Role of gut microbiota in the 5-hydroxytryptamine signal transduction mechanism | Metabolism and Translational Medicine

Role of gut microbiota in the 5-hydroxytryptamine signal transduction mechanism

Authors

  • gaofei Hu Peking University
  • Yujie Zhu
  • Shusi Ding
  • Lemin Zheng

DOI:

https://doi.org/10.54844/mtm.2023.0344

Keywords:

gut microbiota, 5-hydroxytryptamine, inflammation, 5-hydroxyindoleacetic acid

Abstract

As a neurotransmitter, 5-hydroxytryptamine (5-HT) plays a variety of regulatory roles in the brain, including affecting mood, memory, circadian rhythm, and other aspects. However, the brain contains only approximately 10% of the 5-HT in humans. Ninety percent of the 5-HT is produced and stored in the gut. 5-HT in the gut is involved in the regulation of irritable bowel syndrome (IBS), colitis, obesity, and other diseases. Therefore, a comprehensive understanding of the bioactivity of 5-HT is necessary. The gut microbiota has been reported to affect 5-HT synthesis and function, but the exact mechanism remains unclear. This review focuses on the production of 5-HT, its relationship with gut microbiota, its mechanism of action in different disease progression processes, and the biological effects of its derivative 5-hydroxyindoleacetic acid.

Downloads

Published

2023-06-30

How to Cite

1.
Hu gaofei, Zhu Y, Ding S, Zheng L. Role of gut microbiota in the 5-hydroxytryptamine signal transduction mechanism. Metabol Transl Med. 2023;1. doi:10.54844/mtm.2023.0344

Issue

Section

Review Article

Downloads

Download data is not yet available.
REVIEW ARTICLE

Role of gut microbiota in the 5-hydroxytryptamine signal transduction mechanism


Gaofei Hu1, Yujie Zhu1, Shusi Ding2, Lemin Zheng1,2,*

1The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China

2China National Clinical Research Center for Neurological Diseases, Tiantan Hospital; Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing 100070, China


*Corresponding Author:

Lemin Zheng, The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Health Sciences Center Peking University, NO.38, Xueyuan Road, Haidian District, Beijing 100191, China. Email: zhengl@bjmu.edu.cn; https://orcid.org/0000-0003-4473-6936


Received: 26 February 2023 Revised: 10 April 2023 Accepted: 15 May 2023 Published: 30 June 2023


ABSTRACT

As a neurotransmitter, 5-hydroxytryptamine (5-HT) plays a variety of regulatory roles in the brain, including affecting mood, memory, circadian rhythm, and other aspects. However, the brain contains only approximately 10% of the 5-HT in humans. Ninety percent of the 5-HT is produced and stored in the gut. 5-HT in the gut is involved in the regulation of irritable bowel syndrome (IBS), colitis, obesity, and other diseases. Therefore, a comprehensive understanding of the bioactivity of 5-HT is necessary. The gut microbiota has been reported to affect 5-HT synthesis and function, but the exact mechanism remains unclear. This review focuses on the production of 5-HT, its relationship with gut microbiota, its mechanism of action in different disease progression processes, and the biological effects of its derivative 5-hydroxyindoleacetic acid.

Key words: gut microbiota, 5-hydroxytryptamine, inflammation, 5-hydroxyindoleacetic acid

INTRODUCTION: GUT MICROBIOTA

As a research hotspot in recent years, the gut microbiota is active in nutrition, immunology, neurology, and other fields. Due to the wide variety and large number of gut microbiotas, people are driven to study their interaction with humans. Generally, the bacteria in the human intestine are mainly from Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Verrucomicrobia, and Fusobacteria, among which Firmicutes and Bacteroidetes dominate the ecological niche.[1] For a long time, it has been believed that the fetus is sterile when it is still in the mother, but researchers have found bacteria and other microorganisms in the placenta, umbilical cord, amniotic fluid, and even the meconium of newborns.[2,3] It can be said that the gut microbiota accompanies life, and it will constantly change dynamically with changes in individual diet and living habits regardless of infancy, youth, or old age.[4] It is difficult to elaborate on the mechanisms by which the gut microbiota affects humans because the gut microbiota may play a regulatory role either directly or indirectly. Kuziel et al.[5] divided the direct regulation of microorganisms into three situations: small molecule metabolites, ligands for pattern recognition, and molecules that act by influencing adaptive immunity. Because of the existence of various complex regulatory mechanisms, the gut microbiota is also known as an important source of metabolites in humans. Currently, more in-depth studies include short-chain fatty acids (SCFAs) such as acetic acid, propionic acid and butyric acid, 5-hydroxytryptamine (5-HT, serotonin) metabolized from tryptophan, and a variety of bile acids. Under normal circumstances, SCFAs can be metabolized by intestinal bacteria through dietary fiber and interact with the G protein-coupled receptor (GPCR) 41, GPCR43, and GPCR109A on the surface of intestinal epithelial cells and immune cells, which play an important role in maintaining the homeostasis of the internal environment.[6] In addition, branched-chain fatty acids (BCFAs), such as isobutyrate, 2-methylbutyrate, and isovalerate, as well as propionate intermediates lactic acid and succinate, can still be produced by some bacteria and exert biological effects on the human despite their relatively low abundance.[7]

Tryptophan in humans is mainly involved in metabolic pathways, including kynurenine, indole derivatives, and 5-HT, which have been proven to be directly or indirectly controlled by gut microbiota. The critical role of the gut microbiota in stimulating the activity of indoleamine 2,3-dioxygenase 1 (IDO1), a rate-limiting enzyme of kynurenine metabolism, has been clearly demonstrated, and the end products of its metabolism can be associated with host neurotransmission, inflammation, and immune responses.[8] Indole derivatives are molecules that are directly converted by the gut microbiota. For example, indole-3-aldehyde (IAld), indole-3-acid-acetic acid (IAA), indole-3-propionic acid (IPA), indole-3-acetaldehyde (IAAld), and indole-acrylic acid are all ligands of aryl hydrocarbon receptor (AhR).[9,10] This signal often acts on epithelial cell renewal, barrier integrity, and many immune cells.[11] 5-HT is also an important gastrointestinal signaling molecule that can transmit signals from the intestine to the nervous system by acting on at least fourteen 5-HT receptor subtypes,[12] affecting intestinal peristalsis and movement, secretion,[8] platelet aggregation,[13] cardiac function[14] and immune response.[15]

5-HT PRODUCTION PATHWAY

Most of the 5-HT in the human body is synthesized, stored, and released by a special intestinal epithelial cell subtype, enterochromaffin cells, which usually exist in the intestinal mucosal epithelium.[12] Enterochromaffin cells use Trp hydroxylase 1 (TpH1) to generate 5-hydroxytryptophan (5-HTP), which then generates 5-HT under the action of aromatic amino acid decarboxylase.[8] In addition, a small part of 5-HT is produced in the brain as a neurotransmitter through Trp hydroxylase 2 (TpH2) in serotonergic neurons, which plays an important role in neuroregulation. 5-HT in the gut also mediates cell-to-cell signaling, although 5-HT produced in the periphery cannot enter the brain because of the blood-brain barrier. Enterochromaffin cells act as sensors in the gut. When chemical changes or mechanical stimuli occur in the environment,[16] enterochromaffin will capture the signal and selectively release 5-HT. Studies have shown that the basement membrane is the main site of 5-HT release in enterochromaffin cells, as in most types of enteroendocrine cells.[17] The secreted 5-HT reaches the lumen of the gastrointestinal tract and the bloodstream, where it is absorbed and concentrated by platelets. Because platelets lack the TpH to produce 5-HT, they cannot synthesize 5-HT, so 5-HT entering the circulatory system is the only source of 5-HT.[12] 5-HT that arrives in the gut will exert its biological function through 5-hydroxytryptamine receptors (5-HTR). For example, motor neurons express 5-HTR1A, 5-HTR1B/1D, 5-HTR2A, 5-HTR2B, 5-HTR3 and 5-HTR4, and intestinal muscle cells express 5-HTR1B/1D, 5-HTR2A, 5-HTR2B, 5-HTR4 and 5-HTR7. Primary afferent neurons (PANs) that affect intestinal sensitivity express 5-HTR3, 5-HTR4, and 5-HTR7.[18] Because of the existence of multiple receptors, 5-HT can play different regulatory functions. In addition, since there is no extracellular enzyme for 5-HT degradation, transporters are needed to recycle 5-HT and then allow intracellular enzymes to play a role in degradation. The serotonin reuptake transporter (SERT) serves this function and is expressed in almost all intestinal epithelial cells to transport 5-HT through a sodium- and chlorine-dependent mechanism, thereby removing excess 5-HT from the interstitium.[1922] This clearance is important for 5-HT-mediated signaling.

5-HYDROXYINDOLEACETIC ACID (5-HIAA)

5-HIAA is a product of 5-HT metabolism by monoamine oxidase (MAO). Early studies focused on the correlation between the decreased content of 5-HIAA in cerebrospinal fluid (CSF) and mental diseases such as suicide and depression. The most common biomarker of suicide was the decreased concentration of 5-HIAA in CSF of suicide cases.[23] It was also found that the level of 5-HIAA in CSF was lower in suicide attempters, especially in violent suicide attempters.[24] The accumulation of HIAA in manic depressive patients may decrease in both manic and depressive periods.[25] In addition, lower CSF HIAA levels, a previous history of depression, and greater functional disability are associated with a higher risk of depression in Parkinson's disease (PD).[26] Here, 5-HIAA reflects the turnover and metabolism of 5-HT, especially the decreased content in CSF, which indicates the deficiency of the central nervous system (CNS) 5-HT function, which is the basis of some recurrent psychopathological disorders.[27] 5-HIAA is a detection indicator of 5-HT, which also appears in the study of gastrointestinal and pancreatic carcinoid endocrine tumors.[28] Previous studies have found elevated plasma 5-HIAA concentrations in patients with metabolic syndrome, suggesting that 5-HT may play an important role in the development of cardiovascular disease in patients with metabolic syndrome. Fasting blood glucose was positively correlated with the logarithm of plasma 5-HIAA concentration, while high-density lipoprotein cholesterol was negatively correlated.[29] Similarly, the authors suggested that 5-HIAA is a level indicator of 5-HT, suggesting that 5-HT mediates vasoconstriction and induces platelet activation, which may promote atherosclerosis.

5-HIAA is considered an indicator of 5-HT degradation, but its function as a metabolite itself has not been experimentally proven. It has been considered a waste metabolite of 5-HT and has no biological activity. Recent experimental studies have revealed novel biological functions of 5-HIAA. Rosser et al.[30] found that butyrate supplementation inhibited arthritis by increasing the level of the 5-HT-derived metabolite 5-HIAA and activating the AhR, a newly identified transcriptional marker of regulatory B-cell function, as well as inhibiting germinal center B-cell and plasmablastic differentiation. Giovanni et al.[31] found that 5-HIAA is a GPCR35 ligand and neutrophil chemoattractant and determined the role of 5-HIAA produced by platelets and mast cells in cell recruitment to inflammatory sites and bacterial clearance. Interestingly, in a transgenic APPSWE mouse Alzheimer's disease (AD) model and a phosphamide-induced brain neutral endopeptidase inhibition mouse model, repeated chronic treatment with 5-HIAA significantly reduced brain amyloid β-protein (Aβ) and improved the cognitive ability of APPSWE mice. 5-HIAA inhibited the mitogen-activated extracellular signal-regulated kinase/extracellular signal-regulated kinase (MEK/ERK) pathway. This mechanism appears to interact with the mitogen-activated protein kinase (MAPK) pathway.[32] Since then, 5-HIAA should no longer be regarded as an inactive metabolite of 5-HT, and the focus should be shifted to its biological function. The next step is to study its extensive and sufficient mechanism.

GUT MICROBIOTA AFFECT 5-HT PRODUCTION

It has been shown that 5-HT synthesis in the gut is regulated by the gut microbiota (Figure 1). Using TpH1 knockout mice, it was found that the level of 5-HT in the gut and blood decreased by more than 90%,[33] indicating that less than 10% of peripheral 5-HT is directly through the synthesis of microorganisms in mice or TpH2-mediated biosynthesis. By using germ-free (GF) mice, it can be found that the production process of 5-HT in the colon of GF mice is impaired and the concentration of 5-HT in the blood is decreased,[34] which verifies that the gut microbiota is involved in 5-HT synthesis to a certain extent. Current studies suggest that gut microbiota may induce the transcription of TpH1, thereby affecting the production of 5-HT in the gut. In addition, SCFAs produced by microbiota have also been reported to induce the expression of TpH1 in endocrine tumor BON and RIN14B cells.[7] Some secondary bile acids transformed by microorganisms, such as deoxycholic acid, also stimulated the production of 5-HT.[34]In vitro culture, Corynebacterium spp., Streptococcus spp., Escherichia coli, etc., can synthesize 5-HT.[35] However, this process does not depend on TpH, instead, tryptophan decarboxylates to form tryptamine. At present, there is no gut microbiota that can directly synthesize 5-HT in the human intestine. However, Valles-Colomer et al.[36] found that nearly 20% of gut bacteria have the potential to synthesize 5-HT using metagenomic data from a large cohort, which also provides direction and guidance for further exploration of the role of gut microbiota in the 5-HT pathway in the future. Selective serotonin reuptake inhibitors (SSRIs), a class of drugs commonly used to treat patients with depression, tend to increase the concentration of 5-HT in the environment by blocking reuptake during cellular signaling. Lukić et al.[37] found that the use of various selective serum reabsorption inhibitors in mice significantly changed the microbial composition in the body, and supplementation of Ruminococcus flavefaciens with duloxetine weakened the antidepressant treatment effect of the drug. This effect may be induced by changes in synaptic and mitochondrial gene expression to alter neuronal function.

Figure 1

Figure 1. Gut microbiota stimulates 5-HT production by producing short-chain fatty acids and bile acids. In intestinal enterochromaffin cells, tryptophan generates 5-HTP under the action of the TpH1 enzyme and is then metabolized to 5-HT by AAAD. In addition to being stored in intestinal enterochromaffin cells, 5-HT also enters the intestinal lumen and blood and is absorbed by blood platelets. Furthermore, 5-HT is also metabolized to 5-HIAA by MAO in the liver. Although 5-HIAA has long been considered a metabolic waste, its biological functional activity has not been realized until recently. SCFAs: short-chain fatty acids; TpH1: Trp hydroxylase 1; 5-HTP: 5-hydroxytryptophan; AAAD: Aromatic L-amino acid decarboxylase; 5-HT: 5-hydroxytryptamine; MAO: monoamine oxidase; 5-HIAA: 5-hydroxyindoleacetic acid.

5-HT AND DISEASE

Irritable bowel syndrome (IBS)

As a common gastrointestinal disease, IBS is a cluster of symptoms caused by a variety of diseases. Its clinical manifestations are usually intermittent episodes of abdominal pain, abdominal distension, and changes in bowel habits and stool traits. Since patients with this disease often experience pain or discomfort, their quality of life is significantly reduced.[3840] At present, the pathogenesis of the disease is not clear, so it is generally only symptomatic treatment, that is, treating the symptoms rather than the root cause. Some pathophysiological studies on IBS have found that patients' intestinal motility capacity changes and sensitivity increases, and they are more likely to be "stressed" by some external factors that cause gastrointestinal discomfort. Among these factors are changes in mood, such as depression, anxiety, and depression, which have been shown to be related to the 5-HT system.[41] Coates et al.[22] found that patients with ulcerative colitis (UC), irritable bowel syndrome with diarrhea (IBS-D), and irritable bowel syndrome with constipation (IBS-C) had decreased signals regarding 5-HT in rectal samples compared with healthy controls (HCs). Including mucosal 5-HT, TpH1 mRNA, 5-HT transporter mRNA, and 5-HT transporter immunoreactivity were significantly reduced. The researchers believe that these results suggest that alterations in the 5-HT system in the intestinal wall may directly contribute to the symptoms associated with IBS and other gastrointestinal dysfunction. However, studies have reported an increasing trend of 5-HT levels in the colon of patients with IBS, possibly due to the decreased expression of SERT in the colon,[42] which makes monoamine oxidase A (MAOA), which should be responsible for degrading 5-HT in the colon tissue, unable to play its role. SERT knockout mice also suffer from watery diarrhea, enhanced colonic motility, and visceral allergy.[21] These results suggest that when the absorption capacity of 5-HT by colonic cells decreases, the concentration of 5-HT increases, which may promote the development of IBS.

Cao et al.[43] used the supernatant of Lactobacillus rhamnosus GG to treat a rat model of post-infectious irritable bowel syndrome (PI-IBS) induced by Campylobacter jejuni infection and found that SERT mRNA and protein levels in intestinal tissues were higher than those in control and phosphate buffered saline (PBS) gavage rats. Gao et al.[44] found that gavage of fecal supernatant from IBS-D patients to young mice resulted in a significant decrease in SERT expression and an increase in 5-HT levels in the colonic mucosa of mice. The mechanism is that endotoxin in the IBS environment stimulates the synthesis and release of prostaglandin E2 (PGE2) from enteroblasts, which then acts on intestinal epithelial cells to downregulate SERT, leading to an increase in mucosal 5-HT. To test this hypothesis, mice were intragastrically injected with the lipopolysaccharide (LPS)-containing antagonist lipopolysaccharide-Rhodobacter sphaeroides (LPS-RS) or with LPS-removed fecal supernatant and were found to prevent the downregulation of SERT induced by IBS-D fecal supernatant. These studies suggest that intestinal microbiota disorder can affect the normal function of 5-HT signaling, which provides a new idea for the treatment of IBS.

Obesity

With the continuous improvement of living standards and changes in eating habits, obesity has become an urgent problem to be solved worldwide.[45] Obese people tend to have a higher risk of metabolic diseases, including heart disease, atherosclerosis, diabetes, and fatty liver, than HCs. Since obesity is caused in most cases by an imbalance between energy intake and expenditure, it is important to understand the causes of excess nutrition and the factors driving food intake.[46] It is known that 5-HT produced in the brain uses CNS to reduce appetite to regulate energy balance. Since 5-HT produced in the periphery cannot cross the blood-brain barrier, although the gut microbiota can affect the production of 5-HT in the gut, it is still not directly involved in the regulation of the CNS. However, there is considerable evidence that peripheral 5-HT is closely related to energy metabolism. Simansky et al.[47] used rats as a model and found that rats achieved satiety faster in a behaviorally specific manner by injecting 5-HT in the periphery. Crane et al.[48] used mice with TpH1 gene knockout or mice with LP533401, a small molecule that inhibits gut-derived TpH1, to feed a high-fat diet (HFD) and found that obesity, insulin resistance, and non-alcoholic fatty liver disease (NAFLD) could be prevented, while brown adipose tissue (BAT) exhibited greater energy expenditure. The specific mechanism is to protect or reverse the development of HFD-induced obesity and blood glucose by activating uncoupling protein-1 (UCP1)-mediated thermogenesis. Related experiments in Caenorhabditis elegans have shown that, unlike the regulation of feeding, the regulation of fat by 5-HT relies on a 5-HT-gated chloride channel encoded by mod-1 and a GPCRs initiated by the ser-6 signaling cascade, which ultimately promotes lipolysis around fat storage.[49] These studies suggest that peripheral 5-HT may affect host metabolism in a manner independent of central effects.

Recent studies have targeted 5-HTRs, which have been reported to ameliorate insulin resistance by inhibiting 5-HT signaling in adipose tissue and thereby reducing lipolysis in visceral adipocytes. Treatment of HFD-fed mice with an antagonist of 5-HTR2B was found to improve insulin resistance, visceral adipose tissue inflammation, and hepatic steatosis.[50] Increased 5-HTR2B expression was also found in white adipose tissue of obese individuals and was positively correlated with metabolic parameters. In addition, there are also studies on 5-HTR6, which are targeted to develop a new generation of safe and more effective anti-obesity drugs.[51] The gut microbiota, as an important factor affecting 5-HT signaling, may also be used as a target to develop biological oral agents for the treatment of obesity in the future.

Colitis and other inflammatory related diseases

Increasing evidence suggests that immune cells in the peripheral system, including macrophages, T cells, and dendritic cells, may be involved in the production, storage, and function of 5-HT.[52] Aune et al.[53] found that inhibiting the synthesis of 5-HT could inhibit the proliferation of human T cells stimulated by interleukin-2 (IL-2), while adding 5-HTP, a precursor of 5-HT, could reverse the inhibitory effect on T cell proliferation. In addition, 5-HTR1A receptor antagonists also reduced T cell cytokine production in vitro in mouse models, thereby reducing cell-mediated immunity and in vivo contact sensitivity responses. Similar results were found in the study of the 5-HTR1B receptor, where Yin et al.[54] used an antagonist of the 5-HT1B receptor to inhibit the proliferation of human and mouse primary helper T cells and human helper T cell lines. The 5-HT2A receptor was found to be an immune activator. When treated with a 5-HT2A receptor agonist and depleted of 5-HT in T cells of C57BL/6 mice, the production of IL-2 and interferon-γ (IFN-γ) in T cells was enhanced.[55] Not only does the 5-HT receptor exist on the surface of T cells, but the SERT responsible for transport is also expressed. Studies using the selective 5-HT reuptake inhibitor fluoxetine in rats have found that it can significantly increase the expression of the 5-HT transporter and the number of lymphocytes and may lead to a significant reduction in the concentration of cyclic adenosine monophosphate (cAMP) in lymphocytes and inhibit T cell activation and proliferation.[56] Both human and mouse T cells have the potential to store, degrade and synthesize 5-HT. O'Connell et al.[57] suggested that at the site of inflammation, activated T cells first release 5-HT, and dendritic cells can absorb, store and retransport 5-HT to naive T cells, thereby regulating the proliferation and differentiation of T cells. The involvement of T cells in the development of intestinal inflammation is also mediated by 5-HT. In inflammatory bowel disease (IBD), Th1 and Th17 helper cells often overreact. When TpH1 is knocked down in colitis mice, the secretion of inflammatory factors by dendritic cells decreases, and the levels of the proinflammatory cytokines IL-17 and IFN-γ secreted by T cells also decrease.[58] When 5-HT is supplemented, both the inflammatory response and disease progression are promoted, indicating that 5-HT plays an important role in the pathogenesis of colitis. In addition to TpH1 knockout mice, Ghia et al. also used a 5-HT synthesis inhibitor to construct an experimental colitis model of para-chlorophenylalanine (PCPA) and found that 5-HT stimulated macrophages in mice to produce pro-inflammatory cytokines, a process that was inhibited by inhibitors of nuclear factor κB.[59] The 5-HT precursor, 5-HTP, restores 5-HT levels and exacerbates colitis in TpH1 knockout mice. In addition, the synthetic food colorant Allura Red AC (ARAC) has been found to induce colitis through 5-HT in the colon.[60]

5-HT also plays an important role in other types of immune cells, such as macrophages, basophils, monocytes, and mast cells. Durk et al.[61] found mRNA expression of 5-HTR1E, 5-HTR2A, 5-HTR3, 5-HTR4, and 5-HTR7 in monocytes by reverse transcription and polymerase chain reaction (PCR). Among them, 5-HTR3, 5-HTR4, and 5-HTR7 are the main receptors of 5-HT for monocytes, which can activate and regulate the release of different chemokines and inflammatory factors. In macrophages, 5-HT can inhibit the release of inflammatory factors induced by LPS and upregulate M2 polarization-related genes such as SERPINB2, THBS1, STAB1, and COL23A1. The expression of M1-related genes such as INHBA, CCR2, MMP12, SERPINE1, CD1B and ALDH1A2 was decreased.[62] In addition, 5-HTR2B and 5-HTR7 are involved in the maintenance of the anti-inflammatory state of macrophages. Mast cells can produce 5-HT by expressing TpH1,[63] and the expression level of TpH1 is higher than that of other immune cells.[64] Kushnir-Sukhov et al.[62] also found that mast cells express multiple 5-HTRs, including 5-HTR1A, 5-HTR1B, 5-HTR1E, 5-HTR2A, and 5-HTR2B and identified 5-HTR1A as the main receptor mediating the effect of 5-HT on mast cells. When the 5-HTR1A gene is knocked down, the accumulation of mast cells (MCs) induced by 5-HT in the mouse dermis is inhibited. The relationship between gut microbiota and the peripheral immune system is complex and important. Not only 5-HT but also intestinal bacteria play a role in regulating inflammation-related factors by producing SCFAs, trimethylamine N-oxide, and reactive oxygen species.[65] Therefore, a better understanding of the functional mechanisms of the gut microbiota in various immune responses is necessary for determining the development of diseases.

Central nervous system diseases

In the CNS, most serotonergic neurons are located in the nuclei of the median raphe, including the descending medullary neurons of the raphe obscurus, magnus, pallidus, and their ascending pontine and mesencephalic counterparts in the raphe pontis, centralis, and dorsalis. Descending projections of the raphe nucleus affect spinal cord and brain stem mechanisms, which play an important role in central regulation of pain and pathological pain syndromes, in many cases originating from the gut. Ascending raphe projections point to higher integrated functional areas and are associated with advanced nervous function, including the regulation of mood, sleep, sex, and appetite.[12]

In addition, since 5-HT is impermeable across the blood-brain barrier, central serotonergic neurons are separated from peripheral serotonergic neurons, platelets, and enterochromaffin cells. It is believed that enterochromaffin cell-neural junctions are very different from neuromuscular junctions or any other synapse in the central or peripheral nervous system. 5-HT secreted by enterochromaffin cells acts locally in a paracrine manner. Intestinal serotonergic neurons are considered descending interneurons that innervate follower cells in both plexuses, and they do not project to smooth muscle or other effectors. Enterochromaffin cells are moving like the rest of the gastrointestinal epithelium, and nerves are not good at innervating moving targets. Therefore, conventional synapses cannot be found on them. In addition, enterochromaffin cells cannot focus their released transmitters as precisely as neurons. Actually, the high secretion of 5-HT by enterochromaffin cells may compensate for the lack of close enterochromaffin cell-neuron connections. Thus, the concentration of local 5-HT in the mucosa is quite high, suggesting that the function of mucosal 5-HT is unlikely to be limited to nerve stimulation. For example, 5-HT is thought to affect the secretion and proliferation of crypt epithelial cells.[12]

In the gut, 5-HT activates both intrinsic and extrinsic PANs to regulate various physiological processes, including inflammatory responses, intrinsic peristaltic and secretory reflexes, and activation of extrinsic sensory nerves that transmit information to the brain, leading to the sensation of nausea and discomfort in the sensory CNS.[12,66] Cirrito et al. also found that 5-HT signaling may have the ability to alter Aβ levels and plaques in the brain. They gave mice SSRI and found that Aβ levels in the interstitial fluid were reduced by 25%. Direct injection of 5-HT into the hippocampus also reduced Aβ levels in the interstitial fluid.[67] A clinical cohort study of patients with PD has also shown an association between 5-HT and tremor behavior in patients with PD. Loss of serotonin transporters in the brain stem may aggravate the severity of tremor.[68] Based on this result, drugs that regulate 5-HT signal should be investigated in the treatment of Parkinson's related tremor. Among the receptors, 5-HTR3 and 5-HTR4 are closely related to functional gastrointestinal disorders.[69] Electrophysiological and anatomical studies have shown that 5-HTR4 is located in nerve terminals throughout the enteric nervous system (ENS) and mediates excitatory synaptic transmission. Activation of these presynaptic 5-HTR4 leads to an increase in the number of transmitters, such as acetylcholine, released from these terminals, which can enhance the reflex response.[7072]

In IBS, inhibition of 5-HTR3 located on intrinsic sensory neurons reduces motor and secretory reflex activity and reduces activation of extrinsic sensory neurons that transmit signals to the CNS, suppressing sensory signals that cause pain and discomfort.[73] In rotavirus-induced diarrhea, 5-HTR3 has been shown to play a role by influencing intestinal motility, sensory neuron activation, cytokine secretion, and inflammatory responses, resulting in a reduced number of diarrhea days and diarrhea mouse counts in 5-HT3 receptor knockout mice compared with wild-type mice. However, vagal signaling to the vomiting center also occurs in the absence of 5-HTR3, indicating a complicated interaction between intestinal 5-HT and neural systems in the gut.[69,74,75]

Depression

Depression is a major cause of disability worldwide and is a heterogeneous suite of states characterized by sad mood and anhedonia (an inability to experience pleasure). Currently, depression is generally considered a systemic disorder that can be triggered by different factors that ultimately target the 5-HT system in vulnerable individuals.[7678] The therapeutic agents for treating depression are mainly antidepressants, most of which are SSRIs or serotonin-norepinephrine reuptake inhibitors (SNRIs). These agents are thought to act in part by increasing synaptic levels of monoamines, primarily 5-HT and norepinephrine, and by subsequent activation of serotonergic and noradrenergic postsynaptic receptors and autoreceptors. The therapeutic benefits of increased levels of monoamines were discovered through the efficacy of MAO inhibitors and tricyclic antidepressants in treating depression. However, antidepressants are only partially effective in moderate and more severe adult depression, suggesting that the monoamine hypothesis only partially explains depression. Although the idea that a single neurochemical causes depression is now considered simplistic, the low 5-HT hypothesis (also referred to as the "monoamine hypothesis") still lies at the foundation of most research on depression.[79,80]

The bidirectional crosstalk between the CNS, ENS, and gastrointestinal tract is referred to as the gut-brain axis (GBA), which connects the emotional and cognitive centers of the brain with the peripheral functions of the gastrointestinal tract. Accumulating evidence supports the theory that the gut microbiota plays an important role in the modulation of the GBA by directly or indirectly affecting the production of metabolites.[66,7880] 5-HT is a key element of this axis and acts as a neurotransmitter in the CNS and ENS in the intestinal wall. Recent studies have shown that the gut microbiome or microbial species may affect the onset of depressive disorder by modulating neurotransmitters such as γ-aminobutyric acid (GABA) and 5-HT.

Using genome-wide shotgun metagenomics and untargeted metabolomics approaches, Yang et al.[81] identified significant differences in the abundance of gut microbiota between patients with major depressive disorder (MDD) and HCs. Patients with MDD were mainly characterized by increased abundance of the genus Bacteroides and decreased abundance of the genera Blautia and Eubacterium. Disrupted microbial genes and fecal metabolites were consistently mapped to amino acid (γ-aminobutyrate, phenylalanine, and tryptophan) metabolism. Therefore, a balance needs to be maintained between bacterial utilization of tryptophan and the tryptophan necessary for 5-HT synthesis in both enteric and CNS.

Given the role of 5-HT/tryptophan influenced by bacteria in the progression of depression disorder, manipulation of the gut microbiome can make a difference. In line with this notion, GF mice robustly and reproducibly display less anxiety-like or depression-like behaviors than their conventionally colonized counterparts,[82,83] with hippocampal concentrations of 5-HT and its major metabolite, 5-HIAA, significantly elevated in male GF animals,[84] while the anxious state can be reinstated with the introduction of a microbiota prior to critical time windows. Interestingly, although the peripheral availability of tryptophan returned to baseline values, colonization of GF animals after weaning was not sufficient to reverse the CNS neurochemical consequences of early deletion of microbiota in adulthood. Moreover, circulating levels of tryptophan decreased when these animals were introduced tryptophan metabolism-producing bacteria, such as B. infantis, into their gut.[70] Another study found that the gut microbiota from MDD patients transmits depressive behavior to AntiBiotics (ABX)-treated mice. Specifically, B. uniformis, B. fragilis, and B. caccae can impair hippocampal neurogenesis and reduce hippocampal 5-HT levels.[85] These results show that CNS neural conduction may be severely disturbed by the lack of normal gut microbiota.

In addition to probiotics that directly restore certain microbiota to retain the level of 5-HT,[85,86] drugs and prebiotics for interfering with 5-HT metabolism in the gut microbiota can also make sense. For example, Morinda officinalis oligosaccharides (MOOs), an oral drug approved in China for the treatment of depression, have been found to alleviate depression by increasing 5-HTP in the gut microbiota. Specifically, MOO increased tryptophan hydroxylase levels and inhibited 5-HTP decarboxylase activity in the gut microbiota, which accelerated 5-HTP production from tryptophan and reduced 5-HT generation, thus leading to the accumulation of 5-HTP. The increased 5-HTP from the gut microbiota was then absorbed to the blood, passed across the blood-brain barrier, and improved 5-HT levels in the brain.[87]

CONCLUSION

The mechanism of 5-HT action in the nervous system has been relatively well studied, and targeted inhibitors of the reuptake process are also used as drugs to treat patients with depression. Although the 5-HT produced in the gut cannot directly act on the brain through the blood-brain barrier, it plays a role in immune inflammation, obesity, IBS, and other aspects through a variety of 5-HTRs expressed in the intestinal tissue. With the understanding of the link between gut microbiota and 5-HT, gut bacteria are considered to be a key link in 5-HT signaling. It has also been found that the use of antidepressants targeting the 5-HT signaling pathway alters the composition of the gut microbiota, decreasing richness and increasing β diversity. However, the mechanisms by which antidepressants affect the microbiota remain elusive. Possible mechanisms include inhibitors acting as efflux pumps affecting quorum sensing in the microbiota community.[88]

Although a number of studies have systematically explained that gut microbiota affects 5-HT in the occurrence and development of various diseases, the specific molecular mechanism behind it remains to be studied. For example, which bacteria play the most important role in indirectly or even directly producing 5-HT, what enzymes are involved, and whether the key enzymes can be modified? In addition, with the continuous development of technology, more and more methods have been used to study gut microbiota. The cell wall of bacteria can be labeled by D-amino acids with fluorescence to observe the three-dimensional imaging of gastrointestinal bacteria in mice.[89] Unlike ABX, phage has host specificity, so researchers have made use of phage specific knockout bacteria, which can provide effective help to study the role of a certain type of bacteria in gut microbiota.[90] Furthermore, in the process of studying intestinal bacteria, how to make the bacteria better colonize the stomach is also an urgent problem to be solved. By using biological orthogonality, molecules capable of biological orthogonality reaction can be added to the surface of the bacteria to be intragastric and the original colonizing bacteria in the host intestinal tract respectively, so that the bacteria can better colonize.[91]

The relationship between the gut microbiota and host is complex and changeable, and a series of factors, such as diet, daily schedule, and emotional changes, can be sensitively captured and responded to by the gut microbiota. Whether it is 5-HT or SCFAs, the current understanding is still insufficient to regard gut microbiota as a separate means to treat related diseases, and more is to make probiotics for adjuvant treatment. It is very important to determine more possible mechanisms and bacterial species that play a dominant role in the corresponding regulatory system. At the same time, the use of some relatively simple artificial bacterial communities may help people understand the relationship between different bacterial species to better facilitate the regulation of their composition. Diseases such as colitis and IBS, which currently seriously affect the quality of life of patients, may be a breakthrough in the future because of the study of gut microbiota. An increasing number of researchers use more standardized and rigorous quantitative indicators and technical methods to explore the causal relationship between gut microbiota and host metabolism. It is believed that such strategies will greatly promote the development of gut microbiota.

DECLARATION

Acknowledgement

Parts of the figure were drawn using pictures from scidraw.io (upload by Chilton, 2020) and Servier Medical Art.

Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/ accessed on 29 June 2023).

Author contributions

Hu GF: Conceptualization, Investigation, Writing—Original draft. Zhu YJ: Data curation, Formal analysis. Ding SS: Methodology, Writing—Review and Editing. Zheng LM: Supervision, Project administration, Funding acquisition.

Source of funding

This work was supported by the National Natural Science Foundation of China (No.91639108, No.81770272, No.81970425, No.91839302), the National High Technology Research and Development Program of China (No.2020YFA0803700), Hangzhou Qianjiang Distinguished Expert Project (Prof. Lemin Zheng), and the Fundamental Research Funds.

Informed consent

Not applicable.

Ethical approval

Not applicable.

Conflict of interest

Lemin Zheng is the Editor-in-Chief of the journal. The article was subject to the journal's standard procedures, with peer review handled independently of the Editor-in-Chief and the affiliated research groups.

Data sharing

Not applicable.

REFERENCES

  1. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308(5728):1635–1638.    DOI: 10.1126/science.1110591    PMID: 15831718
  2. Jiménez E, Marín ML, Martín R, et al. Is meconium from healthy newborns actually sterile? Res Microbiol. 2008;159(3):187–193.    DOI: 10.1016/j.resmic.2007.12.007    PMID: 18281199
  3. Satokari R, Grönroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol. 2009;48(1):8–12.    DOI: 10.1111/j.1472-765X.2008.02475.x    PMID: 19018955
  4. Derrien M, Alvarez AS, de Vos WM. The gut microbiota in the first decade of life. Trends Microbiol. 2019;27(12):997–1010.    DOI: 10.1016/j.tim.2019.08.001    PMID: 31474424
  5. Kuziel GA, Rakoff-Nahoum S. The gut microbiome. Curr Biol. 2022;32(6):R257–R264.    DOI: 10.1016/j.cub.2022.02.023    PMID: 35349808
  6. Sivaprakasam S, Prasad PD, Singh N. Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis. Pharmacol Ther. 2016;164:144–151.    DOI: 10.1016/j.pharmthera.2016.04.007    PMID: 27113407
  7. Krautkramer KA, Fan J, Bäckhed F. Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol. 2021;19(2):77–94.    DOI: 10.1038/s41579-020-0438-4    PMID: 32968241
  8. Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6):716–724.    DOI: 10.1016/j.chom.2018.05.003    PMID: 29902437
  9. Alexeev EE, Lanis JM, Kao DJ, et al. Microbiota-derived indole metabolites promote human and murine intestinal homeostasis through regulation of interleukin-10 receptor. Am J Pathol. 2018;188(5):1183–1194.    DOI: 10.1016/j.ajpath.2018.01.011    PMID: 29454749
  10. Hubbard TD, Murray IA, Perdew GH. Indole and tryptophan metabolism: Endogenous and dietary routes to Ah receptor activation. Drug Metab Dispos. 2015;43(10):1522–1535.    DOI: 10.1124/dmd.115.064246    PMID: 26041783
  11. Lamas B, Natividad JM, Sokol H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal Immunol. 2018;11(4):1024–1038.    DOI: 10.1038/s41385-018-0019-2    PMID: 29626198
  12. Gershon MD, Tack J. The serotonin signaling system: From basic understanding to drug development for functional GI disorders. Gastroenterology. 2007;132(1):397–414.    DOI: 10.1053/j.gastro.2006.11.002    PMID: 17241888
  13. Mercado CP, Quintero MV, Li Y, et al. A serotonin-induced N-glycan switch regulates platelet aggregation. Sci Rep. 2013;3:2795.    DOI: 10.1038/srep02795    PMID: 24077408
  14. Côté F, Thévenot E, Fligny C, et al. Disruption of the nonneuronal tph1 gene demonstrates the importance of peripheral serotonin in cardiac function. Proc Natl Acad Sci U S A. 2003;100(23):13525–13530.    DOI: 10.1073/pnas.2233056100    PMID: 14597720
  15. Baganz NL, Blakely RD. A dialogue between the immune system and brain, spoken in the language of serotonin. ACS Chem Neurosci. 2013;4(1):48–63.    DOI: 10.1021/cn300186b    PMID: 23336044
  16. Raybould HE, Cooke HJ, Christofi FL. Sensory mechanisms: Transmitters, modulators and reflexes. Neurogastroenterol Motil. 2004;16(Suppl 1):60–63.    DOI: 10.1111/j.1743-3150.2004.00477.x    PMID: 15066007
  17. Mawe GM, Hoffman JM. Serotonin signalling in the gut—functions, dysfunctions and therapeutic targets. Nat Rev Gastroenterol Hepatol. 2013;10(8):473–486.    DOI: 10.1038/nrgastro.2013.105    PMID: 23797870
  18. Sanger GJ. 5-hydroxytryptamine and the gastrointestinal tract: where next? Trends Pharmacol Sci. 2008;29(9):465–471.    DOI: 10.1016/j.tips.2008.06.008    PMID: 19086255
  19. Wade PR, Chen J, Jaffe B, Kassem IS, Blakely RD, Gershon MD. Localization and function of a 5-HT transporter in crypt epithelia of the gastrointestinal tract. J Neurosci. 1996;16(7):2352–2364.    DOI: 10.1523/JNEUROSCI.16-07-02352.1996    PMID: 8601815
  20. Chen JX, Pan H, Rothman TP, Wade PR, Gershon MD. Guinea pig 5-HT transporter: Cloning, expression, distribution, and function in intestinal sensory reception. Am J Physiol. 1998;275(3):G433–G448.    DOI: 10.1152/ajpgi.1998.275.3.G433    PMID: 9724254
  21. Chen JJ, Li Z, Pan H, et al. Maintenance of serotonin in the intestinal mucosa and Ganglia of mice that lack the high-affinity serotonin transporter: Abnormal intestinal motility and the expression of cation transporters. J Neurosci. 2001;21(16):6348–6361.    DOI: 10.1523/JNEUROSCI.21-16-06348.2001    PMID: 11487658
  22. Coates MD, Mahoney CR, Linden DR, et al. Molecular defects in mucosal serotonin content and decreased serotonin reuptake transporter in ulcerative colitis and irritable bowel syndrome. Gastroenterology. 2004;126(7):1657–1664.    DOI: 10.1053/j.gastro.2004.03.013    PMID: 15188158
  23. Maris RW. Suicide. Lancet. 2002;360(9329):319–326.    DOI: 10.1016/S0140-6736(02)09556-9    PMID: 12147388
  24. Sharma R, Tikka SK, Yadav AK, Bhute AR, Dhamija P, Bastia BK. Cerebrospinal fluid monoamine metabolite concentrations in suicide attempt: A meta-analysis. Asian J Psychiatr. 2021;62:102711.    DOI: 10.1016/j.ajp.2021.102711    PMID: 34090249
  25. Jr WE, Wehr TR, Gillin JC, Post RM, Goodwin FK, van Kammen DP. The switch process in manic-depressive psychosis. Ann Intern Med. 1977;87(3):319–335.    DOI: 10.7326/0003-4819-87-3-319    PMID: 20015
  26. Cummings JL. Depression and Parkinson's disease: A review. Am J Psychiatry. 1992;149(4):443–454.
  27. Shannon C, Schwandt ML, Champoux M, et al. Maternal absence and stability of individual differences in CSF 5-HIAA concentrations in rhesus monkey infants. Am J Psychiatry. 2005;162(9):1658–1664.    DOI: 10.1176/appi.ajp.162.9.1658    PMID: 16135625
  28. Tomassetti P, Migliori M, Lalli S, Campana D, Tomassetti V, Corinaldesi R. Epidemiology, clinical features and diagnosis of gastroenteropancreatic endocrine tumours. Ann Oncol. 2001;12(Suppl 2):S95–S99.    DOI: 10.1093/annonc/12.suppl_2.s95    PMID: 11762360
  29. Fukui M, Tanaka M, Toda H, et al. High plasma 5-hydroxyindole-3-acetic acid concentrations in subjects with metabolic syndrome. Diabetes Care. 2012;35(1):163–167.    DOI: 10.2337/dc11-1619    PMID: 22100965
  30. Rosser EC, Piper CJM, Matei DE, et al. Microbiota-derived metabolites suppress arthritis by amplifying aryl-hydrocarbon receptor activation in regulatory B cells. Cell Metab. 2020;31(4):837–851.e10.    DOI: 10.1016/j.cmet.2020.03.003    PMID: 32213346
  31. Giovanni M, Tam H, Valet C, Xu Y, Looney MR, Cyster JG. GPR35 promotes neutrophil recruitment in response to serotonin metabolite 5-HIAA. Cell. 2022;185(5):815–830.e19.    DOI: 10.1016/j.cell.2022.01.010    PMID: 35148838
  32. Klein C, Roussel G, Brun S, et al. 5-HIAA induces neprilysin to ameliorate pathophysiology and symptoms in a mouse model for Alzheimer's disease. Acta Neuropathol Commun. 2018;6(1):136.    DOI: 10.1186/s40478-018-0640-z    PMID: 30537985
  33. Savelieva KV, Zhao S, Pogorelov VM, et al. Genetic disruption of both tryptophan hydroxylase genes dramatically reduces serotonin and affects behavior in models sensitive to antidepressants. PLoS One. 2008;3(10):e3301.    DOI: 10.1371/journal.pone.0003301    PMID: 18923670
  34. Yano JM, Yu K, Donaldson GP, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161(2):264–276.    DOI: 10.1016/j.cell.2015.02.047    PMID: 25860609
  35. Roshchina VV. New trends and perspectives in the evolution of neurotransmitters in microbial, plant, and animal cells. Adv Exp Med Biol. 2016;874:25–77.    DOI: 10.1007/978-3-319-20215-0_2    PMID: 26589213
  36. Valles-Colomer M, Falony G, Darzi Y, et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. 2019;4(4):623–632.    DOI: 10.1038/s41564-018-0337-x    PMID: 30718848
  37. Lukić I, Getselter D, Ziv O, et al. Antidepressants affect gut microbiota and Ruminococcus flavefaciens is able to abolish their effects on depressive-like behavior. Transl Psychiatry. 2019;9(1):133.    DOI: 10.1038/s41398-019-0466-x    PMID: 30967529
  38. Drossman DA, McKee DC, Sandler RS, et al. Psychosocial factors in the irritable bowel syndrome. A multivariate study of patients and nonpatients with irritable bowel syndrome. Gastroenterology. 1988;95(3):701–708.    DOI: 10.1016/s0016-5085(88)80017-9    PMID: 3396817
  39. Talley NJ, Boyce PM, Jones M. Predictors of health care seeking for irritable bowel syndrome: a population based study. Gut. 1997;41(3):394–398.    DOI: 10.1136/gut.41.3.394    PMID: 9378398
  40. Drossman DA, Patrick DL, Whitehead WE, et al. Further validation of the IBS-QOL: A disease-specific quality-of-life questionnaire. Am J Gastroenterol. 2000;95(4):999–1007.    DOI: 10.1111/j.1572-0241.2000.01941.x    PMID: 10763950
  41. Grayson M. Irritable bowel syndrome: 4 big questions. Nature. 2016;533(7603):S118.    DOI: 10.1038/533S118a    PMID: 27191493
  42. Dunlop SP, Coleman NS, Blackshaw E, et al. Abnormalities of 5-hydroxytryptamine metabolism in irritable bowel syndrome. Clin Gastroenterol Hepatol. 2005;3(4):349–357.    DOI: 10.1016/s1542-3565(04)00726-8    PMID: 15822040
  43. Cao YN, Feng LJ, Liu YY, et al. Effect of Lactobacillus rhamnosus GG supernatant on serotonin transporter expression in rats with post-infectious irritable bowel syndrome. World J Gastroenterol. 2018;24(3):338–350.    DOI: 10.3748/wjg.v24.i3.338    PMID: 29391756
  44. Gao J, Xiong T, Grabauskas G, Owyang C. Mucosal serotonin reuptake transporter expression in irritable bowel syndrome is modulated by gut microbiota via mast cell-prostaglandin E2. Gastroenterology. 2022;162(7):1962–1974.e6.    DOI: 10.1053/j.gastro.2022.02.016    PMID: 35167867
  45. Galen KA, Ter Horst KW, Booij J, la Fleur SE, Serlie MJ. The role of central dopamine and serotonin in human obesity: lessons learned from molecular neuroimaging studies. Metabolism. 2018;85:325–339.    DOI: 10.1016/j.metabol.2017.09.007    PMID: 28970033
  46. Swinburn B, Sacks G, Ravussin E. Increased food energy supply is more than sufficient to explain the US epidemic of obesity. Am J Clin Nutr. 2009;90(6):1453–1456.    DOI: 10.3945/ajcn.2009.28595    PMID: 19828708
  47. Simansky KJ, Jakubow J, Sisk FC, Vaidya AH, Eberle-Wang K. Peripheral serotonin is an incomplete signal for eliciting satiety in sham-feeding rats. Pharmacol Biochem Behav. 1992;43(3):847–854.    DOI: 10.1016/0091-3057(92)90417-e    PMID: 1448479
  48. Crane JD, Palanivel R, Mottillo EP, et al. Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis. Nat Med. 2015;21(2):166–172.    DOI: 10.1038/nm.3766    PMID: 25485911
  49. Srinivasan S, Sadegh L, Elle IC, Christensen AGL, Faergeman NJ, Ashrafi K. Serotonin regulates C. elegans fat and feeding through independent molecular mechanisms. Cell Metab. 2008;7(6):533–544.    DOI: 10.1016/j.cmet.2008.04.012    PMID: 18522834
  50. Choi WG, Choi W, Oh TJ, et al. Inhibiting serotonin signaling through HTR2B in visceral adipose tissue improves obesity-related insulin resistance. J Clin Invest. 2021;131(23):e145331.    DOI: 10.1172/JCI145331    PMID: 34618686
  51. Heal DJ, Smith SL, Fisas A, Codony X, Buschmann H. Selective 5-HT6 receptor ligands: Progress in the development of a novel pharmacological approach to the treatment of obesity and related metabolic disorders. Pharmacol Ther. 2008;117(2):207–231.    DOI: 10.1016/j.pharmthera.2007.08.006    PMID: 18068807
  52. Wu H, Denna TH, Storkersen JN, Gerriets VA. Beyond a neurotransmitter: The role of serotonin in inflammation and immunity. Pharmacol Res. 2019;140:100–114.    DOI: 10.1016/j.phrs.2018.06.015    PMID: 29953943
  53. Aune TM, Golden HW, McGrath KM. Inhibitors of serotonin synthesis and antagonists of serotonin 1A receptors inhibit T lymphocyte function in vitro and cell-mediated immunity in vivo. J Immunol. 1994;153(2):489–498.    PMID: 8021490
  54. Yin J, Albert RH, Tretiakova AP, Jameson BA. 5-HT(1B) receptors play a prominent role in the proliferation of T-lymphocytes. J Neuroimmunol. 2006;181(1-2):68–81.    DOI: 10.1016/j.jneuroim.2006.08.004    PMID: 17011639
  55. Inoue M, Okazaki T, Kitazono T, Mizushima M, Omata M, Ozaki S. Regulation of antigen-specific CTL and Th1 cell activation through 5-Hydroxytryptamine 2A receptor. Int Immunopharmacol. 2011;11(1):67–73.    DOI: 10.1016/j.intimp.2010.10.007    PMID: 20971187
  56. Fazzino F, Urbina M, Cedeño N, Lima L. Fluoxetine treatment to rats modifies serotonin transporter and cAMP in lymphocytes, CD4+ and CD8+ subpopulations and interleukins 2 and 4. Int Immunopharmacol. 2009;9(4):463–467.    DOI: 10.1016/j.intimp.2009.01.011    PMID: 19189865
  57. O'Connell PJ, Wang X, Leon-Ponte M, Griffiths C, Pingle SC, Ahern GP. A novel form of immune signaling revealed by transmission of the inflammatory mediator serotonin between dendritic cells and T cells. Blood. 2006;107(3):1010–1017.    DOI: 10.1182/blood-2005-07-2903    PMID: 16223770
  58. Li N, Ghia JE, Wang H, et al. Serotonin activates dendritic cell function in the context of gut inflammation. Am J Pathol. 2011;178(2):662–671.    DOI: 10.1016/j.ajpath.2010.10.028    PMID: 21281798
  59. Ghia JE, Li N, Wang H, et al. Serotonin has a key role in pathogenesis of experimental colitis. Gastroenterology. 2009;137(5):1649–1660.    DOI: 10.1053/j.gastro.2009.08.041    PMID: 19706294
  60. Kwon YH, Banskota S, Wang H, et al. Author Correction: chronic exposure to synthetic food colorant Allura Red AC promotes susceptibility to experimental colitis via intestinal serotonin in mice. Nat Commun. 2023;14(1):3125.    DOI: 10.1038/s41467-023-38903-w    PMID: 37253738
  61. Dürk T, Panther E, Müller T, et al. 5-Hydroxytryptamine modulates cytokine and chemokine production in LPS-primed human monocytes via stimulation of different 5-HTR subtypes. Int Immunol. 2005;17(5):599–606.    DOI: 10.1093/intimm/dxh242    PMID: 15802305
  62. de las Casas-Engel M, Domínguez-Soto A, Sierra-Filardi E, et al. Serotonin skews human macrophage polarization through HTR2B and HTR7. J Immunol. 2013;190(5):2301–2310.    DOI: 10.4049/jimmunol.1201133    PMID: 23355731
  63. Kushnir-Sukhov NM, Brown JM, Wu Y, Kirshenbaum A, Metcalfe DD. Human mast cells are capable of serotonin synthesis and release. J Allergy Clin Immunol. 2007;119(2):498–499.    DOI: 10.1016/j.jaci.2006.09.003    PMID: 17291861
  64. Kushnir-Sukhov NM, Gilfillan AM, Coleman JW, et al. 5-hydroxytryptamine induces mast cell adhesion and migration. J Immunol. 2006;177(9):6422–6432.    DOI: 10.4049/jimmunol.177.9.6422    PMID: 17056574
  65. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535(7610):65–74.    DOI: 10.1038/nature18847    PMID: 27383981
  66. O'Mahony SM, Clarke G, Borre YE, Dinan TG, Cryan JF. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:32–48.    DOI: 10.1016/j.bbr.2014.07.027    PMID: 25078296
  67. Cirrito JR, Disabato BM, Restivo JL, et al. Serotonin signaling is associated with lower amyloid-β levels and plaques in transgenic mice and humans. Proc Natl Acad Sci U S A. 2011;108(36):14968–14973.    DOI: 10.1073/pnas.1107411108    PMID: 21873225
  68. Pasquini J, Ceravolo R, Qamhawi Z, et al. Progression of tremor in early stages of Parkinson's disease: a clinical and neuroimaging study. Brain. 2018;141(3):811–821.    DOI: 10.1093/brain/awx376    PMID: 29365117
  69. Hagbom M, Hellysaz A, Istrate C, et al. The 5-HT3 receptor affects rotavirus-induced motility. J Virol. 2021;95(15):e0075121.    DOI: 10.1128/JVI.00751-21    PMID: 33980599
  70. Liu M, Geddis MS, Wen Y, Setlik W, Gershon MD. Expression and function of 5-HT4 receptors in the mouse enteric nervous system. Am J Physiol Gastrointest Liver Physiol. 2005;289(6):G1148–G1163.    DOI: 10.1152/ajpgi.00245.2005    PMID: 16037544
  71. Pan H, Galligan JJ. 5-HT1A and 5-HT4 receptors mediate inhibition and facilitation of fast synaptic transmission in enteric neurons. Am J Physiol. 1994;266(2 Pt 1):G230–G238.    DOI: 10.1152/ajpgi.1994.266.2.G230    PMID: 8141296
  72. Galligan JJ, Pan H, Messori E. Signalling mechanism coupled to 5-hydroxytryptamine4 receptor-mediated facilitation of fast synaptic transmission in the guinea-pig ileum myenteric plexus. Neurogastroenterol Motil. 2003;15(5):523–529.    DOI: 10.1046/j.1365-2982.2003.00428.x    PMID: 14507352
  73. Kozlowski CM, Green A, Grundy D, Boissonade FM, Bountra C. The 5-HT(3) receptor antagonist alosetron inhibits the colorectal distention induced depressor response and spinal c-fos expression in the anaesthetised rat. Gut. 2000;46(4):474–480.    DOI: 10.1136/gut.46.4.474    PMID: 10716675
  74. Hagbom M, Istrate C, Engblom D, et al. Rotavirus stimulates release of serotonin (5-HT) from human enterochromaffin cells and activates brain structures involved in nausea and vomiting. PLoS Pathog. 2011;7(7):e1002115.    DOI: 10.1371/journal.ppat.1002115    PMID: 21779163
  75. Cremon C, Carini G, Wang B, et al. Intestinal serotonin release, sensory neuron activation, and abdominal pain in irritable bowel syndrome. Am J Gastroenterol. 2011;106(7):1290–1298.    DOI: 10.1038/ajg.2011.86    PMID: 21427712
  76. Dell'Osso L, Carmassi C, Mucci F, Marazziti D. Depression, serotonin and tryptophan. Curr Pharm Des. 2016;22(8):949–954.    DOI: 10.2174/1381612822666151214104826    PMID: 26654774
  77. Andrews PW, Bharwani A, Lee KR, Fox M, Thomson JA. Is serotonin an upper or a downer? The evolution of the serotonergic system and its role in depression and the antidepressant response. Neurosci Biobehav Rev. 2015;51:164–188.    DOI: 10.1016/j.neubiorev.2015.01.018    PMID: 25625874
  78. Jenkins TA, Nguyen JCD, Polglaze KE, Bertrand PP. Influence of tryptophan and serotonin on mood and cognition with a possible role of the gut-brain axis. Nutrients. 2016;8(1):56.    DOI: 10.3390/nu8010056    PMID: 26805875
  79. Gao K, Mu CL, Farzi A, Zhu WY. Tryptophan metabolism: A link between the gut microbiota and brain. Adv Nutr. 2020;11(3):709–723.    DOI: 10.1093/advances/nmz127    PMID: 31825083
  80. Collins SM, Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology. 2009;136(6):2003–2014.    DOI: 10.1053/j.gastro.2009.01.075    PMID: 19457424
  81. Yang J, Zheng P, Li Y, et al. Landscapes of bacterial and metabolic signatures and their interaction in major depressive disorders. Sci Adv. 2020;6(49):eaba8555.    DOI: 10.1126/sciadv.aba8555    PMID: 33268363
  82. Diaz Heijtz R, Wang S, Anuar F, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108(7):3047–3052.    DOI: 10.1073/pnas.1010529108    PMID: 21282636
  83. Lukić I, Getselter D, Koren O, Elliott E. Role of tryptophan in microbiota-induced depressive-like behavior: evidence from tryptophan depletion study. Front Behav Neurosci. 2019;13:123.    DOI: 10.3389/fnbeh.2019.00123    PMID: 31231198
  84. Clarke G, Grenham S, Scully P, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry. 2013;18(6):666–673.    DOI: 10.1038/mp.2012.77    PMID: 22688187
  85. Dinan TG, Stanton C, Cryan JF. Psychobiotics: a novel class of psychotropic. Biol Psychiatry. 2013;74(10):720–726.    DOI: 10.1016/j.biopsych.2013.05.001    PMID: 23759244
  86. Logan AC, Katzman M. Major depressive disorder: Probiotics may be an adjuvant therapy. Med Hypotheses. 2005;64(3):533–538.    DOI: 10.1016/j.mehy.2004.08.019    PMID: 15617861
  87. Zhang ZW, Gao CS, Zhang H, et al. Morinda officinalis oligosaccharides increase serotonin in the brain and ameliorate depression via promoting 5-hydroxytryptophan production in the gut microbiota. Acta Pharm Sin B. 2022;12(8):3298–3312.    DOI: 10.1016/j.apsb.2022.02.032    PMID: 35967282
  88. Munoz-Bellido JL, Munoz-Criado S, Garcìa-Rodrìguez JA. Antimicrobial activity of psychotropic drugs: selective serotonin reuptake inhibitors. Int J Antimicrob Agents. 2000;14(3):177–180.    DOI: 10.1016/s0924-8579(99)00154-5    PMID: 10773485
  89. Wang W, Lin L, Du Y, et al. Assessing the viability of transplanted gut microbiota by sequential tagging with D-amino acid-based metabolic probes. Nat Commun. 2019;10:1317.
  90. Hsu BB, Gibson TE, Yeliseyev V, et al. Dynamic modulation of the gut microbiota and metabolome by bacteriophages in a mouse model. Cell Host Microbe. 2019;25(6):803–814.e5.    DOI: 10.1016/j.chom.2019.05.001    PMID: 31175044
  91. Song WF, Yao WQ, Chen QW, Zheng D, Han ZY, Zhang XZ. In situ bioorthogonal conjugation of delivered bacteria with gut inhabitants for enhancing probiotics colonization. ACS Cent Sci. 2022;8(9):1306–1317.    DOI: 10.1021/acscentsci.2c00533    PMID: 36188344