1
|
Lu X, Liu Q, Deng Y, Wu J, Mu X, Yang X, Zhang T, Luo C, Li Z, Tang S, Hu Y, Du Q, Xu J, Xie R. Research progress on the roles of dopamine and dopamine receptors in digestive system diseases. J Cell Mol Med 2024; 28:e18154. [PMID: 38494840 PMCID: PMC10945074 DOI: 10.1111/jcmm.18154] [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: 11/15/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 03/19/2024] Open
Abstract
Dopamine (DA) is a neurotransmitter synthesized in the human body that acts on multiple organs throughout the body, reaching them through the blood circulation. Neurotransmitters are special molecules that act as messengers by binding to receptors at chemical synapses between neurons. As ligands, they mainly bind to corresponding receptors on central or peripheral tissue cells. Signalling through chemical synapses is involved in regulating the activities of various body systems. Lack of DA or a decrease in DA levels in the brain can lead to serious diseases such as Parkinson's disease, schizophrenia, addiction and attention deficit disorder. It is widely recognized that DA is closely related to neurological diseases. As research on the roles of brain-gut peptides in human physiology and pathology has deepened in recent years, the regulatory role of neurotransmitters in digestive system diseases has gradually attracted researchers' attention, and research on DA has expanded to the field of digestive system diseases. This review mainly elaborates on the research progress on the roles of DA and DRs related to digestive system diseases. Starting from the biochemical and pharmacological properties of DA and DRs, it discusses the therapeutic value of DA- and DR-related drugs for digestive system diseases.
Collapse
Affiliation(s)
- Xianmin Lu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Qi Liu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Ya Deng
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Jiangbo Wu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Xingyi Mu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Xiaoxu Yang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Ting Zhang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Chen Luo
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Zhuo Li
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Siqi Tang
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Yanxia Hu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Qian Du
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Jingyu Xu
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Rui Xie
- Department of Gastroenterology, Digestive Disease HospitalAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative InnovAffiliated Hospital of Zunyi Medical Universityation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
2
|
Gupta S, Dinesh S, Sharma S. Bridging the Mind and Gut: Uncovering the Intricacies of Neurotransmitters, Neuropeptides, and their Influence on Neuropsychiatric Disorders. Cent Nerv Syst Agents Med Chem 2024; 24:2-21. [PMID: 38265387 DOI: 10.2174/0118715249271548231115071021] [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: 07/11/2023] [Revised: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND The gut-brain axis (GBA) is a bidirectional signaling channel that facilitates communication between the gastrointestinal tract and the brain. Recent research on the gut-brain axis demonstrates that this connection enables the brain to influence gut function, which in turn influences the brain and its cognitive functioning. It is well established that malfunctioning of this axis adversely affects both systems' ability to operate effectively. OBJECTIVE Dysfunctions in the GBA have been associated with disorders of gut motility and permeability, intestinal inflammation, indigestion, constipation, diarrhea, IBS, and IBD, as well as neuropsychiatric and neurodegenerative disorders like depression, anxiety, schizophrenia, autism, Alzheimer's, and Parkinson's disease. Multiple research initiatives have shown that the gut microbiota, in particular, plays a crucial role in the GBA by participating in the regulation of a number of key neurochemicals that are known to have significant effects on the mental and physical well-being of an individual. METHODS Several studies have investigated the relationship between neuropsychiatric disorders and imbalances or disturbances in the metabolism of neurochemicals, often leading to concomitant gastrointestinal issues and modifications in gut flora composition. The interaction between neurological diseases and gut microbiota has been a focal point within this research. The novel therapeutic interventions in neuropsychiatric conditions involving interventions such as probiotics, prebiotics, and dietary modifications are outlined in this review. RESULTS The findings of multiple studies carried out on mice show that modulating and monitoring gut microbiota can help treat symptoms of such diseases, which raises the possibility of the use of probiotics, prebiotics, and even dietary changes as part of a new treatment strategy for neuropsychiatric disorders and their symptoms. CONCLUSION The bidirectional communication between the gut and the brain through the gut-brain axis has revealed profound implications for both gastrointestinal and neurological health. Malfunctions in this axis have been connected to a range of disorders affecting gut function as well as cognitive and neuropsychiatric well-being. The emerging understanding of the role of gut microbiota in regulating key neurochemicals opens up possibilities for novel treatment approaches for conditions like depression, anxiety, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Saumya Gupta
- Department of Bioinformatics, BioNome, Bengaluru, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru, India
| |
Collapse
|
3
|
Basiji K, Sendani AA, Ghavami SB, Farmani M, Kazemifard N, Sadeghi A, Lotfali E, Aghdaei HA. The critical role of gut-brain axis microbiome in mental disorders. Metab Brain Dis 2023; 38:2547-2561. [PMID: 37436588 DOI: 10.1007/s11011-023-01248-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/30/2023] [Indexed: 07/13/2023]
Abstract
The Gut-brain axis is a bidirectional neural and humoral signaling that plays an important role in mental disorders and intestinal health and connects them as well. Over the past decades, the gut microbiota has been explored as an important part of the gastrointestinal tract that plays a crucial role in the regulation of most functions of various human organs. The evidence shows several mediators such as short-chain fatty acids, peptides, and neurotransmitters that are produced by the gut may affect the brain's function directly or indirectly. Thus, dysregulation in this microbiome community can give rise to several diseases such as Parkinson's disease, depression, irritable bowel syndrome, and Alzheimer's disease. So, the interactions between the gut and the brain are significantly considered, and also it provides a prominent subject to investigate the causes of some diseases. In this article, we reviewed and focused on the role of the largest and most repetitive bacterial community and their relevance with some diseases that they have mentioned previously.
Collapse
Affiliation(s)
- Kimia Basiji
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Aghamohammadi Sendani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nesa Kazemifard
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Lotfali
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Reyes-Martínez S, Segura-Real L, Gómez-García AP, Tesoro-Cruz E, Constantino-Jonapa LA, Amedei A, Aguirre-García MM. Neuroinflammation, Microbiota-Gut-Brain Axis, and Depression: The Vicious Circle. J Integr Neurosci 2023; 22:65. [PMID: 37258450 DOI: 10.31083/j.jin2203065] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 06/02/2023] Open
Abstract
Depression is the leading cause of disability worldwide, contributing to the global disease burden. From above, it is a priority to investigate models that fully explain its physiopathology to develop new treatments. In the last decade, many studies have shown that gut microbiota (GM) dysbiosis influences brain functions and participate, in association with immunity, in the pathogenesis of depression. Thereby, GM modulation could be a novel therapeutic target for depression. This review aims to evidence how the GM and the immune system influence mental illness, particularly depression. Here, we focus on the communication mechanisms between the intestine and the brain and the impact on the development of neuroinflammation contributing to the development of Major Depressive Disorder (MDD). However, most of the current findings are in animal models, suggesting the need for studies in humans. In addition, more analysis of metabolites and cytokines are needed to identify new pathophysiological mechanisms improving anti-depression treatments.
Collapse
Affiliation(s)
- Sandy Reyes-Martínez
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Ciudad de México, Mexico
| | - Lorena Segura-Real
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Ciudad de México, Mexico
| | - Ana Pamela Gómez-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Ciudad de México, Mexico
| | - Emiliano Tesoro-Cruz
- Unidad de Investigación Biomédica en Infectología e Inmunología, Hospital de Infectología, Centro Médico Nacional "La Raza", IMSS, Col. La Raza, 02990 Ciudad de México, Mexico
| | - Luis A Constantino-Jonapa
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Ciudad de México, Mexico
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Interdisciplinary Internal Medicine Unit, Careggi University Hospital, 50134 Florence, Italy
| | - María M Aguirre-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Ciudad de México, Mexico
| |
Collapse
|
5
|
Begum N, Mandhare A, Tryphena KP, Srivastava S, Shaikh MF, Singh SB, Khatri DK. Epigenetics in depression and gut-brain axis: A molecular crosstalk. Front Aging Neurosci 2022; 14:1048333. [PMID: 36583185 PMCID: PMC9794020 DOI: 10.3389/fnagi.2022.1048333] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Gut-brain axis is a dynamic, complex, and bidirectional communication network between the gut and brain. Changes in the microbiota-gut-brain axis are responsible for developing various metabolic, neurodegenerative, and neuropsychiatric disorders. According to clinical and preclinical findings, the gut microbiota is a significant regulator of the gut-brain axis. In addition to interacting with intestinal cells and the enteric nervous system, it has been discovered that microbes in the gut can modify the central nervous system through metabolic and neuroendocrine pathways. The metabolites of the gut microbiome can modulate a number of diseases by inducing epigenetic alteration through DNA methylation, histone modification, and non-coding RNA-associated gene silencing. Short-chain fatty acids, especially butyrate, are well-known histone deacetylases inhibitors. Similarly, other microbial metabolites such as folate, choline, and trimethylamine-N-oxide also regulate epigenetics mechanisms. Furthermore, various studies have revealed the potential role of microbiome dysbiosis and epigenetics in the pathophysiology of depression. Hence, in this review, we have highlighted the role of gut dysbiosis in epigenetic regulation, causal interaction between host epigenetic modification and the gut microbiome in depression and suggest microbiome and epigenome as a possible target for diagnosis, prevention, and treatment of depression.
Collapse
Affiliation(s)
- Nusrat Begum
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Aniket Mandhare
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Kamatham Pushpa Tryphena
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India,*Correspondence: Saurabh Srivastava,
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia,Mohd Farooq Shaikh,
| | - Shashi Bala Singh
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Cellular and Molecular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India,Dharmendra Kumar Khatri,
| |
Collapse
|
6
|
Diviccaro S, Giatti S, Cioffi L, Falvo E, Herian M, Caruso D, Melcangi RC. Gut Inflammation Induced by Finasteride Withdrawal: Therapeutic Effect of Allopregnanolone in Adult Male Rats. Biomolecules 2022; 12:1567. [PMID: 36358917 PMCID: PMC9687671 DOI: 10.3390/biom12111567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 07/29/2023] Open
Abstract
The treatment with finasteride (i.e., an inhibitor of 5α-reductase) may be associated with different side effects (i.e., depression, anxiety, cognitive impairment and sexual dysfunction) inducing the so-called post finasteride syndrome (PFS). Moreover, previous observations in PFS patients and an experimental model showed alterations in gut microbiota populations, suggesting an inflammatory environment. To confirm this hypothesis, we have explored the effect of chronic treatment with finasteride (i.e., for 20 days) and its withdrawal (i.e., for 1 month) on the levels of steroids, neurotransmitters, pro-inflammatory cytokines and gut permeability markers in the colon of adult male rat. The obtained data demonstrate that the levels of allopregnanolone (ALLO) decreased after finasteride treatment and after its withdrawal. Following the drug suspension, the decrease in ALLO levels correlates with an increase in IL-1β and TNF-α, serotonin and a decrease in dopamine. Importantly, ALLO treatment is able to counteract some of these alterations. The relation between ALLO and GABA-A receptors and/or pregnenolone (ALLO precursor) could be crucial in their mode of action. These observations provide an important background to explore further the protective effect of ALLO in the PFS experimental model and the possibility of its translation into clinical therapy.
Collapse
|
7
|
Chen Y, Xu J, Chen Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021; 13:nu13062099. [PMID: 34205336 PMCID: PMC8234057 DOI: 10.3390/nu13062099] [Citation(s) in RCA: 384] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence indicates that gut microbiota is important in the regulation of brain activity and cognitive functions. Microbes mediate communication among the metabolic, peripheral immune, and central nervous systems via the microbiota–gut–brain axis. However, it is not well understood how the gut microbiome and neurons in the brain mutually interact or how these interactions affect normal brain functioning and cognition. We summarize the mechanisms whereby the gut microbiota regulate the production, transportation, and functioning of neurotransmitters. We also discuss how microbiome dysbiosis affects cognitive function, especially in neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease.
Collapse
Affiliation(s)
- Yijing Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Y.C.); (J.X.)
| | - Jinying Xu
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Y.C.); (J.X.)
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Y.C.); (J.X.)
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing 100049, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen 518057, China
- Correspondence: ; Tel.: +86-755-26925498
| |
Collapse
|
8
|
Feng XY, Xue H, Guo ZH, Yan JT, Liu S, Zhu JX. Dopamine and Gastrointestinal Mucosa Function. DOPAMINE IN THE GUT 2021:87-131. [DOI: 10.1007/978-981-33-6586-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
9
|
Feng X, Yan J, Li G, Liu J, Fan R, Li S, Zheng L, Zhang Y, Zhu J. Source of dopamine in gastric juice and luminal dopamine-induced duodenal bicarbonate secretion via apical dopamine D 2 receptors. Br J Pharmacol 2020; 177:3258-3272. [PMID: 32154577 PMCID: PMC7312307 DOI: 10.1111/bph.15047] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/02/2020] [Accepted: 02/07/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Dopamine protects the duodenal mucosa. Here we have investigated the source of dopamine in gastric juice and the mechanism underlying the effects of luminal dopamine on duodenal bicarbonate secretion (DBS) in rodents. EXPERIMENTAL APPROACH Immunofluorescence, UPLC-MS/MS, gastric incubation and perfusion were used to detect gastric-derived dopamine. Immunofluorescence and RT-PCR were used to examine the expression of dopamine receptors in the duodenal mucosa. Real-time pH titration and pHi measurement were performed to investigate DBS. KEY RESULTS H+ -K+ -ATPase was co-localized with tyrosine hydroxylase and dopamine transporters in gastric parietal cells. Dopamine was increased in in vivo gastric perfusate after intravenous infusion of histamine and in gastric mucosa incubated, in vitro, with bethanechol chloride or tyrosine. D2 receptors were the most abundant dopamine receptors in rat duodenum, mainly distributed on the apical membrane of epithelial cells. Luminal dopamine increased DBS in a concentration-dependent manner, an effect mimicked by a D2 receptor agonist quinpirole and inhibited by the D2 receptor antagonist L741,626, in vivo D2 receptor siRNA and in D2 receptor -/- mice. Dopamine and quinpirole raised the duodenal enterocyte pHi . Quinpirole-evoked DBS and PI3K/Akt activity were inhibited by calcium chelator BAPTA-AM or in D2 receptor-/- mice. CONCLUSION AND IMPLICATIONS Dopamine in the gastric juice is derived from parietal cells and is secreted along with gastric acid. On arrival in the duodenal lumen, dopamine increased DBS via an apical D2 receptor- and calcium-dependent pathway. Our data provide novel insights into the protective effects of dopamine on the duodenal mucosa.
Collapse
Affiliation(s)
- Xiao‐Yan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Jing‐Ting Yan
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Guang‐Wen Li
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Jing‐Hua Liu
- Grade 2017 Clinical Medicine, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Rui‐Fang Fan
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Shi‐Chao Li
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Li‐Fei Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Yue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| | - Jin‐Xia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical ScienceCapital Medical UniversityBeijingChina
| |
Collapse
|
10
|
Li Y, Zhang Y, Zhang XL, Feng XY, Liu CZ, Zhang XN, Quan ZS, Yan JT, Zhu JX. Dopamine promotes colonic mucus secretion through dopamine D 5 receptor in rats. Am J Physiol Cell Physiol 2019; 316:C393-C403. [PMID: 30624983 DOI: 10.1152/ajpcell.00261.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Dopamine regulates gastrointestinal mucosal barrier. Mucus plays important roles in the protection of intestinal mucosa. Here, the regulatory effect of dopamine on rat colonic mucus secretion was investigated. RT-PCR, immunofluorescence, Periodic Acid-Schiff reagent assay, Alcian blue-Periodic Acid-Schiff staining, and enzyme-linked immunosorbent assay were used to observe the expression of dopamine receptor and the direct effect of dopamine on the colonic mucus. Mice injected intraperitoneally with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) destroying enteric dopamine (DA) neurons, rats microinjected with 6-hydroxydopamine (6-OHDA) into the bilateral substantia nigra damaging central dopaminergic neurons, and dopamine D5 receptor-downregulated transgenic mice were used to detect the effect of endogenous enteric dopamine or dopamine receptors on distal colonic mucus. Our results indicated that D5 immunoreactivity was widely distributed on the colonic goblet cells. Dopamine dose-dependently increased rat distal colonic mucus secretion in vitro. D1-like receptor antagonist SCH23390 inhibited dopamine (1 μΜ)-induced distal colonic mucus secretion. D1-like receptor agonist SKF38393 promoted mucin 2 (MUC2) secretion and increased the intracellular cAMP level of colonic mucosa. D5 receptor-downregulated transgenic mice showed a decreased colonic MUC2 content. MPTP-treated mice exhibited lower colonic dopamine content and decreased colonic mucus content. 6-OHDA rats had an increase in the dopamine content in colonic mucosa but decreases in the protein levels of D1 and D5 receptors and MUC2 content in the colonic mucosa. These findings reveal that dopamine is able to promote distal colonic mucus secretion through the D5 receptor, which provides important evidence to better understand the possible role of dopamine in the colonic mucosal barrier.
Collapse
Affiliation(s)
- Yun Li
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China.,Department of Immunology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Yue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Xiao-Li Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Xiao-Yan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Chen-Zhe Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Xiu-Neng Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Zhu-Sheng Quan
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Jing-Ting Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| | - Jin-Xia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University , Beijing , China
| |
Collapse
|
11
|
Interactions Between Bacteria and the Gut Mucosa: Do Enteric Neurotransmitters Acting on the Mucosal Epithelium Influence Intestinal Colonization or Infection? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:121-41. [DOI: 10.1007/978-3-319-20215-0_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Catecholamine-Directed Epithelial Cell Interactions with Bacteria in the Intestinal Mucosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:79-99. [DOI: 10.1007/978-3-319-20215-0_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
13
|
Feng XY, Li Y, Li LS, Li XF, Zheng LF, Zhang XL, Fan RF, Song J, Hong F, Zhang Y, Zhu JX. Dopamine D1 receptors mediate dopamine-induced duodenal epithelial ion transport in rats. Transl Res 2013; 161:486-94. [PMID: 23276732 DOI: 10.1016/j.trsl.2012.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/01/2012] [Accepted: 12/04/2012] [Indexed: 02/08/2023]
Abstract
Dopamine (DA) is synthesized in gastrointestinal epithelial cells and performs important regulatory effects on the duodenal mucosa. However, the underlying mechanism remains largely unknown. The present study investigated the effect of DA on the duodenal epithelial ion transport in rats by means of short-circuit current (ISC), real-time pH titration, enzyme-linked immunosorbent assay, and immunohistochemistry. The results indicate that basolateral, but not apical, application of DA induced a concentration-dependent ISC downward deflection with an apparent IC50 of 5.34 μmol/L. Basolateral application of dopaminergic receptor D1 (D1) antagonist, SCH-23390, inhibited DA-induced change in ISC (△ISC) in a dose-dependent manner. D1 agonist, SKF38393, mimicked the effect of DA on the ISC. The clear immunoreactivity of D1 subtype D5 (D1b) was at the both apical and basolatoral sides of Brunner's glands and intestinal crypts. Basolateral pretreatment with adenylate cyclase inhibitor, MDL12330A, significantly inhibited DA- and forskolin-induced △ISC. DA and SKF38393 increased the level of intracellular cyclic adenosine monophosphate (cAMP) from 1.55 ± 0.11 to 2.07 ± 0.11 and 5.91 ± 0.25 pmol/L·mg(-1), respectively. Furthermore, the serosal DA-induced △ISC was remarkably inhibited by apical administration of K(+) channel blockers, Ba(2+) and tetraethylammonium, but not by Cl(-) channel blockers. Serosal DA and D1 agonist did not affect duodenal HCO3(-) secretion. In conclusion, the present results demonstrate that serosal DA is able to promote rat duodenal epithelial K(+) secretion, not HCO3(-) secretion through D1-mediated and cAMP-dependent pathway. The study provides a new insight in the modulation of DA on the ion transport of duodenal epithelia in rats.
Collapse
Affiliation(s)
- Xiao-Yan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Deachapunya C, Poonyachoti S, Krishnamra N. Site-specific regulation of ion transport by prolactin in rat colon epithelium. Am J Physiol Gastrointest Liver Physiol 2012; 302:G1199-206. [PMID: 22403794 DOI: 10.1152/ajpgi.00143.2011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The effect of prolactin (PRL) on ion transport across the rat colon epithelium was investigated using Ussing chamber technique. PRL (1 μg/ml) induced a sustained decrease in short-circuit current (I(sc)) in the distal colon with an EC(50) value of 100 ng/ml and increased I(sc) in the proximal colon with an EC(50) value of 49 ng/ml. In the distal colon, the PRL-induced decrease in I(sc) was not affected by Na(+) channel blocker amiloride or Cl(-) channel blockers, NPPB, DPC, or DIDS, added mucosally. However, the response was inhibited by mucosal application of K(+) channel blockers glibenclamide, quinidine, and chromanol 293B, whereas other K(+) channel blockers, Ba(2+), tetraethylammonium, clotrimazole, and apamin, failed to have effects. The PRL-induced decrease in I(sc) was also inhibited by Na(+)-K(+)-2Cl(-) transporter inhibitor bumetanide, Ba(2+), and chromanol 293B applied serosally. In the transverse and proximal colon, the PRL-induced increase in I(sc) was suppressed by DPC, glibenclamide, and bumetanide, but not by NPPB, DIDS, or amiloride. The PRL-induced changes in I(sc) in both distal and proximal colon were abolished by JAK2 inhibitor AG490, but not BAPTA-AM, the Ca(2+) chelating agent, or phosphatidylinositol 3-kinase inhibitor wortmannin. These results suggest a segment-specific effect of PRL in rat colon, by activation of K(+) secretion in the distal colon and activation of Cl(-) secretion in the transverse and proximal colon. Both PRL actions are mediated by JAK-STAT-dependent pathway, but not phosphatidylinositol 3-kinase pathway or Ca(2+) mobilization. These findings suggest a role of PRL in the regulation of electrolyte transport in mammalian colon.
Collapse
Affiliation(s)
- Chatsri Deachapunya
- Department of Physiology, Faculty of Medicine, Srinakharinwirot University, Sukhumvit 23, Wattana, Bangkok 10110, Thailand.
| | | | | |
Collapse
|
15
|
Colonic potassium handling. Pflugers Arch 2010; 459:645-56. [PMID: 20143237 DOI: 10.1007/s00424-009-0781-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Revised: 12/23/2009] [Accepted: 12/25/2009] [Indexed: 10/19/2022]
Abstract
Homeostatic control of plasma K+ is a necessary physiological function. The daily dietary K+ intake of approximately 100 mmol is excreted predominantly by the distal tubules of the kidney. About 10% of the ingested K+ is excreted via the intestine. K+ handling in both organs is specifically regulated by hormones and adapts readily to changes in dietary K+ intake, aldosterone and multiple local paracrine agonists. In chronic renal insufficiency, colonic K+ secretion is greatly enhanced and becomes an important accessory K+ excretory pathway. During severe diarrheal diseases of different causes, intestinal K+ losses caused by activated ion secretion may become life threatening. This topical review provides an update of the molecular mechanisms and the regulation of mammalian colonic K+ absorption and secretion. It is motivated by recent results, which have identified the K+ secretory ion channel in the apical membrane of distal colonic enterocytes. The directed focus therefore covers the role of the apical Ca2+ and cAMP-activated BK channel (KCa1.1) as the apparently only secretory K+ channel in the distal colon.
Collapse
|
16
|
Morphine-induced physiological and behavioral responses in mice lacking G protein-coupled receptor kinase 6. Drug Alcohol Depend 2009; 104:187-96. [PMID: 19497686 PMCID: PMC2771341 DOI: 10.1016/j.drugalcdep.2009.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 04/23/2009] [Accepted: 04/27/2009] [Indexed: 11/23/2022]
Abstract
G protein-coupled receptor kinases (GRKs) are a family of intracellular proteins that desensitize and regulate the responsiveness of G protein-coupled receptors (GPCRs). In the present study, we assessed the contribution of GRK6 to the regulation and responsiveness of the G protein-coupled mu-opioid receptor (microOR) in response to morphine in vitro and in vivo using mice lacking GRK6. In cell culture, overexpression of GRK6 facilitates morphine-induced beta-arrestin2 (betaarrestin2) recruitment and receptor internalization, suggesting that this kinase may play a role in regulating the microOR. In vivo, we find that acute morphine treatment induces greater locomotor activation but less constipation in GRK6 knockout (GRK6-KO) mice compared to their wild-type (WT) littermates. The GRK6-KO mice also appear to be "presensitized" to the locomotor stimulating effects induced by chronic morphine treatment, yet these animals do not display more conditioned place preference than WT mice do. Furthermore, several other morphine-mediated responses which were evaluated, including thermal antinociception, analgesic tolerance, and physical dependence, were not affected by ablation of the GRK6 gene. Collectively, these results suggest that GRK6 may play a role in regulating some, but not all morphine-mediated responses. In addition, these findings underscore that the contribution of a particular regulatory factor to receptor function can differ based upon the specific cell composition and physiology assessed, and illustrate the need for using caution when interpreting the importance of interactions observed in cell culture.
Collapse
|
17
|
Du JY, Zuo WL, Ruan YC, Yang ZH, Chen MH, Chen SL, Li S, Wu ZL, Xiang H, Zhou WL. Stimulating Effects of Dopamine on Chloride Transport Across the Rat Caudal Epididymal Epithelium in Culture1. Biol Reprod 2009; 80:13-23. [DOI: 10.1095/biolreprod.108.068346] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
18
|
Zhang XH, Zhang XF, Zhang JQ, Tian YM, Xue H, Yang N, Zhu JX. β-Adrenoceptors, but not dopamine receptors, mediate dopamine-induced ion transport in late distal colon of rats. Cell Tissue Res 2008; 334:25-35. [DOI: 10.1007/s00441-008-0661-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 06/04/2008] [Indexed: 11/24/2022]
|
19
|
Zhang GH, Zhu JX, Xue H, Fan J, Chen X, Tsang LL, Chung YW, Xing Y, Chan HC. Dopamine stimulates Cl− absorption coupled with HCO3− secretion in rat late distal colon. Eur J Pharmacol 2007; 570:188-95. [PMID: 17597606 DOI: 10.1016/j.ejphar.2007.05.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 05/04/2007] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
Freshly isolated rat colonic mucosa close to anus (the late distal colonic mucosa) was used to investigate the effect of dopamine on the rat late distal colonic ion transport by means of short-circuit current (I(SC)) recording and reverse transcription PCR (RT-PCR) analysis. The results showed that the basolateral addition of dopamine (0.1-1000 micromol/l) produced a concentration-dependent downward deflection in I(SC) with an apparent EC(50) of 20.06 micromol/l in the late distal colon. The dopamine-induced I(SC) decrease was abolished by bilateral removal of Cl(-) or HCO(3)(-), apical Cl(-) replacement and apical pretreatment with non-specific Cl(-) channel blocker/transporter inhibitor, DPC (1 mmol/l) or glibenclamide (1 mmol/l), and reversed by subsequent addition of glibenclamide. Removal of basolateral Na(+) or reducing basolateral HCO(3)(-) (3 mmol/l) as well as basolateral pretreatment with DIDS (4,4'-didsothio- cyanostilbene-2, 2'-disulfonic acid) (250 micromol/l), an inhibitor of NBC or AE, could also inhibit the dopamine-induced I(SC) response. However, apical pretreatment with epithelial Na(+) channel blocker, amiloride (10 micromol/l), Ca(2+)-dependent Cl(-) channel blocker/anion exchanger, DIDS (100 micromol/l), or putative K(+) blockers such as Ba(2+) (5 mmol/l), TEA (tetraethylammonium) (5 mmol/l) or 293B (trans-6-cyano-4- (N-ethylsulfonyl-N-methylamino)-3-hydroxy-2,2-dimethyl-chromane) (10 micromol/l) did not significantly affect the dopamine-induced I(SC) response. RT-PCR results showed the expression of anion exchanger, SLC26A3, but not SLC26A6, in rat late distal colon. In conclusion, the present results suggest that dopamine may promote rat late distal colonic epithelial Cl(-) absorption coupled with HCO(3)(-) secretion, which may be mediated by apical electrogenic anion exchanger, SLC26A3, and require basolateral entry of HCO(3)(-) through Na(+)-HCO(3)(-) cotransporter. The present findings reveal a previously unreported dopamine-regulated anion transport process in rat late distal colon, which may have implication in Parkinson's disease.
Collapse
Affiliation(s)
- Gui Hong Zhang
- Epithelial Cell Biology Research Center, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|