1
|
Métivier L, Vivien D, Goy R, Agin V, Bui E, Benbrika S. Plasminogen Activator Inhibitor-1 in the Pathophysiology of Late Life Depression. Int J Geriatr Psychiatry 2024; 39:e70015. [PMID: 39578639 DOI: 10.1002/gps.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Abstract
INTRODUCTION Late life depression (LLD) is characterized by specific clinical features including a high frequency of vascular form and frequent antidepressant treatment resistance. The expression and functions of the serine protease inhibitor, Plasminogen Activator Inhibitor-1 (PAI-1) is known to be altered by aging, vascular damage, insulin levels associated with a sedentary lifestyle, chronic stress leading to hypercortisolemia, and inflammatory changes linked to stress responses. These phenomena would be implicated in LLD like vascular depression. This article thus aims to review the existing literature regarding the association between LLD and plasmatic levels of PAI-1, a marker of hypofibrinolysis. We hypothesize that increased age would be associated with changes in PAI-1 plasma level and function which influence LLD pathogenesis and its treatment. RESULTS Although a large number of studies on PAI-1 changes in the elderly exist, studies about its implications in LLD are sparse. Despite heterogeneous findings regarding the direction of variation in plasmatic PAI-1 levels among elderly participants with LLD, all studies demonstrated an association between PAI-1 levels and current or remitted depressive symptoms. Moreover, disruptions in the concentrations of other biological markers influencing PAI-1 expression, such as cytokines or adipokines, were also observed, notably an increase in the levels of interleukins 6 and 8. DISCUSSION LLD genesis appears to be influenced by PAI-1 regulatory loops which are implicated in senescence or cell death. The resistance to antidepressant treatment appears to be linked to distinct biological profiles involving inflammatory and fibrinolytic factors. Taken together these data suggest that PAI-1 pathway may be a promising target of treatment development efforts for LLD, and depression in general.
Collapse
Affiliation(s)
- L Métivier
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
| | - D Vivien
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - R Goy
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
| | - V Agin
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - E Bui
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
- CHU de CAEN Normandie, Service de Psychiatrie, Centre Esquirol, Caen, France
| | - S Benbrika
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
- CHU de CAEN Normandie, Service de Psychiatrie, Centre Esquirol, Caen, France
| |
Collapse
|
2
|
He Y, Wang K, Su N, Yuan C, Zhang N, Hu X, Fu Y, Zhao F. Microbiota-gut-brain axis in health and neurological disease: Interactions between gut microbiota and the nervous system. J Cell Mol Med 2024; 28:e70099. [PMID: 39300699 PMCID: PMC11412916 DOI: 10.1111/jcmm.70099] [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: 04/18/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
Along with mounting evidence that gut microbiota and their metabolites migrate endogenously to distal organs, the 'gut-lung axis,' 'gut-brain axis,' 'gut-liver axis' and 'gut-renal axis' have been established. Multiple animal recent studies have demonstrated gut microbiota may also be a key susceptibility factor for neurological disorders such as Alzheimer's disease, Parkinson's disease and autism. The gastrointestinal tract is innervated by the extrinsic sympathetic and vagal nerves and the intrinsic enteric nervous system, and the gut microbiota interacts with the nervous system to maintain homeostatic balance in the host gut. A total of 1507 publications on the interactions between the gut microbiota, the gut-brain axis and neurological disorders are retrieved from the Web of Science to investigate the interactions between the gut microbiota and the nervous system and the underlying mechanisms involved in normal and disease states. We provide a comprehensive overview of the effects of the gut microbiota and its metabolites on nervous system function and neurotransmitter secretion, as well as alterations in the gut microbiota in neurological disorders, to provide a basis for the possibility of targeting the gut microbiota as a therapeutic agent for neurological disorders.
Collapse
Affiliation(s)
- Yuhong He
- Department of Operating RoomChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Ke Wang
- Department of Operating RoomChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Niri Su
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Chongshan Yuan
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Naisheng Zhang
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Xiaoyu Hu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Yunhe Fu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Jilin UniversityChangchunJilinChina
| | - Feng Zhao
- Department of Operating RoomChina‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
3
|
Shen L, Zhong X, Ji H, Yang S, Jin J, Lyu C, Ren Y, Xiao Y, Zhang Y, Fang S, Lin N, Tou J, Shu Q, Lai D. Macrophage α7nAChR alleviates the inflammation of neonatal necrotizing enterocolitis through mTOR/NLRP3/IL-1β pathway. Int Immunopharmacol 2024; 139:112590. [PMID: 38996778 DOI: 10.1016/j.intimp.2024.112590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/08/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Neonatal necrotizing enterocolitis (NEC) is one of the most prevalent and severe intestinal emergencies in newborns. The inflammatory activation of macrophages is associated with the intestinal injury of NEC. The neuroimmune regulation mediated by α7 nicotinic acetylcholine receptor (α7nAChR) plays an important role in regulating macrophage activation and inflammation progression, but in NEC remains unclear. This study aims to explore the effect of macrophage α7nAChR on NEC. METHODS Mice NEC model were conducted with high-osmolarity formula feeding, hypoxia, and cold stimulation. The α7nAChR agonist PNU-282987 and mTOR inhibitor rapamycin were treated by intraperitoneal injections in mice. The expression and distribution of macrophages, α7nAChR, and phospho-mammalian target of rapamycin (p-mTOR) in the intestines of NEC patients and mice was assessed using immunohistochemistry, immunofluorescence, and flow cytometry. The expression of NLRP3, activated caspase-1 and IL-1β in mice intestines was detected by flow cytometry, western blot or ELISA. In vitro, the mouse RAW264.7 macrophage cell line was also cultured followed by various treatments. Expression of p-mTOR, NLRP3, activated caspase-1, and IL-1β in macrophages was determined. RESULTS Macrophages accumulated in the intestines and the expression of α7nAChR in the mucosal and submucosal layers of the intestines was increased in both the NEC patients and mice. The p-mTOR and CD68 were increased and co-localized in intestines of NEC patients. In vitro, α7nAChR agonist PNU-282987 significantly reduced the increase of NLRP3, activated caspase-1, and IL-1β in macrophages. PNU-282987 also significantly reduced the increase of p-mTOR. The effect was blocked by AMPK inhibitor compound C. The expression of NLRP3, activated caspase-1, and IL-1β was inhibited after mTOR inhibitor rapamycin treatment. In NEC model mice, PNU-282987 reduced the expression of p-mTOR, NLRP3, activated caspase-1, and IL-1β in the intestine. Meanwhile, rapamycin significantly attenuated NLRP3 activation and the release of IL-1β. Moreover, the proportion of intestinal macrophages and intestinal injury decreased after PNU-282987 treatment. CONCLUSION Macrophage α7nAChR activation mitigates NLRP3 inflammasome activation by modulating mTOR phosphorylation, and subsequently alleviates intestinal inflammation and injury in NEC.
Collapse
Affiliation(s)
- Leiting Shen
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Xiaohui Zhong
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Haosen Ji
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Sisi Yang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jingyi Jin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Chengjie Lyu
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yichao Ren
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yi Xiao
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Yuebai Zhang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Shu Fang
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Nan Lin
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
4
|
Yip JLK, Balasuriya GK, Hill-Yardin EL, Spencer SJ. The gut-brain and gut-macrophage contribution to gastrointestinal dysfunction with systemic inflammation. Brain Behav Immun 2024; 119:867-877. [PMID: 38750700 DOI: 10.1016/j.bbi.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024] Open
Abstract
The gastrointestinal tract is one of the main organs affected during systemic inflammation and disrupted gastrointestinal motility is a major clinical manifestation. Many studies have investigated the involvement of neuroimmune interactions in regulating colonic motility during localized colonic inflammation, i.e., colitis. However, little is known about how the enteric nervous system and intestinal macrophages contribute to dysregulated motility during systemic inflammation. Given that systemic inflammation commonly results from the innate immune response against bacterial infection, we mimicked bacterial infection by administering lipopolysaccharide (LPS) to rats and assessed colonic motility using ex vivo video imaging techniques. We utilized the Cx3cr1-Dtr rat model of transient depletion of macrophages to investigate the role of intestinal macrophages in regulating colonic motility during LPS infection. To investigate the role of inhibitory enteric neurotransmission on colonic motility following LPS, we applied the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (NOLA). Our results confirmed an increase in colonic contraction frequency during LPS-induced systemic inflammation. However, neither the depletion of intestinal macrophages, nor the suppression of inhibitory enteric nervous system activity impacted colonic motility disruption during inflammation. This implies that the interplay between the enteric nervous system and intestinal macrophages is nuanced, and complex, and further investigation is needed to clarify their joint roles in colonic motility.
Collapse
Affiliation(s)
- Jackson L K Yip
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Gayathri K Balasuriya
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia; Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Asgari R, Bazzazan MA, Karimi Jirandehi A, Yousefzadeh S, Alaei M, Keshavarz Shahbaz S. Peyer's Patch: Possible target for modulating the Gut-Brain-Axis through microbiota. Cell Immunol 2024; 401-402:104844. [PMID: 38901288 DOI: 10.1016/j.cellimm.2024.104844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/05/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
The gastrointestinal (GI) tract and the brain form bidirectional nervous, immune, and endocrine communications known as the gut-brain axis. Several factors can affect this axis; among them, various studies have focused on the microbiota and imply that alterations in microbiota combinations can influence both the brain and GI. Also, many studies have shown that the immune system has a vital role in varying gut microbiota combinations. In the current paper, we will review the multidirectional effects of gut microbiota, immune system, and nervous system on each other. Specifically, this review mainly focuses on the impact of Peyer's patches as a critical component of the gut immune system on the gut-brain axis through affecting the gut's microbial composition. In this way, some factors were discussed as proposed elements of missing gaps in this field.
Collapse
Affiliation(s)
- Reza Asgari
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Mohammad Amin Bazzazan
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Ashkan Karimi Jirandehi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Salar Yousefzadeh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Masood Alaei
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical science, Qazvin, Iran
| | - Sanaz Keshavarz Shahbaz
- USERN Office, Qazvin University of Medical science, Qazvin, Iran; Cellular and Molecular Research Center, Research Institute for prevention of Non- Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
6
|
Chen BR, Wu T, Chen TH, Wang Y. Neuroimmune interactions and their roles in neurodegenerative diseases. FUNDAMENTAL RESEARCH 2024; 4:251-261. [PMID: 38933502 PMCID: PMC11197660 DOI: 10.1016/j.fmre.2023.04.002] [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: 07/21/2022] [Revised: 02/10/2023] [Accepted: 04/03/2023] [Indexed: 06/28/2024] Open
Abstract
The nervous system possesses bidirectional, sophisticated and delicate communications with the immune system. These neuroimmune interactions play a vitally important role in the initiation and development of many disorders, especially neurodegenerative diseases. Although scientific advancements have made tremendous progress in this field during the last few years, neuroimmune communications are still far from being elucidated. By organizing recent research, in this review, we discuss the local and intersystem neuroimmune interactions and their roles in Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Unveiling these will help us gain a better understanding of the process of interplay inside the body and how the organism maintains homeostasis. It will also facilitate a view of the diseases from a holistic, pluralistic and interconnected perspective, thus providing a basis of developing novel and effective methods to diagnose, intervene and treat diseases.
Collapse
Affiliation(s)
- Bai-Rong Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Ting Wu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Ting-Hui Chen
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Boem F, Greslehner GP, Konsman JP, Chiu L. Minding the gut: extending embodied cognition and perception to the gut complex. Front Neurosci 2024; 17:1172783. [PMID: 38260022 PMCID: PMC10800657 DOI: 10.3389/fnins.2023.1172783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/30/2023] [Indexed: 01/24/2024] Open
Abstract
Scientific and philosophical accounts of cognition and perception have traditionally focused on the brain and external sense organs. The extended view of embodied cognition suggests including other parts of the body in these processes. However, one organ has often been overlooked: the gut. Frequently conceptualized as merely a tube for digesting food, there is much more to the gut than meets the eye. Having its own enteric nervous system, sometimes referred to as the "second brain," the gut is also an immune organ and has a large surface area interacting with gut microbiota. The gut has been shown to play an important role in many physiological processes, and may arguably do so as well in perception and cognition. We argue that proposals of embodied perception and cognition should take into account the role of the "gut complex," which considers the enteric nervous, endocrine, immune, and microbiota systems as well as gut tissue and mucosal structures. The gut complex is an interface between bodily tissues and the "internalized external environment" of the gut lumen, involved in many aspects of organismic activity beyond food intake. We thus extend current embodiment theories and suggest a more inclusive account of how to "mind the gut" in studying cognitive processes.
Collapse
Affiliation(s)
- Federico Boem
- Section Philosophy, University of Twente, Enschede, Netherlands
| | | | - Jan Pieter Konsman
- IMMUNOlogy from CONcepts and ExPeriments to Translation, CNRS UMR, University of Bordeaux, Bordeaux, France
| | - Lynn Chiu
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Zhang Z, Cheng N, Liang J, Deng Y, Xiang P, Hei Z, Li X. Gut microbiota changes in animal models of spinal cord injury: a preclinical systematic review and meta-analysis. Ann Med 2023; 55:2269379. [PMID: 37851840 PMCID: PMC10586076 DOI: 10.1080/07853890.2023.2269379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND An increasing number of studies show that the intestinal flora is closely related to spinal cord injury. Many researchers are exploring the changes in the richness, diversity, and evenness of intestinal flora in spinal cord injury animal models to identify the characteristic bacteria. METHODS A comprehensive literature search was conducted using three databases: PubMed, Embase, and Web of Science. A meta-analysis was performed using R 4.3.1 to evaluate the comparison of microbiota diversity, richness, and evenness and the relative abundance of intestinal microbiota in animals with spinal cord injury and blank controls. RESULTS Fifteen studies were included in the meta-analysis, of which 12 involved gut microbiota distribution indicators and 11 included intestinal microflora relative abundance indicators. Meta-analysis of high-dimensional indicators describing the distribution of the gut microbiota identified a substantial decline in the evenness and richness of the intestinal flora. In addition, the Actinobacteria phylum and Erysipelotrichales and Clostridiales orders were significantly different between the spinal cord injury and sham groups; therefore, they may be the characteristic bacteria in spinal cord injury models. CONCLUSION Our meta-analysis suggested that the gut microbiota in the spinal cord injury animal model group was altered compared with that in the control group, with varying degrees of changes in richness and evenness and potentially pathogenic characteristic flora. More rigorous methodological studies are needed because of the high heterogeneity and limited sample size. Further research is needed to clinically apply intestinal microbiota and potentially guide fecal microbiota transplantation therapy.
Collapse
Affiliation(s)
- Zhenye Zhang
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Nan Cheng
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianfen Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yifan Deng
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ping Xiang
- Department of Medical Quality Management, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiang Li
- Department of Anesthesiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
9
|
He Y, Zhang M, Dai C, Yu L. Comparison of the Gut Microbial Communities of Domestic and Wild Mallards ( Anas platyrhynchos) Based on High-Throughput Sequencing Technology. Animals (Basel) 2023; 13:2956. [PMID: 37760356 PMCID: PMC10525502 DOI: 10.3390/ani13182956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Mallards (Anas platyrhynchos) are currently one of the most popular species in rare bird breeding in several southern provinces of China, but there have been no studies comparing the gut microbial communities of domestic and wild mallards. In this study, 16S rRNA gene high-throughput sequencing technology was used to compare the composition and diversity of gut microbial communities in domestic and wild mallards. Alpha diversity analysis showed significant differences in gut microbial communities between the two groups of mallards, and the diversity and richness of gut microbial communities were significantly higher in wild mallards than in domestic mallards. Beta diversity analysis showed that the two groups of stool samples were mostly separated on the principal coordinate analysis (PCoA) plot. In domestic mallards, Firmicutes (68.0% ± 26.5%) was the most abundant bacterial phylum, followed by Proteobacteria (24.5% ± 22.9%), Bacteroidetes (3.1% ± 3.2%), Fusobacteria (2.2% ± 5.9%), and Actinobacteria (1.1% ± 1.8%). The dominant bacterial phyla in wild mallards were Firmicutes (79.0% ± 10.2%), Proteobacteria (12.9% ± 9.5%), Fusobacteria (3.4% ± 2.5%), and Bacteroidetes (2.8% ± 2.4%). At the genus level, a total of 10 dominant genera (Streptococcus, Enterococcus, Clostridium, Lactobacillus, Soilbacillus, Bacillus, Acinetobacter, Comamonas, Shigella, and Cetobacterium) with an average relative abundance greater than 1% were detected in the fecal samples of both groups. The average relative abundance of five potential pathogenic genera (Streptococcus, Enterococcus, Acinetobacter, Comamonas, and Shigella) was higher in domestic mallards than in wild mallards. The enrichment of pathogenic bacteria in the intestinal tract of domestic mallards should be of sufficient concern.
Collapse
Affiliation(s)
- Yaoyin He
- Animal Science and Technology College, Guangxi University, Nanning 530004, China; (Y.H.); (M.Z.)
| | - Minghui Zhang
- Animal Science and Technology College, Guangxi University, Nanning 530004, China; (Y.H.); (M.Z.)
| | - Chuanyin Dai
- Key Laboratory of Ecology of Rare and Endangered Species and Environmental Protection (Guangxi Normal University), Ministry of Education, Guilin 541006, China;
| | - Lijiang Yu
- Animal Science and Technology College, Guangxi University, Nanning 530004, China; (Y.H.); (M.Z.)
| |
Collapse
|
10
|
Yan H, Zhao S, Huang HX, Xie P, Cai XH, Qu YD, Zhang W, Luo JQ, Zhang L, Li X. Systematic Mendelian randomization study of the effect of gut microbiome and plasma metabolome on severe COVID-19. Front Immunol 2023; 14:1211612. [PMID: 37662924 PMCID: PMC10468967 DOI: 10.3389/fimmu.2023.1211612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
Background COVID-19 could develop severe respiratory symptoms in certain infected patients, especially in the patients with immune disorders. Gut microbiome and plasma metabolome act important immunological modulators in the human body and could contribute to the immune responses impacting the progression of COVID-19. However, the causal relationship between specific intestinal bacteria, metabolites and severe COVID-19 remains not clear. Methods Based on two-sample Mendelian randomization (MR) framework, the causal effects of 131 intestinal taxa and 452 plasma metabolites on severe COVID-19 were evaluated. Single nucleotide polymorphisms (SNPs) strongly associated with the abundance of intestinal taxa and the concentration of plasma metabolites had been utilized as the instrument variables to infer whether they were causal factors of severe COVID-19. In addition, mediation analysis was conducted to find the potential association between the taxon and metabolite, and further colocalization analysis had been performed to validate the causal relationships. Results MR analysis identified 13 taxa and 53 metabolites, which were significantly associated with severe COVID-19 as causal factors. Mediation analysis revealed 11 mediated relationships. Myo-inositol, 2-stearoylglycerophosphocholine, and alpha-glutamyltyrosine, potentially contributed to the association of Howardella and Ruminiclostridium 6 with severe COVID-19, respectively. Butyrivibrio and Ruminococcus gnavus could mediate the association of myo-inositol and N-acetylalanine, respectively. In addition, Ruminococcus torques abundance was colocalized with severe COVID-19 (PP.H4 = 0.77) and the colon expression of permeability related protein RASIP1 (PP.H4 = 0.95). Conclusions Our study highlights the potential causal relationships between gut microbiome, plasma metabolome and severe COVID-19, which potentially serve as clinical biomarkers for risk stratification and prognostication and benefit the mechanism mechanistic investigation of severe COVID-19.
Collapse
Affiliation(s)
- Han Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Si Zhao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Han-Xue Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pan Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin-He Cai
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yun-Dan Qu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian-Quan Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Longbo Zhang
- Departments of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, United States
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan, China
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Zhang J, Xiao Y, Wang H, Zhang H, Chen W, Lu W. Lactic acid bacteria-derived exopolysaccharide: Formation, immunomodulatory ability, health effects, and structure-function relationship. Microbiol Res 2023; 274:127432. [PMID: 37320895 DOI: 10.1016/j.micres.2023.127432] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Exopolysaccharides (EPSs) synthesized by lactic acid bacteria (LAB) have implications for host health and act as food ingredients. Due to the variability of LAB-EPS (lactic acid bacteria-derived exopolysaccharide) gene clusters, especially the glycosyltransferase genes that determine monosaccharide composition, the structure of EPS is very rich. EPSs are synthesized by LAB through the extracellular synthesis pathway and the Wzx/Wzy-dependent pathway. LAB-EPS has a strong immunomodulatory ability. The EPSs produced by different genera of LAB, especially Lactobacillus, Leuconostoc, and Streptococcus, have different immunomodulatory abilities because of their specific structures. LAB-EPS possesses other health effects, including antitumor, antioxidant, intestinal barrier repair, antimicrobial, antiviral, and cholesterol-lowering activities. The bioactivities of LAB-EPS are tightly related to their structures such us monosaccharide composition, glycosidic bonds, and molecular weight (MW). For the excellent physicochemical property, LAB-EPS acts as product improvers in dairy, bakery food, and meat in terms of stability, emulsification, thickening, and gelling. We systematically summarize the detailed process of EPS from synthesis to application, with emphasis on physiological mechanisms of EPS, and specific structure-function relationship, which provides theoretical support for the potential commercial value in the pharmaceutical, chemical, food, and cosmetic industries.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
12
|
Bai S, Zhang G, Han Y, Ma J, Bai B, Gao J, Zhang Z. Ginsenosides and Polysaccharides from Ginseng Co-Fermented with Multi-Enzyme-Coupling Probiotics Improve In Vivo Immunomodulatory Effects. Nutrients 2023; 15:nu15112434. [PMID: 37299397 DOI: 10.3390/nu15112434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
The active components of ginseng, such as ginsenosides and polysaccharides, have high therapeutic value in treating cancer, decreasing obesity, and enhancing immunity. However, simple primary ginseng treatment cannot maximize this medicinal potential. Therefore, in this study, Panax ginseng was co-fermented with multi-enzyme-coupling probiotics to obtain a fermentation broth with higher levels of ginsenosides, polysaccharides, and probiotics. When compared to other treatment methods for cyclophosphamide-induced immunosuppression in mice, the results reveal that the P. ginseng fermentation broth treated with multi-enzyme-coupling probiotics could significantly improve the immune function of immunosuppressive mice and restore intestinal flora stability. Overall, this processing method will provide a novel strategy for promoting the application of ginseng and the relief of immunosuppression.
Collapse
Affiliation(s)
- Shaowei Bai
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Guangyun Zhang
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yaqin Han
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jianwei Ma
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Bing Bai
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jingjie Gao
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zuoming Zhang
- Key Laboratory for Molecular Enzymology & Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
13
|
Labrenz F, Merz CJ, Icenhour A. Connecting dots in disorders of gut-brain interaction: the interplay of stress and sex hormones in shaping visceral pain. Front Psychiatry 2023; 14:1204136. [PMID: 37275987 PMCID: PMC10235543 DOI: 10.3389/fpsyt.2023.1204136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Visceral pain and stress are tightly intertwined bodily and emotional phenomena, which enable a flexible adaptation to environmental challenges by activating a response repertoire to restore homeostasis along the gut-brain axis. However, visceral pain and stress can persist widely independent of the initial cause, acquiring independent disease values and posing major health burdens as predominant features in disorders of gut-brain interaction (DGBI). Epidemiological data consistently documents an increased prevalence for women to suffer from chronic visceral pain, possibly shaped by sex hormones and modulated by stress and its biological and psychosocial correlates. Yet, mechanisms underlying the complex interactions between altered visceroception, stress and sex remain widely elusive, especially in clinical populations with DGBI. We herein selectively review mechanisms of interactions between stress and sex in the complex pathophysiology of DGBI. A particular emphasis is laid on visceral pain, in which stress constitutes a major risk factor as well as mediator, and sex-related differences are particularly pronounced. Building on the neurobiology of stress and mechanisms of gut-brain interactions, we highlight putative target mechanisms via which visceral pain and stress may converge with sex effects into a triad. Accommodating a global demographic shift, we propose a lifespan perspective in future research, which may enable a more fine-tuned evaluation of this complex interplay exerting distinct challenges during vulnerable developmental phases. This viewpoint may advance our understanding of pathophysiological processes and can ultimately inspire novel tailored prevention strategies and therapeutic approaches in the treatment of chronic visceral pain and DGBI across the lifespan.
Collapse
Affiliation(s)
- Franziska Labrenz
- Department of Medical Psychology and Medical Sociology, Ruhr University Bochum, Bochum, Germany
| | - Christian J. Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Adriane Icenhour
- Department of Medical Psychology and Medical Sociology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
14
|
Bagyánszki M, Bódi N. Key elements determining the intestinal region-specific environment of enteric neurons in type 1 diabetes. World J Gastroenterol 2023; 29:2704-2716. [PMID: 37274063 PMCID: PMC10237112 DOI: 10.3748/wjg.v29.i18.2704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Diabetes, as a metabolic disorder, is accompanied with several gastrointestinal (GI) symptoms, like abdominal pain, gastroparesis, diarrhoea or constipation. Serious and complex enteric nervous system damage is confirmed in the background of these diabetic motility complaints. The anatomical length of the GI tract, as well as genetic, developmental, structural and functional differences between its segments contribute to the distinct, intestinal region-specific effects of hyperglycemia. These observations support and highlight the importance of a regional approach in diabetes-related enteric neuropathy. Intestinal large and microvessels are essential for the blood supply of enteric ganglia. Bidirectional morpho-functional linkage exists between enteric neurons and enteroglia, however, there is also a reciprocal communication between enteric neurons and immune cells on which intestinal microbial composition has crucial influence. From this point of view, it is more appropriate to say that enteric neurons partake in multidirectional communication and interact with these key players of the intestinal wall. These interplays may differ from segment to segment, thus, the microenvironment of enteric neurons could be considered strictly regional. The goal of this review is to summarize the main tissue components and molecular factors, such as enteric glia cells, interstitial cells of Cajal, gut vasculature, intestinal epithelium, gut microbiota, immune cells, enteroendocrine cells, pro-oxidants, antioxidant molecules and extracellular matrix, which create and determine a gut region-dependent neuronal environment in diabetes.
Collapse
Affiliation(s)
- Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| |
Collapse
|
15
|
Aldossari AA, Ansari MA, Nadeem A, Attia SM, Bakheet SA, Al-Ayadhi LY, Alanazi MM, Shahid M, Alwetaid MY, Hussein MH, Ahmad SF. Upregulation of Inflammatory Mediators in Peripheral Blood CD40 + Cells in Children with Autism Spectrum Disorder. Int J Mol Sci 2023; 24:ijms24087475. [PMID: 37108638 PMCID: PMC10138695 DOI: 10.3390/ijms24087475] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is a common and severe neurodevelopmental disorder in early childhood, defined as social and communication deficits and repetitive and stereotypic behaviours. The aetiology is unknown in most cases. However, several studies have identified immune dysregulation as potentially promoting ASD. Among the numerous immunological findings in ASD, reports of increased pro-inflammatory markers remain the most consistently observed. C-C chemokine receptor type 1 (CCR1) activation is pro-inflammatory in several neurological disorders. Previous evidence has implied that the expression of chemokine receptors, inflammatory mediators, and transcription factors play a pivotal role in several neuroinflammatory disorders. There have also been reports on the association between increased levels of proinflammatory cytokines and ASD. In this study, we aimed to investigate the possible involvement of CCR1, inflammatory mediators, and transcription factor expression in CD40+ cells in ASD compared to typically developing controls (TDC). Flow cytometry analysis was used to determine the levels of CCR1-, IFN-γ-, T-box transcription factor (T-bet-), IL-17A-, retinoid-related orphan receptor gamma t (RORγt-), IL-22- and TNF-α-expressing CD40 cells in PBMCs in children with ASD and the TDC group. We further examined the mRNA and protein expression levels of CCR1 using real-time PCR and western blot analysis. Our results revealed that children with ASD had significantly increased numbers of CD40+CCR1+, CD40+IFN-γ+, CD40+T-bet+, CD40+IL-17A+, CD40+RORγt+, CD4+IL-22+, and CD40+TNF-α+ cells compared with the TDC group. Furthermore, children with ASD had higher CCR1 mRNA and protein expression levels than those in the TDC group. These results indicate that CCR1, inflammatory mediators, and transcription factors expressed in CD40 cells play vital roles in disease progression.
Collapse
Affiliation(s)
- Abdullah A Aldossari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila Y Al-Ayadhi
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Y Alwetaid
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa H Hussein
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
16
|
Mayer EA, Ryu HJ, Bhatt RR. The neurobiology of irritable bowel syndrome. Mol Psychiatry 2023; 28:1451-1465. [PMID: 36732586 PMCID: PMC10208985 DOI: 10.1038/s41380-023-01972-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023]
Abstract
Irritable bowel syndrome (IBS) is the most prevalent disorder of brain-gut interactions that affects between 5 and 10% of the general population worldwide. The current symptom criteria restrict the diagnosis to recurrent abdominal pain associated with altered bowel habits, but the majority of patients also report non-painful abdominal discomfort, associated psychiatric conditions (anxiety and depression), as well as other visceral and somatic pain-related symptoms. For decades, IBS was considered an intestinal motility disorder, and more recently a gut disorder. However, based on an extensive body of reported information about central, peripheral mechanisms and genetic factors involved in the pathophysiology of IBS symptoms, a comprehensive disease model of brain-gut-microbiome interactions has emerged, which can explain altered bowel habits, chronic abdominal pain, and psychiatric comorbidities. In this review, we will first describe novel insights into several key components of brain-gut microbiome interactions, starting with reported alterations in the gut connectome and enteric nervous system, and a list of distinct functional and structural brain signatures, and comparing them to the proposed brain alterations in anxiety disorders. We will then point out the emerging correlations between the brain networks with the genomic, gastrointestinal, immune, and gut microbiome-related parameters. We will incorporate this new information into a systems-based disease model of IBS. Finally, we will discuss the implications of such a model for the improved understanding of the disorder and the development of more effective treatment approaches in the future.
Collapse
Affiliation(s)
- Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Departments of Medicine, Psychiatry and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Hyo Jin Ryu
- A.T. Still University School of Osteopathic Medicine in Arizona, Meza, AZ, USA
| | - Ravi R Bhatt
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine at USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Yao H, Fu X, Xu Q, Li T, Li Y, Kang Y, Wu Q. The macrophages regulate intestinal motility dysfunction through the PGE2 Ptger3 axis during Klebsiella pneumonia sepsis. Front Immunol 2023; 14:1147674. [PMID: 37063880 PMCID: PMC10090685 DOI: 10.3389/fimmu.2023.1147674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
IntroductionGut motility dysfunction, the most common complication of post-septic organ dysfunction, depends on immune and neuronal cells. This study aimed to investigate the mechanisms that activate these cells and the contribution of macrophages to the recovery of intestinal motility dysfunction after sepsis.Materials and methodsPostoperative gut motility dysfunction was induced by establishing Klebsiella pneumonia sepsis in mice with selective deletion of neutrophils and macrophages in the gut. The distribution of orally administered fluorescein isothiocyanate-dextran and carmine excretion time was used to determine the severity of small bowel disease. The effect of macrophages on intestinal motility was evaluated after prostaglandin E2 therapy.ResultsWe found that muscular neutrophil infiltration leading to neuronal loss in the intestine muscle triggered intestinal motility dysfunction after pneumonia sepsis; however, reduced neutrophil infiltration did not improve intestinal motility dysfunction. Moreover, macrophage depletion aggravated gut motility dysfunction. The addition of macrophages directly to a smooth muscle was responsible for the recovery of intestinal motility.ConclusionOur results suggest that a direct interaction between macrophages and smooth muscle is neurologically independent of the restoration of intestinal dysmotility.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan Kang
- *Correspondence: Yan Kang, ; Qin Wu,
| | - Qin Wu
- *Correspondence: Yan Kang, ; Qin Wu,
| |
Collapse
|
18
|
Abo-Shaban T, Sharna SS, Hosie S, Lee CYQ, Balasuriya GK, McKeown SJ, Franks AE, Hill-Yardin EL. Issues for patchy tissues: defining roles for gut-associated lymphoid tissue in neurodevelopment and disease. J Neural Transm (Vienna) 2023; 130:269-280. [PMID: 36309872 PMCID: PMC10033573 DOI: 10.1007/s00702-022-02561-x] [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: 08/30/2022] [Accepted: 10/20/2022] [Indexed: 10/31/2022]
Abstract
Individuals diagnosed with neurodevelopmental conditions such as autism spectrum disorder (ASD; autism) often experience tissue inflammation as well as gastrointestinal dysfunction, yet their underlying causes remain poorly characterised. Notably, the largest components of the body's immune system, including gut-associated lymphoid tissue (GALT), lie within the gastrointestinal tract. A major constituent of GALT in humans comprises secretory lymphoid aggregates known as Peyer's patches that sense and combat constant exposure to pathogens and infectious agents. Essential to the functions of Peyer's patches is its communication with the enteric nervous system (ENS), an intrinsic neural network that regulates gastrointestinal function. Crosstalk between these tissues contribute to the microbiota-gut-brain axis that altogether influences mood and behaviour. Increasing evidence further points to a critical role for this signalling axis in neurodevelopmental homeostasis and disease. Notably, while the neuroimmunomodulatory functions for Peyer's patches are increasingly better understood, functions for tissues of analogous function, such as caecal patches, remain less well characterised. Here, we compare the structure, function and development of Peyer's patches, as well as caecal and appendix patches in humans and model organisms including mice to highlight the roles for these essential tissues in health and disease. We propose that perturbations to GALT function may underlie inflammatory disorders and gastrointestinal dysfunction in neurodevelopmental conditions such as autism.
Collapse
Affiliation(s)
- T Abo-Shaban
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - S S Sharna
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, USA
| | - S Hosie
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - C Y Q Lee
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - G K Balasuriya
- Department of Physiology and Cell Biology, Kobe University School of Medicine, 7-5-1 Kusunoki-Cho, Chuo, Kobe, 650-0017, Japan
| | - S J McKeown
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - A E Franks
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - E L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
19
|
Schill EM, Floyd AN, Newberry RD. Neonatal development of intestinal neuroimmune interactions. Trends Neurosci 2022; 45:928-941. [PMID: 36404456 PMCID: PMC9683521 DOI: 10.1016/j.tins.2022.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/19/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022]
Abstract
Interactions between the enteric nervous system (ENS), immune system, and gut microbiota regulate intestinal homeostasis in adults, but their development and role(s) in early life are relatively underexplored. In early life, these interactions are dynamic, because the mucosal immune system, microbiota, and the ENS are developing and influencing each other. Moreover, disrupting gut microbiota and gut immune system development, and potentially ENS development, by early-life antibiotic exposure increases the risk of diseases affecting the gut. Here, we review the development of the ENS and immune/epithelial cells, and identify potential critical periods for their interactions and development. We also highlight knowledge gaps that, when addressed, may help promote intestinal homeostasis, including in the settings of early-life antibiotic exposure.
Collapse
Affiliation(s)
- Ellen Merrick Schill
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA; Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA.
| | - Alexandria N Floyd
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Rodney D Newberry
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
20
|
Telocytes and Macrophages in the Gut: From Morphology to Function. Do the Two Cell Types Interact with Each Other? Which Helps Which? Int J Mol Sci 2022; 23:ijms23158435. [PMID: 35955569 PMCID: PMC9369081 DOI: 10.3390/ijms23158435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 01/27/2023] Open
Abstract
Telocytes and macrophages are ubiquitous cells located in loose connective tissues and share the same mesenchymal origin. Despite these common elements, depending on where they reside, these two cell types are profoundly different in terms of their morphology and functions. The purpose of this review is to provide an update on the knowledge regarding telocytes and macrophages in the gut, where their presence and significance have long been underestimated or misunderstood. The focus will be on the possibility that these two cell types interact with each other and on the potential meaning of these interactions. Based on the complexity of the topic, the variety of possible methodological approaches and the expertise of the author, the point of view in the discussion of the literature data will be mainly morphological. Furthermore, considering the relatively recent period in which these cell types have acquired a primary role in gastrointestinal functions, the attention will be greatly confined to those articles published in the last decade. The microbiota, another main protagonist in this context, will be mentioned only in passing. It is hoped that this review, although not exhaustive, will highlight the importance of macrophages and telocytes in the complex mechanisms that ensure intestinal functions.
Collapse
|
21
|
Middelhoff M, Valenti G, Tomassoni L, Ochiai Y, Belin B, Takahashi R, Malagola E, Nienhüser H, Finlayson M, Hayakawa Y, Zamechek LB, Renz BW, Westphalen CB, Quante M, Margolis KG, Sims PA, Laise P, Califano A, Rao M, Gershon MD, Wang TC. Adult enteric Dclk1-positive glial and neuronal cells reveal distinct responses to acute intestinal injury. Am J Physiol Gastrointest Liver Physiol 2022; 322:G583-G597. [PMID: 35319286 PMCID: PMC9109794 DOI: 10.1152/ajpgi.00244.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 01/31/2023]
Abstract
Intestinal ganglionic cells in the adult enteric nervous system (ENS) are continually exposed to stimuli from the surrounding microenvironment and need at times to respond to disturbed homeostasis following acute intestinal injury. The kinase DCLK1 and intestinal Dclk1-positive cells have been reported to contribute to intestinal regeneration. Although Dclk1-positive cells are present in adult enteric ganglia, their cellular identity and response to acute injury have not been investigated in detail. Here, we reveal the presence of distinct Dclk1-tdTom+/CD49b+ glial-like and Dclk1-tdTom+/CD49b- neuronal cell types in adult myenteric ganglia. These ganglionic cells demonstrate distinct patterns of tracing over time yet show a similar expansion in response to elevated serotonergic signaling. Interestingly, Dclk1-tdTom+ glial-like and neuronal cell types appear resistant to acute irradiation injury-mediated cell death. Moreover, Dclk1-tdTom+/CD49b+ glial-like cells show prominent changes in gene expression profiles induced by injury, in contrast to Dclk1-tdTom+/CD49b- neuronal cell types. Finally, subsets of Dclk1-tdTom+/CD49b+ glial-like cells demonstrate prominent overlap with Nestin and p75NTR and strong responses to elevated serotonergic signaling or acute injury. These findings, together with their role in early development and their neural crest-like gene expression signature, suggest the presence of reserve progenitor cells in the adult Dclk1 glial cell lineage.NEW & NOTEWORTHY The kinase DCLK1 identifies glial-like and neuronal cell types in adult murine enteric ganglia, which resist acute injury-mediated cell death yet differ in their cellular response to injury. Interestingly, Dclk1-labeled glial-like cells show prominent transcriptional changes in response to injury and harbor features reminiscent of previously described enteric neural precursor cells. Our data thus add to recently emerging evidence of reserve cellular plasticity in the adult enteric nervous system.
Collapse
Affiliation(s)
- Moritz Middelhoff
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Giovanni Valenti
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Lorenzo Tomassoni
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Yosuke Ochiai
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Bryana Belin
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Ryota Takahashi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Henrik Nienhüser
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Finlayson
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Leah B Zamechek
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Bernhard W Renz
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Munich, Germany
| | - C Benedikt Westphalen
- Department of Internal Medicine, Comprehensive Cancer Center, Hospital of the University of Munich, Munich, Germany
| | - Michael Quante
- Klinik für Innere Medizin II, Gastrointestinale Onkologie, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Kara G Margolis
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York
| | - Peter A Sims
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York
- Department of Biochemistry and Molecular Biophysics, Columbia University College of Physicians and Surgeons, New York, New York
| | - Pasquale Laise
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York
- DarwinHealth Inc., New York, New York
| | - Andrea Califano
- Department of Systems Biology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Meenakshi Rao
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children´s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| |
Collapse
|
22
|
Cang W, Wu J, Ding R, Wang W, Li N, Shi H, Shi L, Lee Y, Wu R. Potential of Probiotics as an Adjunct for Patients with Major Depressive Disorder. Mol Nutr Food Res 2022; 66:e2101057. [PMID: 35286767 DOI: 10.1002/mnfr.202101057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/12/2022] [Indexed: 11/06/2022]
Abstract
Major depressive disorder (MDD) is an enfeebling disease with a lifetime incidence of 20%. While accumulating studies implicate a correlation between the disease and gut microbiota, data show that not every patient responded to probiotic treatments. To comprehensively assess the potential role of probiotics in MDD, this study first summarizes the current pathological hypothesis of the disease from a life-stage perspective, focuses on the potential role of "depression gut microbiota." Currently available managements are then briefly summarized and novel bio-materials having potential therapeutic effects on MDD are also evaluated. To harness the positive effect of probiotics, prebiotics, and postbiotics, clinical evidence and their applications on MDD patients are listed. Factors that may counteract the pre/probiotic applications, such as diet, physiology, gender difference, and use of antibiotics and antidepressants are also discussed. The endocannabinoid (eCBs) system may be promising targets for probiotic therapy. More evidence is needed to demonstrate the hierarchical factors in the complex network driving the disease, and probiotic can be one promising adjunct for patients with MDD.
Collapse
Affiliation(s)
- Weihe Cang
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, P. R. China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, P. R. China.,Engineering Research Center of Food Fermentation Technology, Liaoning, 110866, P. R. China.,Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, P. R. China
| | - Ruixue Ding
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, P. R. China.,Engineering Research Center of Food Fermentation Technology, Liaoning, 110866, P. R. China.,Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, P. R. China
| | - Weiming Wang
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, 150036, P. R. China
| | - Na Li
- Children's Neurorehabilitation Laboratory, Shenyang Children's Hospital, Shenyang, 110033, P. R. China
| | - Haisu Shi
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, P. R. China.,Engineering Research Center of Food Fermentation Technology, Liaoning, 110866, P. R. China.,Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, P. R. China
| | - Lin Shi
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, P. R. China.,Engineering Research Center of Food Fermentation Technology, Liaoning, 110866, P. R. China.,Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, P. R. China
| | - Yuankun Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, P. R. China.,Engineering Research Center of Food Fermentation Technology, Liaoning, 110866, P. R. China.,Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, P. R. China
| |
Collapse
|
23
|
Almeida PP, Valdetaro L, Thomasi BBDM, Stockler-Pinto MB, Tavares-Gomes AL. High-fat diets on the enteric nervous system: Possible interactions and mechanisms underlying dysmotility. Obes Rev 2022; 23:e13404. [PMID: 34873814 DOI: 10.1111/obr.13404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/25/2021] [Accepted: 11/15/2021] [Indexed: 01/09/2023]
Abstract
Obesity is a chronic disease that affects various physiological systems. Among them, the gastrointestinal tract appears to be a main target of this disease. High-fat diet (HFD) animal models can help recapitulate the classic signs of obesity and present a series of gastrointestinal alterations, mainly dysmotility. Because intestinal motility is governed by the enteric nervous system (ENS), enteric neurons, and glial cells have been studied in HFD models. Given the importance of the ENS in general gut physiology, this review aims to discuss the relationship between HFD-induced neuroplasticity and gut dysmotility observed in experimental models. Furthermore, we highlight components of the gut environment that might influence enteric neuroplasticity, including gut microbiota, enteric glio-epithelial unit, serotonin release, immune cells, and disturbances such as inflammation and oxidative stress.
Collapse
Affiliation(s)
| | - Luisa Valdetaro
- Postgraduate Program in Neurosciences, Fluminense Federal University, Niterói, Brazil
| | | | - Milena Barcza Stockler-Pinto
- Postgraduate Program in Cardiovascular Sciences, Fluminense Federal University, Niterói, Brazil.,Postgraduate Program in Nutrition Sciences, Fluminense Federal University, Niterói, Brazil
| | | |
Collapse
|
24
|
Saldana-Morales FB, Kim DV, Tsai MT, Diehl GE. Healthy Intestinal Function Relies on Coordinated Enteric Nervous System, Immune System, and Epithelium Responses. Gut Microbes 2022; 13:1-14. [PMID: 33929291 PMCID: PMC8096330 DOI: 10.1080/19490976.2021.1916376] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
During both health and disease, a coordinated response between the epithelium, immune system, and enteric nervous system is required for proper intestinal function. While each system responds to a number of common stimuli, their coordinated responses support digestion as well as responses and recovery following injury or pathogenic infections. In this review, we discuss how individual responses to common signals work together to support these critical functions.
Collapse
Affiliation(s)
- Fatima B. Saldana-Morales
- Graduate School of Biomedical Sciences, Baylor College of Medicine, HoustonTXUSA,Immunology Program of the Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NYUSA
| | - Dasom V. Kim
- Immunology Program of the Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NYUSA,Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Ming-Ting Tsai
- Graduate School of Biomedical Sciences, Baylor College of Medicine, HoustonTXUSA,Immunology Program of the Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NYUSA
| | - Gretchen E. Diehl
- Immunology Program of the Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NYUSA,Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA,CONTACT Gretchen E. Diehl Immunology Program of the Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10021, USA. Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, Cornell University, New York, NY, USA
| |
Collapse
|
25
|
Enteric neuroimmune interactions coordinate intestinal responses in health and disease. Mucosal Immunol 2022; 15:27-39. [PMID: 34471248 PMCID: PMC8732275 DOI: 10.1038/s41385-021-00443-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 02/04/2023]
Abstract
The enteric nervous system (ENS) of the gastrointestinal (GI) tract interacts with the local immune system bidirectionally. Recent publications have demonstrated that such interactions can maintain normal GI functions during homeostasis and contribute to pathological symptoms during infection and inflammation. Infection can also induce long-term changes of the ENS resulting in the development of post-infectious GI disturbances. In this review, we discuss how the ENS can regulate and be regulated by immune responses and how such interactions control whole tissue physiology. We also address the requirements for the proper regeneration of the ENS and restoration of GI function following the resolution of infection.
Collapse
|
26
|
Fung C, Cools B, Malagola S, Martens T, Tack J, Kazwiny Y, Vanden Berghe P. Luminal short-chain fatty acids and 5-HT acutely activate myenteric neurons in the mouse proximal colon. Neurogastroenterol Motil 2021; 33:e14186. [PMID: 34121274 DOI: 10.1111/nmo.14186] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 04/03/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Gastrointestinal (GI) function is critically dependent on the control of the enteric nervous system (ENS), which is situated within the gut wall and organized into two ganglionated nerve plexuses: the submucosal and myenteric plexus. The ENS is optimally positioned and together with the intestinal epithelium, is well-equipped to monitor the luminal contents such as microbial metabolites and to coordinate appropriate responses accordingly. Despite the heightened interest in the gut microbiota and its influence on intestinal physiology and pathophysiology, how they interact with the host ENS remains unclear. METHODS Using full-thickness proximal colon preparations from transgenic Villin-CreERT2;R26R-GCaMP3 and Wnt1-Cre;R26R-GCaMP3 mice, which express a fluorescent Ca2+ indicator in their intestinal epithelium or in their ENS, respectively, we examined the effects of key luminal microbial metabolites (SCFAs and 5-HT) on the mucosa and underlying enteric neurons. KEY RESULTS We show that the SCFAs acetate, propionate, and butyrate, as well as 5-HT can, to varying extents, acutely elicit epithelial and neuronal Ca2+ responses. Furthermore, SCFAs exert differential effects on submucosal and myenteric neurons. Additionally, we found that submucosal ganglia are predominantly aligned along the striations of the transverse mucosal folds in the proximal colon. CONCLUSIONS & INFERENCES Taken together, our study demonstrates that different microbial metabolites, including SCFAs and 5-HT, can acutely stimulate Ca2+ signaling in the mucosal epithelium and in enteric neurons.
Collapse
Affiliation(s)
- Candice Fung
- Laboratory for Enteric NeuroScience (LENS) Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Bert Cools
- Laboratory for Enteric NeuroScience (LENS) Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Sergio Malagola
- Laboratory for Enteric NeuroScience (LENS) Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Tobias Martens
- Laboratory for Enteric NeuroScience (LENS) Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Youcef Kazwiny
- Laboratory for Enteric NeuroScience (LENS) Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS) Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Singh R, Zogg H, Ro S. Role of microRNAs in Disorders of Gut-Brain Interactions: Clinical Insights and Therapeutic Alternatives. J Pers Med 2021; 11:jpm11101021. [PMID: 34683162 PMCID: PMC8541612 DOI: 10.3390/jpm11101021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Disorders of gut–brain interactions (DGBIs) are heterogeneous in nature and intertwine with diverse pathophysiological mechanisms. Regular functioning of the gut requires complex coordinated interplay between a variety of gastrointestinal (GI) cell types and their functions are regulated by multiple mechanisms at the transcriptional, post-transcriptional, translational, and post-translational levels. MicroRNAs (miRNAs) are small non-coding RNA molecules that post-transcriptionally regulate gene expression by binding to specific mRNA targets to repress their translation and/or promote the target mRNA degradation. Dysregulation of miRNAs might impair gut physiological functions leading to DGBIs and gut motility disorders. Studies have shown miRNAs regulate gut functions such as visceral sensation, gut immune response, GI barrier function, enteric neuronal development, and GI motility. These biological processes are highly relevant to the gut where neuroimmune interactions are key contributors in controlling gut homeostasis and functional defects lead to DGBIs. Although extensive research has explored the pathophysiology of DGBIs, further research is warranted to bolster the molecular mechanisms behind these disorders. The therapeutic targeting of miRNAs represents an attractive approach for the treatment of DGBIs because they offer new insights into disease mechanisms and have great potential to be used in the clinic as diagnostic markers and therapeutic targets. Here, we review recent advances regarding the regulation of miRNAs in GI pacemaking cells, immune cells, and enteric neurons modulating pathophysiological mechanisms of DGBIs. This review aims to assess the impacts of miRNAs on the pathophysiological mechanisms of DGBIs, including GI dysmotility, impaired intestinal barrier function, gut immune dysfunction, and visceral hypersensitivity. We also summarize the therapeutic alternatives for gut microbial dysbiosis in DGBIs, highlighting the clinical insights and areas for further exploration. We further discuss the challenges in miRNA therapeutics and promising emerging approaches.
Collapse
Affiliation(s)
| | | | - Seungil Ro
- Correspondence: ; Tel.: +1-775-784-1462; Fax: +1-775-784-6903
| |
Collapse
|
28
|
Yip JL, Balasuriya GK, Spencer SJ, Hill-Yardin EL. The Role of Intestinal Macrophages in Gastrointestinal Homeostasis: Heterogeneity and Implications in Disease. Cell Mol Gastroenterol Hepatol 2021; 12:1701-1718. [PMID: 34506953 PMCID: PMC8551786 DOI: 10.1016/j.jcmgh.2021.08.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022]
Abstract
Intestinal macrophages play a key role in the gut immune system and the regulation of gastrointestinal physiology, including gut motility and secretion. Their ability to keep the gut from chronic inflammation despite constantly facing foreign antigens has been an important focus in gastrointestinal research. However, the heterogeneity of intestinal macrophages has impeded our understanding of their specific roles. It is now becoming clear that subsets of intestinal macrophages play diverse roles in various gastrointestinal diseases. This occurs through a complex interplay between cytokine production and enteric nervous system activation that differs for each pathologic condition. Key diseases and disorders in which intestinal macrophages play a role include postoperative ileus, inflammatory bowel disease, necrotizing enterocolitis, as well as gastrointestinal disorders associated with human immunodeficiency virus and Parkinson's disease. Here, we review the identification of intestinal macrophage subsets based on their origins and functions, how specific subsets regulate gut physiology, and the potential for these heterogeneous subpopulations to contribute to disease states. Furthermore, we outline the potential for these subpopulations to provide unique targets for the development of novel therapies for these disorders.
Collapse
Affiliation(s)
| | | | - Sarah J. Spencer
- School of Health and Biomedical Sciences,Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Royal Melbourne Instutite of Technology, Melbourne, Victoria, Australia
| | - Elisa L. Hill-Yardin
- School of Health and Biomedical Sciences,Correspondence Address correspondence to: Elisa L. Hill-Yardin, PhD, School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia.
| |
Collapse
|
29
|
Holland AM, Bon-Frauches AC, Keszthelyi D, Melotte V, Boesmans W. The enteric nervous system in gastrointestinal disease etiology. Cell Mol Life Sci 2021; 78:4713-4733. [PMID: 33770200 PMCID: PMC8195951 DOI: 10.1007/s00018-021-03812-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/20/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
A highly conserved but convoluted network of neurons and glial cells, the enteric nervous system (ENS), is positioned along the wall of the gut to coordinate digestive processes and gastrointestinal homeostasis. Because ENS components are in charge of the autonomous regulation of gut function, it is inevitable that their dysfunction is central to the pathophysiology and symptom generation of gastrointestinal disease. While for neurodevelopmental disorders such as Hirschsprung, ENS pathogenesis appears to be clear-cut, the role for impaired ENS activity in the etiology of other gastrointestinal disorders is less established and is often deemed secondary to other insults like intestinal inflammation. However, mounting experimental evidence in recent years indicates that gastrointestinal homeostasis hinges on multifaceted connections between the ENS, and other cellular networks such as the intestinal epithelium, the immune system, and the intestinal microbiome. Derangement of these interactions could underlie gastrointestinal disease onset and elicit variable degrees of abnormal gut function, pinpointing, perhaps unexpectedly, the ENS as a diligent participant in idiopathic but also in inflammatory and cancerous diseases of the gut. In this review, we discuss the latest evidence on the role of the ENS in the pathogenesis of enteric neuropathies, disorders of gut-brain interaction, inflammatory bowel diseases, and colorectal cancer.
Collapse
Affiliation(s)
- Amy Marie Holland
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Ana Carina Bon-Frauches
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Daniel Keszthelyi
- Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM-School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Werend Boesmans
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
- Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
30
|
Margolis KG, Cryan JF, Mayer EA. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021; 160:1486-1501. [PMID: 33493503 PMCID: PMC8634751 DOI: 10.1053/j.gastro.2020.10.066] [Citation(s) in RCA: 430] [Impact Index Per Article: 107.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/07/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
The gut-brain axis plays an important role in maintaining homeostasis. Many intrinsic and extrinsic factors influence signaling along this axis, modulating the function of both the enteric and central nervous systems. More recently the role of the microbiome as an important factor in modulating gut-brain signaling has emerged and the concept of a microbiota-gut-brain axis has been established. In this review, we highlight the role of this axis in modulating enteric and central nervous system function and how this may impact disorders such as irritable bowel syndrome and disorders of mood and affect. We examine the overlapping biological constructs that underpin these disorders with a special emphasis on the neurotransmitter serotonin, which plays a key role in both the gastrointestinal tract and in the brain. Overall, it is clear that although animal studies have shown much promise, more progress is necessary before these findings can be translated for diagnostic and therapeutic benefit in patient populations.
Collapse
Affiliation(s)
- Kara G. Margolis
- Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Irving Medical Center, New York, NY,Corresponding author:
| | - John F. Cryan
- Department of Anatomy & Neuroscience, University College Cork, Ireland, APC Microbiome Ireland, University College Cork, Ireland
| | - Emeran A. Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vachte and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
31
|
Ebselen prevents cigarette smoke-induced gastrointestinal dysfunction in mice. Clin Sci (Lond) 2021; 134:2943-2957. [PMID: 33125061 PMCID: PMC7676466 DOI: 10.1042/cs20200886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/16/2020] [Accepted: 10/30/2020] [Indexed: 11/30/2022]
Abstract
Gastrointestinal (GI) dysfunction is a common comorbidity of chronic obstructive
pulmonary disease (COPD) for which a major cause is cigarette smoking (CS). The
underlying mechanisms and precise effects of CS on gut contractility, however,
are not fully characterised. Therefore, the aim of the present study was to
investigate whether CS impacts GI function and structure in a mouse model of
CS-induced COPD. We also aimed to investigate GI function in the presence of
ebselen, an antioxidant that has shown beneficial effects on lung inflammation
resulting from CS exposure. Mice were exposed to CS for 2 or 6 months. GI
structure was analysed by histology and immunofluorescence. After 2 months of CS
exposure, ex vivo gut motility was analysed using video-imaging
techniques to examine changes in colonic migrating motor complexes (CMMCs). CS
decreased colon length in mice. Mice exposed to CS for 2 months had a higher
frequency of CMMCs and a reduced resting colonic diameter but no change in
enteric neuron numbers. Ten days cessation after 2 months CS reversed CMMC
frequency changes but not the reduced colonic diameter phenotype. Ebselen
treatment reversed the CS-induced reduction in colonic diameter. After 6 months
CS, the number of myenteric nitric-oxide producing neurons was significantly
reduced. This is the first evidence of colonic dysmotility in a mouse model of
CS-induced COPD. Dysmotility after 2 months CS is not due to altered neuron
numbers; however, prolonged CS-exposure significantly reduced enteric neuron
numbers in mice. Further research is needed to assess potential therapeutic
applications of ebselen in GI dysfunction in COPD.
Collapse
|
32
|
Interleukin-6 produced by enteric neurons regulates the number and phenotype of microbe-responsive regulatory T cells in the gut. Immunity 2021; 54:499-513.e5. [PMID: 33691135 DOI: 10.1016/j.immuni.2021.02.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 11/02/2020] [Accepted: 02/05/2021] [Indexed: 12/22/2022]
Abstract
The immune and enteric nervous (ENS) systems monitor the frontier with commensal and pathogenic microbes in the colon. We investigated whether FoxP3+ regulatory T (Treg) cells functionally interact with the ENS. Indeed, microbe-responsive RORγ+ and Helios+ subsets localized in close apposition to nitrergic and peptidergic nerve fibers in the colon lamina propria (LP). Enteric neurons inhibited in vitro Treg (iTreg) differentiation in a cell-contact-independent manner. A screen of neuron-secreted factors revealed a role for interleukin-6 (IL-6) in modulating iTreg formation and their RORγ+ proportion. Colonization of germfree mice with commensals, especially RORγ+ Treg inducers, broadly diminished colon neuronal density. Closing the triangle, conditional ablation of IL-6 in neurons increased total Treg cells but decreased the RORγ+ subset, as did depletion of two ENS neurotransmitters. Our findings suggest a regulatory circuit wherein microbial signals condition neuronal density and activation, thus tuning Treg cell generation and immunological tolerance in the gut.
Collapse
|
33
|
Kushch ММ, Makhotyna DS, Fesenko IA, Savenko MM, Ulianytska AY. Microstructure of the nerve plexus of the muscular membrane of the gut of domestic ducks (Anas platyrhynchos domesticus) of different ages. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
As evidenced by the publications of recent years, contrary to the existing dogma about the immutability of the state of the enteric nervous system during the postnatal period of ontogenesis, the population of intestinal neurons is a dynamic formation, decreasing with age and changing due to the action of environmental factors. The current article presents the results of study of the microscopic structure of the nerve plexus of the muscular membrane of the enteric nervous system of domestic ducks (Anas platyrhynchos domesticus) of the black white–breasted breed, of nine age groups of 1–365 days of age. The topography, number, area of nerve nodes, as well as the density of neurons in them were determined on transverse sections of the duodenum, jejunum, ileum, caecum and rectum. For the purpose of a generalized assessment of the morphofunctional state of the nerve plexuses, two parameters were determined: the average age indicator of the gut and intestines. The average age indicator of a certain structure of each intestine was determined as the arithmetical average of its nine age indicators. The average age indicator of a certain gut structure was determined as the arithmetic average of the average age indicator of the structure of all five intestines. It has been established that the nerve plexus of the muscular membrane (myenteric, plexus Auerbachi) of the gut of domestic ducks, in contrast to mammals, is not located between the layers of the muscular membrane, but in its outer layer. On a transverse section of the gut wall, the myenteric ganglia and cords that connect have a predominantly elliptical shape. Despite a significant increase with age in the diameter and thickness of the gut wall, the total number of myenteric ganglia changed little, increasing or decreasing with varying degrees of reliability relative to the previous age. In the gut of ducks, during the first year of the postnatal period of ontogenesis, the smallest number of myenteric ganglia was found in the cecum, and the largest – in the ileum. The general pattern of the dynamics of the size of the myenteric ganglia of the gut of ducks was an increase in their area with age. Moreover, this indicator reached the greatest value at different ages of ducks: at 30 days of age in the ileum and cecum, at 180 days of age – in the rectum and at 365 days of age – in the duodenum. The smallest area of the myenteric ganglia was found in the jejunum, and the largest – in the duodenum and ileum. The smallest number of neurons in the ganglion was found in the cecum, and the largest – in the rectum, the lowest density of neurons in the ganglion was found in the cecum, and the largest – in the jejunum. The general quantitative pattern of neurons in the ganglion was the decrease in their density with age. Changes in the morphometric parameters of the ganglia of the nerve plexus of the muscular membrane of the ducks’ gut indicate the plasticity of the enteric nervous system, its ability to dynamically respond to the action of factors of the internal and external environment. It is promising to study the state of the submucous nerve plexus, as well as the cellular composition of the population of neurons of the enteric nervous system of domestic and wild poultry.
Collapse
|
34
|
Kang YN, Fung C, Vanden Berghe P. Gut innervation and enteric nervous system development: a spatial, temporal and molecular tour de force. Development 2021; 148:148/3/dev182543. [PMID: 33558316 DOI: 10.1242/dev.182543] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During embryonic development, the gut is innervated by intrinsic (enteric) and extrinsic nerves. Focusing on mammalian ENS development, in this Review we highlight how important the different compartments of this innervation are to assure proper gut function. We specifically address the three-dimensional architecture of the innervation, paying special attention to the differences in development along the longitudinal and circumferential axes of the gut. We review recent information about the formation of both intrinsic innervation, which is fairly well-known, as well as the establishment of the extrinsic innervation, which, despite its importance in gut-brain signaling, has received much less attention. We further discuss how external microbial and nutritional cues or neuroimmune interactions may influence development of gut innervation. Finally, we provide summary tables, describing the location and function of several well-known molecules, along with some newer factors that have more recently been implicated in the development of gut innervation.
Collapse
Affiliation(s)
- Yi-Ning Kang
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven 3000, Belgium
| | - Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven 3000, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven 3000, Belgium
| |
Collapse
|
35
|
Kulkarni S, Kurapati S, Bogunovic M. Neuro-innate immune interactions in gut mucosal immunity. Curr Opin Immunol 2021; 68:64-71. [PMID: 33130386 PMCID: PMC11095515 DOI: 10.1016/j.coi.2020.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 12/14/2022]
Abstract
The gastrointestinal (GI) tract performs a set of vital physiological functions related to food and water consumption. To help regulate these complex physiological processes, the GI tract is innervated by extensive neural networks. The GI tract also serves as the largest immune organ aimed to protect hosts from harmful microbes and toxins ingested with food. It emerges that the enteric nervous and immune systems are highly integrated to optimize digestion while reinforcing immune protection. In this review, we will discuss key cellular players involved in the neuro-immune interactions within the GI mucosa with the focus on the recently uncovered neural pathways that regulate mucosal immunity in a context relevant to GI health and disease.
Collapse
Affiliation(s)
- Subhash Kulkarni
- Department of Medicine, Center for Neurogastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Sravya Kurapati
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States; Penn State Biomedical Sciences Ph.D. Program, Penn State University College of Medicine, Hershey, PA, United States
| | - Milena Bogunovic
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States.
| |
Collapse
|
36
|
Fung C, Vanden Berghe P. Functional circuits and signal processing in the enteric nervous system. Cell Mol Life Sci 2020; 77:4505-4522. [PMID: 32424438 PMCID: PMC7599184 DOI: 10.1007/s00018-020-03543-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/13/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
The enteric nervous system (ENS) is an extensive network comprising millions of neurons and glial cells contained within the wall of the gastrointestinal tract. The major functions of the ENS that have been most studied include the regulation of local gut motility, secretion, and blood flow. Other areas that have been gaining increased attention include its interaction with the immune system, with the gut microbiota and its involvement in the gut-brain axis, and neuro-epithelial interactions. Thus, the enteric circuitry plays a central role in intestinal homeostasis, and this becomes particularly evident when there are faults in its wiring such as in neurodevelopmental or neurodegenerative disorders. In this review, we first focus on the current knowledge on the cellular composition of enteric circuits. We then further discuss how enteric circuits detect and process external information, how these signals may be modulated by physiological and pathophysiological factors, and finally, how outputs are generated for integrated gut function.
Collapse
Affiliation(s)
- Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.
| |
Collapse
|
37
|
Mast Cell Mediators as Pain Triggers in Migraine: Comparison of Histamine and Serotonin in the Activation of Primary Afferents in the Meninges in Rats. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s11055-020-00983-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Lee CYQ, Franks AE, Hill-Yardin EL. Autism-associated synaptic mutations impact the gut-brain axis in mice. Brain Behav Immun 2020; 88:275-282. [PMID: 32485290 DOI: 10.1016/j.bbi.2020.05.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
Interactions between the gut microbiome and the brain affect mood and behaviour in health and disease. Using preclinical animal models, recent discoveries begin to explain how bacteria in the gut influence our mood as well as highlighting new findings relevant to autism. Autism-associated gene mutations known to alter synapse function in the CNS also affect inflammatory response and modify the enteric nervous system resulting in abnormal gastrointestinal motility and structure. Strikingly, these mutations additionally affect the gut microbiome in mice. This review describes the changes in gut physiology and microbiota in mouse models of autism with modified synapse function. The rationale for different regions of the gastrointestinal tract having variable susceptibility to dysfunction is also discussed. To dissect underlying biological mechanisms involving gut-brain axis dysfunction in preclinical models, a range of multidisciplinary approaches are required. This research will provide insights into the role of the gut-brain axis in health and neurodevelopmental disorders including autism.
Collapse
Affiliation(s)
| | - Ashley E Franks
- School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
39
|
Kulkarni S, Kurapati S, Bogunovic M. 'Nervous' Immunity: Walking the Tightrope. Trends Immunol 2020; 41:359-362. [PMID: 32249062 DOI: 10.1016/j.it.2020.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/19/2020] [Indexed: 11/16/2022]
Abstract
There is a major gap in our understanding of how the intestinal immune and nervous systems are integrated to regulate protective adaptations to enteric infections while maintaining tissue homeostasis. Three recent complementary reports published in Cell (2020) provide new mechanistic insights into how this enteric neuro-immune crosstalk may occur.
Collapse
Affiliation(s)
- Subhash Kulkarni
- Department of Medicine, Center for Neurogastroenterology, Johns Hopkins University - School of Medicine, Baltimore, MD, USA.
| | - Sravya Kurapati
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA; Department of Microbiology and Immunology, Penn State University College of Medicine, Hershey, PA, USA
| | - Milena Bogunovic
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
40
|
Sharna SS, Balasuriya GK, Hosie S, Nithianantharajah J, Franks AE, Hill-Yardin EL. Altered Caecal Neuroimmune Interactions in the Neuroligin-3 R451C Mouse Model of Autism. Front Cell Neurosci 2020; 14:85. [PMID: 32327975 PMCID: PMC7160799 DOI: 10.3389/fncel.2020.00085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
The intrinsic nervous system of the gut interacts with the gut-associated lymphoid tissue (GALT) via bidirectional neuroimmune interactions. The caecum is an understudied region of the gastrointestinal (GI) tract that houses a large supply of microbes and is involved in generating immune responses. The caecal patch is a lymphoid aggregate located within the caecum that regulates microbial content and immune responses. People with Autism Spectrum Disorder (ASD; autism) experience serious GI dysfunction, including inflammatory disorders, more frequently than the general population. Autism is a highly prevalent neurodevelopmental disorder defined by the presence of repetitive behavior or restricted interests, language impairment, and social deficits. Mutations in genes encoding synaptic adhesion proteins such as the R451C missense mutation in neuroligin-3 (NL3) are associated with autism and impair synaptic transmission. We previously reported that NL3R451C mice, a well-established model of autism, have altered enteric neurons and GI dysfunction; however, whether the autism-associated R451C mutation alters the caecal enteric nervous system and immune function is unknown. We assessed for gross anatomical changes in the caecum and quantified the proportions of caecal submucosal and myenteric neurons in wild-type and NL3R451C mice using immunofluorescence. In the caecal patch, we assessed total cellular density as well as the density and morphology of Iba-1 labeled macrophages to identify whether the R451C mutation affects neuro-immune interactions. NL3R451C mice have significantly reduced caecal weight compared to wild-type mice, irrespective of background strain. Caecal weight is also reduced in mice lacking Neuroligin-3. NL3R451C caecal ganglia contain more neurons overall and increased numbers of Nitric Oxide (NO) producing neurons (labeled by Nitric Oxide Synthase; NOS) per ganglion in both the submucosal and myenteric plexus. Overall caecal patch cell density was unchanged however NL3R451C mice have an increased density of Iba-1 labeled enteric macrophages. Macrophages in NL3R451C were smaller and more spherical in morphology. Here, we identify changes in both the nervous system and immune system caused by an autism-associated mutation in Nlgn3 encoding the postsynaptic cell adhesion protein, Neuroligin-3. These findings provide further insights into the potential modulation of neural and immune pathways.
Collapse
Affiliation(s)
- Samiha Sayed Sharna
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | | | - Suzanne Hosie
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | | | - Ashley E Franks
- School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
41
|
Qin L, Xiang J, Xiong F, Wang G, Zou H, Li W, Li M, Wu S. Effects of Bacillus licheniformis on the growth, antioxidant capacity, intestinal barrier and disease resistance of grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2020; 97:344-350. [PMID: 31846776 DOI: 10.1016/j.fsi.2019.12.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 06/10/2023]
Abstract
To study the effect of dietary supplementation of Bacillus licheniformis FA6 on the growth, survival and intestinal health of grass carp, we assessed the antioxidant capacity, intestinal barrier, expression levels of immune genes, and the resistance to Aeromonas hydrophila AH-1 infection. Experimental setup comprised three groups (90 specimens each; average initial weight = 16.5 g): the control group was fed the basal diet without B. licheniformis, the low-dose (LD) group was supplemented with B. licheniformis at the concentration of 1 × 105 cfu/g, and the high-dose (HD) group with 1 × 106 cfu/g. After 56 days of growth trial, the challenge test with A. hydrophila AH-1 was conducted for 14 days. The results revealed that the grass carp in LD group and HD group had significantly (p < 0.05) improved percent weight gain (PWG) and specific growth rate (SGR) parameters. Additionally, the antioxidant status was improved, which included increased superoxide dismutase (SOD) activity and decreased malondialdehyde (MDA) levels in the serum, and upregulated mRNA levels of antioxidant enzymes MnSOD and catalase (CAT) in the intestine. Meanwhile, B. licheniformis FA6 supplementation groups exhibited a decreased mRNA expression of proinflammatory cytokines (such as IL-1β and TNF-α) and increased the expression of anti-inflammatory cytokine IL-10. Histological (villi length was increased) and gene expression (qPCR: upregulated ZO-1, occludin, and claudin-c) analyses suggested improved functioning of the intestinal barrier. Post-challenge mortality rates in LD and HD groups were significantly lower (56.6% and 70% respectively) than in the control group (100%). Overall, these results indicated that dietary supplementation of B. licheniformis FA6 can improve growth, antioxidant capacity, intestinal barrier functions and disease resistance of grass carp.
Collapse
Affiliation(s)
- Lu Qin
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinhua Xiang
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fan Xiong
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China
| | - Guitang Wang
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Zou
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenxiang Li
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ming Li
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shangong Wu
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, and State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
42
|
Affiliation(s)
- Luc Maroteaux
- UMR-S839 INSERM, Sorbonne Université, Institut du Fer à Moulin, Paris, France
| | - Fusun Kilic
- Departments of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
43
|
Chesné J, Cardoso V, Veiga-Fernandes H. Neuro-immune regulation of mucosal physiology. Mucosal Immunol 2019; 12:10-20. [PMID: 30089849 DOI: 10.1038/s41385-018-0063-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
Abstract
Mucosal barriers constitute major body surfaces that are in constant contact with the external environment. Mucosal sites are densely populated by a myriad of distinct neurons and immune cell types that sense, integrate and respond to multiple environmental cues. In the recent past, neuro-immune interactions have been reported to play central roles in mucosal health and disease, including chronic inflammatory conditions, allergy and infectious diseases. Discrete neuro-immune cell units act as building blocks of this bidirectional multi-tissue cross-talk, ensuring mucosal tissue health and integrity. Herein, we will focus on reciprocal neuro-immune interactions in the airways and intestine. Such neuro-immune cross-talk maximizes sensing and integration of environmental aggressions, which can be considered an important paradigm shift in our current views of mucosal physiology and immune regulation.
Collapse
Affiliation(s)
- Julie Chesné
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038, Lisboa, Portugal
| | - Vânia Cardoso
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038, Lisboa, Portugal
| | | |
Collapse
|
44
|
Gabriel T, Paul S, Berger A, Massoubre C. Anorexia Nervosa and Autism Spectrum Disorders: Future Hopes Linked to Mucosal Immunity. Neuroimmunomodulation 2019; 26:265-275. [PMID: 31715599 DOI: 10.1159/000502997] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/28/2019] [Indexed: 11/19/2022] Open
Abstract
Mental health is becoming a public health priority worldwide. Anorexia nervosa and autism spectrum disorders are 2 important types of childhood disorders with a bad prognosis. They share cognitive impairments and, in both cases, the microbiota appears to be a crucial factor. Alteration of the microbiota-gut-brain axis is an appealing hypothesis to define new pathophysiological mechanisms. Mucosal immunity plays a key role between the microbiota and the brain. The mucosal immune system receives and integrates messages from the intestinal microenvironment and the microbiota and then transmits the information to the nervous system. Abnormalities in this sensorial system may be involved in the natural history of mental diseases and might play a role in their maintenance. This review aims to highlight data about the relationship between intestinal mucosal immunity and these disorders. We show that shared cognitive impairments could be found in these 2 disorders, which both present dysbiosis. This literature review provides details on the immune status of anorexic and autistic patients, with a focus on intestinal mucosal factors. Finally, we suggest future research hypotheses that seem important for understanding the implication of the gut-brain-axis in psychiatric diseases.
Collapse
Affiliation(s)
- Tristan Gabriel
- Laboratoire d'Immunologie, GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
- Centre Référent des Troubles du Comportement Alimentaire, CHU Saint Etienne Hôpital Nord, Saint-Etienne, France
| | - Stéphane Paul
- Laboratoire d'Immunologie, GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Anne Berger
- Laboratoire d'Immunologie, GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Catherine Massoubre
- Centre Référent des Troubles du Comportement Alimentaire, CHU Saint Etienne Hôpital Nord, Saint-Etienne, France,
| |
Collapse
|
45
|
Kulkarni S, Ganz J, Bayrer J, Becker L, Bogunovic M, Rao M. Advances in Enteric Neurobiology: The "Brain" in the Gut in Health and Disease. J Neurosci 2018; 38:9346-9354. [PMID: 30381426 PMCID: PMC6209840 DOI: 10.1523/jneurosci.1663-18.2018] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/20/2018] [Accepted: 09/22/2018] [Indexed: 12/14/2022] Open
Abstract
The enteric nervous system (ENS) is a large, complex division of the peripheral nervous system that regulates many digestive, immune, hormonal, and metabolic functions. Recent advances have elucidated the dynamic nature of the mature ENS, as well as the complex, bidirectional interactions among enteric neurons, glia, and the many other cell types that are important for mediating gut behaviors. Here, we provide an overview of ENS development and maintenance, and focus on the latest insights gained from the use of novel model systems and live-imaging techniques. We discuss major advances in the understanding of enteric glia, and the functional interactions among enteric neurons, glia, and enteroendocrine cells, a large class of sensory epithelial cells. We conclude by highlighting recent work on muscularis macrophages, a group of immune cells that closely interact with the ENS in the gut wall, and the importance of neurological-immune system communication in digestive health and disease.
Collapse
Affiliation(s)
- Subhash Kulkarni
- Department of Medicine, The John Hopkins University School of Medicine, Baltimore, Maryland 21205,
| | - Julia Ganz
- Department of Integrative Biology, Michigan State University, East Lansing, Michigan 48824
| | - James Bayrer
- Department of Pediatrics, University of California, San Francisco, San Francisco, California 94143
| | - Laren Becker
- Department of Medicine, Stanford University, Stanford, California 94305
| | - Milena Bogunovic
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania 17033, and
| | - Meenakshi Rao
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
46
|
Wang W, Liu Y, Yang Y, Wang A, Sharshov K, Li Y, Cao M, Mao P, Li L. Comparative analyses of the gut microbiota among three different wild geese species in the genus Anser. J Basic Microbiol 2018; 58:543-553. [PMID: 29668076 DOI: 10.1002/jobm.201800060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/27/2018] [Accepted: 04/02/2018] [Indexed: 12/31/2022]
Abstract
In this study, we characterized for the first time the gut microbiota of Greylag geese (Anser anser) using high-throughput 16S rRNA gene sequencing technology. The results showed that the phyla Firmicutes (78.55%), Fusobacteria (9.38%), Proteobacteria (7.55%), Bacteroidetes (1.82%), Cyanobacteria (1.44%), and Actinobacteria (0.61%) dominated the gut microbial communities in the Greylag geese. Then, the variations of gut microbial community structures and functions among the three geese species, Greylag geese, Bar-headed geese (Anser indicus), and Swan geese (Anser cygnoides), were explored. The greatest gut microbial diversity was found in Bar-headed geese group, while other two groups had the least. The dominant bacterial phyla across all samples were Firmicutes and Proteobacteria, but several characteristic bacterial phyla and genera associated with each group were also detected. At all taxonomic levels, the microbial community structure of Swan geese was different from those of Greylag geese and Bar-headed geese, whereas the latter two groups were less different. Functional KEGG categories and pathways associated with carbohydrate metabolism, energy metabolism, and amino acid metabolism were differentially expressed among different geese species. Taken together, this study could provide valuable information to the vast, and yet little explored, research field of wild birds gut microbiome.
Collapse
Affiliation(s)
- Wen Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xi'ning, Qinghai Province, China
| | - Yingbao Liu
- College of Life Science, Yangtze University, Jingzhou, Hubei Province, China
| | - Yongsheng Yang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xi'ning, Qinghai Province, China
| | - Aizhen Wang
- College of Eco-Environmental Engineering, Qinghai University, Xi'ning, Qinghai Province, China
| | - Kirill Sharshov
- Research Institute of Experimental and Clinical Medicine, Novosibirsk, Russia
| | - Yao Li
- College of Eco-Environmental Engineering, Qinghai University, Xi'ning, Qinghai Province, China
| | - Mengyao Cao
- KunLun College of Qinghai University, Xi'ning, Qinghai Province, China
| | - Puzhen Mao
- KunLun College of Qinghai University, Xi'ning, Qinghai Province, China
| | - Laixing Li
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xi'ning, Qinghai Province, China
| |
Collapse
|
47
|
Hirschsprung disease - integrating basic science and clinical medicine to improve outcomes. Nat Rev Gastroenterol Hepatol 2018; 15:152-167. [PMID: 29300049 DOI: 10.1038/nrgastro.2017.149] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hirschsprung disease is defined by the absence of enteric neurons at the end of the bowel. The enteric nervous system (ENS) is the intrinsic nervous system of the bowel and regulates most aspects of bowel function. When the ENS is missing, there are no neurally mediated propulsive motility patterns, and the bowel remains contracted, causing functional obstruction. Symptoms of Hirschsprung disease include constipation, vomiting, abdominal distension and growth failure. Untreated disease usually causes death in childhood because bloodstream bacterial infections occur in the context of bowel inflammation (enterocolitis) or bowel perforation. Current treatment is surgical resection of the bowel to remove or bypass regions where the ENS is missing, but many children have problems after surgery. Although the anatomy of Hirschsprung disease is simple, many clinical features remain enigmatic, and diagnosis and management remain challenging. For example, the age of presentation and the type of symptoms that occur vary dramatically among patients, even though every affected child has missing neurons in the distal bowel at birth. In this Review, basic science discoveries are linked to clinical manifestations of Hirschsprung disease, including partial penetrance, enterocolitis and genetics. Insights into disease mechanisms that might lead to new prevention, diagnostic and treatment strategies are described.
Collapse
|
48
|
Buhner S, Barki N, Greiter W, Giesbertz P, Demir IE, Ceyhan GO, Zeller F, Daniel H, Schemann M. Calcium Imaging of Nerve-Mast Cell Signaling in the Human Intestine. Front Physiol 2017; 8:971. [PMID: 29238306 PMCID: PMC5712982 DOI: 10.3389/fphys.2017.00971] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/14/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction: It is suggested that an altered microenvironment in the gut wall alters communication along a mast cell nerve axis. We aimed to record for the first time signaling between mast cells and neurons in intact human submucous preparations. Methods: We used the Ca2+ sensitive dye Fluo-4 AM to simultaneously image changes in intracellular calcium [Ca+2]i (%ΔF/F) in neurons and mast cells. Data are presented as median with interquartile ranges (25/75%). Results: We recorded nerve responses in 29 samples upon selective activation of 223 mast cells by IgE receptor cross linking with the antibody mAb22E7. Mast cells responded to mAb22E7 with a median [Ca+2]i increase of 20% (11/39) peaking 90 s (64/144) after the application. Only very few neurons responded and the median percentage of responding neuronal area was 0% (0/5.9). Mast cell activation remained in the presence of the fast sodium channel blocker tetrodotoxin. Specific neuronal activation by transmural electrical field stimulation (EFS) in 34 samples evoked instantaneously [Ca+2]i signals in submucous neurons. This was followed by a [Ca+2]i peak response of 8%ΔF/F (4/15) in 33% of 168 mast cells in the field of view. The mast cell response was abolished by the nerve blocker tetrododoxin, reduced by the Calcitonin Gene-Related Peptide receptor 1 antagonist BIBN-4096 and the Vasoactive Intestinal Peptide receptor antagonist PG97-269, but not by blockade of the neurokinin receptors 1-3. Conclusion: The findings revealed bidirectional signaling between mast cells and submucous neurons in human gut. In our macroscopically normal preparations a nerve to mast cell signaling was very prominent whereas a mast cell to nerve signaling was rather rare.
Collapse
Affiliation(s)
- Sabine Buhner
- Human Biology, Technische Universität München, Freising, Germany
| | - Natasja Barki
- Human Biology, Technische Universität München, Freising, Germany
| | - Wolfgang Greiter
- Human Biology, Technische Universität München, Freising, Germany
| | - Pieter Giesbertz
- Molecular Nutrition Unit, Technische Universität München, Freising, Germany
| | - Ihsan E. Demir
- Department of General Surgery, University Hospital Rechts der Isar, Technische Universität München, Munich, Germany
| | - Güralp O. Ceyhan
- Department of General Surgery, University Hospital Rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Hannelore Daniel
- Molecular Nutrition Unit, Technische Universität München, Freising, Germany
| | - Michael Schemann
- Human Biology, Technische Universität München, Freising, Germany
| |
Collapse
|
49
|
Abstract
The evolution of a nervous system as a control system of the body's functions is a key innovation of animals. Its fundamental units are neurons, highly specialized cells dedicated to fast cell-cell communication. Neurons pass signals to other neurons, muscle cells, or gland cells at specialized junctions, the synapses, where transmitters are released from vesicles in a Ca2+-dependent fashion to activate receptors in the membrane of the target cell. Reconstructing the origins of neuronal communication out of a more simple process remains a central challenge in biology. Recent genomic comparisons have revealed that all animals, including the nerveless poriferans and placozoans, share a basic set of genes for neuronal communication. This suggests that the first animal, the Urmetazoan, was already endowed with neurosecretory cells that probably started to connect into neuronal networks soon afterward. Here, we discuss scenarios for this pivotal transition in animal evolution.
Collapse
Affiliation(s)
- Frederique Varoqueaux
- Département des Neurosciences Fondamentales, Université de Lausanne, Lausanne, CH-1005 Switzerland; ,
| | - Dirk Fasshauer
- Département des Neurosciences Fondamentales, Université de Lausanne, Lausanne, CH-1005 Switzerland; ,
| |
Collapse
|
50
|
Wekerle H. Brain Autoimmunity and Intestinal Microbiota: 100 Trillion Game Changers. Trends Immunol 2017; 38:483-497. [DOI: 10.1016/j.it.2017.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 02/17/2017] [Accepted: 03/31/2017] [Indexed: 02/07/2023]
|