receptors decreases butyrate-dependent electrolyte absorption(Reference Reigstad, Salmonson and Rainey119,Reference Fukumoto, Tatewaki and Yamada125) . These results indicate that the regulation of electrolyte absorption by SCFA is mediated by multiple pathways including gut hormone modulations.

SCFA have also been associated with tuning of intestinal transit(Reference Fukumoto, Tatewaki and Yamada125). Acute effect of SCFA on gut motility is hormone dependent with an important role of PYY(Reference Cherbut, Ferrier and Roze126,Reference Cuche, Cuber and Malbert127) . Moreover, germ-free animals have decreased gut motility which is partially restored by SCFA infusion in the colonic lumen, with butyrate having the highest effect(Reference Vincent, Wang and Parsons128). The gut motility dysfunction in germ-free mouse could be partially explained by the highly dysregulated gut endocrine functions. However, no difference could be found in non-producing serotonin mouse model using TPH1 knockout mice(Reference Vincent, Wang and Parsons128). This suggests that serotonin might not play an important role in the SCFA-dependent regulation of gut motility and effects previously described could be minor compared to other pathways(Reference Fukumoto, Tatewaki and Yamada125). Interestingly, SCFA, and mostly butyrate, have a direct effect on gut motility through the regulation of enteric neurons(Reference Cherbut, Ferrier and Roze126). Indeed, some enteric neurons express GPR41 and can therefore respond to SCFA(Reference Nohr, Pedersen and Gille106). Additionally, HDAC inhibition by butyrate increases gut motility in the long term by increasing the number of acetylcholine and substance P positive neurons, highlighting the importance of distinct mechanisms triggering similar effects(Reference Soret, Chevalier and De Coppet129).

Butyrate and other SCFA are therefore important regulators of EEC functions, both by acutely stimulating gut hormone secretion, and modulating their production. Indeed, SCFA increase EEC subpopulation cell numbers and regulate gene expression. Different mechanisms including receptor activation and HDAC inhibition are involved in these functions, highlighting the important and diverse roles of SCFA as signalling molecules. Modulations of gut hormones participate in many roles of SCFA on host physiology including gut homoeostasis.

Barrier function and immune responses

In the past decade, SCFA have attracted considerable attention for their impact on host immune responses and barrier functions. SCFA play one of their major roles by maintaining an environment favourable for commensal bacteria and controlling pathogens’ growth. By stabilising the transcription factor HIF, butyrate increases VO2 by IEC favouring the physiologic hypoxia in the colon(Reference Kelly, Zheng and Campbell130). Maintenance of the colonic anaerobic environment is key to favour the anaerobe commensal component of the gut microbiota and control the pathogens’ level such as Salmonella in a virtuous cycle(Reference Faber, Tran and Byndloss131Reference Rivera-Chavez, Zhang and Faber133). However, enteric pathogens such as Salmonella enterica serovar Typhimurium are highly adapted to the colonic environment and utilise the gut microbiota-derived butyrate to compete with resident bacteria(Reference Bronner, Faber and Olsan134). Besides effect on the O2 level in the intestinal tract, butyrate promotes the epithelial barrier functions by reducing the epithelial permeability via HIF(Reference Kelly, Zheng and Campbell130). Moreover, butyrate reduces epithelial permeability by the regulation of IL-10 receptor, occludin, zonulin and claudins, reinforcing the tight junctions and the trans-epithelial resistance in vitro (Reference Zheng, Kelly and Battista135,Reference Wang, Wang and Wang136) . Another important mechanism involved in the epithelial barrier function is the modulation of the mucus layer thickness protecting the mucosa. In the colon, MUC2 is the predominant mucin glycoprotein produced by the goblet cells. Treatment with butyrate increases MUC2 production both in vitro and in human colonic biopsies(Reference Hamer, Jonkers and Venema32,Reference Gaudier, Jarry and Blottiere137) . SCFA enhance the epithelial barrier functions by modulating antimicrobial peptide secretion by the gut epithelium. Butyrate increases the level of colonic LL-37 in vitro and in vivo (Reference Hase, Eckmann and Leopard138,Reference Raqib, Sarker and Bergman139) . Activation of GPR43 by butyrate induce RegIIIγ and β-defensins expression by the activation of the mTOR pathway and STAT3 phosphorylation in mouse IEC(Reference Zhao, Chen and Wu140). The modulations of β-defensins in epithelial cells rely on the inhibition of HDAC(Reference Fischer, Sechet and Friedman141). Interestingly, SCFA and butyrate in particular, promote antimicrobial peptides targeting both Gram-positive and -negative bacteria.

It is now clear that gut microbiota plays an important role in intestinal homoeostasis by controlling the human immune response notably by the production of SCFA. Indeed, SCFA have a global anti-inflammatory effect by up-regulating both anti-inflammatory and down-regulating pro-inflammatory cytokines by different mechanisms and consequently promoting mucosal homoeostasis(Reference Maslowski, Vieira and Ng142). This anti-inflammatory effect can be mediated by IEC as binding of SCFA to GPR43 and GPR109a induces Ca2+ efflux and membrane hyperpolarisation which activate the inflammasome-activating protein NLRP3 thereby inducing the release of IL-18 with a protective effect on a dextran sulfate sodium colitis mouse model(Reference Macia, Tan and Vieira143). In vitro experiments demonstrate that the increase of protein acetylation by butyrate decreases IL-8 production in IEC(Reference Huang, Katz and Martin144). Moreover, butyrate, and to a lesser extent propionate, upregulate the production of TGFβ1 in IEC, a cytokine promoting anti-inflammatory regulatory T cells (Treg)(Reference Atarashi, Tanoue and Oshima145,Reference Atarashi, Tanoue and Shima146) . Our group has shown that butyrate acts independently of the main GPCR, via its HDAC inhibition property and the SP1 transcription factor present on the human TGFβ1 promoter(Reference Martin-Gallausiaux, Beguet-Crespel and Marinelli28). Moreover, in mice, fibre supplementation promotes vitamin A metabolism in small intestine epithelial cells by increasing RALDH-1. The production of retinoic acid by epithelial cells, the active metabolite of vitamin A, is crucial for the tolerogenic imprinting of dendritic cells (DC)(Reference Goverse, Molenaar and Macia147).

The impact of SCFA goes beyond the epithelial cells, with similar mechanisms reported in macrophages and DC. In mice, macrophage stimulation with butyrate imprints through HDAC3 inhibition, a metabolic reprogramming and elevates antimicrobial peptides. Hence, upon stimulation, antimicrobial peptides belonging to the S100 family, ficolin and lysozyme are increased(Reference Schulthess, Pandey and Capitani148). Here again, butyrate has a stronger antimicrobial effect than propionate and no protective impact is detected with acetate. Butyrate treatment of DC derived from human donors, decreases their capacity to present antigens and increases IL-10 production leading to a tolerogenic phenotype(Reference Liu, Li and Min149). Upon lipopolysaccharide treatment, butyrate induces the IL-23 production by DC thus promoting the differentiation of naive T lymphocytes into pro-inflammatory Th17(Reference Berndt, Zhang and Owyang150). Another study showed that DC treated with butyrate induce the differentiation of naive T lymphocytes into anti-inflammatory Tr1 producers of IL-10(Reference Kaisar, Pelgrom and van der Ham151). By regulating the transcriptional activity, butyrate decreases the inflammatory response of macrophages exposed to inflammatory microbial molecules such as lipopolysaccharide and induces their polarisation through a M2 anti-inflammatory phenotype(Reference Chang, Hao and Offermanns152,Reference Ji, Shu and Zheng153) . Similarly, butyrate-dependent activation of GPR109a increases the tolerogenic response of colonic macrophages and DC reducing colonic inflammation and promoting homoeostasis(Reference Singh, Gurav and Sivaprakasam154). Furthermore, it has been shown that butyrate pre-treatment down-regulates nitric oxide, IL-6 and IL-12 in mice independently of Toll-like receptor and GPCR pathways. Neutrophil migration is increased upon treatment with SCFA, in a GPR43-dependent mechanism(Reference Vinolo, Ferguson and Kulkarni155).

Treg are critical for limiting intestinal inflammation and have thus been subject of considerable attention to improve diseases such as inflammatory bowel disease. Many studies showed that Treg depend on microbiota-derived signals for proper development and function(Reference Atarashi, Tanoue and Oshima145,Reference Atarashi, Tanoue and Shima146,Reference Geuking, Cahenzli and Lawson156,Reference Round and Mazmanian157) . Recently, several groups identified SCFA as key metabolites for promoting differentiation of naive T lymphocytes into Treg cells in the intestine(Reference Arpaia, Campbell and Fan71,Reference Atarashi, Tanoue and Oshima145,Reference Atarashi, Tanoue and Shima146,Reference Chang, Hao and Offermanns152,Reference Singh, Gurav and Sivaprakasam154,Reference Furusawa, Obata and Fukuda158,Reference Smith, Howitt and Panikov159) . By interacting directly with naive T cells, butyrate and propionate increase the acetylation of the promoter of the transcription factor Foxp3 essential for the differentiation of Treg, leading to an increase of Foxp3 expression(Reference Arpaia, Campbell and Fan71,Reference Chang, Hao and Offermanns152,Reference Furusawa, Obata and Fukuda158) . Another group suggested that propionate might induce the same changes via GPR43(Reference Arpaia, Campbell and Fan71,Reference Smith, Howitt and Panikov159) . Moreover, butyrate-dependent activation of GPR109a increases the tolerogenic response of colonic macrophages and DC, promoting Treg and IL-10-producing T cells(Reference Singh, Gurav and Sivaprakasam154). Interestingly, SCFA increase the TGFβ1 production by IEC via its HDAC inhibition property thus promoting the Treg differentiation in the gut(Reference Martin-Gallausiaux, Beguet-Crespel and Marinelli28,Reference Atarashi, Tanoue and Oshima145,Reference Atarashi, Tanoue and Shima146) . Altogether, these studies highlight that the molecular mechanisms induced by SCFA to control Treg-development are complex and involve many cell types involved in the tolerogenic environment such as myeloid cells and IEC.

The impact of SCFA on other lymphocyte populations such as B cells has not been as extensively studied than their Treg counterparts. Acetate supplementation in mice increases intestinal IgA in a GPR43 dependent mechanism(Reference Wu, Sun and Chen160). Dietary fibres and SCFA enhance antibody response to bacteria by supporting B cell differentiation into plasma B cells via the increase of histone acetylation and of B cell metabolism(Reference Kim, Qie and Park161,Reference Sanchez, Moroney and Gan162) . Mechanistically, it is through the downregulation of B cell AID and Blimp1, dependent on their HDAC inhibitory activity that SCFA inhibited class-switch DNA recombination, somatic hypermutation and plasma cell differentiation. Interestingly, SCFA also modulate the fate of B-cell-producing autoantibodies and reduce autoimmunity in lupus-prone mice(Reference Sanchez, Moroney and Gan162).

Conclusion

The past decade of biological research through a combination of translation-focused animal models and studies in human subjects has highlighted the overarching roles that the gut microbiota plays in human health. It has become clear that dysbiotic microbiota is associated with a wide range of pathologies such as obesity, diabetes, CVD, autoimmune diseases and neuronal disorders. Despite the lack of evidence in human subjects, causality has been demonstrated in rodent models. Factors such as antibiotics use, modern sanitation, quality of diet and environmental factors linked with the lifestyle changes that occurred in the past century in developed societies are suggested to contribute to a decrease in the diversity of the human microbiome(Reference Moskowitz and Devkota163).

Diet and nutritional status are important determinants in human health. Numerous studies have shown that diet modulates the composition and functions of the microbiota in human subjects and animal models(Reference David, Maurice and Carmody164Reference De Filippo, Cavalieri and Di Paola166). These interventional studies showed that microbiota composition is dynamic, can shift rapidly to dietary changes and that this shift is individual dependent and depends on the microbiota diversity of the donor. Thus, the role of diet in shaping microbiota is changing our view of the strategies to take to improve the systemic health. Indeed, it is thought that nutritional interventions could manipulate the microbial ecology and consequently modulate human physiology with beneficial health outcomes. However, what constitutes an optimal health-promoting microbiota and how individuals with distinct microbiota can achieve such a level of diversity are still open questions.

As discussed in this review, the gut microbial metabolites SCFA are well known to exert a wide beneficial impact to the host(Reference Canfora, Jocken and Blaak167,Reference Sanna, van Zuydam and Mahajan168) . Hence, fibre-induced increase of SCFA-producing bacteria has been proposed to play an important role in the prevention and treatment of many diseases. Supporting this idea, clinical studies reported that prebiotics and dietary fibres increased the relative abundance of these beneficial SCFA-producing bacteria and butyrate fermentation, leading to the improvement of type-2 diabetes and ulcerative colitis(Reference Haller169,Reference Zhao, Zhang and Ding170) . However, the microbiota produces a vast number of metabolites that modulate host responses, sometimes in synergy with SCFA(Reference Larraufie, Dore and Lapaque121). Many studies support the benefits of increasing both the amount and the variety of dietary fibres ingested but it is difficult to establish whether it is a direct role of SCFA or the increased bacterial diversity that impact host homoeostasis. As the microbiota is a complex ecosystem, much work remains to be done to investigate fully the functions of SCFA alone or with other beneficial metabolites in physiology and pathophysiology.

Acknowledgements

The authors would like to thank all the members of the ‘functionality of the intestinal ecosystem’ team for helpful discussions.

Financial Support

This work was supported by the by Institut national de recherche pour l'agriculture, l'alimentation et l'environnement and by grants funded by EU-FP7 METACARDIS (HEALTH-F4-2012-305312), by the ANR FunMetagen (ANR-11-BSV6-0013).

Conflict of Interest

None.

Authorship

The authors had joint responsibility for all aspects of preparation of this paper.

Footnotes

These authors contributed equally to this work.

References

Reid, G, Younes, JA, Van der Mei, HC et al. (2011) Microbiota restoration: natural and supplemented recovery of human microbial communities. Nat Rev Microbiol 9, 2738.CrossRefGoogle ScholarPubMed
Lepage, P, Leclerc, MC, Joossens, M et al. (2013) A metagenomic insight into our gut's microbiome. Gut 62, 146158.CrossRefGoogle ScholarPubMed
Gomez de Aguero, M, Ganal-Vonarburg, SC, Fuhrer, T et al. (2016) The maternal microbiota drives early postnatal innate immune development. Science 351, 12961302.CrossRefGoogle ScholarPubMed
Rakoff-Nahoum, S, Kong, Y, Kleinstein, SH et al. (2015) Analysis of gene–environment interactions in postnatal development of the mammalian intestine. Proc Natl Acad Sci USA 112, 19291936.CrossRefGoogle ScholarPubMed
van de Guchte, M, Blottiere, HM, Dore, J (2018) Humans as holobionts: implications for prevention and therapy. Microbiome 6, 81.CrossRefGoogle ScholarPubMed
Atarashi, K & Honda, K (2011) Microbiota in autoimmunity and tolerance. Curr Opin Immunol 23, 761768.CrossRefGoogle ScholarPubMed
Miyake, S, Kim, S, Suda, W et al. (2015) Dysbiosis in the gut microbiota of patients with multiple sclerosis, with a striking depletion of species belonging to Clostridia XIVa and IV clusters. PLoS ONE 10, e0137429.CrossRefGoogle ScholarPubMed
Benakis, C, Brea, D, Caballero, S et al. (2016) Commensal microbiota affects ischemic stroke outcome by regulating intestinal gammadelta T cells. Nat Med, 22, 516–23.CrossRefGoogle ScholarPubMed
Qin, J, Li, R, Raes, J et al. (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 5965.CrossRefGoogle ScholarPubMed
Qin, N, Yang, F, Li, A et al. (2014) Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 5964.CrossRefGoogle ScholarPubMed
Le Chatelier, E, Nielsen, T, Qin, J et al. (2013) Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541546.CrossRefGoogle ScholarPubMed
Karlsson, FH, Fak, F, Nookaew, I et al. (2012) Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun 3, 1245.CrossRefGoogle ScholarPubMed
Pozuelo, M, Panda, S, Santiago, A et al. (2015) Reduction of butyrate- and methane-producing microorganisms in patients with irritable bowel syndrome. Sci Rep 5, 12693.CrossRefGoogle ScholarPubMed
Vatanen, T, Franzosa, EA, Schwager, R et al. (2018) The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature 562, 589594.CrossRefGoogle ScholarPubMed
Garron, ML & Henrissat, B (2019) The continuing expansion of CAZymes and their families. Curr Opin Chem Biol 53, 8287.CrossRefGoogle ScholarPubMed
Duncan, SH, Holtrop, G, Lobley, GE et al. (2004) Contribution of acetate to butyrate formation by human faecal bacteria. Br J Nutr 91, 915923.CrossRefGoogle ScholarPubMed
Pryde, SE, Duncan, SH, Hold, GL et al. (2002) The microbiology of butyrate formation in the human colon. FEMS Microbiol Lett 217, 133139.CrossRefGoogle ScholarPubMed
Cummings, JH (1981) Short chain fatty acids in the human colon. Gut 22, 763779.CrossRefGoogle ScholarPubMed
Macfarlane, GT & Macfarlane, S (1997) Human colonic microbiota: ecology, physiology and metabolic potential of intestinal bacteria. Scand J Gastroenterol Suppl. 222, 39.CrossRefGoogle ScholarPubMed
Dalile, B, Van Oudenhove, L, Vervliet, B et al. (2019) The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol 16, 461478.CrossRefGoogle ScholarPubMed
Bourassa, MW, Alim, I, Bultman, SJ et al. (2016) Butyrate, neuroepigenetics and the gut microbiome: can a high fiber diet improve brain health? Neurosci Lett 625, 5663.CrossRefGoogle ScholarPubMed
Smith, EA & Macfarlane, GT (1997) Dissimilatory amino acid metabolism in human colonic bacteria. Anaerobe 3, 327337.CrossRefGoogle ScholarPubMed
Davila, AM, Blachier, F, Gotteland, M et al. (2013) Re-print of ‘Intestinal luminal nitrogen metabolism: role of the gut microbiota and consequences for the host’. Pharmacol Res 69, 114126.CrossRefGoogle Scholar
Flint, HJ (2016) Gut microbial metabolites in health and disease. Gut Microbes 7, 187188.CrossRefGoogle ScholarPubMed
Louis, P, Hold, GL & Flint, HJ (2014) The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol 12, 661672.CrossRefGoogle ScholarPubMed
Louis, P & Flint, HJ (2009) Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol Lett 294, 18.CrossRefGoogle ScholarPubMed
Martin-Gallausiaux, C, Larraufie, P, Jarry, A et al. (2018) Butyrate produced by commensal bacteria down-regulates indolamine 2,3-dioxygenase 1 (IDO-1) expression via a dual mechanism in human intestinal epithelial cells. Front Immunol 9, 2838.CrossRefGoogle Scholar
Martin-Gallausiaux, C, Beguet-Crespel, F, Marinelli, L et al. (2018) Butyrate produced by gut commensal bacteria activates TGF-beta1 expression through the transcription factor SP1 in human intestinal epithelial cells. Sci Rep 8, 9742.CrossRefGoogle ScholarPubMed
Belzer, C, Chia, LW, Aalvink, S et al. (2017) Microbial metabolic networks at the mucus layer lead to diet-independent butyrate and vitamin B12 production by intestinal symbionts. mBio 8, e0077017.CrossRefGoogle ScholarPubMed
Reichardt, N, Duncan, SH, Young, P et al. (2014) Phylogenetic distribution of three pathways for propionate production within the human gut microbiota. ISME J 8, 13231335.CrossRefGoogle ScholarPubMed
Duncan, SH, Louis, P, Thomson, JM et al. (2009) The role of pH in determining the species composition of the human colonic microbiota. Environ Microbiol 11, 21122122.CrossRefGoogle ScholarPubMed
Hamer, HM, Jonkers, D, Venema, K et al. (2008) Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther 27, 104119.CrossRefGoogle ScholarPubMed
Dostal, A, Lacroix, C, Bircher, L et al. (2015) Iron modulates butyrate production by a child gut microbiota in vitro. mBio 6, e01453–e01415.CrossRefGoogle ScholarPubMed
Dostal, A, Lacroix, C, Pham, VT et al. (2014) Iron supplementation promotes gut microbiota metabolic activity but not colitis markers in human gut microbiota-associated rats. Br J Nutr 111, 21352145.CrossRefGoogle Scholar
Conn, AR, Fell, DI & Steele, RD (1983) Characterization of alpha-keto acid transport across blood–brain barrier in rats. Am J Physiol 245, E253E260.Google ScholarPubMed
Frost, G, Sleeth, ML, Sahuri-Arisoylu, M et al. (2014) The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun 5, 3611.CrossRefGoogle Scholar
Cummings, JH, Pomare, EW, Branch, WJ et al. (1987) Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28, 12211227.CrossRefGoogle ScholarPubMed
Bergman, EN (1990) Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol Rev 70, 567590.CrossRefGoogle ScholarPubMed
Sivaprakasam, S, Bhutia, YD, Yang, S et al. (2017) Short-chain fatty acid transporters: role in colonic homeostasis. Compr Physiol 8, 299314.CrossRefGoogle ScholarPubMed
Sepponen, K, Ruusunen, M, Pakkanen, JA et al. (2007) Expression of CD147 and monocarboxylate transporters MCT1, MCT2 and MCT4 in porcine small intestine and colon. Vet J 174, 122128.CrossRefGoogle ScholarPubMed
Bloemen, JG, Venema, K, van de Poll, MC et al. (2009) Short chain fatty acids exchange across the gut and liver in humans measured at surgery. Clin Nutr 28, 657661.CrossRefGoogle Scholar
van der Beek, CM, Bloemen, JG, van den Broek, MA, et al. (2015) Hepatic uptake of rectally administered butyrate prevents an increase in systemic butyrate concentrations in humans. J Nutr 145, 20192024.CrossRefGoogle ScholarPubMed
McNeil, NI, Cummings, JH James, WP (1979) Rectal absorption of short chain fatty acids in the absence of chloride. Gut 20, 400403.CrossRefGoogle ScholarPubMed
Binder, HJ & Mehta, P (1989) Short-chain fatty acids stimulate active sodium and chloride absorption in vitro in the rat distal colon. Gastroenterology 96, 989996.CrossRefGoogle ScholarPubMed
Lutz, T & Scharrer, E (1991) Effect of short-chain fatty acids on calcium absorption by the rat colon. Exp Physiol 76, 615618.CrossRefGoogle ScholarPubMed
Srinivas, SR, Gopal, E, Zhuang, L et al. (2005) Cloning and functional identification of slc5a12 as a sodium-coupled low-affinity transporter for monocarboxylates (SMCT2). Biochem J 392, 655664.CrossRefGoogle Scholar
Coady, MJ, Chang, MH, Charron, FM et al. (2004) The human tumour suppressor gene SLC5A8 expresses a Na+-monocarboxylate cotransporter. J Physiol 557, 719731.CrossRefGoogle ScholarPubMed
Gupta, N, Martin, PM, Prasad, PD et al. (2006) SLC5A8 (SMCT1)-mediated transport of butyrate forms the basis for the tumor suppressive function of the transporter. Life Sci 78, 24192425.CrossRefGoogle ScholarPubMed
Matthews, JB, Hassan, I, Meng, S et al. (1998) Na-K-2Cl cotransporter gene expression and function during enterocyte differentiation. Modulation of Cl secretory capacity by butyrate. J Clin Invest 101, 20722079.CrossRefGoogle ScholarPubMed
Musch, MW, Bookstein, C, Xie, Y et al. (2001) SCFA Increase intestinal Na absorption by induction of NHE3 in rat colon and human intestinal C2/bbe cells. Am J Physiol: Gastrointest Liver Physiol 280, G687G693.Google ScholarPubMed
Amin, MR, Dudeja, PK, Ramaswamy, K et al. (2007) Involvement of Sp1 and Sp3 in differential regulation of human NHE3 promoter activity by sodium butyrate and IFN-gamma/TNF-alpha. Am J Physiol: Gastrointest Liver Physiol 293, G374G382.Google ScholarPubMed
Subramanya, SB, Rajendran, VM, Srinivasan, P et al. (2007) Differential regulation of cholera toxin-inhibited Na-H exchange isoforms by butyrate in rat ileum. Am J Physiol: Gastrointest Liver Physiol 293, G857G863.Google ScholarPubMed
Gelis, L, Jovancevic, N, Veitinger, S et al. (2016) Functional characterization of the odorant receptor 51E2 in human melanocytes. J Biol Chem 291, 1777217786.CrossRefGoogle ScholarPubMed
Puhl, HL III, Won, YJ, Lu, VB et al. (2015) Human GPR42 is a transcribed multisite variant that exhibits copy number polymorphism and is functional when heterologously expressed. Sci Rep 5, 12880.CrossRefGoogle ScholarPubMed
Le Poul, E, Loison, C, Struyf, S et al. (2003) Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 278, 2548125489.CrossRefGoogle ScholarPubMed
Brown, AJ, Goldsworthy, SM, Barnes, AA et al. (2003) The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 278, 1131211319.CrossRefGoogle ScholarPubMed
Thangaraju, M, Cresci, GA, Liu, K et al. (2009) GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res 69, 28262832.CrossRefGoogle Scholar
Hudson, BD, Tikhonova, IG, Pandey, SK et al. (2012) Extracellular ionic locks determine variation in constitutive activity and ligand potency between species orthologs of the free fatty acid receptors FFA2 and FFA3. J Biol Chem 287, 4119541209.CrossRefGoogle ScholarPubMed
Dorsam, RT & Gutkind, JS (2007) G-protein-coupled receptors and cancer. Nat Rev Cancer 7, 7994.CrossRefGoogle ScholarPubMed
Priori, D, Colombo, M, Clavenzani, P et al. (2015) The olfactory receptor OR51E1 is present along the gastrointestinal tract of pigs, co-localizes with enteroendocrine cells and is modulated by intestinal microbiota. PLoS ONE 10, e0129501.CrossRefGoogle ScholarPubMed
Han, YE, Kang, CW, Oh, JH et al. (2018) Olfactory receptor OR51E1 mediates GLP-1 secretion in human and rodent enteroendocrine L cells. J Endocr Soc 2, 12511258.CrossRefGoogle ScholarPubMed
Fleischer, J, Bumbalo, R, Bautze, V et al. (2015) Expression of odorant receptor Olfr78 in enteroendocrine cells of the colon. Cell Tissue Res 361, 697710.CrossRefGoogle ScholarPubMed
Pluznick, JL (2013) Renal and cardiovascular sensory receptors and blood pressure regulation. Am J Physiol Renal Physiol 305, F439F444.CrossRefGoogle ScholarPubMed
Basson, MD, Liu, YW, Hanly, AM et al. (2000) Identification and comparative analysis of human colonocyte short-chain fatty acid response genes. J Gastrointest Surg 4, 501512.CrossRefGoogle ScholarPubMed
Rada-Iglesias, A, Enroth, S, Ameur, A et al. (2007) Butyrate mediates decrease of histone acetylation centered on transcription start sites and down-regulation of associated genes. Genome Res 17, 708719.CrossRefGoogle ScholarPubMed
Donohoe, DR, Collins, LB, Wali, A et al. (2012) The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell 48, 612626.CrossRefGoogle ScholarPubMed
Candido, EP, Reeves, R & Davie, JR (1978) Sodium butyrate inhibits histone deacetylation in cultured cells. Cell 14, 105113.CrossRefGoogle ScholarPubMed
Sealy, L & Chalkley, R (1978) The effect of sodium butyrate on histone modification. Cell 14, 115121.CrossRefGoogle ScholarPubMed
Donohoe, DR & Bultman, SJ (2012) Metaboloepigenetics: interrelationships between energy metabolism and epigenetic control of gene expression. J Cell Physiol 227, 31693177.CrossRefGoogle ScholarPubMed
Bose, P, Dai, Y & Grant, S (2014) Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights. Pharmacol Ther 143, 323336.CrossRefGoogle ScholarPubMed
Arpaia, N, Campbell, C, Fan, X et al. (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451455.CrossRefGoogle ScholarPubMed
Thakur, BK, Dasgupta, N, Ta, A et al. (2016) Physiological TLR5 expression in the intestine is regulated by differential DNA binding of Sp1/Sp3 through simultaneous Sp1 dephosphorylation and Sp3 phosphorylation by two different PKC isoforms. Nucleic Acids Res 44, 56585672.CrossRefGoogle ScholarPubMed
Fellows, R, Denizot, J, Stellato, C et al. (2018) Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases. Nat Commun 9, 105.CrossRefGoogle ScholarPubMed
Boffa, LC, Gruss, RJ & Allfrey, VG (1981) Manifold effects of sodium butyrate on nuclear function. Selective and reversible inhibition of phosphorylation of histones H1 and H2A and impaired methylation of lysine and arginine residues in nuclear protein fractions. J Biol Chem 256, 96129621.CrossRefGoogle ScholarPubMed
Mathew, OP, Ranganna, K & Yatsu, FM (2010) Butyrate, an HDAC inhibitor, stimulates interplay between different posttranslational modifications of histone H3 and differently alters G1-specific cell cycle proteins in vascular smooth muscle cells. Biomed Pharmacother 64, 733740.CrossRefGoogle ScholarPubMed
Parker, MI, de Haan, JB & Gevers, W (1986) DNA hypermethylation in sodium butyrate-treated WI-38 fibroblasts. J Biol Chem 261, 27862790.CrossRefGoogle ScholarPubMed
Alex, S, Lange, K, Amolo, T et al. (2013) Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating peroxisome proliferator-activated receptor gamma. Mol Cell Biol 33, 13031316.CrossRefGoogle ScholarPubMed
Marinelli, L, Martin-Gallausiaux, C, Bourhis, J et al. (2019) Identification of the novel role of butyrate as AhR ligand in human intestinal epithelial cells. Sci Rep 9, 643.CrossRefGoogle ScholarPubMed
Fleming, SE, Choi, SY & Fitch, MD (1991) Absorption of short-chain fatty acids from the rat cecum in vivo. J Nutr 121, 17871797.CrossRefGoogle ScholarPubMed
Ardawi, MS & Newsholme, EA (1985) Fuel utilization in colonocytes of the rat. Biochem J 231, 713719.CrossRefGoogle ScholarPubMed
Roediger, WE (1980) Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 21, 793798.CrossRefGoogle ScholarPubMed
Furuta, GT, Turner, JR, Taylor, CT et al. (2001) Hypoxia-inducible factor 1-dependent induction of intestinal trefoil factor protects barrier function during hypoxia. J Exp Med 193, 10271034.CrossRefGoogle ScholarPubMed
Andriamihaja, M, Chaumontet, C, Tome, D et al. (2009) Butyrate metabolism in human colon carcinoma cells: implications concerning its growth-inhibitory effect. J Cell Physiol 218, 5865.CrossRefGoogle ScholarPubMed
Blouin, JM, Penot, G, Collinet, M et al. (2011) Butyrate elicits a metabolic switch in human colon cancer cells by targeting the pyruvate dehydrogenase complex. Int J Cancer 128, 25912601.CrossRefGoogle ScholarPubMed
Donohoe, DR, Garge, N, Zhang, X et al. (2011) The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab 13, 517526.CrossRefGoogle ScholarPubMed
Zambell, KL, Fitch, MD & Fleming, SE (2003) Acetate and butyrate are the major substrates for de novo lipogenesis in rat colonic epithelial cells. J Nutr 133, 35093515.CrossRefGoogle Scholar
Park, JH, Kotani, T, Konno, T et al. (2016) Promotion of intestinal epithelial cell turnover by commensal bacteria: role of short-chain fatty acids. PLoS ONE 11, e0156334.CrossRefGoogle ScholarPubMed
Augeron, C & Laboisse, CL (1984) Emergence of permanently differentiated cell clones in a human colonic cancer cell line in culture after treatment with sodium butyrate. Cancer Res 44, 39613969.Google Scholar
Barnard, JA & Warwick, G (1993) Butyrate rapidly induces growth inhibition and differentiation in HT-29 cells. Cell Growth Differ 4, 495501.Google ScholarPubMed
Kaiko, GE, Ryu, SH, Koues, OI et al. (2016) The colonic crypt protects stem cells from microbiota-derived metabolites. Cell 167, 1137.CrossRefGoogle ScholarPubMed
Csordas, A (1996) Butyrate, aspirin and colorectal cancer. Eur J Cancer Prev 5, 221231.CrossRefGoogle ScholarPubMed
Sengupta, S, Muir, JG & Gibson, PR (2006) Does butyrate protect from colorectal cancer? J Gastroenterol Hepatol 21, 209218.CrossRefGoogle ScholarPubMed
Arun, KB, Madhavan, A, Reshmitha, TR, et al. (2019) Short chain fatty acids enriched fermentation metabolites of soluble dietary fibre from Musa paradisiaca drives HT29 colon cancer cells to apoptosis. PLoS ONE 14, e0216604.Google Scholar
Matthews, GM, Howarth, GS & Butler, RN (2012) Short-chain fatty acids induce apoptosis in colon cancer cells associated with changes to intracellular redox state and glucose metabolism. Chemotherapy 58, 102109.CrossRefGoogle ScholarPubMed
Okabe, S, Okamoto, T, Zhao, CM et al. (2014) Acetic acid induces cell death: an in vitro study using normal rat gastric mucosal cell line and rat and human gastric cancer and mesothelioma cell lines. J Gastroenterol Hepatol 29, Suppl. 4, 6569.CrossRefGoogle Scholar
Verma, SP, Agarwal, A & Das, P (2018) Sodium butyrate induces cell death by autophagy and reactivates a tumor suppressor gene DIRAS1 in renal cell carcinoma cell line UOK146. In Vitro Cell Dev Biol Anim 54, 295303.CrossRefGoogle ScholarPubMed
Kim, K, Kwon, O, Ryu, TY et al. (2019) Propionate of a microbiota metabolite induces cell apoptosis and cell cycle arrest in lung cancer. Mol Med Rep 20, 15691574.Google ScholarPubMed
Wang, TT, Chiang, AS, Chu, JJ et al. (1998) Concomitant alterations in distribution of 70 kDa heat shock proteins, cytoskeleton and organelles in heat shocked 9L cells. Int J Biochem Cell Biol 30, 745759.CrossRefGoogle ScholarPubMed
Bindels, LB, Porporato, P, Dewulf, EM et al. (2012) Gut microbiota-derived propionate reduces cancer cell proliferation in the liver. Br J Cancer 107, 13371344.CrossRefGoogle ScholarPubMed
Casanova, MR, Azevedo-Silva, J, Rodrigues, LR et al. (2018) Colorectal cancer cells increase the production of short chain fatty acids by Propionibacterium freudenreichii impacting on cancer cells survival. Front Nutr 5, 44.CrossRefGoogle ScholarPubMed
Gribble, FM & Reimann, F (2019) Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat Rev Endocrinol 15, 226237.CrossRefGoogle ScholarPubMed
Cani, PD, Amar, J, Iglesias, MA et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 17611772.CrossRefGoogle ScholarPubMed
Samuel, BS, Shaito, A, Motoike, T et al. (2008) Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc Natl Acad Sci USA 105, 1676716772.CrossRefGoogle ScholarPubMed
Karaki, S, Tazoe, H, Hayashi, H et al. (2008) Expression of the short-chain fatty acid receptor, GPR43, in the human colon. J Mol Histol 39, 135142.CrossRefGoogle ScholarPubMed
Lu, VB, Gribble, FM & Reimann, F (2018) Free fatty acid receptors in enteroendocrine cells. Endocrinology 159, 28262835.CrossRefGoogle ScholarPubMed
Nohr, MK, Pedersen, MH, Gille, A et al. (2013) GPR41/FFAR3 And GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology 154, 35523564.CrossRefGoogle ScholarPubMed
Tolhurst, G, Heffron, H, Lam, YS et al. (2012) Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 61, 364371.CrossRefGoogle ScholarPubMed
Bolognini, D, Tobin, AB, Milligan, G et al. (2016) The pharmacology and function of receptors for short-chain fatty acids. Mol Pharmacol 89, 388398.CrossRefGoogle ScholarPubMed
Hudson, BD, Due-Hansen, ME, Christiansen, E et al. (2013) Defining the molecular basis for the first potent and selective orthosteric agonists of the FFA2 free fatty acid receptor. J Biol Chem 288, 1729617312.CrossRefGoogle ScholarPubMed
Psichas, A, Sleeth, ML, Murphy, KG et al. (2015) The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes 39, 424429.CrossRefGoogle ScholarPubMed
Lee, EY, Zhang, X, Miyamoto, J et al. (2018) Gut carbohydrate inhibits GIP secretion via a microbiota/SCFA/FFAR3 pathway. J Endocrinol 239, 267276.CrossRefGoogle Scholar
Ang, Z, Xiong, D, Wu, M et al. (2018) FFAR2-FFAR3 Receptor heteromerization modulates short-chain fatty acid sensing. FASEB J 32, 289303.CrossRefGoogle ScholarPubMed
Chambers, ES, Morrison, DJ & Frost, G (2015) Control of appetite and energy intake by SCFA: what are the potential underlying mechanisms? Proc Nutr Soc 74, 328336.CrossRefGoogle ScholarPubMed
Tazoe, H, Otomo, Y, Karaki, S et al. (2009) Expression of short-chain fatty acid receptor GPR41 in the human colon. Biomed Res 30, 149156.CrossRefGoogle ScholarPubMed
Roberts, GP, Larraufie, P, Richards, P et al. (2019) Comparison of human and murine enteroendocrine cells by transcriptomic and peptidomic profiling. Diabetes 68, 10621072.CrossRefGoogle ScholarPubMed
Larraufie, P, Martin-Gallausiaux, C, Lapaque, N et al. (2018) SCFA strongly stimulate PYY production in human enteroendocrine cells. Sci Rep 8, 74.CrossRefGoogle ScholarPubMed
Brooks, L, Viardot, A, Tsakmaki, A et al. (2017) Fermentable carbohydrate stimulates FFAR2-dependent colonic PYY cell expansion to increase satiety. Mol Metab 6, 4860.CrossRefGoogle ScholarPubMed
Petersen, N, Reimann, F, Bartfeld, S et al. (2014) Generation of L cells in mouse and human small intestine organoids. Diabetes 63, 410420.CrossRefGoogle ScholarPubMed
Reigstad, CS, Salmonson, CE, Rainey, JF III et al. (2015) Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J 29, 13951403.CrossRefGoogle ScholarPubMed
Zhou, J, Martin, RJ, Tulley, RT et al. (2008) Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents. Am J Physiol Endocrinol Metab 295, E1160E1166.CrossRefGoogle Scholar
Larraufie, P, Dore, J, Lapaque, N et al. (2017) TLR ligands and butyrate increase PYY expression through two distinct but inter-regulated pathways. Cell Microbiol 19, e12648.CrossRefGoogle ScholarPubMed
Cox, HM (2007) Neuropeptide Y receptors; antisecretory control of intestinal epithelial function. Auton Neurosci 133, 7685.CrossRefGoogle ScholarPubMed
Pais, R, Rievaj, J, Larraufie, P et al. (2016) Angiotensin II type 1 receptor-dependent GLP-1 and PYY secretion in mice and humans. Endocrinology 157, 38213831.CrossRefGoogle ScholarPubMed
Okuno, M, Nakanishi, T, Shinomura, Y et al. (1992) Peptide YY enhances NaCl and water absorption in the rat colon in vivo. Experientia 48, 4750.CrossRefGoogle ScholarPubMed
Fukumoto, S, Tatewaki, M, Yamada, T et al. (2003) Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats. Am J Physiol Regul Integr Comp Physiol 284, R1269R1276.CrossRefGoogle ScholarPubMed
Cherbut, C, Ferrier, L, Roze, C et al. (1998) Short-chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. Am J Physiol 275, G1415G1422.Google ScholarPubMed
Cuche, G, Cuber, JC & Malbert, CH (2000) Ileal short-chain fatty acids inhibit gastric motility by a humoral pathway. Am J Physiol: Gastrointest Liver Physiol 279, G925G930.Google ScholarPubMed
Vincent, AD, Wang, XY, Parsons, SP et al. (2018) Abnormal absorptive colonic motor activity in germ-free mice is rectified by butyrate, an effect possibly mediated by mucosal serotonin. Am J Physiol: Gastrointest Liver Physiol 315, G896G907.Google ScholarPubMed
Soret, R, Chevalier, J, De Coppet, P et al. (2010) Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology 138, 17721782.CrossRefGoogle ScholarPubMed
Kelly, CJ, Zheng, L, Campbell, EL et al. (2015) Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 17, 662671.CrossRefGoogle ScholarPubMed
Faber, F, Tran, L, Byndloss, MX et al. (2016) Host-mediated sugar oxidation promotes post-antibiotic pathogen expansion. Nature 534, 697699.CrossRefGoogle ScholarPubMed
Litvak, Y, Byndloss, MX, Tsolis, RM et al. (2017) Dysbiotic proteobacteria expansion: a microbial signature of epithelial dysfunction. Curr Opin Microbiol 39, 16.CrossRefGoogle ScholarPubMed
Rivera-Chavez, F, Zhang, LF, Faber, F et al. (2016) Depletion of butyrate-producing Clostridia from the gut microbiota drives an aerobic luminal expansion of Salmonella. Cell Host Microbe 19, 443454.CrossRefGoogle ScholarPubMed
Bronner, DN, Faber, F, Olsan, EE et al. (2018) Genetic ablation of butyrate utilization attenuates gastrointestinal Salmonella disease. Cell Host Microbe 23, 266273 e264.CrossRefGoogle ScholarPubMed
Zheng, L, Kelly, CJ, Battista, KD et al. (2017) Microbial-derived butyrate promotes epithelial barrier function through IL-10 receptor-dependent repression of claudin-2. J Immunol 199, 29762984.CrossRefGoogle ScholarPubMed
Wang, HB, Wang, PY, Wang, X et al. (2012) Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein claudin-1 transcription. Dig Dis Sci 57, 31263135.CrossRefGoogle ScholarPubMed
Gaudier, E, Jarry, A, Blottiere, HM et al. (2004) Butyrate specifically modulates MUC gene expression in intestinal epithelial goblet cells deprived of glucose. Am J Physiol: Gastrointest Liver Physiol 287, G1168G1174.Google ScholarPubMed
Hase, K, Eckmann, L, Leopard, JD et al. (2002) Cell differentiation is a key determinant of cathelicidin LL-37/human cationic antimicrobial protein 18 expression by human colon epithelium. Infect Immun 70, 953963.CrossRefGoogle ScholarPubMed
Raqib, R, Sarker, P, Bergman, P et al. (2006) Improved outcome in shigellosis associated with butyrate induction of an endogenous peptide antibiotic. Proc Natl Acad Sci USA 103, 91789183.CrossRefGoogle ScholarPubMed
Zhao, Y, Chen, F, Wu, W et al. (2018) GPR43 Mediates microbiota metabolite SCFA regulation of antimicrobial peptide expression in intestinal epithelial cells via activation of mTOR and STAT3. Mucosal Immunol 11, 752762.CrossRefGoogle ScholarPubMed
Fischer, N, Sechet, E, Friedman, R et al. (2016) Histone deacetylase inhibition enhances antimicrobial peptide but not inflammatory cytokine expression upon bacterial challenge. Proc Natl Acad Sci USA 113, E2993E3001.CrossRefGoogle Scholar
Maslowski, KM, Vieira, AT, Ng, A et al. (2009) Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 12821286.CrossRefGoogle ScholarPubMed
Macia, L, Tan, J, Vieira, AT et al. (2015) Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun 6, 6734.CrossRefGoogle ScholarPubMed
Huang, N, Katz, JP, Martin, DR et al. (1997) Inhibition of IL-8 gene expression in Caco-2 cells by compounds which induce histone hyperacetylation. Cytokine 9, 2736.CrossRefGoogle ScholarPubMed
Atarashi, K, Tanoue, T, Oshima, K et al. (2013) Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500, 232236.CrossRefGoogle ScholarPubMed
Atarashi, K, Tanoue, T, Shima, T et al. (2011) Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337341.CrossRefGoogle ScholarPubMed
Goverse, G, Molenaar, R, Macia, L et al. (2017) Diet-derived short chain fatty acids stimulate intestinal epithelial cells To induce mucosal tolerogenic dendritic cells. J Immunol 198, 21722181.CrossRefGoogle ScholarPubMed
Schulthess, J, Pandey, S, Capitani, M et al. (2019) The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 50, 432445. e437.CrossRefGoogle ScholarPubMed
Liu, L, Li, L, Min, J et al. (2012) Butyrate interferes with the differentiation and function of human monocyte-derived dendritic cells. Cell Immunol 277, 6673.CrossRefGoogle ScholarPubMed
Berndt, BE, Zhang, M, Owyang, SY et al. (2012) Butyrate increases IL-23 production by stimulated dendritic cells. Am J Physiol: Gastrointest Liver Physiol 303, G1384G1392.Google ScholarPubMed
Kaisar, MMM, Pelgrom, LR, van der Ham, AJ et al. (2017) Butyrate conditions human dendritic cells to prime type 1 regulatory T cells via both histone deacetylase inhibition and G protein-coupled receptor 109A signaling. Front Immunol 8, 1429.CrossRefGoogle Scholar
Chang, PV, Hao, L, Offermanns, S et al. (2014) The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci USA 111, 22472252.CrossRefGoogle ScholarPubMed
Ji, J, Shu, D, Zheng, M et al. (2016) Microbial metabolite butyrate facilitates M2 macrophage polarization and function. Sci Rep 6, 24838.CrossRefGoogle ScholarPubMed
Singh, N, Gurav, A, Sivaprakasam, S et al. (2014) Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40, 128139.CrossRefGoogle ScholarPubMed
Vinolo, MA, Ferguson, GJ, Kulkarni, S et al. (2011) SCFA induce mouse neutrophil chemotaxis through the GPR43 receptor. PLoS ONE 6, e21205.CrossRefGoogle Scholar
Geuking, MB, Cahenzli, J, Lawson, MA et al. (2011) Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34, 794806.CrossRefGoogle ScholarPubMed
Round, JL & Mazmanian, SK (2010) Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA 107, 1220412209.CrossRefGoogle ScholarPubMed
Furusawa, Y, Obata, Y, Fukuda, S et al. (2013) Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446450.CrossRefGoogle ScholarPubMed
Smith, PM, Howitt, MR, Panikov, N et al. (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569573.CrossRefGoogle ScholarPubMed
Wu, W, Sun, M, Chen, F et al. (2017) Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol 10, 946956.CrossRefGoogle ScholarPubMed
Kim, M, Qie, Y, Park, J et al. (2016) Gut microbial metabolites fuel host antibody responses. Cell Host Microbe 20, 202214.CrossRefGoogle ScholarPubMed
Sanchez, HN, Moroney, JB, Gan, H et al. (2020) B cell-intrinsic epigenetic modulation of antibody responses by dietary fiber-derived short-chain fatty acids. Nat Commun 11, 60.CrossRefGoogle ScholarPubMed
Moskowitz, JE & Devkota, S (2019) Determinants of microbial antibiotic susceptibility: the commensal gut microbiota perspective. Cell Host Microbe 26, 574576.CrossRefGoogle ScholarPubMed
David, LA, Maurice, CF, Carmody, RN et al. (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559563.CrossRefGoogle ScholarPubMed
Cotillard, A, Kennedy, SP, Kong, LC et al. (2013) Dietary intervention impact on gut microbial gene richness. Nature 500, 585588.CrossRefGoogle ScholarPubMed
De Filippo, C, Cavalieri, D, Di Paola, M et al. (2010) Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA 107, 1469114696.CrossRefGoogle ScholarPubMed
Canfora, EE, Jocken, JW & Blaak, EE (2015) Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 11, 577591.CrossRefGoogle ScholarPubMed
Sanna, S, van Zuydam, NR, Mahajan, A et al. (2019) Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet 51, 600605.CrossRefGoogle ScholarPubMed
Haller, D (2010) Nutrigenomics and IBD: the intestinal microbiota at the cross-road between inflammation and metabolism. J Clin Gastroenterol 44, Suppl. 1, S6S9.CrossRefGoogle ScholarPubMed
Zhao, L, Zhang, F, Ding, X et al. (2018) Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 359, 11511156.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. (a) Functional impact of SCFA on the host. (b) Mechanisms: (1) G protein-coupled receptor (GPCR)-dependent signalling, (2) histone and transcription factor acetylation by SCFA and (3) role of butyrate as a ligand of transcription factors. AhR, aryl hydrocarbon receptor; ARNT, aryl hydrocarbon receptor nuclear translocator; HAT, histone acetyltransferase; K/HDAC, lysine/histone deacetylase; MAPK, mitogen-activated protein kinase; PLC, phospholipase C; TF, transcription factor; XRE, xenobiotic response element.