Skip to main content
EMBO Reports logoLink to EMBO Reports
. 2022 Aug 29;23(10):e55664. doi: 10.15252/embr.202255664

Local and systemic effects of microbiome‐derived metabolites

Igor Spivak 1,2, , Leviel Fluhr 1, , Eran Elinav 1,3,
PMCID: PMC9535759  PMID: 36031866

Abstract

Commensal microbes form distinct ecosystems within their mammalian hosts, collectively termed microbiomes. These indigenous microbial communities broadly expand the genomic and functional repertoire of their host and contribute to the formation of a “meta‐organism.” Importantly, microbiomes exert numerous biochemical reactions synthesizing or modifying multiple bioactive small molecules termed metabolites, which impact their host's physiology in a variety of contexts. Identifying and understanding molecular mechanisms of metabolite–host interactions, and how their disrupted signaling can contribute to diseases, may enable their therapeutic application, a modality termed “postbiotic” therapy. In this review, we highlight key examples of effects of bioactive microbe‐associated metabolites on local, systemic, and immune environments, and discuss how these may impact mammalian physiology and associated disorders. We outline the challenges and perspectives in understanding the potential activity and function of this plethora of microbially associated small molecules as well as possibilities to harness them toward the promotion of personalized precision therapeutic interventions.

Keywords: commensals, immune, metabolites, microbiome, postbiotic

Subject Categories: Metabolism; Microbiology, Virology & Host Pathogen Interaction; Signal Transduction


Microbiomes synthesize or modify multiple bioactive small molecules, which impact their host's physiology. This review highlights key effects of bioactive microbe‐associated metabolites and discusses how they may impact mammalian physiology and associated disorders.

graphic file with name EMBR-23-e55664-g004.jpg

Introduction

Commensal microorganism consortia, collectively termed the microbiome, inhabit multiple mucosal sites of their mammalian host, and significantly contribute to the “holobiont” in cell numbers and genome size (Tierney et al, 2019). The microbiome carries out multiple roles, such as outcompeting pathogens in their habitat, providing signals of homeostatic or perturbed surrounding conditions, and producing, modulating, and degrading a wide range of small soluble bioactive molecules (herein referred to as metabolites, see Table 1), which feature multiple effects on the host. Microbial metabolism often involves the utilization of components not accessible to host metabolic enzymes, such as complex carbohydrates, but can also compete with host metabolic systems for substrates. Microbial products and processes are highly interconnected with the host's own metabolic function in contributing to the host's physiology and homeostasis (Thaiss et al, 2016b), complementing the host in degrading complex molecules (Cani et al, 2019), sensing different conditions in their environment, and in the regulation of commensal communities (Krautkramer et al, 2021). The microbial‐produced metabolite repertoire is dependent on host genetic and ecological diversity (Costello et al, 2012; Goodrich et al, 2014), geographic location (Hehemann et al, 2010), age (Yatsunenko et al, 2012; Sato et al, 2021), diet (Kolodziejczyk et al, 2019; Alexander & Turnbaugh, 2020), and other lifestyle‐related factors (Bajaj, 2019). Certain classes of bacterial metabolites, including short‐chain fatty acids (SCFAs) and bile acids are mostly linked to beneficial effects on their host's health (Koh & Bäckhed, 2020). Different end‐products of microbial tryptophan metabolism can have either a beneficial or harmful impact (Agus et al, 2018; Paeslack et al, 2022). Others, such as some amino acid derivates or trimethylamine‐oxide (TMAO) are largely associated with noxious effects contributing to disease pathogenesis (Agus et al, 2021). Insights into the mechanism of action and modulation of these microbial products may enable their integration as therapeutics (Chaluvadi et al, 2016), a modality recently termed postbiotic treatment (Aguilar‐Toalá et al, 2018). A recent consensus statement by the International Scientific Association of Probiotics and Prebiotics (ISAPP) proposed to integrate supplementation with inactivated microorganisms (Salminen et al, 2021) into this term. However, this inclusive, industry‐backed definition remains highly controversial (Aguilar‐Toalá et al, 2021), with many microbiome researchers relating to “postbiotic” therapy only in the context of a well‐defined and evidence‐based small molecule intervention (Cullin et al, 2021; Vrzáčková et al, 2021; Box 1).

Table 1.

Different classes of microbial products, disease contexts, and organismal platforms of conducted studies.

Category Molecule Source(s) Platforms of study Disease context
Short chain fatty acids (SCFAs) Butyrate, propionate, acetate Carbohydrates (diet) In vitro, rodents, human: microbial transfer, metabolite supplementation Intestinal inflammation, metabolic syndrome, adiposity, hypertension, atherosclerosis, ischemic stroke, chronic kidney disease, type‐1 Diabetes mellitus
Carboxylic acid intermediates Lactate, succinate Carbohydrates (diet) In vitro, rodents, human: microbial transfer Metabolic syndrome, intestinal epithelial regeneration, bacterial vaginosis
Amino acids and derivatives Branched‐chain amino acids (BCAAs), niacin/nicotinamide, 5‐aminovaleric acid, dimethylglycine, acetylglycine Amino acids (diet and microbial de‐novo synthesis) In vitro, rodents, human: observational Adiposity, cardiovascular events, amyotrophic lateral sclerosis (ALS), intestinal inflammation and carcinogenesis, atherosclerosis, myocardial infarction, stroke
Taurine Primary bile acids (host metabolism) In vitro, rodents Intestinal inflammation
Trimethylamine‐oxide (TMAO) Host metabolism of microbial trimethylamine (TMA) In vitro, rodents, human: observational Metabolic syndrome, type‐2 Diabetes mellitus, liver steatosis, atherosclerosis, myocardial infarction, ischemic stroke, thrombosis
Pattern receptor recognition (PRR) ligands Lipopolysaccharide (LPS), peptidoglycan, lipoteichoic acid (LTA), polysaccharide A, bacterial DNA, secreted microbial proteins Structural components of microbes In vitro, rodents, human: observational Intestinal inflammation, infection and carcinogenesis, adiposity, liver steatosis, inflammation and fibrosis, acute liver failure, thrombosis, metabolic syndrome, type‐1 Diabetes mellitus
Tryptophan metabolites Indole‐3‐propionic acid (I3PA), indole‐3‐aldehyde (IAId), indoxylsulfate (IS), tryptamine Amino acids (diet) In vitro, rodents, human: observational Intestinal inflammation, fungal infection, hypertension, chronic kidney disease, type‐1 Diabetes mellitus, atopic dermatitis
Flavonoids Quercetin, apigenin, naringenin Polyphenols (diet) In vitro, rodents Adiposity, intestinal inflammation and infection
Secondary bile acids lithocholic acid (LCA), deoxycholic acid (DCA) & derivatives Primary bile acids (host metabolism) In vitro, rodents, human: observational Liver steatosis, inflammation and fibrosis, metabolic syndrome, adiposity, colon cancer, malabsorption and micro‐nutrient deficiency, C. difficile infection
Other classes Tetrahydrobiopterin (BH4), folate, sphingolipids, amyloids Microbial de‐novo synthesis, amino acids (diet) In vitro, rodents Metabolic syndrome, intestinal inflammation, Parkinson's disease, autism spectrum disorder (ASD)

ALS, amyotrophic lateral sclerosis; ASD, autism spectrum disorder; BCAAs, branched‐chain amino acids; BH4, tetrahydrobiopterin; DCA, deoxycholic acid; I3PA, indole‐3‐propionic acid; IAId, indole‐3‐aldehyde; IS, indoxylsulfate; LCA, lithocholic acid; LPS, lipopolysaccharide; LTA, lipoteichoic acid; SCFA, short chain fatty acids; TMA, trimethylamine, TMAO, trimethylamine‐oxide.

Box 1. In need of answers.

Will consensus be reached on a clear definition and classification of therapeutic products derived from microbes or targeting microbial metabolism?

Is the role of certain microbial metabolites in health and disease correlative or causative?

Are the current analytical pipelines for microbiomes and their metabolites reflecting the in vivo conditions and can they be reproduced across studies?

What are the translational limitations of microbial metabolite focused studies in animal models to the human setting?

What host factors contribute to beneficial or deleterious effects of microbial metabolites?

Are personalized approaches to make use of the effects of microbial metabolites for health benefits superior to resource‐saving one‐for‐all strategies?

In this review, we aim to exemplify major mechanisms of action of microbially modulated metabolites, impacting host physiology and disease both locally at their site of production and following systemic distribution. Rather than focusing on the description of metabolite classes and their impact on selected organs (addressed in excellent reviews (Wahlström et al, 2016; Borghi et al, 2020; van der Hee & Wells, 2021)), we illuminate the differential roles played by key microbial metabolites in their local environment of production, as compared to remote effects mediated by metabolite influx into the host's systemic circulation.

Effects of microbial metabolites on their local environment

As metabolically active organisms, microbes and their immediate neighboring host tissues have co‐evolved to take advantage of each other's unique metabolic capabilities. Metabolites secreted and modulated by both constituents of the “holobiont” are used in various fashions, ranging from energy sources, sensing of nutrients, directing physiologic responses or preventing the expansion of malicious microorganisms (Fig 1). At times, detrimental processes such as invasive pathogen infections or neoplasia may “hijack” metabolites and their signaling pathways in conferring a competitive advantage to themselves. Key examples of local metabolite functions are depicted below.

Figure 1. Molecules of microbial origin interact with the local environment at their site of production.

Figure 1

The indigenous microbiome secretes amino acids and tryptophan metabolites maintaining its own stability through tolerogenic signals received from the host's epithelium. Other commensal factors such as bile acids, lactic acid, or short chain fatty acids (SCFAs) can counteract the overgrowth of pathobionts. Besides supplying micronutrients including vitamin B9, iron, and zinc directly to the host, the microbiome also modifies the absorption of dietary components in the gastrointestinal tract: conjugated bile acids increase the solubility of lipids, while lactic acid contributes to maintain pH levels preventing the absorption of toxic ammonia. Bacterial products such as SCFAs, flavonoids, tryptophan metabolites, amino acids, and toll‐like receptor (TLR)‐ligands contribute to fortify the intestinal barrier, preventing bacteria from dislocating into deeper mucosal layers. Intestinal epithelial cells (IECs) utilize SCFAs as their main metabolic precursors for the harvest of nutrients. They also utilize SCFAs and bile acids in the regulation of metabolic circuits resulting in the production and systemic deployment of glucose or ceramides. SCFAs or tryptamine can promote intestinal motility activating feedback‐loops involving the epithelium, local neurons, and smooth muscles. While lactate and TLR‐agonists can progress regenerative processes of the epithelium after injury, bile acids have the potential to drive proliferative signals facilitating tumor development. SCFA, short chain fatty acid; TLR, toll‐like receptor; Trp, tryptophan. (Created with BioRender.com).

Intestinal barrier function

Microbes inhabit large mucosal surfaces within their host. While commensals contribute to host barrier function in preventing colonization and translocation of exogenous pathogens, the large commensal biomass itself must be regulated by the host barrier in preventing harmful microbial invasion. The microbiome also provides crucial stimuli to the host to maintain the function of this important barrier. For example, tight junctions (TJ) and their cytoskeleton anchors constitute an important component of the intestinal epithelial barrier. Upregulation of their production can be induced by various gut bacteria‐induced metabolites in mouse models, such as SCFAs (Wang et al, 2020), products of tryptophan‐degradation, like indole‐3‐propionic acid (I3PA; Venkatesh et al, 2014), the flavonoid quercetin (Suzuki & Hara, 2009; Carrasco‐Pozo et al, 2013), and amino acids (Grosheva et al, 2020). Metabolite‐induced fortification of gut barrier function can increase intestinal resistance to several noxious events modeled in rodents, such as inflammation (Wang et al, 2020) or drug‐induced injury (Singh et al, 2017). Bacterial toll‐like receptor (TLR)‐ligands activate Nod‐like receptor pyrin domain‐containing protein 6 (NLRP6) inflammasomes in “sentinel” goblet cells (senGCs) in lower crypts of the colon. This triggers mucus secretion and luminal expulsion of senGCs, both fortifying the mucus layer to prevent bacteria from breaching the intestinal barrier (Birchenough et al, 2016). The entry of microbes into the systemic circulation is also limited by the gut vascular barrier (GVB; Spadoni et al, 2015). Bile acids that are agonists of the farnesoid X receptor (FXR) have a stabilizing effect on the GVB in mouse models of liver injury (Sorribas et al, 2019). Bile acid deconjugation through gut microbes generally impacts the bile acid pools toward FXR agonists in mice and humans (Tremaroli et al, 2015; Parséus et al, 2017; Quinn et al, 2020) and therefore could play a role in maintaining the GVB. A combinatorial impact of multiple, yet uncharacterized barrier‐strengthening and disrupting metabolites (Grosheva et al, 2020) likely contributes to differences in barrier function observed in different clinical and pathological contexts and merits future mechanistic studies.

Energy source

Microbial metabolites may also serve as sources of energy for their surrounding tissue. For example, as a result of co‐evolution with their neighboring microbiome, intestinal epithelial cells (IECs) have adapted to harness nutrients in a variety of metabolic processes, such as oxidation, lipogenesis or the generation of ketone bodies, and amino acids (Bugaut, 1987; Bergman, 1990). SCFAs generated through microbial carbohydrate fermentation are utilized by IECs as energy sources (den Besten et al, 2013a). Additionally, a study in rats found vasoactive intestinal peptide (VIP) inducing epithelial gluconeogenesis, while propionate led to an increase of VIP‐expressing neurons in the gut submucosal plexus (De Vadder et al, 2015). SCFAs are also able to engage in epithelial lipid metabolism. Acetate can be transformed into the metabolic intermediate Acetyl‐CoA in IECs. This leads to activation of the anti‐lipogenic master regulator AMP‐activated protein kinase (AMPK) and therefore to increased fatty acid oxidation and downregulated lipid production. As a consequence, fewer lipids are shuttled to the lymph and systemically available as chylomicrons (Araújo et al, 2020). Acetyl‐CoA derived from L‐lactate of microbial origin features an opposite effect on lipid metabolism in IECs. Its transformation to Malonyl‐CoA results in intracellular lipogenesis and decreased oxidation of lipids. Surprisingly, this leads to lower secretion of lipids from IECs despite an increased storage (Araújo et al, 2020). However, lipogenesis triggered by microbial metabolites can also have undesirable outcomes in downstream organs. Exposure to bile acids that act as FXR‐agonists leads to IEC‐production of ceramides, lipid molecules consisting of a sphingosine and fatty acid, contributing to hepatic lipogenesis in mice (Jiang et al, 2015). The beneficiary effect of the antidiabetic drug metformin depleting B. fragilis recently reported in a human cohort of Type‐2‐diabetics (Sun et al, 2018) could be a result of disruptions of microbial folate and methionine metabolism (Induri et al, 2022). Through this mechanism, the ability of Bacteroides to transform bile acids that fuel ceramide production is abrogated (Sun et al, 2018).

Microbially modulated bile acids can also alter the response of the endocrine pancreas to incretins released from intestinal L‐cells in response to dietary signals. Mouse models found bile acids deconjugated by Clostridium and Bacteroides to bind the membrane G protein‐coupled bile acid receptor 1 (TGR5) on L‐cells, inducing the release of insulinotropic glucagon‐like peptide 1 (GLP‐1; Pathak et al, 2018), possibly through inhibition of local glycolysis (Trabelsi et al, 2015). In contrast, bile acid engagement with the nuclear receptor FXR in L‐cells leads to a downregulation of SCFA‐receptors, thereby abrogating their positive effects on GLP‐1 release (Ducastel et al, 2020).

Intestinal motility

Both commensals, their dietary exposures (Dey et al, 2015), and microbial metabolite repertoires are involved in gut motility and the control of luminal content. SCFAs are able to trigger the release of the hormone peptide YY (PYY) from L‐cells. As a late postprandial signal, PYY in murine ex vivo mucosa led to an inhibition of chloride driving fluid secretion into the gut lumen. It also interacts with both layers of the enteric smooth muscle as well as neurons belonging to the vagal afferent system leading to an antisecretory response and inhibition of gastro‐intestinal motility in vivo (Tough et al, 2011). SCFAs can also induce pro‐contractile responses. Neurons located in the rat intestinal myenteric plexus bind butyrate through monocarboxylate transporter 2 (MCT2) receptors, resulting in propulsive contractions (Soret et al, 2010). The same effect is observed upon acetylcholine release triggered from IECs upon exposure to propionate (Yajima et al, 2011). SCFAs are also capable to act on central nervous sensory and sympathetic ganglia in mice through transcriptional regulation, modulating gastrointestinal motility through a gut–brain circuit (Muller et al, 2020). A different bacterial metabolite, tryptamine derived from tryptophan by R. gnavus and C. sporogenes, exerts pro‐contractile signals (Bhattarai et al, 2018). Upon binding serotonin‐receptors, adenylate cyclase activation leads to luminal secretion of Cl and HCO3 ions as well as water, leading to a shortened colonic transit time in humanized mice (Bhattarai et al, 2018).

Intestinal digestion and absorption

Microbial metabolites may modify the digestion and absorption of dietary compounds. Bile acids are responsible for the emulsification of hydrophobic fat droplets into micelles and therefore heavily influence the digestion and absorption of lipids and fat‐soluble vitamins (A, D, E, K). Microbial deconjugation of bile acids tends to reduce their emulsifying potential (Swann et al, 2011). However, most of the absorption of dietary lipids and vitamins takes place in the small intestine (Iqbal & Hussain, 2009), while bacterial deconjugation through bile salt hydrolases is commonly found in the large intestine (Guzior & Quinn, 2021). This geographical distinction can drastically change after surgical creation of blind loops or the congenital occurrence of large diverticula in the small intestine. These regions of blind loops, isolated from the regular gut transit, can undergo an overgrowth of anaerobic bacteria capable of bile salt deconjugation. The consequential impaired emulsification can eventually lead to lipid maldigestion causing diarrhea and a deficiency of a number of vitamins due to malabsorption (Quigley et al, 2020). Furthermore, microbial‐derived products also feature a regulatory impact on intestinal lipid absorption and digestion, as these processes were found defective in germ‐free mice. As such, bacterial metabolites may trigger the release of enzymes involved in digestion and transepithelial shuttling of lipids (Martinez‐Guryn et al, 2018) and the maturation of lymphatic vessels draining lipids absorbed in the intestine (Suh et al, 2019). The production of SCFAs from colonic microbial fermentation results in local acidification, which may augment absorption. One example is the shift from ammonia (NH3) toward the less diffusable cationic ammonium (NH4 +) under acidic conditions. This principle has been applied for more than half a century in the treatment of hepatic encephalopathy, in which ammonia cannot be detoxified due to poor liver function. Lactulose, a nonabsorbable disaccharide, contributes to acidic conditions upon microbial digestion in the colon and trapping of ammonia (Agostini et al, 1972).

Gut commensals are also capable of producing micronutrients. Though these compounds maintain numerous host metabolic, cellular, and immune functions, only few insights exist about the systemic impact of their bacterial synthesis (Biesalski, 2016). A defective gut bacterial production of folic acid was recently suggested to be linked to the development of Parkinson's disease (PD) in a human study cohort (Rosario et al, 2021), in line with previous findings of lowered systemic folate levels in patients with PD (dos Santos et al, 2009) and PD‐like symptoms in mouse models of folic acid deficiency (Duan et al, 2002). The microbial generation of SCFAs through carbohydrate fermentation may impact the intestinal bioavailability and absorption of certain trace elements. The supplementation of dietary fiber leads to an increase of trans‐epithelial iron transport in the cecum in a rat model of anemia (Lobo et al, 2014), while improving the bioavailability of zinc in another study (Scholz‐Ahrens & Schrezenmeir, 2002). Nevertheless, the impact of metabolites of microbial origin in determining the bioavailability of trace elements merits further studies, especially in the human setting (Coudray et al, 1997).

Protection against pathogens and pathobionts

Microbial metabolites are involved in the reaction against the overgrowth and invasion of potentially noxious organisms while allowing indigenous commensals to harbor their niches. For example, intestinal infection with C. difficile is often triggered by a disruption of eubiotic communities through broad‐spectrum antibiotic treatment. C. scindens was found to counteract the blossom of this pathobiont through deconjugation of the bile acid cholic acid (Buffie et al, 2015). The augmentation of a healthy intestinal microbial community through fecal microbial transplantation (FMT) is effective as treatment of therapy‐resistant C. difficile infection. Metabolites derived from the transplanted microbiome, such as the bile acids deoxycholic acid (DCA) and lithocholic acid (LCA), and the SCFAs propionate and butyrate may contribute to this beneficial effect (Seekatz et al, 2018).

Microbial communities can induce tolerogenic signals in their host, thereby enabling the persistence of microbes with beneficial effects and preventing the overgrowth of pathogenic strains. Tryptophan metabolites produced by lactobacilli lead to Il‐22 release upon aryl hydrocarbon receptor (AHR) signaling from intestinal immune cells, counteracting Candida colonization through an amplified antifungal immune response (De Luca et al, 2010; Zelante et al, 2013). Vice versa, Il‐22 secretion also serves to maintain a community rich in bacterial AHR‐ligand producers: Defective Il‐22 signaling in mice led to dysbiosis concomitant with low AHR‐ligand production and colitis susceptibility, while in humans, lower gut microbial AHR activation and a single nucleotide polymorphism linked to impaired Il‐22 function were associated with inflammatory bowel disease (Lamas et al, 2016). A similar effect was observed upon gut microbial taurine production in mice inducing epithelial NLRP6 inflammasome activation and Il‐18 secretion. Subsequently, the release of antimicrobial peptides (AMPs) counteracted an overgrowth of bacteria that lead to an inflammatory reaction and sustained the commensal taurine producers (Levy et al, 2015).

Beyond the gut, lactic acid produced in the vaginal mucosal surface by resident lactobacilli (most effectively by the crispatus species) is present in its protonated form at pH levels < 3.9. In this state, it exerts direct antimicrobial effects protective of bacterial overgrowth (Tachedjian et al, 2017), coupled with virucidal effects on HIV and HSV‐2 (Conti et al, 2009; Aldunate et al, 2013). A recent first‐in‐human clinical trial suggested that restoration of the vaginal microbiome with vaginal microbiome transplantation (VMT) successfully reestablishes lactic acid‐producing communities and reverses the overgrowth of anaerobes, collectively improving intractable and recurrent bacterial vaginosis and its associated symptoms (Lev‐Sagie et al, 2019). The contribution of metabolite shifts to this effect merit further studies.

Epithelial cell replenishment

Epithelial cells are frequently replaced through differentiation and proliferation of intestinal stem cells. This enables intestinal regeneration after injury, but, when excessive and unregulated, also increases susceptibility toward colorectal cancer development. Bacterial‐produced lactate is involved in a number of proliferative processes in the intestinal epithelium. It interacts with Paneth cells through the receptor GPR81, leading to epithelial expansion and maintenance of intestinal stem cells (ISCs) in mice through Wnt/β‐catenin signals (Lee et al, 2018). Recognition of bacterial components through specialized pattern recognition receptors (PRRs) plays an important role in all phases of gut epithelial regeneration after injury: In mouse models of chemical colitis it was involved in initial epithelial restitution (Fukata et al, 2006; Normand et al, 2011), proliferation (Hsu et al, 2010; Zaki et al, 2010), and eventually differentiation (Podolsky et al, 2009; Round et al, 2011). Mice subjected to starvation exert a hyperproliferation of IECs upon refeeding triggered by lactate as a metabolite derived from Lactobacillus murinus (Okada et al, 2013). Additionally, chronic inflammatory or premalignant conditions may trigger proliferative signals from microbial metabolites that can induce oncogenic pathways. For example, intestinal adenoma‐prone ApcMin mouse exposure to deoxycholic acid, a product of bacterial bile acid deconjugation, may trigger the intestinal adenoma–carcinoma pathway concomitant with activation of the inflammatory Il6‐STAT3 axis (Wang et al, 2019). Sensing of bacterial components through TLR2 and nucleotide binding and oligomerization domain 2 (NOD2) induces the expression of major histocompatibility complex (MHC) class II in ISCs. This contributed to immune surveillance of the intestinal epithelium and prevented the formation of tumors under premalignant conditions in mice (Beyaz et al, 2021).

Systemic impacts of microbial products on the host

In addition to the aforementioned local mechanisms of metabolite activity, their distribution throughout the body by portal and systemic circulation may result in interaction with a variety of remote organs and cells, where they may influence their host's systemic homeostasis and risk of disease (Fig 2, Table 1). In the below section, we exemplify some of these effects by focusing on metabolites impacting systemic features of host metabolism. In a separate section, immune‐mediated effects will be discussed.

Figure 2. Microbial metabolites systemically impact host physiology.

Figure 2

Numerous metabolic functions of the host are influenced by products of microbial origin. Short‐chain fatty acids (SCFAs) can act as precursors for lipid or glucose production in the liver. SCFAs and bile acids may also induce hepatic pathways to degrade glucose and lipids. In muscle, SCFAs can increase the net uptake of glucose and induce the transformation to type‐1 fibers capable of increased fatty acid oxidation. Additional improvements of muscle metabolism can be induced by bile acids or amino acids, such as a higher energy expenditure. Adipose tissue may respond to SCFAs or bile acids modulating the transformation of white adipose tissue (WAT) to brown adipose tissue (BAT) to enhance thermogenesis. Microbe‐associated molecular patterns activating toll‐like receptors (TLRs) and nucleotide binding and oligomerization domain receptors (NLRs) as well as microbiome‐modulated amino acids may trigger steato‐inflammation in adipose tissue. Amino acids of bacterial origin, particularly trimethylamine‐oxide (TMAO) and branched‐chain amino acids (BCAAs), are linked to the development of atherosclerosis, myocardial infarction or stroke. In patients suffering from chronic kidney disease (CKD), SCFAs may modulate disease progression, while an impaired degradation of the tryptophan metabolite indoxylsulfate (I3S) could aggravate uremic symptoms. In the central nervous system (CNS), microbial metabolic dysfunction resulting in a lack of nicotinamide in amyotrophic lateral sclerosis (ALS) or an excess of amyloids in Parkinson's disease (PD) contribute to neurodegeneration and worsening motor functions. Behavioral disorders, such as autism spectrum disorder (ASD) are accompanied by alterations of the microbial production of amino acids, such as 5‐aminovaleric acid and taurine upon microbiome transfer to germ‐free mice or tetrahydrobiopterin (BH4) in a mouse model of ASD. ASD, autism spectrum disorder; BAT, brown adipose tissue; BCAA, branched‐chain amino acid; CKD, chronic kidney disease; CNS, central nervous system; CVS, cardiovascular system; MACE, major adverse cardiovascular event; NLR, nucleotide binding and oligomerization domain receptor; SCFA, short chain fatty acid; TLR, toll‐like receptor; TMAO, trimethylamine‐oxide; Trp, tryptophan; WAT: white adipose tissue. (Created with BioRender.com).

Glucose metabolism

Every cell in the mammalian body is capable of utilizing glucose as a source of energy. Systemically distributed microbial products act as metabolic precursors and in signaling cascades modifying their hosts' glucose metabolism. The potential of SCFAs to act as gluconeogenic substrates through incorporation into the tricarboxylic acid (TCA) cycle has already been mentioned in the previous local metabolic section. Systemically, radiolabeling suggests that propionate may also contribute to a net glucose synthesis (den Besten et al, 2013b). The intestinal metabolic capacity significantly contributes to energy homeostasis, with SCFAs converted to glucose in IECs inducing decreased bodyweight, improved insulin sensitivity, and ameliorated hepatic glycogenolysis in mice (De Vadder et al, 2014, 2016). Propionate interacting with periportal afferent neurons was suggested to drive this effect (De Vadder et al, 2014). SCFAs can act as precursors of two major energy sources of skeletal muscle, glucose and fatty acids. They are also involved in muscle cell uptake of glucose through an increase of intracellular transport (Yamashita et al, 2009). SCFAs can induce a switch toward the fatty‐acid oxidizing type‐I fiber, facilitating glucose clearance and increasing insulin sensitivity in mice (Gao et al, 2009).

Additionally, microbially deconjugated secondary bile acids have beneficial metabolic effects on skeletal muscles. In a mouse model, TGR5‐triggered cAMP‐dependent thyroid hormone‐activated enzyme type 2 iodothyronine deiodinase (D2), thereby leading to a higher energy expenditure (Watanabe et al, 2006). Amino‐acid derivatives undergo metabolic conversions through both host and microbial enzymes. Trimethylamine‐oxide (TMAO) is generated in the liver from trimethylamine, which is produced by gut microbes from multiple precursors including choline, carnitine, and betaine (Chhibber‐Goel et al, 2016). In rodents, TMAO may modulate glucose homeostasis via induction of the unfolded protein response (UPR), though the downstream results remain debated. Work by Dumas et al (2017) suggests that TMAO induces an improved insulin secretion and glucose tolerance in mouse models of metabolic dysfunction, while others (Chen et al, 2019b) argue that TMAO interacts with the UPR‐effector PERK, thereby disrupting glucose tolerance. A study in non‐diabetic humans did not find serum levels of TMAO to be associated with fasting glucose or insulin resistance, but suggested that it may constitute an independent predictor of meeting one of the prediabetes‐defining criteria (Roy et al, 2020).

Adiposity

Historic observations in human subjects suggested a correlation between plasma SCFA levels and adipose tissue mass (Björntorp & Hood, 1966). In agreement, a study comparing gut microbial community structure in lean and obese twins (Turnbaugh et al, 2009), and another utilizing FMT from obese donors or those successfully reducing weight following bariatric surgery (Tremaroli et al, 2015) concluded that gut microbes from obese subjects harbor the capacity to harvest energy through enhanced carbohydrate fermentation and generation of SCFAs. FMT from lean and obese human twins into germ‐free mice demonstrated a decline in adiposity in recipients of microbiomes from lean individuals, accompanied by an increased capacity to metabolize carbohydrates and higher propionate and butyrate levels (Ridaura et al, 2013). In contrast, human FMT from lean to obese subjects failed to alter SCFA levels (Mocanu et al, 2021) or induce sustainable metabolic improvements (Zhang et al, 2019). Other studies in mice suggested that higher SCFA availability may induce white adipose tissue (WAT) browning and an expansion of beige adipose tissue (BAT; Li et al, 2017, 2019; Weitkunat et al, 2017), potentially mediated by fatty acid oxidation (Gao et al, 2009), phosphorylation of AMPK, or reduction of PPARγ‐signaling (den Besten et al, 2015; Gao et al, 2019). Besides a direct impact on adipose tissue, SCFAs can cross the blood–brain barrier (Li et al, 2019) and modulate neural circuits by releasing neuropeptide Y (NPY) that drives BAT activation and upregulation of thermogenesis (Li et al, 2018) in mouse models of obesity. Moreover, the release of the peptide hormone leptin from adipocytes, best known for its effects on satiety, is triggered by SCFAs (Xiong et al, 2004; Zaibi et al, 2010).

Additionally, microbially modulated bile acids may participate in regulation of adiposity (Tremaroli et al, 2015). The exposure of mice to cold temperatures led to changes in the gut microbiome and bile acid profile triggering lipolysis (Ziętak et al, 2016; Worthmann et al, 2017). Switches in the microbial bile acid metabolic capacity can result in functional reprogramming of the adipose tissue (Li et al, 2013; Jiang et al, 2015; Pathak et al, 2018) enabling thermogenesis, lipolysis, and mitochondrial uncoupling (Broeders et al, 2015; Somm et al, 2017; Velazquez‐Villegas et al, 2018). TMAO (originating from a bacterially produced precursor) may induce or aggravate hepatic steatosis (Chen et al, 2016; Tan et al, 2019; León‐Mimila et al, 2021). A possible mechanism for this effect may involve microbial producers of TMA (the TMAO precursor) outcompeting the host for choline. The hepatic and visceral adipogenesis are a result of impaired DNA‐methylation as well as decreased availability of phosphatidylcholine for lipoprotein synthesis (Romano et al, 2017).

In other contexts, dysbiosis in cigarette‐smoking humans and smoke‐exposed mice may increase the microbial conversion of the amino acid glycine into betaine and subsequently dimethylglycine (DMG), concurrently depleting acetylglycine (ACG). Upon smoking cessation, the dysbiosis in mice remained persistent, while the active smoking‐related anorexigenic molecules dissipated, with a combination of increased DMG and depleted ACG contributing to the exacerbation of smoking cessation‐related weight gain mediated through increased energy harvest. Interestingly, these metabolite effects may also be active upon supplementation to non‐smoking obese mice (Fluhr et al, 2021). Another common and poorly understood obesity pattern in humans, involves recurring and gradually exacerbating cycles of obesity, also termed “yoyo” obesity. In mouse models of this obesity pattern, apigenin and naringenin, flavonoids modulated by the gut microbiome, are depleted upon induction of obesity, but fail to replenish upon successful dieting. Depletion of these flavonoids contributes to a susceptibility to excessive weight regain upon exposure to further bouts of obesity by impaired regulation of BAT decoupling of oxidation‐phosphorylation, driving excessive net fat accumulation (Thaiss et al, 2016a).

Lipid metabolism

Hyperlipidemia, a component of the metabolic syndrome and a contributor to cardiovascular morbidity, may likewise be influenced by the microbiome (Zmora et al, 2018). Several metabolites may impact the metabolism of lipids at sites distant from their production. The liver, exposed to an array of metabolites originating from the gut through the portal vein, exerts a key role in lipid metabolism. The SCFAs butyrate and acetate supplemented to mice were shuttled from the gut and served as precursors for palmitate and cholesterol synthesis in the liver (den Besten et al, 2013b). Whether this function additionally contributes to obesity (Samuel et al, 2008) or fatty liver in humans (Chambers et al, 2019) remains debatable. This is due to the fact that numerous other studies found SCFAs potentially modulating lipid levels through several mechanisms including an activation of the master regulator AMPK/SREBP‐1 axis (Gao et al, 2009; den Besten et al, 2015; Wu et al, 2019), increased β‐oxidation of fatty acids (Gao et al, 2009; den Besten et al, 2015), and long‐chain fatty acid (LCFA)‐induced modulation of the ω6/ω3 ratio (Weitkunat et al, 2016). Sphingolipids (SL) produced by gut bacteria downregulate the host production of SLs in the liver (Johnson et al, 2020), a process linked to metabolic disorders such as insulin resistance and nonalcoholic steatohepatitis (Apostolopoulou et al, 2018).

Cardiovascular system

Cardiovascular disease (CVD) complications, including myocardial infarction (MI) or stroke, constitute a major source of morbidity and mortality in individuals suffering from the above features of the metabolic syndrome (Alberti et al, 2006; Roth et al, 2020). In the past decade, the microbiome and its metabolites were suggested to contribute to the pathogenesis of CVD. For example, gut microbiomes of omnivorous people harbored a higher capacity of processing L‐carnitine compared to those of vegetarians and vegans, leading to increased systemic levels of TMAO (Koeth et al, 2013). Higher plasma L‐carnitine and TMAO levels were identified as risk factors for coronary atherosclerosis and major cardiovascular events (Koeth et al, 2013; Senthong et al, 2016). Increased levels of TMAO were also associated with higher risk of stroke in the presence of hypertension or atrial fibrillation (Nie et al, 2018; Liang et al, 2019) A possible mechanistic explanation of this increased risk (Zhu et al, 2016) included a direct impact of TMAO on the activation of platelets, a critical step in the emergence of clot formation driving ischemic cardiovascular events. As such, FMT of gut microbial communities featuring enhanced capability of TMAO production to humanized mice induced a pro‐thrombotic vascular dysfunction (Zhu et al, 2016). Structural components of bacteria can influence clot formation through PRR‐interaction: Platelet TLR2 was found to mediate aggregation in vitro upon S. pneumoniae recognition (Keane et al, 2010), while in mice, LPS triggered platelet TLR4 expression (Aslam et al, 2006) resulting in adhesion and accumulation in the lung capillaries (Andonegui et al, 2005). Sensing bacterial signals through NOD‐receptors on platelets induced MAPK and NO‐signaling pathways eventually increasing clot formation in mesenteric vessels in a mouse model of thrombosis (Zhang et al, 2015). Branched‐chain amino acids (BCAAs) were also linked to cardiovascular events like MI or stroke, although this effect may have been indirectly driven by impaired glucose metabolism (Tobias et al, 2018). Humans with acute ischemic stroke were found to harbor alterations of gut microbial communities associated with decreased SCFA production (Tan et al, 2021). In rats that developed similar dysbiotic traits upon ischemic stroke, replenishment of SCFA levels through FMT or direct butyrate supplementation led to improved neurological outcomes (Chen et al, 2019a). Another small study (Hayashi et al, 2021b) suggested that gut microbial community alterations were linked with an enhanced BCAA synthesis in people suffering from heart failure. Interestingly, tryptophan derivatives of bacterial and host origin may impact cardiovascular health in opposing manners. AHR‐agonists of bacterial origin like indole‐3‐acetic acid (IAA) alleviated hypertension in mice (Wilck et al, 2017), while host production of kynurenines from tryptophan was associated with acute coronary events in a study cohort of elderly humans (Eussen et al, 2015). Microbial metabolites may also prevent atherosclerosis. A genome‐wide association study suggested that microbially produced butyrate is negatively associated with atherosclerotic cardiovascular disease (Jie et al, 2017). Likewise, studies in rodents (Marques et al, 2017; Wang et al, 2017) and human diabetics (Roshanravan et al, 2017) pointed toward the potential role of SCFAs to attenuate high blood pressure as well as chronic injury of the heart and kidneys. However, these suggested microbiome‐driven effects on blood pressure (Chen et al, 2020; Cook & Chappell, 2021) may also be indirectly driven by altered weight and glucose metabolism.

Systemic nonmetabolic effects

In addition to the above systemic metabolite‐driven effects on host metabolism, microbial‐modulated metabolites may modify a variety of other physiological and pathological processes in numerous cells and organs. For example, in the brain, a mouse model of amyotrophic lateral sclerosis (ALS) featured dysbiosis in the gut preceding the motor symptoms (Blacher et al, 2019). A decrease of A. muciniphila in ALS‐bearing mice was accompanied by decreased systemic levels of nicotinamide, a metabolic disturbance that was also found in a cohort of ALS patients. Akkermansia or nicotinamide supplementation improved motor symptoms and triggered protective transcriptional neural pathways in the mouse ALS model, encouraging a future testing of this and other differentially abundant metabolites in human ALS clinical trials. Likewise, microbiome‐associated metabolites may modify disease course in other central nervous system disorders. The aggregation of the amyloid protein alpha‐synuclein (aSyn) is associated with several neurodegenerative disorders, including Parkinson's disease (PD). An amyloidogenic product secreted by Enterobactericae named “Curli” may increase the formation of aSyn‐aggregates, while colonizing an aSyn‐overexpressing mouse line with a Curli producing E. coli strain worsened the PD‐like motor and intestinal impairments (Sampson et al, 2020). Other metabolite alterations correlated with autism spectrum disorder (ASD). 5‐aminovaleric acid and taurine alleviated symptoms in an ASD mouse model and were suggested as candidates to modify behavioral patterns in ASD patients (Sharon et al, 2019). Altered bacterial tetrahydrobiopterin (BH4) synthesis was associated with behavioral disorders in a mouse ASD model, while targeted replenishment with BH4 improved ASD‐like behavioral deficits (Buffington et al, 2021). These interesting observations merit further mechanistic validation in mice and humans. In the kidneys, the tryptophan derivate indoxylsulfate was found to accumulate in subjects suffering from chronic kidney disease (CKD), thereby contributing to uremic symptoms (Devlin et al, 2016). A probiotic intervention with a Lactobacillus strain increased renal SCFA levels, resulting in protective effects from fibrosis and chronic renal dysfunction after ischemia‐reperfusion injury in mice. A subsequent administration of the probiotic to a cohort of CKD patients in a placebo‐controlled study reduced the decline of kidney function over the period of observation (Zhu et al, 2021). Other examples of systemic metabolite‐induced effects are reviewed elsewhere (Needham et al, 2020; Rossi et al, 2020; Schupack et al, 2022).

Microbial metabolites shaping host immunity

Metabolite impacts on the mammalian immune system can be both local and systemic and are central in shaping microbiome‐host interactions and their physiological consequences. A variety of microbial recognition patterns of the host's innate and adaptive immune mechanisms are able to induce a myriad of host responses ranging from anti‐microbial activity to maintenance of tolerance and commensalism (Fig 3). While microbial surface components and nucleic acids are central in innate immune system microbial recognition, their metabolites constitute important additional means of signaling with and modulation of both innate and adaptive immune cells.

Figure 3. Microbiome‐modulated metabolites modify the innate and adaptive immune response.

Figure 3

Metabolite signaling contributes to a balance between tolerogenic and inflammatory immune reactions. Intestinal epithelial cells (IECs) sense short chain fatty acids (SCFAs); and other microbial metabolites through toll‐like receptors (TLRs), nucleotide binding and oligomerization domain receptors (NLRs), inflammasomes and Dectin1, in inducing tolerance to commensals versus inflammatory responses against pathogens. The secretion of antimicrobial peptides (AMPs) from Paneth cells and IECs upon sensing of amino acids, SCFAs, and microbial‐associated molecular patterns by TLRs‐ and NLRs contributes to protecting the gut from pathobiont bloom and invasion. Both monocytes and macrophages can recognize SCFAs, tryptophan metabolites, or TLR‐ligands affecting the differentiation of surrounding dendritic cells (DCs) or triggering the release of either pro‐ or anti‐inflammatory cytokines. In type 3 innate lymphoid cells 3 (ILC3s), recognition of SCFAs, bile acids, and aryl hydrocarbon receptor (AHR)‐ligands through specialized cell surface receptors also leads to the promotion of anti‐inflammatory cytokine patterns. Adaptive immune cells also interact with microbial metabolites. Classical dendritic cells (cDCs) recognize components of Gram‐positive bacteria through TLR2, while plasmacytoid dendritic cells (pDCs) express AHR and G‐protein coupled receptors (GPRs) interacting with tryptophan metabolites, SCFAs, and niacin. These antigen‐presenting cell (APC)‐signals modulate the activation of B and T cells. T cells also directly react to microbial signals in shaping their immunomodulatory phenotype: the differentiation of naïve T cells to regulatory T cells (Tregs) is reinforced by SCFAs and bile acids. Also, Tregs sense SCFAs and bacterial components through GPRs and TLR2 resulting in proliferation and the secretion of anti‐inflammatory cytokines. SCFAs can lead to an immunoglobulin A (IgA)‐class switch in B cells, so that secreted IgA in the intestinal lumen can counteract bacterial dissemination. Natural killer T (NKT) cells are polarized toward an immunomodulatory state counteracting inflammation in reaction to branched‐chain amino acids (BCAAs). CD8+ T cell sensing of SCFAs regulates their differentiation into memory cells and cytotoxic anti‐tumor activity. AHR, aryl hydrocarbon receptor; AMP, antimicrobial peptide; BCAA, branched‐chain amino acid; cDC, classical dendritic cell; DC, dendritic cell; GPR, G‐protein coupled receptor; IEC, intestinal epithelial cell; IgA, immunoglobulin A; MΦ, macrophage; NKT, natural killer T cell; NLR, nucleotide binding and oligomerization domain receptor; NOD, nucleotide binding and oligomerization domain; PC, Paneth cell; pDC, plasmacytoid dendritic cell; SCFA, short chain fatty acid; TCA, tricarboxylic acid cycle; TGR5, G protein‐coupled bile acid receptor 1; TLR, toll‐like receptor; TMAO, trimethylamine‐oxide; Treg, regulatory T cell; Trp, tryptophan. (Created with BioRender.com).

Modulation of the innate immune system

The innate immune system is tasked with sensing of microbiome commensals, helping the host to differentiate between “friend and foe” signals, and directing responses to these signals that can range from tolerance to inflammation. Microbial‐modulated metabolites are involved in these processes. IECs, increasingly considered an integral part of the mucosal immune system, serve as a critical communication hub, together with professional immune cells, in sensing and reacting to commensals and pathogens (Pott & Hornef, 2012; Soderholm & Pedicord, 2019). The most common specialized luminal receptors for the interaction with molecules of microbial origin are the G‐protein coupled receptors (GPRs) 41, 43, and 109 sensing SCFAs, as well as TLRs recognizing a variety of bacterial surface structures (Zheng et al, 2022). Several other surface receptors such as Dectin‐1 (Gantner et al, 2003), CD14 (Jiang et al, 2005), and CD36 (Hoebe et al, 2005) closely interact with TLRs in sensing components of putative pathogens. Additionally, intracellular detection and signaling is mediated by NOD‐like receptors (NLRs) and multiprotein‐complexes involving certain NLRs, termed inflammasomes. Collectively, integration of these diverse signals leads to tolerogenic versus inflammatory reactions (Rakoff‐Nahoum et al, 2004; Macho Fernandez et al, 2011; Macia et al, 2015; Wang et al, 2020).

IEC crosstalk with bacterial metabolites is also essential for the secretion of AMPs. IEC‐sensing of SCFAs (Zhao et al, 2018; Hayashi et al, 2021a) or the amino acid taurine activating NRLP6‐inflammasomes (Levy et al, 2015) trigger the release of AMPs. This increases the resistance toward noxious toxin‐producing C. difficile strains (Hayashi et al, 2021a) and attenuates the severity of experimentally induced colitis (Levy et al, 2015) in mice. Paneth cells can also independently respond with AMP‐secretion upon microbial components sensed through TLRs on their surface or intracellularly through the NOD2 pattern recognition receptor (Ogura et al, 2003). This contributes to lower intestinal penetration of bacteria and decreases colonization with pathobionts (Vaishnava et al, 2008).

However, the recognition of bacterial components through TLRs on host cells can have detrimental outcomes in cases of chronic inflammation, such as ulcerative colitis (UC). As such, TLR4 is upregulated in colon tumors found in UC‐patients as well as in a mouse model of inflammation‐associated colon cancer (AOM‐DSS; Fukata et al, 2007). The latter provides a potential mechanism linking TLR4 activation to the COX‐PGE‐EGFR pathway in promoting cancer (Fukata et al, 2007).

The innate immune system is also involved in responses to molecules of bacterial origin at sites remote from their production. Myeloid cells have emerged as critical hubs sensing and responding to bacterial signals. Innate lymphoid cells (ILCs) can be found in the subepithelial layers of organs harboring microbial communities. Their interaction with a variety of microbial products modulates their impact on innate immune response mechanisms. For example, SCFAs can influence type 3 innate lymphoid cells (ILC3s) through histone‐deacetylase (HDAC)‐inhibition (Yang et al, 2020), GPR43‐binding (Chun et al, 2019) or triggering of neutrophils to induce inflammasome activation (Fachi et al, 2020). In mouse models, this resulted in a protective effect from gut inflammatory injury (Chun et al, 2019; Yang et al, 2020), and infection (Chun et al, 2019; Fachi et al, 2020). Moreover, ILC3s exert anti‐inflammatory effects upon interaction with primary bile acids through TGR5 (Qi et al, 2019) as well as bacterial tryptophan derivatives binding AHR (Laurans et al, 2018; Hendrikx et al, 2019).

The liver serves as an immunologic “first‐pass” gate keeper, by responding to an array of metabolites influxing from the gut through the portal vein. Kupffer cells, the tissue‐resident macrophages in the liver, can respond to bacterial products while inducing distinct responses in their surrounding cell populations. Kupffer cell sensing of bacterial components through TLRs plays a role in triggering liver inflammation during liver steatosis (Henao‐Mejia et al, 2012; Carpino et al, 2020) as well as in acute liver failure (Kolodziejczyk et al, 2020). On the other hand, the abrogation of bacterial signaling through TLR‐ligands in germ‐free animals may lead to an increased susceptibility to hepatic fibrosis (Mazagova et al, 2015).

Adipose tissue (AT) also contains macrophages exerting an important role in regulating local inflammation and lipid storage. Their inflammatory responses can be modified by certain molecules of microbial origin. SCFAs may cause AT‐macrophages to abolish the secretion of various pro‐inflammatory cytokines and chemokines (Al‐Lahham et al, 2012; Ohira et al, 2013), including the adipokine resistin (Curat et al, 2006; Al‐Lahham et al, 2010). Increased gut permeability during obesity is driven by dietary compounds, perturbations of gut microbial communities, and the local immune response of the host, leading to endotoxemia (Serino et al, 2012; Tilg et al, 2020). In the AT, this results in a TLR4‐dependent recruitment of macrophages, triggering inflammation (Caesar et al, 2015). In addition, sensing of microbial patterns through TLR4 as well as NRLP3 in AT may trigger mitochondrial dysfunction upregulating fat storage processes, closely related to metabolic syndrome (Okla et al, 2018).

Modulation of the adaptive immune system

The adaptive immune response is an antigen‐specific and memory‐inducing immune reaction that evolved in vertebrates. Co‐evolution with complex microbial communities contributes to proper development and function of the adaptive immune response (McFall‐Ngai, 2007), while defective microbial colonization leads to an altered adaptive immunity, thereby triggering susceptibility toward infections (Mazmanian et al, 2005; Hall et al, 2008; Ivanov et al, 2009) and reduced efficiency of vaccinations (Korpe & Petri, 2012). Therefore, it is not surprising that microbial metabolites may shape functions of the adaptive immune system, in particular those involving several T cell subsets.

For example, SCFAs may impact intestinal T cell function by inducing their differentiation into anti‐inflammatory regulatory T cells (Tregs), while preventing a shift toward the pro‐inflammatory Th1/17 phenotype. This effect is mediated through HDAC‐inhibition (Arpaia et al, 2013) and metabolic reprogramming toward fatty acid oxidation (Hao et al, 2021). Similarly, SCFAs signaling via GPRs may directly target intestinal Tregs, leading to their increase in number and proliferative capacity through HDAC‐inhibition (Smith et al, 2013). To harness the local immunoregulatory potential of SCFAs, attempts of a postbiotic treatment were conducted in inflammatory bowel disease involving local administration of butyrate to patients suffering from ulcerous proctosigmoiditis (Scheppach et al, 1992; Scheppach & German‐Austrian SCFA Study Group, 1996). However, only a few of the heterogeneous and rather small‐scale studies were able to detect a decrease of inflammatory activity (Jamka et al, 2021).

The potential of SCFAs to facilitate an extrathymic development of anti‐inflammatory Tregs (Arpaia et al, 2013) can promote delivering immuno‐metabolic signals from gut microbes to distant effector organs. In mouse AT, butyrate leads to a Treg‐triggered alleviation of steato‐inflammation (Sato et al, 2020). In high‐fat diet induced obesity, SCFA‐triggered Tregs elicit weight loss and improve insulin sensitivity (Mandaliya et al, 2021). Inflammation in the course of pulmonary allergic hypersensitivity modeled in mice responds to increased SCFA levels leading to induction of higher numbers of Tregs (Trompette et al, 2014; Zaiss et al, 2015). In the pancreas, Tregs may alleviate the destruction of insulin‐producing β‐cells observed in the NOD1 mouse model of type‐1 Diabetes mellitus (T1DM). This effect is dependent on gut microbial SCFAs and AHR‐ligands, which, in turn induce pancreatic islet secretion of antimicrobial peptides (AMPs; Sun et al, 2015; Miani et al, 2018).

Importantly, SCFAs may also contribute to other adaptive immune effects. SCFAs may induce the inflammatory Th1/Th17 CD4 T cell phenotype in the setting of C. rodentium infection (Kim et al, 2013). Also, they may promote urethritis (Park et al, 2016), stimulate cytotoxic CD8+ lymphocytes (Luu et al, 2018), possibly contributing to their anti‐tumor effects, and impact B cell‐ and antibody‐related responses. An activation of GPR41 and GPR109 in DCs stimulates B cells to an IgA‐class switch and secretion of IgA into the gut lumen, thereby inhibiting bacterial dissemination into distant organs in the course of experimental colitis (Isobe et al, 2020).

Bile acids modified by gut commensals may also impact the differentiation and activity of T cells. Iso‐desoxycholic acid induces anti‐inflammatory FoxP3+/RoRγt+ Tregs by blocking nuclear FXR‐signaling in interacting DCs (Campbell et al, 2020). Both 3‐oxo lithocholic acid (LCA) and iso‐allo‐LCA direct T cells toward an anti‐inflammatory Treg phenotype while preventing Th17 differentiation (Hang et al, 2019). The mechanism by which both bile acids induce Tregs includes enhancement of mitochondrial function, resulting in transcription through histone acetylation at the promoter of FoxP3 (Hang et al, 2019).

While sensing of bacterial components by TLRs is mainly attributed to innate immune functions, it can also have an impact on inflammatory pathways driven by adaptive immune responses. Polysaccharide A (PSA), a component of the capsule of B. fragilis, induces a tolerogenic Treg phenotype through multiple mechanisms, including binding to TLR2 on plasmacytoid dendritic cells (pDCs; Dasgupta et al, 2014) or FoxP3+ CD4+ T cells (Round et al, 2011), as well as directly promoting the secretion of the anti‐inflammatory cytokine Il‐10 from T cells (Mazmanian et al, 2008). In the pancreas, microbial components interacting with different TLRs in mouse models can either exacerbate or alleviate autoimmune injury, a hallmark of T1DM. TLR2 may induce a microbiome‐dependent inflammatory impairment, while TLR4 exerts a protective tolerogenic effect on pancreatic islets (Burrows et al, 2015).

Niacin, produced by commensal bacteria in the gut from nicotinamide adenine dinucleotide (NAD), can activate local DCs and macrophages by interacting with their membrane receptor GPR109a (Singh et al, 2014). This subsequently drives a differentiation of naïve T cells into Tregs (Singh et al, 2014). These effects reduce gut inflammation and the subsequent carcinogenesis induced in the azoxymethane‐dextran sodium sulfate (AOM‐DSS) mouse model (Singh et al, 2014). Bacterial tryptophan metabolites signaling through AHR in the gut can induce CD4+8+ double‐positive intraepithelial lymphocytes (IEL; Cervantes‐Barragan et al, 2017) as well as non‐monocyte‐derived DCs (Kinnebrew et al, 2012), both associated with reduced intestinal inflammation. In the skin epithelia, supplementation with the tryptophan derivate indole‐3‐aldehyde (IAId) results in reduced inflammation caused by a mouse model of atopic dermatitis (Yu et al, 2019). This bacterial metabolite is decreased in the skin of people suffering from this disease (Yu et al, 2019).

Other metabolites of bacterial origin induce effector T cells. Glycosphingolipids, generated from BCAAs, lead to the development of NKT‐cells in the gut toward an immunomodulatory state capable of ameliorating colitis in mice (Oh et al, 2021). SCFA derivatives can be incorporated into the TCA of CD8+ T cells to enhance their metabolic capacity through mitochondrial oxidative phosphorylation, eventually improving survival and leading to polarization toward memory cells upon activation through antigen encounter (Bachem et al, 2019). Harnessing this beneficial effect may enable to sustain T cell memory after vaccinations or enable in vitro induction of memory chimeric antigen receptor (CAR) T cells in optimizing cancer immunotherapy.

Challenges and perspectives in the study of microbiome‐associated metabolites

As outlined in this review, metabolites derived from or modulated by microbial commensals impact their eukaryote host in a variety of local and systemic manners, which may affect host immunity, metabolism, and disease susceptibility. Disentangling the underlying mechanisms of these complex metabolite effects and harnessing them as microbiome‐targeted interventions remains complicated and elusive in most cases (Box 1).

Mechanistically studying metabolite impacts on host physiology and disease is complicated by multiple challenges. These include, among others, the lack of standardization of analytical pipelines, mainly those involving microbiome sequencing (Beresford‐Jones et al, 2022), and difficulties in culturing of many commensals, resulting in over‐reliance on their genomic characterization without sufficient evidence of causality (Cani, 2018). Moreover, functional analyses of bacterial metabolism, usually performed within the in vitro setting, does not account for metabolite impacts on the host and on neighboring microbes. Additionally, characterizing the “rules of engagement” of complex networks of microbial communities and their broad spectrum of metabolite secretomes necessitates the development of sophisticated computational tools and machine learning processes predicting the impact of metabolite consortia on distinct physiological responses.

Many approaches striving to determine metabolite causation and molecular mechanisms impacting disease processes heavily rely on rodent models. However, translational generalization of their findings to the human setting is impaired by the limited shared bacterial taxa (Chung et al, 2012), difficulties in reproducibility attributed to variations in microbial communities throughout animal vending and housing institutions (Beresford‐Jones et al, 2022), constrains in colonization and function upon transfer of human bacteria into mice (Lundberg et al, 2020) and variable effects of microbial metabolites on host physiology across species (Koh & Bäckhed, 2020). Furthermore, in some cases, secretion of microbial metabolites and their downstream impact on the host are investigated using unphysiological doses and application routes, thereby reducing the potential to translate findings from animal studies into the human setting. In human studies, a major challenge includes a high inter‐individual variability in microbiome composition and metabolite landscape, stemming from a multitude of environmental, immune, and genomic variations such as diet, ethnicity, and geography.

Regardless of these difficulties and challenges, the insight into the ubiquitous functions of microbially modulated metabolites has opened a new window of opportunity toward improving human health. In contrast to attempts to define a “core microbiome” by taxonomic similarity (Neu et al, 2021), the shared metabolite landscape of microbes and their host may allow a better functional classification of microbiome contributions in distinct clinical contexts (Beresford‐Jones et al, 2022). Metabolites derived from or modified by commensals may play central roles in driving “personalized” interventions. Within the Personalized Nutrition Project, microbial features were harnessed to develop a prediction platform of glycemic responses (Zeevi et al, 2015). This outperformed common dietary approaches in an intervention targeting the postprandial glucose levels in healthy and (pre‐)diabetic subjects (Zeevi et al, 2015; Ben‐Yacov et al, 2021; Rein et al, 2022). A diet leading to the blossom of strains with the capacity of carbohydrate fermentation (Zeevi et al, 2015) as well as propionate production (Rein et al, 2022) was associated with metabolic improvements, corresponding to previous reports (Qin et al, 2012; Louis & Flint, 2017). However, a dietary pattern promoting strains from the Alistipes phylum and Bacteriodetes genus was found to have detrimental metabolic effects, contradicting prior studies which associated these microbial signatures with leanness (Turnbaugh et al, 2006, 2009). This underlines that personalized approaches focused on the interplay of diet and microbiome have the potential to guide future strategies in the prevention and treatment of metabolic disorders. Moreover, probiotic supplementation steadily increases in popularity, despite unequivocal results from studies with methodological quality issues, evidence of colonization resistance, and a lack of assessment of long‐term adverse outcomes (Suez et al, 2019). Compounds secreted by probiotics play an important role in inhibiting the reconstitution of the gut microbiome after antibiotic therapy in susceptible subjects (Suez et al, 2018). The inter‐individual variability in effects of probiotics adds a perspective of a personalized approach in refining their use, which is currently based on inconclusive evidence (McFarland, 2014). Eventually, postbiotic interventions can be tailored to balance disturbances in the metabolic function of the resident microbiome. A selective supplementation of metabolites, as demonstrated in preclinical models of weight gain after dieting and smoking cessation (Thaiss et al, 2016a; Fluhr et al, 2021), is expected to be increasingly researched also in the human setting, with an outlook to be incorporated into the precision medicine toolbox.

Author contributions

Eran Elinav: Conceptualization; supervision; funding acquisition; writing – original draft; writing – review and editing. Igor Spivak: Conceptualization; data curation; writing – original draft; writing – review and editing. Leviel Fluhr: Conceptualization; data curation; writing – original draft; writing – review and editing.

Disclosure and competing interests statement

EE is a scientific founder of DayTwo and BiomX, and a paid consultant to Hello Inside and Aposense. The remaining authors declare no competing interests.

Acknowledgments

The authors thank the members of the Elinav lab, Weizmann Institute of Science, and members of the DKFZ microbiome and cancer division for insightful discussions. IS is supported by the Deutsche Forschungsgemeinschaft (SP 1902/1‐1). EE is supported by the Leona M. and Harry B. Helmsley Charitable Trust; Adelis Foundation; Ben B. and Joyce E. Eisenberg Foundation; Estate of Bernard Bishin for the WIS‐Clalit Program; Jeanne and Joseph Nissim Center for Life Sciences Research; Miel de Botton; The Vera Rosenberg Schwartz Research Fellow Chair; Swiss Society Institute for Cancer Prevention Research; Belle S. and Irving E. Meller Center for the Biology of Aging; Sagol Institute for Longevity Research; Sagol Weizmann‐MIT Bridge Program; Norman E Alexander Family M Foundation Coronavirus Research Fund; Mike and Valeria Rosenbloom Foundation; Daniel Morris Trust; Isidore and Penny Myers Foundation; Vainboim Family; and by grants funded by the European Research Council; Israel Science Foundation; Israel Ministry of Science and Technology; Israel Ministry of Health; The German‐Israeli Helmholtz International Research School: Cancer‐TRAX (HIRS‐0003); Helmholtz Association's Initiative and Networking Fund; Minerva Foundation; Garvan Institute; European Crohn's and Colitis Organization; Deutsch‐Israelische Projektkooperation; IDSA Foundation; WIS‐MIT grant; Emulate; Charlie Teo Foundation; Mark Foundation for Cancer Research, and Welcome Trust. EE is the incumbent of the Sir Marc and Lady Tania Feldmann Professorial Chair of immunology; a senior fellow, Canadian Institute of Advanced Research (CIFAR); and an international scholar, The Bill & Melinda Gates Foundation and Howard Hughes Medical Institute (HHMI). Open Access funding enabled and organized by Projekt DEAL.

EMBO reports (2022) 23: e55664

Correction added on 22 March 2023, after first online publication: The copyright line was changed.

References

  1. Agostini L, Down PF, Murison J, Wrong OM (1972) Faecal ammonia and pH during lactulose administration in man: comparison with other cathartics. Gut 13: 859–866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aguilar‐Toalá JE, Garcia‐Varela R, Garcia HS, Mata‐Haro V, González‐Córdova AF, Vallejo‐Cordoba B, Hernández‐Mendoza A (2018) Postbiotics: an evolving term within the functional foods field. Trends Food Sci Technol 75: 105–114 [Google Scholar]
  3. Aguilar‐Toalá JE, Arioli S, Behare P, Belzer C, Berni Canani R, Chatel J‐M, D'Auria E, de Freitas MQ, Elinav E, Esmerino EA et al (2021) Postbiotics – when simplification fails to clarify. Nat Rev Gastroenterol Hepatol 18: 825–826 [DOI] [PubMed] [Google Scholar]
  4. Agus A, Planchais J, Sokol H (2018) Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23: 716–724 [DOI] [PubMed] [Google Scholar]
  5. Agus A, Clément K, Sokol H (2021) Gut microbiota‐derived metabolites as central regulators in metabolic disorders. Gut 70: 1174–1182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Alberti KGMM, Zimmet P, Shaw J (2006) Metabolic syndrome–a new world‐wide definition. A Consensus Statement from the International Diabetes Federation. Diabet Med 23: 469–480 [DOI] [PubMed] [Google Scholar]
  7. Aldunate M, Tyssen D, Johnson A, Zakir T, Sonza S, Moench T, Cone R, Tachedjian G (2013) Vaginal concentrations of lactic acid potently inactivate HIV. J Antimicrob Chemother 68: 2015–2025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Alexander M, Turnbaugh PJ (2020) Deconstructing mechanisms of diet‐microbiome‐immune interactions. Immunity 53: 264–276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Al‐Lahham SH, Roelofsen H, Priebe M, Weening D, Dijkstra M, Hoek A, Rezaee F, Venema K, Vonk RJ (2010) Regulation of adipokine production in human adipose tissue by propionic acid. Eur J Clin Invest 40: 401–407 [DOI] [PubMed] [Google Scholar]
  10. Al‐Lahham S, Roelofsen H, Rezaee F, Weening D, Hoek A, Vonk R, Venema K (2012) Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest 42: 357–364 [DOI] [PubMed] [Google Scholar]
  11. Andonegui G, Kerfoot SM, McNagny K, Ebbert KVJ, Patel KD, Kubes P (2005) Platelets express functional Toll‐like receptor‐4. Blood 106: 2417–2423 [DOI] [PubMed] [Google Scholar]
  12. Apostolopoulou M, Gordillo R, Koliaki C, Gancheva S, Jelenik T, De Filippo E, Herder C, Markgraf D, Jankowiak F, Esposito I et al (2018) Specific hepatic sphingolipids relate to insulin resistance, oxidative stress, and inflammation in nonalcoholic steatohepatitis. Diabetes Care 41: 1235–1243 [DOI] [PubMed] [Google Scholar]
  13. Araújo JR, Tazi A, Burlen‐Defranoux O, Vichier‐Guerre S, Nigro G, Licandro H, Demignot S, Sansonetti PJ (2020) Fermentation products of commensal bacteria alter enterocyte lipid metabolism. Cell Host Microbe 27: 358–375 [DOI] [PubMed] [Google Scholar]
  14. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ et al (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T‐cell generation. Nature 504: 451–455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Aslam R, Speck ER, Kim M, Crow AR, Bang KWA, Nestel FP, Ni H, Lazarus AH, Freedman J, Semple JW (2006) Platelet Toll‐like receptor expression modulates lipopolysaccharide‐induced thrombocytopenia and tumor necrosis factor‐alpha production in vivo . Blood 107: 637–641 [DOI] [PubMed] [Google Scholar]
  16. Bachem A, Makhlouf C, Binger KJ, de Souza DP, Tull D, Hochheiser K, Whitney PG, Fernandez‐Ruiz D, Dähling S, Kastenmüller W et al (2019) Microbiota‐derived short‐chain fatty acids promote the memory potential of antigen‐activated CD8+ T cells. Immunity 51: 285–297 [DOI] [PubMed] [Google Scholar]
  17. Bajaj JS (2019) Alcohol, liver disease and the gut microbiota. Nat Rev Gastroenterol Hepatol 16: 235–246 [DOI] [PubMed] [Google Scholar]
  18. Ben‐Yacov O, Godneva A, Rein M, Shilo S, Kolobkov D, Koren N, Cohen Dolev N, Travinsky Shmul T, Wolf BC, Kosower N et al (2021) Personalized postprandial glucose response‐targeting diet versus mediterranean diet for glycemic control in prediabetes. Diabetes Care 44: 1980–1991 [DOI] [PubMed] [Google Scholar]
  19. Beresford‐Jones BS, Forster SC, Stares MD, Notley G, Viciani E, Browne HP, Boehmler DJ, Soderholm AT, Kumar N, Vervier K et al (2022) The mouse gastrointestinal bacteria catalogue enables translation between the mouse and human gut microbiotas via functional mapping. Cell Host Microbe 30: 124–138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bergman EN (1990) Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol Rev 70: 567–590 [DOI] [PubMed] [Google Scholar]
  21. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D‐J, Bakker BM (2013a) The role of short‐chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 54: 2325–2340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. den Besten G, Lange K, Havinga R, van Dijk TH, Gerding A, van Eunen K, Müller M, Groen AK, Hooiveld GJ, Bakker BM et al (2013b) Gut‐derived short‐chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am J Physiol Gastrointest Liver Physiol 305: G900‐10 [DOI] [PubMed] [Google Scholar]
  23. den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud D‐J et al (2015) Short‐chain fatty acids protect against high‐fat diet‐induced obesity via a PPARγ‐dependent switch from lipogenesis to fat oxidation. Diabetes 64: 2398–2408 [DOI] [PubMed] [Google Scholar]
  24. Beyaz S, Chung C, Mou H, Bauer‐Rowe KE, Xifaras ME, Ergin I, Dohnalova L, Biton M, Shekhar K, Eskiocak O et al (2021) Dietary suppression of MHC class II expression in intestinal epithelial cells enhances intestinal tumorigenesis. Cell Stem Cell 28: 1922–1935 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR, Till L, Grover M, Linden DR, Akiba Y, Kandimalla KK, Zachos NC et al (2018) Gut microbiota‐produced tryptamine activates an epithelial G‐protein‐coupled receptor to increase colonic secretion. Cell Host Microbe 23: 775–785 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Biesalski HK (2016) Nutrition meets the microbiome: micronutrients and the microbiota. Ann N Y Acad Sci 1372: 53–64 [DOI] [PubMed] [Google Scholar]
  27. Birchenough GMH, Nyström EEL, Johansson MEV, Hansson GC (2016) A sentinel goblet cell guards the colonic crypt by triggering Nlrp6‐dependent Muc2 secretion. Science 352: 1535–1542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Björntorp P, Hood B (1966) Studies on adipose tissue from obese patients with or without diabetes mellitus. I. Release of glycerol and free fatty acids. Acta Med Scand 179: 221–227 [DOI] [PubMed] [Google Scholar]
  29. Blacher E, Bashiardes S, Shapiro H, Rothschild D, Mor U, Dori‐Bachash M, Kleimeyer C, Moresi C, Harnik Y, Zur M et al (2019) Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 572: 474–480 [DOI] [PubMed] [Google Scholar]
  30. Borghi M, Puccetti M, Pariano M, Renga G, Stincardini C, Ricci M, Giovagnoli S, Costantini C, Romani L (2020) Tryptophan as a central hub for host/microbial symbiosis. Int J Tryptophan Res 13: 1178646920919755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Broeders EPM, Nascimento EBM, Havekes B, Brans B, Roumans KHM, Tailleux A, Schaart G, Kouach M, Charton J, Deprez B et al (2015) The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Cell Metab 22: 418–426 [DOI] [PubMed] [Google Scholar]
  32. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, No D, Liu H, Kinnebrew M, Viale A et al (2015) Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile . Nature 517: 205–208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Buffington SA, Dooling SW, Sgritta M, Noecker C, Murillo OD, Felice DF, Turnbaugh PJ, Costa‐Mattioli M (2021) Dissecting the contribution of host genetics and the microbiome in complex behaviors. Cell 184: 1740–1756 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bugaut M (1987) Occurrence, absorption and metabolism of short chain fatty acids in the digestive tract of mammals. Comp Biochem Physiol B 86: 439–472 [DOI] [PubMed] [Google Scholar]
  35. Burrows MP, Volchkov P, Kobayashi KS, Chervonsky AV (2015) Microbiota regulates type 1 diabetes through Toll‐like receptors. Proc Natl Acad Sci U S A 112: 9973–9977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Caesar R, Tremaroli V, Kovatcheva‐Datchary P, Cani PD, Bäckhed F (2015) Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling. Cell Metab 22: 658–668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Campbell C, McKenney PT, Konstantinovsky D, Isaeva OI, Schizas M, Verter J, Mai C, Jin W‐B, Guo C‐J, Violante S et al (2020) Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 581: 475–479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Cani PD (2018) Human gut microbiome: hopes, threats and promises. Gut 67: 1716–1725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Cani PD, Van Hul M, Lefort C, Depommier C, Rastelli M, Everard A (2019) Microbial regulation of organismal energy homeostasis. Nat Metab 1: 34–46 [DOI] [PubMed] [Google Scholar]
  40. Carpino G, Del Ben M, Pastori D, Carnevale R, Baratta F, Overi D, Francis H, Cardinale V, Onori P, Safarikia S et al (2020) Increased liver localization of lipopolysaccharides in human and experimental NAFLD. Hepatology 72: 470–485 [DOI] [PubMed] [Google Scholar]
  41. Carrasco‐Pozo C, Morales P, Gotteland M (2013) Polyphenols protect the epithelial barrier function of Caco‐2 cells exposed to indomethacin through the modulation of occludin and zonula occludens‐1 expression. J Agric Food Chem 61: 5291–5297 [DOI] [PubMed] [Google Scholar]
  42. Cervantes‐Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S et al (2017) Lactobacillus reuteri induces gut intraepithelial CD4+CD8αα+ T cells. Science 357: 806–810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Chaluvadi S, Hotchkiss AT, Yam KL (2016) Chapter 36 – Gut microbiota: impact of probiotics, prebiotics, synbiotics, pharmabiotics, and postbiotics on human health. In Probiotics, Prebiotics, and Synbiotics, Watson RR, Preedy VR (eds), pp 515–523. London: Academic Press; [Google Scholar]
  44. Chambers ES, Byrne CS, Rugyendo A, Morrison DJ, Preston T, Tedford C, Bell JD, Thomas L, Akbar AN, Riddell NE et al (2019) The effects of dietary supplementation with inulin and inulin‐propionate ester on hepatic steatosis in adults with non‐alcoholic fatty liver disease. Diabetes Obes Metab 21: 372–376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Chen R, Xu Y, Wu P, Zhou H, Lasanajak Y, Fang Y, Tang L, Ye L, Li X, Cai Z et al (2019a) Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol Res 148: 104403 [DOI] [PubMed] [Google Scholar]
  46. Chen S, Henderson A, Petriello MC, Romano KA, Gearing M, Miao J, Schell M, Sandoval‐Espinola WJ, Tao J, Sha B et al (2019b) Trimethylamine N‐oxide binds and activates PERK to promote metabolic dysfunction. Cell Metab 30: 1141–1151 [DOI] [PubMed] [Google Scholar]
  47. Chen X, Li P, Liu M, Zheng H, He Y, Chen M‐X, Tang W, Yue X, Huang Y, Zhuang L et al (2020) Gut dysbiosis induces the development of pre‐eclampsia through bacterial translocation. Gut 69: 513–522 [DOI] [PubMed] [Google Scholar]
  48. Chen Y‐M, Liu Y, Zhou R‐F, Chen X‐L, Wang C, Tan X‐Y, Wang L‐J, Zheng R‐D, Zhang H‐W, Ling W‐H et al (2016) Associations of gut‐flora‐dependent metabolite trimethylamine‐N‐oxide, betaine and choline with non‐alcoholic fatty liver disease in adults. Sci Rep 6: 19076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Chhibber‐Goel J, Gaur A, Singhal V, Parakh N, Bhargava B, Sharma A (2016) The complex metabolism of trimethylamine in humans: endogenous and exogenous sources. Expert Rev Mol Med 18: e8 [DOI] [PubMed] [Google Scholar]
  50. Chun E, Lavoie S, Fonseca‐Pereira D, Bae S, Michaud M, Hoveyda HR, Fraser GL, Gallini Comeau CA, Glickman JN, Fuller MH et al (2019) Metabolite‐sensing receptor Ffar2 regulates colonic group 3 innate lymphoid cells and gut immunity. Immunity 51: 871–884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR et al (2012) Gut immune maturation depends on colonization with a host‐specific microbiota. Cell 149: 1578–1593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Conti C, Malacrino C & Mastromarino P (2009) Inhibition of herpes simplex virus type 2 by vaginal lactobacilli. J Physiol Pharmacol 60 Suppl 6: 19–26 [PubMed] [Google Scholar]
  53. Cook KL, Chappell MC (2021) Gut dysbiosis and hypertension: is it cause or effect? J Hypertens 39: 1768–1770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Costello EK, Stagaman K, Dethlefsen L, Bohannan BJM, Relman DA (2012) The application of ecological theory toward an understanding of the human microbiome. Science 336: 1255–1262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Coudray C, Bellanger J, Castiglia‐Delavaud C, Rémésy C, Vermorel M, Rayssignuier Y (1997) Effect of soluble or partly soluble dietary fibres supplementation on absorption and balance of calcium, magnesium, iron and zinc in healthy young men. Eur J Clin Nutr 51: 375–380 [DOI] [PubMed] [Google Scholar]
  56. Cullin N, Azevedo Antunes C, Straussman R, Stein‐Thoeringer CK, Elinav E (2021) Microbiome and cancer. Cancer Cell 39: 1317–1341 [DOI] [PubMed] [Google Scholar]
  57. Curat CA, Wegner V, Sengenès C, Miranville A, Tonus C, Busse R, Bouloumié A (2006) Macrophages in human visceral adipose tissue: increased accumulation in obesity and a source of resistin and visfatin. Diabetologia 49: 744–747 [DOI] [PubMed] [Google Scholar]
  58. Dasgupta S, Erturk‐Hasdemir D, Ochoa‐Reparaz J, Reinecker H‐C, Kasper DL (2014) Plasmacytoid dendritic cells mediate anti‐inflammatory responses to a gut commensal molecule via both innate and adaptive mechanisms. Cell Host Microbe 15: 413–423 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. De Luca A, Zelante T, D'Angelo C, Zagarella S, Fallarino F, Spreca A, Iannitti RG, Bonifazi P, Renauld J‐C, Bistoni F et al (2010) IL‐22 defines a novel immune pathway of antifungal resistance. Mucosal Immunol 3: 361–373 [DOI] [PubMed] [Google Scholar]
  60. De Vadder F, Kovatcheva‐Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, Bäckhed F, Mithieux G (2014) Microbiota‐generated metabolites promote metabolic benefits via gut‐brain neural circuits. Cell 156: 84–96 [DOI] [PubMed] [Google Scholar]
  61. De Vadder F, Kovatcheva‐Datchary P, Zitoun C, Duchampt A, Bäckhed F, Mithieux G (2016) Microbiota‐produced succinate improves glucose homeostasis via intestinal gluconeogenesis. Cell Metab 24: 151–157 [DOI] [PubMed] [Google Scholar]
  62. De Vadder F, Plessier F, Gautier‐Stein A, Mithieux G (2015) Vasoactive intestinal peptide is a local mediator in a gut‐brain neural axis activating intestinal gluconeogenesis. Neurogastroenterol Motil 27: 443–448 [DOI] [PubMed] [Google Scholar]
  63. Devlin AS, Marcobal A, Dodd D, Nayfach S, Plummer N, Meyer T, Pollard KS, Sonnenburg JL, Fischbach MA (2016) Modulation of a circulating uremic solute via rational genetic manipulation of the gut microbiota. Cell Host Microbe 20: 709–715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Dey N, Wagner VE, Blanton LV, Cheng J, Fontana L, Haque R, Ahmed T, Gordon JI (2015) Regulators of gut motility revealed by a gnotobiotic model of diet‐microbiome interactions related to travel. Cell 163: 95–107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Duan W, Ladenheim B, Cutler RG, Kruman II, Cadet JL, Mattson MP (2002) Dietary folate deficiency and elevated homocysteine levels endanger dopaminergic neurons in models of Parkinson's disease. J Neurochem 80: 101–110 [DOI] [PubMed] [Google Scholar]
  66. Ducastel S, Touche V, Trabelsi M‐S, Boulinguiez A, Butruille L, Nawrot M, Peschard S, Chávez‐Talavera O, Dorchies E, Vallez E et al (2020) The nuclear receptor FXR inhibits glucagon‐like peptide‐1 secretion in response to microbiota‐derived short‐chain fatty acids. Sci Rep 10: 174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Dumas M‐E, Rothwell AR, Hoyles L, Aranias T, Chilloux J, Calderari S, Noll EM, Péan N, Boulangé CL, Blancher C et al (2017) Microbial‐host co‐metabolites are prodromal markers predicting phenotypic heterogeneity in behavior, obesity, and impaired glucose tolerance. Cell Rep 20: 136–148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Eussen SJPM, Ueland PM, Vollset SE, Nygård O, Midttun Ø, Sulo G, Ulvik A, Meyer K, Pedersen ER, Tell GS (2015) Kynurenines as predictors of acute coronary events in the Hordaland Health Study. Int J Cardiol 189: 18–24 [DOI] [PubMed] [Google Scholar]
  69. Fachi JL, Sécca C, Rodrigues PB, Mato FCP, Di Luccia B, Felipe JS, Pral LP, Rungue M, Rocha VM, Sato FT et al (2020) Acetate coordinates neutrophil and ILC3 responses against C. difficile through FFAR2. J Exp Med 217: jem.20190489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Fluhr L, Mor U, Kolodziejczyk AA, Dori‐Bachash M, Leshem A, Itav S, Cohen Y, Suez J, Zmora N, Moresi C et al (2021) Gut microbiota modulates weight gain in mice after discontinued smoke exposure. Nature 600: 713–719 [DOI] [PubMed] [Google Scholar]
  71. Fukata M, Chen A, Klepper A, Krishnareddy S, Vamadevan AS, Thomas LS, Xu R, Inoue H, Arditi M, Dannenberg AJ et al (2006) Cox‐2 is regulated by Toll‐like receptor‐4 (TLR4) signaling: role in proliferation and apoptosis in the intestine. Gastroenterology 131: 862–877 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Fukata M, Chen A, Vamadevan AS, Cohen J, Breglio K, Krishnareddy S, Hsu D, Xu R, Harpaz N, Dannenberg AJ et al (2007) Toll‐like receptor‐4 promotes the development of colitis‐associated colorectal tumors. Gastroenterology 133: 1869–1881 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Gantner BN, Simmons RM, Canavera SJ, Akira S, Underhill DM (2003) Collaborative induction of inflammatory responses by dectin‐1 and Toll‐like receptor 2. J Exp Med 197: 1107–1117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Gao F, Lv Y‐W, Long J, Chen J‐M, He J‐M, Ruan X‐Z, Zhu H‐B (2019) Butyrate improves the metabolic disorder and gut microbiome dysbiosis in mice induced by a high‐fat diet. Front Pharmacol 10: 1040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, Cefalu WT, Ye J (2009) Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 58: 1509–1517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT et al (2014) Human genetics shape the gut microbiome. Cell 159: 789–799 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Grosheva I, Zheng D, Levy M, Polansky O, Lichtenstein A, Golani O, Dori‐Bachash M, Moresi C, Shapiro H, Del Mare‐Roumani S et al (2020) High‐throughput screen identifies host and microbiota regulators of intestinal barrier function. Gastroenterology 159: 1807–1823 [DOI] [PubMed] [Google Scholar]
  78. Guzior DV, Quinn RA (2021) Review: microbial transformations of human bile acids. Microbiome 9: 140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Hall JA, Bouladoux N, Sun CM, Wohlfert EA, Blank RB, Zhu Q, Grigg ME, Berzofsky JA, Belkaid Y (2008) Commensal DNA limits regulatory T cell conversion and is a natural adjuvant of intestinal immune responses. Immunity 29: 637–649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Hang S, Paik D, Yao L, Kim E, Trinath J, Lu J, Ha S, Nelson BN, Kelly SP, Wu L et al (2019) Bile acid metabolites control TH17 and Treg cell differentiation. Nature 576: 143–148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Hao F, Tian M, Zhang X, Jin X, Jiang Y, Sun X, Wang Y, Peng P, Liu J, Xia C et al (2021) Butyrate enhances CPT1A activity to promote fatty acid oxidation and iTreg differentiation. Proc Natl Acad Sci U S A 118: e2014681118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Hayashi A, Nagao‐Kitamoto H, Kitamoto S, Kim CH, Kamada N (2021a) The butyrate‐producing bacterium clostridium butyricum suppresses Clostridioides difficile infection via neutrophil‐ and antimicrobial cytokine‐dependent but GPR43/109a‐independent mechanisms. J Immunol 206: 1576–1585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Hayashi T, Yamashita T, Takahashi T, Tabata T, Watanabe H, Gotoh Y, Shinohara M, Kami K, Tanaka H, Matsumoto K et al (2021b) Uncovering the role of gut microbiota in amino acid metabolic disturbances in heart failure through metagenomic analysis. Front Cardiovasc Med 8: 789325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. van der Hee B, Wells JM (2021) Microbial regulation of host physiology by short‐chain fatty acids. Trends Microbiol 29: 700–712 [DOI] [PubMed] [Google Scholar]
  85. Hehemann J‐H, Correc G, Barbeyron T, Helbert W, Czjzek M, Michel G (2010) Transfer of carbohydrate‐active enzymes from marine bacteria to Japanese gut microbiota. Nature 464: 908–912 [DOI] [PubMed] [Google Scholar]
  86. Henao‐Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ et al (2012) Inflammasome‐mediated dysbiosis regulates progression of NAFLD and obesity. Nature 482: 179–185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Hendrikx T, Duan Y, Wang Y, Oh J‐H, Alexander LM, Huang W, Stärkel P, Ho SB, Gao B, Fiehn O et al (2019) Bacteria engineered to produce IL‐22 in intestine induce expression of REG3G to reduce ethanol‐induced liver disease in mice. Gut 68: 1504–1515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, Sovath S, Shamel L, Hartung T, Zähringer U et al (2005) CD36 is a sensor of diacylglycerides. Nature 433: 523–527 [DOI] [PubMed] [Google Scholar]
  89. Hsu D, Fukata M, Hernandez YG, Sotolongo JP, Goo T, Maki J, Hayes LA, Ungaro RC, Chen A, Breglio KJ et al (2010) Toll‐like receptor 4 differentially regulates epidermal growth factor‐related growth factors in response to intestinal mucosal injury. Lab Invest 90: 1295–1305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Induri SNR, Kansara P, Thomas SC, Xu F, Saxena D, Li X (2022) The gut microbiome, metformin, and aging. Annu Rev Pharmacol Toxicol 62: 85–108 [DOI] [PubMed] [Google Scholar]
  91. Iqbal J, Hussain MM (2009) Intestinal lipid absorption. Am J Physiol Endocrinol Metab 296: E1183–E1194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Isobe J, Maeda S, Obata Y, Iizuka K, Nakamura Y, Fujimura Y, Kimizuka T, Hattori K, Kim Y‐G, Morita T et al (2020) Commensal‐bacteria‐derived butyrate promotes the T‐cell‐independent IgA response in the colon. Int Immunol 32: 243–258 [DOI] [PubMed] [Google Scholar]
  93. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV et al (2009) Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139: 485–498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Jamka M, Kokot M, Kaczmarek N, Bermagambetova S, Nowak JK, Walkowiak J (2021) The effect of sodium butyrate enemas compared with placebo on disease activity, endoscopic scores, and histological and inflammatory parameters in inflammatory bowel diseases: a systematic review of randomised controlled trials. Complement Med Res 28: 344–356 [DOI] [PubMed] [Google Scholar]
  95. Jiang C, Xie C, Lv Y, Li J, Krausz KW, Shi J, Brocker CN, Desai D, Amin SG, Bisson WH et al (2015) Intestine‐selective farnesoid X receptor inhibition improves obesity‐related metabolic dysfunction. Nat Commun 6: 10166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Jiang Z, Georgel P, Du X, Shamel L, Sovath S, Mudd S, Huber M, Kalis C, Keck S, Galanos C et al (2005) CD14 is required for MyD88‐independent LPS signaling. Nat Immunol 6: 565–570 [DOI] [PubMed] [Google Scholar]
  97. Jie Z, Xia H, Zhong S‐L, Feng Q, Li S, Liang S, Zhong H, Liu Z, Gao Y, Zhao H et al (2017) The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun 8: 845 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Johnson EL, Heaver SL, Waters JL, Kim BI, Bretin A, Goodman AL, Gewirtz AT, Worgall TS, Ley RE (2020) Sphingolipids produced by gut bacteria enter host metabolic pathways impacting ceramide levels. Nat Commun 11: 2471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Keane C, Tilley D, Cunningham A, Smolenski A, Kadioglu A, Cox D, Jenkinson HF, Kerrigan SW (2010) Invasive Streptococcus pneumoniae trigger platelet activation via Toll‐like receptor 2. J Thromb Haemost 8: 2757–2765 [DOI] [PubMed] [Google Scholar]
  100. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH (2013) Short‐chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 145: 396–406 [DOI] [PubMed] [Google Scholar]
  101. Kinnebrew MA, Buffie CG, Diehl GE, Zenewicz LA, Leiner I, Hohl TM, Flavell RA, Littman DR, Pamer EG (2012) Interleukin 23 production by intestinal CD103(+)CD11b(+) dendritic cells in response to bacterial flagellin enhances mucosal innate immune defense. Immunity 36: 276–287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L et al (2013) Intestinal microbiota metabolism of L‐carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19: 576–585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Koh A, Bäckhed F (2020) From association to causality: the role of the gut microbiota and its functional products on host metabolism. Mol Cell 78: 584–596 [DOI] [PubMed] [Google Scholar]
  104. Kolodziejczyk AA, Federici S, Zmora N, Mohapatra G, Dori‐Bachash M, Hornstein S, Leshem A, Reuveni D, Zigmond E, Tobar A et al (2020) Acute liver failure is regulated by MYC‐ and microbiome‐dependent programs. Nat Med 26: 1899–1911 [DOI] [PubMed] [Google Scholar]
  105. Kolodziejczyk AA, Zheng D, Elinav E (2019) Diet–microbiota interactions and personalized nutrition. Nat Rev Microbiol 17: 742–753 [DOI] [PubMed] [Google Scholar]
  106. Korpe PS, Petri WA Jr (2012) Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med 18: 328–336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Krautkramer KA, Fan J, Bäckhed F (2021) Gut microbial metabolites as multi‐kingdom intermediates. Nat Rev Microbiol 19: 77–94 [DOI] [PubMed] [Google Scholar]
  108. Lamas B, Richard ML, Leducq V, Pham H‐P, Michel M‐L, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM et al (2016) CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 22: 598–605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Laurans L, Venteclef N, Haddad Y, Chajadine M, Alzaid F, Metghalchi S, Sovran B, Denis RGP, Dairou J, Cardellini M et al (2018) Genetic deficiency of indoleamine 2,3‐dioxygenase promotes gut microbiota‐mediated metabolic health. Nat Med 24: 1113–1120 [DOI] [PubMed] [Google Scholar]
  110. Lee Y‐S, Kim T‐Y, Kim Y, Lee S‐H, Kim S, Kang SW, Yang J‐Y, Baek I‐J, Sung YH, Park Y‐Y et al (2018) Microbiota‐derived lactate accelerates intestinal stem‐cell‐mediated epithelial development. Cell Host Microbe 24: 833–846 [DOI] [PubMed] [Google Scholar]
  111. León‐Mimila P, Villamil‐Ramírez H, Li XS, Shih DM, Hui ST, Ocampo‐Medina E, López‐Contreras B, Morán‐Ramos S, Olivares‐Arevalo M, Grandini‐Rosales P et al (2021) Trimethylamine N‐oxide levels are associated with NASH in obese subjects with type 2 diabetes. Diabetes Metab 47: 101183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Lev‐Sagie A, Goldman‐Wohl D, Cohen Y, Dori‐Bachash M, Leshem A, Mor U, Strahilevitz J, Moses AE, Shapiro H, Yagel S et al (2019) Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat Med 25: 1500–1504 [DOI] [PubMed] [Google Scholar]
  113. Levy M, Thaiss CA, Zeevi D, Dohnalová L, Zilberman‐Schapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y et al (2015) Microbiota‐modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell 163: 1428–1443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Li B, Li L, Li M, Lam SM, Wang G, Wu Y, Zhang H, Niu C, Zhang X, Liu X et al (2019) Microbiota depletion impairs thermogenesis of brown adipose tissue and browning of white adipose tissue. Cell Rep 26: 2720–2737 [DOI] [PubMed] [Google Scholar]
  115. Li F, Jiang C, Krausz KW, Li Y, Albert I, Hao H, Fabre KM, Mitchell JB, Patterson AD, Gonzalez FJ (2013) Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat Commun 4: 2384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Li G, Xie C, Lu S, Nichols RG, Tian Y, Li L, Patel D, Ma Y, Brocker CN, Yan T et al (2017) Intermittent fasting promotes white adipose browning and decreases obesity by shaping the gut microbiota. Cell Metab 26: 672–685 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Li Z, Yi C‐X, Katiraei S, Kooijman S, Zhou E, Chung CK, Gao Y, van den Heuvel JK, Meijer OC, Berbée JFP et al (2018) Butyrate reduces appetite and activates brown adipose tissue via the gut‐brain neural circuit. Gut 67: 1269–1279 [DOI] [PubMed] [Google Scholar]
  118. Liang Z, Dong Z, Guo M, Shen Z, Yin D, Hu S, Hai X (2019) Trimethylamine N‐oxide as a risk marker for ischemic stroke in patients with atrial fibrillation. J Biochem Mol Toxicol 33: e22246 [DOI] [PubMed] [Google Scholar]
  119. Lobo AR, Gaievski EHS, De Carli E, Alvares EP, Colli C (2014) Fructo‐oligosaccharides and iron bioavailability in anaemic rats: the effects on iron species distribution, ferroportin‐1 expression, crypt bifurcation and crypt cell proliferation in the caecum. Br J Nutr 112: 1286–1295 [DOI] [PubMed] [Google Scholar]
  120. Louis P, Flint HJ (2017) Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol 19: 29–41 [DOI] [PubMed] [Google Scholar]
  121. Lundberg R, Toft MF, Metzdorff SB, Hansen CHF, Licht TR, Bahl MI, Hansen AK (2020) Human microbiota‐transplanted C57BL/6 mice and offspring display reduced establishment of key bacteria and reduced immune stimulation compared to mouse microbiota‐transplantation. Sci Rep 10: 7805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Luu M, Weigand K, Wedi F, Breidenbend C, Leister H, Pautz S, Adhikary T, Visekruna A (2018) Regulation of the effector function of CD8+ T cells by gut microbiota‐derived metabolite butyrate. Sci Rep 8: 14430 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Macho Fernandez E, Valenti V, Rockel C, Hermann C, Pot B, Boneca IG, Grangette C (2011) Anti‐inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2‐mediated recognition of a specific peptidoglycan‐derived muropeptide. Gut 60: 1050–1059 [DOI] [PubMed] [Google Scholar]
  124. Macia L, Tan J, Vieira AT, Leach K, Stanley D, Luong S, Maruya M, Ian McKenzie C, Hijikata A, Wong C et al (2015) Metabolite‐sensing receptors GPR43 and GPR109A facilitate dietary fibre‐induced gut homeostasis through regulation of the inflammasome. Nat Commun 6: 6734 [DOI] [PubMed] [Google Scholar]
  125. Mandaliya DK, Patel S, Seshadri S (2021) The combinatorial effect of acetate and propionate on high‐fat diet induced diabetic inflammation or metaflammation and T cell polarization. Inflammation 44: 68–79 [DOI] [PubMed] [Google Scholar]
  126. Marques FZ, Nelson E, Chu P‐Y, Horlock D, Fiedler A, Ziemann M, Tan JK, Kuruppu S, Rajapakse NW, El‐Osta A et al (2017) High‐fiber diet and acetate supplementation change the gut microbiota and prevent the development of hypertension and heart failure in hypertensive mice. Circulation 135: 964–977 [DOI] [PubMed] [Google Scholar]
  127. Martinez‐Guryn K, Hubert N, Frazier K, Urlass S, Musch MW, Ojeda P, Pierre JF, Miyoshi J, Sontag TJ, Cham CM et al (2018) Small intestine microbiota regulate host digestive and absorptive adaptive responses to dietary lipids. Cell Host Microbe 23: 458–469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Mazagova M, Wang L, Anfora AT, Wissmueller M, Lesley SA, Miyamoto Y, Eckmann L, Dhungana S, Pathmasiri W, Sumner S et al (2015) Commensal microbiota is hepatoprotective and prevents liver fibrosis in mice. FASEB J 29: 1043–1055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL (2005) An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122: 107–118 [DOI] [PubMed] [Google Scholar]
  130. Mazmanian SK, Round JL, Kasper DL (2008) A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453: 620–625 [DOI] [PubMed] [Google Scholar]
  131. McFall‐Ngai M (2007) Adaptive immunity: care for the community. Nature 445: 153 [DOI] [PubMed] [Google Scholar]
  132. McFarland LV (2014) Use of probiotics to correct dysbiosis of normal microbiota following disease or disruptive events: a systematic review. BMJ Open 4: e005047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Miani M, Le Naour J, Waeckel‐Enée E, Verma SC, Straube M, Emond P, Ryffel B, van Endert P, Sokol H, Diana J (2018) Gut microbiota‐stimulated innate lymphoid cells support β‐defensin 14 expression in pancreatic endocrine cells, preventing autoimmune diabetes. Cell Metab 28: 557–572 [DOI] [PubMed] [Google Scholar]
  134. Mocanu V, Zhang Z, Deehan EC, Kao DH, Hotte N, Karmali S, Birch DW, Samarasinghe KK, Walter J, Madsen KL (2021) Fecal microbial transplantation and fiber supplementation in patients with severe obesity and metabolic syndrome: a randomized double‐blind, placebo‐controlled phase 2 trial. Nat Med 27: 1272–1279 [DOI] [PubMed] [Google Scholar]
  135. Muller PA, Schneeberger M, Matheis F, Wang P, Kerner Z, Ilanges A, Pellegrino K, Del Mármol J, Castro TBR, Furuichi M et al (2020) Microbiota modulate sympathetic neurons via a gut‐brain circuit. Nature 583: 441–446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Needham BD, Kaddurah‐Daouk R, Mazmanian SK (2020) Gut microbial molecules in behavioural and neurodegenerative conditions. Nat Rev Neurosci 21: 717–731 [DOI] [PubMed] [Google Scholar]
  137. Neu AT, Allen EE, Roy K (2021) Defining and quantifying the core microbiome: challenges and prospects. Proc Natl Acad Sci U S A 118: e2104429118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Nie J, Xie L, Zhao B‐X, Li Y, Qiu B, Zhu F, Li G‐F, He M, Wang Y, Wang B et al (2018) Serum trimethylamine N‐oxide concentration is positively associated with first stroke in hypertensive patients. Stroke 49: 2021–2028 [DOI] [PubMed] [Google Scholar]
  139. Normand S, Delanoye‐Crespin A, Bressenot A, Huot L, Grandjean T, Peyrin‐Biroulet L, Lemoine Y, Hot D, Chamaillard M (2011) Nod‐like receptor pyrin domain‐containing protein 6 (NLRP6) controls epithelial self‐renewal and colorectal carcinogenesis upon injury. Proc Natl Acad Sci U S A 108: 9601–9606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Ogura Y, Lala S, Xin W, Smith E, Dowds TA, Chen FF, Zimmermann E, Tretiakova M, Cho JH, Hart J et al (2003) Expression of NOD2 in Paneth cells: a possible link to Crohn's ileitis. Gut 52: 1591–1597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Oh SF, Praveena T, Song H, Yoo J‐S, Jung D‐J, Erturk‐Hasdemir D, Hwang YS, Lee CC, Le Nours J, Kim H et al (2021) Host immunomodulatory lipids created by symbionts from dietary amino acids. Nature 600: 302–307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Ohira H, Fujioka Y, Katagiri C, Mamoto R, Aoyama‐Ishikawa M, Amako K, Izumi Y, Nishiumi S, Yoshida M, Usami M et al (2013) Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J Atheroscler Thromb 20: 425–442 [DOI] [PubMed] [Google Scholar]
  143. Okada T, Fukuda S, Hase K, Nishiumi S, Izumi Y, Yoshida M, Hagiwara T, Kawashima R, Yamazaki M, Oshio T et al (2013) Microbiota‐derived lactate accelerates colon epithelial cell turnover in starvation‐refed mice. Nat Commun 4: 1654 [DOI] [PubMed] [Google Scholar]
  144. Okla M, Zaher W, Alfayez M, Chung S (2018) Inhibitory effects of toll‐like receptor 4, NLRP3 inflammasome, and interleukin‐1β on white adipocyte browning. Inflammation 41: 626–642 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Paeslack N, Mimmler M, Becker S, Gao Z, Khuu MP, Mann A, Malinarich F, Regen T, Reinhardt C (2022) Microbiota‐derived tryptophan metabolites in vascular inflammation and cardiovascular disease. Amino Acids 10.1007/s00726-022-03161-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Park J, Goergen CJ, HogenEsch H, Kim CH (2016) Chronically elevated levels of short‐chain fatty acids induce T cell‐mediated ureteritis and hydronephrosis. J Immunol 196: 2388–2400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Parséus A, Sommer N, Sommer F, Caesar R, Molinaro A, Ståhlman M, Greiner TU, Perkins R, Bäckhed F (2017) Microbiota‐induced obesity requires farnesoid X receptor. Gut 66: 429–437 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Pathak P, Xie C, Nichols RG, Ferrell JM, Boehme S, Krausz KW, Patterson AD, Gonzalez FJ, Chiang JYL (2018) Intestine farnesoid X receptor agonist and the gut microbiota activate G‐protein bile acid receptor‐1 signaling to improve metabolism. Hepatology 68: 1574–1588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Podolsky DK, Gerken G, Eyking A, Cario E (2009) Colitis‐associated variant of TLR2 causes impaired mucosal repair because of TFF3 deficiency. Gastroenterology 137: 209–220 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Pott J, Hornef M (2012) Innate immune signalling at the intestinal epithelium in homeostasis and disease. EMBO Rep 13: 684–698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, Wang L, Zhang Y, Liang X, Wang L et al (2019) Gut microbiota‐bile acid‐interleukin‐22 axis orchestrates polycystic ovary syndrome. Nat Med 25: 1225–1233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D et al (2012) A metagenome‐wide association study of gut microbiota in type 2 diabetes. Nature 490: 55–60 [DOI] [PubMed] [Google Scholar]
  153. Quigley EMM, Murray JA, Pimentel M (2020) AGA clinical practice update on small intestinal bacterial overgrowth: expert review. Gastroenterology 159: 1526–1532 [DOI] [PubMed] [Google Scholar]
  154. Quinn RA, Melnik AV, Vrbanac A, Fu T, Patras KA, Christy MP, Bodai Z, Belda‐Ferre P, Tripathi A, Chung LK et al (2020) Global chemical effects of the microbiome include new bile‐acid conjugations. Nature 579: 123–129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Rakoff‐Nahoum S, Paglino J, Eslami‐Varzaneh F, Edberg S, Medzhitov R (2004) Recognition of commensal microflora by toll‐like receptors is required for intestinal homeostasis. Cell 118: 229–241 [DOI] [PubMed] [Google Scholar]
  156. Rein M, Ben‐Yacov O, Godneva A, Shilo S, Zmora N, Kolobkov D, Cohen‐Dolev N, Wolf B‐C, Kosower N, Lotan‐Pompan M et al (2022) Effects of personalized diets by prediction of glycemic responses on glycemic control and metabolic health in newly diagnosed T2DM: a randomized dietary intervention pilot trial. BMC Med 20: 56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR et al (2013) Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 341: 1241214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Romano KA, Martinez‐Del Campo A, Kasahara K, Chittim CL, Vivas EI, Amador‐Noguez D, Balskus EP, Rey FE (2017) Metabolic, epigenetic, and transgenerational effects of gut bacterial choline consumption. Cell Host Microbe 22: 279–290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Rosario D, Bidkhori G, Lee S, Bedarf J, Hildebrand F, Le Chatelier E, Uhlen M, Ehrlich SD, Proctor G, Wüllner U et al (2021) Systematic analysis of gut microbiome reveals the role of bacterial folate and homocysteine metabolism in Parkinson's disease. Cell Rep 34: 108807 [DOI] [PubMed] [Google Scholar]
  160. Roshanravan N, Mahdavi R, Alizadeh E, Jafarabadi M, Hedayati M, Ghavami A, Alipour S, Alamdari N, Barati M, Ostadrahimi A (2017) Effect of butyrate and inulin supplementation on glycemic status, lipid profile and glucagon‐like peptide 1 level in patients with type 2 diabetes: a randomized double‐blind, placebo‐controlled trial. Horm Metab Res 49: 886–891 [DOI] [PubMed] [Google Scholar]
  161. Rossi T, Vergara D, Fanini F, Maffia M, Bravaccini S, Pirini F (2020) Microbiota‐derived metabolites in tumor progression and metastasis. Int J Mol Sci 21: 5786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Roth GA, Mensah GA, Johnson CO, Giovanni A, Enrico A, Baddour LM, Barengo NC, Beaton AZ, Benjamin EJ, Benziger CP et al (2020) Global burden of cardiovascular diseases and risk factors, 1990–2019. J Am Coll Cardiol 76: 2982–3021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, Mazmanian SK (2011) The Toll‐like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332: 974–977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Roy S, Yuzefpolskaya M, Nandakumar R, Colombo PC, Demmer RT (2020) Plasma Trimethylamine‐N‐oxide and impaired glucose regulation: Results from The Oral Infections, Glucose Intolerance and Insulin Resistance Study (ORIGINS). PLoS One 15: e0227482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Salminen S, Collado MC, Endo A, Hill C, Lebeer S, Quigley EMM, Sanders ME, Shamir R, Swann JR, Szajewska H et al (2021) The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat Rev Gastroenterol Hepatol 18: 649–667 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Sampson TR, Challis C, Jain N, Moiseyenko A, Ladinsky MS, Shastri GG, Thron T, Needham BD, Horvath I, Debelius JW et al (2020) A gut bacterial amyloid promotes α‐synuclein aggregation and motor impairment in mice. Elife 9: e53111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Samuel BS, Shaito A, Motoike T, Rey FE, Backhed F, Manchester JK, Hammer RE, Williams SC, Crowley J, Yanagisawa M 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 U S A 105: 16767–16772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. dos Santos EF, Busanello ENB, Miglioranza A, Zanatta A, Barchak AG, Vargas CR, Saute J, Rosa C, Carrion MJ, Camargo D et al (2009) Evidence that folic acid deficiency is a major determinant of hyperhomocysteinemia in Parkinson's disease. Metab Brain Dis 24: 257–269 [DOI] [PubMed] [Google Scholar]
  169. Sato FT, Yap YA, Crisma AR, Portovedo M, Murata GM, Hirabara SM, Ribeiro WR, Marcantonio Ferreira C, Cruz MM, Pereira JNB et al (2020) Tributyrin attenuates metabolic and inflammatory changes associated with obesity through a GPR109A‐dependent mechanism. Cell 9: 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Sato Y, Atarashi K, Plichta DR, Arai Y, Sasajima S, Kearney SM, Suda W, Takeshita K, Sasaki T, Okamoto S et al (2021) Novel bile acid biosynthetic pathways are enriched in the microbiome of centenarians. Nature 599: 458–464 [DOI] [PubMed] [Google Scholar]
  171. Scheppach W, German‐Austrian SCFA Study Group (1996) Treatment of distal ulcerative colitis with short‐chain fatty acid enemas a placebo‐controlled trial. Dig Dis Sci 41: 2254–2259 [DOI] [PubMed] [Google Scholar]
  172. Scheppach W, Sommer H, Kirchner T, Paganelli GM, Bartram P, Christl S, Richter F, Dusel G, Kasper H (1992) Effect of butyrate enemas on the colonic mucosa in distal ulcerative colitis. Gastroenterology 103: 51–56 [DOI] [PubMed] [Google Scholar]
  173. Scholz‐Ahrens KE, Schrezenmeir J (2002) Inulin, oligofructose and mineral metabolism — experimental data and mechanism. Br J Nutr 87: S179 [DOI] [PubMed] [Google Scholar]
  174. Schupack DA, Mars RAT, Voelker DH, Abeykoon JP, Kashyap PC (2022) The promise of the gut microbiome as part of individualized treatment strategies. Nat Rev Gastroenterol Hepatol 19: 7–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Seekatz AM, Theriot CM, Rao K, Chang Y‐M, Freeman AE, Kao JY, Young VB (2018) Restoration of short chain fatty acid and bile acid metabolism following fecal microbiota transplantation in patients with recurrent Clostridium difficile infection. Anaerobe 53: 64–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Senthong V, Li XS, Hudec T, Coughlin J, Wu Y, Levison B, Wang Z, Hazen SL, Tang WHW (2016) Plasma trimethylamine N‐Oxide, a gut microbe‐generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J Am Coll Cardiol 67: 2620–2628 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Serino M, Luche E, Gres S, Baylac A, Bergé M, Cenac C, Waget A, Klopp P, Iacovoni J, Klopp C et al (2012) Metabolic adaptation to a high‐fat diet is associated with a change in the gut microbiota. Gut 61: 543–553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Sharon G, Cruz NJ, Kang D‐W, Gandal MJ, Wang B, Kim Y‐M, Zink EM, Casey CP, Taylor BC, Lane CJ et al (2019) Human gut microbiota from autism spectrum disorder promote behavioral symptoms in mice. Cell 177: 1600–1618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Singh DP, Borse SP, Nivsarkar M (2017) Overcoming the exacerbating effects of ranitidine on NSAID‐induced small intestinal toxicity with quercetin: providing a complete GI solution. Chem Biol Interact 272: 53–64 [DOI] [PubMed] [Google Scholar]
  180. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH et al (2014) Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40: 128–139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly‐Y M, Glickman JN, Garrett WS (2013) The microbial metabolites, short‐chain fatty acids, regulate colonic Treg cell homeostasis. Science 341: 569–573 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Soderholm AT, Pedicord VA (2019) Intestinal epithelial cells: at the interface of the microbiota and mucosal immunity. Immunology 158: 267–280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Somm E, Henry H, Bruce SJ, Aeby S, Rosikiewicz M, Sykiotis GP, Asrih M, Jornayvaz FR, Denechaud PD, Albrecht U et al (2017) β‐Klotho deficiency protects against obesity through a crosstalk between liver, microbiota, and brown adipose tissue. JCI Insight 2: e91809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, Neunlist M (2010) Short‐chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology 138: 1772–1782 [DOI] [PubMed] [Google Scholar]
  185. Sorribas M, Jakob MO, Yilmaz B, Li H, Stutz D, Noser Y, de Gottardi A, Moghadamrad S, Hassan M, Albillos A et al (2019) FXR modulates the gut‐vascular barrier by regulating the entry sites for bacterial translocation in experimental cirrhosis. J Hepatol 71: 1126–1140 [DOI] [PubMed] [Google Scholar]
  186. Spadoni I, Zagato E, Bertocchi A, Paolinelli R, Hot E, Di Sabatino A, Caprioli F, Bottiglieri L, Oldani A, Viale G et al (2015) A gut‐vascular barrier controls the systemic dissemination of bacteria. Science 350: 830–834 [DOI] [PubMed] [Google Scholar]
  187. Suez J, Zmora N, Segal E, Elinav E (2019) The pros, cons, and many unknowns of probiotics. Nat Med 25: 716–729 [DOI] [PubMed] [Google Scholar]
  188. Suez J, Zmora N, Zilberman‐Schapira G, Mor U, Dori‐Bachash M, Bashiardes S, Zur M, Regev‐Lehavi D, Ben‐Zeev Brik R, Federici S et al (2018) Post‐antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 174: 1406–1423 [DOI] [PubMed] [Google Scholar]
  189. Suh SH, Choe K, Hong SP, Jeong S‐H, Mäkinen T, Kim KS, Alitalo K, Surh CD, Koh GY, Song J‐H (2019) Gut microbiota regulates lacteal integrity by inducing VEGF‐C in intestinal villus macrophages. EMBO Rep 20: e46927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Sun J, Furio L, Mecheri R, van der Does AM, Lundeberg E, Saveanu L, Chen Y, van Endert P, Agerberth B, Diana J (2015) Pancreatic β‐cells limit autoimmune diabetes via an immunoregulatory antimicrobial peptide expressed under the influence of the gut microbiota. Immunity 43: 304–317 [DOI] [PubMed] [Google Scholar]
  191. Sun L, Xie C, Wang G, Wu Y, Wu Q, Wang X, Liu J, Deng Y, Xia J, Chen B et al (2018) Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat Med 24: 1919–1929 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Suzuki T, Hara H (2009) Quercetin enhances intestinal barrier function through the assembly of zonnula Occludens‐2, Occludin, and Claudin‐1 and the expression of Claudin‐4 in Caco‐2 cells. J Nutr 139: 965–974 [DOI] [PubMed] [Google Scholar]
  193. Swann JR, Want EJ, Geier FM, Spagou K, Wilson ID, Sidaway JE, Nicholson JK & Holmes E (2011) Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc Natl Acad Sci U S A 108 Suppl 1: 4523–4530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Tachedjian G, Aldunate M, Bradshaw CS, Cone RA (2017) The role of lactic acid production by probiotic Lactobacillus species in vaginal health. Res Microbiol 168: 782–792 [DOI] [PubMed] [Google Scholar]
  195. Tan C, Wu Q, Wang H, Gao X, Xu R, Cui Z, Zhu J, Zeng X, Zhou H, He Y et al (2021) Dysbiosis of gut microbiota and short‐chain fatty acids in acute ischemic stroke and the subsequent risk for poor functional outcomes. JPEN J Parenter Enteral Nutr 45: 518–529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Tan X, Liu Y, Long J, Chen S, Liao G, Wu S, Li C, Wang L, Ling W, Zhu H (2019) Trimethylamine N‐Oxide aggravates liver steatosis through modulation of bile acid metabolism and inhibition of farnesoid X receptor signaling in nonalcoholic fatty liver disease. Mol Nutr Food Res 63: e1900257 [DOI] [PubMed] [Google Scholar]
  197. Thaiss CA, Itav S, Rothschild D, Meijer MT, Levy M, Moresi C, Dohnalová L, Braverman S, Rozin S, Malitsky S et al (2016a) Persistent microbiome alterations modulate the rate of post‐dieting weight regain. Nature 540: 544–551 [DOI] [PubMed] [Google Scholar]
  198. Thaiss CA, Levy M, Korem T, Dohnalová L, Shapiro H, Jaitin DA, David E, Winter DR, Gury‐BenAri M, Tatirovsky E et al (2016b) Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 167: 1495–1510 [DOI] [PubMed] [Google Scholar]
  199. Tierney BT, Yang Z, Luber JM, Beaudin M, Wibowo MC, Baek C, Mehlenbacher E, Patel CJ, Kostic AD (2019) The landscape of genetic content in the gut and oral human microbiome. Cell Host Microbe 26: 283–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Tilg H, Zmora N, Adolph TE, Elinav E (2020) The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 20: 40–54 [DOI] [PubMed] [Google Scholar]
  201. Tobias DK, Lawler PR, Harada PH, Demler OV, Ridker PM, Manson JE, Cheng S, Mora S (2018) Circulating branched‐chain amino acids and incident cardiovascular disease in a prospective cohort of US women. Circ Genom Precis Med 11: e002157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Tough IR, Forbes S, Tolhurst R, Ellis M, Herzog H, Bornstein JC, Cox HM (2011) Endogenous peptide YY and neuropeptide Y inhibit colonic ion transport, contractility and transit differentially via Y₁ and Y₂ receptors. Br J Pharmacol 164: 471–484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Trabelsi M‐S, Daoudi M, Prawitt J, Ducastel S, Touche V, Sayin SI, Perino A, Brighton CA, Sebti Y, Kluza J et al (2015) Farnesoid X receptor inhibits glucagon‐like peptide‐1 production by enteroendocrine L cells. Nat Commun 6: 7629 [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Tremaroli V, Karlsson F, Werling M, Ståhlman M, Kovatcheva‐Datchary P, Olbers T, Fändriks L, le Roux CW, Nielsen J, Bäckhed F (2015) Roux‐en‐Y gastric bypass and vertical banded gastroplasty induce long‐term changes on the human gut microbiome contributing to fat mass regulation. Cell Metab 22: 228–238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom‐Bru C, Blanchard C, Junt T, Nicod LP, Harris NL et al (2014) Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 20: 159–166 [DOI] [PubMed] [Google Scholar]
  206. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP et al (2009) A core gut microbiome in obese and lean twins. Nature 457: 480–484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI (2006) An obesity‐associated gut microbiome with increased capacity for energy harvest. Nature 444: 1027–1031 [DOI] [PubMed] [Google Scholar]
  208. Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV (2008) Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host‐microbial interface. Proc Natl Acad Sci U S A 105: 20858–20863 [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Velazquez‐Villegas LA, Perino A, Lemos V, Zietak M, Nomura M, Pols TWH, Schoonjans K (2018) TGR5 signalling promotes mitochondrial fission and beige remodelling of white adipose tissue. Nat Commun 9: 245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo MR, Phillips RS et al (2014) Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll‐like receptor 4. Immunity 41: 296–310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Vrzáčková N, Ruml T, Zelenka J (2021) Postbiotics, metabolic signaling, and cancer. Molecules 26: 1528 [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Wahlström A, Sayin SI, Marschall H‐U, Bäckhed F (2016) Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab 24: 41–50 [DOI] [PubMed] [Google Scholar]
  213. Wang L, Zhu Q, Lu A, Liu X, Zhang L, Xu C, Liu X, Li H, Yang T (2017) Sodium butyrate suppresses angiotensin II‐induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin–angiotensin system. J Hypertens 35: 1899 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Wang RX, Lee JS, Campbell EL, Colgan SP (2020) Microbiota‐derived butyrate dynamically regulates intestinal homeostasis through regulation of actin‐associated protein synaptopodin. Proc Natl Acad Sci U S A 117: 11648–11657 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Wang S, Dong W, Liu L, Xu M, Wang Y, Liu T, Zhang Y, Wang B, Cao H (2019) Interplay between bile acids and the gut microbiota promotes intestinal carcinogenesis. Mol Carcinog 58: 1155–1167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, Messaddeq N, Harney JW, Ezaki O, Kodama T et al (2006) Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439: 484–489 [DOI] [PubMed] [Google Scholar]
  217. Weitkunat K, Schumann S, Nickel D, Kappo KA, Petzke KJ, Kipp AP, Blaut M, Klaus S (2016) Importance of propionate for the repression of hepatic lipogenesis and improvement of insulin sensitivity in high‐fat diet‐induced obesity. Mol Nutr Food Res 60: 2611–2621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Weitkunat K, Stuhlmann C, Postel A, Rumberger S, Fankhänel M, Woting A, Petzke KJ, Gohlke S, Schulz TJ, Blaut M et al (2017) Short‐chain fatty acids and inulin, but not guar gum, prevent diet‐induced obesity and insulin resistance through differential mechanisms in mice. Sci Rep 7: 6109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, Haase S, Mähler A, Balogh A, Markó L et al (2017) Salt‐responsive gut commensal modulates TH17 axis and disease. Nature 551: 585–589 [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Worthmann A, John C, Rühlemann MC, Baguhl M, Heinsen F‐A, Schaltenberg N, Heine M, Schlein C, Evangelakos I, Mineo C et al (2017) Cold‐induced conversion of cholesterol to bile acids in mice shapes the gut microbiome and promotes adaptive thermogenesis. Nat Med 23: 839–849 [DOI] [PubMed] [Google Scholar]
  221. Wu Y, Ma N, Song P, He T, Levesque C, Bai Y, Zhang A, Ma X (2019) Grape seed proanthocyanidin affects lipid metabolism via changing gut microflora and enhancing propionate production in weaned pigs. J Nutr 149: 1523–1532 [DOI] [PubMed] [Google Scholar]
  222. Xiong Y, Miyamoto N, Shibata K, Valasek MA, Motoike T, Kedzierski RM, Yanagisawa M (2004) Short‐chain fatty acids stimulate leptin production in adipocytes through the G protein‐coupled receptor GPR41. Proc Natl Acad Sci U S A 101: 1045–1050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Yajima T, Inoue R, Matsumoto M, Yajima M (2011) Non‐neuronal release of ACh plays a key role in secretory response to luminal propionate in rat colon. J Physiol 589: 953–962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Yamashita H, Maruta H, Jozuka M, Kimura R, Iwabuchi H, Yamato M, Saito T, Fujisawa K, Takahashi Y, Kimoto M et al (2009) Effects of acetate on lipid metabolism in muscles and adipose tissues of type 2 diabetic Otsuka Long‐Evans Tokushima Fatty (OLETF) rats. Biosci Biotechnol Biochem 73: 570–576 [DOI] [PubMed] [Google Scholar]
  225. Yang W, Yu T, Huang X, Bilotta AJ, Xu L, Lu Y, Sun J, Pan F, Zhou J, Zhang W et al (2020) Intestinal microbiota‐derived short‐chain fatty acids regulation of immune cell IL‐22 production and gut immunity. Nat Commun 11: 4457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez‐Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP et al (2012) Human gut microbiome viewed across age and geography. Nature 486: 222–227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Yu J, Luo Y, Zhu Z, Zhou Y, Sun L, Gao J, Sun J, Wang G, Yao X, Li W (2019) A tryptophan metabolite of the skin microbiota attenuates inflammation in patients with atopic dermatitis through the aryl hydrocarbon receptor. J Allergy Clin Immunol 143: 2108–2119 [DOI] [PubMed] [Google Scholar]
  228. Zaibi MS, Stocker CJ, O'Dowd J, Davies A, Bellahcene M, Cawthorne MA, Brown AJH, Smith DM, Arch JRS (2010) Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett 584: 2381–2386 [DOI] [PubMed] [Google Scholar]
  229. Zaiss MM, Rapin A, Lebon L, Dubey LK, Mosconi I, Sarter K, Piersigilli A, Menin L, Walker AW, Rougemont J et al (2015) The intestinal microbiota contributes to the ability of helminths to modulate allergic inflammation. Immunity 43: 998–1010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, Kanneganti T‐D (2010) The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity 32: 379–391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, Ben‐Yacov O, Lador D, Avnit‐Sagi T, Lotan‐Pompan M et al (2015) Personalized nutrition by prediction of glycemic responses. Cell 163: 1079–1094 [DOI] [PubMed] [Google Scholar]
  232. Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D'Angelo C, Massi‐Benedetti C, Fallarino F et al (2013) Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin‐22. Immunity 39: 372–385 [DOI] [PubMed] [Google Scholar]
  233. Zhang S, Zhang S, Hu L, Zhai L, Xue R, Ye J, Chen L, Cheng G, Mruk J, Kunapuli SP et al (2015) Nucleotide‐binding oligomerization domain 2 receptor is expressed in platelets and enhances platelet activation and thrombosis. Circulation 131: 1160–1170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Zhang Z, Mocanu V, Cai C, Dang J, Slater L, Deehan EC, Walter J, Madsen KL (2019) Impact of fecal microbiota transplantation on obesity and metabolic syndrome—a systematic review. Nutrients 11: 2291 [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Zhao Y, Chen F, Wu W, Sun M, Bilotta AJ, Yao S, Xiao Y, Huang X, Eaves‐Pyles TD, Golovko G 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: 752–762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Zheng X, Cai X, Hao H (2022) Emerging targetome and signalome landscape of gut microbial metabolites. Cell Metab 34: 35–58 [DOI] [PubMed] [Google Scholar]
  237. Zhu H, Cao C, Wu Z, Zhang H, Sun Z, Wang M, Xu H, Zhao Z, Wang Y, Pei G et al (2021) The probiotic L. casei Zhang slows the progression of acute and chronic kidney disease. Cell Metab 33: 1926–1942 [DOI] [PubMed] [Google Scholar]
  238. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M et al (2016) Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165: 111–124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Ziętak M, Kovatcheva‐Datchary P, Markiewicz LH, Ståhlman M, Kozak LP, Bäckhed F (2016) Altered microbiota contributes to reduced diet‐induced obesity upon cold exposure. Cell Metab 23: 1216–1223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Zmora N, Suez J, Elinav E (2018) You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol 16: 35–56 [DOI] [PubMed] [Google Scholar]

Articles from EMBO Reports are provided here courtesy of Nature Publishing Group

RESOURCES