1
|
Milešević M, Matić Jelić I, Rumenović V, Ivanjko N, Vukičević S, Bordukalo-Nikšić T. The Influence of BMP6 on Serotonin and Glucose Metabolism. Int J Mol Sci 2024; 25:7842. [PMID: 39063084 PMCID: PMC11276723 DOI: 10.3390/ijms25147842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Previous studies have suggested a potential role of bone morphogenetic protein 6 (BMP6) in glucose metabolism, which also seems to be regulated by serotonin (5-hydroxytryptamine, 5HT), a biogenic amine with multiple roles in the organism. In this study, we explored possible interactions between BMP6, serotonin, and glucose metabolism regulation. The effect of BMP6 or 5HT on pancreatic β-cells has been studied in vitro using the INS-1 832/13 rat insulinoma cell line. Studies in vivo have been performed on mice with the global deletion of the Bmp6 gene (BMP6-/-) and included glucose and insulin tolerance tests, gene expression studies using RT-PCR, immunohistochemistry, and ELISA analyses. We have shown that BMP6 and 5HT treatments have the opposite effect on insulin secretion from INS-1 cells. The effect of BMP6 on the 5HT system in vivo depends on the tissue studied, with no observable systemic effect on peripheral 5HT metabolism. BMP6 deficiency does not cause diabetic changes, although a mild difference in insulin tolerance test between BMP6-/- and WT mice was observed. In conclusion, BMP6 does not directly influence glucose metabolism, but there is a possibility that its deletion causes slowly developing changes in glucose and serotonin metabolism, which would become more expressed with ageing.
Collapse
Affiliation(s)
| | | | | | | | | | - Tatjana Bordukalo-Nikšić
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (M.M.); (I.M.J.); (V.R.); (N.I.); (S.V.)
| |
Collapse
|
2
|
Jiang L, Han D, Hao Y, Song Z, Sun Z, Dai Z. Linking serotonin homeostasis to gut function: Nutrition, gut microbiota and beyond. Crit Rev Food Sci Nutr 2024; 64:7291-7310. [PMID: 36861222 DOI: 10.1080/10408398.2023.2183935] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Serotonin (5-HT) produced by enterochromaffin (EC) cells in the digestive tract is crucial for maintaining gut function and homeostasis. Nutritional and non-nutritional stimuli in the gut lumen can modulate the ability of EC cells to produce 5-HT in a temporal- and spatial-specific manner that toning gut physiology and immune response. Of particular interest, the interactions between dietary factors and the gut microbiota exert distinct impacts on gut 5-HT homeostasis and signaling in metabolism and the gut immune response. However, the underlying mechanisms need to be unraveled. This review aims to summarize and discuss the importance of gut 5-HT homeostasis and its regulation in maintaining gut metabolism and immune function in health and disease with special emphasis on different types of nutrients, dietary supplements, processing, and gut microbiota. Cutting-edge discoveries in this area will provide the basis for the development of new nutritional and pharmaceutical strategies for the prevention and treatment of serotonin homeostasis-related gut and systematic disorders and diseases.
Collapse
Affiliation(s)
- Lili Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Youling Hao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhuan Song
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhiyuan Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
3
|
Cai J, Cheung J, Cheung SWM, Chin KTC, Leung RWK, Lam RST, Sharma R, Yiu JHC, Woo CW. Butyrate acts as a positive allosteric modulator of the 5-HT transporter to decrease availability of 5-HT in the ileum. Br J Pharmacol 2024; 181:1654-1670. [PMID: 38129963 DOI: 10.1111/bph.16305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Radiation therapy-induced gastrointestinal distress is partly associated with the elimination of gut microbiota. The effectiveness of 5-HT receptor antagonists to treat radiation therapy-induced emesis implies a pathophysiological role of 5-HT. Peripheral 5-HT is derived from intestinal epithelium. We have investigated the role of gut microbiota in regulating intestinal 5-HT availability. EXPERIMENTAL APPROACH A radiation therapy murine model accompanied by faecal microbiota transplantation from donors fed different diets was investigated, and mouse ileal organoids were used for mechanistic studies. The clinical relevance was validated by a small-scale human study. KEY RESULTS Short-term high-fat diet (HFD) induced gut bacteria to produce butyrate. Irradiated mice receiving HFD-induced microbiome had the lowest ileal levels of 5-HT, compared with other recipients. Treatment with butyrate increased 5-HT uptake in mouse ileal organoids, assayed by the real-time tracking of a fluorescent substrate for monoamine transporters. Silencing the 5-HT transporter (SERT) in the organoids abolished butyrate-stimulated 5-HT uptake. The competitive tests using different types of selective 5-HT reuptake inhibitors suggested that butyrate acted as a positive allosteric modulator of SERT. In human gut microbiota, butyrate production was associated with the interconversion between acetate and butyrate. Faecal contents of both acetate and butyrate were negatively associated with serum 5-HT, but only butyrate was positively correlated with body mass index in humans. CONCLUSION AND IMPLICATIONS Short-term HFD may be beneficial for alleviating gastrointestinal reactions by increasing butyrate to suppress local 5-HT levels and providing energy to cancer patients given radiation therapy.
Collapse
Affiliation(s)
- Jieling Cai
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jamie Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Samson W M Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Karie T C Chin
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ricky W K Leung
- Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ronald S T Lam
- Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Rakesh Sharma
- Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jensen H C Yiu
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Connie W Woo
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Micon Analytics, Toronto, Canada
| |
Collapse
|
4
|
Sun Y, Zhang S, Nie Q, He H, Tan H, Geng F, Ji H, Hu J, Nie S. Gut firmicutes: Relationship with dietary fiber and role in host homeostasis. Crit Rev Food Sci Nutr 2023; 63:12073-12088. [PMID: 35822206 DOI: 10.1080/10408398.2022.2098249] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Firmicutes and Bacteroidetes are the predominant bacterial phyla colonizing the healthy human gut. Accumulating evidence suggests that dietary fiber plays a crucial role in host health, yet most studies have focused on how the dietary fiber affects health through gut Bacteroides. More recently, gut Firmicutes have been found to possess many genes responsible for fermenting dietary fiber, and could also interact with the intestinal mucosa and thereby contribute to homeostasis. Consequently, the relationship between dietary fiber and Firmicutes is of interest, as well as the role of Firmicutes in host health. In this review, we summarize the current knowledge regarding the molecular mechanism of dietary fiber degradation by gut Firmicutes and explain the communication pathway of the dietary fiber-Firmicutes-host axis, and the beneficial effects of dietary fiber-induced Firmicutes and their metabolites on health. A better understanding of the dialogue sustained by the dietary fiber-Firmicutes axis and the host could provide new insights into probiotic therapy and novel dietary interventions aimed at increasing the abundance of Firmicutes (such as Faecalibacterium, Lactobacillus, and Roseburia) to promote health.
Collapse
Affiliation(s)
- Yonggan Sun
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Shanshan Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Huijun He
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Huizi Tan
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Haihua Ji
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Dothel G, Barbaro MR, Di Vito A, Ravegnini G, Gorini F, Monesmith S, Coschina E, Benuzzi E, Fuschi D, Palombo M, Bonomini F, Morroni F, Hrelia P, Barbara G, Angelini S. New insights into irritable bowel syndrome pathophysiological mechanisms: contribution of epigenetics. J Gastroenterol 2023; 58:605-621. [PMID: 37160449 PMCID: PMC10307698 DOI: 10.1007/s00535-023-01997-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
Irritable bowel syndrome (IBS) is a complex multifactorial condition including alterations of the gut-brain axis, intestinal permeability, mucosal neuro-immune interactions, and microbiota imbalance. Recent advances proposed epigenetic factors as possible regulators of several mechanisms involved in IBS pathophysiology. These epigenetic factors include biomolecular mechanisms inducing chromosome-related and heritable changes in gene expression regardless of DNA coding sequence. Accordingly, altered gut microbiota may increase the production of metabolites such as sodium butyrate, a prominent inhibitor of histone deacetylases. Patients with IBS showed an increased amount of butyrate-producing microbial phila as well as an altered profile of methylated genes and micro-RNAs (miRNAs). Importantly, gene acetylation as well as specific miRNA profiles are involved in different IBS mechanisms and may be applied for future diagnostic purposes, especially to detect increased gut permeability and visceromotor dysfunctions. In this review, we summarize current knowledge of the role of epigenetics in IBS pathophysiology.
Collapse
Affiliation(s)
- Giovanni Dothel
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Connect By Circular Lab SRL, Madrid, Spain
| | | | - Aldo Di Vito
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Francesca Gorini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sarah Monesmith
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Emma Coschina
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Eva Benuzzi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Marta Palombo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bonomini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabiana Morroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| | - Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Inter-Departmental Center for Health Sciences & Technologies, CIRI-SDV, University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
Jadhav VV, Han J, Fasina Y, Harrison SH. Connecting gut microbiomes and short chain fatty acids with the serotonergic system and behavior in Gallus gallus and other avian species. Front Physiol 2022; 13:1035538. [PMID: 36406988 PMCID: PMC9667555 DOI: 10.3389/fphys.2022.1035538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/03/2022] [Indexed: 12/05/2022] Open
Abstract
The chicken gastrointestinal tract has a diverse microbial community. There is increasing evidence for how this gut microbiome affects specific molecular pathways and the overall physiology, nervous system and behavior of the chicken host organism due to a growing number of studies investigating conditions such as host diet, antibiotics, probiotics, and germ-free and germ-reduced models. Systems-level investigations have revealed a network of microbiome-related interactions between the gut and state of health and behavior in chickens and other animals. While some microbial symbionts are crucial for maintaining stability and normal host physiology, there can also be dysbiosis, disruptions to nutrient flow, and other outcomes of dysregulation and disease. Likewise, alteration of the gut microbiome is found for chickens exhibiting differences in feather pecking (FP) behavior and this alteration is suspected to be responsible for behavioral change. In chickens and other organisms, serotonin is a chief neuromodulator that links gut microbes to the host brain as microbes modulate the serotonin secreted by the host's own intestinal enterochromaffin cells which can stimulate the central nervous system via the vagus nerve. A substantial part of the serotonergic network is conserved across birds and mammals. Broader investigations of multiple species and subsequent cross-comparisons may help to explore general functionality of this ancient system and its increasingly apparent central role in the gut-brain axis of vertebrates. Dysfunctional behavioral phenotypes from the serotonergic system moreover occur in both birds and mammals with, for example, FP in chickens and depression in humans. Recent studies of the intestine as a major site of serotonin synthesis have been identifying routes by which gut microbial metabolites regulate the chicken serotonergic system. This review in particular highlights the influence of gut microbial metabolite short chain fatty acids (SCFAs) on the serotonergic system. The role of SCFAs in physiological and brain disorders may be considerable because of their ability to cross intestinal as well as the blood-brain barriers, leading to influences on the serotonergic system via binding to receptors and epigenetic modulations. Examinations of these mechanisms may translate into a more general understanding of serotonergic system development within chickens and other avians.
Collapse
Affiliation(s)
- Vidya V. Jadhav
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC, United States
| | - Jian Han
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC, United States
| | - Yewande Fasina
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, United States,*Correspondence: Yewande Fasina, ; Scott H. Harrison,
| | - Scott H. Harrison
- Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC, United States,*Correspondence: Yewande Fasina, ; Scott H. Harrison,
| |
Collapse
|
7
|
Iimura S, Takasugi S. Sensory Processing Sensitivity and Gastrointestinal Symptoms in Japanese Adults. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9893. [PMID: 36011526 PMCID: PMC9408471 DOI: 10.3390/ijerph19169893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Sensory processing sensitivity is a personality or temperamental trait defined as individual differences in the tendency to perceive and process both positive and negative stimuli and experiences. Studies have shown that high sensitivity is correlated with psychosocial health, including depression and anxiety. However, its relationship with physical health has not been clarified. To fill this gap, using a large sample size with sufficient statistical power, an adult sample not including university students, and a range of covariates, this study examined the association between gastrointestinal symptoms as an indicator of physical health and sensory processing sensitivity. METHODS In this cross-sectional study, the participants were 863 Japanese adults (female = 450; male = 413; Mage = 30.4 years; SD = 4.9) who completed a web-based questionnaire. We statistically controlled for sociodemographic characteristics and examined whether sensory processing sensitivity is correlated with gastrointestinal symptoms. RESULTS The results showed that highly sensitive individuals were more likely to experience a wide range of gastrointestinal symptoms in the past week, including reflux symptoms, abdominal pain, indigestion symptoms, diarrhea symptoms, and constipation symptoms, even when statistically controlling for the participants' sociodemographic characteristics. CONCLUSIONS Our findings suggest that high sensory processing sensitivity is associated with physical health. Some of the potential causes of this are also discussed.
Collapse
Affiliation(s)
- Shuhei Iimura
- Soka University, 1-236 Tangi-machi, Hachioji, Tokyo 192-8577, Japan
| | - Satoshi Takasugi
- R&D Division, Meiji Co., Ltd., 1-29-1 Nanakuni, Hachioji, Tokyo 192-0919, Japan
| |
Collapse
|
8
|
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; 162:1962-1974.e6. [PMID: 35167867 PMCID: PMC9117493 DOI: 10.1053/j.gastro.2022.02.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/17/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Increased colonic serotonin (5-HT) level and decreased serotonin reuptake transporter (SERT) expression in irritable bowel syndrome (IBS) may contribute to diarrhea and visceral hypersensitivity. We investigated whether mucosal SERT is modulated by gut microbiota via a mast cell-prostaglandin E2 (PGE2) pathway. METHODS C57Bl/6 mice received intracolonic infusion of fecal supernatant (FS) from healthy controls or patients with diarrhea-predominant irritable bowel syndrome (IBS-D). The role of mast cells was studied in mast cell-deficient mice. Colonic organoids and/or mast cells were used for in vitro experiments. SERT expression was measured by quantitative polymerase chain reaction and Western blot. Visceromotor responses to colorectal distension and colonic transit were assessed. RESULTS Intracolonic infusion of IBS-D FS in mice caused an increase in mucosal 5-HT compared with healthy control FS, accompanied by ∼50% reduction in SERT expression. Mast cell stabilizers, cyclooxygenase-2 inhibitors, and PGE2 receptor antagonist prevented SERT downregulation. Intracolonic infusion of IBS-D FS failed to reduce SERT expression in mast cell-deficient (W/Wv) mice. This response was restored by mast cell reconstitution. The downregulation of SERT expression evoked by IBS FS was prevented by lipopolysaccharide (LPS) antagonist LPS from Rhodobacter sphaeroides and a bacterial trypsin inhibitor. In vitro LPS treatment caused increased cyclooxygenase-2 expression and PGE2 release from cultured mouse mast cells. Intracolonic infusion of IBS-D FS in mice reduced colonic transit, increased fecal water content, and increased visceromotor responses to colorectal distension. Ondansetron prevented these changes. CONCLUSIONS Fecal LPS acting in concert with trypsin in patients with IBS-D stimulates mucosal mast cells to release PGE2, which downregulates mucosal SERT, resulting in increased mucosal 5-HT. This may contribute to diarrhea and abdominal pain common in IBS.
Collapse
Affiliation(s)
| | | | | | - Chung Owyang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
9
|
Horvath TD, Ihekweazu FD, Haidacher SJ, Ruan W, Engevik KA, Fultz R, Hoch KM, Luna RA, Oezguen N, Spinler JK, Haag AM, Versalovic J, Engevik MA. Bacteroides ovatus colonization influences the abundance of intestinal short chain fatty acids and neurotransmitters. iScience 2022; 25:104158. [PMID: 35494230 PMCID: PMC9038548 DOI: 10.1016/j.isci.2022.104158] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 11/01/2021] [Accepted: 03/23/2022] [Indexed: 12/18/2022] Open
Abstract
Gut microbes can synthesize multiple neuro-active metabolites. We profiled neuro-active compounds produced by the gut commensal Bacteroides ovatus in vitro and in vivo by LC-MS/MS. We found that B. ovatus generates acetic acid, propionic acid, isobutyric acid, and isovaleric acid. In vitro, B. ovatus consumed tryptophan and glutamate and synthesized the neuro-active compounds glutamine and GABA. Consistent with our LC-MS/MS-based in vitro data, we observed elevated levels of acetic acid, propionic acid, isobutyric acid, and isovaleric acid in the intestines of B. ovatus mono-associated mice compared with germ-free controls. B. ovatus mono-association also increased the concentrations of intestinal GABA and decreased the concentrations of tryptophan and glutamine compared with germ-free controls. Computational network analysis revealed unique links between SCFAs, neuro-active compounds, and colonization status. These results highlight connections between microbial colonization and intestinal neurotransmitter concentrations, suggesting that B. ovatus selectively influences the presence of intestinal neurotransmitters.
Collapse
Affiliation(s)
- Thomas D. Horvath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Faith D. Ihekweazu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, TX, USA
| | - Sigmund J. Haidacher
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, TX, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Robert Fultz
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Kathleen M. Hoch
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Ruth Ann Luna
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Numan Oezguen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Jennifer K. Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Anthony M. Haag
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Melinda A. Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, 173 Ashley Ave, BSB 621, Charleston, SC 29425, USA
| |
Collapse
|
10
|
Subramanian VS, Teafatiller T, Moradi H, Marchant JS. Histone deacetylase inhibitors regulate vitamin C transporter functional expression in intestinal epithelial cells. J Nutr Biochem 2021; 98:108838. [PMID: 34403723 DOI: 10.1016/j.jnutbio.2021.108838] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/04/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023]
Abstract
Intestinal absorption of vitamin C in humans is mediated via the sodium-dependent vitamin C transporters (hSVCT1 and hSVCT2). hSVCT1 and hSVCT2 are localized at the apical and basolateral membranes, respectively, of polarized intestinal epithelia. Studies have identified low plasma levels of vitamin C and decreased expression of hSVCT1 in patients with several inflammatory conditions including inflammatory bowel disease (IBD). Investigating the underlying mechanisms responsible for regulating hSVCT1 expression are critical for understanding vitamin C homeostasis, particularly in conditions where suboptimal vitamin C levels detrimentally affect human health. Previous research has shown that hSVCT1 expression is regulated at the transcriptional level, however, little is known about epigenetic regulatory pathways that modulate hSVCT1 expression in the intestine. In this study, we found that hSVCT1 expression and function were significantly decreased in intestinal epithelial cells by the histone deacetylase inhibitors (HDACi), valproic acid (VPA), and sodium butyrate (NaB). Further, expression of transcription factor HNF1α, which is critical for SLC23A1 promoter activity, was significantly down regulated in VPA-treated cells. Chromatin immunoprecipitation (ChIP) assays showed significantly increased enrichment of tetra-acetylated histone H3 and H4 within the SLC23A1 promoter following VPA treatment. In addition, knockdown of HDAC isoforms two, and three significantly decreased hSVCT1 functional expression. Following VPA administration to mice, functional expression of SVCT1 in the jejunum was significantly decreased. Collectively, these in vitro and in vivo studies demonstrate epigenetic regulation of SVCT1 expression in intestinal epithelia partly mediated through HDAC isoforms two and three.
Collapse
Affiliation(s)
| | - Trevor Teafatiller
- Department of Medicine, University of California, Irvine, California, USA
| | - Hamid Moradi
- Department of Medicine, University of California, Irvine, California, USA; Tibor Rubin VA Medical Center, Long Beach, California, USA
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
11
|
Tarashi S, Karimipoor M, Siadat SD, Fuso A. Epigenetic modifications in host-bacterial dialogues: more than meets the eye. Epigenomics 2021; 14:5-9. [PMID: 34676788 DOI: 10.2217/epi-2021-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Samira Tarashi
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, 13169-43551, Iran.,Mycobacteriology & Pulmonary Research Department, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Morteza Karimipoor
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, 13169-43551, Iran.,Mycobacteriology & Pulmonary Research Department, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, 00161, Italy
| |
Collapse
|
12
|
Gerbeth L, Glauben R. Histone Deacetylases in the Inflamed Intestinal Epithelium-Promises of New Therapeutic Strategies. Front Med (Lausanne) 2021; 8:655956. [PMID: 33842512 PMCID: PMC8032862 DOI: 10.3389/fmed.2021.655956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/22/2022] Open
Abstract
The intestinal epithelium is a complex, dynamic barrier that separates luminal contents from the immune compartment while mediating nutrient absorption and controlled passage of antigens to convey oral tolerance. A compromised epithelial barrier often leads to inflammation because immune cells in the lamina propria come into direct contact with luminal antigens. Defects in epithelial cell function were also shown to be involved in the etiology of inflammatory bowel diseases. These are severe, chronically relapsing inflammatory conditions of the gastrointestinal tract that also increase the risk of developing colorectal cancer. Despite major efforts of the scientific community, the precise causes and drivers of these conditions still remain largely obscured impeding the development of a permanent cure. Current therapeutic approaches mostly focus on alleviating symptoms by targeting immune cell signaling. The protein family of histone deacetylases (HDACs) has gained increasing attention over the last years, as HDAC inhibitors were shown to be potent tumor cell suppressors and also alleviate morbid inflammatory responses. Recent research continuously identifies new roles for specific HDACs suggesting that HDACs influence the cell signaling network from many different angles. This makes HDACs very interesting targets for therapeutic approaches but predicting effects after system manipulations can be difficult. In this review, we want to provide a comprehensive overview of current knowledge about the individual roles of HDACs in the intestinal epithelium to evaluate their therapeutic potential for inflammatory conditions of the gut.
Collapse
Affiliation(s)
- Lorenz Gerbeth
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Medical Biotechnology, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Rainer Glauben
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
13
|
Hartstra AV, Schüppel V, Imangaliyev S, Schrantee A, Prodan A, Collard D, Levin E, Dallinga-Thie G, Ackermans MT, Winkelmeijer M, Havik SR, Metwaly A, Lagkouvardos I, Nier A, Bergheim I, Heikenwalder M, Dunkel A, Nederveen AJ, Liebisch G, Mancano G, Claus SP, Benítez-Páez A, la Fleur SE, Bergman JJ, Gerdes V, Sanz Y, Booij J, Kemper E, Groen AK, Serlie MJ, Haller D, Nieuwdorp M. Infusion of donor feces affects the gut-brain axis in humans with metabolic syndrome. Mol Metab 2020; 42:101076. [PMID: 32916306 PMCID: PMC7536740 DOI: 10.1016/j.molmet.2020.101076] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/31/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Increasing evidence indicates that intestinal microbiota play a role in diverse metabolic processes via intestinal butyrate production. Human bariatric surgery data suggest that the gut-brain axis is also involved in this process, but the underlying mechanisms remain unknown. METHODS We compared the effect of fecal microbiota transfer (FMT) from post-Roux-en-Y gastric bypass (RYGB) donors vs oral butyrate supplementation on (123I-FP-CIT-determined) brain dopamine transporter (DAT) and serotonin transporter (SERT) binding as well as stable isotope-determined insulin sensitivity at baseline and after 4 weeks in 24 male and female treatment-naïve metabolic syndrome subjects. Plasma metabolites and fecal microbiota were also determined at these time points. RESULTS We observed an increase in brain DAT after donor FMT compared to oral butyrate that reduced this binding. However, no effect on body weight and insulin sensitivity was demonstrated after post-RYGB donor feces transfer in humans with metabolic syndrome. Increases in fecal levels of Bacteroides uniformis were significantly associated with an increase in DAT, whereas increases in Prevotella spp. showed an inverse association. Changes in the plasma metabolites glycine, betaine, methionine, and lysine (associated with the S-adenosylmethionine cycle) were also associated with altered striatal DAT expression. CONCLUSIONS Although more and larger studies are needed, our data suggest a potential gut microbiota-driven modulation of brain dopamine and serotonin transporters in human subjects with obese metabolic syndrome. These data also suggest the presence of a gut-brain axis in humans that can be modulated. NTR REGISTRATION 4488.
Collapse
Affiliation(s)
- Annick V Hartstra
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Valentina Schüppel
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Sultan Imangaliyev
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Andrei Prodan
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Didier Collard
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Geesje Dallinga-Thie
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Maaike Winkelmeijer
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Stefan R Havik
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Ilias Lagkouvardos
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Anika Nier
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Austria
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Freising, Germany
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Gerhard Liebisch
- Department of Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Giulia Mancano
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Sandrine P Claus
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Alfonso Benítez-Páez
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Susanne E la Fleur
- Laboratory of Endocrinology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Elles Kemper
- Department of Clinical Pharmacy, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany; ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Holton NW, Singhal M, Kumar A, Ticho AL, Manzella CR, Malhotra P, Jarava D, Saksena S, Dudeja PK, Alrefai WA, Gill RK. Hepatocyte nuclear factor-4α regulates expression of the serotonin transporter in intestinal epithelial cells. Am J Physiol Cell Physiol 2020; 318:C1294-C1304. [PMID: 32348179 PMCID: PMC7311735 DOI: 10.1152/ajpcell.00477.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022]
Abstract
The serotonin transporter (SERT) functions to regulate the availability of serotonin (5-HT) in the brain and intestine. An intestine-specific mRNA variant arising from a unique transcription start site and alternative promoter in the SERT gene has been identified (iSERT; spanning exon 1C). A decrease in SERT is implicated in several gut disorders, including inflammatory bowel diseases (IBD). However, little is known about mechanisms regulating the iSERT variant, and a clearer understanding is warranted for targeting SERT for the treatment of gut disorders. The current studies examined the expression of iSERT across different human intestinal regions and investigated its regulation by HNF4α (hepatic nuclear factor-4α), a transcription factor important for diverse cellular functions. iSERT mRNA abundance was highest in the human ileum and Caco-2 cell line. iSERT mRNA expression was downregulated by loss of HNF4α (but not HNF1α, HNF1β, or FOXA1) in Caco-2 cells. Overexpression of HNF4α increased iSERT mRNA concomitant with an increase in SERT protein. Progressive promoter deletion and site-directed mutagenesis revealed that the HNF4α response element spans nucleotides -1,163 to -1150 relative to the translation start site. SERT mRNA levels in the intestine were drastically reduced in the intestine-specific HNF4α-knockout mice relative to HNF4αFL/FL mice. Both HNF4α and SERT mRNA levels were also downregulated in mouse model of ileitis (SAMP) compared with AKR control mice. These results establish the transcriptional regulation of iSERT at the gut-specific internal promoter (hSERTp2) and have identified HNF4α as a critical modulator of basal SERT expression in the intestine.
Collapse
Affiliation(s)
- Nathaniel W Holton
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Megha Singhal
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Alexander L Ticho
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Christopher R Manzella
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Pooja Malhotra
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - David Jarava
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Pradeep K Dudeja
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Waddah A Alrefai
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
15
|
Deryabina IB, Andrianov VV, Muranova LN, Bogodvid TK, Gainutdinov KL. Effects of Thryptophan Hydroxylase Blockade by P-Chlorophenylalanine on Contextual Memory Reconsolidation after Training of Different Intensity. Int J Mol Sci 2020; 21:E2087. [PMID: 32197439 PMCID: PMC7139692 DOI: 10.3390/ijms21062087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 01/09/2023] Open
Abstract
The processes of memory formation and its storage are extremely dynamic. Therefore, the determination of the nature and temporal evolution of the changes that underlie the molecular mechanisms of retrieval and cause reconsolidation of memory is the key to understanding memory formation. Retrieval induces the plasticity, which may result in reconsolidation of the original memory and needs critical molecular events to stabilize the memory or its extinction. 4-Chloro-DL-phenylalanine (P-chlorophenylalanine-PCPA) depresses the most limiting enzyme of serotonin synthesis the tryptophan hydroxylase. It is known that PCPA reduces the serotonin content in the brain up to 10 times in rats (see Methods). We hypothesized that the PCPA could behave the similar way in snails and could reduce the content of serotonin in snails. Therefore, we investigated the effect of PCPA injection on contextual memory reconsolidation using a protein synthesis blocker in snails after training according to two protocols of different intensities. The results obtained in training according to the first protocol using five electrical stimuli per day for 5 days showed that reminding the training environment against the background of injection of PCPA led to a significant decrease in contextual memory. At the same time, the results obtained in training according to the second protocol using three electrical stimuli per day for 5 days showed that reminding the training environment against the injection of PCPA did not result in a significant change in contextual memory. The obtain results allowed us to conclude that the mechanisms of processes developed during the reconsolidation of contextual memory after a reminding depend both on the intensity of learning and on the state of the serotonergic system.
Collapse
Affiliation(s)
- Irina B. Deryabina
- Laboratory of Neuroreabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (I.B.D.); (V.V.A.); (L.N.M.); (T.K.B.)
| | - Viatcheslav V. Andrianov
- Laboratory of Neuroreabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (I.B.D.); (V.V.A.); (L.N.M.); (T.K.B.)
- Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of Sciences, 420000 Kazan, Russia
| | - Lyudmila N. Muranova
- Laboratory of Neuroreabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (I.B.D.); (V.V.A.); (L.N.M.); (T.K.B.)
| | - Tatiana K. Bogodvid
- Laboratory of Neuroreabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (I.B.D.); (V.V.A.); (L.N.M.); (T.K.B.)
- Department of Biomedical Sciences, Volga Region State Academy of Physical Culture, Sport and Tourism, 420000 Kazan, Russia
| | - Khalil L. Gainutdinov
- Laboratory of Neuroreabilitation of Motor Disorders, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420000 Kazan, Russia; (I.B.D.); (V.V.A.); (L.N.M.); (T.K.B.)
- Laboratory of Spin Physics and Spin Chemistry, Zavoisky Physical-Technical Institute of the Russian Academy of Sciences, 420000 Kazan, Russia
| |
Collapse
|
16
|
Manzella C, Singhal M, Ackerman M, Alrefai WA, Saksena S, Dudeja PK, Gill RK. Serotonin transporter untranslated regions influence mRNA abundance and protein expression. GENE REPORTS 2020; 18. [PMID: 34113740 DOI: 10.1016/j.genrep.2019.100513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The serotonin transporter (SERT, SLC6A4) is a Na+-dependent transporter that regulates the availability of serotonin (5-HT, 5-hydroxytryptamine), a key neurotransmitter and hormone in the brain and the intestine. The human SERT gene consists of two alternate promoters that drive expression of an identical SERT protein. However, there are different mRNA transcript variants derived from these two promoters that differ in their 5' untranslated region (5'UTR), which is the region of the mRNA upstream from the protein-coding region. Two of these transcripts contain exon-1a and are abundant in neuronal tissue, whereas the third transcript contains exon-1c and is abundant in the intestine. The 3'UTR is nearly identical among the transcripts. Current studies tested the hypothesis that the UTRs of SERT influence its expression in intestinal epithelial cells (IECs) by controlling mRNA or protein levels. The SERT UTRs were cloned into luciferase reporter plasmids and luciferase mRNA and activity were measured following transient transfection of the UTR constructs into the model IEC Caco-2. Luciferase activity and mRNA abundance were higher than the empty vector for two of the three 5'UTR variants. Calculation of translation index (luciferase activity divided by the relative luciferase mRNA level) revealed that the exon-1a containing 5'UTRs had enhanced translation when compared to the exon-1c containing 5'UTR which exhibited a low translation efficiency. Compared to the empty vector, the SERT 3'UTR markedly decreased luciferase activity. In silico analysis of the SERT 3'UTR revealed many conserved potential miRNA binding sites that may be responsible for this decrease. In conclusion, we have shown that the UTRs of SERT regulate mRNA abundance and protein expression. Delineating the molecular basis by which the UTRs of SERT influence its expression will lead to an increased understanding of post-transcriptional regulation of SERT in GI disorders associated with altered 5-HT availability.
Collapse
Affiliation(s)
- Christopher Manzella
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Megha Singhal
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Max Ackerman
- University of Illinois at Chicago, College of Liberal Arts and Sciences, United States of America
| | - Waddah A Alrefai
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| | - Seema Saksena
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| | - Pradeep K Dudeja
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| | - Ravinder K Gill
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| |
Collapse
|
17
|
Analysis of gut microbiota of obese individuals with type 2 diabetes and healthy individuals. PLoS One 2019; 14:e0226372. [PMID: 31891582 PMCID: PMC6938335 DOI: 10.1371/journal.pone.0226372] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) accounts for 90% of diabetes cases worldwide. The majority of T2DM patients are obese. Dysbiosis in the gut microflora is strongly associated with the pathogenesis of obesity and T2DM; however, the microbiome of obese-T2DM individuals in the Pakistani population remains unexplored. The gut microbiota signature of 60 Pakistani adults was studied using 16S rRNA sequencing targeting V3–V4 hypervariable regions. The sequence analysis revealed that bacteria from Firmicutes were predominant along with those from Clostridia and Negativicutes, whereas bacteria from Verrucomicrobia, Bacteroidetes, Proteobacteria, and Elusimicrobia were less abundant among the obese T2DM patients. These data distinctively vary from those in reports on the Indian population. The difference in gut microbiota could presumably be related to the distinct lifestyle and eastern dietary habits (high carbohydrate and fat intake, low fiber intake) and unregulated antibiotic consumption. This is the first study carried out to understand the gut microbiome and its correlation with individual life style of obese T2DM patients in the Pakistani population.
Collapse
|
18
|
Baudry A, Pietri M, Launay JM, Kellermann O, Schneider B. Multifaceted Regulations of the Serotonin Transporter: Impact on Antidepressant Response. Front Neurosci 2019; 13:91. [PMID: 30809118 PMCID: PMC6379337 DOI: 10.3389/fnins.2019.00091] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
Serotonin transporter, SERT (SLC64A for solute carrier family 6, member A4), is a twelve transmembrane domain (TMDs) protein that assumes the uptake of serotonin (5-HT) through dissipation of the Na+ gradient established by the electrogenic pump Na/K ATPase. Abnormalities in 5-HT level and signaling have been associated with various disorders of the central nervous system (CNS) such as depression, obsessive-compulsive disorder, anxiety disorders, and autism spectrum disorder. Since the 50s, SERT has raised a lot of interest as being the target of a class of antidepressants, the Serotonin Selective Reuptake Inhibitors (SSRIs), used in clinics to combat depressive states. Because of the refractoriness of two-third of patients to SSRI treatment, a better understanding of the mechanisms regulating SERT functions is of priority. Here, we review how genetic and epigenetic regulations, post-translational modifications of SERT, and specific interactions between SERT and a set of diverse partners influence SERT expression, trafficking to and away from the plasma membrane and activity, in connection with the neuronal adaptive cell response to SSRI antidepressants.
Collapse
Affiliation(s)
- Anne Baudry
- INSERM UMR-S 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, Paris, France
| | - Mathea Pietri
- INSERM UMR-S 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, Paris, France
| | - Jean-Marie Launay
- Hôpital Lariboisière, AP-HP, INSERM UMR-S 942, Paris, France.,Pharma Research Department, Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Odile Kellermann
- INSERM UMR-S 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, Paris, France
| | - Benoit Schneider
- INSERM UMR-S 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR-S 1124, Paris, France
| |
Collapse
|
19
|
Vincent AD, Wang XY, Parsons SP, Khan WI, Huizinga JD. Abnormal absorptive colonic motor activity in germ-free mice is rectified by butyrate, an effect possibly mediated by mucosal serotonin. Am J Physiol Gastrointest Liver Physiol 2018; 315:G896-G907. [PMID: 30095295 DOI: 10.1152/ajpgi.00237.2017] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The role of short-chain fatty acids (SCFAs) in the control of colonic motility is controversial. Germ-free (GF) mice are unable to produce these metabolites and serve as a model to study how their absence affects colonic motility. GF transit is slower than controls, and colonization of these mice improves transit and serotonin [5-hydroxytryptamine (5-HT)] levels. Our aim was to determine the role SCFAs play in improving transit and whether this is dependent on mucosal 5-HT signaling. Motility was assessed in GF mice via spatiotemporal mapping. First, motor patterns in the whole colon were measured ex vivo with or without luminal SCFA, and outflow from the colon was recorded to quantify outflow caused by individual propulsive contractions. Second, artificial fecal pellet propulsion was measured. Motility was then assessed in tryptophan hydroxylase-1 (TPH1) knockout (KO) mice, devoid of mucosal 5-HT, with phosphate buffer, butyrate, or propionate intraluminal perfusion. GF mice exhibited a lower proportion of propulsive contractions, lower volume of outflow/contraction, slower velocity of contractions, and slower propulsion of fecal pellets compared with controls. SCFAs changed motility patterns to that of controls in all parameters. Butyrate administration increased the proportion of propulsive contractions in controls yet failed to in TPH1 KO mice. Propionate inhibited propulsive contractions in all mice. Our results reveal significant abnormalities in the propulsive nature of colonic motor patterns in GF mice, explaining the decreased transit time in in vivo studies. We show that butyrate but not propionate activates propulsive motility and that this may require mucosal 5-HT. NEW & NOTEWORTHY Understanding the role that the microbiota play in governing the physiology of colonic motility is lacking. Here, we offer for the first time, to our knowledge, a detailed analysis of colonic motor patterns and pellet propulsion using spatiotemporal mapping in the absence of microbiota. We show a striking difference in germ-free and control phenotypes and attribute this to a lack of fermentation-produced short-chain fatty acid. We then show that butyrate but not propionate can restore motility and that the butyrate effect likely requires mucosal 5-hydroxytryptamine.
Collapse
Affiliation(s)
- Alexander D Vincent
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Xuan-Yu Wang
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Sean P Parsons
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Waliul I Khan
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Jan D Huizinga
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| |
Collapse
|
20
|
Sherman Horev H, Rabinowitz KM, Elad H, Barkan R, Ben-Shachar S, Pasmanik Chor M, Dotan I. Increase in Processing Factors Is Involved in Skewed MicroRNA Expression in Patients with Ulcerative Colitis Who Develop Small Intestine Inflammation after Pouch Surgery. Inflamm Bowel Dis 2018; 24:1045-1054. [PMID: 29688474 DOI: 10.1093/ibd/izy034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND A large-scale increase in microRNA (miRNA) expression was observed in patients with ulcerative colitis who underwent pouch surgery and developed inflammation of the pouch (pouchitis). In this study, we assessed miRNA expression in these patients and investigated how regulation of its expression changes in the setting of pouchitis. METHODS Autologous samples that included mucosal biopsies, peripheral blood cells, and plasma were collected from the patients. Candidate primary and mature miRNA expressions were analyzed by quantitative polymerase chain reaction. A human intestinal epithelial cell line was used to test DICER activity, and the expression of key miRNA processing factors was analyzed by Western blot. miRNA-424 and its potential target serotonin reuptake transporter (SERT) expressions were examined by quantitative reverse transcription polymerase chain reaction and Western blot in human pouch tissues and in a human intestinal epithelial cell line stimulated with inflammatory cytokines TNF-α, IL-1β, and INF-γ. RESULTS Candidate miRNA expression and protein expression of DICER-1, EXPORTIN-5, and AGO-2 were increased in association with pouch inflammation. Similarly, inflammatory cytokines increased protein expression of DICER-1, EXPORTIN-5, and AGO-2 and DICER activity in the epithelial cell line. The miRNA-424 expression increased whereas SERT expression decreased in the patients' mucosa. Similarly, incubation of the epithelial cell line with inflammatory cytokines resulted in increased miRNA-424 and decreased SERT mRNA and protein expression. CONCLUSIONS The miRNA expression and processing are augmented in the inflamed intestinal mucosa of patients with pouchitis. These alterations are accompanied by increased expression of proteins involved in miRNA processing, suggesting that pouch inflammation contributes to miRNA processing and expression.
Collapse
Affiliation(s)
- Hadas Sherman Horev
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren M Rabinowitz
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hofit Elad
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Revital Barkan
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shay Ben-Shachar
- Genetic Institute, Tel Aviv Medical Center, Tel Aviv, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Iris Dotan
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
21
|
Naderi A. SRARP and HSPB7 are epigenetically regulated gene pairs that function as tumor suppressors and predict clinical outcome in malignancies. Mol Oncol 2018; 12:724-755. [PMID: 29577611 PMCID: PMC5928383 DOI: 10.1002/1878-0261.12195] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/27/2018] [Accepted: 03/10/2018] [Indexed: 12/16/2022] Open
Abstract
Deletions of chromosome 1p36 are common in cancers; however, despite extensive studies, there has been limited success for discovering candidate tumor suppressors in this region. SRARP has recently been identified as a novel corepressor of the androgen receptor (AR) and is located on chromosome 1p36. Here, bioinformatics analysis of large tumor datasets was performed to study SRARP and its gene pair, HSPB7. In addition, using cancer cell lines, mechanisms of SRARP and HSPB7 regulation and their molecular functions were investigated. This study demonstrated that SRARP and HSPB7 are a gene pair located 5.2 kb apart on 1p36.13 and are inactivated by deletions and epigenetic silencing in malignancies. Importantly, SRARP and HSPB7 have tumor suppressor functions in clonogenicity and cell viability associated with the downregulation of Akt and ERK. SRARP expression is inversely correlated with genes that promote cell proliferation and signal transduction, which supports its functions as a tumor suppressor. In addition, AR exerts dual regulatory effects on SRARP, and although an increased AR activity suppresses SRARP transcription, a minimum level of AR activity is required to maintain baseline SRARP expression in AR+ cancer cells. Furthermore, as observed with SRARP, HSPB7 interacts with the 14-3-3 protein, presenting a shared molecular feature between SRARP and HSPB7. Of note, genome- and epigenome-wide associations of SRARP and HSPB7 with survival strongly support their tumor suppressor functions. In particular, DNA hypermethylation, lower expression, somatic mutations, and lower copy numbers of SRARP are associated with worse cancer outcome. Moreover, DNA hypermethylation and lower expression of SRARP in normal adjacent tissues predict poor survival, suggesting that SRARP inactivation is an early event in carcinogenesis. In summary, SRARP and HSPB7 are tumor suppressors that are commonly inactivated in malignancies. SRARP inactivation is an early event in carcinogenesis that is strongly associated with worse survival, presenting potential translational applications.
Collapse
Affiliation(s)
- Ali Naderi
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| |
Collapse
|
22
|
El Aidy S, Ramsteijn AS, Dini-Andreote F, van Eijk R, Houwing DJ, Salles JF, Olivier JDA. Serotonin Transporter Genotype Modulates the Gut Microbiota Composition in Young Rats, an Effect Augmented by Early Life Stress. Front Cell Neurosci 2017; 11:222. [PMID: 28824378 PMCID: PMC5540888 DOI: 10.3389/fncel.2017.00222] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/13/2017] [Indexed: 02/01/2023] Open
Abstract
The neurotransmitter serotonin (5-HT) plays a vital regulatory role in both the brain and gut. 5-HT is crucial for regulating mood in the brain as well as gastrointestinal motility and secretion peripherally. Alterations in 5-HT transmission have been linked to pathological symptoms in both intestinal and psychiatric disorders and selective 5-HT transporter (5-HTT) inhibitors, affecting the 5-HT system by blocking the 5-HT transporter (5-HTT) have been successfully used to treat CNS- and intestinal disorders. Humans that carry the short allele of the 5-HTT-linked polymorphic region (5-HTTLPR) are more vulnerable to adverse environmental stressors, in particular early life stress. Although, early life stress has been shown to alter the composition of the gut microbiota, it is not known whether a lower 5-HTT expression is also associated with an altered microbiome composition. To investigate this, male and female wild type (5-HTT+/+), heterozygous (5-HTT+/-), and knockout (5-HTT-/-) 5-HT transporter rats were maternally separated for 6 h a day from postnatal day 2 till 15. On postnatal day 21, fecal samples were collected and the impact of 5-HTT genotype and maternal separation (MS) on the microbiome was analyzed using high-throughput sequencing of the bacterial 16S rRNA gene. MS showed a shift in the ratio between the two main bacterial phyla characterized by a decrease in Bacteroidetes and an increase in Firmicutes. Interestingly, the 5-HTT genotype caused a greater microbal dysbiosis (microbial imbalance) compared with MS. A significant difference in microbiota composition was found segregating 5-HTT-/- apart from 5-HTT+/- and 5-HTT+/+ rats. Moreover, exposure of rats with 5-HTT diminished expression to MS swayed the balance of their microbiota away from homeostasis to 'inflammatory' type microbiota characterized by higher abundance of members of the gut microbiome including Desulfovibrio, Mucispirillum, and Fusobacterium, all of which are previously reported to be associated with a state of intestinal inflammation, including inflammation associated with MS and brain disorders like multiple depressive disorders. Overall, our data show for the first time that altered expression of 5-HTT induces disruptions in male and female rat gut microbes and these 5-HTT genotype-related disruptions are augmented when combined with early life stress.
Collapse
Affiliation(s)
- Sahar El Aidy
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of GroningenGroningen, Netherlands
| | - Anouschka S Ramsteijn
- Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of GroningenGroningen, Netherlands
| | - Francisco Dini-Andreote
- Microbial Ecology Cluster, Genomics Research in Ecology and Evolution in Nature, Groningen Institute for Evolutionary Life Sciences, University of GroningenGroningen, Netherlands
| | - Roel van Eijk
- Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of GroningenGroningen, Netherlands
| | - Danielle J Houwing
- Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of GroningenGroningen, Netherlands
| | - Joana F Salles
- Microbial Ecology Cluster, Genomics Research in Ecology and Evolution in Nature, Groningen Institute for Evolutionary Life Sciences, University of GroningenGroningen, Netherlands
| | - Jocelien D A Olivier
- Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of GroningenGroningen, Netherlands
| |
Collapse
|
23
|
Iurescia S, Seripa D, Rinaldi M. Looking Beyond the 5-HTTLPR Polymorphism: Genetic and Epigenetic Layers of Regulation Affecting the Serotonin Transporter Gene Expression. Mol Neurobiol 2016; 54:8386-8403. [DOI: 10.1007/s12035-016-0304-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 11/16/2016] [Indexed: 01/01/2023]
|
24
|
Interplay between gut microbiota, its metabolites and human metabolism: Dissecting cause from consequence. Trends Food Sci Technol 2016. [DOI: 10.1016/j.tifs.2016.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Jin DC, Cao HL, Xu MQ, Wang SN, Wang YM, Yan F, Wang BM. Regulation of the serotonin transporter in the pathogenesis of irritable bowel syndrome. World J Gastroenterol 2016; 22:8137-8148. [PMID: 27688655 PMCID: PMC5037082 DOI: 10.3748/wjg.v22.i36.8137] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/28/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Serotonin (5-HT) and the serotonin transporter (SERT) have earned a tremendous amount of attention regarding the pathogenesis of irritable bowel syndrome (IBS). Considering that enteric 5-HT is responsible for the secretion, motility and perception of the bowel, the involvement of altered enteric 5-HT metabolism in the pathogenesis of IBS has been elucidated. Higher 5-HT availability is commonly associated with depressed SERT mRNA in patients with IBS compared with healthy controls. The expression difference of SERT between IBS patients and healthy controls might suggest that SERT plays an essential role in IBS pathogenesis, and SERT was expected to be a novel therapeutic target for IBS. Progress in this area has begun to illuminate the complex regulatory mechanisms of SERT in the etiology of IBS. In this article, current insights regarding the regulation of SERT in IBS are provided, including aspects of SERT gene polymorphisms, microRNAs, immunity and inflammation, gut microbiota, growth factors, among others. Potential SERT-directed therapies for IBS are also described. The potential regulators of SERT are of clinical importance and are important for better understanding IBS pathophysiology and therapeutic strategies.
Collapse
|
26
|
Tada Y, Ishihara S, Kawashima K, Fukuba N, Sonoyama H, Kusunoki R, Oka A, Mishima Y, Oshima N, Moriyama I, Yuki T, Ishikawa N, Araki A, Harada Y, Maruyama R, Kinoshita Y. Downregulation of serotonin reuptake transporter gene expression in healing colonic mucosa in presence of remaining low-grade inflammation in ulcerative colitis. J Gastroenterol Hepatol 2016; 31:1443-52. [PMID: 26676714 DOI: 10.1111/jgh.13268] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/19/2015] [Accepted: 12/05/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM The serotonin reuptake transporter (SERT) terminates serotonin activity by removing it from interstitial space. Downregulated colonic SERT expression has been reported in irritable bowel disease (IBS), and symptoms resembling IBS occur in cases of inflammatory bowel disease (IBD) in remission; thus, a common pathogenesis for IBS and IBD is possible. However, little is known regarding SERT expression in colonic mucosa of IBD patients during healing. METHODS Twenty-two ulcerative colitis (UC) patients underwent colonoscopy examinations, during which inflamed mucosa was distinguished from that undergoing healing. Healing mucosa was classified into regular and irregular vessel patterns by narrowband imaging magnifying colonoscopy. Expressions of SERT and various inflammation-related genes in biopsy samples were assessed using a polymerase chain reaction array system and real-time polymerase chain reaction. Colitis model mice were established by administration of dextran sodium sulfate or transfer of CD4(+) T cells isolated from SAMP1 mice, then time-course changes of SERT and inflammatory gene expressions were observed in colonic mucosa. RESULTS In UC patients, SERT expression in inflamed mucosa was significantly lower than in healing mucosa. SERT expression was decreased in healing mucosa with an irregular vessel pattern with mildly increased levels of inflammatory gene expression. In mice, SERT expression was suppressed in inflamed mucosa and continuously observed with low-grade mucosal inflammation during colitis healing. CONCLUSIONS Sserotonin reuptake transporter expression is downregulated in healing colonic mucosa of UC patients and that suppression may be dependent on the presence of remaining low-grade colonic inflammation.
Collapse
Affiliation(s)
- Yasumasa Tada
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Shunji Ishihara
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Kousaku Kawashima
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Nobuhiko Fukuba
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Hiroki Sonoyama
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Ryusaku Kusunoki
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Akihiko Oka
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Yoshiyuki Mishima
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Naoki Oshima
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Ichiro Moriyama
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan.,Cancer Center, Shimane University Hospital, Izumo, Japan
| | - Takafumi Yuki
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| | - Noriyoshi Ishikawa
- Department of Organ Pathology, Shimane University Faculty of Medicine, Matsue, Japan
| | - Asuka Araki
- Department of Organ Pathology, Shimane University Faculty of Medicine, Matsue, Japan
| | - Yuji Harada
- Department of Pathology Laboratory, Shimane University Hospital, Izumo, Japan
| | - Riruke Maruyama
- Department of Organ Pathology, Shimane University Faculty of Medicine, Matsue, Japan
| | - Yoshikazu Kinoshita
- Department of Internal Medicine II, Shimane University Faculty of Medicine, Matsue, Japan
| |
Collapse
|
27
|
Priyamvada S, Anbazhagan AN, Kumar A, Soni V, Alrefai WA, Gill RK, Dudeja PK, Saksena S. Lactobacillus acidophilus stimulates intestinal P-glycoprotein expression via a c-Fos/c-Jun-dependent mechanism in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2016; 310:G599-608. [PMID: 26867563 PMCID: PMC4836133 DOI: 10.1152/ajpgi.00210.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 02/05/2016] [Indexed: 02/08/2023]
Abstract
Our previous studies showed that Lactobacillus acidophilus (LA) culture supernatant (CS) increased P-glycoprotein [Pgp/multidrug resistance 1 (MDR1)] function, expression, and promoter activity in Caco-2 cells. The current studies were designed to elucidate the molecular mechanisms mediating the stimulatory effects of LA CS on Pgp promoter activity. Deletion analysis indicated that the LA CS response element(s) is located in the -172/+428-bp region, and sequence analysis of this region revealed three potential binding sites for c-Fos or c-Jun: proximal activating protein (AP) 1a (-119/-98 bp), distal AP1b (-99/-78 bp), and AP1c (+175/+196 bp). LA CS (24 h) showed an approximately twofold increase in the protein expression of c-Fos and c-Jun in Caco-2 cells. Electrophoretic mobility shift assay showed that LA CS markedly increased the binding of Caco-2 nuclear proteins to AP1a and AP1b, but not AP1c. The DNA-protein complex was completely eliminated by c-Fos antibody, while c-Jun antibody partially eliminated the complex. Chromatin immunoprecipitation analysis also showed that LA CS enhanced the association of c-Fos and c-Jun (by ∼4- and 1.5-fold, respectively) with endogenous Pgp promoter in Caco-2 cells (p-172/+1). Interestingly, overexpression of c-Fos or c-Jun activated Pgp promoter by nearly twofold each. This increase was further enhanced (∼14-fold) when c-Fos and c-Jun were simultaneously overexpressed, suggesting that the presence of one of these transcription factors potentiates the effect of the other. These studies, for the first time, provide evidence for the involvement of c-Fos/c-Jun in stimulation of Pgp gene expression by LA CS in the human intestine.
Collapse
Affiliation(s)
- Shubha Priyamvada
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Arivarasu N. Anbazhagan
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Anoop Kumar
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Vikas Soni
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Waddah A. Alrefai
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and ,2Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Ravinder K. Gill
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Pradeep K. Dudeja
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and ,2Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
28
|
Bailey C, Ruaux C, Stang BV, Valentine BA. Expression of serotonin, chromogranin-A, serotonin receptor-2B, tryptophan hydroxylase-1, and serotonin reuptake transporter in the intestine of dogs with chronic enteropathy. J Vet Diagn Invest 2016; 28:271-8. [PMID: 27026108 DOI: 10.1177/1040638715618232] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Serotonin regulates many intestinal motor and sensory functions. Altered serotonergic metabolism has been described in human gastrointestinal diseases. The objective of our study was to compare expression of several components of the serotonergic system [serotonin (5-HT), serotonin reuptake transporter protein (SERT), tryptophan hydroxylase-1 (TPH-1), 5-HT receptor2B (5-HT2B)] and the enterochromaffin cell marker chromogranin-A (CgA) in the intestinal mucosa between dogs with chronic enteropathy and healthy controls. Serotonin and CgA expression were determined by immunohistochemistry using banked and prospectively obtained, paraffin-embedded canine gastrointestinal biopsies (n = 11), and compared to a control group of canine small intestinal sections (n = 10). Expression of SERT, TPH-1, and 5-HT2B were determined via real-time reverse transcription (qRT)-PCR using prospectively collected endoscopic duodenal biopsies (n = 10) and compared to an additional control group of control duodenal biopsies (n = 8, control group 2) showing no evidence of intestinal inflammation. Dogs with chronic enteropathies showed strong staining for both 5-HT and CgA. Mean positive cells per high power field (HPF) were significantly increased for both compounds in dogs with chronic enteropathies (p < 0.001 for 5-HT; p < 0.05 for CgA). The number of 5-HT-positive and CgA-positive cells/HPF showed significant correlation in the entire group of dogs, including both diseased and healthy individuals (Pearson r(2) = 0.2433, p = 0.016). No significant differences were observed for SERT, TPH-1, or 5-HT2B expression; however, dogs with chronic enteropathy showed greater variability in expression of TPH-1 and 5-HT2B We conclude that components of the neuroendocrine system show altered expression in the intestinal mucosa of dogs with chronic enteropathy. These changes may contribute to nociception and clinical signs in these patients.
Collapse
Affiliation(s)
- Candice Bailey
- Departments of Clinical Sciences (Bailey, Ruaux, Stang), School of Veterinary Medicine, Oregon State University, Corvallis, ORBiomedical Sciences (Valentine), School of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Craig Ruaux
- Departments of Clinical Sciences (Bailey, Ruaux, Stang), School of Veterinary Medicine, Oregon State University, Corvallis, ORBiomedical Sciences (Valentine), School of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Bernadette V Stang
- Departments of Clinical Sciences (Bailey, Ruaux, Stang), School of Veterinary Medicine, Oregon State University, Corvallis, ORBiomedical Sciences (Valentine), School of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Beth A Valentine
- Departments of Clinical Sciences (Bailey, Ruaux, Stang), School of Veterinary Medicine, Oregon State University, Corvallis, ORBiomedical Sciences (Valentine), School of Veterinary Medicine, Oregon State University, Corvallis, OR
| |
Collapse
|
29
|
Chang WH, Lee LT, Lee IH, Chi MH, Chen KC, Chiu NT, Yao WJ, Chen PS, Yang YK. Which demographic variables are necessary to correct in neuroimaging studies of serotonin transporter availability? A SPECT study with [¹²³I]ADAM. Psychiatry Res 2015; 231:320-4. [PMID: 25670644 DOI: 10.1016/j.pscychresns.2015.01.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/07/2014] [Accepted: 01/14/2015] [Indexed: 11/27/2022]
Abstract
Previous studies have shown that many demographic variables influence serotonin transporter (SERT) availability as assessed by single photon emission computed tomography (SPECT). The aim of this study was to explore which demographic variables influenced the SERT availability most in a SPECT study with [(123)I]ADAM. Ninety-five healthy volunteers were recruited. Age, sex, smoking, alcohol intake, educational level, body mass index, seasonal change, and SERT availability were recorded and then analyzed by multivariate linear regression. Age was the only variable that was significantly associated with SERT availability (calculate: (midbrain-cerebellum)/cerebellum). Furthermore, the inverse correlation of age and SERT availability may be present only before the age of 47. Age should be a covariate in SERT-related neuroimaging analyses, particularly in participants under the age of 47 years.
Collapse
Affiliation(s)
- Wei Hung Chang
- Department of Psychiatry, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin, Taiwan; Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Lan-Ting Lee
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I Hui Lee
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Addiction Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Mei Hung Chi
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kao Chin Chen
- Department of Psychiatry, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin, Taiwan; Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Addiction Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Nan Tsing Chiu
- Department of Nuclear Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei Jen Yao
- Department of Nuclear Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Addiction Research Center, National Cheng Kung University, Tainan, Taiwan
| | - Yen Kuang Yang
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Addiction Research Center, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
30
|
Hartstra AV, Bouter KEC, Bäckhed F, Nieuwdorp M. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care 2015; 38:159-65. [PMID: 25538312 DOI: 10.2337/dc14-0769] [Citation(s) in RCA: 431] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The worldwide prevalence of obesity and type 2 diabetes mellitus (T2DM) continues to rise at an alarming pace. Recently the potential role of the gut microbiome in these metabolic disorders has been identified. Obesity is associated with changes in the composition of the intestinal microbiota, and the obese microbiome seems to be more efficient in harvesting energy from the diet. Lean male donor fecal microbiota transplantation (FMT) in males with metabolic syndrome resulted in a significant improvement in insulin sensitivity in conjunction with an increased intestinal microbial diversity, including a distinct increase in butyrate-producing bacterial strains. Such differences in gut microbiota composition might function as early diagnostic markers for the development of T2DM in high-risk patients. Products of intestinal microbes such as butyrate may induce beneficial metabolic effects through enhancement of mitochondrial activity, prevention of metabolic endotoxemia, and activation of intestinal gluconeogenesis via different routes of gene expression and hormone regulation. Future research should focus on whether bacterial products (like butyrate) have the same effects as the intestinal bacteria that produce it, in order to ultimately pave the way for more successful interventions for obesity and T2DM. The rapid development of the currently available techniques, including use of fecal transplantations, has already shown promising results, so there is hope for novel therapies based on the microbiota in the future.
Collapse
Affiliation(s)
- Annick V Hartstra
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Kristien E C Bouter
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Göteborg, Göteborg, Sweden Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Max Nieuwdorp
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands Wallenberg Laboratory, Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Göteborg, Göteborg, Sweden
| |
Collapse
|
31
|
Grinkevich LN, Vorobiova OV. Role of modulatory mediator serotonin in induction of epigenetic processes during long-term memory formation in Helix. ACTA ACUST UNITED AC 2014. [DOI: 10.1134/s2079059714060094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Takahashi K. Influence of bacteria on epigenetic gene control. Cell Mol Life Sci 2014; 71:1045-54. [PMID: 24132510 PMCID: PMC11113846 DOI: 10.1007/s00018-013-1487-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 01/26/2023]
Abstract
Cellular information is inherited by daughter cells through epigenetic routes in addition to genetic routes. Epigenetics, which is primarily mediated by inheritable DNA methylation and histone post-translational modifications, involves changes in the chromatin structure important for regulating gene expression. It is widely known that epigenetic control of gene expression plays an essential role in cell differentiation processes in vertebrates. Furthermore, because epigenetic changes can occur reversibly depending on environmental factors in differentiated cells, they have recently attracted considerable attention as targets for disease prevention and treatment. These environmental factors include diet, exposure to bacteria or viruses, and air pollution, of which this review focuses on the influence of bacteria on epigenetic gene control in a host. Host-bacterial interactions not only occur upon pathogenic bacterial infection but also continuously exist between commensal bacteria and the host. These bacterial stimuli play an essential role in various biological responses involving external stimuli and in maintaining physiological homeostasis by altering epigenetic markers and machinery.
Collapse
Affiliation(s)
- Kyoko Takahashi
- Food and Physiological Functions Laboratory, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa-shi, Kanagawa, 252-0880, Japan,
| |
Collapse
|