1
|
Breßer M, Siemens KD, Schneider L, Lunnebach JE, Leven P, Glowka TR, Oberländer K, De Domenico E, Schultze JL, Schmidt J, Kalff JC, Schneider A, Wehner S, Schneider R. Macrophage-induced enteric neurodegeneration leads to motility impairment during gut inflammation. EMBO Mol Med 2025; 17:301-335. [PMID: 39762650 PMCID: PMC11822118 DOI: 10.1038/s44321-024-00189-w] [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: 05/14/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
Current studies pictured the enteric nervous system and macrophages as modulators of neuroimmune processes in the inflamed gut. Expanding this view, we investigated the impact of enteric neuron-macrophage interactions on postoperative trauma and subsequent motility disturbances, i.e., postoperative ileus. In the early postsurgical phase, we detected strong neuronal activation, followed by transcriptional and translational signatures indicating neuronal death and synaptic damage. Simultaneously, our study revealed neurodegenerative profiles in macrophage-specific transcriptomes after postoperative trauma. Validating the role of resident and monocyte-derived macrophages, we depleted macrophages by CSF-1R-antibodies and used CCR2-/- mice, known for reduced monocyte infiltration, in POI studies. Only CSF-1R-antibody-treated animals showed decreased neuronal death and lessened synaptic decay, emphasizing the significance of resident macrophages. In human gut samples taken early and late during abdominal surgery, we substantiated the mouse model data and found reactive and apoptotic neurons and dysregulation in synaptic genes, indicating a species' overarching mechanism. Our study demonstrates that surgical trauma activates enteric neurons and induces neurodegeneration, mediated by resident macrophages, introducing neuroprotection as an option for faster recovery after surgery.
Collapse
Affiliation(s)
- Mona Breßer
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kevin D Siemens
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Patrick Leven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Tim R Glowka
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristin Oberländer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Elena De Domenico
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim Schmidt
- Department of General, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
2
|
Santhosh S, Zanoletti L, Stamp LA, Hao MM, Matteoli G. From diversity to disease: unravelling the role of enteric glial cells. Front Immunol 2024; 15:1408744. [PMID: 38957473 PMCID: PMC11217337 DOI: 10.3389/fimmu.2024.1408744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 07/04/2024] Open
Abstract
Enteric glial cells (EGCs) are an essential component of the enteric nervous system (ENS) and play key roles in gastrointestinal development, homeostasis, and disease. Derived from neural crest cells, EGCs undergo complex differentiation processes regulated by various signalling pathways. Being among the most dynamic cells of the digestive system, EGCs react to cues in their surrounding microenvironment and communicate with various cell types and systems within the gut. Morphological studies and recent single cell RNA sequencing studies have unveiled heterogeneity among EGC populations with implications for regional functions and roles in diseases. In gastrointestinal disorders, including inflammatory bowel disease (IBD), infections and cancer, EGCs modulate neuroplasticity, immune responses and tumorigenesis. Recent evidence suggests that EGCs respond plastically to the microenvironmental cues, adapting their phenotype and functions in disease states and taking on a crucial role. They exhibit molecular abnormalities and alter communication with other intestinal cell types, underscoring their therapeutic potential as targets. This review delves into the multifaceted roles of EGCs, particularly emphasizing their interactions with various cell types in the gut and their significant contributions to gastrointestinal disorders. Understanding the complex roles of EGCs in gastrointestinal physiology and pathology will be crucial for the development of novel therapeutic strategies for gastrointestinal disorders.
Collapse
Affiliation(s)
- Sneha Santhosh
- Department of Chronic Diseases, Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa Zanoletti
- Department of Chronic Diseases, Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Lincon A. Stamp
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Marlene M. Hao
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism (CHROMETA), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Leuven Institute for Single-cell Omics (LISCO), KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Varley AN, Browning KN. Gastrointestinal dysfunction in the valproic acid induced model of social deficit in rats. Auton Neurosci 2024; 253:103161. [PMID: 38461695 PMCID: PMC11128350 DOI: 10.1016/j.autneu.2024.103161] [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: 12/06/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
Autism spectrum disorder (ASD) has increased in incidence over the past several decades, and is associated with a range of co-morbidities including gastrointestinal (GI) dysfunctions including gastroesophageal reflux, abdominal pain, bloating, constipation and/or diarrhea. Several animal models have been used that replicate several aspects of ASD but no single model has been able to replicate the entire disease pathophysiology. In humans, prenatal exposure to valproic acid (VPA) has been identified as a significant risk factor and rodent models have shown that in utero VPA exposure leads to behavioral deficits in offspring. The present study aimed to investigate whether in utero exposure to VPA induces GI dysfunction in rats. Timed pregnant Sprague-Dawley rats were injected with a single dose of VPA at embryonic day 12.5. Both male and female offspring subsequently underwent behavioral studies and assessment of GI function in adulthood. In utero VPA treatment induced social deficits in both male and female offspring, decreasing sociability and social novelty. Histological examination showed that VPA treated offspring had decreased thickness of GI muscle and mucosa, while immunohistochemical studies showed a decrease in myenteric neuron number in the fundus. Functional studies showed that both male and female VPA offspring had a delay in gastric emptying compared to vehicle treated offspring. Results of the current study suggest that the rat VPA model of behavioral deficits may be a convenient model by which both mechanistic and functional insights into GI dysfunction may be studied.
Collapse
Affiliation(s)
- Ashley N Varley
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, PA, United States of America
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States of America.
| |
Collapse
|
4
|
Truong Thuy Nguyen V, Taheri N, Choi EL, Kellogg TA, Linden DR, Hayashi Y. Insulin-Like Growth Factor1 Preserves Gastric Pacemaker Cells and Motor Function in Aging via ERK1/2 Activation. Cell Mol Gastroenterol Hepatol 2023; 16:369-383. [PMID: 37301443 PMCID: PMC10372898 DOI: 10.1016/j.jcmgh.2023.06.002] [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: 11/29/2022] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND & AIMS Impaired gastric motor function in the elderly causes reduced food intake leading to frailty and sarcopenia. We previously found that aging-related impaired gastric compliance was mainly owing to depletion of interstitial cells of Cajal (ICC), pacemaker cells, and neuromodulator cells. These changes were associated with reduced food intake. Transformation-related protein 53-induced suppression of extracellular signal-regulated protein kinase (ERK)1/2 in ICC stem cell (ICC-SC) cell-cycle arrest is a key process for ICC depletion and gastric dysfunction during aging. Here, we investigated whether insulin-like growth factor 1 (IGF1), which can activate ERK in gastric smooth muscles and invariably is reduced with age, could mitigate ICC-SC/ICC loss and gastric dysfunction in klotho mice, a model of accelerated aging. METHODS Klotho mice were treated with the stable IGF1 analog LONG R3 recombinant human (rh) IGF1 (150 μg/kg intraperitoneally twice daily for 3 weeks). Gastric ICC/ICC-SC and signaling pathways were studied by flow cytometry, Western blot, and immunohistochemistry. Gastric compliance was assessed in ex vivo systems. Transformation-related protein 53 was induced with nutlin 3a and ERK1/2 signaling was activated by rhIGF-1 in the ICC-SC line. RESULTS LONG R3 rhIGF1 treatment prevented reduced ERK1/2 phosphorylation and gastric ICC/ICC-SC decrease. LONG R3 rhIGF1 also mitigated the reduced food intake and impaired body weight gain. Improved gastric function by LONG R3 rhIGF1 was verified by in vivo systems. In ICC-SC cultures, rhIGF1 mitigated nutlin 3a-induced reduced ERK1/2 phosphorylation and cell growth arrest. CONCLUSIONS IGF1 can mitigate age-related ICC/ICC-SC loss by activating ERK1/2 signaling, leading to improved gastric compliance and increased food intake in klotho mice.
Collapse
Affiliation(s)
- Vy Truong Thuy Nguyen
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Negar Taheri
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Egan L Choi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Todd A Kellogg
- Department of Surgery, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - David R Linden
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Yujiro Hayashi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota.
| |
Collapse
|
5
|
López-Pingarrón L, Almeida H, Soria-Aznar M, Reyes-Gonzales MC, Rodríguez-Moratinos AB, Muñoz-Hoyos A, García JJ. Interstitial Cells of Cajal and Enteric Nervous System in Gastrointestinal and Neurological Pathology, Relation to Oxidative Stress. Curr Issues Mol Biol 2023; 45:3552-3572. [PMID: 37185756 PMCID: PMC10136929 DOI: 10.3390/cimb45040232] [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: 03/11/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The enteric nervous system (ENS) is organized into two plexuses-submucosal and myenteric-which regulate smooth muscle contraction, secretion, and blood flow along the gastrointestinal tract under the influence of the rest of the autonomic nervous system (ANS). Interstitial cells of Cajal (ICCs) are mainly located in the submucosa between the two muscle layers and at the intramuscular level. They communicate with neurons of the enteric nerve plexuses and smooth muscle fibers and generate slow waves that contribute to the control of gastrointestinal motility. They are also involved in enteric neurotransmission and exhibit mechanoreceptor activity. A close relationship appears to exist between oxidative stress and gastrointestinal diseases, in which ICCs can play a prominent role. Thus, gastrointestinal motility disorders in patients with neurological diseases may have a common ENS and central nervous system (CNS) nexus. In fact, the deleterious effects of free radicals could affect the fine interactions between ICCs and the ENS, as well as between the ENS and the CNS. In this review, we discuss possible disturbances in enteric neurotransmission and ICC function that may cause anomalous motility in the gut.
Collapse
Affiliation(s)
- Laura López-Pingarrón
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Henrique Almeida
- i3S-Instituto de Investigação e Inovação em Saúde, Porto University, 4200-135 Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
- Department of Obstetrics and Gynecology, Hospital-CUF Porto, 4100-180 Porto, Portugal
| | - Marisol Soria-Aznar
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Marcos C Reyes-Gonzales
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | | | - Antonio Muñoz-Hoyos
- Department of Pediatrics, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Joaquín J García
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
6
|
Xia Y, Prokop S, Bell BM, Gorion KMM, Croft CL, Nasif L, Xu G, Riffe CJ, Manaois AN, Strang KH, Quintin SS, Paterno G, Tansey MG, Borchelt DR, Golde TE, Giasson BI. Pathogenic tau recruits wild-type tau into brain inclusions and induces gut degeneration in transgenic SPAM mice. Commun Biol 2022; 5:446. [PMID: 35550593 PMCID: PMC9098443 DOI: 10.1038/s42003-022-03373-1] [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: 12/01/2021] [Accepted: 04/14/2022] [Indexed: 01/04/2023] Open
Abstract
Pathological tau inclusions are neuropathologic hallmarks of many neurodegenerative diseases. We generated and characterized a transgenic mouse model expressing pathogenic human tau with S320F and P301S aggregating mutations (SPAM) at transgene levels below endogenous mouse tau protein levels. This mouse model develops a predictable temporal progression of tau pathology in the brain with biochemical and ultrastructural properties akin to authentic tau inclusions. Surprisingly, pathogenic human tau extensively recruited endogenous mouse tau into insoluble aggregates. Despite the early onset and rapid progressive nature of tau pathology, major neuroinflammatory and transcriptional changes were only detectable at later time points. Moreover, tau SPAM mice are the first model to develop loss of enteric neurons due to tau accumulation resulting in a lethal phenotype. With moderate transgene expression, rapidly progressing tau pathology, and a highly predictable lethal phenotype, the tau SPAM model reveals new associations of tau neurotoxicity in the brain and intestinal tract.
Collapse
Affiliation(s)
- Yuxing Xia
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Stefan Prokop
- grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Department of Pathology, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Brach M. Bell
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Kimberly-Marie M. Gorion
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Cara L. Croft
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Lith Nasif
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Guilian Xu
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Cara J. Riffe
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Alyssa N. Manaois
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Kevin H. Strang
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Stephan S. Quintin
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Giavanna Paterno
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Malú Gámez Tansey
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - David R. Borchelt
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Todd E. Golde
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| | - Benoit I. Giasson
- grid.15276.370000 0004 1936 8091Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610 USA ,grid.15276.370000 0004 1936 8091McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610 USA
| |
Collapse
|
7
|
Zizzo MG, Cicio A, Raimondo S, Alessandro R, Serio R. Age-related differences of γ-aminobutyric acid (GABA)ergic transmission in human colonic smooth muscle. Neurogastroenterol Motil 2022; 34:e14248. [PMID: 34432349 PMCID: PMC9285353 DOI: 10.1111/nmo.14248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/10/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Enteric neurons undergo to functional changes during aging. We investigated the possible age-associated differences in enteric γ-aminobutyric acid (GABA)ergic transmission evaluating function and distribution of GABAergic receptors in human colon. METHODS Mechanical responses to GABA and GABA receptor agonists on slow phasic contractions were examined in vitro as changes in isometric tension in colonic muscle strips from young (<65 years old) and aged patients (>65 years old). GABAergic receptor expression was assessed by quantitative RT-PCR. KEY RESULTS In both preparations GABA induced an excitatory effect, consisting in an increase in the basal tone, antagonized by the GABAA receptor antagonist, bicuculline, and potentiated by phaclofen, GABAB receptor antagonist.Tetrodotoxin (TTX) and atropine-sensitive contractile responses to GABA and GABAA receptor agonist, muscimol, were more pronounced in old compared to young subjects. Baclofen, GABAB receptor agonist, induced a TTX-sensitive reduction of the amplitude of the spontaneous. Nω-nitro-l-arginine methyl ester (L-NAME), nitric oxide (NO) synthase inhibitor abolished the inhibitory responses in old preparations, but a residual responses persisted in young preparations, which in turn was abolished by suramin, purinergic receptor antagonist. α3-GABAA receptor subunit expression tends to change in an age-dependent manner. CONCLUSIONS AND INFERENCES Our results reveal age-related differences in GABAergic transmission in human colon. At all the age tested GABA regulates muscular contractility modulating the activity of the intrinsic neurons. Activation of GABAA receptor, through acetylcholine release, induces contraction, which increases in amplitude with age. GABAB receptor activation leads to neural release of NO and purines, being a loss of purinergic-component in aged group.
Collapse
Affiliation(s)
- Maria Grazia Zizzo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF)University of PalermoViale delle Scienze,ed 16Palermo90128Italy,ATeN (Advanced Technologies Network) CenterUniversity of PalermoViale delle Scienze, ed 18Palermo90128Italy
| | - Adele Cicio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF)University of PalermoViale delle Scienze,ed 16Palermo90128Italy
| | - Stefania Raimondo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D)University of PalermoSection of Biology and GeneticsPalermo90133Italy
| | - Riccardo Alessandro
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bi.N.D)University of PalermoSection of Biology and GeneticsPalermo90133Italy
| | - Rosa Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF)University of PalermoViale delle Scienze,ed 16Palermo90128Italy
| |
Collapse
|
8
|
Gau J, Patel P, Pan J, Kao T. Analyzing fecal loading and retention patterns by abdominal X-rays of hospitalized older adults: A retrospective study. Aging Med (Milton) 2022; 5:38-44. [PMID: 35309161 PMCID: PMC8917260 DOI: 10.1002/agm2.12199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 12/17/2022] Open
Abstract
Background Aging may affect ascending colon (AC) differently from descending colon (DC) and increase the risk of fecal loading (FL) in AC. Methods Patients aged ≥65 years admitted to a community hospital were analyzed by abdominal x-ray for fecal loads and stool retention patterns. FL was scored between 0 and 5 (severe) on each segment of colon with a possible total score 20. Mean segment scores ≥3.5 were designated as high scores for both AC and DC. Logistic regression was performed between groups to identify factors associated with FL patterns. Results Groups identified were high FL in both AC and DC (N = 21, 17.2%), FL predominantly in AC (N = 38, 31.1%), low FL in both AC and DC (N=60, 49.2%), and FL low in AC and high in DC (N = 3, 2.5%). Among 71 patients with total FL scores ≥13 (indicating significant stool retention), 37 (52.1%) had the FL predominantly in AC. Patients prescribed antibiotic(s) prior to hospitalization had lower odds of FL predominantly in AC (adjusted odds ratio = 0.18, 95% confidence interval = 0.04-0.84) compared to the group of low FL in both AC and DC with the adjustment of confounders. Conclusion This study found that 52.1% of those with significant stool retention on x-ray had the FL predominantly in AC. Antibiotic use was associated with lower odds of having FL predominately in AC. This study provided insights of FL distribution in colon and AC could be an area for significant stool burden in older adults with stool retention.
Collapse
Affiliation(s)
- Jen‐Tzer Gau
- Department of Primary Care/geriatric medicineOhio University Heritage College of Osteopathic Medicine (OU‐HCOM)AthensOhioUSA
- Ohio University Heritage College of Osteopathic Medicine (OU‐HCOM)AthensOhioUSA
| | - Parth Patel
- Ohio University Heritage College of Osteopathic Medicine (OU‐HCOM)AthensOhioUSA
- Present address:
SUNY Upstate Medical University HospitalSyracuseNew YorkUSA
| | - Jen‐Jung Pan
- Gastroenterology, Hepatology and Liver TransplantationBanner‐University Medical Center PhoenixPhoenixArizonaUSA
| | - Tzu‐Cheg Kao
- Uniformed Services University of the Health SciencesBethesdaMarylandUSA
| |
Collapse
|
9
|
Special Considerations for the Management of Disorders of Gut-Brain Interaction in Older Adults. CURRENT TREATMENT OPTIONS IN GASTROENTEROLOGY 2022; 20:582-593. [PMID: 36406807 PMCID: PMC9652122 DOI: 10.1007/s11938-022-00403-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 11/13/2022]
Abstract
Purpose of Review The world's population is aging rapidly, with 40% of patients seen in US gastroenterology (GI) clinics being 60 years or older. Many gastrointestinal problems are more common or unique to the older adult because of progressive damage to the structure and function of the GI tract. Until recently, the epidemiology of disorders of gut-brain interaction (such as irritable bowel syndrome and functional dyspepsia) was not well-characterized. Recent Findings Forty percent of persons worldwide have disorders of gut-brain interaction (DGBI), with varying global patterns of incidence in older adults. There are multiple first-line approaches to managing DGBI which can also be combined including pharmacologic (e.g., neuromodulators) and nonpharmacologic approaches including dietary therapies and brain-gut behavioral therapies. However, there are considerations clinicians must account for when offering each approach related to unique biopsychosocial factors in the older adult population. In this review, we aim to critically review recent literature on the pathophysiology, epidemiology, and special considerations for diagnosing and managing DGBI in the older adult population. Summary There have been many advances in the management of DGBI over the past decades. Given the increase in the number of older adults in the USA and worldwide, there is an urgent need for evidence-based guidance to help providers guide comprehensive care for specifically our aging patient population with respect to DGBI.
Collapse
|
10
|
Seguella L, Gulbransen BD. Enteric glial biology, intercellular signalling and roles in gastrointestinal disease. Nat Rev Gastroenterol Hepatol 2021; 18:571-587. [PMID: 33731961 PMCID: PMC8324524 DOI: 10.1038/s41575-021-00423-7] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
One of the most transformative developments in neurogastroenterology is the realization that many functions normally attributed to enteric neurons involve interactions with enteric glial cells: a large population of peripheral neuroglia associated with enteric neurons throughout the gastrointestinal tract. The notion that glial cells function solely as passive support cells has been refuted by compelling evidence that demonstrates that enteric glia are important homeostatic cells of the intestine. Active signalling mechanisms between enteric glia and neurons modulate gastrointestinal reflexes and, in certain circumstances, function to drive neuroinflammatory processes that lead to long-term dysfunction. Bidirectional communication between enteric glia and immune cells contributes to gastrointestinal immune homeostasis, and crosstalk between enteric glia and cancer stem cells regulates tumorigenesis. These neuromodulatory and immunomodulatory roles place enteric glia in a unique position to regulate diverse gastrointestinal disease processes. In this Review, we discuss current concepts regarding enteric glial development, heterogeneity and functional roles in gastrointestinal pathophysiology and pathophysiology, with a focus on interactions with neurons and immune cells. We also present a working model to differentiate glial states based on normal function and disease-induced dysfunctions.
Collapse
Affiliation(s)
- Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Brian D Gulbransen
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
11
|
Guimaraes de Souza Melo C, Nelisis Zanoni J, Raquel Garcia de Souza S, Zignani I, de Lima Leite A, Domingues Heubel A, Vanessa Colombo Martins Perles J, Afonso Rabelo Buzalaf M. Global Proteomic Profile Integrated to Quantitative and Morphometric Assessment of Enteric Neurons: Investigation of the Mechanisms Involved in the Toxicity Induced by Acute Fluoride Exposure in the Duodenum. Neurotox Res 2021; 39:800-814. [PMID: 33689147 DOI: 10.1007/s12640-020-00296-9] [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: 06/22/2020] [Revised: 09/20/2020] [Accepted: 10/11/2020] [Indexed: 10/21/2022]
Abstract
The enteric nervous system is responsible for controlling the gastrointestinal tract (GIT) functions. Enteric neuropathies are highly correlated to the development of several intestinal disturbances. Fluoride (F) is extensively applied for dental health improvement and its ingestion can promote systemic toxicity with mild to severe GIT symptomatology and neurotoxicity. Although F harmful effects have been published, there is no information regarding noxiousness of a high acute F exposure (25 mg F/kg) on enteric neurons and levels of expression of intestinal proteins in the duodenum. Quantitative proteomics of the duodenum wall associated to morphometric and quantitative analysis of enteric neurons displayed F effects of a high acute exposure. F-induced myenteric neuroplasticity was characterized by a decrease in the density of nitrergic neurons and morphometric alterations in the general populations of neurons, nitrergic neurons, and substance P varicosities. Proteomics demonstrated F-induced alterations in levels of expression of 356 proteins correlated to striated muscle cell differentiation; generation of precursor metabolites and energy; NADH and glutathione metabolic process and purine ribonucleoside triphosphate biosynthesis. The neurochemical role of several intestinal proteins was discussed specially related to the modulation of enteric neuroplasticity. The results provide a new perspective on cell signaling pathways of gastrointestinal symptomatology promoted by acute F toxicity.
Collapse
Affiliation(s)
| | | | | | - Isabela Zignani
- Department of Morphophysiological Sciences, State University of Maringá, Paraná, Brazil
| | - Aline de Lima Leite
- Department of Biological Sciences, School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | | | | |
Collapse
|
12
|
Derkinderen P, Rolli-Derkinderen M, Chapelet G, Neunlist M, Noble W. Tau in the gut, does it really matter? J Neurochem 2021; 158:94-104. [PMID: 33569813 DOI: 10.1111/jnc.15320] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022]
Abstract
The enteric nervous system plays a critical role in the regulation of gastrointestinal tract functions and is often referred to as the 'second brain' because it shares many features with the central nervous system. These similarities include among others a large panel of neurotransmitters, a large population of glial cells and a susceptibility to neurodegeneration. This close homology between the central and enteric nervous systems suggests that a disease process affecting the central nervous system could also involve its enteric counterpart. This was already documented in Parkinson's disease, the most common synucleinopathy, in which alpha-synuclein deposits are reported in the enteric nervous system in the vast majority of patients. Tau is another key protein involved in neurodegenerative disorders of the brain. Whether changes in tau also occur in the enteric nervous system during gut or brain disorders has just begun to be explored. The scope of the present article is therefore to review existing studies on the expression and phosphorylation pattern of tau in the enteric nervous system under physiological and pathological conditions and to discuss the possible occurrence of 'enteric tauopathies'.
Collapse
Affiliation(s)
- Pascal Derkinderen
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France.,Department of Neurology, CHU Nantes, Nantes, France
| | - Malvyne Rolli-Derkinderen
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Guillaume Chapelet
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France.,Clinical Gerontology Department, CHU Nantes, Nantes, France
| | - Michel Neunlist
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Université de Nantes, Nantes, France
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| |
Collapse
|
13
|
Sinagra E, Pellegatta G, Maida M, Rossi F, Conoscenti G, Pallio S, Alloro R, Raimondo D, Anderloni A. Could Chronic Idiopatic Intestinal Pseudo-Obstruction Be Related to Viral Infections? J Clin Med 2021; 10:268. [PMID: 33450988 PMCID: PMC7828444 DOI: 10.3390/jcm10020268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic idiopathic intestinal pseudo-obstruction (CIIPO) is a disease characterized by symptoms and signs of small bowel obstruction in the absence of displayable mechanical obstruction. Due to the known neuropathic capacity of several viruses, and their localization in the intestine, it has been hypothesized that such viruses could be involved in the pathogenesis of CIIPO. The most frequently involved viruses are John Cunningham virus, Herpesviridae, Flaviviruses, Epstein-Barr virus and Citomegalovirus. Therefore, the present narrative review aims to sum up some new perspectives in the etiology and pathophysiology of CIIPO.
Collapse
Affiliation(s)
- Emanuele Sinagra
- Gastroenterology and Endoscopy Unit, Fondazione Istituto San Raffaele-Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy; (F.R.); (G.C.); (D.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Division of Gastroenterology, Humanitas Clinical and Research Center (IRCCS), 20089 Rozzano, Italy; (G.P.); (A.A.)
| | - Marcello Maida
- Gastroenterology and Endoscopy Unit, S. Elia-Raimondi Hospital, 93100 Caltanissetta, Italy;
| | - Francesca Rossi
- Gastroenterology and Endoscopy Unit, Fondazione Istituto San Raffaele-Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy; (F.R.); (G.C.); (D.R.)
| | - Giuseppe Conoscenti
- Gastroenterology and Endoscopy Unit, Fondazione Istituto San Raffaele-Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy; (F.R.); (G.C.); (D.R.)
| | - Socrate Pallio
- Endoscopy Unit, University Hospital Policlinic G. Martino, 98125 Messina, Italy;
| | - Rita Alloro
- Division of General and Oncologic Surgery, Department of Surgical, Oncological and Oral Sciences (DICHIRONS), University of Palermo, 90133 Palermo, Italy;
| | - Dario Raimondo
- Gastroenterology and Endoscopy Unit, Fondazione Istituto San Raffaele-Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy; (F.R.); (G.C.); (D.R.)
| | - Andrea Anderloni
- Digestive Endoscopy Unit, Division of Gastroenterology, Humanitas Clinical and Research Center (IRCCS), 20089 Rozzano, Italy; (G.P.); (A.A.)
| |
Collapse
|
14
|
Sohrabi M, Pecoraro HL, Combs CK. Gut Inflammation Induced by Dextran Sulfate Sodium Exacerbates Amyloid-β Plaque Deposition in the AppNL-G-F Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2021; 79:1235-1255. [PMID: 33427741 PMCID: PMC8122495 DOI: 10.3233/jad-201099] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although it is known that the brain communicates with the gastrointestinal (GI) tract via the well-established gut-brain axis, the influence exerted by chronic intestinal inflammation on brain changes in Alzheimer's disease (AD) is not fully understood. We hypothesized that increased gut inflammation would alter brain pathology of a mouse model of AD. OBJECTIVE Determine whether colitis exacerbates AD-related brain changes. METHODS To test this idea, 2% dextran sulfate sodium (DSS) was dissolved in the drinking water and fed ad libitum to male C57BL/6 wild type and AppNL-G-F mice at 6-10 months of age for two cycles of three days each. DSS is a negatively charged sulfated polysaccharide which results in bloody diarrhea and weight loss, changes similar to human inflammatory bowel disease (IBD). RESULTS Both wild type and AppNL-G-F mice developed an IBD-like condition. Brain histologic and biochemical assessments demonstrated increased insoluble Aβ1-40/42 levels along with the decreased microglial CD68 immunoreactivity in DSS treated AppNL-G-F mice compared to vehicle treated AppNL-G-F mice. CONCLUSION These data demonstrate that intestinal dysfunction is capable of altering plaque deposition and glial immunoreactivity in the brain. This study increases our knowledge of the impact of peripheral inflammation on Aβ deposition via an IBD-like model system.
Collapse
Affiliation(s)
- Mona Sohrabi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND
| | - Heidi L. Pecoraro
- Veterinary Diagnostic Laboratory, North Dakota State University, Fargo ND
| | - Colin K. Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND
| |
Collapse
|
15
|
Seifi M, Swinny JD. Developmental and age-dependent plasticity of GABA A receptors in the mouse colon: Implications in colonic motility and inflammation. Auton Neurosci 2019; 221:102579. [PMID: 31445405 DOI: 10.1016/j.autneu.2019.102579] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/15/2019] [Accepted: 08/10/2019] [Indexed: 12/16/2022]
Abstract
Lifelong functional plasticity of the gastrointestinal (GI) tract is essential for health, yet the underlying molecular mechanisms are poorly understood. The enteric nervous system (ENS) regulates all aspects of the gut function, via a range of neurotransmitter pathways, one of which is the GABA-GABAA receptor (GABAAR) system. We have previously shown that GABAA receptor subunits are differentially expressed within the ENS and are involved in regulating various GI functions. We have also shown that these receptors are involved in mediating stress-induced colonic inflammation. However, the expression and function of intestinal GABAARs, at different ages, is largely unexplored and was the focus of this study. Here we show that the impact of GABAAR activation on colonic contractility changes from early postnatal period through to late adulthood, in an age-dependant manner. We also show that the highest levels of expression for all GABAAR subunits is evident at postnatal day (P) 10 apart from the α3 subunit which increased with age. This increase in the α3 subunit expression in late adulthood (18 months old) is accompanied by an increase in the expression of inflammatory markers within the mouse colon. Finally, we demonstrate that the deletion of the α3 subunit prevents the increase in the expression of colonic inflammatory markers associated with healthy ageing. Collectively, the data provide the first demonstration of the molecular and functional plasticity of the GI GABAAR system over the course of a lifetime, and its possible role in mediating the age-induced colonic inflammation associated with healthy ageing.
Collapse
Affiliation(s)
- Mohsen Seifi
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, PO1 2DT, UK; School of Sport, Health and Social Scinces, Solent University, SO14 0YN, UK.
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, PO1 2DT, UK
| |
Collapse
|
16
|
Kim JE, Yun WB, Lee ML, Choi JY, Park JJ, Kim HR, Song BR, Hong JT, Song HK, Hwang DY. Synergic Laxative Effects of an Herbal Mixture of Liriope platyphylla, Glycyrrhiza uralensis, and Cinnamomum cassia in Loperamide-Induced Constipation of Sprague Dawley Rats. J Med Food 2019; 22:294-304. [PMID: 30724689 DOI: 10.1089/jmf.2018.4234] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Constipation is an acute or chronic illness attributed to various causes, ranging from lifestyle habits to side effects of a disease. To improve the laxative effects of some traditional medicines, herbal mixtures of Liriope platyphylla, Glycyrrhiza uralensis, and Cinnamomum cassia (LGC) were evaluated for their mechanism of action and therapeutic effects in loperamide (Lop)-induced constipated Sprague Dawley rats by examining alterations in excretion parameters, histological structure, mucin secretion, and related protein levels. Food intake and water consumption were constant for all animals. We observed that the Lop+LGC-treated group had significantly greater excretion of stool and urine than was observed in the Lop+Vehicle-treated group. Administration of LGC in the constipation model restored the intestinal transit ratio to normal levels, and increased the number of goblet cells, mucosal layer, and muscle thickness. Mucin secretion was greater in the Lop+LGC-treated group than in the Lop+Vehicle-treated group, and the expression of MUC2 and AQP8 genes were also increased. In addition, reverse transcription polymerase chain reaction and Western blot revealed an increase in the muscarinic acetylcholine receptors (mAChRs) in the Lop+LGC-treated group compared to the Lop+Vehicle-treated group. Furthermore, compared with the Lop+Vehicle-treated group, treatment with LGC reduced the phosphorylation of PKC and PI3K, and expression of Gα protein, but increased levels of IP3. Our results suggest that the traditional herbal mixture of LGC induces a potent laxative effect in Lop-induced constipation through mucosal tissue changes and mucin production. We also demonstrated that the laxative effect of LGC is closely related to the expression of mAChR and its downstream signals, suggesting the possibility of developing a constipation-laxative agent using LGC.
Collapse
Affiliation(s)
- Ji Eun Kim
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Woo Bin Yun
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Mi Lim Lee
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Jun Young Choi
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Jin Ju Park
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Hye Ryeong Kim
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Bo Ram Song
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Jin Tae Hong
- 2 College of Pharmacy, Chungbuk National University, Chungju, Korea
| | - Hyun Keun Song
- 3 Biomedical Science Institute, Changwon National University, Changwon-si, Gyeongsangnam-do, Korea
| | - Dae Youn Hwang
- 1 Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| |
Collapse
|
17
|
Garrido-Gil P, Dominguez-Meijide A, Moratalla R, Guerra MJ, Labandeira-Garcia JL. Aging-related dysregulation in enteric dopamine and angiotensin system interactions: implications for gastrointestinal dysfunction in the elderly. Oncotarget 2018. [PMID: 29541380 PMCID: PMC5834264 DOI: 10.18632/oncotarget.24330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gastrointestinal dysfunction is a common problem in the elderly. Aging-related changes in interactions between local dopaminergic and renin-angiotensin systems (RAS) have been observed in the brain, renal and vascular tissues. However, it is not known if these interactions also occur in the gut, and are dysregulated with aging. We showed a mutual regulation between the colonic dopaminergic system and RAS using young and aged mice deficient for major angiotensin and dopamine receptors. Aged rats showed a marked decrease in colonic dopamine D2 receptor expression, together with an increase in angiotensin type 1 (AT1) receptor expression, a decrease in angiotensin type 2 (AT2) receptor expression (i.e. an increase in the RAS pro-inflammatory arm activity), and increased levels of inflammatory and oxidative markers. Aged rats also showed increased levels of colonic dopamine and noradrenalin, and a marked decrease in acetylcholine and serotonin levels. The present observations contribute to explain an aging-related pro-inflammatory state and dysregulation in gastrointestinal function, which may be counteracted by treatment of aged animals with the AT1 receptor blocker candesartan.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| | - Antonio Dominguez-Meijide
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| | - Rosario Moratalla
- Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain.,Instituto Cajal, Madrid, Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| |
Collapse
|
18
|
Wells CI, O’Grady G, Bissett IP. Acute colonic pseudo-obstruction: A systematic review of aetiology and mechanisms. World J Gastroenterol 2017; 23:5634-5644. [PMID: 28852322 PMCID: PMC5558126 DOI: 10.3748/wjg.v23.i30.5634] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 06/29/2017] [Accepted: 07/22/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To critically review the literature addressing the definition, epidemiology, aetiology and pathophysiology of acute colonic pseudo-obstruction (ACPO). METHODS A systematic search was performed to identify articles investigating the aetiology and pathophysiology of ACPO. A narrative synthesis of the evidence was undertaken. RESULTS No consistent approach to the definition or reporting of ACPO has been developed, which has led to overlapping investigation with other conditions. A vast array of risk factors has been identified, supporting a multifactorial aetiology. The pathophysiological mechanisms remain unclear, but are likely related to altered autonomic regulation of colonic motility, in the setting of other predisposing factors. CONCLUSION Future research should aim to establish a clear and consistent definition of ACPO, and elucidate the pathophysiological mechanisms leading to altered colonic function. An improved understanding of the aetiology of ACPO may facilitate the development of targeted strategies for its prevention and treatment.
Collapse
|
19
|
Patel BA, Fidalgo S, Wang C, Parmar L, Mandona K, Panossian A, Flint MS, Ranson RN, Saffrey MJ, Yeoman MS. The TNF-α antagonist etanercept reverses age-related decreases in colonic SERT expression and faecal output in mice. Sci Rep 2017; 7:42754. [PMID: 28198447 DOI: 10.1038/srep42754] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
Treatment for chronic constipation in older people is challenging and the condition has a major impact on quality of life. A lack of understanding about the causes of this condition has hampered the development of effective treatments. 5-HT is an important pro-kinetic agent in the colon. We examined whether alterations in colonic 5-HT signalling underlie age-related changes in faecal output in mice and whether these changes were due to an increase in TNF-α. Components of the 5-HT signalling system (5-HT, 5-HIAA, SERT) and TNF-α expression were examined in the distal colon of 3, 12, 18 and 24-month old mice and faecal output and water content monitored under control conditions and following the administration of etanercept (TNF-α inhibitor; 1 mg Kg-1). Faecal output and water content were reduced in aged animals. Age increased mucosal 5-HT availability and TNF-α expression and decreased mucosal SERT expression and 5-HIAA. Etanercept treatment of old mice reversed these changes, suggesting that age-related changes in TNFα expression are an important regulator of mucosal 5-HT signalling and pellet output and water content in old mice. These data point to "anti-TNFα" drugs as potential treatments for age-related chronic constipation.
Collapse
Affiliation(s)
- Bhavik Anil Patel
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| | - Sara Fidalgo
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| | - Chunfang Wang
- Department of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Leena Parmar
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| | - Kasonde Mandona
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| | - Annabelle Panossian
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| | - Melanie S Flint
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| | - Richard N Ranson
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK
| | - M Jill Saffrey
- Department of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Mark S Yeoman
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, BN2 4GJ, UK
| |
Collapse
|
20
|
Gastrointestinal Dysfunctions in Parkinson's Disease: Symptoms and Treatments. PARKINSONS DISEASE 2016; 2016:6762528. [PMID: 28050310 PMCID: PMC5168460 DOI: 10.1155/2016/6762528] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/16/2016] [Indexed: 12/21/2022]
Abstract
A diagnosis of Parkinson's disease is classically established after the manifestation of motor symptoms such as rigidity, bradykinesia, and tremor. However, a growing body of evidence supports the hypothesis that nonmotor symptoms, especially gastrointestinal dysfunctions, could be considered as early biomarkers since they are ubiquitously found among confirmed patients and occur much earlier than their motor manifestations. According to Braak's hypothesis, the disease is postulated to originate in the intestine and then spread to the brain via the vagus nerve, a phenomenon that would involve other neuronal types than the well-established dopaminergic population. It has therefore been proposed that peripheral nondopaminergic impairments might precede the alteration of dopaminergic neurons in the central nervous system and, ultimately, the emergence of motor symptoms. Considering the growing interest in the gut-brain axis in Parkinson's disease, this review aims at providing a comprehensive picture of the multiple gastrointestinal features of the disease, along with the therapeutic approaches used to reduce their burden. Moreover, we highlight the importance of gastrointestinal symptoms with respect to the patients' responses towards medical treatments and discuss the various possible adverse interactions that can potentially occur, which are still poorly understood.
Collapse
|
21
|
Guha I, Slamova I, Chun S, Clegg A, Golos M, Thrasivoulou C, Simons JP, Al-Shawi R. The effects of short-term JNK inhibition on the survival and growth of aged sympathetic neurons. Neurobiol Aging 2016; 46:138-48. [PMID: 27490965 DOI: 10.1016/j.neurobiolaging.2016.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/30/2016] [Accepted: 06/24/2016] [Indexed: 11/27/2022]
Abstract
During the course of normal aging, certain populations of nerve growth factor (NGF)-responsive neurons become selectively vulnerable to cell death. Studies using dissociated neurons isolated from neonates have shown that c-Jun N-terminal kinases (JNKs) are important in regulating the survival and neurite outgrowth of NGF-responsive sympathetic neurons. Unlike neonatal neurons, adult sympathetic neurons are not dependent on NGF for their survival. Moreover, the NGF precursor, proNGF, is neurotoxic for aging but not young adult NGF-responsive neurons. Because of these age-related differences, the effects of JNK inhibition on the survival and growth of sympathetic neurons isolated from aged mice were studied. Aged neurons, as well as glia, were found to be dependent on JNK for their growth but not their survival. Conversely, proNGF neurotoxicity was JNK-dependent and mediated by the p75-interacting protein NRAGE, whereas neurite outgrowth was independent of NRAGE. These results have implications for the potential use of JNK inhibitors as therapies for ameliorating age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Isa Guha
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Ivana Slamova
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Soyon Chun
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Arthur Clegg
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Michal Golos
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Chris Thrasivoulou
- Research Department of Cell and Developmental Biology, University College London, London, UK
| | - J Paul Simons
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK.
| | - Raya Al-Shawi
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK.
| |
Collapse
|
22
|
Vather R, O'Grady G, Arkwright JW, Rowbotham DS, Cheng LK, Dinning PG, Bissett IP. Restoration of normal colonic motor patterns and meal responses after distal colorectal resection. Br J Surg 2016; 103:451-61. [DOI: 10.1002/bjs.10074] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/30/2015] [Accepted: 11/05/2015] [Indexed: 12/12/2022]
Abstract
Abstract
Background
Colorectal resections alter colonic motility, including disruption of control by neural or bioelectrical cell networks. The long-term impact of surgical resections and anastomoses on colonic motor patterns has, however, never been assessed accurately. Fibreoptic high-resolution colonic manometry was employed to define motility in patients who had undergone distal colorectal resection.
Methods
Recruited patients had undergone distal colorectal resections more than 12 months previously, and had normal bowel function. Manometry was performed in the distal colon (36 sensors; 1-cm intervals), with 2-h recordings taken before and after a meal, with comparison to controls. Analysis quantified all propagating events and frequencies (cyclical, short single, and long single motor patterns), including across anastomoses.
Results
Fifteen patients and 12 controls were recruited into the study. Coordinated propagating events directly traversed the healed anastomoses in nine of 12 patients with available data, including antegrade and retrograde cyclical, short single and long single patterns. Dominant frequencies in the distal colon were similar in patients and controls (2–3 cycles/min) (antegrade P = 0·482; retrograde P = 0·178). Compared with values before the meal, the mean(s.d.) number of dominant cyclical retrograde motor patterns increased in patients after the meal (2·1(2·7) versus 32·6(31·8) in 2 h respectively; P < 0·001), similar to controls (P = 0·178), although the extent of propagation was 41 per cent shorter in patients, by a mean of 3·4 cm (P = 0·003). Short and long single propagating motor patterns were comparable between groups in terms of frequency, velocity, extent and amplitude.
Conclusion
Motility patterns and meal responses are restored after distal colorectal resection in patients with normal bowel function. Coordinated propagation across healed anastomoses may indicate regeneration of underlying cellular networks.
Collapse
Affiliation(s)
- R Vather
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - G O'Grady
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - J W Arkwright
- School of Computer Science, Engineering and Mathematics, Flinders University, Adelaide, South Australia
| | - D S Rowbotham
- Department of Gastroenterology and Hepatology, Auckland District Health Board, Auckland, New Zealand
| | - L K Cheng
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - P G Dinning
- Department of Human Physiology, Flinders University, Adelaide, South Australia
- Department of Gastroenterology and Surgery, Flinders Medical Centre, Adelaide, South Australia
| | - I P Bissett
- Department of Surgery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
23
|
Puig KL, Lutz BM, Urquhart SA, Rebel AA, Zhou X, Manocha GD, Sens M, Tuteja AK, Foster NL, Combs CK. Overexpression of mutant amyloid-β protein precursor and presenilin 1 modulates enteric nervous system. J Alzheimers Dis 2015; 44:1263-78. [PMID: 25408221 PMCID: PMC6295343 DOI: 10.3233/jad-142259] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder histologically characterized by amyloid-β (Aβ) protein accumulation and activation of associated microglia. Although these features are well described in the central nervous system, the process and consequences of Aβ accumulation in the enteric nervous system have not been extensively studied. We hypothesized that Aβ also may accumulate in the enteric nervous system and lead to immune cell activation and neuronal dysfunction in the digestive tract not unlike that observed in diseased brain. To test this hypothesis, ileums of the small intestine of thirteen month old AβPP/PS1 and C57BL/6 (wild type) mice were collected and analyzed using immunohistochemistry, western blot analysis, cytokine arrays, and ELISA. AβPP/PS1 mice demonstrated no differences in intestinal motility or water absorption but elevated luminal IgA levels compared to wild type mice. They also had increased protein levels of AβPP and the proteolytic enzyme, BACE, corresponding to an increase in Aβ1-40 in the intestinal lysate as well as an increase in both Aβ1-40 and Aβ1-42 in the stool. This correlated with increased protein markers of proinflammatory and immune cell activation. Histologic analysis localized AβPP within enteric neurons but also intestinal epithelial cells with elevated Aβ immunoreactivity in the AβPP/PS1 mice. The presence of AβPP, Aβ, and CD68 immunoreactivity in the intestines of some patients with neuropathologically-confirmed AD are consistent with the findings in this mouse model. These data support the hypothesis that in AD the intestine, much like the brain, may develop proinflammatory and immune changes related to AβPP and Aβ.
Collapse
Affiliation(s)
- Kendra L. Puig
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Brianna M. Lutz
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Siri A. Urquhart
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Andrew A. Rebel
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xudong Zhou
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Gunjan D. Manocha
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - MaryAnn Sens
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Ashok K. Tuteja
- Division of Gastroenterology, University of Utah, Salt Lake City, UT, USA
| | - Norman L. Foster
- Center for Alzheimer’s Care, Imaging and Research, Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Colin K. Combs
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
24
|
Gombash SE, Cowley CJ, Fitzgerald JA, Hall JCE, Mueller C, Christofi FL, Foust KD. Intravenous AAV9 efficiently transduces myenteric neurons in neonate and juvenile mice. Front Mol Neurosci 2014; 7:81. [PMID: 25360081 PMCID: PMC4197761 DOI: 10.3389/fnmol.2014.00081] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/22/2014] [Indexed: 12/12/2022] Open
Abstract
Gene therapies for neurological diseases with autonomic or gastrointestinal involvement may require global gene expression. Gastrointestinal complications are often associated with Parkinson's disease and autism. Lewy bodies, a pathological hallmark of Parkinson's brains, are routinely identified in the neurons of the enteric nervous system (ENS) following colon biopsies from patients. The ENS is the intrinsic nervous system of the gut, and is responsible for coordinating the secretory and motor functions of the gastrointestinal tract. ENS dysfunction can cause severe patient discomfort, malnourishment, or even death as in intestinal pseudo-obstruction (Ogilvie syndrome). Importantly, ENS transduction following systemic vector administration has not been thoroughly evaluated. Here we show that systemic injection of AAV9 into neonate or juvenile mice results in transduction of 25-57% of ENS myenteric neurons. Transgene expression was prominent in choline acetyltransferase positive cells, but not within vasoactive intestinal peptide or neuronal nitric oxide synthase cells, suggesting a bias for cells involved in excitatory signaling. AAV9 transduction in enteric glia is very low compared to CNS astrocytes. Enteric glial transduction was enhanced by using a glial specific promoter. Furthermore, we show that AAV8 results in comparable transduction in neonatal mice to AAV9 though AAV1, 5, and 6 are less efficient. These data demonstrate that systemic AAV9 has high affinity for peripheral neural tissue and is useful for future therapeutic development and basic studies of the ENS.
Collapse
Affiliation(s)
- Sara E Gombash
- Department of Neuroscience, Ohio State University Columbus, OH, USA
| | | | | | - Jodie C E Hall
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Ohio State University Columbus, OH, USA
| | - Christian Mueller
- Department of Pediatrics, Gene Therapy Center, University of Massachusetts Medical School Worcester, MA, USA
| | | | - Kevin D Foust
- Department of Neuroscience, Ohio State University Columbus, OH, USA
| |
Collapse
|
25
|
Jo HJ, Kim N, Nam RH, Kang JM, Kim JH, Choe G, Lee HS, Park JH, Chang H, Kim H, Lee MY, Kim YS, Kim JS, Jung HC. Fat deposition in the tunica muscularis and decrease of interstitial cells of Cajal and nNOS-positive neuronal cells in the aged rat colon. Am J Physiol Gastrointest Liver Physiol 2014; 306:G659-69. [PMID: 24525022 DOI: 10.1152/ajpgi.00304.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Little is known about the time course of aging on interstitial cells of Cajal (ICC) of colon. The aim of this study was to investigate the change of morphology, ICC, and neuronal nitric oxide synthase (nNOS)-immunoreactive cells in the aged rat. The proximal colon of 344 Fischer rats at four different ages (6, 31, 74 wk, and 2 yr) were studied. The immunoreactivity of c-Kit, nNOS, anti-protein gene product 9.5, and synaptophysin were counted after immunohistochemistry. The c-kit, stem cell factor (ligand of Kit), and nNOS mRNA were measured by real-time PCR. c-Kit and nNOS protein were assessed by Western blot. Isovolumetric contractile force measurement and electrical field stimulation (EFS) were conducted. The area of intramuscular fat deposition significantly increased with age after 31 wk. c-Kit-immunoreactive ICC and nNOS-immunoreactive neurons and nerve fibers significantly declined with age. mRNA and protein expression of c-kit and nNOS decreased with aging. The functional study showed that the spontaneous contractility was decreased in aged rat, whereas EFS responses in the presence of atropine and L-NG-Nitroarginine methyl ester were increased in aged rat. In conclusion, the decrease of proportion of proper smooth muscle, the density of ICC and nNOS-immunoreactive neuronal fibers, and the number of nNOS-immunoreactive neurons during the aging process may explain the aging-associated colonic dysmotility.
Collapse
|
26
|
Patel PJ, Singh SK, Panaich S, Cardozo L. The aging gut and the role of prebiotics, probiotics, and synbiotics: A review. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.jcgg.2013.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Martin-Cano FE, Camello-Almaraz C, Acuña-Castroviejo D, Pozo MJ, Camello PJ. Age-related changes in mitochondrial function of mouse colonic smooth muscle: beneficial effects of melatonin. J Pineal Res 2014; 56:163-74. [PMID: 24313280 DOI: 10.1111/jpi.12109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 12/03/2013] [Indexed: 01/13/2023]
Abstract
Aging is a multifactorial process that involves biochemical, structural, and functional changes in mitochondria. The ability of melatonin to palliate the alterations induced by aging is based on its chronobiologic, antioxidant, and mitochondrial effects. There is little information about the effects of melatonin on the in situ mitochondrial network of aging cells and its physiological implications. We have studied the ability of melatonin to prevent the functional alterations of in situ mitochondria of smooth muscle cells and its impact on contractility. Mitochondrial membrane potential was recorded in isolated colonic smooth muscle cells from young mice (3 month old), aged mice (22-24-month old), and aged mice treated with melatonin (starting at 14-month age). Aging induced a partial mitochondrial depolarization in resting conditions and reduced the depolarizing response to cellular stimulation. Use of oligomycin indicated that aging enhanced the resting activity of the mitochondrial ATP synthase, whereas in young cells, the enzyme operated mainly in reverse mode. Melatonin treatment prevented all these changes. Aging reduced both spontaneous and stimulated contraction of colonic strips and shifted the metabolic dependence of contraction from mitochondria to glycolysis, as indicated the use of mitochondrial and glycolysis inhibitors. These functional alterations were also palliated by melatonin treatment. Aging effects were not related to a decrease in Ca2+ store mobilization, because this was enhanced in aged cells and restored by melatonin. In conclusion, melatonin prevents the age induced in situ mitochondrial potential alterations in smooth muscle cells and the associated changes in contractility and metabolism.
Collapse
Affiliation(s)
- Francisco E Martin-Cano
- Department of Physiology, Faculty of Nursing and Occupational Therapy, University of Extremadura, Cáceres, Spain
| | | | | | | | | |
Collapse
|
28
|
Choung RS, Locke GR, Schleck CD, Zinsmeister AR, Talley NJ. The effects of ageing on the onset and disappearance of unexplained abdominal pain: a population-based study. Aliment Pharmacol Ther 2014; 39:217-25. [PMID: 24304163 PMCID: PMC4070656 DOI: 10.1111/apt.12557] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 06/18/2013] [Accepted: 10/30/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND The population ≥65 years is rapidly increasing, but remarkably little is known about the natural history of abdominal pain with ageing. AIM To prospectively evaluate the natural history of abdominal pain (severity and frequency) in a US population, and evaluate potential risk factors (including somatisation) for the onset and disappearance of abdominal pain with increasing age. METHODS Between 1988 and 2004, valid self-report questionnaires that recorded gastrointestinal symptoms including severity and frequency of abdominal pain were mailed to randomly selected cohorts of community residents followed over time. This study identified all respondents who answered abdominal pain questions at an initial and follow-up survey. RESULTS One thousand nine hundred and thirteen subjects were included (mean age in years at first survey: 48 ± 12 (SD), mean age at second survey: 59 ± 13 (SD); 53% female). The onset and disappearance rate of abdominal pain over the follow-up were 18% (95% CI, 16, 20) and 47% (43, 50) respectively. The rates of increasing vs. decreasing abdominal pain score were 18% (16, 20) vs. 21% (20, 23) respectively. While younger age at initial survey was associated with the onset of abdominal pain {vs. subjects without abdominal pain, [OR 0.9 (0.7, 1.0)]}, older age at initial survey and times between surveys were associated with the disappearance of abdominal pain {vs. subjects with abdominal pain, [OR 1.2 (1.0, 1.5)]}. Female gender (OR 1.4 [1.0, 2.1]), higher somatisation scores (OR 5.3 [3.2, 8.7]) and larger changes in somatisation score (OR 2.1 [1.4, 3.2]) were positively associated with the onset of abdominal pain. CONCLUSION Increasing age is associated with the disappearance of abdominal pain in the community.
Collapse
Affiliation(s)
- Rok Seon Choung
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA,Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea, University College of Medicine, Seoul, South Korea
| | - G. Richard Locke
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Cathy D. Schleck
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Alan R. Zinsmeister
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
29
|
Phillips RJ, Hudson CN, Powley TL. Sympathetic axonopathies and hyperinnervation in the small intestine smooth muscle of aged Fischer 344 rats. Auton Neurosci 2013; 179:108-21. [PMID: 24104187 DOI: 10.1016/j.autneu.2013.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/04/2013] [Accepted: 09/09/2013] [Indexed: 01/13/2023]
Abstract
It is well documented that the intrinsic enteric nervous system of the gastrointestinal (GI) tract sustains neuronal losses and reorganizes as it ages. In contrast, age-related remodeling of the extrinsic sympathetic projections to the wall of the gut is poorly characterized. The present experiment, therefore, surveyed the sympathetic projections to the aged small intestine for axonopathies. Furthermore, the experiment evaluated the specific prediction that catecholaminergic inputs undergo hyperplastic changes. Jejunal tissue was collected from 3-, 8-, 16-, and 24-month-old male Fischer 344 rats, prepared as whole mounts consisting of the muscularis, and processed immunohistochemically for tyrosine hydroxylase, the enzymatic marker for norepinephrine, and either the protein CD163 or the protein MHCII, both phenotypical markers for macrophages. Four distinctive sympathetic axonopathy profiles occurred in the small intestine of the aged rat: (1) swollen and dystrophic terminals, (2) tangled axons, (3) discrete hyperinnervated loci in the smooth muscle wall, including at the bases of Peyer's patches, and (4) ectopic hyperplastic or hyperinnervating axons in the serosa/subserosal layers. In many cases, the axonopathies occurred at localized and limited foci, involving only a few axon terminals, in a pattern consistent with incidences of focal ischemic, vascular, or traumatic insult. The present observations underscore the complexity of the processes of aging on the neural circuitry of the gut, with age-related GI functional impairments likely reflecting a constellation of adjustments that range from selective neuronal losses, through accumulation of cellular debris, to hyperplasias and hyperinnervation of sympathetic inputs.
Collapse
Affiliation(s)
- Robert J Phillips
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States.
| | | | | |
Collapse
|
30
|
Gamage PPKM, Ranson RN, Patel BA, Yeoman MS, Saffrey MJ. Myenteric neuron numbers are maintained in aging mouse distal colon. Neurogastroenterol Motil 2013; 25:e495-e505. [PMID: 23517051 DOI: 10.1111/nmo.12114] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/15/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Age-associated myenteric neuronal loss has been described in several species. In some studies,cholinergic neurons have been reported to be selectively vulnerable, whereas nitrergic neurons are spared. Aging of the mouse enteric nervous system(ENS) and the subtypes of mouse myenteric neurons that may be lost have been little studied. We therefore investigated changes in the numbers of total neurons and two neuronal subpopulations in the mouse distal colon during aging. METHODS Wholemount preparations from 3–4-, 12–13-, 18–19-, and 24–25-month-old C57BL/6 mice were double immunolabeled with HuC/D antibody to identify the total neuronal population and antisera to either calbindin or neuronal nitric oxide synthase (nNOS) to identify myenteric neuronal subpopulations. Samples were analyzed by confocal microscopy. New procedures were employed to ensure unbiased counting and to correct for changes in gut dimensions with age and stretch during sample preparation. The density of nerve fibers in the tertiary plexus was also studied. KEY RESULTS No significant change in numbers of total neurons or of either subpopulation with age was measured, but because of gut growth, the density of myenteric neurons decreased between 3–4 and 12–13 months. The density of nNOS-immunoreactive nerve fibers in the tertiary plexus increased significantly with age, up to 18–19 months. Numerous swollen processes of CB and nNOS-immunoreactive neurons were observed in 18–19- and 24–25-month-old animals. Conclusions &Inferences These results indicate that aging does not result in a loss of myenteric neurons in mouse distal colon at the ages studied, although neurodegenerative changes, which may impact on neuronal function, do occur.
Collapse
Affiliation(s)
- P. P. K. M. Gamage
- Department of Life, Health and Chemical Sciences; Biomedical Research Network; Open University; Walton Hall; Milton Keynes; MK7 6AA; UK
| | - R. N. Ranson
- Faculty of Health and Life Sciences; Northumbria University; Ellison Building; Newcastle upon Tyne; NE1 8ST; UK
| | - B. A. Patel
- School of Pharmacy and Biomolecular Sciences, Huxley Building; University of Brighton; Lewes Road; Brighton; BN2 4GJ; UK
| | - M. S. Yeoman
- School of Pharmacy and Biomolecular Sciences, Huxley Building; University of Brighton; Lewes Road; Brighton; BN2 4GJ; UK
| | - M. J. Saffrey
- Department of Life, Health and Chemical Sciences; Biomedical Research Network; Open University; Walton Hall; Milton Keynes; MK7 6AA; UK
| |
Collapse
|
31
|
Chen JH, Zhang Q, Yu Y, Li K, Liao H, Jiang L, Hong L, Du X, Hu X, Chen S, Yin S, Gao Q, Yin X, Luo H, Huizinga JD. Neurogenic and myogenic properties of pan-colonic motor patterns and their spatiotemporal organization in rats. PLoS One 2013; 8:e60474. [PMID: 23577116 PMCID: PMC3618275 DOI: 10.1371/journal.pone.0060474] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 02/26/2013] [Indexed: 02/06/2023] Open
Abstract
Background and Aims Better understanding of intrinsic control mechanisms of colonic motility will lead to better treatment options for colonic dysmotility. The aim was to investigate neurogenic and myogenic control mechanisms underlying pan-colonic motor patterns. Methods Analysis of in vitro video recordings of whole rat colon motility was used to explore motor patterns and their spatiotemporal organizations and to identify mechanisms of neurogenic and myogenic control using pharmacological tools. Results Study of the pan-colonic spatiotemporal organization of motor patterns revealed: fluid-induced or spontaneous rhythmic propulsive long distance contractions (LDCs, 0.4–1.5/min, involving the whole colon), rhythmic propulsive motor complexes (RPMCs) (0.8–2.5/min, dominant in distal colon), ripples (10–14/min, dominant in proximal colon), segmentation and retrograde contractions (0.1–0.8/min, prominent in distal and mid colon). Spontaneous rhythmic LDCs were the dominant pattern, blocked by tetrodotoxin, lidocaine or blockers of cholinergic, nitrergic or serotonergic pathways. Change from propulsion to segmentation and distal retrograde contractions was most prominent after blocking 5-HT3 receptors. In the presence of all neural blockers, bethanechol consistently evoked rhythmic LDC-like propulsive contractions in the same frequency range as the LDCs, indicating the existence of myogenic mechanisms of initiation and propulsion. Conclusions Neurogenic and myogenic control systems orchestrate distinct and variable motor patterns at different regions of the pan-colon. Cholinergic, nitrergic and serotonergic pathways are essential for rhythmic LDCs to develop. Rhythmic motor patterns in presence of neural blockade indicate the involvement of myogenic control systems and suggest a role for the networks of interstitial cells of Cajal as pacemakers.
Collapse
Affiliation(s)
- Ji-Hong Chen
- Department of Gastroenterology and Hepatology, Renmin Hospital of Wuhan University and Wuhan University Institute of Digestive and Liver Diseases, Wuhan, Hubei, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dietary restriction interferes with oxidative status and intrinsic intestinal innervation in aging rats. Nutrition 2013; 29:673-80. [PMID: 23317927 DOI: 10.1016/j.nut.2012.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/20/2012] [Accepted: 09/24/2012] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To evaluate the effects of dietary restriction on oxidative status, the HuC/D-neuronal nitric oxide synthase (nNOS) myenteric neuron population, HuC/D-S100 glial cells, and the morphometry of the small intestine in rats at various ages. METHODS Fifteen Wistar rats were divided into 7-and 12-mo-old control groups and a 12-mo-old experimental group subjected to dietary restrictions (50% of normal ration) for 5 mo. At 7 and 12 mo of age, the animals were anesthetized, and blood was collected to assess the biochemical components and oxidative status. Ileum samples were subjected to double-marker (HuC/D-nNOS and HuC/D-S100) immunostaining and histologic processing to morphometrically analyze intestinal wall elements and determine the metaphase index and rate of caliciform cells. The data were subjected to analysis of variance and the Tukey post hoc test with a 5% significance level. RESULTS Age affected the oxidative status by increasing lipid peroxidation, with no effect on blood components, intrinsic innervation, and intestinal wall elements. The animals subjected to dietary restriction showed improved levels of total cholesterol, triacylglycerols, and oxidative status, with no changes in the nNOS neuron population. However, the dietary restriction dramatically decreased the glial and HuC/D myenteric populations, led to atrophy of the neuronal cell body, induced glial hypertrophy, and decreased the thickness of the intestinal wall. CONCLUSION The high oxidative status of the aging animals was reversed by dietary restriction, which also lowered cholesterol and triacylglycerol levels. The present dietary restriction elicited morpho-quantitative changes in the myenteric plexus and histology of the ileum, with likely effects on intestinal functions.
Collapse
|
33
|
Bernardini N, Ippolito C, Segnani C, Mattii L, Bassotti G, Villanacci V, Blandizzi C, Dolfi A. Histopathology in gastrointestinal neuromuscular diseases: methodological and ontological issues. Adv Anat Pathol 2013; 20:17-31. [PMID: 23232568 DOI: 10.1097/pap.0b013e31827b65c0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gastrointestinal neuromuscular diseases (GINMDs) comprise a heterogenous group of chronic conditions associated with impaired gut motility. These gastrointestinal (GI) disorders, differing for etiopathogenic mechanisms, pathologic lesions, and region of gut involvement, represent a relevant matter for public health, because they are very common, can be disabling, and determine major social and economic burdens. GINMDs are presumed or proven to arise as a result of a dysfunctioning GI neuromuscular apparatus, which includes myenteric ganglia (neurons and glial cells), interstitial cells of Cajal and smooth muscle cells. Despite the presence of symptoms related to gut dysmotility in the clinical phenotype of these patients, in the diagnostic setting scarce attention is usually paid to the morphologic pattern of the GI neuromuscular apparatus. It is also objectively difficult to collect full-thickness gut tissue samples from patients with GINMDs, because their disease, which can be only functional in nature, may not justify invasive diagnostic procedures as a first-line approach. As a consequence, whenever available, bioptic gut specimens, retrieved from these patients, must be regarded as a unique chance for obtaining relevant diagnostic information. On the basis of these arguments, there is an urgent need of standardized and validated histopathologic methods, aiming at overcoming the discrepancies affecting current approaches, which usually lead to conflicting definitions of normality and hamper the identification of disease-specific pathologic patterns. This review article intends to address current methodological and ontological issues in the histopathologic diagnosis of GINMDs, to foster the debate on how to discriminate normal morphology from abnormalities.
Collapse
Affiliation(s)
- Nunzia Bernardini
- Unit of Histology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Korsak K, Dolatshad NF, Silva AT, Saffrey MJ. Ageing of enteric neurons: oxidative stress, neurotrophic factors and antioxidant enzymes. Chem Cent J 2012; 6:80. [PMID: 22857398 PMCID: PMC3469348 DOI: 10.1186/1752-153x-6-80] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 07/10/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Ageing is associated with gastrointestinal dysfunction, which can have a major impact on quality of life of the elderly. A number of changes in the innervation of the gut during ageing have been reported, including neuronal loss and degenerative changes. Evidence indicates that reactive oxygen species (ROS) are elevated in ageing enteric neurons, but that neurotrophic factors may reduce generation of neuronal ROS. Two such factors, glial cell line derived neurotrophic factor (GDNF) and neurotrophin-3 (NT-3) have also been found to protect enteric neurons against oxidative stress induced cell death of enteric ganglion cells in vitro. We have investigated the possible roles of neurotrophic factors further, by examining their expression in the gut during ageing, and by analysing their effects on antioxidant enzyme production in cultures of enteric ganglion cells. RESULTS Analysis of the expression of GDNF and its receptors c-Ret and GFR α - 1 in rat gut by RT-PCR showed that expression continues throughout life and into ageing, in both ad libitum(AL) and calorically-restricted (CR) animals. Levels of expression of GDNF and GFR α - 1 were elevated in 24 month AL animals compared to 24 month CR animals, and to 24 CR and 6 month control animals respectively. The related factor Neurturin and its receptor GFR α - 2 were also expressed throughout life, the levels of the GFR - α-2(b) isoform were reduced in 24 m AL animals. Immunolabelling showed that c-Ret and GFR α - 1 proteins were expressed by myenteric neurons in ageing animals. GDNF, but not NT-3, was found to increase expression of Cu/Zn superoxide dismutase and catalase by cultured enteric ganglion cells. CONCLUSIONS The neurotrophic factors GDNF and neurturin and their receptors continue to be expressed in the ageing gut. Changes in the levels of expression of GDNF , GFR α-1 and GFR α-2(b) isoform occurred in 24 m AL animals. GDNF, but not NT-3, increased the levels of antioxidant enzymes in cultured enteric ganglion cells, indicating a possible mechanism for the reported protective effect of GDNF against menadione-induced neuronal apoptosis in the ageing gut. Together these data suggest that GDNF family members may play a protective role in the gut throughout life, and support the suggestion that dysregulation of neurotrophic factor support could contribute to neuronal ageing in the gut.
Collapse
Affiliation(s)
- Kris Korsak
- Department of Life, Health and Chemical Sciences, Biomedical Research Network, The Open University, Milton Keynes MK7 6AA, UK.
| | | | | | | |
Collapse
|
35
|
Hoyle CHV, Saffrey MJ. Effects of aging on cholinergic neuromuscular transmission in isolated small intestine of ad libitum fed and calorically-restricted rats. Neurogastroenterol Motil 2012; 24:586-92. [PMID: 22435850 DOI: 10.1111/j.1365-2982.2012.01913.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Age-associated losses of enteric neurons have been described. In rat ileum, myenteric neurons lost during aging have been reported to be predominantly cholinergic, and caloric restriction (CR) has been shown to protect against these losses. Cholinergic myenteric neurons include excitatory motor neurons, so the aim of this work was to determine whether neuronal loss in ad libitum (AL)-fed animals is reflected in dysfunctional cholinergic neuromuscular transmission, and if CR reduces any such dysfunction. METHODS Effects of electrical field stimulation (EFS) and applied acetylcholine (ACh) were examined in the longitudinal muscle of isolated ileal segments from 6-month-old rats and from 13- and 24-month-old rats fed either AL or CR diets. KEY RESULTS Contractile responses to EFS were abolished by atropine and potentiated by the acetylcholinesterase inhibitor, eserine. Frequency-response relationships were not significantly different amongst the three age-groups. Sensitivity to applied ACh, however, was three-fold lower in the oldest animals (P < 0.05). Eserine potentiated responses to ACh; there were no statistically significant differences amongst the sensitivities to ACh in its presence. No significant differences between AL- and CR-fed animals were measured, although variability was less in CR-fed than in AL-fed groups. CONCLUSIONS & INFERENCES The cholinergic system supplying the rat ileum longitudinal muscle did not appear to be impaired in old age. Decreased sensitivity to applied ACh in old tissues may have been due to increased acetylcholinesterase activity. Caloric restriction had no significant effect on responses to EFS or applied ACh. The implications of these results are discussed.
Collapse
Affiliation(s)
- C H V Hoyle
- Department of Anatomy and Developmental Biology, University College London, London, UK
| | | |
Collapse
|
36
|
Korsak K, Silva AT, Saffrey MJ. Differing effects of NT-3 and GDNF on dissociated enteric ganglion cells exposed to hydrogen peroxide in vitro. Neurosci Lett 2012; 517:102-6. [PMID: 22548772 DOI: 10.1016/j.neulet.2012.04.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 03/26/2012] [Accepted: 04/13/2012] [Indexed: 01/19/2023]
Abstract
Oxidative stress is widely recognized to contribute to neuronal death during various pathological conditions and ageing. In the enteric nervous system (ENS), reactive oxygen species have been implicated in the mechanism of age-associated neuronal loss. The neurotrophic factors, neurotrophin 3 (NT-3) and glial cell line-derived neurotrophic factor (GDNF), are important in the development of enteric neurons and continue to be expressed in the gut throughout life. It has therefore been suggested that they may have a neuroprotective role in the ENS. We investigated the potential of NT-3 and GDNF to prevent the death of enteric ganglion cells in dissociated cell culture after exposure to hydrogen peroxide (H(2)O(2)). H(2)O(2) treatment resulted in a dose-dependent death of enteric neurons and glial cells, as demonstrated by MTS assay, bis-benzimide and propidium iodide staining and immunolabelling. Cultures treated with NT-3 prior to exposure showed reduced cell death compared to untreated control or GDNF-treated cultures. GDNF treatment did not affect neuronal survival in H(2)O(2)-treated cultures. These results suggest that NT-3 is able to enhance the survival of enteric ganglion cells exposed to oxidative stress.
Collapse
Affiliation(s)
- Kris Korsak
- Department of Life, Health and Chemical Sciences, The Open University, UK
| | | | | |
Collapse
|
37
|
Phillips RJ, Powley TL. Macrophages associated with the intrinsic and extrinsic autonomic innervation of the rat gastrointestinal tract. Auton Neurosci 2012; 169:12-27. [PMID: 22436622 DOI: 10.1016/j.autneu.2012.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 12/28/2022]
Abstract
Interactions between macrophages and the autonomic innervation of gastrointestinal (GI) tract smooth muscle have received little experimental attention. To better understand this relationship, immunohistochemistry was performed on GI whole mounts from rats at three ages. The phenotypes, morphologies, and distributions of gut macrophages are consistent with the cells performing extensive housekeeping functions in the smooth muscle layers. Specifically, a dense population of macrophages was located throughout the muscle wall where they were distributed among the muscle fibers and along the vasculature. Macrophages were also associated with ganglia and connectives of the myenteric plexus and with the sympathetic innervation. Additionally, these cells were in tight registration with the dendrites and axons of the myenteric neurons as well as the varicosities along the length of the sympathetic axons, suggestive of a contribution by the macrophages to the homeostasis of both synapses and contacts between the various elements of the enteric circuitry. Similarly, macrophages were involved in the presumed elimination of neuropathies as indicated by their association with dystrophic neurons and neurites which are located throughout the myenteric plexus and smooth muscle wall of aged rats. Importantly, the patterns of macrophage-neuron interactions in the gut paralleled the much more extensively characterized interactions of macrophages (i.e., microglia) and neurons in the CNS. The present observations in the PNS as well as extrapolations from homologous microglia in the CNS suggest that GI macrophages play significant roles in maintaining the nervous system of the gut in the face of wear and tear, disease, and aging.
Collapse
Affiliation(s)
- Robert J Phillips
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana 47907-2081, USA
| | | |
Collapse
|
38
|
Pathological correlates of gastrointestinal dysfunction in Parkinson's disease. Neurobiol Dis 2011; 46:559-64. [PMID: 22048068 DOI: 10.1016/j.nbd.2011.10.014] [Citation(s) in RCA: 185] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 10/11/2011] [Accepted: 10/16/2011] [Indexed: 12/11/2022] Open
Abstract
Gastrointestinal dysfunction is a prominent manifestation of Parkinson's disease (PD). Gastrointestinal symptoms in PD include reduced salivation, dysphagia, impaired gastric emptying, constipation, and defecatory dysfunction. Constipation may precede the development of somatic motor symptoms of PD for several years. Neuropathological studies show early accumulation of abnormal alpha-synuclein (α-SYN) containing inclusions (Lewy neurites) in the enteric nervous system (ENS) and dorsal motor nucleus of the vagus (DMV) both in PD and in incidental Lewy body disease (ILBD). These findings provided the basis for the hypothesis that α-SYN pathology progresses in a centripetal, prion-like fashion, from the ENS to the DMV and then to more rostral areas of the central nervous system. Colonic biopsies may show accumulation α-SYN immunoreactive Lewy neurites in the submucosal plexus of PD patients. Salivary gland involvement is prominent in PD and α-SYN pathology can be detected both at autopsy and in minor salivary gland biopsies.
Collapse
|
39
|
Asuzu DT, Hayashi Y, Izbeki F, Popko LN, Young DL, Bardsley MR, Lorincz A, Kuro-o M, Linden DR, Farrugia G, Ordog T. Generalized neuromuscular hypoplasia, reduced smooth muscle myosin and altered gut motility in the klotho model of premature aging. Neurogastroenterol Motil 2011; 23:e309-23. [PMID: 21605285 PMCID: PMC3149585 DOI: 10.1111/j.1365-2982.2011.01730.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Gastrointestinal symptoms, particularly constipation, increase with aging, but their underlying mechanisms are poorly understood due to lack of experimental models. Previously we established the progeric klotho mouse as a model of aging-associated anorexia and gastric dysmotility. We also detected reduced fecal output in these animals; therefore, the aim of this study was to investigate in vivo function and cellular make-up of the small intestinal and colonic neuromuscular apparatus. METHODS Klotho expression was studied by RT-PCR and immunohistochemistry. Motility was assessed by dye transit and bead expulsion. Smooth muscle and neuron-specific gene expression was studied by Western immunoblotting. Interstitial cells of Cajal (ICC) and precursors were analyzed by flow cytometry, confocal microscopy, and three-dimensional reconstruction. HuC/D(+) myenteric neurons were enumerated by fluorescent microscopy. KEY RESULTS Klotho protein was detected in neurons, smooth muscle cells, and some ICC classes. Small intestinal transit was slower but whole-gut transit of klotho mice was accelerated due to faster colonic transit and shorter intestinal lengths, apparent only after weaning. Fecal water content remained normal despite reduced output. Smooth muscle myosin expression was reduced. ICC, ICC precursors, as well as nitrergic and cholinergic neurons maintained their normal proportions in the shorter intestines. CONCLUSIONS & INFERENCES Progeric klotho mice express less contractile proteins and develop generalized intestinal neuromuscular hypoplasia mainly arising from stunted postweaning growth. As reduced fecal output in these mice occurs in the presence of accelerated colonic and whole-gut transit, it likely reflects reduced food intake rather than intestinal dysmotility.
Collapse
Affiliation(s)
- David T. Asuzu
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Ferenc Izbeki
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Laura N. Popko
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - David L. Young
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael R. Bardsley
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Andrea Lorincz
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David R. Linden
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamas Ordog
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
40
|
Bitar K, Greenwood-Van Meerveld B, Saad R, Wiley JW. Aging and gastrointestinal neuromuscular function: insights from within and outside the gut. Neurogastroenterol Motil 2011; 23:490-501. [PMID: 21320236 PMCID: PMC3094479 DOI: 10.1111/j.1365-2982.2011.01678.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Affiliation(s)
- K Bitar
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA Research Administration, VA Medical Center, Oklahoma City, OK, USA
| | | | | | | |
Collapse
|
41
|
Wiskur B, Greenwood-Van Meerveld B. The aging colon: the role of enteric neurodegeneration in constipation. Curr Gastroenterol Rep 2011; 12:507-12. [PMID: 20878508 DOI: 10.1007/s11894-010-0139-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Constipation is a common problem in the elderly, and abnormalities in the neural innervation of the colon play a significant role in abnormalities in colonic motility leading to delayed colonic transit. The scope of this review encompasses the latest advances to enhance our understanding of the aging colon with emphasis on enteric neurodegeneration, considered a likely cause for the development of constipation in the aging gut in animal models. Neural innervation of the colon and the effects of aging on intrinsic and extrinsic nerves innervating the colonic smooth muscle is discussed. Evidence supporting the concept that neurologic disorders, such as Parkinson's disease, not only affect the brain but also cause neurodegeneration within the enteric nervous system leading to colonic dysmotility is presented. Further research is needed to investigate the influence of aging on the gastrointestinal tract and to develop novel approaches to therapy directed at protecting the enteric nervous system from neurodegeneration.
Collapse
Affiliation(s)
- Brandt Wiskur
- VA Medical Center, Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | |
Collapse
|
42
|
Hwang SJ, Durnin L, Dwyer L, Rhee PL, Ward SM, Koh SD, Sanders KM, Mutafova-Yambolieva VN. β-nicotinamide adenine dinucleotide is an enteric inhibitory neurotransmitter in human and nonhuman primate colons. Gastroenterology 2011; 140:608-617.e6. [PMID: 20875415 PMCID: PMC3031738 DOI: 10.1053/j.gastro.2010.09.039] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 08/03/2010] [Accepted: 09/17/2010] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS An important component of enteric inhibitory neurotransmission is mediated by a purine neurotransmitter, such as adenosine 5'-triphosphate (ATP), binding to P2Y1 receptors and activating small conductance K(+) channels. In murine colon β-nicotinamide adenine dinucleotide (β-NAD) is released with ATP and mimics the pharmacology of inhibitory neurotransmission better than ATP. Here β-NAD and ATP were compared as possible inhibitory neurotransmitters in human and monkey colons. METHODS A small-volume superfusion assay and high-pressure liquid chromatography with fluorescence detection were used to evaluate spontaneous and nerve-evoked overflow of β-NAD, ATP, and metabolites. Postjunctional responses to nerve stimulation, β-NAD and ATP were compared using intracellular membrane potential and force measurements. Effects of β-NAD on smooth muscle cells (SMCs) were recorded by patch clamp. P2Y receptor transcripts were assayed by reverse transcription polymerase chain reaction. RESULTS In contrast to ATP, overflow of β-NAD evoked by electrical field stimulation correlated with stimulation frequency and was diminished by the neurotoxins, tetrodotoxin, and ω-conotoxin GVIA. Inhibitory junction potentials and responses to exogenous β-NAD, but not ATP, were blocked by P2Y receptor antagonists suramin, pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS), 2'-deoxy-N6-methyladenosine 3',5'-bisphosphate (MRS 2179), and (1R,2S,4S,5S)-4-[2-Iodo-6-(methylamino)-9H-purin-9-yl]-2-(phosphonooxy)bicyclo[3.1.0]hexane-1-methanol dihydrogen phosphate ester tetraammonium salt (MRS 2500). β-NAD activated nonselective cation currents in SMCs, but failed to activate outward currents. CONCLUSIONS β-NAD meets the criteria for a neurotransmitter better than ATP in human and monkey colons and therefore may contribute to neural regulation of colonic motility. SMCs are unlikely targets for inhibitory purine neurotransmitters because dominant responses of SMCs were activation of net inward, rather than outward, current.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557
| | - Leonie Durnin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557
| | - Laura Dwyer
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557
| | - Poong-Lyul Rhee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, South Korea
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557
| | | |
Collapse
|
43
|
Morphological changes in the enteric nervous system of aging and APP23 transgenic mice. Brain Res 2011; 1378:43-53. [PMID: 21241669 DOI: 10.1016/j.brainres.2011.01.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 12/22/2010] [Accepted: 01/10/2011] [Indexed: 12/31/2022]
Abstract
Gastrointestinal motility disorders often pose a debilitating problem, especially in elderly patients. In addition, they are frequently occurring co-morbidities in dementia. Whereas a failing enteric nervous system has already been shown to be involved in gastrointestinal motility disorders and in Parkinson's disease, a relationship with the neurodegenerative process of Alzheimer's disease was not yet shown. Therefore, we sought to document quantitative changes in the distribution of βIII-tubulin (general neuronal marker), Substance P, neuronal nitric oxide synthase (NOS), glial fibrillary acidic protein (GFAP) and S-100 immunoreactivity in addition to a qualitative assessment of the presence of amyloid in the small and large intestines of 6, 12 and 18-month-old wild type and transgenic Thy-1-APP23 mice. Amyloid deposits were seen in the vasculature, the mucosal and muscle layer of both heterozygous and wild type mice. Amyloidβ₁₋₄₂ could not be detected, pointing to a different amyloid composition than that found in senile plaques in the mice's brains. The finding of an increased density of βIII-tubulin-, Substance P- and NOS-IR-nerve fibres in heterozygous mice could not undoubtedly be related to amyloid deposition or to an activation of glial cells. Therefore, the alterations at the level of the enteric nervous system and the deposition of amyloid seem not primarily involved in the pathogenesis of Alzheimer's disease. At most they are secondary related to the neurodegenerative process. Additionally, our data could not show extensive neuronal or glial cell loss associated with aging, in contrast to other reports. Instead an increase in S100-IR was observed in senescent mice.
Collapse
|
44
|
Colonic biopsies to assess the neuropathology of Parkinson's disease and its relationship with symptoms. PLoS One 2010; 5:e12728. [PMID: 20856865 PMCID: PMC2939055 DOI: 10.1371/journal.pone.0012728] [Citation(s) in RCA: 317] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 08/24/2010] [Indexed: 12/14/2022] Open
Abstract
Background The presence of Lewy bodies and Lewy neurites (LN) has been demonstrated in the enteric nervous system (ENS) of Parkinson's disease (PD) patients. The aims of the present research were to use routine colonoscopy biopsies (1) to analyze, in depth, enteric pathology throughout the colonic submucosal plexus (SMP), and (2) to correlate the pathological burden with neurological and gastrointestinal (GI) symptoms. Methodology/Principal Findings A total of 10 control and 29 PD patients divided into 3 groups according to disease duration were included. PD and GI symptoms were assessed using the Unified Parkinson's Disease Rating Scale part III and the Rome III questionnaire, respectively. Four biopsies were taken from the ascending and descending colon during the course of a total colonoscopy. Immunohistochemical analysis was performed using antibodies against phosphorylated alpha-synuclein, neurofilaments NF 220 kDa (NF) and tyrosine hydroxylase (TH). The density of LN, labeled by anti-phosphorylated alpha-synuclein antibodies, was evaluated using a quantitative rating score. Lewy pathology was apparent in the colonic biopsies from 21 patients and in none of the controls. A decreased number of NF-immunoreactive neurons per ganglion was observed in the SMP of PD patients compared to controls. The amount of LN in the ENS was inversely correlated with neuronal count and positively correlated with levodopa-unresponsive features and constipation. Conclusion/Significance Analysis of the ENS by routine colonoscopy biopsies is a useful tool for pre-mortem neuropathological diagnosis of PD, and also provides insight into the progression of motor and non-motor symptoms.
Collapse
|
45
|
Müller-Lissner S, Rykx A, Kerstens R, Vandeplassche L. A double-blind, placebo-controlled study of prucalopride in elderly patients with chronic constipation. Neurogastroenterol Motil 2010; 22:991-8, e255. [PMID: 20529205 DOI: 10.1111/j.1365-2982.2010.01533.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Constipation affects up to 50% of the elderly; this study evaluates the efficacy, safety, and tolerability of the selective 5-HT(4) agonist prucalopride in chronically constipated elderly patients. METHODS Three hundred chronic constipation patients aged >or=65 years were randomized to prucalopride (1, 2, or 4 mg once daily) or placebo for 4 weeks. The primary endpoint was the percentage of patients with >or=3 spontaneous complete bowel movements (SCBM) per week. Secondary endpoints included the percentage with an increase of >or=1 SCBM per week, BM frequency, constipation-related symptoms, quality of life (QoL), safety, and tolerability. KEY RESULTS More patients achieved >or=3 SCBM per week with prucalopride than with placebo. This difference was largest and significant during the first week of 4 mg prucalopride (P <or= 0.05). Significantly more patients in each prucalopride group achieved an increase of >or=1 SCBM per week from baseline vs placebo (e.g. 60% with 1 mg prucalopride vs 34% with placebo at week 4; P <or= 0.05). More patients had improvement in PAC-QOL satisfaction score of >or=1 with 1 mg prucalopride than with placebo (P <or= 0.05); the same was true for PAC-SYM stool symptoms (1 and 4 mg prucalopride; P <or= 0.05). Treatment-emergent adverse events were similar between groups: the most frequently reported with prucalopride were headache and gastrointestinal events. There were no clinically significant differences between prucalopride and placebo for vital signs, laboratory assessments, or ECG variables. CONCLUSIONS & INFERENCES Prucalopride, in the dose-range tested (1-4 mg once daily), has beneficial effects on bowel movements, symptoms, and QoL, and is safe and well-tolerated in elderly patients with chronic constipation.
Collapse
|
46
|
Izbeki F, Asuzu DT, Lorincz A, Bardsley MR, Popko LN, Choi KM, Young DL, Hayashi Y, Linden DR, Kuro-o M, Farrugia G, Ordog T. Loss of Kitlow progenitors, reduced stem cell factor and high oxidative stress underlie gastric dysfunction in progeric mice. J Physiol 2010; 588:3101-17. [PMID: 20581042 PMCID: PMC2956948 DOI: 10.1113/jphysiol.2010.191023] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 06/24/2010] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal functions decline with ageing leading to impaired quality of life, and increased morbidity and mortality. Neurodegeneration is believed to underlie ageing-associated dysmotilities but the mechanisms have not been fully elucidated. We used progeric mice deficient in the anti-ageing peptide Klotho to investigate the contribution of key cell types of the gastric musculature to ageing-associated changes in stomach function and the underlying mechanisms. Klotho expression, enteric neurons, interstitial cells of Cajal (ICC), smooth muscle cells and electrical activity were assessed by immunofluorescence, confocal microscopy, 3-dimensional reconstruction, flow cytometry, quantitative RT-PCR, Western immunoblotting and intracellular recordings. Gastric emptying of solids was analysed by the [13C]octanoic acid breath test. Circulating and tissue trophic factors were measured by enzyme immunoassays and quantitative RT-PCR. The role of oxidative stress was investigated in organotypic cultures. Klotho expression was detected in gastric glands, myenteric neurons and smooth muscle cells. Progeric Klotho-deficient mice had profound loss of ICC and ICC stem cells without a significant decrease in neuron counts, expression of neuronal nitric oxide synthase or smooth muscle myosin. Slow wave amplitude and nitrergic inhibitory junction potentials were reduced while solid emptying was unchanged. Klotho-deficient mice were marantic and had low insulin, insulin-like growth factor-I and membrane-bound stem cell factor. Klotho deficiency accentuated oxidative stress and ICC loss. We conclude that Klotho-deficient, progeric mice display a gastric phenotype resembling human ageing and involving profound ICC loss. Klotho protects ICC by preserving their precursors, limiting oxidative stress, and maintaining nutritional status and normal levels of trophic factors important for ICC differentiation.
Collapse
Affiliation(s)
- Ferenc Izbeki
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Guggenheim 10, 200 1st Street SW, Rochester, MN 55906, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Age-related changes in substance P-immunoreactive nerve structures of the rat recto-anal region. Open Med (Wars) 2010. [DOI: 10.2478/s11536-009-0098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe recto-anal region is innervated by extrinsic and intrinsic nerves and a number of neuropeptides including substance P (SP) have been suggested to participate in the regulation of intestinal movements. We examined the age-related changes in the distribution of SP-immunoreactive nerve structures in the distal part of the rat large intestine. Using immunohistochemistry, the presence of SP was studied in fresh tissues from Wistar rats at different ages taken at three sampling sites, the distal rectum, anal canal and internal anal sphincter. In the 15-day old rats the myenteric plexus of the distal rectum and anal canal was well outlined by numerous SP-immunoreactive varicose nerve fibres encircling immunonegative perikarya. In the circular muscle layer, nerve fibres and small nerve bundles ran parallel to the muscle cells, while in the longitudinal muscle layer, only occasional nerve fibres were seen. At the level of the internal anal sphincter, no myenteric ganglia were present. Here, thin varicose fibers ran parallel to the smooth muscle cells. In the 3-month old rats, a larger number of intensely staining SP-immunoreactive nerve fibres were found and in the circular muscle layer, thicker nerve strands were observed. In the 26-month old rats, the density and staining intensity of SP-immunopositive nerve fibres in the myenteric plexus was lower than in the 3-month-old rats. Similar changes in the SP-immunostained fibres in the internal anal sphincter were observed. Degenerative alterations in SP-containing fibres during aging appear to play a role in ano-rectal motility and sphincter control.
Collapse
|
48
|
Phillips RJ, Walter GC, Powley TL. Age-related changes in vagal afferents innervating the gastrointestinal tract. Auton Neurosci 2010; 153:90-8. [PMID: 19665435 PMCID: PMC2818053 DOI: 10.1016/j.autneu.2009.07.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Revised: 07/11/2009] [Accepted: 07/13/2009] [Indexed: 12/16/2022]
Abstract
Recent progress in understanding visceral afferents, some of it reviewed in the present issue, serves to underscore how little is known about the aging of the visceral afferents in the gastrointestinal (GI) tract. In spite of the clinical importance of the issue-with age, GI function often becomes severely compromised-only a few initial observations on age-related structural changes of visceral afferents are available. Primary afferent cell bodies in both the nodose ganglia and dorsal root ganglia lose Nissl material and accumulate lipofucsin, inclusions, aggregates, and tangles. Additionally, in changes that we focus on in the present review, vagal visceral afferent terminals in both the muscle wall and the mucosa of the GI tract exhibit age-related structural changes. In aged animals, both of the vagal terminal types examined, namely intraganglionic laminar endings and villus afferents, exhibit dystrophic or regressive morphological changes. These neuropathies are associated with age-related changes in the structural integrity of the target organs of the affected afferents, suggesting that local changes in trophic environment may give rise to the aging of GI innervation. Given the clinical relevance of GI tract aging, a more complete understanding both of how aging alters the innervation of the gut and of how such changes might be mitigated should be made research priorities.
Collapse
Affiliation(s)
- Robert J Phillips
- Purdue University, Ingestive Behavior Research Center, Department of Psychological Sciences, West Lafayette, IN 47907-2081, USA.
| | | | | |
Collapse
|
49
|
Abstract
Colonic sensorimotor dysfunction is recognized as the principal pathophysiological mechanism underpinning chronic constipation. This review addresses current understanding derived from both human and animal studies, with particular reference made to methods of investigation.
Collapse
Affiliation(s)
- P. G. Dinning
- Department of Medicine, University of New South Wales, St George Hospital, Sydney, Australia
| | - T. K. Smith
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - S. M. Scott
- Queen Mary University London, Barts and the London School of Medicine & Dentistry, London, UK
| |
Collapse
|
50
|
Postnatal maturation of the gastrointestinal tract: A functional and immunohistochemical study in the guinea-pig ileum at weaning. Neurosci Lett 2009; 467:105-10. [DOI: 10.1016/j.neulet.2009.10.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 09/26/2009] [Accepted: 10/05/2009] [Indexed: 11/20/2022]
|