Next Article in Journal
Reduced Granule Cell Proliferation and Molecular Dysregulation in the Cerebellum of Lysosomal Acid Phosphatase 2 (ACP2) Mutant Mice
Next Article in Special Issue
Lectin-Mediated Bacterial Modulation by the Intestinal Nematode Ascaris suum
Previous Article in Journal
Cannabidiol and Other Cannabinoids in Demyelinating Diseases
Previous Article in Special Issue
Interaction of Intestinal Bacteria with Human Rotavirus during Infection in Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of the Gut Microbiota in the Gut–Brain Axis in Obesity: Mechanisms and Future Implications

1
Department of Endocrinology and Metabolism, Amsterdam UMC, location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
2
Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(6), 2993; https://doi.org/10.3390/ijms22062993
Submission received: 28 February 2021 / Revised: 12 March 2021 / Accepted: 13 March 2021 / Published: 15 March 2021
(This article belongs to the Special Issue Strategies Targeting Microbiota Modulation)

Abstract

:
Interaction between the gut and the brain is essential for energy homeostasis. In obesity, this homeostasis is disrupted, leading to a positive energy balance and weight gain. Obesity is a global epidemic that affects individual health and strains the socioeconomic system. Microbial dysbiosis has long been reported in obesity and obesity-related disorders. More recent literature has focused on the interaction of the gut microbiota and its metabolites on human brain and behavior. Developing strategies that target the gut microbiota could be a future approach for the treatment of obesity. Here, we review the microbiota–gut–brain axis and possible therapeutic options.

1. Introduction

The gut microbiota consist of trillions of microorganisms within the gastrointestinal tract and contain over 100 times more genes than the human genome [1]. Diverse and stable microbiota is crucial for human health and essential for gut homeostasis, metabolism of nutrients, supply of microbial metabolites, and defense against pathogens [2]. Microbial dysbiosis, defined as a shift in the composition of bacteria from a diverse to a maladaptive and pathogenic profile, is associated with several pathologic conditions (e.g., obesity, type 2 diabetes mellitus, and cardiovascular disease) [3,4,5,6]. Over the last decades, the prevalence of obesity, defined as body mass index (BMI) ≥30 kg/m2, has dramatically increased. According to the World Health Organization, 650 million adults had obesity in 2016 [7]. Obesity occurs when energy intake exceeds energy expenditure and is a complex multifactorial condition that includes both biological and environmental factors. Obesity is a major health problem and is associated with increased morbidity and mortality [8,9] as well as high socioeconomic burden [10]. This has led to extensive research. However, currently besides bariatric surgery, successful treatment options are still limited [11].
Obesity is characterized by perturbations in homeostatic and hedonic feeding behavior [12]. During feeding, there is a complex feedback system between the gut and the brain, also known as the gut–brain axis. The importance of the gut–brain axis in energy homeostasis has long been reported [13,14]. More recent research has started to uncover the link between the gut microbiota and the central nervous system (CNS) [15]. Studies with germ-free mice have shown that the gut microbiota is essential for brain development and behavior [16,17].
Here, we aim to review the interaction between the gut microbiota and CNS in obesity and will highlight several underlying mechanisms and possible treatment strategies.

2. Gut Microbiota in Obesity

In healthy adults, the gut microbiota is dominated by organisms from the Bacteroidetes and Firmicutes phyla [18]. In individuals with obesity, lower relative proportions of Bacteroidetes and higher proportions Firmicutes have been observed compared with lean controls, with a significant increase of Bacteroidetes after weight loss [3]. Additionally, comparison of twins with normal weight or obesity showed a lower bacterial diversity and Bacteroidetes proportion but higher Actinobacteria in obesity. However, there was no significant difference in Firmicutes proportion [19]. Conversely, a meta-analysis did not find a clear association between the Firmicutes/Bacteroidetes ratio and obesity, underscoring a potentially more complex system involved [20]. Nevertheless, several studies have reported that weight loss can reciprocally influence the gut microbiota composition [21,22]. In this regard, it has been suggested that in individuals with obesity, the gut microbiota may be more efficient in harvesting energy, leading to more fat storage compared with lean individuals [23,24]. Indeed, research in mice revealed that colonization of germ-free mice with microbiota of conventionally raised mice significantly increased body fat despite reduced food intake [23]. Moreover, the gut microbiota from human or mice donors with obesity resulted in greater increase in body fat than the microbiota from lean donors in mice [24,25]. However, findings are inconsistent, and it is not clear if and how this will translate to humans [26].
Obesity is characterized by the presence of low-grade inflammation, which increases the risk for comorbidities, such as insulin resistance, cardiovascular disease, and cancer [27,28]. The precise mechanisms behind this inflammatory state are still largely unknown, but several factors have been implicated, including the gut microbiota. It is thought that changes in gut microbiota composition in obesity lead to increased gut permeability [29]. Disruption of this intestinal barrier leads to passage of components, such as lipopolysaccharide (LPS), an endotoxin produced by the lysis of Gram-negative bacteria, into the systemic circulation. These elevated plasma levels of LPS can initiate inflammatory cascades in adipose tissue, leading to the production of proinflammatory cytokines [30], and can also induce neuroinflammation [31]. It has indeed been shown that humans with obesity have significantly higher plasma LPS levels after a high-fat meal compared with lean controls [32], further underscoring the potential contributing role of the gut microbiota in the low-grade inflammatory state in human obesity.

3. The Microbiota–Gut–Brain Axis

The gut–brain axis is a complex bidirectional pathway that is essential for metabolic homeostasis. The gut can interact with the CNS to transfer information on nutritional status through a variety of mechanisms, including enteroendocrine cells (EECs), the vagus nerve (VN), and the enteric nervous system (ENS). Microbial-produced metabolites can modulate these signals. In this section, we will provide a brief overview of the microbiota–gut–brain axis.
During feeding, several neuroendocrine mechanisms are activated. EECs are located throughout the epithelium of the gut and respond to nutrient and mechanical stimuli by secreting hormones and neurotransmitters that have a key role in metabolism, including serotonin (5-hydroxytryptamine), ghrelin, cholecystokinin (CCK), peptide YY (PYY), and glucagon-like peptide 1 (GLP-1) [33,34]. These endocrine hormones mediate their effect on secretion (insulin, gastric acid, and bile acids), gut motility, and food intake through vagal afferent neurons or ENS or through release into the bloodstream [35,36,37]. Indeed, gut-microbiota-derived metabolites (e.g., short-chain fatty acids (SCFAs)) can modify the release of these hormones and neurotransmitters [38,39].
Beyond hormonal signaling, neuronal orchestration of the gut–brain axis is gaining more and more attention. In this regard, the VN is the longest nerve in the human body and plays a key role in gut–brain signaling [40]. It is a mixed nerve containing approximately 80% afferent and 20% efferent fibers that transmit information from the viscera to the brain and vice versa [41]. Signals from the VN are integrated in the nucleus of the solitary tract (NTS), which then projects signals to areas important for energy balance, such as the hypothalamic arcuate nucleus (ARC), the parabrachial nucleus, and the dorsal motor nucleus [42,43,44]. The VN is involved in food intake and gastrointestinal motility and has anti-inflammatory properties [45]. Its afferent fibers can be activated by gastrointestinal tract (GIT) distension, hormones, and peptides released from the EECs or microbial metabolites [46]. The VN can also be directly inhibited by ghrelin and PYY or stimulated by CCK [47,48,49]. Several studies have shown an association between microbial dysbiosis in obesity and VN innervation and signaling [50,51]. Moreover, older studies have suggested that (gastric) vagotomy in humans affects body weight, thus underscoring the driving role of VN in human metabolism and food intake [52], a finding that more recently has been endorsed in mice [53].
Connected to the VN is the enteric nervous system, which comprises a neural network located throughout the GIT, which controls gut motility, blood flow, and secretion. It communicates with the CNS (e.g., via the VN) but is also capable of detecting and integrating information and functioning independently [54,55]. For example, animal studies have shown that with a severed vagus nerve, the ENS continues to function [56]. The gut microbiota may signal the ENS directly through microbial-produced neurotransmitters and SCFAs or indirectly through the effect of bacterial metabolites on the EECs. Studies in germ-free mice have shown that the gut microbiota is involved in the maturation of the ENS, and also during adult age, ENS might be sensitive to microbial intervention [57,58,59].
Moreover, the dietary nutrients that are converted into plasma metabolites by the gut microbiota include short-chain fatty acids (SCFAs lactate, butyrate, propionate, acetate, and succinate), as well as gamma-aminobutyric acid (GABA), dopamine, and serotonin, which all have important functions in the gut–brain axis. Specifically, the SCFAs that are derived from dietary fibers are essential signaling molecules by binding to G protein-coupled receptors (GPR), GPR43, and GPR41 [60]. They have a role in satiety, may increase energy expenditure [61], and have also shown to have a possible role in CNS inflammation [62]. Additionally, butyrate is an important energy source for the colonocytes, but also has a strong epigenetic effect (via methylation as HDAC inhibitor) [63]. Although the majority of intestinally produced SCFAs are consumed in the intestine, a small part can stimulate the secretion of PYY and GLP-1 through interaction with EECs [64,65], thus indirectly affecting satiety. Additionally, SCFAs can suppress food intake by stimulating the VN [66] and centrally after passing the blood–brain barrier (BBB) through the circulatory system. A study in mice showed that acetate can cross the BBB and induce anorexigenic signals in the hypothalamus [67]. Besides modulating satiety, studies in obese mice also found that acetate and butyrate can increase thermogenesis and energy expenditure [61,68]. Additionally, a study in mice suggested that SCFA-mediated GPR43 activation inhibits fat accumulation in adipose tissue and promotes metabolism of glucose in the liver and muscle [69], and propionate and butyrate have been demonstrated to induce intestinal gluconeogenesis in rats [70]. In this regard, research providing insight into the role of SCFAs in obesity has been performed. For example, a study with dietary supplementation of SCFAs in mice prevented high-fat-diet-induced weight gain [71]. In humans, individuals with obesity have higher fecal concentrations of SCFAs compared with lean individuals [72,73]. However, the relevance of fecal concentrations is debatable as it does not directly correlate with circulating SCFAs [74]. Several human trials have suggested a positive effect of SCFAs on energy expenditure, glucose metabolism, and appetite [75,76,77]. Furthermore, ex vivo treatment of human adipose tissue biopsies with propionic acid increased the expression of the anorexigenic hormone leptin and reduced inflammatory factors [78,79]. Most studies in humans are associative, and our group recently showed that oral administration of SCFA butyrate does not improve glucose metabolism or vagal tone in metabolic syndrome subjects [76,80]. Additional human trials are needed to further identify the possible role of other SCFAs as intervention in obesity and obesity-related metabolic disorders.
In summary, there is a complex interaction between the gut and the CNS to maintain energy homeostasis, which can be modulated by the microbiota and its metabolites (Figure 1).

4. Gut Microbiota and Metabolites and Feeding Behavior

The consumption of food is essential for survival. Signals of hunger and satiety motivate behavior to obtain an adequate intake of nutrients. In vertebrates, food intake is regulated by two pathways: the homeostatic and hedonic pathways. The homeostatic pathway controls energy balance and energy metabolism by mediators such as leptin and ghrelin, while the hedonic pathway is involved in the motivational aspects of food (i.e., pleasure and reward) [81]. Even though these are separate pathways, interaction between the two as well as with the gut microbiota has been reported [82].

4.1. Homeostatic Pathway

Homeostatic control of feeding is focused on energy homeostasis. Before, during, or after ingesting a meal, neural, substrate-driven, and hormonal pathways signal the brain on the nutrient status. The ARC plays a central role in food intake pathways and receives input from circulating metabolic substrates that pass the BBB and through vagal afferents, which activate the NTS, inducing a signal to the ARC [83,84]. There are two major neuronal populations that play an important role in food intake and energy balance. One group consists of the orexigenic GABAergic neurons that express neuropeptide Y (NPY) and agouti-related protein (AgRP), which during acute activation increase food intake and reduce energy expenditure [85]. The second group consists of glutamatergic anorexigenic neurons that express pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) [86]. Chronic POMC neuron activation suppresses feeding behavior. Activation of POMC neurons leads to alpha-melanocyte-stimulating hormone (α-MSH) release, which then activates melanocortin-4-receptor (MC4R) and the melanocortin satiety pathway [87]. AgRP is an inverse agonist of the MC4R [88].
With regard to hormonal regulation of the homeostatic pathway ghrelin, an orexigenic hormone, mainly produced in the stomach by the EECs, rises before meals and is suppressed after food ingestion [89,90]. Ghrelin increases gastric motility and gastric acid secretion and simulates food intake, but also is involved in glucose metabolism, learning and memory, taste, and reward [91]. Moreover, ghrelin passes the BBB and activates NPY/AgRP neurons. In addition, ghrelin mediates GABA release from the NPY/AgRP neurons, leading to inhibition of POMC/CART. The effect of ghrelin on reward-driven feeding has more recently been described [92,93]. In obese individuals, plasma ghrelin concentrations are lower than in lean controls, suggesting that these plasma concentrations do not contribute to the increased food intake [94]. In line with this, leptin, an anorexigenic hormone, is produced by white adipose tissue, and circulating plasma concentrations are related to body fat mass [95]. Leptin is a marker for long-term energy balance and signals the hypothalamus after crossing the BBB [96]. A decrease in plasma leptin concentrations has also been described as a hunger signal [97,98]. Leptin inhibits hunger signals by binding to leptin receptors in the ARC, leading to activation of POMC/CART neurons and inhibition of NPY/AgRP neurons [99]. Plasma leptin concentrations are higher in individuals with obesity compared with lean controls [95]. However, this does not lead to reduced food intake, which suggests that there is resistance to the effects of leptin. This is in line with studies with exogenous administration of leptin in obesity that have failed to show weight loss [100]. Possible mechanisms, such as reduced transport across the BBB, have been hypothesized [101]. However, leptin resistance has been observed with intact leptin transport across the BBB [102]. Impaired hypothalamic signaling has been implied to underlie leptin resistance in obesity [103,104]. Therefore, although leptin is a key signaling factor during starvation, its precise role in overfeeding is still unclear [105].
Although the involved gut microbiota are not known, previous studies have shown that specific bacterial strains are associated with intestinal hormone concentrations. For example, quantities of Bifidobacterium and Lactobacillus negatively correlated with ghrelin concentrations and positively correlated with plasma leptin concentrations [106]. Furthermore, in Helicobacter pylori-infected patients who underwent eradication therapy, an increase in the Bacteroidetes/Firmicutes ratio significantly correlated with a decrease in ghrelin concentrations, suggesting that changes in the microbiota composition could affect plasma ghrelin concentrations [107]. Additionally, a recent study showed that SCFAs were able to attenuate ghrelin-mediated signaling through the growth hormone secretagogue receptor-1a [108]. In this regard, in mice SCFAs were found to stimulate leptin production in cultured adipocyte cells through the activation of GPR41 and also increased circulating leptin concentrations after oral administration [109]. Germ-free mice have shown to have higher leptin sensitivity, resulting in greater weight loss after leptin injection, compared with conventionally raised mice on a chow diet [110], and hypothalamic expression of suppressor of cytokine signaling 3, which is associated with leptin resistance, was higher in the conventionally raised mice [110]. A recent study found that supplementation with Lactobacillus rhamnosus GG for 10 weeks decreased the proportion of Proteobacteria and restored exogenous leptin responsiveness in high-fat-diet mice [111].
Besides orexigenic hormonal gut–brain circuits, the previously mentioned anorexigenic peptides PYY, CCK, and GLP-1 (mainly produced by the intestinal EECs and postprandially released) also play an important regulatory role. After a meal, an increase of plasma concentrations of PYY inhibits NPY/AgRP by binding to the neuropeptide Y receptor type 2 (Y2R). This removes their inhibition on the POMC neurons, which reduces food intake [112,113]. GLP-1 plays an important role in glucose regulation by enhancing glucose-induced insulin secretion and lowering glucagon secretion, and therefore, GLP-1 receptor agonists (GLP-1RAs) have been developed as a treatment for type 2 diabetes [114]. GLP-1RAs have been shown to also induce weight loss by the reduction of food intake and gastric emptying [115,116]. In rodents, GLP-1RAs exert this effect by activating POMC/CART neurons and indirectly inhibiting NPY/AgRP through GABAergic transmission [117]. Ongoing studies are currently investigating the possibility of weight management with GLP-1RAs [118], and a recent randomized trial has reported a significant reduction in weight after treatment with the GLP-1 analogue semaglutide once weekly for 68 weeks [119]. Furthermore, GLP-1 exerts its effect on satiety and gastric emptying through GLP-1 receptors in the vagal nerve [120].
Moreover, CCK secretion is regulated by GLP-1, and CCK is involved in digestion by stimulating gallbladder contraction, pancreatic secretion, delaying gastric emptying and increasing satiety [121]. It was the first gut hormone that was shown to induce satiety and decrease food intake in humans [49,122]. CCK reduces food intake through activation of the vagus nerve after binding the vagal CCK-A receptors [123,124]. However, in humans, stimulation with a CCK-A receptor agonist did not result in weight loss [125], thus limiting its therapeutic benefit. A recent study in mice showed that colonization with dysbiotic microbiota led to decreased vagal innervation at the NTS, which was associated with loss of CCK-induced satiety [126]. To our knowledge, currently, little is known about the possible effects of the gut microbiota on the regulation of CCK release. However, humans with obesity who underwent Roux-en-Y gastric bypass surgery had no changes in plasma CCK even though their microbial composition significantly changed [127].
In this regard, there is more evidence suggesting that the next-generation (gut microbiota produced) intestinally derived metabolites do play a larger role in the regulation of host appetite and metabolism [128]. For example, caseinolytic protease B produced by Escherichia coli is an antigen-mimetic of α-MSH [129]. As described above, α-MSH has a central role, but local signaling in the gut has also been suggested after a study showed stimulation of the MC4R in the gut EECs induced PYY and GLP-1 release [130]. Another study found that in mice, α-MSH was upregulated after nutrient-induced E. coli growth and led to increased plasma PYY and GLP-1 release and activated POMC neurons [131]. Finally, imidazole propionate (ImP, derived from gut-microbiota-processed dietary histidine) results in insulin resistance and subsequent type 2 diabetes [132,133]. Moreover, recent data showed that ImP is present in the forebrains of mice, thus underscoring that it can pass the BBB and therefore affect neurodevelopmental trajectories, including the gut–brain axis [134,135]. Moreover, histidine, the precursor to ImP, has been suggested to affect GLP-1 secretion [136]. Taken together, the homeostatic pathway of feeding is a complex interaction between the periphery and the CNS. Although more research is needed, there is accumulating evidence of an interplay with the gut microbiota and the secretion and function of endogenous (incretin) hormonal potentiation.

4.2. Hedonic Pathway and Microbiota

An abnormal hedonic drive and enhanced motivation, both elements of the reward system, have also been shown to play a major role in obesity [137,138,139]. The mesolimbic dopamine system centered in the striatum modulates reward and has close connection with the homeostatic system [12]. Food intake stimulates dopamine release, and the rewarding sensation can promote feeding behavior [140]. Again, leptin and ghrelin have both been identified to affect mesolimbic dopaminergic activity [93,141]. Moreover, lower binding potential of striatal dopamine D2 receptors (D2R) in humans with obesity compared with lean controls has been reported in both animal studies and single photon emission computed tomography (SPECT) studies in humans [142,143]. Additionally, it was found that individuals with obesity who were presented pictures of high-calorie food showed increased activation of regions involved in reward and motivation [144] but had less activation from actual food consumption [145]. It has been hypothesized that decreased dopaminergic signaling leads to overconsumption to compensate for lower reward feedback during feeding [146,147]. However, there is also evidence showing hyper-responsivity of reward regions, thus increasing the risk for overeating, which suggests that the response to food cues in obesity is a dynamic model that shifts over time [148].
Dopamine is involved in neurological processes, including cognition, motor function, learning, and reward, and also regulates various peripheral functions, such as insulin release [149]. In the gastrointestinal tract, dopamine reduces gut motility, modulates mucosal blood flow, and stimulates exocrine secretions [150]. A substantial portion of peripheral circulating dopamine is produced in the gastrointestinal tract [151]. Altered gut microbiota composition has been reported in several central disorders with dysregulated dopaminergic transmission, such as anxiety, depression, and Parkinson’s disease [152,153]. It is unknown whether peripherally produced dopamine has direct effects on dopaminergic pathways in the brain. It has been hypothesized that the proinflammatory state in microbial dysbiosis could contribute to these disorders [31,152]. However, evidence on a causal effect in humans is scarce.
An overview of the homeostatic and hedonic pathways and possible ways of interaction from the gut microbiota is summarized in Figure 2.

5. Other Neurotransmitters

Besides dopamine, the gut bacteria can modulate and produce several neurotransmitters, including GABA and serotonin. In this section, we will briefly summarize their function and how gut microbiota composition may affect them.

5.1. Serotonin

Serotonin has a key role both in the brain and peripherally, including modulating satiety, anxiety, and mood and stimulating peristalsis, secretion, and vasodilation, respectively [154,155,156]. There is also evidence suggesting the role of serotonin in the regulation of glucose and lipid metabolism [157,158]. Serotonin cannot pass the BBB; thus production from tryptophan takes place separately within the periphery and the CNS. Tryptophan is an essential amino acid obtained from dietary proteins and is a precursor to several metabolites, including serotonin [159]. It is well established that the majority of peripheral serotonin is produced in the enterochromaffin cells in the (small) intestinal EECs [156,160]. Within the hypothalamus, serotonin mediates its food intake suppressant effects through inhibition of NPY/AgRP and activation of POMC neurons. However, the complete interaction between serotonin and satiety signals is not yet fully understood [155]. The serotonin 2C receptor agonist, lorcaserin, was found to significantly improve weight loss in individuals with obesity [161], but was recently withdrawn from the market after reports of increased cancer risk [162]. In obesity, evidence of decreased central serotonin signaling has been reported [147].
In the periphery, increased circulating plasma serotonin concentrations have been found in individuals with obesity [163]. Crane et al. found that inhibition of peripheral serotonin synthesis enhanced thermogenesis in adipose tissue, causing increased energy expenditure and protection against high-fat-diet-induced obesity in mice [164]. Gut microbes have a regulatory effect on the peripheral concentrations of serotonin [165]. The role of serotonin in obesity is complex, and the role the microbiota have on central serotonin is still unclear. However, our recent pilot study using SPECT imaging in humans suggested that the gut microbiota can directly or indirectly affect brain serotonin (and dopamine) transporter binding potential in humans with obesity via sympathetic tone [80].

5.2. GABA

GABA is the main inhibitory neurotransmitter. GABA is produced from its counter glutamate, which functions as an excitatory neurotransmitter. GABA has been implicated in the hypothalamic control of food intake [166,167]. Peripheral GABA production can be stimulated by several bacteria of the microbiota, such as Bifidobacterium and Lactobacillus [168]. Animal studies have indicated that treatment modulating the microbiota might improve metabolic health and behavior. For example, treatment with Lactobacillus rhamnosus induced alterations in GABA mRNA in the brain and reduced anxiety- and depression-related behavior via the regulation of vagal nerve tone [169], and obese mice that received the GABA-producing Lactobacillus brevis improved metabolically and had less depressive-like behavior [170]. Finally, we recently showed that in humans with obesity treated with fecal microbiota transplantation (FMT) from lean donors, plasma GABA concentrations increased [171].

6. Therapeutic Options for Modulation of the Gut–Brain Axis via Microbial Communities

Whereas gut microbiota dysbiosis seems to play a role in obesity, possibly affecting fat storage and inflammation and modulating the gut–brain axis, we now aim to discuss novel therapeutic options.
Diet plays an important role in the composition and diversity of the gut microbiome and has been identified to affect colonization, maturation, and microbiome diversity [172]. A population-based study revealed significant associations between diet and gut microbiota variation [173]. Especially Western-style diets (characterized by high intake of saturated fats and sucrose and low amounts of fiber) have been associated with metabolic disease and obesity [174] and can lead to shifts in the microbiota composition [175]. For example, high-fat diets have shown to alter the gut microbiota and increase intestinal pathogens in both animal and human studies [176,177]. However, adverse diet-related effects are likely to be person specific as subjects with higher baseline microbial diversity seemed to be more resistant to adverse microbial changes induced by high-fat [178] and sugar-containing diets [179]. In contrast, dietary nondigestible fibers (responsible for SCFA production) promote intestinal health. A meta-analysis found that a dietary fiber intervention resulted in higher levels of Bifidobacterium spp. and Lactobacillus spp., but did not affect SCFA concentrations [180]. Future studies are required to improve our understanding of the underlying mechanisms of how specific dietary components affect the gut microbiota composition and subsequent health consequences.
In this regard, interventions with probiotics and prebiotics and FMT are discussed.

6.1. Probiotics

Probiotics are defined as “live microorganisms that confer a health benefit on the host when administered in adequate amounts” [181]. Intervention with probiotics might be an option for the treatment of obesity and obesity-related disorders, although it is a challenge to get viable bacterial strains into the gut because of the low pH of the stomach. Nevertheless, studies in animals and humans have shown that probiotics are associated with weight change [182,183]. Even though study results are not always consistent, a large meta-analysis showed that probiotic intervention was associated with a reduction in body weight in subjects with obesity [184]. The most commonly used probiotics are Bifidobacterium and Lactobacillus genera; however, their effect on obesity seems to be species and strain specific [183]. In line with this, oral administration with probiotics for 4 weeks was associated with affected midbrain connectivity assessed with functional magnetic resonance imaging in healthy female subjects [185]. More recently, Akkermansia muciniphila, a strain that has been negatively associated with obesity [186], was tested in a randomized placebo-controlled study. Although it was well tolerated and improved insulin sensitivity, there was no effect on fasting glucose or body weight when compared with placebo. Additionally, this study did not investigate aspects of the gut–brain axis [187]. All in all, it is too early to conclude that probiotics can help to improve metabolism and body weight in humans.

6.2. Prebiotics

Prebiotics are defined as “selectively fermented ingredients that result in specific changes in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefits upon host health” [181]. Prebiotics are present in plants used for food or can be industrially produced and may be considered a future target against obesity [188]. Treatment with prebiotics in human trials led to a reduction of metabolic endotoxemia [189,190] and increased SCFA production [191] in individuals with obesity. The prebiotics inulin-type fructans have been shown to increase the growth of the beneficial lactobacilli and bifidobacteria and increase (postprandial) GLP-1 and PYY response with a concomitant decreased serum ghrelin concentration, which could affect food intake [192,193]. However, human trials studying the effect of prebiotics on weight loss have had contradictory results [194,195]. Even though accumulating data are becoming available, the precise mechanisms between prebiotics and obesity are still unclear.

6.3. Fecal Microbiota Transplantation

Altering the gut microbiota through FMT has increasingly gained interest as a possible treatment for chronic diseases. During FMT, the stool of a heathy donor is infused into the patient’s intestinal tract. FMT has been successfully used as treatment for recurrent Clostridium difficile infections [196]. More recent studies found a possible role of fresh donor FMT in the treatment of peripheral insulin resistance [171], with a specific important role in the metabolic status of the FMT donor [197]. The transplantation of fecal microbiota from twins discordant for obesity into germ-free mice led to increased body and fat mass if the donor was obese [25]. In contrast, two placebo-controlled pilot studies in humans with obesity found that treatment with FMT capsules from lean donors did not result in significant weight loss [198] or improve metabolic health [199]. Likewise, a randomized placebo-controlled trial in adolescents with obesity did not find an effect of FMT on weight loss [200]. However, exploratory post hoc analyses did show a significant decrease in subjects with metabolic syndrome and low baseline microbiota diversity [201]. As these two studies used a different route of administering donor feces (frozen capsules instead of duodenal infusion of fresh feces potentially losing beneficial strains) and did not assess the metabolic status of the FMT donor, future studies will have to shed light on the efficacy of donor FMT on human metabolism and the CNS.

7. Conclusions

Currently, there are limited effective treatments available for obesity. There is accumulating evidence of the role of the gut microbiota in obesity and its interaction with the gut–brain axis. Manipulation of the gut microbiota might be a novel therapeutic option in treating obesity. However, mechanisms linking the gut microbiota and the CNS in obesity are complex. Better understanding of these mechanisms could lead to the development of microbiota-targeted therapies, such as probiotics, prebiotics, and FMT.

Funding

Jamie van Son and Max Nieuwdorp are supported by a Novo Nordisk Foundation CAMIT grant 2018. Max Nieuwdorp is supported by a ZonMw VICI (grant number 09150182010020). None of the funding bodies had any role in writing of the manuscript of selection of the literature.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

Max Nieuwdorp is in the Scientific Advisory Board of Caelus Pharmaceuticals, the Netherlands, and Kaleido Biosciences, USA. None of these are directly relevant to the current paper. There are no patents, products in development, or marketed products to declare. The other authors declare no competing financial interests.

References

  1. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [Green Version]
  2. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
  3. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
  4. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
  5. Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal, J.M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J.; et al. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Karlsson, F.H.; Fak, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Backhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012, 3, 1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. World Health Organization. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 1 March 2021).
  8. Berrington de Gonzalez, A.; Hartge, P.; Cerhan, J.R.; Flint, A.J.; Hannan, L.; MacInnis, R.J.; Moore, S.C.; Tobias, G.S.; Anton-Culver, H.; Freeman, L.B.; et al. Body-mass index and mortality among 1.46 million white adults. N. Engl. J. Med. 2010, 363, 2211–2219. [Google Scholar] [CrossRef] [Green Version]
  9. Abdelaal, M.; le Roux, C.W.; Docherty, N.G. Morbidity and mortality associated with obesity. Ann. Transl. Med. 2017, 5, 161. [Google Scholar] [CrossRef] [Green Version]
  10. Wang, Y.C.; McPherson, K.; Marsh, T.; Gortmaker, S.L.; Brown, M. Health and economic burden of the projected obesity trends in the USA and the UK. Lancet 2011, 378, 815–825. [Google Scholar] [CrossRef]
  11. Kushner, R.F. Weight Loss Strategies for Treatment of Obesity: Lifestyle Management and Pharmacotherapy. Prog. Cardiovasc. Dis. 2018, 61, 246–252. [Google Scholar] [CrossRef]
  12. Berthoud, H.R.; Munzberg, H.; Morrison, C.D. Blaming the Brain for Obesity: Integration of Hedonic and Homeostatic Mechanisms. Gastroenterology 2017, 152, 1728–1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hussain, S.S.; Bloom, S.R. The regulation of food intake by the gut-brain axis: Implications for obesity. Int. J. Obes. 2013, 37, 625–633. [Google Scholar] [CrossRef] [Green Version]
  14. Naslund, E.; Hellstrom, P.M. Appetite signaling: From gut peptides and enteric nerves to brain. Physiol. Behav. 2007, 92, 256–262. [Google Scholar] [CrossRef]
  15. Kuwahara, A.; Matsuda, K.; Kuwahara, Y.; Asano, S.; Inui, T.; Marunaka, Y. Microbiota-gut-brain axis: Enteroendocrine cells and the enteric nervous system form an interface between the microbiota and the central nervous system. Biomed. Res. 2020, 41, 199–216. [Google Scholar] [CrossRef] [PubMed]
  16. Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [Green Version]
  17. Diaz Heijtz, R.; Wang, S.; Anuar, F.; Qian, Y.; Bjorkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [Green Version]
  19. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [Green Version]
  20. Sze, M.A.; Schloss, P.D. Looking for a Signal in the Noise: Revisiting Obesity and the Microbiome. mBio 2016, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Santacruz, A.; Marcos, A.; Warnberg, J.; Marti, A.; Martin-Matillas, M.; Campoy, C.; Moreno, L.A.; Veiga, O.; Redondo-Figuero, C.; Garagorri, J.M.; et al. Interplay between weight loss and gut microbiota composition in overweight adolescents. Obesity 2009, 17, 1906–1915. [Google Scholar] [CrossRef] [Green Version]
  22. Nadal, I.; Santacruz, A.; Marcos, A.; Warnberg, J.; Garagorri, J.M.; Moreno, L.A.; Martin-Matillas, M.; Campoy, C.; Marti, A.; Moleres, A.; et al. Shifts in clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents. Int. J. Obes. 2009, 33, 758–767. [Google Scholar] [CrossRef] [Green Version]
  23. Backhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef] [Green Version]
  24. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  25. Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef] [Green Version]
  26. Duncan, S.H.; Lobley, G.E.; Holtrop, G.; Ince, J.; Johnstone, A.M.; Louis, P.; Flint, H.J. Human colonic microbiota associated with diet, obesity and weight loss. Int. J. Obes. 2008, 32, 1720–1724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Reilly, S.M.; Saltiel, A.R. Adapting to obesity with adipose tissue inflammation. Nat. Rev. Endocrinol. 2017, 13, 633–643. [Google Scholar] [CrossRef] [PubMed]
  28. Hersoug, L.G.; Moller, P.; Loft, S. Gut microbiota-derived lipopolysaccharide uptake and trafficking to adipose tissue: Implications for inflammation and obesity. Obes. Rev. 2016, 17, 297–312. [Google Scholar] [CrossRef] [PubMed]
  29. De La Serre, C.B.; Ellis, C.L.; Lee, J.; Hartman, A.L.; Rutledge, J.C.; Raybould, H.E. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G440–G448. [Google Scholar] [CrossRef]
  30. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  31. Zhao, J.; Bi, W.; Xiao, S.; Lan, X.; Cheng, X.; Zhang, J.; Lu, D.; Wei, W.; Wang, Y.; Li, H.; et al. Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci. Rep. 2019, 9, 5790. [Google Scholar] [CrossRef] [Green Version]
  32. Harte, A.L.; Varma, M.C.; Tripathi, G.; McGee, K.C.; Al-Daghri, N.M.; Al-Attas, O.S.; Sabico, S.; O’Hare, J.P.; Ceriello, A.; Saravanan, P.; et al. High fat intake leads to acute postprandial exposure to circulating endotoxin in type 2 diabetic subjects. Diabetes Care 2012, 35, 375–382. [Google Scholar] [CrossRef] [Green Version]
  33. Gribble, F.M.; Reimann, F. Enteroendocrine Cells: Chemosensors in the Intestinal Epithelium. Annu. Rev. Physiol. 2016, 78, 277–299. [Google Scholar] [CrossRef]
  34. Worthington, J.J.; Reimann, F.; Gribble, F.M. Enteroendocrine cells-sensory sentinels of the intestinal environment and orchestrators of mucosal immunity. Mucosal Immunol. 2018, 11, 3–20. [Google Scholar] [CrossRef] [PubMed]
  35. Symonds, E.L.; Peiris, M.; Page, A.J.; Chia, B.; Dogra, H.; Masding, A.; Galanakis, V.; Atiba, M.; Bulmer, D.; Young, R.L.; et al. Mechanisms of activation of mouse and human enteroendocrine cells by nutrients. Gut 2015, 64, 618–626. [Google Scholar] [CrossRef] [Green Version]
  36. Chin, A.; Svejda, B.; Gustafsson, B.I.; Granlund, A.B.; Sandvik, A.K.; Timberlake, A.; Sumpio, B.; Pfragner, R.; Modlin, I.M.; Kidd, M. The role of mechanical forces and adenosine in the regulation of intestinal enterochromaffin cell serotonin secretion. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 302, G397–G405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bellono, N.W.; Bayrer, J.R.; Leitch, D.B.; Castro, J.; Zhang, C.; O’Donnell, T.A.; Brierley, S.M.; Ingraham, H.A.; Julius, D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell 2017, 170, 185–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Nohr, M.K.; Pedersen, M.H.; Gille, A.; Egerod, K.L.; Engelstoft, M.S.; Husted, A.S.; Sichlau, R.M.; Grunddal, K.V.; Poulsen, S.S.; Han, S.; et al. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. Endocrinology 2013, 154, 3552–3564. [Google Scholar] [CrossRef]
  39. Modasia, A.; Parker, A.; Jones, E.; Stentz, R.; Brion, A.; Goldson, A.; Defernez, M.; Wileman, T.; Ashley Blackshaw, L.; Carding, S.R. Regulation of Enteroendocrine Cell Networks by the Major Human Gut Symbiont Bacteroides thetaiotaomicron. Front. Microbiol. 2020, 11, 575595. [Google Scholar] [CrossRef] [PubMed]
  40. Fulling, C.; Dinan, T.G.; Cryan, J.F. Gut Microbe to Brain Signaling: What Happens in Vagus. Neuron 2019, 101, 998–1002. [Google Scholar] [CrossRef] [Green Version]
  41. Bonaz, B.; Sinniger, V.; Pellissier, S. The Vagus Nerve in the Neuro-Immune Axis: Implications in the Pathology of the Gastrointestinal Tract. Front. Immunol. 2017, 8, 1452. [Google Scholar] [CrossRef]
  42. Zhang, X.; Fogel, R. Involvement of glutamate in gastrointestinal vago-vagal reflexes initiated by gastrointestinal distention in the rat. Auton. Neurosci. 2003, 103, 19–37. [Google Scholar] [CrossRef]
  43. Ueno, H.; Nakazato, M. Mechanistic relationship between the vagal afferent pathway, central nervous system and peripheral organs in appetite regulation. J. Diabetes Investig. 2016, 7, 812–818. [Google Scholar] [CrossRef]
  44. Roman, C.W.; Derkach, V.A.; Palmiter, R.D. Genetically and functionally defined NTS to PBN brain circuits mediating anorexia. Nat. Commun. 2016, 7, 11905. [Google Scholar] [CrossRef] [PubMed]
  45. Bonaz, B.; Sinniger, V.; Pellissier, S. Anti-inflammatory properties of the vagus nerve: Potential therapeutic implications of vagus nerve stimulation. J. Physiol. 2016, 594, 5781–5790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Raybould, H.E. Gut chemosensing: Interactions between gut endocrine cells and visceral afferents. Auton. Neurosci. 2010, 153, 41–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Cui, R.J.; Li, X.; Appleyard, S.M. Ghrelin inhibits visceral afferent activation of catecholamine neurons in the solitary tract nucleus. J. Neurosci. 2011, 31, 3484–3492. [Google Scholar] [CrossRef] [Green Version]
  48. Koda, S.; Date, Y.; Murakami, N.; Shimbara, T.; Hanada, T.; Toshinai, K.; Niijima, A.; Furuya, M.; Inomata, N.; Osuye, K.; et al. The role of the vagal nerve in peripheral PYY3-36-induced feeding reduction in rats. Endocrinology 2005, 146, 2369–2375. [Google Scholar] [CrossRef] [Green Version]
  49. Lieverse, R.J.; Jansen, J.B.; Masclee, A.A.; Lamers, C.B. Satiety effects of a physiological dose of cholecystokinin in humans. Gut 1995, 36, 176–179. [Google Scholar] [CrossRef] [Green Version]
  50. De La Serre, C.B.; de Lartigue, G.; Raybould, H.E. Chronic exposure to Low dose bacterial lipopolysaccharide inhibits leptin signaling in vagal afferent neurons. Physiol. Behav. 2015, 139, 188–194. [Google Scholar] [CrossRef] [Green Version]
  51. Sen, T.; Cawthon, C.R.; Ihde, B.T.; Hajnal, A.; DiLorenzo, P.M.; de La Serre, C.B.; Czaja, K. Diet-driven microbiota dysbiosis is associated with vagal remodeling and obesity. Physiol. Behav. 2017, 173, 305–317. [Google Scholar] [CrossRef] [Green Version]
  52. Kral, J.G.; Gortz, L.; Hermansson, G.; Wallin, G.S. Gastroplasty for obesity: Long-term weight loss improved by vagotomy. World J. Surg. 1993, 17, 75–78. [Google Scholar] [CrossRef]
  53. Balbo, S.L.; Ribeiro, R.A.; Mendes, M.C.; Lubaczeuski, C.; Maller, A.C.; Carneiro, E.M.; Bonfleur, M.L. Vagotomy diminishes obesity in cafeteria rats by decreasing cholinergic potentiation of insulin release. J. Physiol. Biochem. 2016, 72, 625–633. [Google Scholar] [CrossRef] [Green Version]
  54. Furness, J.B.; Callaghan, B.P.; Rivera, L.R.; Cho, H.J. The enteric nervous system and gastrointestinal innervation: Integrated local and central control. Adv. Exp. Med. Biol. 2014, 817, 39–71. [Google Scholar] [CrossRef]
  55. Fung, C.; Vanden Berghe, P. Functional circuits and signal processing in the enteric nervous system. Cell. Mol. Life Sci. 2020, 77, 4505–4522. [Google Scholar] [CrossRef]
  56. Li, Y.; Owyang, C. Musings on the wanderer: What’s new in our understanding of vago-vagal reflexes? V. Remodeling of vagus and enteric neural circuitry after vagal injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2003, 285, G461–G469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. De Vadder, F.; Grasset, E.; Manneras Holm, L.; Karsenty, G.; Macpherson, A.J.; Olofsson, L.E.; Backhed, F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc. Natl. Acad. Sci. USA 2018, 115, 6458–6463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Di Giancamillo, A.; Vitari, F.; Bosi, G.; Savoini, G.; Domeneghini, C. The chemical code of porcine enteric neurons and the number of enteric glial cells are altered by dietary probiotics. Neurogastroenterol. Motil. 2010, 22, e271–e278. [Google Scholar] [CrossRef] [PubMed]
  59. Kabouridis, P.S.; Lasrado, R.; McCallum, S.; Chng, S.H.; Snippert, H.J.; Clevers, H.; Pettersson, S.; Pachnis, V. Microbiota controls the homeostasis of glial cells in the gut lamina propria. Neuron 2015, 85, 289–295. [Google Scholar] [CrossRef] [Green Version]
  60. Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef] [Green Version]
  61. Gao, Z.; Yin, J.; Zhang, J.; Ward, R.E.; Martin, R.J.; Lefevre, M.; Cefalu, W.T.; Ye, J. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 2009, 58, 1509–1517. [Google Scholar] [CrossRef] [Green Version]
  62. Huuskonen, J.; Suuronen, T.; Nuutinen, T.; Kyrylenko, S.; Salminen, A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. Br. J. Pharmacol. 2004, 141, 874–880. [Google Scholar] [CrossRef]
  63. Wong, J.M.; de Souza, R.; Kendall, C.W.; Emam, A.; Jenkins, D.J. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  64. Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef] [Green Version]
  65. Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.M.; Reimann, F.; Blottiere, H.M. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74. [Google Scholar] [CrossRef] [PubMed]
  66. Goswami, C.; Iwasaki, Y.; Yada, T. Short-chain fatty acids suppress food intake by activating vagal afferent neurons. J. Nutr. Biochem. 2018, 57, 130–135. [Google Scholar] [CrossRef]
  67. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Sahuri-Arisoylu, M.; Brody, L.P.; Parkinson, J.R.; Parkes, H.; Navaratnam, N.; Miller, A.D.; Thomas, E.L.; Frost, G.; Bell, J.D. Reprogramming of hepatic fat accumulation and ‘browning’ of adipose tissue by the short-chain fatty acid acetate. Int. J. Obes. 2016, 40, 955–963. [Google Scholar] [CrossRef]
  69. Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Backhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Lu, Y.; Fan, C.; Li, P.; Lu, Y.; Chang, X.; Qi, K. Short Chain Fatty Acids Prevent High-fat-diet-induced Obesity in Mice by Regulating G Protein-coupled Receptors and Gut Microbiota. Sci. Rep. 2016, 6, 37589. [Google Scholar] [CrossRef] [Green Version]
  72. Schwiertz, A.; Taras, D.; Schafer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef]
  73. Teixeira, T.F.; Grzeskowiak, L.; Franceschini, S.C.; Bressan, J.; Ferreira, C.L.; Peluzio, M.C. Higher level of faecal SCFA in women correlates with metabolic syndrome risk factors. Br. J. Nutr. 2013, 109, 914–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. den Besten, G.; Havinga, R.; Bleeker, A.; Rao, S.; Gerding, A.; van Eunen, K.; Groen, A.K.; Reijngoud, D.J.; Bakker, B.M. The short-chain fatty acid uptake fluxes by mice on a guar gum supplemented diet associate with amelioration of major biomarkers of the metabolic syndrome. PLoS ONE 2014, 9, e107392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Chambers, E.S.; Byrne, C.S.; Aspey, K.; Chen, Y.; Khan, S.; Morrison, D.J.; Frost, G. Acute oral sodium propionate supplementation raises resting energy expenditure and lipid oxidation in fasted humans. Diabetes Obes. Metab. 2018, 20, 1034–1039. [Google Scholar] [CrossRef] [PubMed]
  76. Bouter, K.; Bakker, G.J.; Levin, E.; Hartstra, A.V.; Kootte, R.S.; Udayappan, S.D.; Katiraei, S.; Bahler, L.; Gilijamse, P.W.; Tremaroli, V.; et al. Differential metabolic effects of oral butyrate treatment in lean versus metabolic syndrome subjects. Clin. Transl. Gastroenterol. 2018, 9, 155. [Google Scholar] [CrossRef]
  77. Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef] [Green Version]
  78. Al-Lahham, S.H.; Roelofsen, H.; Priebe, M.; Weening, D.; Dijkstra, M.; Hoek, A.; Rezaee, F.; Venema, K.; Vonk, R.J. Regulation of adipokine production in human adipose tissue by propionic acid. Eur. J. Clin. Investig. 2010, 40, 401–407. [Google Scholar] [CrossRef] [PubMed]
  79. Al-Lahham, S.; Roelofsen, H.; Rezaee, F.; Weening, D.; Hoek, A.; Vonk, R.; Venema, K. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur. J. Clin. Invest. 2012, 42, 357–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Hartstra, A.V.; Schuppel, V.; Imangaliyev, S.; Schrantee, A.; Prodan, A.; Collard, D.; Levin, E.; Dallinga-Thie, G.; Ackermans, M.T.; Winkelmeijer, M.; et al. Infusion of donor feces affects the gut-brain axis in humans with metabolic syndrome. Mol. Metab. 2020, 42, 101076. [Google Scholar] [CrossRef] [PubMed]
  81. Saper, C.B.; Chou, T.C.; Elmquist, J.K. The need to feed: Homeostatic and hedonic control of eating. Neuron 2002, 36, 199–211. [Google Scholar] [CrossRef] [Green Version]
  82. Lutter, M.; Nestler, E.J. Homeostatic and hedonic signals interact in the regulation of food intake. J. Nutr. 2009, 139, 629–632. [Google Scholar] [CrossRef] [Green Version]
  83. Schwartz, G.J. Brainstem integrative function in the central nervous system control of food intake. Forum. Nutr. 2010, 63, 141–151. [Google Scholar] [CrossRef]
  84. Langlet, F.; Levin, B.E.; Luquet, S.; Mazzone, M.; Messina, A.; Dunn-Meynell, A.A.; Balland, E.; Lacombe, A.; Mazur, D.; Carmeliet, P.; et al. Tanycytic VEGF-A boosts blood-hypothalamus barrier plasticity and access of metabolic signals to the arcuate nucleus in response to fasting. Cell Metab. 2013, 17, 607–617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Loh, K.; Herzog, H.; Shi, Y.C. Regulation of energy homeostasis by the NPY system. Trends Endocrinol. Metab. 2015, 26, 125–135. [Google Scholar] [CrossRef]
  86. Morton, G.J.; Cummings, D.E.; Baskin, D.G.; Barsh, G.S.; Schwartz, M.W. Central nervous system control of food intake and body weight. Nature 2006, 443, 289–295. [Google Scholar] [CrossRef] [PubMed]
  87. Cone, R.D. Anatomy and regulation of the central melanocortin system. Nat. Neurosci. 2005, 8, 571–578. [Google Scholar] [CrossRef]
  88. Ollmann, M.M.; Wilson, B.D.; Yang, Y.K.; Kerns, J.A.; Chen, Y.; Gantz, I.; Barsh, G.S. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 1997, 278, 135–138. [Google Scholar] [CrossRef]
  89. Cummings, D.E.; Purnell, J.Q.; Frayo, R.S.; Schmidova, K.; Wisse, B.E.; Weigle, D.S. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 2001, 50, 1714–1719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Williams, D.L.; Cummings, D.E.; Grill, H.J.; Kaplan, J.M. Meal-related ghrelin suppression requires postgastric feedback. Endocrinology 2003, 144, 2765–2767. [Google Scholar] [CrossRef] [Green Version]
  91. Muller, T.D.; Nogueiras, R.; Andermann, M.L.; Andrews, Z.B.; Anker, S.D.; Argente, J.; Batterham, R.L.; Benoit, S.C.; Bowers, C.Y.; Broglio, F.; et al. Ghrelin. Mol. Metab. 2015, 4, 437–460. [Google Scholar] [CrossRef]
  92. Egecioglu, E.; Jerlhag, E.; Salome, N.; Skibicka, K.P.; Haage, D.; Bohlooly, Y.M.; Andersson, D.; Bjursell, M.; Perrissoud, D.; Engel, J.A.; et al. Ghrelin increases intake of rewarding food in rodents. Addict. Biol. 2010, 15, 304–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Skibicka, K.P.; Hansson, C.; Alvarez-Crespo, M.; Friberg, P.A.; Dickson, S.L. Ghrelin directly targets the ventral tegmental area to increase food motivation. Neuroscience 2011, 180, 129–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tschop, M.; Weyer, C.; Tataranni, P.A.; Devanarayan, V.; Ravussin, E.; Heiman, M.L. Circulating ghrelin levels are decreased in human obesity. Diabetes 2001, 50, 707–709. [Google Scholar] [CrossRef] [Green Version]
  95. Considine, R.V.; Sinha, M.K.; Heiman, M.L.; Kriauciunas, A.; Stephens, T.W.; Nyce, M.R.; Ohannesian, J.P.; Marco, C.C.; McKee, L.J.; Bauer, T.L.; et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 1996, 334, 292–295. [Google Scholar] [CrossRef]
  96. Jequier, E. Leptin signaling, adiposity, and energy balance. Ann. N. Y. Acad. Sci. 2002, 967, 379–388. [Google Scholar] [CrossRef] [PubMed]
  97. Keim, L.N.; Stern, J.S.; Havel, P.J. Relation between circulating leptin concentrations and appetite during a prolonged, moderate energy deficit in women. Am. J. Clin. Nutr. 1998, 68, 794–801. [Google Scholar] [CrossRef] [PubMed]
  98. Mars, M.; De Graaf, C.; De Groot, C.P.G.M.; Van Rossum, C.T.M.; Kok, F.J. Fasting leptin and appetite responses induced by a 4-day 65%-energy-restricted diet. Int. J. Obes. 2006, 30, 122–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Elias, C.F.; Aschkenasi, C.; Lee, C.; Kelly, J.; Ahima, R.S.; Bjorbæk, C.; Flier, J.S.; Saper, C.B.; Elmquist, J.K. Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron 1999, 23, 775–786. [Google Scholar] [CrossRef] [Green Version]
  100. Zelissen, P.M.; Stenlof, K.; Lean, M.E.; Fogteloo, J.; Keulen, E.T.; Wilding, J.; Finer, N.; Rossner, S.; Lawrence, E.; Fletcher, C.; et al. Effect of three treatment schedules of recombinant methionyl human leptin on body weight in obese adults: A randomized, placebo-controlled trial. Diabetes Obes. Metab. 2005, 7, 755–761. [Google Scholar] [CrossRef]
  101. Schwartz, M.W.; Peskind, E.; Raskind, M.; Boyko, E.J.; Porte, D., Jr. Cerebrospinal fluid leptin levels: Relationship to plasma levels and to adiposity in humans. Nat. Med. 1996, 2, 589–593. [Google Scholar] [CrossRef]
  102. Harrison, L.; Schriever, S.C.; Feuchtinger, A.; Kyriakou, E.; Baumann, P.; Pfuhlmann, K.; Messias, A.C.; Walch, A.; Tschop, M.H.; Pfluger, P.T. Fluorescent blood-brain barrier tracing shows intact leptin transport in obese mice. Int. J. Obes. 2019, 43, 1305–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Enriori, P.J.; Evans, A.E.; Sinnayah, P.; Jobst, E.E.; Tonelli-Lemos, L.; Billes, S.K.; Glavas, M.M.; Grayson, B.E.; Perello, M.; Nillni, E.A.; et al. Diet-induced obesity causes severe but reversible leptin resistance in arcuate melanocortin neurons. Cell Metab. 2007, 5, 181–194. [Google Scholar] [CrossRef]
  104. Munzberg, H.; Flier, J.S.; Bjorbaek, C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology 2004, 145, 4880–4889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Flier, J.S.; Maratos-Flier, E. Leptin’s Physiologic Role: Does the Emperor of Energy Balance Have No Clothes? Cell Metab. 2017, 26, 24–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Queipo-Ortuno, M.I.; Seoane, L.M.; Murri, M.; Pardo, M.; Gomez-Zumaquero, J.M.; Cardona, F.; Casanueva, F.; Tinahones, F.J. Gut microbiota composition in male rat models under different nutritional status and physical activity and its association with serum leptin and ghrelin levels. PLoS ONE 2013, 8, e65465. [Google Scholar] [CrossRef] [PubMed]
  107. Yanagi, H.; Tsuda, A.; Matsushima, M.; Takahashi, S.; Ozawa, G.; Koga, Y.; Takagi, A. Changes in the gut microbiota composition and the plasma ghrelin level in patients with Helicobacter pylori-infected patients with eradication therapy. BMJ Open Gastroenterol. 2017, 4, e000182. [Google Scholar] [CrossRef] [Green Version]
  108. Torres-Fuentes, C.; Golubeva, A.V.; Zhdanov, A.V.; Wallace, S.; Arboleya, S.; Papkovsky, D.B.; El Aidy, S.; Ross, P.; Roy, B.L.; Stanton, C.; et al. Short-chain fatty acids and microbiota metabolites attenuate ghrelin receptor signaling. FASEB J. 2019, 33, 13546–13559. [Google Scholar] [CrossRef] [Green Version]
  109. Xiong, Y.; Miyamoto, N.; Shibata, K.; Valasek, M.A.; Motoike, T.; Kedzierski, R.M.; Yanagisawa, M. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc. Natl. Acad. Sci. USA 2004, 101, 1045–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Schele, E.; Grahnemo, L.; Anesten, F.; Hallen, A.; Backhed, F.; Jansson, J.O. The gut microbiota reduces leptin sensitivity and the expression of the obesity-suppressing neuropeptides proglucagon (Gcg) and brain-derived neurotrophic factor (Bdnf) in the central nervous system. Endocrinology 2013, 154, 3643–3651. [Google Scholar] [CrossRef] [Green Version]
  111. Cheng, Y.C.; Liu, J.R. Effect of Lactobacillus rhamnosus GG on Energy Metabolism, Leptin Resistance, and Gut Microbiota in Mice with Diet-Induced Obesity. Nutrients 2020, 12, 2557. [Google Scholar] [CrossRef] [PubMed]
  112. Schellekens, H.; Finger, B.C.; Dinan, T.G.; Cryan, J.F. Ghrelin signalling and obesity: At the interface of stress, mood and food reward. Pharmacol. Ther. 2012, 135, 316–326. [Google Scholar] [CrossRef]
  113. Batterham, R.L.; Cowley, M.A.; Small, C.J.; Herzog, H.; Cohen, M.A.; Dakin, C.L.; Wren, A.M.; Brynes, A.E.; Low, M.J.; Ghatei, M.A.; et al. Gut hormone PYY(3-36) physiologically inhibits food intake. Nature 2002, 418, 650–654. [Google Scholar] [CrossRef]
  114. Drucker, D.J.; Nauck, M.A. The incretin system: Glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006, 368, 1696–1705. [Google Scholar] [CrossRef]
  115. Astrup, A.; Rossner, S.; Van Gaal, L.; Rissanen, A.; Niskanen, L.; Al Hakim, M.; Madsen, J.; Rasmussen, M.F.; Lean, M.E.; Group, N.N.S. Effects of liraglutide in the treatment of obesity: A randomised, double-blind, placebo-controlled study. Lancet 2009, 374, 1606–1616. [Google Scholar] [CrossRef]
  116. Blundell, J.; Finlayson, G.; Axelsen, M.; Flint, A.; Gibbons, C.; Kvist, T.; Hjerpsted, J.B. Effects of once-weekly semaglutide on appetite, energy intake, control of eating, food preference and body weight in subjects with obesity. Diabetes Obes. Metab. 2017, 19, 1242–1251. [Google Scholar] [CrossRef]
  117. Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sorensen, J.; Cowley, M.A.; Dalboge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Investig. 2014, 124, 4473–4488. [Google Scholar] [CrossRef] [Green Version]
  118. Kushner, R.F.; Calanna, S.; Davies, M.; Dicker, D.; Garvey, W.T.; Goldman, B.; Lingvay, I.; Thomsen, M.; Wadden, T.A.; Wharton, S.; et al. Semaglutide 2.4 mg for the Treatment of Obesity: Key Elements of the STEP Trials 1 to 5. Obesity 2020, 28, 1050–1061. [Google Scholar] [CrossRef] [PubMed]
  119. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021. [Google Scholar] [CrossRef] [PubMed]
  120. Krieger, J.P.; Arnold, M.; Pettersen, K.G.; Lossel, P.; Langhans, W.; Lee, S.J. Knockdown of GLP-1 Receptors in Vagal Afferents Affects Normal Food Intake and Glycemia. Diabetes 2016, 65, 34–43. [Google Scholar] [CrossRef] [Green Version]
  121. Rehfeld, J.F.; Friis-Hansen, L.; Goetze, J.P.; Hansen, T.V. The biology of cholecystokinin and gastrin peptides. Curr. Top. Med. Chem. 2007, 7, 1154–1165. [Google Scholar] [CrossRef] [PubMed]
  122. Gibbs, J.; Young, R.C.; Smith, G.P. Cholecystokinin elicits satiety in rats with open gastric fistulas. Nature 1973, 245, 323–325. [Google Scholar] [CrossRef]
  123. Corp, E.S.; McQuade, J.; Moran, T.H.; Smith, G.P. Characterization of type A and type B CCK receptor binding sites in rat vagus nerve. Brain Res. 1993, 623, 161–166. [Google Scholar] [CrossRef]
  124. Smith, G.P.; Jerome, C.; Cushin, B.J.; Eterno, R.; Simansky, K.J. Abdominal vagotomy blocks the satiety effect of cholecystokinin in the rat. Science 1981, 213, 1036–1037. [Google Scholar] [CrossRef] [PubMed]
  125. Jordan, J.; Greenway, F.L.; Leiter, L.A.; Li, Z.; Jacobson, P.; Murphy, K.; Hill, J.; Kler, L.; Aftring, R.P. Stimulation of cholecystokinin-A receptors with GI181771X does not cause weight loss in overweight or obese patients. Clin. Pharmacol. Ther. 2008, 83, 281–287. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, J.S.; Kirkland, R.A.; Lee, S.H.; Cawthon, C.R.; Rzepka, K.W.; Minaya, D.M.; de Lartigue, G.; Czaja, K.; de La Serre, C.B. Gut microbiota composition modulates inflammation and structure of the vagal afferent pathway. Physiol. Behav. 2020, 225, 113082. [Google Scholar] [CrossRef] [PubMed]
  127. Federico, A.; Dallio, M.; Tolone, S.; Gravina, A.G.; Patrone, V.; Romano, M.; Tuccillo, C.; Mozzillo, A.L.; Amoroso, V.; Misso, G.; et al. Gastrointestinal Hormones, Intestinal Microbiota and Metabolic Homeostasis in Obese Patients: Effect of Bariatric Surgery. In Vivo 2016, 30, 321–330. [Google Scholar]
  128. Fetissov, S.O. Role of the gut microbiota in host appetite control: Bacterial growth to animal feeding behaviour. Nat. Rev. Endocrinol. 2017, 13, 11–25. [Google Scholar] [CrossRef]
  129. Tennoune, N.; Chan, P.; Breton, J.; Legrand, R.; Chabane, Y.N.; Akkermann, K.; Jarv, A.; Ouelaa, W.; Takagi, K.; Ghouzali, I.; et al. Bacterial ClpB heat-shock protein, an antigen-mimetic of the anorexigenic peptide alpha-MSH, at the origin of eating disorders. Transl. Psychiatry 2014, 4, e458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Panaro, B.L.; Tough, I.R.; Engelstoft, M.S.; Matthews, R.T.; Digby, G.J.; Moller, C.L.; Svendsen, B.; Gribble, F.; Reimann, F.; Holst, J.J.; et al. The melanocortin-4 receptor is expressed in enteroendocrine L cells and regulates the release of peptide YY and glucagon-like peptide 1 in vivo. Cell Metab. 2014, 20, 1018–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Breton, J.; Tennoune, N.; Lucas, N.; Francois, M.; Legrand, R.; Jacquemot, J.; Goichon, A.; Guerin, C.; Peltier, J.; Pestel-Caron, M.; et al. Gut Commensal E. coli Proteins Activate Host Satiety Pathways following Nutrient-Induced Bacterial Growth. Cell Metab. 2016, 23, 324–334. [Google Scholar] [CrossRef] [Green Version]
  132. Molinaro, A.; Bel Lassen, P.; Henricsson, M.; Wu, H.; Adriouch, S.; Belda, E.; Chakaroun, R.; Nielsen, T.; Bergh, P.O.; Rouault, C.; et al. Imidazole propionate is increased in diabetes and associated with dietary patterns and altered microbial ecology. Nat. Commun. 2020, 11, 5881. [Google Scholar] [CrossRef]
  133. Koh, A.; Molinaro, A.; Stahlman, M.; Khan, M.T.; Schmidt, C.; Manneras-Holm, L.; Wu, H.; Carreras, A.; Jeong, H.; Olofsson, L.E.; et al. Microbially Produced Imidazole Propionate Impairs Insulin Signaling through mTORC1. Cell 2018, 175, 947–961. [Google Scholar] [CrossRef] [Green Version]
  134. Swann, J.R.; Spitzer, S.O.; Diaz Heijtz, R. Developmental Signatures of Microbiota-Derived Metabolites in the Mouse Brain. Metabolites 2020, 10, 172. [Google Scholar] [CrossRef]
  135. Heiss, C.N.; Olofsson, L.E. The role of the gut microbiota in development, function and disorders of the central nervous system and the enteric nervous system. J. Neuroendocrinol. 2019, 31, e12684. [Google Scholar] [CrossRef]
  136. Leech, C.A.; Habener, J.F. Regulation of glucagon-like peptide-1 receptor and calcium-sensing receptor signaling by L-histidine. Endocrinology 2003, 144, 4851–4858. [Google Scholar] [CrossRef] [Green Version]
  137. Stice, E.; Figlewicz, D.P.; Gosnell, B.A.; Levine, A.S.; Pratt, W.E. The contribution of brain reward circuits to the obesity epidemic. Neurosci. Biobehav. Rev. 2013, 37, 2047–2058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Schultes, B.; Ernst, B.; Wilms, B.; Thurnheer, M.; Hallschmid, M. Hedonic hunger is increased in severely obese patients and is reduced after gastric bypass surgery. Am. J. Clin. Nutr. 2010, 92, 277–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ribeiro, G.; Camacho, M.; Santos, O.; Pontes, C.; Torres, S.; Oliveira-Maia, A.J. Association between hedonic hunger and body-mass index versus obesity status. Sci. Rep. 2018, 8, 5857. [Google Scholar] [CrossRef] [Green Version]
  140. Lenoir, M.; Serre, F.; Cantin, L.; Ahmed, S.H. Intense sweetness surpasses cocaine reward. PLoS ONE 2007, 2, e698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Leinninger, G.M.; Jo, Y.H.; Leshan, R.L.; Louis, G.W.; Yang, H.; Barrera, J.G.; Wilson, H.; Opland, D.M.; Faouzi, M.A.; Gong, Y.; et al. Leptin acts via leptin receptor-expressing lateral hypothalamic neurons to modulate the mesolimbic dopamine system and suppress feeding. Cell Metab. 2009, 10, 89–98. [Google Scholar] [CrossRef] [Green Version]
  142. Volkow, N.D.; Wang, G.J.; Telang, F.; Fowler, J.S.; Thanos, P.K.; Logan, J.; Alexoff, D.; Ding, Y.S.; Wong, C.; Ma, Y.; et al. Low dopamine striatal D2 receptors are associated with prefrontal metabolism in obese subjects: Possible contributing factors. Neuroimage 2008, 42, 1537–1543. [Google Scholar] [CrossRef] [Green Version]
  143. Johnson, P.M.; Kenny, P.J. Dopamine D2 receptors in addiction-like reward dysfunction and compulsive eating in obese rats. Nat. Neurosci. 2010, 13, 635–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Stoeckel, L.E.; Weller, R.E.; Cook, E.W., 3rd; Twieg, D.B.; Knowlton, R.C.; Cox, J.E. Widespread reward-system activation in obese women in response to pictures of high-calorie foods. Neuroimage 2008, 41, 636–647. [Google Scholar] [CrossRef] [PubMed]
  145. Stice, E.; Spoor, S.; Bohon, C.; Veldhuizen, M.G.; Small, D.M. Relation of reward from food intake and anticipated food intake to obesity: A functional magnetic resonance imaging study. J. Abnorm. Psychol. 2008, 117, 924–935. [Google Scholar] [CrossRef] [Green Version]
  146. Blum, K.; Thanos, P.K.; Gold, M.S. Dopamine and glucose, obesity, and reward deficiency syndrome. Front. Psychol. 2014, 5, 919. [Google Scholar] [CrossRef] [Green Version]
  147. Van Galen, K.A.; Ter Horst, K.W.; Booij, J.; la Fleur, S.E.; Serlie, M.J. The role of central dopamine and serotonin in human obesity: Lessons learned from molecular neuroimaging studies. Metabolism 2018, 85, 325–339. [Google Scholar] [CrossRef] [PubMed]
  148. Burger, K.S.; Stice, E. Variability in reward responsivity and obesity: Evidence from brain imaging studies. Curr. Drug Abuse Rev. 2011, 4, 182–189. [Google Scholar] [CrossRef] [Green Version]
  149. Garcia Barrado, M.J.; Iglesias Osma, M.C.; Blanco, E.J.; Carretero Hernandez, M.; Sanchez Robledo, V.; Catalano Iniesta, L.; Carrero, S.; Carretero, J. Dopamine modulates insulin release and is involved in the survival of rat pancreatic beta cells. PLoS ONE 2015, 10, e0123197. [Google Scholar] [CrossRef]
  150. Rubi, B.; Maechler, P. Minireview: New roles for peripheral dopamine on metabolic control and tumor growth: Let’s seek the balance. Endocrinology 2010, 151, 5570–5581. [Google Scholar] [CrossRef] [Green Version]
  151. Eisenhofer, G.; Aneman, A.; Friberg, P.; Hooper, D.; Fandriks, L.; Lonroth, H.; Hunyady, B.; Mezey, E. Substantial production of dopamine in the human gastrointestinal tract. J. Clin. Endocrinol. Metab. 1997, 82, 3864–3871. [Google Scholar] [CrossRef]
  152. Simpson, C.A.; Diaz-Arteche, C.; Eliby, D.; Schwartz, O.S.; Simmons, J.G.; Cowan, C.S.M. The gut microbiota in anxiety and depression—A systematic review. Clin. Psychol. Rev. 2020, 83, 101943. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, D.; Zhao, D.; Ali Shah, S.Z.; Wu, W.; Lai, M.; Zhang, X.; Li, J.; Guan, Z.; Zhao, H.; Li, W.; et al. The Role of the Gut Microbiota in the Pathogenesis of Parkinson’s Disease. Front. Neurol. 2019, 10, 1155. [Google Scholar] [CrossRef] [Green Version]
  154. Jenkins, T.A.; Nguyen, J.C.; Polglaze, K.E.; Bertrand, P.P. Influence of Tryptophan and Serotonin on Mood and Cognition with a Possible Role of the Gut-Brain Axis. Nutrients 2016, 8, 56. [Google Scholar] [CrossRef]
  155. Voigt, J.P.; Fink, H. Serotonin controlling feeding and satiety. Behav. Brain Res. 2015, 277, 14–31. [Google Scholar] [CrossRef]
  156. Mawe, G.M.; Hoffman, J.M. Serotonin signalling in the gut--functions, dysfunctions and therapeutic targets. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 473–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Watanabe, H.; Nakano, T.; Saito, R.; Akasaka, D.; Saito, K.; Ogasawara, H.; Minashima, T.; Miyazawa, K.; Kanaya, T.; Takakura, I.; et al. Serotonin Improves High Fat Diet Induced Obesity in Mice. PLoS ONE 2016, 11, e0147143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Watanabe, H.; Akasaka, D.; Ogasawara, H.; Sato, K.; Miyake, M.; Saito, K.; Takahashi, Y.; Kanaya, T.; Takakura, I.; Hondo, T.; et al. Peripheral serotonin enhances lipid metabolism by accelerating bile acid turnover. Endocrinology 2010, 151, 4776–4786. [Google Scholar] [CrossRef] [Green Version]
  159. Fernstrom, J.D. Large neutral amino acids: Dietary effects on brain neurochemistry and function. Amino Acids 2013, 45, 419–430. [Google Scholar] [CrossRef]
  160. Ruddick, J.P.; Evans, A.K.; Nutt, D.J.; Lightman, S.L.; Rook, G.A.; Lowry, C.A. Tryptophan metabolism in the central nervous system: Medical implications. Expert Rev. Mol. Med. 2006, 8, 1–27. [Google Scholar] [CrossRef]
  161. Bohula, E.A.; Wiviott, S.D.; McGuire, D.K.; Inzucchi, S.E.; Kuder, J.; Im, K.; Fanola, C.L.; Qamar, A.; Brown, C.; Budaj, A.; et al. Cardiovascular Safety of Lorcaserin in Overweight or Obese Patients. N. Engl. J. Med. 2018, 379, 1107–1117. [Google Scholar] [CrossRef]
  162. Mahase, E. Weight loss pill praised as “holy grail” is withdrawn from US market over cancer link. BMJ 2020, 368, m705. [Google Scholar] [CrossRef]
  163. Young, R.L.; Lumsden, A.L.; Martin, A.M.; Schober, G.; Pezos, N.; Thazhath, S.S.; Isaacs, N.J.; Cvijanovic, N.; Sun, E.W.L.; Wu, T.; et al. Augmented capacity for peripheral serotonin release in human obesity. Int. J. Obes. 2018, 42, 1880–1889. [Google Scholar] [CrossRef]
  164. Crane, J.D.; Palanivel, R.; Mottillo, E.P.; Bujak, A.L.; Wang, H.; Ford, R.J.; Collins, A.; Blumer, R.M.; Fullerton, M.D.; Yabut, J.M.; et al. Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nat. Med. 2015, 21, 166–172. [Google Scholar] [CrossRef] [Green Version]
  165. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [Green Version]
  166. Delgado, T.C. Glutamate and GABA in Appetite Regulation. Front. Endocrinol. 2013, 4, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Meng, F.; Han, Y.; Srisai, D.; Belakhov, V.; Farias, M.; Xu, Y.; Palmiter, R.D.; Baasov, T.; Wu, Q. New inducible genetic method reveals critical roles of GABA in the control of feeding and metabolism. Proc. Natl. Acad. Sci. USA 2016, 113, 3645–3650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  169. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [Green Version]
  170. Patterson, E.; Ryan, P.M.; Wiley, N.; Carafa, I.; Sherwin, E.; Moloney, G.; Franciosi, E.; Mandal, R.; Wishart, D.S.; Tuohy, K.; et al. Gamma-aminobutyric acid-producing lactobacilli positively affect metabolism and depressive-like behaviour in a mouse model of metabolic syndrome. Sci. Rep. 2019, 9, 16323. [Google Scholar] [CrossRef] [PubMed]
  171. Kootte, R.S.; Levin, E.; Salojarvi, J.; Smits, L.P.; Hartstra, A.V.; Udayappan, S.D.; Hermes, G.; Bouter, K.E.; Koopen, A.M.; Holst, J.J.; et al. Improvement of Insulin Sensitivity after Lean Donor Feces in Metabolic Syndrome Is Driven by Baseline Intestinal Microbiota Composition. Cell Metab. 2017, 26, 611–619. [Google Scholar] [CrossRef] [Green Version]
  172. Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef]
  173. Zhernakova, A.; Kurilshikov, A.; Bonder, M.J.; Tigchelaar, E.F.; Schirmer, M.; Vatanen, T.; Mujagic, Z.; Vila, A.V.; Falony, G.; Vieira-Silva, S.; et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 2016, 352, 565–569. [Google Scholar] [CrossRef] [Green Version]
  174. Martinez, K.B.; Leone, V.; Chang, E.B. Western diets, gut dysbiosis, and metabolic diseases: Are they linked? Gut Microbes 2017, 8, 130–142. [Google Scholar] [CrossRef] [Green Version]
  175. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
  176. Wan, Y.; Wang, F.; Yuan, J.; Li, J.; Jiang, D.; Zhang, J.; Li, H.; Wang, R.; Tang, J.; Huang, T.; et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: A 6-month randomised controlled-feeding trial. Gut 2019, 68, 1417–1429. [Google Scholar] [CrossRef] [Green Version]
  177. Wolters, M.; Ahrens, J.; Romani-Perez, M.; Watkins, C.; Sanz, Y.; Benitez-Paez, A.; Stanton, C.; Gunther, K. Dietary fat, the gut microbiota, and metabolic health—A systematic review conducted within the MyNewGut project. Clin. Nutr. 2019, 38, 2504–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Lang, J.M.; Pan, C.; Cantor, R.M.; Tang, W.H.W.; Garcia-Garcia, J.C.; Kurtz, I.; Hazen, S.L.; Bergeron, N.; Krauss, R.M.; Lusis, A.J. Impact of Individual Traits, Saturated Fat, and Protein Source on the Gut Microbiome. mBio 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  179. Salonen, A.; Lahti, L.; Salojarvi, J.; Holtrop, G.; Korpela, K.; Duncan, S.H.; Date, P.; Farquharson, F.; Johnstone, A.M.; Lobley, G.E.; et al. Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men. ISME J. 2014, 8, 2218–2230. [Google Scholar] [CrossRef] [PubMed]
  180. So, D.; Whelan, K.; Rossi, M.; Morrison, M.; Holtmann, G.; Kelly, J.T.; Shanahan, E.R.; Staudacher, H.M.; Campbell, K.L. Dietary fiber intervention on gut microbiota composition in healthy adults: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2018, 107, 965–983. [Google Scholar] [CrossRef] [Green Version]
  181. Guarner, F.; Khan, A.G.; Garisch, J.; Eliakim, R.; Gangl, A.; Thomson, A.; Krabshuis, J.; Lemair, T.; Kaufmann, P.; de Paula, J.A.; et al. World Gastroenterology Organisation Global Guidelines: Probiotics and prebiotics October 2011. J. Clin. Gastroenterol. 2012, 46, 468–481. [Google Scholar] [CrossRef]
  182. Sivamaruthi, B.S.; Kesika, P.; Suganthy, N.; Chaiyasut, C. A Review on Role of Microbiome in Obesity and Antiobesity Properties of Probiotic Supplements. Biomed Res. Int. 2019, 2019, 3291367. [Google Scholar] [CrossRef] [PubMed]
  183. Million, M.; Angelakis, E.; Paul, M.; Armougom, F.; Leibovici, L.; Raoult, D. Comparative meta-analysis of the effect of Lactobacillus species on weight gain in humans and animals. Microb. Pathog. 2012, 53, 100–108. [Google Scholar] [CrossRef] [PubMed]
  184. Borgeraas, H.; Johnson, L.K.; Skattebu, J.; Hertel, J.K.; Hjelmesaeth, J. Effects of probiotics on body weight, body mass index, fat mass and fat percentage in subjects with overweight or obesity: A systematic review and meta-analysis of randomized controlled trials. Obes. Rev. 2018, 19, 219–232. [Google Scholar] [CrossRef] [Green Version]
  185. Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain-Raspaud, S.; Trotin, B.; Naliboff, B.; et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 1394–1401.e4. [Google Scholar] [CrossRef] [Green Version]
  186. Zhou, Q.; Zhang, Y.; Wang, X.; Yang, R.; Zhu, X.; Zhang, Y.; Chen, C.; Yuan, H.; Yang, Z.; Sun, L. Gut bacteria Akkermansia is associated with reduced risk of obesity: Evidence from the American Gut Project. Nutr. Metab. 2020, 17, 90. [Google Scholar] [CrossRef]
  187. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  188. Geurts, L.; Neyrinck, A.M.; Delzenne, N.M.; Knauf, C.; Cani, P.D. Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: Novel insights into molecular targets and interventions using prebiotics. Benef. Microbes 2014, 5, 3–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Parnell, J.A.; Klancic, T.; Reimer, R.A. Oligofructose decreases serum lipopolysaccharide and plasminogen activator inhibitor-1 in adults with overweight/obesity. Obesity 2017, 25, 510–513. [Google Scholar] [CrossRef] [Green Version]
  190. Dewulf, E.M.; Cani, P.D.; Claus, S.P.; Fuentes, S.; Puylaert, P.G.; Neyrinck, A.M.; Bindels, L.B.; de Vos, W.M.; Gibson, G.R.; Thissen, J.P.; et al. Insight into the prebiotic concept: Lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 2013, 62, 1112–1121. [Google Scholar] [CrossRef] [PubMed]
  191. van der Beek, C.M.; Canfora, E.E.; Kip, A.M.; Gorissen, S.H.M.; Olde Damink, S.W.M.; van Eijk, H.M.; Holst, J.J.; Blaak, E.E.; Dejong, C.H.C.; Lenaerts, K. The prebiotic inulin improves substrate metabolism and promotes short-chain fatty acid production in overweight to obese men. Metabolism 2018, 87, 25–35. [Google Scholar] [CrossRef]
  192. Parnell, J.A.; Reimer, R.A. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am. J. Clin. Nutr. 2009, 89, 1751–1759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Delzenne, N.M.; Cani, P.D.; Daubioul, C.; Neyrinck, A.M. Impact of inulin and oligofructose on gastrointestinal peptides. Br. J. Nutr. 2005, 93 (Suppl. 1), S157–S161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Hess, A.L.; Benitez-Paez, A.; Blaedel, T.; Larsen, L.H.; Iglesias, J.R.; Madera, C.; Sanz, Y.; Larsen, T.M.; MyNewGut Consortium. The effect of inulin and resistant maltodextrin on weight loss during energy restriction: A randomised, placebo-controlled, double-blinded intervention. Eur. J. Nutr. 2020, 59, 2507–2524. [Google Scholar] [CrossRef]
  195. Nicolucci, A.C.; Hume, M.P.; Martinez, I.; Mayengbam, S.; Walter, J.; Reimer, R.A. Prebiotics Reduce Body Fat and Alter Intestinal Microbiota in Children Who Are Overweight or With Obesity. Gastroenterology 2017, 153, 711–722. [Google Scholar] [CrossRef] [Green Version]
  196. Li, Y.T.; Cai, H.F.; Wang, Z.H.; Xu, J.; Fang, J.Y. Systematic review with meta-analysis: Long-term outcomes of faecal microbiota transplantation for Clostridium difficile infection. Aliment. Pharmacol. Ther. 2016, 43, 445–457. [Google Scholar] [CrossRef] [Green Version]
  197. de Groot, P.; Scheithauer, T.; Bakker, G.J.; Prodan, A.; Levin, E.; Khan, M.T.; Herrema, H.; Ackermans, M.; Serlie, M.J.M.; de Brauw, M.; et al. Donor metabolic characteristics drive effects of faecal microbiota transplantation on recipient insulin sensitivity, energy expenditure and intestinal transit time. Gut 2020, 69, 502–512. [Google Scholar] [CrossRef] [Green Version]
  198. Allegretti, J.R.; Kassam, Z.; Mullish, B.H.; Chiang, A.; Carrellas, M.; Hurtado, J.; Marchesi, J.R.; McDonald, J.A.K.; Pechlivanis, A.; Barker, G.F.; et al. Effects of Fecal Microbiota Transplantation With Oral Capsules in Obese Patients. Clin. Gastroenterol. Hepatol. 2020, 18, 855–863. [Google Scholar] [CrossRef] [PubMed]
  199. Yu, E.W.; Gao, L.; Stastka, P.; Cheney, M.C.; Mahabamunuge, J.; Torres Soto, M.; Ford, C.B.; Bryant, J.A.; Henn, M.R.; Hohmann, E.L. Fecal microbiota transplantation for the improvement of metabolism in obesity: The FMT-TRIM double-blind placebo-controlled pilot trial. PLoS Med. 2020, 17, e1003051. [Google Scholar] [CrossRef] [PubMed]
  200. Leong, K.S.W.; Jayasinghe, T.N.; Wilson, B.C.; Derraik, J.G.B.; Albert, B.B.; Chiavaroli, V.; Svirskis, D.M.; Beck, K.L.; Conlon, C.A.; Jiang, Y.; et al. Effects of Fecal Microbiome Transfer in Adolescents With Obesity: The Gut Bugs Randomized Controlled Trial. JAMA Netw. Open 2020, 3, e2030415. [Google Scholar] [CrossRef]
  201. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojarvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143, 913–916.e7. [Google Scholar] [CrossRef]
Figure 1. Microbiota–gut–brain axis. The gut–brain axis is a bidirectional pathway that integrates signals from the VN, hormones (e.g., GLP-1, PYY, CCK, ghrelin) secreted from the EECs and signals from the ENS. The microbiota and its metabolites, such as SCFAs, can influence the gut–brain axis through modulating these pathways. Gut permeability due to altered gut microbial composition in obesity leads to LPS leakage and endotoxemia, which in turn can induce peripheral and neuroinflammation. GLP-1, glucagon-like peptide 1; PYY, peptide YY; CCK, cholecystokinin; SCFAs, short-chain fatty acids; LPS, lipopolysaccharide; EEC, enteroendocrine cell; ENS, enteric nervous system.
Figure 1. Microbiota–gut–brain axis. The gut–brain axis is a bidirectional pathway that integrates signals from the VN, hormones (e.g., GLP-1, PYY, CCK, ghrelin) secreted from the EECs and signals from the ENS. The microbiota and its metabolites, such as SCFAs, can influence the gut–brain axis through modulating these pathways. Gut permeability due to altered gut microbial composition in obesity leads to LPS leakage and endotoxemia, which in turn can induce peripheral and neuroinflammation. GLP-1, glucagon-like peptide 1; PYY, peptide YY; CCK, cholecystokinin; SCFAs, short-chain fatty acids; LPS, lipopolysaccharide; EEC, enteroendocrine cell; ENS, enteric nervous system.
Ijms 22 02993 g001
Figure 2. Possible mechanisms through which the gut microbiota could affect the homeostatic and hedonic pathways. The homeostatic pathway includes the processing of signals from white adipose tissue-derived leptin and GIT-produced peptides, PYY, CCK, GLP-1, and ghrelin, directly or through the VN. The hedonic pathway, which includes motivation- and reward-driven behavior through dopaminergic signaling, can be stimulated by food intake and visual and olfactory stimuli. Although the gut microbiota’s role in these pathways is still largely uncertain, several mechanisms have been hypothesized, indicated by the red dotted arrows. Altered gut microbiota composition could affect plasma leptin concentrations and sensitivity and alter the release of peptides. Additionally, microbial dysbiosis can lead to neuroinflammation, which could possibly alter both the homeostatic pathway and central dopaminergic signaling. GIT, gastrointestinal tract; ARC, arcuate nucleus; NPY, neuropeptide Y; AgRP, agouti-related protein; POMC, pro-opiomelanocortin; CART, cocaine- and amphetamine-regulated transcript; PYY, peptide YY; CCK, cholecystokinin; GLP-1, glucagon-like peptide-1.
Figure 2. Possible mechanisms through which the gut microbiota could affect the homeostatic and hedonic pathways. The homeostatic pathway includes the processing of signals from white adipose tissue-derived leptin and GIT-produced peptides, PYY, CCK, GLP-1, and ghrelin, directly or through the VN. The hedonic pathway, which includes motivation- and reward-driven behavior through dopaminergic signaling, can be stimulated by food intake and visual and olfactory stimuli. Although the gut microbiota’s role in these pathways is still largely uncertain, several mechanisms have been hypothesized, indicated by the red dotted arrows. Altered gut microbiota composition could affect plasma leptin concentrations and sensitivity and alter the release of peptides. Additionally, microbial dysbiosis can lead to neuroinflammation, which could possibly alter both the homeostatic pathway and central dopaminergic signaling. GIT, gastrointestinal tract; ARC, arcuate nucleus; NPY, neuropeptide Y; AgRP, agouti-related protein; POMC, pro-opiomelanocortin; CART, cocaine- and amphetamine-regulated transcript; PYY, peptide YY; CCK, cholecystokinin; GLP-1, glucagon-like peptide-1.
Ijms 22 02993 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

van Son, J.; Koekkoek, L.L.; La Fleur, S.E.; Serlie, M.J.; Nieuwdorp, M. The Role of the Gut Microbiota in the Gut–Brain Axis in Obesity: Mechanisms and Future Implications. Int. J. Mol. Sci. 2021, 22, 2993. https://doi.org/10.3390/ijms22062993

AMA Style

van Son J, Koekkoek LL, La Fleur SE, Serlie MJ, Nieuwdorp M. The Role of the Gut Microbiota in the Gut–Brain Axis in Obesity: Mechanisms and Future Implications. International Journal of Molecular Sciences. 2021; 22(6):2993. https://doi.org/10.3390/ijms22062993

Chicago/Turabian Style

van Son, Jamie, Laura L. Koekkoek, Susanne E. La Fleur, Mireille J. Serlie, and Max Nieuwdorp. 2021. "The Role of the Gut Microbiota in the Gut–Brain Axis in Obesity: Mechanisms and Future Implications" International Journal of Molecular Sciences 22, no. 6: 2993. https://doi.org/10.3390/ijms22062993

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop