Next Article in Journal
Tendon 3D Scaffolds Establish a Tailored Microenvironment Instructing Paracrine Mediated Regenerative Amniotic Epithelial Stem Cells Potential
Next Article in Special Issue
Brain-Derived Neurotrophic Factor (BDNF) as an Indicator for Effects of Cognitive Behavioral Therapy (CBT): A Systematic Review
Previous Article in Journal
Radiologist versus Non-Radiologist Detection of Lymph Node Metastasis in Papillary Thyroid Carcinoma by Ultrasound: A Meta-Analysis
Previous Article in Special Issue
New Insights into Cerebral Vessel Disease Landscapes at Single-Cell Resolution: Pathogenetic and Therapeutic Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Involvement of Intestinal Enteroendocrine Cells in Neurological and Psychiatric Disorders

1
Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
2
Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(10), 2577; https://doi.org/10.3390/biomedicines10102577
Submission received: 31 July 2022 / Revised: 21 September 2022 / Accepted: 21 September 2022 / Published: 14 October 2022
(This article belongs to the Special Issue Molecular Research in Neurological and Psychiatric Disease)

Abstract

:
Neurological and psychiatric patients have increased dramatically in number in the past few decades. However, effective treatments for these diseases and disorders are limited due to heterogeneous and unclear pathogenic mechanisms. Therefore, further exploration of the biological aspects of the disease, and the identification of novel targets to develop alternative treatment strategies, is urgently required. Systems-level investigations have indicated the potential involvement of the brain–gut axis and intestinal microbiota in the pathogenesis and regulation of neurological and psychiatric disorders. While intestinal microbiota is crucial for maintaining host physiology, some important sensory and regulatory cells in the host should not be overlooked. Intestinal epithelial enteroendocrine cells (EECs) residing in the epithelium throughout intestine are the key regulators orchestrating the communication along the brain-gut-microbiota axis. On one hand, EECs sense changes in luminal microorganisms via microbial metabolites; on the other hand, they communicate with host body systems via neuroendocrine molecules. Therefore, EECs are believed to play important roles in neurological and psychiatric disorders. This review highlights the involvement of EECs and subtype cells, via secretion of endocrine molecules, in the development and regulation of neurological and psychiatric disorders, including Parkinson’s disease (PD), schizophrenia, visceral pain, neuropathic pain, and depression. Moreover, the current paper summarizes the potential mechanism of EECs in contributing to disease pathogenesis. Examination of these mechanisms may inspire and lead to the development of new aspects of treatment strategies for neurological and psychiatric disorders in the future.

1. Introduction

The number of patients suffering from neurological and psychiatric disorders has increased dramatically in the past few decades. According to the World Health Organization (WHO) epidemiology statistics, the number of Parkinson’s disease (PD) patients has doubled within the last 25 years [1]. Moreover, recent updates from the WHO indicate that there are nearly a billion people suffering from mental disorders, while approximately 280 million people suffer from depression around the world [2,3]. However, effective treatments for neurological and psychiatric disorders are currently limited due to the heterogeneous disease pathogenesis and targets of treatments. For example, it has been shown that visceral pain or depression patients sometimes express resistance to treatment [4,5,6,7]. Therefore, in order to develop novel therapeutic strategies, the exploration of novel aspects of neurological or psychiatric pathogenic mechanisms is urgently required.
Enteroendocrine cells (EECs) are chemosensory cells residing in the intestinal epithelium, and they function as important sensors monitoring changes in the lumen of the gastrointestinal (GI) tract. The EECs orchestrate not only the communication with luminal microorganisms via microbial metabolites, but also the communication with host body systems via neuroendocrine hormones (Figure 1 and Figure 2). For example, epithelial EECs continuously respond to short-chain fatty acids (SCFAs) generated by luminal microorganisms via free fatty acid receptor 2 and 3 (FFAR2/3). Following this transceptor or receptor activation, EECs secrete pre-made peptide hormones to conduct paracrine and endocrine functions [8,9,10,11]. Further, the neuropod structure of EECs allows direct or indirect signal transductions to enteric glia cells and enteric neurons [12,13]. An increasing body of evidence indicates the unique involvement or pathogenic role of the gut–brain axis and gut microbiota in neurological and psychiatric disorders, in which EECs might participate [14,15,16,17,18,19].
The present paper reviews the current understanding and newly published evidence regarding how EECs and their peptide hormones are involved in neurological and psychiatric disorders. Moreover, the current paper focuses on the mechanism of EECs to summarize the potential pathways for the development of new aspects of treatment strategies in the future. The papers in the present review were generally selected under the scope of EECs and endocrine molecules including serotonin, glucagon-like peptide 1 (GLP1), glucagon-like peptide 2 (GLP2), peptide YY (PYY), as well as their involvement in particular neurological psychiatric disorders, such as PD, schizophrenia, visceral pain, neuropathic pain, and depression.

2. Enteroendocrine Cell Functions That Might Be Related to Neurological and Psychiatric Disorders

EECs are located in the epithelium throughout the GI tract. They dynamically produce and store various peptide hormones and bioactive components, depending on the intestinal segments and epithelial homeostasis status. The regulation of EECs’ content profile, as well as their functions in energy metabolism and roles as incretins, has been reviewed elsewhere [20,21]. EECs can be further categorized into multiple subtypes, depending on their endocrine molecules production and secretion. For example, G cells can be identified by the secretion of gastrin; K cells uniquely secrete gastric inhibitory peptides; L cells produce and secrete GLP1, GLP2, PYY, and oxyntomodulin; I cells produce cholecystokinin (CCK); N cells secrete neurotensin; S cells secrete secretin; enterochromaffin cells (ECs) secrete serotonin; and enterochromaffin-like cells secrete histamine [20,21]. Here, we only focus on the subtypes of EECs that are potentially involved in neurological and psychiatric disorders.
L cells mostly secrete GLP1, GLP2, PYY, and oxyntomodulin, although PYY might also be co-expressed with gastrin, which is mostly secreted by G cells [22]. GLP1, GLP2, and PYY, which are secreted by L cells, and serotonin, secreted predominantly by ECs (Figure 2), are discussed in this paper. GLP1 and GLP2 have been shown to correlate with multiple neurological disorders. Serotonin also showed a correlation with depression and visceral pain, although this is still under debate. Besides incretin functions, GLP1 has been shown to exert anti-inflammatory effects in both the GI tract and central nervous system (CNS) [23,24,25]. Moreover, GLP1 possesses neuroprotective effects and triggers neurogenesis [26,27,28,29,30,31]. New evidence suggested that GLP1 and glucagon-like peptide 1 receptor (GLP1R), a receptor of GLP1, have protective effects on hypothalamic inflammation and leptin sensitivity in mice [25,32]. Despite the well-known source and their effects within the CNS, the GLP1 derived from intestinal EECs has also been suggested to play a role in neurological pathology, due to the feature wherein GLP1 is able to pass through the brain–blood barrier [33,34,35]. Similar to GLP1, L-cell-secreted GLP2 also possesses anti-inflammation effects [36,37,38]. In cows, GLP2 administration increased the intestinal villi height, mucosal surface, and proliferating cells, and decreased inflammation [39]. Further, GLP2 has a neuroprotective effect and can trigger neurogenesis in a similar manner as GLP1 [29,37,40,41,42]. Interestingly, the anti-inflammatory effects of other components of EEC content have recently been revealed, including PYY [43,44] and serotonin [45,46], which are likely associated with neuroinflammation. Therefore, accumulating evidence suggests that EECs and ECs could play important pathogenic and regulatory roles in neurological and psychiatric disorders.

3. Enteroendocrine Cells in Parkinson’s Disease

PD is a common movement disorder that was originally characterized as a neurodegenerative disorder due to the loss of dopaminergic neurons and accumulated aggregation of α-synuclein fibrils (called Lewy bodies) (reviewed elsewhere previously [47]). However, studies have shown a new pathogenic aspect of PD, which could be linked to intestinal disorders, as well as to changes in intestinal microbiota and metabolites [15,48,49]. For instance, inflammatory bowel disease (IBD) has increased by 22 to 35% regarding the incidence of PD [50]. In addition, Sampson et al. reported that the GI microbiota was required for motor deficits, microglia activation, and α-synuclein pathology (PD symptoms), in a germ-free mice model overexpressing α-synuclein. Further, their results indicated that the microbial metabolites produced in PD patients enhanced the pathophysiology of PD [15]. Although with a negative correlation, others also found an association among the GI microbiota, the total faecal SCFAs, and PD incidence [48]. Researchers hypothesized that the origin of PD might lie in the enteric nervous system (ENS) [51,52]. Accordingly, α-synuclein was detected in GI mucosa in early PD patients [53,54].
Given the important luminal chemo-sensing and neuroendocrine functions of EECs, these recent results point to a hypothesis that EECs contribute to and regulate the pathogenesis of PD. Interestingly, in human intestinal tissue, the α-synuclein that triggers PD was colocalized with EECs, such as L cells and K cells [55,56]. Although the authors have not confirmed the original secretion location of the α-synuclein, the data in these studies strengthen the possibility of EECs’ involvement in PD progression.
Two potential mechanisms of EECs’ contribution in PD pathogenesis have been proposed. On one hand, the EECs are likely to be a source of α-synuclein, which is generated in response to specific microbial activation. Thereafter, the α-synuclein is transported into the brain via nerves, leading to the accumulation of α-synuclein [57] (Figure 3a). In line with this hypothesis, a very recent research work revealed the potential mechanisms. The authors identified an increased population of microorganism Akkermansia muciniphila in the guts of PD patients. The metabolites of this microorganism initiated α-synuclein aggregation in EECs, via activation of ryanodine receptor (RyR), calcium ion (Ca2+) release, and increased mitochondrial reactive oxygen species (ROS) generation [58] (Figure 3b). Moreover, a newly published paper indicated that another microbial metabolite, sodium butyrate, increased the α-synuclein mRNA expression in EECs through the autophagy-related 5 (Atg5) dependent autophagy pathway [59]. Holmqvist et al. provided evidence that α-synuclein was able to move from the intestine to the brain in rats [60]. Further, the transportation of α-synuclein from EECs to neurons requires GTPase called Ras-related protein Rab-35 (Rab35) and cell-to-cell contact, which is in line with the EECs’ characteristics [61].
On the other hand, the EECs’ secretion could also be suppressed by alterations in luminal SCFA concentrations and profiles. This could be the consequence of changes in specific microbes, which then increase the systemic inflammation, and this eventually enhances the progression of PD [62,63,64]. It was suggested that sodium butyrate increased the pro-inflammatory cytokines and α-synuclein mRNA expression in an EECs cell line and neuroblast cell line treated with EECs conditional medium [59]. Further, the EECs facilitate α-synuclein transport, which could trigger inflammation responses in microglia [65,66]. In contrast, a study in a PD mouse model suggested that the oral administration of butyrate could have protective effects on the neurobehavioral impairment via increased EEC activities, such as increased colonic GLP1 expression and brain GLP1R gene expression [67]. A recent animal study also indicated the neuroprotective effect of GLP1, triggered by chlorogenic acid [31]. These conflicting characteristics of EECs might be due to the variations in EECs’ homeostasis status or the hormone composition of EECs. In other words, the EECs that secrete GLP1 could be beneficial in terms of inflammation reduction, while the EECs that cannot secrete GLP1 but produce α-synuclein could be harmful. However, the detailed mechanism for either hypothesis is still unclear, especially regarding the extent to which EECs contribute to inflammation in PD patients. Future study will be needed to investigate the detailed mechanisms of EECs in PD progression.

4. Enteroendocrine Cells in Schizophrenia

Schizophrenia is a complex neurodevelopmental disorder that could be significantly defined by observations of psychosis signs. In most cases, schizophrenia patients present paranoid delusions and auditory hallucinations [68]. Due to the complexity of neurodevelopment and schizophrenia, the mechanism behind schizophrenia remains unknown [16].
Schizophrenia has been suggested to be associated with the impaired function and structure of synapses [69,70]. Moreover, the microbiota is also associated with synaptogenesis and synapse maturation [16,71]. While the dietary manipulation and intestinal SCFAs enhancement in schizophrenia patients have been discussed elsewhere [72], there have been fewer connections identified between schizophrenia and luminal SCFAs and EECs.
A recent study provided new evidence that linked schizophrenia and epithelial EECs [73]. Uellendahl-Werth et al. reported the susceptibility genes shared between EECs and schizophrenia. Their results indicated that protein phosphatase 3 catalytic subunit alpha (PPP3CA) is the shared susceptibility locus for IBD (Crohn’s disease and ulcerative colitis) and schizophrenia. The genes were expressed in restricted tissues, including neurons in the brain, intestinal epithelial EECs, and Paneth cells in the ileum, colon, and rectum [73]. The authors also provided two possible mechanisms by which PPP3CA in EECs contributes to disease pathology. First, EEC modulation altered the neuronal signal transduction in the striatum. Second, the EECs modulated inflammation responses. Several studies discussed the beneficial effects of the GLP1 (EECs product) agonists on metabolic disorders in schizophrenia patients [74], while others also revealed the potential neuroprotective effect of GLP1 agonists [75,76]. In contrast, several controversial data also suggested that the GLP1 agonists did not improve the cognition or psychosocial function in schizophrenia patients [77]. These conflicts might be due to dosage differences or variations in GLP1 agonists; for example, the differences between Bydureon and Liraglutide [76,77].

5. Enteroendocrine Cells in Visceral Pain and Neuropathic Pain

Visceral pain is a severe form of pain originating from the internal organs. However, it is generally difficult to localize. Among heterogenous pathogenic hypotheses, the neurological dysfunction is significantly linked to visceral pain [78,79,80,81,82]. In fact, visceral pain and neuropathic pain are mostly characterized by hypersensitivity to stimulus, potentially due to hypersensitivity of primary sensory afferent neurons and dysregulation of neurotransmission [79,83].
Visceral pain is often correlated with digestive disorders such as IBD or irritable bowel syndrome (IBS) [84,85,86,87]. Due to the complicity of disease pathology, visceral pain sometimes shows resistance to treatment, especially to opioid drugs. In the worst-case scenario, opioid drugs might even worsen the disease symptoms [4,5]. Therefore, an understanding of the novel biological aspects, such as intestinal microbiota and epithelial EECs, in visceral pain would improve the therapeutic treatments. The relationship between the intestinal microbiota and visceral pain modulation has been discussed recently [88,89]. We highlight the potential connections of EECs to visceral pain via unique proteins and peptide hormones, including serotonin, GLP1, PYY, and Guanylate cyclase 2C (GUCY2C) (Figure 4).
Serotonin is predominantly (90%) secreted by ECs in the intestinal epithelium. It could activate the receptors on serotonergic neurons and trigger the enteric nerve system activity for pain [90]. Numerous studies indicate that serotonin signalling is associated with neuron hypersensitivity to pain. An increased number of ECs has been observed in IBS patients, who usually suffer from pain symptoms [91]. Further, the blockage of serotonin signalling by 5-hydroxytryptamine 3 (5-HT3) receptor antagonists reduced pain in IBS patients [92]. Subcutaneous or tissue injection of serotonin induced the hyperalgesia response and interacted with the endocannabinoid system, which further exacerbated pain [93,94,95,96]. The mechanism of serotonin-induced hypersensitivity has been investigated in the past few decades. Serotonin is known to activate 5-HT3 receptors, thus inhibiting the expression of catecholamine-O-methyltransferase (COMT), which contributes to the downregulation of the pain perception and sensitivity [94,97,98,99,100]. Moreover, a recent study provided new evidence of serotonin-mediated visceral hypersensitivity, which worked via 5-hydroxytryptamine 7 (5-HT7) dependent mucosal neurite outgrowth [101]. Therefore, EECs could play important roles in the pathogenesis and severity of visceral pain. Alterations in the characteristics of EECs (especially ECs) might be an effective target for pain treatment. However, the detailed mechanism is still unknown. Future studies are needed to investigate this aspect.
GLP1 is secreted by not only EECs, but also in the CNS system. GLP1 and its receptor have also been suggested to be associated with neuropathic pain and visceral pain in numerous studies, working mainly through the modulation of inflammation. Recent studies showed that the activation of the GLP1/GLP1R axis improved recognition memory impairment, neuroinflammation, and neurological pain via regulating the AMP-activated protein kinase/nuclear factor kappa B (AMPK/NF-κB) pathway [102,103]. Further, the GLP1R agonist decreases pain hypersensitivity through decreasing pro-inflammatory factors and increasing microglia anti-inflammatory factors, such as interleukin 10 (IL-10), cluster of differentiation 206 (CD206), interleukin 4 (IL-4), and arginase 1 (Arg1) [102,104,105,106,107,108]. New research claimed that the gene regulation in response to GLP1R activation is an effective strategy in new treatments for neuropathic pain, by confirming that the GLP1R pathway is involved in pain hypersensitivity mediated by microglia activation [109]. Considering the inter-organ communication though nerve and endocrine systems, regulation of GLP1 and its receptor in the intestine and CNS system could synergistically improve neural pain sensitivity. Similar to neuropathic pain, the GLP1 agonist is also able to decrease visceral pain. In animal models, a GLP1 analogue or GLP1R agonist improved the visceral pain hypersensitivity in rats [110,111]. New evidence in clinical trials has shown that the subcutaneous injection of a GLP1 analogue, ROSE-010, decreased pain hypersensitivity [112,113]. Although these exogenous peptide treatment data strongly support the connection between GLP1 and visceral pain, less research has been performed to understand the effects of endogenous EECs-derived GLP1 in mediating visceral pain. More investigations are needed to identify the potential EECs targets in developing visceral pain treatments.
PYY is mainly expressed in EECs. However, there are only limited data on the relationship between PYY and visceral pain. Neuropeptide Y is in the same family as PYY but is secreted mainly by neurons. Although the neuropeptide Y inhibits the transmission of pain in the spinal cord and brain stem [114], the relationship between PYY and neural pain is still unclear. In IBS patients, PYY cell density was decreased, which has been proposed as a potential biomarker for the disease [115,116]. In a recent study, Hassan et al. used PYY knockout mice to investigate the relationship between pain, PYY, and the Y2 receptor. Their data suggested that the Y2 receptor antagonist and knockout of PYY increased visceral pain [117]. However, future studies are needed to confirm the effect of PYY on visceral pain and to investigate the details of the mechanism.
Finally, the hypothesis of GUCY2C signalling has been linked to visceral pain pathogenesis. A recent study suggested that GUCY2C-enriched intestinal neuropod cells could modulate visceral pain [118]. Further, GUCY2C agonists decreased pain through increasing the cytoplasm cyclic guanosine monophosphate (cGMP) synthesis from guanosine triphosphate (GTP), as well as through releasing the cGMP from the basolateral membrane of the epithelium to the ENS [119,120,121,122,123]. Therefore, GUCY2C agonists have been proposed as a potential treatment for visceral pain (well reviewed previously) [124]. Not surprisingly, GUCY2C is expressed in whole intestinal epithelial cells, including EECs [125,126]. Given the fact that EECs are close to and actively communicate with ENS neurons, one would strongly expect EECs-derived GUCY2C to modulate visceral pain [13,127]. However, there is not yet a clear understanding of EECs’ involvement in GUCY2C-modulated visceral pain. Future research is required to address this.

6. Enteroendocrine Cells in Depression

Depression is a common disease that affects up to 350 million people around the world [128]. Although depression is a neurological disease, it has been believed to be related to gastrointestinal disorders. Evidence suggests that constipation is a common comorbidity in depression patients [129]. Further, chronic constipation patients have a 33% of incidence of major depression [129,130]. In depression patients, a high number of ECs has been observed, which indicates a potential relationship between depression, ECs, and serotonin production [91].
In contrast to above hypothesis, serotonin deficiency has long been believed to be one of the potential mechanisms of depression, due to the fact that effective medicines have been serotonin-related [131]. Although a recent systematic review questioned the serotonin deficiency hypothesis due to a lack of sufficient supportive data, it might be due to the heterogenous nature of depression among different studies [132]. In fact, several studies have suggested a relationship between depression and the metabolism of tryptophan, a precursor of serotonin [133,134]. Further, selective serotonin reuptake inhibitor (SSRIs) drugs have been used as treatments for depression patients. Therefore, serotonin is still a potential mechanism and pathway for depression. In addition, recent studies suggested that the modulation of intestinal serotonin metabolism through ECs and oral probiotics improved depressive symptoms in an animal model [14,135,136,137]. The potential mechanism has been proposed to be associated with tryptophan hydroxylase and tryptophan metabolism. For instance, mice fed with probiotics showed increased tryptophan hydroxylase 1 mRNA expression in the colon, and the treatment alleviated depressive behaviour in mice with induced chronic stress [135]. However, more research and evidence are needed to investigate the detailed mechanism of ECs involved in depression.
Recent studies suggest that other EEC products, namely GLP1 and GLP2, are also potentially involved in depression. In stressed mice models, GLP2 played a role in regulating monoamine pathways, which in turn exhibited anti-depressive effects [138,139,140]. Similar to GLP2, GLP1 has also shown anti-depressive effects. Both intraperitoneal administration of a GLP1 analogue (Liraglutide) and or oral delivery of an enhancer (metformin) of endogenous GLP1 secretion have shown an anti-depressive effect in animal models [141,142]. The potential mechanisms of GLP1 in depression have been well reviewed in [143]. Briefly, there are four potential mechanisms of GLP1 involved in depression treatment. First, neuroinflammation is modulated by GLP1. Second, the dysregulation of neurotransmitters is modulated by GLP1. Third, the neurogenesis caused by depression is modulated through GLP1. Finally, GLP1′s regulation of depression induces synaptic dysfunction and memory loss [143]. Given the fact that GLP1 might be produced via a multi-organ system, as well as the dynamically regulated EECs activity in response to the brain–gut–microbiota axis, one should not overlook the potential contribution and significance of EECs-derived GLP1 in modulating depression. Further, since the microbiota metabolites trigger EECs to secrete GLP1 and GLP2, this increases the possibility of EECs serving as mediating regulators between the microbiota and enteric nerve system. However, research is needed to provide evidence to support this hypothesis and investigate the details of the involved mechanism.

7. Conclusions

In the present paper we summarized the direct and indirect involvement mechanisms of the EECs in neurological and psychiatric disorders, and discussed the potential treatments. Besides the accumulation of EECs-derived α-synuclein that exacerbates the disease progression in PD, most of the disorders showed significant associations with dysregulation of the neuroendocrine molecules (such as GLP1, GLP2, PYY, serotonin, etc.) produced by EECs and subtype cells. Most of the current treatment strategies focus on administrating exogenous agonists or analogues (GLP1 in schizophrenia, visceral pain hypersensitivity, and depression) and receptor antagonist (serotonin in visceral pain) of these molecules. Alternatively, optimizing the endogenous production of these neuroendocrine molecules could also be considered for developing novel therapeutic strategy. Accumulating evidence connect brain–gut–microbiota axis to the pathogenesis and regulation of neurological and psychiatric disorders. Future investigation should focus on characterizing healthy EECs and reshaping EECs homeostasis in diseases. Intestinal EECs serve as significant source of neuroendocrine molecules. The number and content profiling of EECs depends on intrinsic factors, such as intestinal epithelial stem cells, and the extrinsic microenvironment, such as luminal microbiota. Therefore, the new strategy could be focusing on the differentiation and homeostasis of EECs in the intestinal epithelium, as well as optimizing the EECs functions via regulation of microbiota and nutrition, especially the probiotics and prebiotics. However, the detailed mechanisms are still unclear due to the limitations of techniques and current evidence. The present paper humbly provides a direction for future studies.

Author Contributions

L.Y. (Conceptualization and Writing—Original Draft); Y.L. (Project Administration, Writing—Review and Editing). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Parkinson Disease. Available online: https://www.who.int/news-room/fact-sheets/detail/parkinson-disease (accessed on 16 May 2022).
  2. WHO. Depression. Available online: https://www.who.int/news-room/fact-sheets/detail/depression (accessed on 16 May 2022).
  3. WHO. Nearly One Billion People Have a Mental Disorder: WHO. UN News, 17 June 2022. [Google Scholar]
  4. Hanson, K.A.; Loftus, E.V., Jr.; Harmsen, W.S.; Diehl, N.N.; Zinsmeister, A.R.; Sandborn, W.J. Clinical features and outcome of patients with inflammatory bowel disease who use narcotics: A case-control study. Inflamm. Bowel Dis. 2009, 15, 772–777. [Google Scholar] [CrossRef] [PubMed]
  5. Drossman, D.A.; Morris, C.B.; Edwards, H.; Wrennall, C.E.; Weinland, S.R.; Aderoju, A.O.; Kulkarni-Kelapure, R.R.; Hu, Y.J.; Dalton, C.; Bouma, M.H.; et al. Diagnosis, characterization, and 3-month outcome after detoxification of 39 patients with narcotic bowel syndrome. Am. J. Gastroenterol. 2012, 107, 1426–1440. [Google Scholar] [CrossRef] [PubMed]
  6. Mueller, T.I.; Leon, A.C.; Keller, M.B.; Solomon, D.A.; Endicott, J.; Coryell, W.; Warshaw, M.; Maser, J.D. Recurrence after recovery from major depressive disorder during 15 years of observational follow-up. Am. J. Psychiatry 1999, 156, 1000–1006. [Google Scholar] [CrossRef]
  7. Trivedi, M.H.; Fava, M.; Wisniewski, S.R.; Thase, M.E.; Quitkin, F.; Warden, D.; Ritz, L.; Nierenberg, A.A.; Lebowitz, B.D.; Biggs, M.M.; et al. Medication augmentation after the failure of SSRIs for depression. N. Engl. J. Med. 2006, 354, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
  8. 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]
  9. Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424–429. [Google Scholar] [CrossRef] [Green Version]
  10. Tazoe, H.; Otomo, Y.; Karaki, S.; Kato, I.; Fukami, Y.; Terasaki, M.; Kuwahara, A. Expression of short-chain fatty acid receptor GPR41 in the human colon. Biomed. Res. 2009, 30, 149–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. 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] [PubMed] [Green Version]
  12. Bohorquez, D.V.; Samsa, L.A.; Roholt, A.; Medicetty, S.; Chandra, R.; Liddle, R.A. An enteroendocrine cell-enteric glia connection revealed by 3D electron microscopy. PLoS ONE 2014, 9, e89881. [Google Scholar] [CrossRef] [Green Version]
  13. Bohorquez, D.V.; Shahid, R.A.; Erdmann, A.; Kreger, A.M.; Wang, Y.; Calakos, N.; Wang, F.; Liddle, R.A. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. J. Clin. Investig. 2015, 125, 782–786. [Google Scholar] [CrossRef]
  14. Tian, P.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Bifidobacterium with the role of 5-hydroxytryptophan synthesis regulation alleviates the symptom of depression and related microbiota dysbiosis. J. Nutr. Biochem. 2019, 66, 43–51. [Google Scholar] [CrossRef] [PubMed]
  15. Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kelly, J.R.; Minuto, C.; Cryan, J.F.; Clarke, G.; Dinan, T.G. The role of the gut microbiome in the development of schizophrenia. Schizophr. Res. 2021, 234, 4–23. [Google Scholar] [CrossRef]
  17. Socala, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Wlodarczyk, M.; Zielinska, A.; Poleszak, E.; Fichna, J.; Wlaz, P. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef] [PubMed]
  18. Maiuolo, J.; Gliozzi, M.; Musolino, V.; Carresi, C.; Scarano, F.; Nucera, S.; Scicchitano, M.; Oppedisano, F.; Bosco, F.; Ruga, S.; et al. The Contribution of Gut Microbiota-Brain Axis in the Development of Brain Disorders. Front. Neurosci. 2021, 15, 616883. [Google Scholar] [CrossRef] [PubMed]
  19. Suganya, K.; Koo, B.S. Gut-Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef] [PubMed]
  20. Xie, C.; Jones, K.L.; Rayner, C.K.; Wu, T. Enteroendocrine Hormone Secretion and Metabolic Control: Importance of the Region of the Gut Stimulation. Pharmaceutics 2020, 12, 790. [Google Scholar] [CrossRef]
  21. Martin, A.M.; Sun, E.W.; Keating, D.J. Mechanisms controlling hormone secretion in human gut and its relevance to metabolism. J. Endocrinol. 2019, 244, R1–R15. [Google Scholar] [CrossRef] [Green Version]
  22. Upchurch, B.H.; Fung, B.P.; Rindi, G.; Ronco, A.; Leiter, A.B. Peptide YY expression is an early event in colonic endocrine cell differentiation: Evidence from normal and transgenic mice. Development 1996, 122, 1157–1163. [Google Scholar] [CrossRef] [PubMed]
  23. Barreto-Vianna, A.R.C.; Aguila, M.B.; Mandarim-de-Lacerda, C.A. Beneficial effects of liraglutide (GLP1 analog) in the hippocampal inflammation. Metab. Brain Dis. 2017, 32, 1735–1745. [Google Scholar] [CrossRef]
  24. Solmaz, V.; Cinar, B.P.; Yigitturk, G.; Cavusoglu, T.; Taskiran, D.; Erbas, O. Exenatide reduces TNF-alpha expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur. J. Pharmacol. 2015, 765, 482–487. [Google Scholar] [CrossRef] [PubMed]
  25. Heiss, C.N.; Manneras-Holm, L.; Lee, Y.S.; Serrano-Lobo, J.; Hakansson Gladh, A.; Seeley, R.J.; Drucker, D.J.; Backhed, F.; Olofsson, L.E. The gut microbiota regulates hypothalamic inflammation and leptin sensitivity in Western diet-fed mice via a GLP-1R-dependent mechanism. Cell Rep. 2021, 35, 109163. [Google Scholar] [CrossRef] [PubMed]
  26. Perry, T.; Lahiri, D.K.; Chen, D.; Zhou, J.; Shaw, K.T.; Egan, J.M.; Greig, N.H. A novel neurotrophic property of glucagon-like peptide 1: A promoter of nerve growth factor-mediated differentiation in PC12 cells. J. Pharm. Exp. Ther. 2002, 300, 958–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Perry, T.; Haughey, N.J.; Mattson, M.P.; Egan, J.M.; Greig, N.H. Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4. J. Pharm. Exp. Ther. 2002, 302, 881–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. During, M.J.; Cao, L.; Zuzga, D.S.; Francis, J.S.; Fitzsimons, H.L.; Jiao, X.; Bland, R.J.; Klugmann, M.; Banks, W.A.; Drucker, D.J.; et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat. Med. 2003, 9, 1173–1179. [Google Scholar] [CrossRef] [PubMed]
  29. Voss, U.; Sand, E.; Hellstrom, P.M.; Ekblad, E. Glucagon-like peptides 1 and 2 and vasoactive intestinal peptide are neuroprotective on cultured and mast cell co-cultured rat myenteric neurons. BMC Gastroenterol. 2012, 12, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Zhang, L.; Zhang, L.; Li, Y.; Li, L.; Melchiorsen, J.U.; Rosenkilde, M.; Holscher, C. The Novel Dual GLP-1/GIP Receptor Agonist DA-CH5 Is Superior to Single GLP-1 Receptor Agonists in the MPTP Model of Parkinson’s Disease. J. Parkinsons Dis. 2020, 10, 523–542. [Google Scholar] [CrossRef] [PubMed]
  31. Sharma, N.; Soni, R.; Sharma, M.; Chatterjee, S.; Parihar, N.; Mukarram, M.; Kale, R.; Sayyed, A.A.; Behera, S.K.; Khairnar, A. Chlorogenic Acid: A Polyphenol from Coffee Rendered Neuroprotection Against Rotenone-Induced Parkinson’s Disease by GLP-1 Secretion. Mol. Neurobiol. 2022. [Google Scholar] [CrossRef]
  32. Erdogan, M.A.; Erdogan, A.; Erbas, O. The Anti-Seizure Effect of Liraglutide on Ptz-Induced Convulsions Through its Anti-Oxidant and Anti-Inflammatory Properties. Neurochem. Res. 2022. [Google Scholar] [CrossRef]
  33. Kastin, A.J.; Akerstrom, V.; Pan, W. Interactions of Glucagon-Like Peptide-1 (GLP-1) with the Blood-Brain Barrier. J. Mol. Neurosci. 2002, 18, 7–14. [Google Scholar] [CrossRef]
  34. Kastin, A.J.; Akerstrom, V. Entry of exendin-4 into brain is rapid but may be limited at high doses. Int. J. Obes. Relat. Metab. Disord. 2003, 27, 313–318. [Google Scholar] [CrossRef] [Green Version]
  35. Hunter, K.; Holscher, C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012, 13, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cani, P.D.; Possemiers, S.; Van de Wiele, T.; Guiot, Y.; Everard, A.; Rottier, O.; Geurts, L.; Naslain, D.; Neyrinck, A.; Lambert, D.M.; et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009, 58, 1091–1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Nuzzo, D.; Baldassano, S.; Amato, A.; Picone, P.; Galizzi, G.; Caldara, G.F.; Di Carlo, M.; Mule, F. Glucagon-like peptide-2 reduces the obesity-associated inflammation in the brain. Neurobiol. Dis. 2019, 121, 296–304. [Google Scholar] [CrossRef]
  38. Xie, S.; Liu, B.; Fu, S.; Wang, W.; Yin, Y.; Li, N.; Chen, W.; Liu, J.; Liu, D. GLP-2 suppresses LPS-induced inflammation in macrophages by inhibiting ERK phosphorylation and NF-kappaB activation. Cell Physiol. Biochem. 2014, 34, 590–602. [Google Scholar] [CrossRef] [PubMed]
  39. Kvidera, S.K.; Horst, E.A.; Sanz Fernandez, M.V.; Abuajamieh, M.; Ganesan, S.; Gorden, P.J.; Green, H.B.; Schoenberg, K.M.; Trout, W.E.; Keating, A.F.; et al. Characterizing effects of feed restriction and glucagon-like peptide 2 administration on biomarkers of inflammation and intestinal morphology. J. Dairy Sci. 2017, 100, 9402–9417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Li, N.; Liu, B.W.; Ren, W.Z.; Liu, J.X.; Li, S.N.; Fu, S.P.; Zeng, Y.L.; Xu, S.Y.; Yan, X.; Gao, Y.J.; et al. GLP-2 Attenuates LPS-Induced Inflammation in BV-2 Cells by Inhibiting ERK1/2, JNK1/2 and NF-κB Signaling Pathways. Int. J. Mol. Sci. 2016, 17, 190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Zhang, Z.; Hao, L.; Shi, M.; Yu, Z.; Shao, S.; Yuan, Y.; Zhang, Z.; Holscher, C. Neuroprotective Effects of a GLP-2 Analogue in the MPTP Parkinson’s Disease Mouse Model. J. Parkinsons Dis. 2021, 11, 529–543. [Google Scholar] [CrossRef]
  42. Su, Y.; Zhang, Z.; Li, H.; Ma, J.; Sun, L.; Shao, S.; Zhang, Z.; Holscher, C. A GLP-2 Analogue Protects SH-SY5Y and Neuro-2a Cells Against Mitochondrial Damage, Autophagy Impairments and Apoptosis in a Parkinson Model. Drug Res. 2021, 71, 43–50. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Z.; Kuang, X.; Chen, T.; Shen, T.; Wu, J. Peptide YY 3-36 attenuates trinitrobenzene sulfonic acid-induced colitis in mice by modulating Th1/Th2 differentiation. Bioengineered 2022, 13, 10144–10158. [Google Scholar] [CrossRef] [PubMed]
  44. Guzzardi, M.A.; La Rosa, F.; Campani, D.; Cacciato Insilla, A.; Nannipieri, M.; Brunetto, M.R.; Bonino, F.; Iozzo, P. Evidence of a Gastro-Duodenal Effect on Adipose Tissue and Brain Metabolism, Potentially Mediated by Gut-Liver Inflammation: A Study with Positron Emission Tomography and Oral (18)FDG in Mice. Int. J. Mol. Sci. 2022, 23, 2695. [Google Scholar] [CrossRef]
  45. Mota, C.M.D.; Rodrigues-Santos, C.; Fernandez, R.A.R.; Carolino, R.O.G.; Antunes-Rodrigues, J.; Anselmo-Franci, J.A.; Branco, L.G.S. Central serotonin attenuates LPS-induced systemic inflammation. Brain Behav. Immun. 2017, 66, 372–381. [Google Scholar] [CrossRef] [PubMed]
  46. Arnold, W.R.; Carnevale, L.N.; Xie, Z.; Baylon, J.L.; Tajkhorshid, E.; Hu, H.; Das, A. Anti-inflammatory dopamine- and serotonin-based endocannabinoid epoxides reciprocally regulate cannabinoid receptors and the TRPV1 channel. Nat. Commun. 2021, 12, 926. [Google Scholar] [CrossRef] [PubMed]
  47. Davie, C.A. A review of Parkinson’s disease. Br. Med. Bull 2008, 86, 109–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Unger, M.M.; Spiegel, J.; Dillmann, K.U.; Grundmann, D.; Philippeit, H.; Burmann, J.; Fassbender, K.; Schwiertz, A.; Schafer, K.H. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
  49. Hirayama, M.; Ohno, K. Parkinson’s Disease and Gut Microbiota. Ann. Nutr. Metab. 2021, 77 (Suppl. 2), 28–35. [Google Scholar] [CrossRef]
  50. Lin, J.C.; Lin, C.S.; Hsu, C.W.; Lin, C.L.; Kao, C.H. Association Between Parkinson’s Disease and Inflammatory Bowel Disease: A Nationwide Taiwanese Retrospective Cohort Study. Inflamm. Bowel Dis. 2016, 22, 1049–1055. [Google Scholar] [CrossRef] [Green Version]
  51. Braak, H.; Rub, U.; Gai, W.P.; Del Tredici, K. Idiopathic Parkinson’s disease: Possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm. 2003, 110, 517–536. [Google Scholar] [CrossRef]
  52. Del Tredici, K.; Rub, U.; De Vos, R.A.; Bohl, J.R.; Braak, H. Where does parkinson disease pathology begin in the brain? J. Neuropathol. Exp. Neurol. 2002, 61, 413–426. [Google Scholar] [CrossRef]
  53. Shannon, K.M.; Keshavarzian, A.; Mutlu, E.; Dodiya, H.B.; Daian, D.; Jaglin, J.A.; Kordower, J.H. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov. Disord. 2012, 27, 709–715. [Google Scholar] [CrossRef]
  54. Sanchez-Ferro, A.; Rabano, A.; Catalan, M.J.; Rodriguez-Valcarcel, F.C.; Fernandez Diez, S.; Herreros-Rodriguez, J.; Garcia-Cobos, E.; Alvarez-Santullano, M.M.; Lopez-Manzanares, L.; Mosqueira, A.J.; et al. In vivo gastric detection of alpha-synuclein inclusions in Parkinson’s disease. Mov. Disord. 2015, 30, 517–524. [Google Scholar] [CrossRef] [PubMed]
  55. Chandra, R.; Hiniker, A.; Kuo, Y.M.; Nussbaum, R.L.; Liddle, R.A. alpha-Synuclein in gut endocrine cells and its implications for Parkinson’s disease. JCI Insight 2017, 2, e92295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Casini, A.; Mancinelli, R.; Mammola, C.L.; Pannarale, L.; Chirletti, P.; Onori, P.; Vaccaro, R. Distribution of alpha-synuclein in normal human jejunum and its relations with the chemosensory and neuroendocrine system. Eur. J. Histochem. EJH 2021, 65, 3310. [Google Scholar] [CrossRef]
  57. Liddle, R.A. Parkinson’s disease from the gut. Brain Res. 2018, 1693, 201–206. [Google Scholar] [CrossRef] [PubMed]
  58. Amorim Neto, D.P.; Bosque, B.P.; Pereira de Godoy, J.V.; Rodrigues, P.V.; Meneses, D.D.; Tostes, K.; Costa Tonoli, C.C.; Faustino de Carvalho, H.; Gonzalez-Billault, C.; de Castro Fonseca, M. Akkermansia muciniphila induces mitochondrial calcium overload and alpha -synuclein aggregation in an enteroendocrine cell line. iScience 2022, 25, 103908. [Google Scholar] [CrossRef]
  59. Qiao, C.M.; Sun, M.F.; Jia, X.B.; Shi, Y.; Zhang, B.P.; Zhou, Z.L.; Zhao, L.P.; Cui, C.; Shen, Y.Q. Sodium butyrate causes alpha-synuclein degradation by an Atg5-dependent and PI3K/Akt/mTOR-related autophagy pathway. Exp. Cell Res. 2020, 387, 111772. [Google Scholar] [CrossRef]
  60. Holmqvist, S.; Chutna, O.; Bousset, L.; Aldrin-Kirk, P.; Li, W.; Bjorklund, T.; Wang, Z.Y.; Roybon, L.; Melki, R.; Li, J.Y. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 2014, 128, 805–820. [Google Scholar] [CrossRef] [Green Version]
  61. Rodrigues, P.V.; de Godoy, J.V.P.; Bosque, B.P.; Amorim Neto, D.P.; Tostes, K.; Palameta, S.; Garcia-Rosa, S.; Tonoli, C.C.C.; de Carvalho, H.F.; de Castro Fonseca, M. Transcellular propagation of fibrillar alpha-synuclein from enteroendocrine to neuronal cells requires cell-to-cell contact and is Rab35-dependent. Sci. Rep. 2022, 12, 4168. [Google Scholar] [CrossRef]
  62. Tansey, M.G.; Wallings, R.L.; Houser, M.C.; Herrick, M.K.; Keating, C.E.; Joers, V. Inflammation and immune dysfunction in Parkinson disease. Nat. Rev. Immunol. 2022. [Google Scholar] [CrossRef]
  63. Boyle, J.G.; Livingstone, R.; Petrie, J.R. Cardiovascular benefits of GLP-1 agonists in type 2 diabetes: A comparative review. Clin. Sci. 2018, 132, 1699–1709. [Google Scholar] [CrossRef]
  64. Drobny, A.; Ngo, P.A.; Neurath, M.F.; Zunke, F.; Lopez-Posadas, R. Molecular Communication Between Neuronal Networks and Intestinal Epithelial Cells in Gut Inflammation and Parkinson’s Disease. Front. Med. 2021, 8, 655123. [Google Scholar] [CrossRef] [PubMed]
  65. Alvarez-Erviti, L.; Couch, Y.; Richardson, J.; Cooper, J.M.; Wood, M.J. Alpha-synuclein release by neurons activates the inflammatory response in a microglial cell line. Neurosci. Res. 2011, 69, 337–342. [Google Scholar] [CrossRef]
  66. Beraud, D.; Hathaway, H.A.; Trecki, J.; Chasovskikh, S.; Johnson, D.A.; Johnson, J.A.; Federoff, H.J.; Shimoji, M.; Mhyre, T.R.; Maguire-Zeiss, K.A. Microglial activation and antioxidant responses induced by the Parkinson’s disease protein alpha-synuclein. J. Neuroimmune Pharm. 2013, 8, 94–117. [Google Scholar] [CrossRef] [Green Version]
  67. Liu, J.; Wang, F.; Liu, S.; Du, J.; Hu, X.; Xiong, J.; Fang, R.; Chen, W.; Sun, J. Sodium butyrate exerts protective effect against Parkinson’s disease in mice via stimulation of glucagon like peptide-1. J. Neurol. Sci. 2017, 381, 176–181. [Google Scholar] [CrossRef] [PubMed]
  68. Insel, T.R. Rethinking schizophrenia. Nature 2010, 468, 187–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Habela, C.W.; Song, H.; Ming, G.L. Modeling synaptogenesis in schizophrenia and autism using human iPSC derived neurons. Mol. Cell Neurosci. 2016, 73, 52–62. [Google Scholar] [CrossRef]
  70. Faludi, G.; Mirnics, K. Synaptic changes in the brain of subjects with schizophrenia. Int. J. Dev. Neurosci. 2011, 29, 305–309. [Google Scholar] [CrossRef] [Green Version]
  71. Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, 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] [Green Version]
  72. Joseph, J.; Depp, C.; Shih, P.B.; Cadenhead, K.S.; Schmid-Schonbein, G. Modified Mediterranean Diet for Enrichment of Short Chain Fatty Acids: Potential Adjunctive Therapeutic to Target Immune and Metabolic Dysfunction in Schizophrenia? Front Neurosci. 2017, 11, 155. [Google Scholar] [CrossRef] [Green Version]
  73. Uellendahl-Werth, F.; Maj, C.; Borisov, O.; Juzenas, S.; Wacker, E.M.; Jorgensen, I.F.; Steiert, T.A.; Bej, S.; Krawitz, P.; Hoffmann, P.; et al. Cross-tissue transcriptome-wide association studies identify susceptibility genes shared between schizophrenia and inflammatory bowel disease. Commun. Biol. 2022, 5, 80. [Google Scholar] [CrossRef]
  74. Kouidrat, Y.; Amad, A. GLP-1 agonists for metabolic disorders in schizophrenia. Schizophr. Res. 2019, 204, 448–449. [Google Scholar] [CrossRef]
  75. McIntyre, R.S.; Powell, A.M.; Kaidanovich-Beilin, O.; Soczynska, J.K.; Alsuwaidan, M.; Woldeyohannes, H.O.; Kim, A.S.; Gallaugher, L.A. The neuroprotective effects of GLP-1: Possible treatments for cognitive deficits in individuals with mood disorders. Behav. Brain Res. 2013, 237, 164–171. [Google Scholar] [CrossRef]
  76. Camkurt, M.A.; Lavagnino, L.; Zhang, X.Y.; Teixeira, A.L. Liraglutide for psychiatric disorders: Clinical evidence and challenges. Horm. Mol. Biol. Clin. Investig. 2018, 36, 1–6. [Google Scholar] [CrossRef]
  77. Ishoy, P.L.; Fagerlund, B.; Broberg, B.V.; Bak, N.; Knop, F.K.; Glenthoj, B.Y.; Ebdrup, B.H. No cognitive-enhancing effect of GLP-1 receptor agonism in antipsychotic-treated, obese patients with schizophrenia. Acta Psychiatr. Scand. 2017, 136, 52–62. [Google Scholar] [CrossRef]
  78. Cohen, S.P.; Mao, J. Neuropathic pain: Mechanisms and their clinical implications. BMJ 2014, 348, f7656. [Google Scholar] [CrossRef] [Green Version]
  79. Sengupta, J.N. Visceral pain: The neurophysiological mechanism. In Handbook of Experimental Pharmacology; Springer: Cham, Switzerland, 2009; pp. 31–74. [Google Scholar] [CrossRef] [Green Version]
  80. Katz, B.L.; Van Houten, T.; Sabouri, A.S. Neuroanatomy and Mechanisms of Visceral Pain. In Interventional Management of Chronic Visceral Pain Syndromes; Elsevier: Amsterdam, The Netherlands, 2021; pp. 5–15. [Google Scholar] [CrossRef]
  81. Ohlmann, H.; Koenen, L.R.; Labrenz, F.; Engler, H.; Theysohn, N.; Langhorst, J.; Elsenbruch, S. Altered Brain Structure in Chronic Visceral Pain: Specific Differences in Gray Matter Volume and Associations With Visceral Symptoms and Chronic Stress. Front. Neurol. 2021, 12, 733035. [Google Scholar] [CrossRef] [PubMed]
  82. Cortelli, P.; Montagna, P. Migraine as a visceral pain. Neurol. Sci. 2009, 30 (Suppl. 1.), S19–S22. [Google Scholar] [CrossRef]
  83. Moshiree, B.; Zhou, Q.; Price, D.D.; Verne, G.N. Central sensitisation in visceral pain disorders. Gut 2006, 55, 905–908. [Google Scholar] [CrossRef] [PubMed]
  84. Bielefeldt, K.; Davis, B.; Binion, D.G. Pain and inflammatory bowel disease. Inflamm. Bowel Dis. 2009, 15, 778–788. [Google Scholar] [CrossRef]
  85. Balmus, I.M.; Ciobica, A.; Cojocariu, R.; Luca, A.C.; Gorgan, L. Irritable Bowel Syndrome and Neurological Deficiencies: Is There A Relationship? The Possible Relevance of the Oxidative Stress Status. Medicina 2020, 56, 175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Moloney, R.D.; O’Mahony, S.M.; Dinan, T.G.; Cryan, J.F. Stress-induced visceral pain: Toward animal models of irritable-bowel syndrome and associated comorbidities. Front. Psychiatry 2015, 6, 15. [Google Scholar] [CrossRef] [Green Version]
  87. Greenwood-Van Meerveld, B.; Johnson, A.C. Stress-Induced Chronic Visceral Pain of Gastrointestinal Origin. Front. Syst. Neurosci. 2017, 11, 86. [Google Scholar] [CrossRef] [Green Version]
  88. Lomax, A.E.; Pradhananga, S.; Sessenwein, J.L.; O’Malley, D. Bacterial modulation of visceral sensation: Mediators and mechanisms. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G363–G372. [Google Scholar] [CrossRef] [PubMed]
  89. Morreale, C.; Bresesti, I.; Bosi, A.; Baj, A.; Giaroni, C.; Agosti, M.; Salvatore, S. Microbiota and Pain: Save Your Gut Feeling. Cells 2022, 11, 971. [Google Scholar] [CrossRef] [PubMed]
  90. 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]
  91. Dunlop, S.P.; Jenkins, D.; Neal, K.R.; Spiller, R.C. Relative importance of enterochromaffin cell hyperplasia, anxiety, and depression in postinfectious IBS. Gastroenterology 2003, 125, 1651–1659. [Google Scholar] [CrossRef]
  92. Camilleri, M.; Boeckxstaens, G. Dietary and pharmacological treatment of abdominal pain in IBS. Gut 2017, 66, 966–974. [Google Scholar] [CrossRef]
  93. Taiwo, Y.O.; Levine, J.D. Serotonin is a directly-acting hyperalgesic agent in the rat. Neuroscience 1992, 48, 485–490. [Google Scholar] [CrossRef]
  94. Sufka, K.J.; Schomburg, F.M.; Giordano, J. Receptor mediation of 5-HT-induced inflammation and nociception in rats. Pharmacol. Biochem. Behav. 1992, 41, 53–56. [Google Scholar] [CrossRef]
  95. Zeitz, K.P.; Guy, N.; Malmberg, A.B.; Dirajlal, S.; Martin, W.J.; Sun, L.; Bonhaus, D.W.; Stucky, C.L.; Julius, D.; Basbaum, A.I. The 5-HT3Subtype of Serotonin Receptor Contributes to Nociceptive Processing via a Novel Subset of Myelinated and Unmyelinated Nociceptors. J. Neurosci. 2002, 22, 1010–1019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Salaga, M.; Binienda, A.; Piscitelli, F.; Mokrowiecka, A.; Cygankiewicz, A.I.; Verde, R.; Malecka-Panas, E.; Kordek, R.; Krajewska, W.M.; Di Marzo, V.; et al. Systemic administration of serotonin exacerbates abdominal pain and colitis via interaction with the endocannabinoid system. Biochem. Pharm. 2019, 161, 37–51. [Google Scholar] [CrossRef]
  97. Diatchenko, L.; Slade, G.D.; Nackley, A.G.; Bhalang, K.; Sigurdsson, A.; Belfer, I.; Goldman, D.; Xu, K.; Shabalina, S.A.; Shagin, D.; et al. Genetic basis for individual variations in pain perception and the development of a chronic pain condition. Hum. Mol. Genet. 2005, 14, 135–143. [Google Scholar] [CrossRef] [Green Version]
  98. Nackley, A.G.; Shabalina, S.A.; Tchivileva, I.E.; Satterfield, K.; Korchynskyi, O.; Makarov, S.S.; Maixner, W.; Diatchenko, L. Human catechol-O-methyltransferase haplotypes modulate protein expression by altering mRNA secondary structure. Science 2006, 314, 1930–1933. [Google Scholar] [CrossRef] [Green Version]
  99. Nackley, A.G.; Tan, K.S.; Fecho, K.; Flood, P.; Diatchenko, L.; Maixner, W. Catechol-O-methyltransferase inhibition increases pain sensitivity through activation of both beta2- and beta3-adrenergic receptors. Pain 2007, 128, 199–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Tsao, D.; Wieskopf, J.S.; Rashid, N.; Sorge, R.E.; Redler, R.L.; Segall, S.K.; Mogil, J.S.; Maixner, W.; Dokholyan, N.V.; Diatchenko, L. Serotonin-induced hypersensitivity via inhibition of catechol O-methyltransferase activity. Mol. Pain 2012, 8, 25. [Google Scholar] [CrossRef] [Green Version]
  101. Chang, W.Y.; Yang, Y.T.; She, M.P.; Tu, C.H.; Lee, T.C.; Wu, M.S.; Sun, C.H.; Hsin, L.W.; Yu, L.C. 5-HT7 receptor-dependent intestinal neurite outgrowth contributes to visceral hypersensitivity in irritable bowel syndrome. Lab. Investig. 2022, 102, 1023–1037. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, L.Q.; Zhang, W.; Li, T.; Yang, T.; Yuan, X.; Zhou, Y.; Zou, Q.; Yang, H.; Gao, F.; Tian, Y.; et al. GLP-1R activation ameliorated novel-object recognition memory dysfunction via regulating hippocampal AMPK/NF-κB pathway in neuropathic pain mice. Neurobiol. Learn Mem. 2021, 182, 107463. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, L.; Zhang, W.; Tian, X. The pleiotropic of GLP-1/GLP-1R axis in central nervous system diseases. Int. J. Neurosci. 2021, 1–38. [Google Scholar] [CrossRef]
  104. Wu, H.Y.; Tang, X.Q.; Mao, X.F.; Wang, Y.X. Autocrine Interleukin-10 Mediates Glucagon-Like Peptide-1 Receptor-Induced Spinal Microglial beta-Endorphin Expression. J. Neurosci. 2017, 37, 11701–11714. [Google Scholar] [CrossRef] [Green Version]
  105. Gong, N.; Xiao, Q.; Zhu, B.; Zhang, C.Y.; Wang, Y.C.; Fan, H.; Ma, A.N.; Wang, Y.X. Activation of spinal glucagon-like peptide-1 receptors specifically suppresses pain hypersensitivity. J. Neurosci. 2014, 34, 5322–5334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wang, Z.Y.; Han, Q.Q.; Deng, M.Y.; Zhao, M.J.; Apryani, E.; Shoaib, R.M.; Wei, D.Q.; Wang, Y.X. Lemairamin, isolated from the Zanthoxylum plants, alleviates pain hypersensitivity via spinal alpha7 nicotinic acetylcholine receptors. Biochem. Biophys. Res. Commun. 2020, 525, 1087–1094. [Google Scholar] [CrossRef] [PubMed]
  107. Ma, L.; Peng, S.; Wei, J.; Zhao, M.; Ahmad, K.A.; Chen, J.; Wang, Y.X. Spinal microglial beta-endorphin signaling mediates IL-10 and exenatide-induced inhibition of synaptic plasticity in neuropathic pain. CNS Neurosci. 2021, 27, 1157–1172. [Google Scholar] [CrossRef] [PubMed]
  108. Tang, X.; Wu, H.; Mao, X.; Li, X.; Wang, Y. The GLP-1 receptor herbal agonist morroniside attenuates neuropathic pain via spinal microglial expression of IL-10 and beta-endorphin. Biochem. Biophys. Res. Commun. 2020, 530, 494–499. [Google Scholar] [CrossRef] [PubMed]
  109. Ma, L.; Ju, P.; Wang, W.; Wei, J.; Wang, W.; Zhao, M.; Ahmad, K.A.; Wang, Y.; Chen, J. Microglial Activation of GLP-1R Signaling in Neuropathic Pain Promotes Gene Expression Adaption Involved in Inflammatory Responses. Neural Plast. 2021, 2021, 9923537. [Google Scholar] [CrossRef] [PubMed]
  110. Nozu, T.; Miyagishi, S.; Kumei, S.; Nozu, R.; Takakusaki, K.; Okumura, T. Glucagon-like peptide-1 analog, liraglutide, improves visceral sensation and gut permeability in rats. J. Gastroenterol. Hepatol. 2018, 33, 232–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. O’Brien, R.; O’Malley, D. The Glucagon-like peptide-1 receptor agonist, exendin-4, ameliorated gastrointestinal dysfunction in the Wistar Kyoto rat model of Irritable Bowel Syndrome. Neurogastroenterol. Motil. 2020, 32, e13738. [Google Scholar] [CrossRef] [PubMed]
  112. Hellstrom, P.M.; Hein, J.; Bytzer, P.; Bjornsson, E.; Kristensen, J.; Schambye, H. Clinical trial: The glucagon-like peptide-1 analogue ROSE-010 for management of acute pain in patients with irritable bowel syndrome: A randomized, placebo-controlled, double-blind study. Aliment Pharm. Ther. 2009, 29, 198–206. [Google Scholar] [CrossRef] [PubMed]
  113. Touny, A.A.; Kenny, E.; Mansson, M.; Webb, D.L.; Hellstrom, P.M. Pain relief and pain intensity response to GLP-1 receptor agonist ROSE-010 in irritable bowel syndrome; clinical study cross-analysis with respect to patient characteristics. Scand. J. Gastroenterol. 2022, 57, 783–791. [Google Scholar] [CrossRef] [PubMed]
  114. Holzer, P.; Reichmann, F.; Farzi, A. Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis. Neuropeptides 2012, 46, 261–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. El-Salhy, M.; Gundersen, D.; Hatlebakk, J.G.; Gilja, O.H.; Hausken, T. Abnormal rectal endocrine cells in patients with irritable bowel syndrome. Regul. Pept. 2014, 188, 60–65. [Google Scholar] [CrossRef] [PubMed]
  116. El-Salhy, M.; Hatlebakk, J.G.; Gilja, O.H.; Hausken, T. Densities of rectal peptide YY and somatostatin cells as biomarkers for the diagnosis of irritable bowel syndrome. Peptides 2015, 67, 12–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Hassan, A.M.; Jain, P.; Mayerhofer, R.; Frohlich, E.E.; Farzi, A.; Reichmann, F.; Herzog, H.; Holzer, P. Visceral hyperalgesia caused by peptide YY deletion and Y2 receptor antagonism. Sci. Rep. 2017, 7, 40968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Barton, J.R.; Alexander, T.; Covarrubias, M.; Waldman, S.A. GUCY2C-Enriched Intestinal Neuropod Cells Modulate Visceral Pain. FASEB J. 2022, 36. [Google Scholar] [CrossRef]
  119. Bryant, A.P.; Busby, R.W.; Bartolini, W.P.; Cordero, E.A.; Hannig, G.; Kessler, M.M.; Pierce, C.M.; Solinga, R.M.; Tobin, J.V.; Mahajan-Miklos, S.; et al. Linaclotide is a potent and selective guanylate cyclase C agonist that elicits pharmacological effects locally in the gastrointestinal tract. Life Sci. 2010, 86, 760–765. [Google Scholar] [CrossRef] [PubMed]
  120. Busby, R.W.; Bryant, A.P.; Bartolini, W.P.; Cordero, E.A.; Hannig, G.; Kessler, M.M.; Mahajan-Miklos, S.; Pierce, C.M.; Solinga, R.M.; Sun, L.J.; et al. Linaclotide, through activation of guanylate cyclase C, acts locally in the gastrointestinal tract to elicit enhanced intestinal secretion and transit. Eur. J. Pharmacol. 2010, 649, 328–335. [Google Scholar] [CrossRef]
  121. Kuhn, M. Molecular Physiology of Membrane Guanylyl Cyclase Receptors. Physiol. Rev. 2016, 96, 751–804. [Google Scholar] [CrossRef] [Green Version]
  122. Silos-Santiago, I.; Hannig, G.; Eutamene, H.; Ustinova, E.E.; Bernier, S.G.; Ge, P.; Graul, C.; Jacobson, S.; Jin, H.; Liong, E.; et al. Gastrointestinal pain: Unraveling a novel endogenous pathway through uroguanylin/guanylate cyclase-C/cGMP activation. Pain 2013, 154, 1820–1830. [Google Scholar] [CrossRef] [Green Version]
  123. Boulete, I.M.; Thadi, A.; Beaufrand, C.; Patwa, V.; Joshi, A.; Foss, J.A.; Eddy, E.P.; Eutamene, H.; Palejwala, V.A.; Theodorou, V.; et al. Oral treatment with plecanatide or dolcanatide attenuates visceral hypersensitivity via activation of guanylate cyclase-C in rat models. World J. Gastroenterol. 2018, 24, 1888–1900. [Google Scholar] [CrossRef]
  124. Brierley, S.M.; Grundy, L.; Castro, J.; Harrington, A.M.; Hannig, G.; Camilleri, M. Guanylate cyclase-C agonists as peripherally acting treatments of chronic visceral pain. Trends Pharm. Sci. 2022, 43, 110–122. [Google Scholar] [CrossRef]
  125. Gallery, M.; Zhang, J.; Bradley, D.P.; Brauer, P.; Cvet, D.; Estevam, J.; Danaee, H.; Greenfield, E.; Li, P.; Manfredi, M.; et al. A monomethyl auristatin E-conjugated antibody to guanylyl cyclase C is cytotoxic to target-expressing cells in vitro and in vivo. PLoS ONE 2018, 13, e0191046. [Google Scholar] [CrossRef] [Green Version]
  126. Danaee, H.; Kalebic, T.; Wyant, T.; Fassan, M.; Mescoli, C.; Gao, F.; Trepicchio, W.L.; Rugge, M. Consistent expression of guanylyl cyclase-C in primary and metastatic gastrointestinal cancers. PLoS ONE 2017, 12, e0189953. [Google Scholar] [CrossRef] [Green Version]
  127. Liddle, R.A. Interactions of Gut Endocrine Cells with Epithelium and Neurons. Compr. Physiol. 2018, 8, 1019–1030. [Google Scholar] [CrossRef] [PubMed]
  128. Smith, K. Mental health: A world of depression. Nature 2014, 515, 181. [Google Scholar] [CrossRef] [Green Version]
  129. Hosseinzadeh, S.T.; Poorsaadati, S.; Radkani, B.; Forootan, M. Psychological disorders in patients with chronic constipation. Gastroenterol. Hepatol. Bed Bench 2011, 4, 159–163. [Google Scholar] [PubMed]
  130. Dipnall, J.F.; Pasco, J.A.; Berk, M.; Williams, L.J.; Dodd, S.; Jacka, F.N.; Meyer, D. Into the Bowels of Depression: Unravelling Medical Symptoms Associated with Depression by Applying Machine-Learning Techniques to a Community Based Population Sample. PLoS ONE 2016, 11, e0167055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Coppen, A. The biochemistry of affective disorders. Br. J. Psychiatry 1967, 113, 1237–1264. [Google Scholar] [CrossRef]
  132. Moncrieff, J.; Cooper, R.E.; Stockmann, T.; Amendola, S.; Hengartner, M.P.; Horowitz, M.A. The serotonin theory of depression: A systematic umbrella review of the evidence. Mol. Psychiatry 2022, 2022, 1–14. [Google Scholar] [CrossRef]
  133. Smith, K.A.; Fairburn, C.G.; Cowen, P.J. Relapse of depression after rapid depletion of tryptophan. Lancet 1997, 349, 915–919. [Google Scholar] [CrossRef]
  134. Ruhe, H.G.; Mason, N.S.; Schene, A.H. Mood is indirectly related to serotonin, norepinephrine and dopamine levels in humans: A meta-analysis of monoamine depletion studies. Mol Psychiatry 2007, 12, 331–359. [Google Scholar] [CrossRef] [Green Version]
  135. Gao, K.; Farzi, A.; Ke, X.; Yu, Y.; Chen, C.; Chen, S.; Yu, T.; Wang, H.; Li, Y. Oral administration of Lactococcus lactis WHH2078 alleviates depressive and anxiety symptoms in mice with induced chronic stress. Food Funct. 2022, 13, 957–969. [Google Scholar] [CrossRef]
  136. Tian, P.; Zhu, H.; Zou, R.; Kong, Q.; Xu, M.; Zhao, J.; Zhang, H.; Chen, W.; Wang, G. An in vitro screening method for probiotics with antidepressant-like effect using the enterochromaffin cell model. Food Funct. 2021, 12, 646–655. [Google Scholar] [CrossRef] [PubMed]
  137. Israelyan, N.; Del Colle, A.; Li, Z.; Park, Y.; Xing, A.; Jacobsen, J.P.R.; Luna, R.A.; Jensen, D.D.; Madra, M.; Saurman, V.; et al. Effects of Serotonin and Slow-Release 5-Hydroxytryptophan on Gastrointestinal Motility in a Mouse Model of Depression. Gastroenterology 2019, 157, 507–521.e504. [Google Scholar] [CrossRef]
  138. Iwai, T.; Hayashi, Y.; Narita, S.; Kasuya, Y.; Jin, K.; Tsugane, M.; Oka, J. Antidepressant-like effects of glucagon-like peptide-2 in mice occur via monoamine pathways. Behav. Brain Res. 2009, 204, 235–240. [Google Scholar] [CrossRef]
  139. Iwai, T.; Ohnuki, T.; Sasaki-Hamada, S.; Saitoh, A.; Sugiyama, A.; Oka, J. Glucagon-like peptide-2 but not imipramine exhibits antidepressant-like effects in ACTH-treated mice. Behav. Brain Res. 2013, 243, 153–157. [Google Scholar] [CrossRef] [PubMed]
  140. Sasaki-Hamada, S.; Nakamura, R.; Nakao, Y.; Akimoto, T.; Sanai, E.; Nagai, M.; Horiguchi, M.; Yamashita, C.; Oka, J.I. Antidepressant-like effects exerted by the intranasal administration of a glucagon-like peptide-2 derivative containing cell-penetrating peptides and a penetration-accelerating sequence in mice. Peptides 2017, 87, 64–70. [Google Scholar] [CrossRef] [PubMed]
  141. Weina, H.; Yuhu, N.; Christian, H.; Birong, L.; Feiyu, S.; Le, W. Liraglutide attenuates the depressive- and anxiety-like behaviour in the corticosterone induced depression model via improving hippocampal neural plasticity. Brain Res. 2018, 1694, 55–62. [Google Scholar] [CrossRef] [Green Version]
  142. Zemdegs, J.; Martin, H.; Pintana, H.; Bullich, S.; Manta, S.; Marques, M.A.; Moro, C.; Laye, S.; Ducrocq, F.; Chattipakorn, N.; et al. Metformin Promotes Anxiolytic and Antidepressant-Like Responses in Insulin-Resistant Mice by Decreasing Circulating Branched-Chain Amino Acids. J. Neurosci. 2019, 39, 5935–5948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Kim, Y.K.; Kim, O.Y.; Song, J. Alleviation of Depression by Glucagon-Like Peptide 1 Through the Regulation of Neuroinflammation, Neurotransmitters, Neurogenesis, and Synaptic Function. Front Pharm. 2020, 11, 1270. [Google Scholar] [CrossRef] [PubMed]
Figure 1. General structure of EECs involved in gut–brain axis. The EECs present receptors on the brush border to sense the microbiota metabolites, toxins, pathogens, and nutrients in the lumen. Enteric glia cells and neurons connect to EECs. The secreted endocrine molecules affect afferent neuron signalling directly and (or) indirectly via EECs enteric glia cells. Efferent neurons bring the signal into the central nervous system. On the other hand, the central nervous system can pass the signal to EECs through efferent neurons. EECs, enteroendocrine cells.
Figure 1. General structure of EECs involved in gut–brain axis. The EECs present receptors on the brush border to sense the microbiota metabolites, toxins, pathogens, and nutrients in the lumen. Enteric glia cells and neurons connect to EECs. The secreted endocrine molecules affect afferent neuron signalling directly and (or) indirectly via EECs enteric glia cells. Efferent neurons bring the signal into the central nervous system. On the other hand, the central nervous system can pass the signal to EECs through efferent neurons. EECs, enteroendocrine cells.
Biomedicines 10 02577 g001
Figure 2. GLP1, 2, and serotonin secretion mechanisms in EECs and ECs, respectively. In ECs, TRPA1, piezol 1/2, Olfr, and ST2 bind to microbiota metabolites, pathogens, or nutrients, which increases calcium levels through directly increasing cAMP or endoplasmic reticulum IP3. Once the calcium level increases, it triggers the release of serotonin. Similar to ECs, EECs sense microbiota metabolites, toxins, pathogens, and nutrients through nutrient transporters and taste receptors. Further, they trigger an increase in calcium and sodium by directly increasing cAMP or endoplasmic reticulum IP3. Once the calcium level increases, it triggers the release of GLP1 and GLP2. cAMP, cyclic adenosine monophosphate; ECs, enterochromaffin cells; EECs, enteroendocrine cells; GLP1, glucagon-like peptide 1; GLP2, glucagon-like peptide 2; IP3, inositol trisphosphate; Olfr, olfactory receptor; TRPA1, transient receptor potential ankyrin 1.
Figure 2. GLP1, 2, and serotonin secretion mechanisms in EECs and ECs, respectively. In ECs, TRPA1, piezol 1/2, Olfr, and ST2 bind to microbiota metabolites, pathogens, or nutrients, which increases calcium levels through directly increasing cAMP or endoplasmic reticulum IP3. Once the calcium level increases, it triggers the release of serotonin. Similar to ECs, EECs sense microbiota metabolites, toxins, pathogens, and nutrients through nutrient transporters and taste receptors. Further, they trigger an increase in calcium and sodium by directly increasing cAMP or endoplasmic reticulum IP3. Once the calcium level increases, it triggers the release of GLP1 and GLP2. cAMP, cyclic adenosine monophosphate; ECs, enterochromaffin cells; EECs, enteroendocrine cells; GLP1, glucagon-like peptide 1; GLP2, glucagon-like peptide 2; IP3, inositol trisphosphate; Olfr, olfactory receptor; TRPA1, transient receptor potential ankyrin 1.
Biomedicines 10 02577 g002
Figure 3. α-synuclein accumulates in Parkinson’s disease through EECs. (a) The general pathway by which EECs trigger α-synuclein transfer into brain. Aggregated α-synuclein produced by EECs is transported into brain through afferent neurons and vagal nerve. (b) Cell signalling of α-synuclein aggregation present in EECs. The EECs present receptors on the brush border to sense the microbiota metabolites. Triggering of endoplasmic reticulum releases calcium through RyR. The increase in calcium induces reactive oxygen species (ROS) synthesis in mitochondria, which further creates α-synuclein aggregates. Microbiota metabolites also increase α-synuclein expression through Atg5 pathway in EECs. Atg5, autophagy-related 5; EECs, enteroendocrine cells; ROS, reactive oxygen species; RyR, ryanodine receptor.
Figure 3. α-synuclein accumulates in Parkinson’s disease through EECs. (a) The general pathway by which EECs trigger α-synuclein transfer into brain. Aggregated α-synuclein produced by EECs is transported into brain through afferent neurons and vagal nerve. (b) Cell signalling of α-synuclein aggregation present in EECs. The EECs present receptors on the brush border to sense the microbiota metabolites. Triggering of endoplasmic reticulum releases calcium through RyR. The increase in calcium induces reactive oxygen species (ROS) synthesis in mitochondria, which further creates α-synuclein aggregates. Microbiota metabolites also increase α-synuclein expression through Atg5 pathway in EECs. Atg5, autophagy-related 5; EECs, enteroendocrine cells; ROS, reactive oxygen species; RyR, ryanodine receptor.
Biomedicines 10 02577 g003
Figure 4. Visceral pain and depression pathologies involved with EECs and ECs. EECs and ECs sense the microbiota metabolites, toxins, pathogens, and nutrients in the lumen to secrete GLP1, 2, and serotonin, respectively. During pathology status, IELs produce proinflammatory cytokines, which enhance the progression of visceral pain and depression. GLP1 and GLP2 secreted by EECs have anti-inflammatory effects. Since the inflammation increases the visceral pain and depression through afferent neurons, the anti-inflammatory effect of GLP1 and GLP2 would decrease the visceral pain and depression. Although it is not clear how serotonin secreted by ECs affects depression through the afferent neurons, it has the effect of anti-inflammation, which might reduce depression. ECs, enterochromaffin cells; EECs, enteroendocrine cells; GLP1, glucagon-like peptide 1; GLP2, glucagon-like peptide 2; IELs, intraepithelial lymphocytes.
Figure 4. Visceral pain and depression pathologies involved with EECs and ECs. EECs and ECs sense the microbiota metabolites, toxins, pathogens, and nutrients in the lumen to secrete GLP1, 2, and serotonin, respectively. During pathology status, IELs produce proinflammatory cytokines, which enhance the progression of visceral pain and depression. GLP1 and GLP2 secreted by EECs have anti-inflammatory effects. Since the inflammation increases the visceral pain and depression through afferent neurons, the anti-inflammatory effect of GLP1 and GLP2 would decrease the visceral pain and depression. Although it is not clear how serotonin secreted by ECs affects depression through the afferent neurons, it has the effect of anti-inflammation, which might reduce depression. ECs, enterochromaffin cells; EECs, enteroendocrine cells; GLP1, glucagon-like peptide 1; GLP2, glucagon-like peptide 2; IELs, intraepithelial lymphocytes.
Biomedicines 10 02577 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yu, L.; Li, Y. Involvement of Intestinal Enteroendocrine Cells in Neurological and Psychiatric Disorders. Biomedicines 2022, 10, 2577. https://doi.org/10.3390/biomedicines10102577

AMA Style

Yu L, Li Y. Involvement of Intestinal Enteroendocrine Cells in Neurological and Psychiatric Disorders. Biomedicines. 2022; 10(10):2577. https://doi.org/10.3390/biomedicines10102577

Chicago/Turabian Style

Yu, Liangen, and Yihang Li. 2022. "Involvement of Intestinal Enteroendocrine Cells in Neurological and Psychiatric Disorders" Biomedicines 10, no. 10: 2577. https://doi.org/10.3390/biomedicines10102577

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