RISING STARS: Endocrine regulation of metabolic homeostasis via the intestine and gut microbiome

in Journal of Endocrinology
Authors:
Rachel K Meyer School of Nutritional Sciences and Wellness, University of Arizona, Tucson, Arizona, USA

Search for other papers by Rachel K Meyer in
Current site
Google Scholar
PubMed
Close
and
Frank A Duca School of Animal and Comparative Biomedical Sciences, College of Agricultural and Life Sciences, University of Arizona, Tucson, Arizona, USA
BIO5 Institute, University of Arizona, Tucson, Arizona, USA

Search for other papers by Frank A Duca in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0003-1760-4081

Correspondence should be addressed to F A Duca: faduca@arizona.edu

This paper is part of a collection of articles highlighting the breadth and depth of research being undertaken across the field of basic endocrinology by early- and mid-career researchers. The collection is published across the Journal of Endocrinology and the Journal of Molecular Endocrinology.

Free access

Sign up for journal news

The gastrointestinal system is now considered the largest endocrine organ, highlighting the importance of gut-derived peptides and metabolites in metabolic homeostasis. Gut peptides are secreted from intestinal enteroendocrine cells in response to nutrients, microbial metabolites, and neural and hormonal factors, and they regulate systemic metabolism via multiple mechanisms. While extensive research is focused on the neuroendocrine effects of gut peptides, evidence suggests that several of these hormones act as endocrine signaling molecules with direct effects on the target organ, especially in a therapeutic setting. Additionally, the gut microbiota metabolizes ingested nutrients and fiber to produce compounds that impact host metabolism indirectly, through gut peptide secretion, and directly, acting as endocrine factors. This review will provide an overview of the role of endogenous gut peptides in metabolic homeostasis and disease, as well as the potential endocrine impact of microbial metabolites on host metabolic tissue function.

Abstract

The gastrointestinal system is now considered the largest endocrine organ, highlighting the importance of gut-derived peptides and metabolites in metabolic homeostasis. Gut peptides are secreted from intestinal enteroendocrine cells in response to nutrients, microbial metabolites, and neural and hormonal factors, and they regulate systemic metabolism via multiple mechanisms. While extensive research is focused on the neuroendocrine effects of gut peptides, evidence suggests that several of these hormones act as endocrine signaling molecules with direct effects on the target organ, especially in a therapeutic setting. Additionally, the gut microbiota metabolizes ingested nutrients and fiber to produce compounds that impact host metabolism indirectly, through gut peptide secretion, and directly, acting as endocrine factors. This review will provide an overview of the role of endogenous gut peptides in metabolic homeostasis and disease, as well as the potential endocrine impact of microbial metabolites on host metabolic tissue function.

Invited Author’s profile

article image

Dr Frank Duca is an assistant professor in the School of Animal and Comparative Biomedical Sciences at the University of Arizona. He obtained his PhD from Pierre and Marie Curie University in 2013, examining the impact of high-fat diets and obesity on gut-brain signaling and the gut microbiome. He was a Banting Postdoctoral Fellow at the Toronto General Hospital Research Institute, under the mentorship of Dr Tony Lam, where he examined how metformin can directly, and indirectly via the gut microbiome, impact hepatic glucose production through a neuronal gut–brain–liver axis. At the University of Arizona, his lab is currently focused on how dietary and environmental exposures can impact gut–brain signaling mechanisms that regulate metabolic homeostasis. His lab is especially interested in how changes in the gut metagenome and metabolome can influence the development of metabolic dysregulation via alterations in nutrient-sensing, vagal signaling, and the central nervous system.

Introduction

Energy and glucose homeostasis are tightly controlled by coordinated neural and endocrine signals that facilitate tissue crosstalk and central nervous system (CNS) integration to regulate food intake, energy expenditure, and glycemia. The liver, pancreas, and adipose tissue are traditionally considered organs of the endocrine system involved in regulating metabolic homeostasis. The endocrine pancreas secretes insulin in response to the postprandial rise in blood glucose, repressing hepatic glucose production and facilitating glucose uptake in adipose tissue and skeletal muscle while glucagon secretion generally opposes these actions (Campbell & Newgard 2021). Further, adipocytes secrete adipokines, including leptin and adiponectin, to regulate food intake and maintain fat stores (Scheja & Heeren 2019). The textbook functions of the gastrointestinal (GI) system are digestion and nutrient absorption; however, the gut is now considered the largest endocrine organ, maintaining energy and glucose homeostasis both directly and indirectly via gut peptides produced by enteroendocrine cells (EECs) and endocrine metabolites produced or altered by the gut microbiota (Ahlman & Nilsson 2001).

EECs are dispersed throughout the GI tract, comprising only 1% of the total intestinal epithelial cell population (Worthington et al. 2018). Despite the low abundance of EECs, they have a major role in the maintenance of energy and glucose homeostasis, evidenced by glucose intolerance in mice lacking normal EEC development (Terry et al. 2014). Gut peptides are secreted in response to the sensing of luminal contents and function to coordinate digestion, nutrient absorption, appetite, energy expenditure, and insulin secretion (Table 1) (Gribble & Reimann 2019). Recent advances have demonstrated the complexity and redundancy of these signaling molecules, as peptides impacting metabolic homeostasis are still being identified while the mechanisms of action and metabolic effects of previous peptides are continually redefined or discovered. While a large proportion of gut peptides act in a paracrine fashion on nearby intestinal epithelial cells or peripheral nerves, like vagal afferent neurons or spinal afferents that can signal to the brain (Wachsmuth et al. 2022), studies suggest many intestinally derived peptides can enter the bloodstream and act in an endocrine fashion. This review focuses on the endocrine signaling capabilities of gut peptides, as other recent reviews have highlighted the role of neural signaling in regulating the metabolic effects (see Duca et al. 2021, Wachsmuth et al. 2022 for more).

Table 1

Summary of intestinal gut peptides.

Peptide EEC type Tissue location of secretion Function References
Serotonin Enterochromaffin cell Throughout the GI tract Regulation of intestinal motility and inflammation, gluconeogenesis and glucose uptake, adipose tissue lipolysis, brown adipose tissue thermogenesis Sumara et al. (2012), Heredia et al. (2013), Margolis et al. (2014), Crane et al. (2015)
CCK I cell Small intestine Regulation of gallbladder contraction, gastric emptying, pancreatic exocrine secretion, brown adipose tissue thermogenesis and hepatic glucose production, decreases food intake Lorenz & Goldman (1982), Li & Owyang (1993), Schwartz et al. (1993), Sonobe et al. (1995), Cheung et al. (2009), Blouet & Schwartz (2012)
GIP K cell (also found in some GLP-1 secreting cells) Small intestine Amplifies glucose-stimulated insulin secretion, promotes β-cell survival and proliferation Kim et al. (2005), Gasbjerg et al. (2019)
Neurotensin N cell Small intestine Increases bile acid reabsorption and gallbladder motility, regulates insulin, somatostatin and glucagon secretion Dolais-Kitabgi et al. (1979), Yamasato & Nakayama (1988), Béraud-Dufour et al. (2010), Li et al. (2021 b)
GLP-1 L cell Small intestine through rectum Amplifies glucose-stimulated insulin secretion, promotes β-cell survival and proliferation, decreases food intake, delays gastric emptying Turton et al. (1996), Davis et al. (1998), Li et al. (2005), Hare et al. (2010), Lamont et al. (2012), Zhang et al. (2022)
GLP-2 L cell Small intestine through rectum Increases epithelial cell proliferation, intestinal barrier function and intestinal hexose transport, inhibits gastric acid secretion Cheeseman & Tsang (1996), Drucker et al. (1996), Wøjdemann et al. (1999), Benjamin et al. (2000)
PYY L cell Small intestine through rectum Inhibits gastric acid secretion, gastric emptying and pancreatic exocrine secretion, decreases food intake Adrian et al. (1985), Grandt et al. (1995), Moran et al. (2005), Challis et al. (2003), Degen et al. (2005)
Oxyntomodulin L cell Colon Decreases food intake, amplifies glucose-stimulated insulin secretion Dakin et al. (2004), Maida et al. (2008)
INSL5a L cell Colon Increases food intake and hepatic glucose production, regulates islet development and insulin secretion Burnicka-Turek et al. (2012), Grosse et al. (2014), Lee et al. (2016), Zaykov et al. (2019)

aSeveral actions of INSL5 are debated; see text for more details.

CCK, cholecystokinin; GI, gastrointestinal; GIP, glucose-dependent insulinotropic peptide; GLP-1, glucagon-like peptide 1; GLP-2, glucagon-like peptide 2; INSL5, insulin-like peptide 5; PYY, peptide YY.

While the role of the intestine in regulating food intake and glucose homeostasis is well documented, the gut microbiota is also now considered a critical component of the intestinal endocrine system (Clarke et al. 2014). The gut microbiota, composed of all bacteria, archaea, and fungi residing in the GI tract, is both directly and indirectly implicated in host metabolic homeostasis (Howard et al. 2022). Many of the effects of the gut microbiota on energy and glucose homeostasis are linked to compounds produced or altered by gut bacteria that act directly on EECs or alternatively enter circulation and target metabolic tissue function (Agus et al. 2021). For example, short-chain fatty acids (SCFAs) produced by gut bacterial fermentation of ingested fiber induce secretion of glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) from EECs, thereby indirectly impacting the gut endocrine system, but also enter the circulation to impact hepatic glucose metabolism (Shimizu et al. 2019), lipid metabolism (Yu et al. 2019), and regulate brown adipose thermogenesis (Cani et al. 2006, Christiansen et al. 2018). In addition, molecular components of microbes, like lipopolysaccharide (LPS), can activate EECs via innate immune recognition (Nguyen et al. 2014, Anhê et al. 2021). As the gut microbiota–metabolome axis impacts host metabolism, this review will discuss several metabolites and bacterial components with endocrine action that participate in the host maintenance of energy and glucose homeostasis. Given the complex interaction of diet, gut microbiota, and the GI tract, it is crucial to better understand how these pathways work in unison to impact host metabolic health.

Gut peptides/hormones

EECs are specialized secretory cells located throughout the GI tract. While EEC subtypes are classically characterized based on the gut peptide they produce (e.g. K-cells secrete glucose-dependent insulinotropic peptide (GIP), L-cells secrete PYY and GLP-1, and I-cells secrete cholecystokinin (CCK); see Fig. 1), it is now accepted that EEC location may more accurately dictate peptide expression based on migration from the crypt to villus (Beumer et al. 2018) and anatomical location (e.g. small intestine vs colon) (Habib et al. 2012). Here, we review the endocrine effect of gut peptides, while there is substantial evidence that many gut peptides act in a paracrine fashion on vagal and spinal afferent neurons innervating the gut to regulate energy and glucose homeostasis (see Duca et al. 2021, Wachsmuth et al. 2022 for more). For example, CCK is a gut hormone secreted by I-cells of the upper small intestine in response to luminal fat and protein, and the CCK receptor is expressed in the GI tract and vagal afferent neurons (Fakhry et al. 2017, Wang et al. 2019). A gut–brain vagal signaling axis is implicated in the effects of CCK on gallbladder contraction (Sonobe et al. 1995), gastric emptying (Schwartz et al. 1993), pancreatic exocrine secretion (Li & Owyang 1993), brown adipose tissue thermogenesis (Blouet & Schwartz 2012), hepatic glucose production (Cheung et al. 2009), and control of feeding behavior (Lorenz & Goldman 1982). However, non-neural signaling pathways for gut peptides are also critical for metabolic homeostasis, especially in the effect of incretin hormones.

Figure 1
Figure 1

Gut peptide secretion and endocrine effects. Enteroendocrine cells (EECs) dispersed throughout the intestine, sense luminal nutrients and microbial metabolites and secrete gut peptides that impact metabolism. K-cells secrete glucose-dependent insulinotropic peptide (GIP); L-cells secrete glucagon-like peptide 1 (GLP-1) and peptide YY (PYY); enterochromaffin cells (EC cells) secrete 5-hydroxytryptamine (5-HT, also known as serotonin); and N-cells secrete neurotensin (NT). Some of these gut peptides, especially GLP-1, PYY, and CCK, impact metabolism via paracrine neuronal signaling. Further, gut peptides enter circulation and act as endocrine factors in the intestine, pancreas, liver, gallbladder, central nervous system, and brown and white adipose tissue. Figure created with BioRender.com.

Citation: Journal of Endocrinology 258, 2; 10.1530/JOE-23-0019

Incretin hormones

There are over 20 known gut peptides secreted by EECs that have both independent and overlapping effects on metabolism. A subset of gut peptides, termed incretins, are released in response to ingested nutrients and perpetuate glucose-stimulated insulin secretion from pancreatic β-cells, accounting for 50–70% of total insulin secretion following meal consumption (Nauck et al. 1986). The incretin hormones, GIP and GLP-1, are secreted in response to meal consumption, with the magnitude of secretion proportional to both rate of nutrient appearance (or rate of gastric emptying) and energy content (Vilsbøll et al. 2003, Ahrén 2022). Traditionally, K-cells located in the duodenum and upper jejunum were thought to exclusively secrete GIP, and L-cells located in the ileum and colon were thought to exclusively secrete GLP-1. However, GIP and GLP-1 have been shown to colocalize in a subset of human, rat, and porcine small intestinal EECs, indicating simultaneous postprandial secretion of these peptides (Mortensen et al. 2003, Habib et al. 2012). GIP, the first identified incretin hormone, is secreted in response to luminal glucose and lipids (Wu et al. 2012, 2017). Interestingly, in humans, GIP secretion is greater in response to fat than carbohydrates, despite the glucose-dependent insulinotropic effect of this peptide (Wu et al. 2017). GLP-1 is secreted in response to ingested macronutrients and fiber, as well as neural and hormonal factors (Wang et al. 2015). Further, GLP-1 secretion in response to nutrients and other secretagogues appears to be specific to L-cell localization in the intestine. For example, L-cells of the small intestine are indispensable for the secretion of ingested nutrient-induced GLP-1 (Sun et al. 2017), whereas colonic L cells mediate GLP-1 secretion in response to activation of the G-protein-coupled receptors, GPR119 and melanocortin 4 receptor (MC4R), metformin, bile acids, as well as maximal LPS-induced GLP-1 secretion (Christiansen et al. 2019, Panaro et al. 2020). In addition, microbial metabolites, such as SCFAs, can induce incretin hormone secretion (see section later). Both GIP and GLP-1 are degraded by dipeptidyl-peptidase 4 (DPP4) within minutes of secretion (Kieffer et al. 1995), such that only a small percentage of these hormones reach systemic circulation, calling into question the endocrine ability of these peptides.

Despite the short half-life, GIP and GLP-1 function to amplify glucose-stimulated insulin secretion via direct activation of the GIP receptor (GIPR) and GLP-1 receptor (GLP-1R) expressed on pancreatic β-cells. Both are members of the B family of G-protein-coupled receptors (GPCRs) and have overlapping signaling mechanisms to potentiate glucose-stimulated insulin secretion (Mayo et al. 2003). The binding of GIP or GLP-1 to their associated receptors induces recruitment and activation of the G as protein, adenylate cyclase activation and elevated intracellular cyclic AMP (cAMP), resulting in protein kinase A (PKA) and exchange protein directly activated by cAMP (EPAC)-mediated potentiation of insulin granular exocytosis (Kashima et al. 2001, Dyachok et al. 2006, Kaihara et al. 2013). Further, GLP-1 and GIP activate divergent, PKA-independent signaling mechanisms to promote β-cell survival and proliferation (Li et al. 2005, Kim et al. 2005). In addition to the insulin-stimulating effects of incretins, GLP-1, but not GIP, inhibits glucagon secretion from α-cells, with equal contributions from glucagon inhibition and insulin secretion on glucose homeostasis (Hare et al. 2010). Although there is evidence for a neural GLP-1-mediated regulation of glucose homeostasis, potentially mediated by hepatic portal vein or gut-innervating GLP-1R-expressing neurons (Balkan & Li 2000, Burcelin et al. 2001, Vahl et al. 2007, Borgmann et al. 2021), recent work involving transgenic mice highlights the importance of pancreatic GLP-1R in glucose homeostasis (Lamont et al. 2012). Indeed, the knockdown of the GLP-1R in β-cells abolishes the effects of GLP-1 on insulin secretion (Smith et al. 2014). Further, whole-body GLP-1R-deficient mice have impaired glucose-stimulated insulin secretion and glucose tolerance, whereas reintroduction of the GLP-1R only in pancreatic islets normalized glucose homeostasis and glucose-stimulated insulin secretion (Lamont et al. 2012), while deletion of GLP-1R in neurons does not impair oral glucose-stimulated insulin secretion (Sisley et al. 2014, Varin et al. 2019), all indicating that GLP-1 likely augments glucose-stimulated insulin secretion in an endocrine fashion via pancreatic rather than neural GLP-1Rs.

In addition to direct receptor binding on pancreatic islet cells, GLP-1 is proposed to act on the peripheral and CNS to induce satiation and decrease food intake postprandially. The GLP-1R is expressed on neurons in the hindbrain and hypothalamus (Turton et al. 1996, Adams et al. 2018), key regions regulating feeding behavior, as well as a subset of vagal afferent neurons in the nodose ganglion (Nakagawa et al. 2004). Both central and peripheral GLP-1 administration decreases food intake and GLP-1 secretion activates vagal afferent neurons (Nakabayashi et al. 1996, Turton et al. 1996, Davis et al. 1998, Buckley et al. 2020), sparking debate regarding the neural circuit involved in the effect of GLP-1 on food intake. However, studies utilizing vagal lesioning and deafferentation prompted in part by the rate of GLP-1 degradation by DDP-4 suggest that endogenous GLP-1 acts as a paracrine peptide through a gut-brain vagal circuit to regulate feeding behavior (Abbott et al. 2005a , Plamboeck et al. 2013, Diepenbroek et al. 2017, Borgmann et al. 2021, Brierley & de Lartigue 2022). Conversely, more recently, the impact of vagal GLP-1R on energy homeostasis has been debated, as viral and transgenic knockout studies have shown a limited role in vagal afferent GLP-1 signaling on energy homeostasis (Sisley et al. 2014, Varin et al. 2019, Brierley et al. 2021). Interestingly, in an elegant study, it was demonstrated that the effects of GLP-1 on food intake and gastric emptying are mediated by GLP-1R-expressing ileal enteric neurons (Zhang et al. 2022). Thus, while the endocrine action of GLP-1 is likely limited to the pancreas, the overall impact of neural endogenous GLP-1 signaling is contentious (see McLean et al. (2021) for more detailed endocrine and paracrine signaling of GLP-1).

Oxyntomodulin, like GLP-1, is derived from posttranslational modifications of proglucagon, is secreted postprandially by colonic EECs, and binds both the GLP-1 and glucagon (GCG) receptor (Baldissera et al. 1988, Baggio et al. 2004). Similar to GLP-1, oxyntomodulin acutely decreases food intake in rodents when administered directly to the brain (Dakin et al. 2001) and peripherally (Dakin et al. 2004), likely dependent on hypothalamic GLP-1R activation (Baggio et al. 2004). Further, oxyntomodulin production is blunted in individuals with type 2 diabetes (T2D) (Wewer Albrechtsen et al. 2016), and treatment with oxyntomodulin is beneficial for glucose homeostasis via the amplification of glucose-stimulated insulin secretion and body weight in individuals with T2D and obesity (Wynne et al. 2005, Maida et al. 2008, Shankar et al. 2018). However, more research is needed into determining the mechanism of action of oxyntomodulin, given it has a longer half-life than GLP-1 (~12 min) (Schjoldager et al. 1988).

Glucagon-like peptide 2

Glucagon-like peptide 2 (GLP-2), co-secreted with GLP-1 from intestinal L cells in response to nutrients (Hartmann et al. 2000), has intestinotrophic as well as metabolic effects. At the intestine, GLP-2 plays a protective role in gut barrier function (Benjamin et al. 2000, Chen et al. 2012, Chang et al. 2021) and enhances nutrient absorption (Meier et al. 2006). Additionally, GLP-2 induces glucagon secretion, but has no effect on glycemia, in healthy individuals, suggesting minimal contribution of this peptide in normal glucose homeostasis (Sørensen et al. 2003, Meier et al. 2006). On the contrary, GLP-2 signaling is an attractive target for obesity-associated hyperglycemia, given that, in rodents with obesity, blocking endogenous GLP-2 action worsens glucose tolerance (Baldassano et al. 2015), and peripheral GLP-2 analog treatment improves glucose tolerance independent of body weight (Ejarque et al. 2021). Further, GLP-2 signaling is necessary and sufficient for the metabolic improvements associated with prebiotic supplementation in high-fat feeding (Cani et al. 2009). The effect of GLP-2 on glucose regulation is hypothesized to occur due to decreased adipose tissue inflammation (Ejarque et al. 2021), improved gut barrier that attenuates metabolic endotoxemia (Cani et al. 2009), and/or neuroendocrine action via activation of Pro-opiomelanocortin-expressing neurons of the hypothalamus (Shi et al. 2013); however, the exact mechanism remains to be fully elucidated.

Peptide YY

PYY is a gut peptide expressed in L-cells of the distal intestine, where it is co-secreted with GLP-1 (Habib et al. 2013). PYY exists in two isoforms: PYY1-36 and PYY3-36, formed by DPP4 mediated N-terminal cleavage following secretion (Mentlein et al. 1993). PYY3-36, the dominant form in circulation postprandially (Grandt et al. 1994), principally binds the Y2 receptor found in the CNS, including the hypothalamus and brain stem, as well as peripheral tissues, including the colon and kidney (Yi et al. 2018). PYY is secreted in response to luminal lipids and protein (Batterham et al. 2006, Mangan et al. 2019), as well as neural and gut microbial factors, including SCFAs (Zhang et al. 1993, Larraufie et al. 2018) (see later). PYY functions to inhibit gastric acid secretion, gastric emptying, and pancreatic exocrine secretion (Adrian et al. 1985, Grandt et al. 1995, Moran et al. 2005). Exogenous PYY administration also decreases food intake in rodents and humans (Challis et al. 2003, Degen et al. 2005), suggesting a role for this peptide in the suppression of food intake following meal consumption. Indeed, mice lacking PYY develop obesity, and replacing PYY via once-daily injection or continuous delivery via osmotic minipump induces weight loss in these mice (Batterham et al. 2006), indicating that PYY is an endogenous regulator of food intake. Mechanistically, PYY is proposed to activate Y2 receptors in the nucleus tractus solitarius and/or the arcuate nucleus of the hypothalamus, activating anorexigenic neurons and inhibiting orexigenic neurons (Gustafson et al. 1997, Batterham et al. 2002, Abbott et al. 2005b , Blevins et al. 2008), indicating a clear endocrine action. However, the hypophagic effect of PYY is likely at least in part mediated by a gut–brain vagal circuit, as vagotomy and midbrain transection abolish the effect of PYY on food intake in rats (Koda et al. 2005).

In addition to energy homeostasis, PYY is also implicated in the control of glucose homeostasis. In the pancreas, PYY is co-expressed with glucagon in α-cells and somatostatin in δ-cells (Böttcher et al. 1989, Khan et al. 2016), suggesting an endocrine effect of PYY on insulin secretion. In accordance with the inhibitory actions of this peptide, PYY inhibits glucose-stimulated insulin secretion in vivo (Böttcher et al. 1989). However, as PYY3-36 has no effect on insulin secretion in isolated islets and the Y2 receptor is not expressed in pancreatic islets (Chandarana et al. 2013), locally secreted PYY1-36 can act directly at the islet, whereas gut-derived PYY3-36 has no direct effect on β-cell insulin secretion. In contrast, peripheral PYY3-36 administration improves glucose tolerance likely via EEC Y2 receptor activation and increased GLP-1 secretion that subsequently increases insulin release (Chandarana et al. 2013). Thus, PYY1-36 secreted within pancreatic islets may represent a negative feedback mechanism for glucose-stimulated insulin secretion.

5-Hydroxytryptamine (serotonin)

While 5-hydroxytryptamine (5-HT, also known as serotonin) is canonically considered a neurotransmitter, serotonin is also synthesized by enterochromaffin (EC) cells of the intestine from tryptophan, a process critically regulated by tryptophan availability, tryptophan hydroxylase (the rate-limiting enzyme in serotonin synthesis), and gut microbial metabolism of tryptophan (Yano et al. 2015, Yabut et al. 2019). Serotonin is secreted from EC cells in response to changes in luminal nutrients, microbial metabolites, or stretch following meal consumption (Reigstad et al. 2015, Martin et al. 2017, Wang et al. 2017). Following secretion, the majority of serotonin is taken up and stored or degraded in platelets (Mercado & Kilic 2010), with a small proportion remaining in plasma to act as a signaling factor in peripheral tissues. Because serotonin typically cannot cross the blood–brain barrier, the actions of peripheral serotonin are distinct from central serotonin; as such, gut-derived serotonin is an independent regulator of metabolic tissue function.

Serotonin primarily acts on peripheral tissues via activation of one of fourteen 5-HT receptors (HTRs), all except one of which are classified as G-protein-coupled receptors (Sahu et al. 2018). Peripheral serotonin participates in intestinal homeostasis, including regulation of gut motility via enteric neuron signaling and intestinal inflammation (Heredia et al. 2013, Margolis et al. 2014). In addition, serotonin is implicated in adipose tissue lipid metabolism, as it promotes adipocyte glucose uptake and decreases lipolysis via HTR2A receptor activation (Hansson et al. 2016), and may also play a role in the inhibition of brown adipose tissue thermogenesis, especially during diet-induced obesity (Crane et al. 2015). However, adipocytes synthesize and reuptake serotonin directly, so many of the actions of serotonin on adipose tissue are attributed to local, adipocyte-derived serotonin (Kinoshita et al. 2010, Oh et al. 2015). Interestingly, serotonin is increased during fasting, promotes hepatic gluconeogenesis and inhibits hepatic glucose uptake, and promotes adipose tissue lipolysis via HTR2B activation in the fasted state (Sumara et al. 2012). On the contrary, during states of nutrient availability, serotonin may increase hepatic triglyceride accumulation (Osawa et al. 2011), providing evidence for the potential of HTR3 antagonists for the treatment of non-alcoholic fatty liver disease (Haub et al. 2011).

Insulin-like peptide 5

As previously mentioned, there are over 20 identified gut peptides, and recent research has identified novel gut peptides as well as the functions of known peptides in metabolism. Among these, insulin-like peptide 5 (INSL5), produced by colonic L-cells (Billing et al. 2018), is secreted during fasting and has orexigenic properties (Grosse et al. 2014, Lewis et al. 2020); however, this effect is inconsistent (Zaykov et al. 2019). Interestingly, INSL5 receptor (relaxin/insulin-like family peptide receptor 4) expressing neurons in the hypothalamus was recently found to play a role in the regulation of feeding behavior associated with INSL5 (Lewis et al. 2022). However, as evidence for INSL5 production or presence in the brain is lacking, the physiological role of these neurons in INSL5-mediated feeding behavior is unclear, and it is unknown if these neurons are targeted by gut-derived INSL5. While the biological role of INSL5 is not fully elucidated, INSL5 signaling may participate in islet development and insulin secretion, as mice lacking INSL5 have decreased basal and glucose-stimulated insulin secretion and smaller pancreatic islets compared to wildtype controls, likely due to decreased INSL5-mediated activation of the relaxin family peptide receptor 4 (Burnicka-Turek et al. 2012). Further, INSL5 expression is regulated by the gut microbiota and may act to increase hepatic glucose production, in accordance with its secretion profile during low nutrient availability (Lee et al. 2016).

Neurotensin

Neurotensin, secreted by enteroendocrine N-cells and hypothalamic neurons (Polak et al. 1977), has major implications in the physiology of the CNS but also plays a role in intestinal and metabolic homeostasis. Neurotensin is secreted primarily in response to luminal lipids (Draviam et al. 1990), acting locally to increase lipid absorption via increasing bile acid reabsorption and gallbladder motility (Yamasato & Nakayama 1988, Gui & Carraway 2001, Li et al. 2021). Further, neurotensin may regulate glucose homeostasis, as systemic neurotensin administration results in hepatic glucose production from glycogenolysis and hyperglycemia (Carraway et al. 1976); this effect is likely due to the regulatory effect of neurotensin on insulin, glucagon, and somatostatin secretion from pancreatic islets (Dolais-Kitabgi et al. 1979, Béraud-Dufour et al. 2010). While peripheral neurotensin certainly plays a role in metabolic homeostasis, much of the research is focused on the effects of intracerebroventricular neurotensin on metabolic and energy homeostasis. Further, as the half-life of this peptide is ~30 s in rodents (Aronin et al. 1982), the endocrine effects of peripheral neurotensin have yet to be fully elucidated but are likely extremely limited.

Gut microbiota

The complex gut microbiota–host relationship integrates intestinal and systemic metabolism, impacting gut peptide secretion and overall metabolic tissue function (Agus et al. 2021). As mentioned earlier, the gut microbiota encompasses all microbes residing in the GI tract. However, the majority of research thus far has focused on the impact of the gut bacteria, while only recently have other microbes, like fungi or bacteriophages, been implicated in regulating host metabolic health (Heisel et al. 2017, Sun et al. 2021a , de Jonge et al. 2022).

Interaction of the gut microbiota and gut peptide signaling

The gut microbiota is a key factor for coordinated gut peptide secretion, as germ-free and antibiotic-treated mice have alterations in nutrient-sensing and chemosensory machinery, EEC number, and gut peptide release (Table 2) (Duca et al. 2012, Lee et al. 2016, Modasia et al. 2020). For example, germ-free mice exhibit dysregulated diurnal GLP-1 secretion and consistently increased circulating basal and fed GLP-1 (Bäckhed et al. 2004, Zarrinpar et al. 2018, Martchenko et al. 2020, Heiss et al. 2021), despite discrepancies in intestinal expression in the literature (Duca et al. 2012, Wichmann et al. 2013). This increase in GLP-1 secretion likely mediates the increase in gut transit time observed in germ-free mice compared to conventional mice (Wichmann et al. 2013); however, this has also been attributed to the modulation of bile acids by intestinal bacteria (Li et al. 2021c ). Similarly, INSL5 expression is increased in antibiotic-treated and germ-free mice (Lee et al. 2016), whereas circulating PYY is decreased in germ-free mice during fasting and in response to ingested lipids (Samuel et al. 2008, Duca et al. 2012), suggesting that the gut microbiota regulate L-cell secretion profiles. Germ-free mice also have decreased colonic tryptophan hydroxylase expression and circulating serotonin (Wikoff et al. 2009, Sjögren et al. 2012, Yano et al. 2015), likely due to the key role of the gut microbiota in tryptophan metabolism and serotonin biosynthesis.

Table 2

Summary of gut peptide and expression in germ-free mice compared to conventional mice.

Peptide GF vs conventional mice References
Serotonin Decreased in circulation Wikoff et al. (2009), Sjögren et al. (2012), Yano et al. (2015)
CCK Increased in circulation Martinez-Guryn et al. (2018)
Decreased expression in the proximal intestine Duca et al. (2012)
GIP Increased GIP+ cells in jejunum and colon Modasia et al. (2020)
Neurotensin No data
GLP-1 Increased in circulation Wichmann et al. (2013), Zarrinpar et al. (2018), Heiss et al. (2021)
Increased cecal and colon Gcg expression Wichmann et al. (2013)
Decreased expression in the proximal intestine Duca et al. (2012)
GLP-2 No data
PYY Decreased in circulation and decreased expression in the proximal intestine Duca et al. (2012)
Decreased in circulation compared to mice colonized with B. thetaiotaomicron and M. smithii Samuel et al. (2008)
Oxyntomodulin No data
INSL5 Increased expression in colon Lee et al. (2016)

CCK, cholecystokinin; GF, germ-free; GI, gastrointestinal; GIP, glucose-dependent insulinotropic peptide; GLP-1, glucagon-like peptide 1; GLP-2, glucagon-like peptide 2; INSL5, insulin-like peptide 5; PYY, peptide YY.

Further, germ-free mice have lower fasting insulin and body weight, and it is therefore proposed that the gut microbiota coordinates nutrient harvest and lipid metabolism and storage as a beneficial survival mechanism (Bäckhed et al. 2004). In addition, modulation, rather than ablation, of the gut microbiome with fermentable fiber supplementation induces GLP-1 secretion and glucose tolerance in healthy animals (Massimino et al. 1998), providing a more physiologically relevant model implicating the importance of the gut microbiota in metabolic homeostasis. Thus, modifying the gut microbiota is a promising therapy for obesity and glucose intolerance. Indeed, fermentable fiber supplementation reduces body weight gain in models of diet-induced obesity (Meyer et al. 2022b ) and improves glucose tolerance and insulin sensitivity in diabetic rodents dependent on GLP-1R signaling (Cani et al. 2005, 2006). However, this effect remains controversial in human studies, as fermentable fiber supplementation in individuals with T2D has shown both no effect on postprandial GLP-1 secretion (Birkeland et al. 2021) and increased postprandial GLP-1 and improved glucose tolerance (Zhao et al. 2018). The impact of fiber on GLP-1 signaling could be due to increased number of L-cells or expression of the preproglucagon gene (Massimino et al. 1998, Everard et al. 2011, Kaji et al. 2011), although alterations in the gut microbiota via fermentable fibers induce a myriad of other effects that could impact host metabolism, such as production and alterations in gut-derived metabolites (Meyer et al. 2022a ). For example, both SCFAs and bile acids have been linked with the effect of dietary fiber on gut peptide secretion and subsequent effects on host metabolic homeostasis (Cani et al. 2006, Makki et al. 2023). The specific signaling pathways for which SCFAs, bile acids, and other gut-derived metabolites are discussed in detail in the following section. Altogether, the gut microbiota has a significant impact on the secretion of gut peptides that can impact metabolic homeostasis.

Gut-derived metabolites

Perhaps the most investigated mechanism by which the gut microbiota impacts energy and glucose homeostasis is via the host metabolome, as gut microbes metabolize dietary components and endogenous substances to produce novel bioactive chemicals (Agus et al. 2021). A notable class of compounds produced by intestinal bacteria is SCFAs generated by gut bacterial fiber fermentation. Specifically, fermentable soluble fibers, including resistant starch, β-glucan, inulin/inulin-type fructans, pectin, and soluble corn fiber, are well-established substrates for SCFA production by intestinal bacteria (Martinez et al. 2021). SCFAs can induce gut peptide secretion locally or can enter the systemic circulation to act on peripheral metabolic tissues like the liver and adipose tissue (den Besten et al. 2015, Li et al. 2018). Further, amino acids from the diet are modified by gut microbial metabolism, resulting in altered circulating metabolites that act as endocrine factors to regulate energy and glucose homeostasis (Hubbard et al. 2015, Jo et al. 2021). For example, branched-chain amino acids (BCAAs) are produced by bacterial metabolism of the amino acids, glycine, serine, or threonine (Amorim Franco & Blanchard 2017, Gojda & Cahova 2021). Similarly, bacterial metabolism of histidine produces imidazole propionate, which regulates hepatic metabolism (Koh et al. 2018), and bacterial metabolism of tryptophan produces tryptamine, indoleacetic acid, indole aldehyde, and others that regulate inflammation and metabolism via cellular signaling mechanisms (Roager & Licht 2018). Endogenous compounds can also be modified by intestinal bacteria. Bile acids, produced in the liver, are modified by gut bacteria via deconjugation by bile salt hydrolase and production of exogenous bile acid species (termed secondary and tertiary bile acids) by coordinated bacterial dihydroxylation, oxidation, and epimerization enzymes and resulting in a diverse bile acid pool (Guzior & Quinn 2021); these modifications impact host receptor signaling to alter gut peptide secretion and tissue metabolism (Ridlon et al. 2014). As these compounds both impact intestinal endocrine function and act as endocrine factors themselves, this review will discuss in detail the effects of metabolites produced or altered by the gut microbiota on host energy and glucose homeostasis.

Short-chain fatty acids

As previously mentioned, fiber consumption induces gut peptide secretion at least in part by increasing SCFA production by gut bacteria. It is thought that SCFAs impact gut peptide secretion via the G-protein coupled receptors GPR41 (FFAR3) and GPR43 (FFAR2) expressed on EECs (Fig. 2, Table 3) (Brooks et al. 2017, Christiansen et al. 2018). Knockout of either FFAR2 or FFAR3 reduces GLP-1 secretion in response to either propionate or acetate (Tolhurst et al. 2012), and activation of a mutant FFAR2-DREADD unresponsive to SCFAs induces GLP-1 secretion similar to propionate administration in wildtype mice (Bolognini et al. 2019). Because the concentration of certain SCFAs, like acetate, in the intestinal lumen is consistently maintained to achieve FFAR2 activation (Cummings et al. 1987), it is proposed that SCFAs may impact gut peptide secretion via basolateral receptor activation. In line with this, FFAR2 has been shown to be expressed on the basolateral EEC membrane, and dietary and vascular SCFAs have differential effects on GLP-1 and PYY secretion (Karaki et al. 2006, Christiansen et al. 2018), indicating that absorption may be necessary for SCFA sensing. However, FFAR3 has a higher affinity for butyrate than acetate and propionate (Brown et al. 2003, Le Poul et al. 2003); therefore, luminal butyrate may be sensed by FFAR3 to induce gut peptide secretion. Nonetheless, it is possible that despite the open-faced nature of EECs to the luminal environment, SCFAs may induce gut peptide secretion via an endocrine mechanism that targets the basolateral side of the EECs; the reasoning and exact pathway for this unique mechanism warrants further investigation.

Figure 2
Figure 2

Signaling mechanisms of gut peptide secretion by microbially produced metabolites. Metabolites produced or altered by the gut microbiota that impact gut peptide secretion include short-chain fatty acids (SCFAs), indole metabolites produced from bacterial metabolism of tryptophan, primary bile acids (BAs) that can be deconjugated by bacterial bile salt hydrolase, and secondary BAs produced by bacterial metabolism of primary Bas, among others. SCFAs are proposed to induce the secretion of glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) via FFAR2 and/or FFAR3; however, some studies suggest that SCFA absorption and basolateral FFAR2 is responsible for SCFA-induced gut peptide secretion. Indole metabolites inhibit voltage-gated K+ channels to increase EEC action potential and intracellular Ca2+, and induce GLP-1 secretion; alternatively, indole metabolites may activate the aryl hydrocarbon receptor (AHR) to induce GLP-1 secretion. Bacterial LPS induces GLP-1 secretion via toll-like receptor 4 (TLR4). Primary BAs primarily activate the farnesoid X receptor (FXR) to inhibit GLP-1 secretion, whereas secondary BAs primarily activate the basolateral G-protein bile acid receptor 1 (Gpbar1, also known as TGR5) to induce gut peptide secretion. Black arrows indicate signaling pathways resulting in the induction of gut peptide secretion; red arrow indicates signaling pathways resulting in the inhibition of gut peptide secretion. Figure created with BioRender.com.

Citation: Journal of Endocrinology 258, 2; 10.1530/JOE-23-0019

Table 3

Effects of microbial metabolites or components on gut peptide secretion.

Compound Effect on gut peptide secretion Proposed mechanism References
SCFA Increased GLP-1 and PYY Activation of FFAR2/FFAR3 Tolhurst et al. (2012), Brooks et al. (2017), Christiansen et al. (2018)
Primary and some secondary bile acids Decreased GLP-1 Activation of FXR Trabelsi et al. (2015), Li et al. (2019b, c)
Secondary bile acids Increased GLP-1 and PYY Activation of TGR5 (Gpbar1) Brighton et al. (2015), Kuhre et al. (2018), Christiansen et al. (2019)
Tryptophan metabolites Increased GLP-1 Activation of AHR Natividad et al. (2018)
Increased GLP-1 (acute) Inhibition of voltage-gated K+ channels (acute) Chimerel et al. (2014)
Decreased GLP-1 (prolonged) Decreased ATP production via inhibition of NADH dehydrogenase
LPS (E. coli) Increased GLP-1 Activation of TLR4 Lebrun et al. (2017), Anhê et al. (2021)
LPS (R. sphaeroides) No effect on GLP-1 secretion No (or possibly antagonistic) effect on TLR4 activation Anhê et al. (2021)

AHR, arylhydrocarbon receptor; FFAR2, free fatty acid receptor 2; FFAR3, free fatty acid receptor 3; FXR, farnesoid X receptor; GLP-1, glucagon-like peptide 1; LPS, lipopolysaccharide; PYY, peptide YY; SCFA, short-chain fatty acids; TGR5, G-protein-coupled bile acid receptor (Gpbar1); TLR4, toll-like receptor 4.

In addition to their role in the stimulation of gut peptide secretion, SCFAs are also absorbed into general circulation and can act as endocrine factors in metabolically active tissues (Fig. 3). The majority of SCFAs are removed via first pass by the liver, where they impact hepatic metabolism. For example, butyrate decreases lipogenesis and increases hepatic oxidative respiration and beta-oxidation via activation of AMP-activated protein kinase (AMPK) (Mollica et al. 2017), dependent on peroxisome proliferator-activated receptor gamma (den Besten et al. 2015). Acetate and propionate can also be used by hepatocytes for ATP production and gluconeogenesis, respectively (Anderson & Bridges 1984, Fujino et al. 2001). A small amount of SCFAs escape hepatocyte uptake and enter the general circulation to regulate adipocyte thermogenesis and browning. Specifically, butyrate, and, to a lesser extent, acetate, are consistently shown to induce adipocyte browning and increase thermogenesis in mice (Gao et al. 2009, Sahuri-Arisoylu et al. 2016, Li et al. 2019a , Wang et al. 2020). However, there are differential effects of circulating acetate compared to acetate derived from adipocytes acting as a paracrine signal (Sun et al. 2021b ), indicating the effect of acetate on adipocyte browning may be dependent on source and concentration. Circulating SCFAs alter tissue metabolism via two primary mechanisms: activation of GPCR signaling and epigenetic regulation. As GPR41 and GPR43 are widely expressed (Brown et al. 2003), these receptors may mediate the endocrine actions of SCFAs in the liver (Aoki et al. 2021) and adipose tissue (Kimura et al. 2013). SCFAs also directly impact gene expression via epigenetic modulation to regulate metabolic function. Indeed, SCFA administration in diet-induced obese mice induces expression of adiponectin and resistin via decreasing CpG methylation at adiponectin and resistin promotor regions (Lu et al. 2018). In addition, butyrate acts as a histone deacetylase (HDAC) inhibitor to alter gene expression (Vidali et al. 1978), including hepatic fibroblast growth factor 21 (FGF21) through HDAC3 inhibition (Li et al. 2012) and skeletal muscle insulin receptor substrate 1 (IRS1), peroxisome proliferator-activated receptor-gamma coactivator-1 alpha, and sirtuins to regulate insulin receptor signaling (Chriett et al. 2019). The effects of butyrate on HDAC inhibition have also been extensively investigated in the context of inflammatory bowel disease (Li et al. 2021a ), gut immunity (Yang et al. 2020), and asthma (Islam et al. 2022), implicating butyrate as a key epigenetic regulator of both metabolic and immune cell function. However, the impact of endogenous SCFA on host metabolism is less resolved, as a majority of the studies outlined above utilize orally or intraperitoneally administrated SCFAs, which is not physiologically relevant. Additionally, several studies suggest SCFAs act in a neutral fashion to regulate host metabolism (Goswami et al. 2018, Li et al. 2018, Muller et al. 2020), and at least one study demonstrated that intravenous administration had no impact on improving energy homeostasis (Li et al. 2018). Future studies examining the metabolic impact of SCFAs should aim to deliver SCFAs directly to the large intestine to more closely mimic endogenous production, or at the very least, should try to replicate post-prandial levels in portal and general circulation (Meyer et al. 2022b ). At least one study though has elegantly demonstrated that endogenous SCFAs derived from dietary fiber fermentation can enter circulation and reach the CNS to impact energy homeostasis (Frost et al. 2014), thus underscoring the need for further investigation.

Figure 3
Figure 3

Microbial metabolites enter circulation and impact metabolic organ function. Microbial metabolites discussed in the text are listed with their metabolic organ targets. BCAAs, branched-chain amino acids; ClpB, caseinolytic peptidase B protein homolog; IMP, imidazole propionate; LPS, lipopolysaccharide; SCFAs, short-chain fatty acids. Figure created with BioRender.com.

Citation: Journal of Endocrinology 258, 2; 10.1530/JOE-23-0019

Bile acids

Bile acid concentrations increase in the intestinal lumen postprandially, playing a critical role in lipid absorption in the proximal small intestine as emulsifying agents. However, it is now known that bile acid signaling is a critical regulator of metabolic homeostasis, via paracrine and endocrine actions that are mediated in part by interactions with the gut bacteria. For example, bile acids regulate food intake through distinct signaling pathways via induction of gut peptide secretion by acting as ligands for both the G-protein coupled bile acid receptor-1 (GPBAR1, also known as TGR5) and farnesoid X receptor (FXR) (Fig. 2, Table 3) (Chiang 2013). Indeed, the effects of intraluminal bile acids on gut peptide secretion are well-documented in rodents (Kuhre et al. 2018, Christiansen et al. 2019) as well as humans (Adrian et al. 1993, Adrian et al. 2012, Hansen et al. 2016) and are induced via TGR5 (Christiansen et al. 2019). TGR5 is highly expressed in the colon, where secondary bile acids are produced; as such, the endogenous ligands of TGR5 are conjugated secondary bile acids produced by gut bacteria from host-derived primary bile acids, with taurine-conjugated lithocholic acid being the most potent agonist (Duboc et al. 2014). While luminal bile acids were thought to induce GLP-1 and PYY secretion dependent on apical TGR5, more recent research suggests that bile acid absorption and basolateral TGR5 are required for the effect of bile acids on gut peptide secretion (Brighton et al. 2015, Kuhre et al. 2018), as intraluminal TGR5 agonism has no effect on gut peptide secretion, but intravascular administration of a TGR5 agonist induces robust GLP-1 responses (Christiansen et al. 2019). Additionally, based on the gut peptide-stimulating effect of TGR5 agonism, this signaling pathway has recently been implicated in treatments for obesity and T2D (Zheng et al. 2021), including fiber supplementation and gastric bypass surgeries (Ding et al. 2016, McGavigan et al. 2017), both of which are associated with increased plasma GLP-1 and attenuated food intake.

FXR is expressed in the ileum where primary bile acid concentrations are the greatest, thus FXR activity is largely regulated by primary bile acid species, with chenodeoxycholic acid being the most potent agonist, and rodent taurine-conjugated beta-muricholic acid a potent FXR antagonist (Makishima et al. 1999, Sayin et al. 2013). Whereas TGR5 induces gut peptide secretion, FXR inhibits proglucagon expression and GLP-1 secretion via interaction with cAMP response element binding protein (CREB) in EECs (Trabelsi et al. 2015, Li et al. 2019b,c). Further, FXR activation impairs SCFA-induced gut peptide secretion via inhibition of FFAR2 signaling (Ducastel et al. 2020), demonstrating complex interactions and converging signaling pathways between different classes of microbial metabolites. Interestingly, despite the antagonistic role of FXR signaling in bile acid- and SCFA-mediated GLP-1 secretion in metabolically healthy individuals, FXR activation promotes weight loss and improvements in glucose regulation following gastric bypass surgery (Ryan et al. 2014) and increases intestinal EEC number ex vivo (Kim et al. 2022), suggesting dynamic FXR signaling dependent on the physiological state. Additionally, FXR is localized in peripheral metabolic tissues (Cariou et al. 2006, Zhang et al. 2014), and it is plausible that the differing metabolic outcomes observed during studies involving FXR are due to action in the intestine vs other tissues like the liver.

Aside from their role in the induction of gut peptides, bile acids can also impact host metabolism in peripheral tissues and within the CNS (Fig. 3). As bile acids undergo enterohepatic circulation, they can both directly and indirectly alter systemic physiology through hepatic and intestinal FXR, respectively; however, the role of FXR remains contentious in individuals with normal metabolic function and metabolic syndrome. For example, while global FXR-deficient mice on a normal chow diet display peripheral insulin resistance and elevated serum free fatty acids (Cariou et al. 2006), mice with global, but not liver-specific, FXR deficiency are protected from diet-induced obesity and insulin resistance (Prawitt et al. 2011). On the contrary, intestinal FXR agonism prevents diet-induced obesity and insulin resistance (Fang et al. 2015), further complicating the role of FXR in obesity and metabolic disease. Intestinal FXR may exert beneficial effects via secretion of FGF19 (rodent FGF15) that acts on the fibroblast growth factor receptor 4 (FGFR4) to control bile acid, glucose, and lipid metabolism (Stroeve et al. 2010), as FGF15/19 represses gluconeogenic enzyme expression and postprandial lipogenesis and induces glycogen synthesis (Kir et al. 2011, Potthoff et al. 2011, Kim et al. 2020). However, bile acids also exert an FGF19-independent effect on hepatic lipid metabolism through FXR, as hepatic FXR deficiency induces hepatic triglyceride accumulation and elevated serum cholesterol, whereas intestinal FXR deficiency has no effect on hepatic or circulating lipids (Schmitt et al. 2015). FGF15/19 is also involved in the adipose tissue thermogenic response to cold (Fang et al. 2015, Morón-Ros et al. 2021). Finally, levels of the FXR agonist taurochenodeoxycholic acid increase with high-fat feeding due to small intestinal gut microbiota modulation and impair insulin action in the dorsal vagal cortex dependent on FXR (Zhang et al. 2021, Meyer et al. 2022a), implicating central FXR in control of glucose homeostasis.

TGR5, on the other hand, is consistently reported to be metabolically beneficial. Following a meal, bile acids increase temporally in the hypothalamus, where TGR5 activation participates in satiety and decreases food intake (Perino et al. 2021). Therefore, TGR5 is a prime target for obesity, as central TGR5 agonism in obesity reduces body weight and food intake and increases energy expenditure via the sympathetic nervous system (Castellanos-Jankiewicz et al. 2021). Peripheral TGR5 also increases energy expenditure in humans (Broeders et al. 2015) and mice via TGR5-mediated intracellular thyroid hormone activation and adipose tissue beiging (Watanabe et al. 2006, Velazquez-Villegas et al. 2018).

Amino acids and derivatives

Large-scale metabolomic studies have identified gut microbiota-related amino acid metabolites that regulate metabolic homeostasis via endocrine action. Among these, BCAAs are essential amino acids derived from the diet or gut bacterial biosynthesis. Following absorption, BCAA catabolism occurs primarily in skeletal muscle, where the activity of the first enzyme in the BCAA catabolic pathway, branched-chain-amino-acid aminotransferase, is high. In healthy individuals, BCAAs, especially leucine, promote protein synthesis and inhibit proteolysis through mammalian target of rapamycin (mTOR) signaling (Suryawan et al. 2008). In the brain, BCAAs compete for transport with other aromatic amino acids (tryptophan, tyrosine, and phenylalanine) and can thus decrease the production of certain neurotransmitters, including serotonin (Gijsman et al. 2002, Choi et al. 2013). In addition, BCAA catabolism results in the production of alanine, a key gluconeogenic amino acid, and can therefore promote hepatic glucose production during starvation when BCAA levels increase (Fig. 3) (Holecek et al. 2016). These metabolic effects provide the basis for BCAA supplementation for athletes; however, human studies suggest that the benefits of BCAAs are limited (Plotkin et al. 2021).

Interestingly, plasma BCAAs are elevated in obesity and correlate with insulin resistance and are a predictor of T2D (Felig et al. 1969, Newgard et al. 2009, Wang et al. 2011b , Vanweert et al. 2021). Evidence suggests that both peripheral and hepatic insulin resistance occurs with elevated BCAAS in obesity and T2D, independent of body weight. BCAA supplementation in diet-induced obesity induces skeletal muscle insulin resistance via phosphorylation of mTOR and IRS1, in accordance with the known functions of BCAAs in skeletal muscle (Newgard et al. 2009). Mechanistically, elevated BCAAs in T2D occur at least in part due to altered expression of enzymes involved in BCAA metabolism in the liver, skeletal muscle, and adipose tissue (She et al. 2007, Lian et al. 2015), as well as due to increased abundance of BCAA-producing bacteria and decreased abundance of bacteria that uptake BCAAs in the gut (Pedersen et al. 2016). In the liver, enzymes that regulate BCAA catabolism also control hepatic lipogenesis; therefore, dysregulated expression of these enzymes could contribute to hepatic insulin resistance (White et al. 2018). On the other hand, strategies that reduce circulating BCAAs, like gastric bypass surgery, improve peripheral insulin sensitivity independent of body weight (Magkos et al. 2013, Lips et al. 2014) at least in part by decreasing muscle fatty acyl CoA and glycine accumulation (White et al. 2016).

In addition to BCAAs, imidazole propionate (IMP), a metabolite produced by gut bacterial histidine metabolism, has recently gained attention in the context of T2D. Individuals with T2D have increased portal vein and peripheral IMP levels (Koh et al. 2018), increased pro-inflammatory gut bacteria (Molinaro et al. 2020), and low gut microbial diversity (Menni et al. 2020). Despite no differences in dietary histidine intake, IMP is positively correlated with saturated fat and negatively correlated with fiber and unsaturated fat consumption in individuals with T2D, indicating that diet-mediated gut microbiota modulation is critical for IMP production (Molinaro et al. 2020). Following absorption, IMP impairs glucose tolerance and insulin signaling in mice through a p38γ mitogen-activated protein kinase (MAPK)-p62-mTOR complex 1 (mTORC1) signaling axis (Fig. 3) (Koh et al. 2018). Interestingly, individuals with T2D and high blood glucose taking metformin have increased IMP levels, and the blood glucose lowering effect of metformin is blunted with IMP pretreatment in mice, dependent on p38γ MAPK-Akt mediated inhibitory AMPK phosphorylation (Koh et al. 2020). Taken together, these data provide a promising framework for therapeutics targeting IMP-producing bacteria for the treatment of T2D.

The amino acid tryptophan is also metabolized by gut bacteria, producing metabolites that impact host receptor activity. While over 95% of dietary tryptophan is metabolized directly by the host via indoleamine 2,3-dioxygenase 1, gut bacteria can metabolize tryptophan into tryptamine and indole metabolites. Production of the metabolite tryptamine in the gut is impacted by bacterial metabolism of tryptophan, as germ-free mice have lower fecal tryptamine than humanized mice (Marcobal et al. 2013), and it is estimated that >10% of individuals harbor gut microbes that express at least one of the enzymes for decarboxylation of tryptophan to produce tryptamine (Williams et al. 2014). Further, metabolic syndrome is associated with blunted production of tryptamine and indole metabolites from dietary tryptophan due to gut microbiome dysbiosis (Natividad et al. 2018). Although a relatively low-abundance metabolite, tryptamine induces serotonin secretion from gut EC cells (Takaki et al. 1985), potentially indirectly impacting peripheral tissue metabolism. Tryptamine is also a proposed therapeutic for gut inflammatory disorders, as it induces mucus secretion from goblet cells via the G-protein coupled serotonin receptor HTR4 (Bhattarai et al. 2020). The effects of tryptophan metabolites on metabolic homeostasis are at least partially dependent on the aryl hydrocarbon receptor (AHR). Indeed, high fat-fed mice treated with either an AHR agonist or Lactobacillus reuteri, a bacteria with high AHR ligand production, have improved gut barrier function and metabolic homeostasis possibly mediated by AHR-induced GLP-1 secretion from EECs (Natividad et al. 2018). However, a previous study found that indole induces acute GLP-1 secretion from EECs via voltage-gated K+ channel inhibition (Chimerel et al. 2014); therefore, multiple potential intersecting pathways may be responsible for indole-mediated GLP-1 secretion (Fig. 2, Table 3). On the other hand, exposure to the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (also known as dioxin) is correlated with hyperglycemia and insulin resistance in humans (Henriksen et al. 1997, Cranmer et al. 2000), and mice expressing a low-binding affinity AHR variant and global AHR deficient mice are resistant to diet-induced obesity and associated metabolic perturbances (Wang et al. 2011a , Kerley-Hamilton et al. 2012, Xu et al. 2015). Taken together, while these data indicate a potential impact of indole metabolites impacting metabolic homeostasis via AHR, there is much to be determined in regards to the site of action and mechanism. Indeed, AHR is expressed in a variety of cell types and is a critical regulator of NFKB inflammatory signaling (Ishihara et al. 2021). Thus, it is possible that there exists a balance of pro- and anti-inflammatory signaling required to maintain homeostasis, which is further dependent on the specific tissue affected. For example, intestinal AHR activation improves intestinal inflammation associated with obesity (Postal et al. 2020) and promotes the secretion of anti-inflammatory cytokines in the intestine to improve gut barrier and metabolic homeostasis in mice challenged with high-fat feeding (Lin et al. 2019). Nonetheless, bacterially derived tryptophan metabolites represent an exciting new area of research in metabolic disease and warrant further research.

Bacterial components

Chronic low-grade inflammation often occurs in obesity and obesity-associated metabolic disorders, at least in part due to LPS exposure. Western-style high-fat diet-feeding and loss of gut barrier integrity in obesity promote LPS absorption, resulting in host low-grade inflammation and impaired glucose homeostasis, termed metabolic endotoxemia (Fig. 3) (Cani et al. 2007a, Pendyala et al. 2012). High-fat diet-fed mice have increased circulating LPS, and chronic LPS exposure increases body weight, worsens glucose tolerance and insulin sensitivity, and increases inflammatory cytokine expression dependent on the cell surface receptor cluster of differentiation 14 (CD14) (Cani et al. 2007a). Both rodent and human obesity is associated with increased adipose tissue expression of proinflammatory cytokines, including tumor necrosis factor-α that is correlated with hyperinsulinemia and inhibits insulin receptor tyrosine kinase activity through IRS-1 (Hotamisligil et al. 1993, 1995, 1996). Further, OFS supplementation in high-fat feeding improves glucose homeostasis, reduces adipose and circulating inflammatory cytokines, and increases gut Bifidobacterium sp. that are negatively associated with endotoxemia, further implicating the gut microbiota composition and in the detrimental inflammatory and metabolic effects of high-fat feeding (Cani et al. 2007b). Circulating LPS forms a complex with LPS-binding protein, which can interact with cell surface receptors CD14, toll-like receptor 4 (TLR4), and toll-like receptor 2 (TLR2), inducing proinflammatory cytokine release (Mohammad & Thiemermann 2020). As such, some reports suggest that CD14 and TLR4-deficient mice are protected from diet-induced obesity and insulin resistance (Kim et al. 2007, Poggi et al. 2007, Roncon-Albuquerque et al. 2008, Jia et al. 2014), whereas others suggest that neither TLR4 or CD14 mediate diet-induced obesity (Young et al. 2012, Dalby et al. 2018). These discrepancies in the literature may be due to differences in knockout tissue specificity, genetic background, or diet and indicate a need to further elucidate the significance of TLR4 in the pathophysiology of metabolic disorders. Interestingly, hexa-acylated LPS derived from Escherichia coli induces GLP-1 secretion from enteroendocrine L-cells in response to intestinal injury to reduce inflammation via TLR4 activation (Fig. 2, Table 3) (Lebrun et al. 2017), whereas penta-acylated LPS from R. sphaeroides has no effect on GLP-1 secretion (Table 3) (Anhê et al. 2021), indicating that the effects of LPS on metabolism are dependent on diet, physiological state, and species-specific LPS type. In addition to TLR4, bacterial LPS agonizes TLR2, altering cellular metabolism and immune cell activation (Kirschning et al. 1998). Further, mice lacking TLR2 are resistant to diet-induced obesity and glucose intolerance (Ehses et al. 2010, Guo et al. 2021), and inhibition of TLR2 signaling improves insulin sensitivity (Caricilli et al. 2008). On the contrary, flagellin, the primary protein found in bacterial flagella, may reduce metabolic endotoxemia via TLR5-mediated gut microbiota remodeling. Upon activation by flagellin, TLR5, expressed in the intestinal epithelium, modulates the presence of pathogenic gut bacteria (Carvalho et al. 2012). Interestingly, mice lacking whole body and intestinal TLR5 develop metabolic endotoxemia, with increased body weight and adiposity and abnormal glucose regulation, as well as susceptibility to colonization with pathogenic bacteria (Vijay-Kumar et al. 2010, Chassaing et al. 2014), implicating an intestinal feedback loop in which pathogenic bacteria stimulate TLR5 that, in turn, impairs pathogenic bacterial growth to regulate intestinal inflammation and prevent metabolic endotoxemia. Together, these studies suggest a role for multiple TLRs in inflammation-associated metabolic perturbances.

The nucleotide-binding oligomerization domain-containing proteins, NOD1 and NOD2, are ubiquitously expressed pattern recognition receptors that recognize bacterial cell wall components, including peptidoglycans from gram-negative and some gram-positive bacteria (Rivers et al. 2019). In particular, NOD1 and NOD2 have been studied in bacterial induction of inflammatory signaling that results in insulin resistance. Expression of NOD1 and NOD2 is elevated in individuals with metabolic syndrome (Shiny et al. 2013, Lappas 2014, Zhou et al. 2015) and diet-induced obese rodents (Sharma et al. 2022), and mice lacking NOD1, but not NOD2, are resistant to diet-induced body weight gain and glucose intolerance (Amar et al. 2011). Further, activation of NOD1 is consistently linked to adipose tissue inflammation and peripheral insulin resistance (Schertzer et al. 2011, Zhao et al. 2011, Zhou et al. 2012). Taken together, inflammatory signaling induced by bacterial activation of TLRs and/or NOD-like receptors may be a promising target for the treatment of metabolic disease.

The bacterial protein, caseinolytic peptidase B protein homolog (ClpB), expressed by E. coli has been identified as an antigen mimetic protein of α-melanocyte stimulating hormone (αMSH) (Tennoune et al. 2014), a key neuropeptide involved in the regulation of food intake. While little is known about the physiological effects of ClpB, this protein has been implicated in the development of eating disorders, and, more recently, obesity, as gut bacterial ClpB-like gene function is negatively correlated with obesity in humans (Arnoriaga-Rodríguez et al. 2020). Further, chronic intragastric E. coli treatment decreases food intake, while treatment with Clpb-deficient E. coli has no effect on food intake (Tennoune et al. 2014); this effect is proposed to be mediated by increased PYY secretion with ClpB exposure (Dominique et al. 2019). Additionally, treatment with a strain Hafnia alvei expressing ClpB with an αMSH-like motif reduces food intake and body weight in diet-induced obese mice, reduces food intake in genetically obese ob/ob mice (Legrand et al. 2020) and improves body weight loss in humans (Déchelotte et al. 2021), providing the foundation for research into novel probiotic ClpB-expressing bacterial strains for obesity.

Conclusions and future perspectives

Given the expanding viewpoint for the GI tract as an important endocrine organ in the regulation of metabolic homeostasis, it is no surprise that several of the most successful treatment options for obesity and diabetes are gut-derived in nature. For example, two classes of drugs, GLP-1R agonists (GLP-1RA), like liraglutide, and DPP4 inhibitors, like sitagliptin, improve T2D via activating GLP-1R signaling mechanisms. Interestingly, GLP-1RAs possess a long half-life, while DPP4 inhibitors increase the half-life of endogenous GLP-1 (Omar & Ahrén 2014, Nauck et al. 2021); therefore, these drugs can target endocrine actions of GLP-1R signaling. For example, it is likely that GLP-1RAs improve glucose homeostasis via amplification of glucose-stimulated insulin secretion at the β-cell and induce significant weight loss via CNS action (Lamont et al. 2012, Varin et al. 2019). More recently, clinical trials investigating both dual GLP-1R/GIPR agonists and GLP-1/glucagon receptor (GCGR) agonists as well as GLP-1R/GIPR/GCGR triagonists indicate positive effects on weight loss and glycemia, with GCGR and GLP-1R agonism promoting weight loss and GIPR agonism negating the effects of glucagon signaling on hepatic glucose production (Capozzi et al. 2018, Coskun et al. 2018, Frias et al. 2018, Ji et al. 2021). For example, the ‘twincretin’ tirzepatide is generally more effective at reducing glycemia and body weight compared to the GLP-1 analog semaglutide with the same safety profile (Vadher et al. 2022), whereas GLP-1R/GIPR/GCGR triagonists show early synergistic effects on metabolism, improving glycemic control and body weight to a greater extent than dual incretin receptor agonists in rodents (Finan et al. 2015). However, with all these current treatments, there are moderate side effects, including nausea, diarrhea, and, to a lesser extent, vomiting, constipation, abdominal pain, and dyspepsia (Filippatos et al. 2014). Therefore, future studies must continue to understand the endocrine action of gut peptide signals, as a better understanding of potential sites of action could lead to more personalized and targeted therapies that limit side effects.

In contrast to the establishment and success of GLP-1-mediated therapies, therapies targeting the vast potential of the gut microbiota are still in infancy. As such, while many studies have highlighted the potential of various probiotics in metabolic homeostasis (Stenman et al. 2014, Bauer et al. 2018), only a few have been successful in clinical trials (Kadooka et al. 2010, Minami et al. 2015, Bernini et al. 2016, Depommier et al. 2019). However, as sequencing efforts become more advanced, there is a greater likelihood that gut bacteria will be discovered that have novel roles in mediating energy and glucose homeostasis. For example, one group has discovered a gut bacteria that is capable of producing ClpB, which could have major implications for metabolism (Tennoune et al. 2014). Additionally, there is the emerging field of bioengineered bacteria, with several groups generating bacteria capable of producing specific metabolites, like leptin and GLP-1, that target metabolic organs to prevent or treat metabolic disease (Bermúdez-Humarán et al. 2007, Arora et al. 2016). Nonetheless, despite these efforts, it is possible that probiotic treatment may be highly personalized, as some individuals are permissive to the colonization of probiotics while others are resistant, depending on their pre-existing gut microbiota (Zmora et al. 2018). Indeed, the gut microbiome is highly complex and individualized, thus baseline gut microbiome and metabolome conditions could influence whether treatments targeting the gut microbiome are successful. For example, an individual’s baseline gut microbiome and metabolome can dictate the successful glucoregulatory effect of exercise, while a machine-learning algorithm can use this information to predict if an individual will ‘respond’ to exercise based on microbial characteristics (Liu et al. 2020). A similar program uses a machine learning algorithm to personalize dietary interventions for glucose tolerance using baseline gut microbial signatures in combination with diet and health information (Berry et al. 2020). Altogether, this highlights the importance of comprehensive clinical studies that incorporate not only phenotypic characteristics but also baseline gut metagenomic and metabolomic analyses to determine if drug–gut interactions dictate the success or failure of treatments toward obesity and diabetes. While the exact mechanisms are not completely elucidated, it is evident that both gut peptides and gut microbiota-derived compounds act as endocrine factors to impact host signaling and metabolic homeostasis, representing a relatively novel and exciting collection of compounds and receptors that can be targeted for treatment of metabolic disease.

Declaration of interest

The authors declare no conflicts of interest.

Funding

This work was supported by the National Institute of Diabetes and Digestive and Kidney Diseases of the NIH (R01DK121804).

References

  • Abbott CR, Monteiro M, Small CJ, Sajedi A, Smith KL, Parkinson JR, Ghatei MA & & Bloom SR 2005a The inhibitory effects of peripheral administration of peptide YY(3–36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Research 1044 127131. (https://doi.org/10.1016/j.brainres.2005.03.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Abbott CR, Small CJ, Kennedy AR, Neary NM, Sajedi A, Ghatei MA & & Bloom SR 2005b Blockade of the neuropeptide Y Y2 receptor with the specific antagonist BIIE0246 attenuates the effect of endogenous and exogenous peptide YY(3–36) on food intake. Brain Research 1043 139144. (https://doi.org/10.1016/j.brainres.2005.02.065)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adams JM, Pei H, Sandoval DA, Seeley RJ, Chang RB, Liberles SD & & Olson DP 2018 Liraglutide modulates appetite and body weight through glucagon-like peptide 1 receptor-expressing glutamatergic neurons. Diabetes 67 15381548. (https://doi.org/10.2337/db17-1385)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adrian TE, Ballantyne GH, Longo WE, Bilchik AJ, Graham S, Basson MD, Tierney RP & & Modlin IM 1993 Deoxycholate is an important releaser of peptide YY and enteroglucagon from the human colon. Gut 34 12191224. (https://doi.org/10.1136/gut.34.9.1219)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adrian TE, Gariballa S, Parekh KA, Thomas SA, Saadi H, Al Kaabi J, Nagelkerke N, Gedulin B & & Young AA 2012 Rectal taurocholate increases L cell and insulin secretion, and decreases blood glucose and food intake in obese type 2 diabetic volunteers. Diabetologia 55 23432347. (https://doi.org/10.1007/s00125-012-2593-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adrian TE, Savage AP, Sagor GR, Allen JM, Bacarese-Hamilton AJ, Tatemoto K, Polak JM & & Bloom SR 1985 Effect of peptide YY on gastric, pancreatic, and biliary function in humans. Gastroenterology 89 494499. (https://doi.org/10.1016/0016-5085(8590442-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Agus A, Clément K & & Sokol H 2021 Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 70 11741182. (https://doi.org/10.1136/gutjnl-2020-323071)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ahlman H & Nilsson 2001 The gut as the largest endocrine organ in the body. Annals of Oncology 12(Supplement 2) S63S68. (https://doi.org/10.1093/annonc/12.suppl_2.s63)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ahrén B 2022 Glucose-dependent insulinotropic polypeptide secretion after oral macronutrient ingestion: the human literature revisited and a systematic study in model experiments in mice. Journal of Diabetes Investigation 13 16551665. (https://doi.org/10.1111/jdi.13836)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, Smirnova N, Bergé M, Sulpice T, Lahtinen S, et al.2011 Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Molecular Medicine 3 559572. (https://doi.org/10.1002/emmm.201100159)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Amorim Franco TM & & Blanchard JS 2017 Bacterial branched-chain amino acid biosynthesis: structures, mechanisms, and drugability. Biochemistry 56 58495865. (https://doi.org/10.1021/acs.biochem.7b00849)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Anderson JW & & Bridges SR 1984 Short-chain fatty acid fermentation products of plant fiber affect glucose metabolism of isolated rat hepatocytes. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine 177 372376. (https://doi.org/10.3181/00379727-177-41958)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Anhê FF, Barra NG, Cavallari JF, Henriksbo BD & & Schertzer JD 2021 Metabolic endotoxemia is dictated by the type of lipopolysaccharide. Cell Reports 36 109691. (https://doi.org/10.1016/j.celrep.2021.109691)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aoki R, Onuki M, Hattori K, Ito M, Yamada T, Kamikado K, Kim YG, Nakamoto N, Kimura I, Clarke JM, et al.2021 Commensal microbe-derived acetate suppresses NAFLD/NASH development via hepatic FFAR2 signalling in mice. Microbiome 9 188. (https://doi.org/10.1186/s40168-021-01125-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arnoriaga-Rodríguez M, Mayneris-Perxachs J, Burokas A, Pérez-Brocal V, Moya A, Portero-Otin M, Ricart W, Maldonado R & & Fernández-Real JM 2020 Gut bacterial ClpB-like gene function is associated with decreased body weight and a characteristic microbiota profile. Microbiome 8 59. (https://doi.org/10.1186/s40168-020-00837-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Aronin N, Carraway RE, Ferris CF, Hammer RA & & Leeman SE 1982 The stability and metabolism of intravenously administered neurotensin in the rat. Peptides 3 637642. (https://doi.org/10.1016/0196-9781(8290164-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arora T, Wegmann U, Bobhate A, Lee YS, Greiner TU, Drucker DJ, Narbad A & & Bäckhed F 2016 Microbially produced glucagon-like peptide 1 improves glucose tolerance in mice. Molecular Metabolism 5 725730. (https://doi.org/10.1016/j.molmet.2016.06.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF & & Gordon JI 2004 The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences of the United States of America 101 1571815723. (https://doi.org/10.1073/pnas.0407076101)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baggio LL, Huang Q, Brown TJ & & Drucker DJ 2004 Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology 127 546558. (https://doi.org/10.1053/j.gastro.2004.04.063)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baldassano S, Rappa F, Amato A, Cappello F & & Mulè F 2015 GLP-2 as beneficial factor in the glucose homeostasis in mice fed a high fat diet. Journal of Cellular Physiology 230 30293036. (https://doi.org/10.1002/jcp.25039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baldissera FG, Holst JJ, Knuhtsen S, Hilsted L & & Nielsen OV 1988 Oxyntomodulin (glicentin-(33–69)): pharmacokinetics, binding to liver cell membranes, effects on isolated perfused pig pancreas, and secretion from isolated perfused lower small intestine of pigs. Regulatory Peptides 21 151166. (https://doi.org/10.1016/0167-0115(8890099-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Balkan B & & Li X 2000 Portal GLP-1 administration in rats augments the insulin response to glucose via neuronal mechanisms. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 279 R1449R1454. (https://doi.org/10.1152/ajpregu.2000.279.4.R1449)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, Brynes AE, Low MJ, Ghatei MA, et al.2002 Gut hormone PYY(3–36) physiologically inhibits food intake. Nature 418 650654. (https://doi.org/10.1038/nature00887)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Batterham RL, Heffron H, Kapoor S, Chivers JE, Chandarana K, Herzog H, Le Roux CW, Thomas EL, Bell JD & & Withers DJ 2006 Critical role for peptide YY in protein-mediated satiation and body-weight regulation. Cell Metabolism 4 223233. (https://doi.org/10.1016/j.cmet.2006.08.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bauer PV, Duca FA, Waise TMZ, Dranse HJ, Rasmussen BA, Puri A, Rasti M, O'brien CA & & Lam TKT 2018 Lactobacillus gasseri in the Upper Small Intestine Impacts an ACSL3-Dependent Fatty Acid-Sensing Pathway Regulating Whole-Body glucose Homeostasis. Cell Metabolism 27 572–587.e6. (https://doi.org/10.1016/j.cmet.2018.01.013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Benjamin MA, Mckay DM, Yang PC, Cameron H & & Perdue MH 2000 Glucagon-like peptide-2 enhances intestinal epithelial barrier function of both transcellular and paracellular pathways in the mouse. Gut 47 112119. (https://doi.org/10.1136/gut.47.1.112)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Béraud-Dufour S, Abderrahmani A, Noel J, Brau F, Waeber G, Mazella J & & Coppola T 2010 Neurotensin is a regulator of insulin secretion in pancreatic beta-cells. International Journal of Biochemistry and Cell Biology 42 16811688. (https://doi.org/10.1016/j.biocel.2010.06.018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bermúdez-Humarán LG, Nouaille S, Zilberfarb V, Corthier G, Gruss A, Langella P & & Issad T 2007 Effects of intranasal administration of a leptin-secreting Lactococcus lactis recombinant on food intake, body weight, and immune response of mice. Applied and Environmental Microbiology 73 53005307. (https://doi.org/10.1128/AEM.00295-07)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bernini LJ, Simao AN, Alfieri DF, Lozovoy MA, Mari NL, De Souza CH, Dichi I & & Costa GN 2016 Beneficial effects of Bifidobacterium lactis on lipid profile and cytokines in patients with metabolic syndrome: a randomized trial. Effects of probiotics on metabolic syndrome. Nutrition 32 716719. (https://doi.org/10.1016/j.nut.2015.11.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Berry SE, Valdes AM, Drew DA, Asnicar F, Mazidi M, Wolf J, Capdevila J, Hadjigeorgiou G, Davies R, Al Khatib H, et al.2020 Human postprandial responses to food and potential for precision nutrition. Nature Medicine 26 964973. (https://doi.org/10.1038/s41591-020-0934-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Beumer J, Artegiani B, Post Y, Reimann F, Gribble F, Nguyen TN, Zeng H, Van Den Born M, Van Es JH & & Clevers H 2018 Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signalling gradient. Nature Cell Biology 20 909916. (https://doi.org/10.1038/s41556-018-0143-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhattarai Y, Jie S, Linden DR, Ghatak S, Mars RAT, Williams BB, Pu M, Sonnenburg JL, Fischbach MA, Farrugia G, et al.2020 Bacterially derived tryptamine increases mucus release by activating a Host Receptor in a Mouse Model of Inflammatory Bowel Disease. iScience 23 101798. (https://doi.org/10.1016/j.isci.2020.101798)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Billing LJ, Smith CA, Larraufie P, Goldspink DA, Galvin S, Kay RG, Howe JD, Walker R, Pruna M, Glass L, et al.2018 Co-storage and release of insulin-like peptide-5, glucagon-like peptide-1 and peptideYY from murine and human colonic enteroendocrine cells. Molecular Metabolism 16 6575. (https://doi.org/10.1016/j.molmet.2018.07.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Birkeland E, Gharagozlian S, Gulseth HL, Birkeland KI, Hartmann B, Holst JJ, Holst R & & Aas AM 2021 Effects of prebiotics on postprandial GLP-1, GLP-2 and glucose regulation in patients with type 2 diabetes: a randomised, double-blind, placebo-controlled crossover trial. Diabetic Medicine 38 e14657. (https://doi.org/10.1111/dme.14657)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Blevins JE, Chelikani PK, Haver AC & & Reidelberger RD 2008 PYY(3–36) induces Fos in the arcuate nucleus and in both catecholaminergic and non-catecholaminergic neurons in the nucleus tractus solitarius of rats. Peptides 29 112119. (https://doi.org/10.1016/j.peptides.2007.11.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Blouet C & & Schwartz GJ 2012 Duodenal lipid sensing activates vagal afferents to regulate non-shivering brown fat thermogenesis in rats. PLoS One 7 e51898. (https://doi.org/10.1371/journal.pone.0051898)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bolognini D, Barki N, Butcher AJ, Hudson BD, Sergeev E, Molloy C, Moss CE, Bradley SJ, Le Gouill C, Bouvier M, et al.2019 Chemogenetics defines receptor-mediated functions of short chain free fatty acids. Nature Chemical Biology 15 489498. (https://doi.org/10.1038/s41589-019-0270-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Borgmann D, Ciglieri E, Biglari N, Brandt C, Cremer AL, Backes H, Tittgemeyer M, Wunderlich FT, Brüning JC & & Fenselau H 2021 Gut-brain communication by distinct sensory neurons differently controls feeding and glucose metabolism. Cell Metabolism 33 14661482.e7. (https://doi.org/10.1016/j.cmet.2021.05.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Böttcher G, Ahrén B, Lundquist I & & Sundler F 1989 Peptide YY: intrapancreatic localization and effects on insulin and glucagon secretion in the mouse. Pancreas 4 282288. (https://doi.org/10.1097/00006676-198906000-00002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brierley DI & & de Lartigue G 2022 Reappraising the role of the vagus nerve in GLP-1-mediated regulation of eating. British Journal of Pharmacology 179 584599. (https://doi.org/10.1111/bph.15603)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brierley DI, Holt MK, Singh A, De Araujo A, Mcdougle M, Vergara M, Afaghani MH, Lee SJ, Scott K, Maske C, et al.2021 Central and peripheral GLP-1 systems independently suppress eating. Nature Metabolism 3 258273. (https://doi.org/10.1038/s42255-021-00344-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brighton CA, Rievaj J, Kuhre RE, Glass LL, Schoonjans K, Holst JJ, Gribble FM & & Reimann F 2015 Bile acids trigger GLP-1 release predominantly by accessing basolaterally located G protein-coupled bile acid receptors. Endocrinology 156 39613970. (https://doi.org/10.1210/en.2015-1321)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Broeders EP, Nascimento EB, Havekes B, Brans B, Roumans KH, 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 Metabolism 22 418426. (https://doi.org/10.1016/j.cmet.2015.07.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brooks L, Viardot A, Tsakmaki A, Stolarczyk E, Howard JK, Cani PD, Everard A, Sleeth ML, Psichas A, Anastasovskaj J, et al.2017 Fermentable carbohydrate stimulates FFAR2-dependent colonic PYY cell expansion to increase satiety. Molecular Metabolism 6 4860. (https://doi.org/10.1016/j.molmet.2016.10.011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, et al.2003 The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. Journal of Biological Chemistry 278 1131211319. (https://doi.org/10.1074/jbc.M211609200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buckley MM, O'brien R, Brosnan E, Ross RP, Stanton C, Buckley JM & & O'malley D 2020 Glucagon-like Peptide-1 secreting L-cells coupled to sensory nerves translate microbial signals to the host rat nervous system. Frontiers in Cellular Neuroscience 14 95. (https://doi.org/10.3389/fncel.2020.00095)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Burcelin R, Da Costa A, Drucker D & & Thorens B 2001 Glucose competence of the hepatoportal vein sensor requires the presence of an activated glucagon-like peptide-1 receptor. Diabetes 50 17201728. (https://doi.org/10.2337/diabetes.50.8.1720)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Burnicka-Turek O, Mohamed BA, Shirneshan K, Thanasupawat T, Hombach-Klonisch S, Klonisch T & & Adham IM 2012 INSL5-deficient mice display an alteration in glucose homeostasis and an impaired fertility. Endocrinology 153 46554665. (https://doi.org/10.1210/en.2012-1161)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Campbell JE & & Newgard CB 2021 Mechanisms controlling pancreatic islet cell function in insulin secretion. Nature Reviews. Molecular Cell Biology 22 142158. (https://doi.org/10.1038/s41580-020-00317-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, et al.2007a Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56 17611772. (https://doi.org/10.2337/db06-1491)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cani PD, Daubioul CA, Reusens B, Remacle C, Catillon G & & Delzenne NM 2005 Involvement of endogenous glucagon-like peptide-1(7–36) amide on glycaemia-lowering effect of oligofructose in streptozotocin-treated rats. Journal of Endocrinology 185 457465. (https://doi.org/10.1677/joe.1.06100)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM & & Burcelin R 2006 Improvement of glucose tolerance and hepatic insulin sensitivity by oligofructose requires a functional glucagon-like peptide 1 receptor. Diabetes 55 14841490. (https://doi.org/10.2337/db05-1360)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, Gibson GR & & Delzenne NM 2007b Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 50 23742383. (https://doi.org/10.1007/s00125-007-0791-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cani PD, Possemiers S, Van De Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM, et al.2009 Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 58 10911103. (https://doi.org/10.1136/gut.2008.165886)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Capozzi ME, Dimarchi RD, Tschöp MH, Finan B & & Campbell JE 2018 Targeting the incretin/glucagon system with triagonists to treat diabetes. Endocrine Reviews 39 719738. (https://doi.org/10.1210/er.2018-00117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Caricilli AM, Nascimento PH, Pauli JR, Tsukumo DM, Velloso LA, Carvalheira JB & & Saad MJ 2008 Inhibition of toll-like receptor 2 expression improves insulin sensitivity and signaling in muscle and white adipose tissue of mice fed a high-fat diet. Journal of Endocrinology 199 399406. (https://doi.org/10.1677/JOE-08-0354)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cariou B, Van Harmelen K, Duran-Sandoval D, Van Dijk TH, Grefhorst A, Abdelkarim M, Caron S, Torpier G, Fruchart JC, Gonzalez FJ, et al.2006 The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. Journal of Biological Chemistry 281 1103911049. (https://doi.org/10.1074/jbc.M510258200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carraway RE, Demers LM & & Leeman SE 1976 Hyperglycemic effect of neurotensin, a hypothalamic peptide. Endocrinology 99 14521462. (https://doi.org/10.1210/endo-99-6-1452)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carvalho FA, Koren O, Goodrich JK, Johansson ME, Nalbantoglu I, Aitken JD, Su Y, Chassaing B, Walters WA, González A, et al.2012 Transient inability to manage proteobacteria promotes chronic gut inflammation in TLR5-deficient mice. Cell Host & Microbe 12 1 39152. (https://doi.org/10.1016/j.chom.2012.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Castellanos-Jankiewicz A, Guzmán-Quevedo O, Fénelon VS, Zizzari P, Quarta C, Bellocchio L, Tailleux A, Charton J, Fernandois D, Henricsson M, et al.2021 Hypothalamic bile acid-TGR5 signaling protects from obesity. Cell Metabolism 33 14831492.e10. (https://doi.org/10.1016/j.cmet.2021.04.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Challis BG, Pinnock SB, Coll AP, Carter RN, Dickson SL & & O'rahilly S 2003 Acute effects of PYY3-36 on food intake and hypothalamic neuropeptide expression in the mouse. Biochemical and Biophysical Research Communications 311 915919. (https://doi.org/10.1016/j.bbrc.2003.10.089)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chandarana K, Gelegen C, Irvine EE, Choudhury AI, Amouyal C, Andreelli F, Withers DJ & & Batterham RL 2013 Peripheral activation of the Y2-receptor promotes secretion of GLP-1 and improves glucose tolerance. Molecular Metabolism 2 142152. (https://doi.org/10.1016/j.molmet.2013.03.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang Y, Deng Q, Zhang Z, Zhao H, Tang J, Chen X, Liu G, Tian G, Cai J & & Jia G 2021 Glucagon-like peptide 2 attenuates intestinal mucosal barrier injury through the MLCK/pMLC signaling pathway in a piglet model. Journal of Cellular Physiology 236 30153032. (https://doi.org/10.1002/jcp.30068)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chassaing B, Ley RE & & Gewirtz AT 2014 Intestinal epithelial cell toll-like receptor 5 regulates the intestinal microbiota to prevent low-grade inflammation and metabolic syndrome in mice. Gastroenterology 147 136377.e17. (https://doi.org/10.1053/j.gastro.2014.08.033)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheeseman CI & & Tsang R 1996 The effect of GIP and glucagon-like peptides on intestinal basolateral membrane hexose transport. American Journal of Physiology 271 G477G482. (https://doi.org/10.1152/ajpgi.1996.271.3.G477)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen X, Zhao HX, Fu XS, Li CP & & Zhong XL 2012 Glucagonlike peptide 2 protects intestinal barrier in severe acute pancreatitis through regulating intestinal epithelial cell proliferation and apoptosis. Pancreas 41 10801085. (https://doi.org/10.1097/MPA.0b013e31824966b0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheung GW, Kokorovic A, Lam CK, Chari M & & Lam TK 2009 Intestinal cholecystokinin controls glucose production through a neuronal network. Cell Metabolism 10 99109. (https://doi.org/10.1016/j.cmet.2009.07.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chiang JY 2013 Bile acid metabolism and signaling. Comprehensive Physiology 3 11911212. (https://doi.org/10.1002/cphy.c120023)

  • Chimerel C, Emery E, Summers DK, Keyser U, Gribble FM & & Reimann F 2014 Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Reports 9 12021208. (https://doi.org/10.1016/j.celrep.2014.10.032)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Choi S, Disilvio B, Fernstrom MH & & Fernstrom JD 2013 Oral branched-chain amino acid supplements that reduce brain serotonin during exercise in rats also lower brain catecholamines. Amino Acids 45 11331142. (https://doi.org/10.1007/s00726-013-1566-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chriett S, Dąbek A, Wojtala M, Vidal H, Balcerczyk A & & Pirola L 2019 Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Scientific Reports 9 742. (https://doi.org/10.1038/s41598-018-36941-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Christiansen CB, Gabe MBN, Svendsen B, Dragsted LO, Rosenkilde MM & & Holst JJ 2018 The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. American Journal of Physiology. Gastrointestinal and Liver Physiology 315 G53G65. (https://doi.org/10.1152/ajpgi.00346.2017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Christiansen CB, Trammell SAJ, Wewer Albrechtsen NJ, Schoonjans K, Albrechtsen R, Gillum MP, Kuhre RE & & Holst JJ 2019 Bile acids drive colonic secretion of glucagon-like-peptide 1 and peptide-YY in rodents. American Journal of Physiology. Gastrointestinal and Liver Physiology 316 G574–G584. (https://doi.org/10.1152/ajpgi.00010.2019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF & & Dinan TG 2014 Minireview: gut microbiota: the neglected endocrine organ. Molecular Endocrinology 28 12211238. (https://doi.org/10.1210/me.2014-1108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coskun T, Sloop KW, Loghin C, Alsina-Fernandez J, Urva S, Bokvist KB, Cui X, Briere DA, Cabrera O, Roell WC, et al.2018 LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: from discovery to clinical proof of concept. Molecular Metabolism 18 314. (https://doi.org/10.1016/j.molmet.2018.09.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Crane JD, Palanivel R, Mottillo EP, Bujak AL, Wang H, Ford RJ, Collins A, Blümer RM, Fullerton MD, Yabut JM, et al.2015 Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nature Medicine 21 166172. (https://doi.org/10.1038/nm.3766)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cranmer M, Louie S, Kennedy RH, Kern PA & & Fonseca VA 2000 Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is associated with hyperinsulinemia and insulin resistance. Toxicological Sciences 56 431436. (https://doi.org/10.1093/toxsci/56.2.431)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cummings JH, Pomare EW, Branch WJ, Naylor CP & & Macfarlane GT 1987 Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28 12211227. (https://doi.org/10.1136/gut.28.10.1221)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dakin CL, Gunn I, Small CJ, Edwards CMB, Hay DL, Smith DM, Ghatei MA & & Bloom SR 2001 Oxyntomodulin inhibits food intake in the rat. Endocrinology 142 42444250. (https://doi.org/10.1210/endo.142.10.8430)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dakin CL, Small CJ, Batterham RL, Neary NM, Cohen MA, Patterson M, Ghatei MA & & Bloom SR 2004 Peripheral oxyntomodulin reduces food intake and body weight gain in rats. Endocrinology 145 26872695. (https://doi.org/10.1210/en.2003-1338)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dalby MJ, Aviello G, Ross AW, Walker AW, Barrett P & & Morgan PJ 2018 Diet induced obesity is independent of metabolic endotoxemia and TLR4 signalling, but markedly increases hypothalamic expression of the acute phase protein, SerpinA3N. Scientific Reports 8 15648. (https://doi.org/10.1038/s41598-018-33928-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Davis HR, Mullins DE, Pines JM, Hoos LM, France CF, Compton DS, Graziano MP, Sybertz EJ, Strader CD & & Van Heek M 1998 Effect of chronic central administration of glucagon-like peptide-1 (7–36) amide on food consumption and body weight in normal and obese rats. Obesity Research 6 147156. (https://doi.org/10.1002/j.1550-8528.1998.tb00329.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Jonge PA, Wortelboer K, Scheithauer TPM, Van Den Born B-JH, Zwinderman AH, Nobrega FL, Dutilh BE, Nieuwdorp M & & Herrema H 2022 Gut virome profiling identifies a widespread bacteriophage family associated with metabolic syndrome. Nature Communications 13 3594. (https://doi.org/10.1038/s41467-022-31390-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Déchelotte P, Breton J, Trotin-Picolo C, Grube B, Erlenbeck C, Bothe G, Fetissov SO & & Lambert G 2021 The probiotic strain H. alvei HA4597(®) improves weight loss in overweight subjects under moderate hypocaloric diet: a proof-of-concept, multicenter randomized, double-blind placebo-controlled study. Nutrients 13. (https://doi.org/10.3390/nu13061902)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Degen L, Oesch S, Casanova M, Graf S, Ketterer S, Drewe J & & Beglinger C 2005 Effect of peptide YY3-36 on food intake in humans. Gastroenterology 129 14301436. (https://doi.org/10.1053/j.gastro.2005.09.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • den Besten G, Bleeker A, Gerding A, Van Eunen K, Havinga R, Van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud DJ, et al.2015 Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARgamma-dependent switch from lipogenesis to fat oxidation. Diabetes 64 23982408. (https://doi.org/10.2337/db14-1213)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, Falony G, Raes J, Maiter D, Delzenne NM, et al.2019 Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nature Medicine 25 10961103. (https://doi.org/10.1038/s41591-019-0495-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Diepenbroek C, Quinn D, Stephens R, Zollinger B, Anderson S, Pan A & & De Lartigue G 2017 Validation and characterization of a novel method for selective vagal deafferentation of the gut. American Journal of Physiology. Gastrointestinal and Liver Physiology 313 G342–G352. (https://doi.org/10.1152/ajpgi.00095.2017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ding L, Sousa KM, Jin L, Dong B, Kim BW, Ramirez R, Xiao Z, Gu Y, Yang Q, Wang J, et al.2016 Vertical sleeve gastrectomy activates GPBAR-1/TGR5 to sustain weight loss, improve fatty liver, and remit insulin resistance in mice. Hepatology 64 760773. (https://doi.org/10.1002/hep.28689)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dolais-Kitabgi J, Kitabgi P, Brazeau P & & Freychet P 1979 Effect of neurotensin on insulin, glucagon, and somatostatin release from isolated pancreatic islets. Endocrinology 105 256260. (https://doi.org/10.1210/endo-105-1-256)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dominique M, Breton J, Guérin C, Bole-Feysot C, Lambert G, Déchelotte P & & Fetissov S 2019 Effects of macronutrients on the in vitro production of ClpB, a bacterial mimetic protein of α-MSH and its possible role in satiety signaling. Nutrients 11. (https://doi.org/10.3390/nu11092115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Draviam EJ, Upp JR, Greeley GH, Townsend CM & & Thompson JC 1990 Effect of oral fat on plasma levels of neurotensin and neurotensin fragments in humans. Characterization by high-pressure liquid chromatography. Digestive Diseases and Sciences 35 200204. (https://doi.org/10.1007/BF01536763)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Drucker DJ, Erlich P, Asa SL & & Brubaker PL 1996 Induction of intestinal epithelial proliferation by glucagon-like peptide 2. Proceedings of the National Academy of Sciences of the United States of America 93 79117916. (https://doi.org/10.1073/pnas.93.15.7911)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duboc H, Taché Y & & Hofmann AF 2014 The bile acid TGR5 membrane receptor: from basic research to clinical application. Digestive and Liver Disease 46 302312. (https://doi.org/10.1016/j.dld.2013.10.021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duca FA, Swartz TD, Sakar Y & & Covasa M 2012 Increased oral detection, but decreased intestinal signaling for fats in mice lacking gut microbiota. PLoS One 7 e39748. (https://doi.org/10.1371/journal.pone.0039748)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duca FA, Waise TMZ, Peppler WT & & Lam TKT 2021 The metabolic impact of small intestinal nutrient sensing. Nature Communications 12 903. (https://doi.org/10.1038/s41467-021-21235-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ducastel S, Touche V, Trabelsi MS, 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. Scientific Reports 10 174. (https://doi.org/10.1038/s41598-019-56743-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dyachok O, Isakov Y, Sågetorp J & & Tengholm A 2006 Oscillations of cyclic AMP in hormone-stimulated insulin-secreting β-cells. Nature 439 349352. (https://doi.org/10.1038/nature04410)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ehses JA, Meier DT, Wueest S, Rytka J, Boller S, Wielinga PY, Schraenen A, Lemaire K, Debray S, Van Lommel L, et al.2010 Toll-like receptor 2-deficient mice are protected from insulin resistance and beta cell dysfunction induced by a high-fat diet. Diabetologia 53 17951806. (https://doi.org/10.1007/s00125-010-1747-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ejarque M, Sabadell-Basallote J, Beiroa D, Calvo E, Keiran N, Nuñez-Roa C, Rodríguez MDM, Sabench F, Del Castillo D, Jimenez V, et al.2021 Adipose tissue is a key organ for the beneficial effects of GLP-2 metabolic function. British Journal of Pharmacology 178 21312145. (https://doi.org/10.1111/bph.15278)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GG, Neyrinck AM, Possemiers S, Van Holle A, Francois P, De Vos WM, et al.2011 Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 60 27752786. (https://doi.org/10.2337/db11-0227)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fakhry J, Wang J, Martins P, Fothergill LJ, Hunne B, Prieur P, Shulkes A, Rehfeld JF, Callaghan B & & Furness JB 2017 Distribution and characterisation of CCK containing enteroendocrine cells of the mouse small and large intestine. Cell and Tissue Research 369 245253. (https://doi.org/10.1007/s00441-017-2612-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fang S, Suh JM, Reilly SM, Yu E, Osborn O, Lackey D, Yoshihara E, Perino A, Jacinto S, Lukasheva Y, et al.2015 Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nature Medicine 21 159165. (https://doi.org/10.1038/nm.3760)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Felig P, Marliss E & & Cahill GF 1969 Plasma amino acid levels and insulin secretion in obesity. New England Journal of Medicine 281 811816. (https://doi.org/10.1056/NEJM196910092811503)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Filippatos TD, Panagiotopoulou TV & & Elisaf MS 2014 Adverse effects of GLP-1 receptor agonists. Review of Diabetic Studies 11 202230. (https://doi.org/10.1900/RDS.2014.11.202)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Finan B, Yang B, Ottaway N, Smiley DL, Ma T, Clemmensen C, Chabenne J, Zhang L, Habegger KM, Fischer K, et al.2015 A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nature Medicine 21 2736. (https://doi.org/10.1038/nm.3761)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C, Urva S, Gimeno RE, Milicevic Z, Robins D, et al.2018 Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 392 21802193. (https://doi.org/10.1016/S0140-6736(1832260-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S, Brody L, Anastasovska J, Ghourab S, Hankir M, Zhang S, et al.2014 The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nature Communications 5 3611. (https://doi.org/10.1038/ncomms4611)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fujino T, Kondo J, Ishikawa M, Morikawa K & & Yamamoto TT 2001 Acetyl-CoA synthetase 2, a mitochondrial matrix enzyme involved in the oxidation of acetate. Journal of Biological Chemistry 276 1142011426. (https://doi.org/10.1074/jbc.M008782200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 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 15091517. (https://doi.org/10.2337/db08-1637)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gasbjerg LS, Helsted MM, Hartmann B, Jensen MH, Gabe MBN, Sparre-Ulrich AH, Veedfald S, Stensen S, Lanng AR, Bergmann NC, et al.2019 Separate and combined glucometabolic effects of endogenous glucose-dependent insulinotropic polypeptide and glucagon-like peptide 1 in healthy individuals. Diabetes 68 906917. (https://doi.org/10.2337/db18-1123)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gijsman HJ, Scarnà A, Harmer CJ, Mctavish SB, Odontiadis J, Cowen PJ & & Goodwin GM 2002 A dose-finding study on the effects of branch chain amino acids on surrogate markers of brain dopamine function. Psychopharmacology 160 192197. (https://doi.org/10.1007/s00213-001-0970-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gojda J & & Cahova M 2021 Gut microbiota as the link between elevated BCAA serum levels and insulin resistance. Biomolecules 11. (https://doi.org/10.3390/biom11101414)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goswami C, Iwasaki Y & & Yada T 2018 Short-chain fatty acids suppress food intake by activating vagal afferent neurons. Journal of Nutritional Biochemistry 57 130135. (https://doi.org/10.1016/j.jnutbio.2018.03.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Grandt D, Schimiczek M, Beglinger C, Layer P, Goebell H, Eysselein VE & & Reeve JR 1994 Two molecular forms of peptide YY (PYY) are abundant in human blood: characterization of a radioimmunoassay recognizing PYY 1–36 and PYY 3–36. Regulatory Peptides 51 151159. (https://doi.org/10.1016/0167-0115(9490204-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Grandt D, Siewert J, Sieburg B, Al Tai O, Schimiczek M, Goebell H, Layer P, Eysselein VE, Reeve JR & & Müller MK 1995 Peptide YY inhibits exocrine pancreatic secretion in isolated perfused rat pancreas by Y1 receptors. Pancreas 10 180186. (https://doi.org/10.1097/00006676-199503000-00012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gribble FM & & Reimann F 2019 Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nature Reviews. Endocrinology 15 226237. (https://doi.org/10.1038/s41574-019-0168-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Grosse J, Heffron H, Burling K, Akhter Hossain M, Habib AM, Rogers GJ, Richards P, Larder R, Rimmington D, Adriaenssens AA, et al.2014 Insulin-like peptide 5 is an orexigenic gastrointestinal hormone. Proceedings of the National Academy of Sciences of the United States of America 111 1113311138. (https://doi.org/10.1073/pnas.1411413111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gui X & & Carraway RE 2001 Enhancement of jejunal absorption of conjugated bile acid by neurotensin in rats. Gastroenterology 120 151160. (https://doi.org/10.1053/gast.2001.20876)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo Z, Zhang Y, Liu C, Youn JY & & Cai H 2021 Toll-like receptor 2 (TLR2) knockout abrogates diabetic and obese phenotypes while restoring endothelial function via inhibition of NOX1. Diabetes 70 21072119. (https://doi.org/10.2337/db20-0591)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gustafson EL, Smith KE, Durkin MM, Walker MW, Gerald C, Weinshank R & & Branchek TA 1997 Distribution of the neuropeptide Y Y2 receptor mRNA in rat central nervous system. Brain Research. Molecular Brain Research 46 223235. (https://doi.org/10.1016/s0169-328x(9700017-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guzior DV & & Quinn RA 2021 Review: microbial transformations of human bile acids. Microbiome 9 140. (https://doi.org/10.1186/s40168-021-01101-1)

  • Habib AM, Richards P, Cairns LS, Rogers GJ, Bannon CA, Parker HE, Morley TC, Yeo GS, Reimann F & & Gribble FM 2012 Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry. Endocrinology 153 30543065. (https://doi.org/10.1210/en.2011-2170)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Habib AM, Richards P, Rogers GJ, Reimann F & & Gribble FM 2013 Co-localisation and secretion of glucagon-like peptide 1 and peptide YY from primary cultured human L cells. Diabetologia 56 14131416. (https://doi.org/10.1007/s00125-013-2887-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansen M, Scheltema MJ, Sonne DP, Hansen JS, Sperling M, Rehfeld JF, Holst JJ, Vilsbøll T & & Knop FK 2016 Effect of chenodeoxycholic acid and the bile acid sequestrant colesevelam on glucagon-like peptide-1 secretion. Diabetes, Obesity and Metabolism 18 571580. (https://doi.org/10.1111/dom.12648)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hansson B, Medina A, Fryklund C, Fex M & & Stenkula KG 2016 Serotonin (5-HT) and 5-HT2A receptor agonists suppress lipolysis in primary rat adipose cells. Biochemical and Biophysical Research Communications 474 357363. (https://doi.org/10.1016/j.bbrc.2016.04.110)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hare KJ, Vilsbøll T, Asmar M, Deacon CF, Knop FK & & Holst JJ 2010 The glucagonostatic and insulinotropic effects of glucagon-like peptide 1 contribute equally to its glucose-lowering action. Diabetes 59 17651770. (https://doi.org/10.2337/db09-1414)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hartmann B, Johnsen AH, Orskov C, Adelhorst K, Thim L & & Holst JJ 2000 Structure, measurement, and secretion of human glucagon-like peptide-2. Peptides 21 7380. (https://doi.org/10.1016/s0196-9781(9900176-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haub S, Ritze Y, Ladel I, Saum K, Hubert A, Spruss A, Trautwein C & & Bischoff SC 2011 Serotonin receptor type 3 antagonists improve obesity-associated fatty liver disease in mice. Journal of Pharmacology and Experimental Therapeutics 339 790798. (https://doi.org/10.1124/jpet.111.181834)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heisel T, Montassier E, Johnson A, Al-Ghalith G, Lin YW, Wei LN, Knights D & & Gale CA 2017 High-fat diet changes fungal microbiomes and interkingdom relationships in the murine gut. mSphere 2. (https://doi.org/10.1128/mSphere.00351-17)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heiss CN, Mannerås-Holm L, Lee YS, Serrano-Lobo J, Håkansson Gladh A, Seeley RJ, Drucker DJ, Bäckhed F & & Olofsson LE 2021 The gut microbiota regulates hypothalamic inflammation and leptin sensitivity in Western diet-fed mice via a GLP-1R-dependent mechanism. Cell Reports 35 109163. (https://doi.org/10.1016/j.celrep.2021.109163)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Henriksen GL, Ketchum NS, Michalek JE & & Swaby JA 1997 Serum dioxin and diabetes mellitus in veterans of Operation Ranch Hand. Epidemiology 8 252258. (https://doi.org/10.1097/00001648-199705000-00005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heredia DJ, Gershon MD, Koh SD, Corrigan RD, Okamoto T & & Smith TK 2013 Important role of mucosal serotonin in colonic propulsion and peristaltic reflexes: in vitro analyses in mice lacking tryptophan hydroxylase 1. Journal of Physiology 591 59395957. (https://doi.org/10.1113/jphysiol.2013.256230)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holecek M, Siman P, Vodenicarovova M & & Kandar R 2016 Alterations in protein and amino acid metabolism in rats fed a branched-chain amino acid- or leucine-enriched diet during postprandial and postabsorptive states. Nutrition and Metabolism 13 12. (https://doi.org/10.1186/s12986-016-0072-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hotamisligil GS, Arner P, Caro JF, Atkinson RL & & Spiegelman BM 1995 Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. Journal of Clinical Investigation 95 24092415. (https://doi.org/10.1172/JCI117936)

    • PubMed
    • Search Google Scholar
    • Export Citation