1
|
Yeoman MS, Fidalgo S, Hobby I, Hafeez A, Ranson RN, Saffrey MJ, Patel BA. Decreases in mucosally-evoked tachykinin signaling pathways can explain age-related reductions in murine colonic motility patterns. Neurogastroenterol Motil 2024; 36:e14891. [PMID: 39155460 DOI: 10.1111/nmo.14891] [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: 03/11/2024] [Revised: 06/19/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Increasing age increases the incidence of chronic constipation and fecal impaction. The contribution of the natural aging process to this phenotype is unclear. This study explored the effects of age on key motility patterns in the murine colon and determined the contribution that altered neurokinin 2 (NK2) -mediated signaling made to the aging phenotype. METHODS Mucosal reflexes, colonic migrating motor complexes (CMMCs) and colonic motility assays were explored in isolated ex vivo colons from 3, 12-14, 18- and 24-months old mice and the NK2-mediated response determined. Electrical field stimulation (EFS) or exogenous drug application were used to explore the role of the mucosa in colonic segments. KEY RESULTS Aging reduced the force of contraction of the distal colon mucosal reflex, the frequency and force of contraction of CMMCs and the NK2-mediated component of both motility patterns. Ondansetron, a 5-HT3 receptor antagonist, blocked a component of both motility patterns in full thickness but not in mucosa-free segments of the distal colon. 5, hydroxytryptamine (5-HT) and EFS-evoked NK2-dependent contractions were reduced with increasing age. Smooth muscle sensitivity to 5-HT or neurokinin A (NKA) was not altered with age. In isolated colon motility assays application of NKA decreased transit time in 24-months colon and the NK2 antagonist GR159897 increased transit times in both 3- and 24-months old colons. CONCLUSIONS AND INFERENCES Aging impairs key motility patterns in the murine colon. These changes involve a decrease in mucosally-evoked NK2-mediated signaling. Targeting NK2-mediated signaling may provide a novel approach to treating age-related motility disorders in the lower bowel.
Collapse
Affiliation(s)
- Mark S Yeoman
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sara Fidalgo
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, UK
| | - India Hobby
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, UK
| | - Ali Hafeez
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, UK
| | - Rachel N Ranson
- Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - M Jill Saffrey
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Bhavik Anil Patel
- Centre for Lifelong Health, School of Applied Sciences, University of Brighton, Brighton, UK
| |
Collapse
|
2
|
Baidoo N, Sanger GJ. The human colon: Evidence for degenerative changes during aging and the physiological consequences. Neurogastroenterol Motil 2024:e14848. [PMID: 38887160 DOI: 10.1111/nmo.14848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The incidence of constipation increases among the elderly (>65 years), while abdominal pain decreases. Causes include changes in lifestyle (e.g., diet and reduced exercise), disease and medications affecting gastrointestinal functions. Degenerative changes may also occur within the colo-rectum. However, most evidence is from rodents, animals with relatively high rates of metabolism and accelerated aging, with considerable variation in time course. In humans, cellular and non-cellular changes in the aging intestine are poorly investigated. PURPOSE To examine all available studies which reported the effects of aging on cellular and tissue functions of human isolated colon, noting the region studied, sex and age of tissue donors and study size. The focus on human colon reflects the ability to access full-thickness tissue over a wide age range, compared with other gastrointestinal regions. Details are important because of natural human variability. We found age-related changes within the muscle, in the enteric and nociceptor innervation, and in the submucosa. Some involve all regions of colon, but the ascending colon appears more vulnerable. Changes can be cell- and sublayer-dependent. Mechanisms are unclear but may include development of "senescent-like" and associated inflammaging, perhaps associated with increased mucosal permeability to harmful luminal contents. In summary, reduced nociceptor innervation can explain diminished abdominal pain among the elderly. Degenerative changes within the colon wall may have little impact on symptoms and colonic functions, because of high "functional reserve," but are likely to facilitate the development of constipation during age-related challenges (e.g., lifestyle, disease, and medications), now operating against a reduced functional reserve.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life Sciences, University of Westminster, London, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth J Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
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
|
Gałęcka I, Szyryńska N, Całka J. Influence of polyethylene terephthalate (PET) microplastic on selected active substances in the intramural neurons of the porcine duodenum. Part Fibre Toxicol 2024; 21:5. [PMID: 38321545 PMCID: PMC10845528 DOI: 10.1186/s12989-024-00566-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: 06/18/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Currently, society and industry generate huge amounts of plastics worldwide. The ubiquity of microplastics is obvious, but its impact on the animal and human organism remains not fully understood. The digestive tract is one of the first barriers between pathogens and xenobiotics and a living organism. Its proper functioning is extremely important in order to maintain homeostasis. The aim of this study was to determine the effect of microplastic on enteric nervous system and histological structure of swine duodenum. The experiment was carried out on 15 sexually immature gilts, approximately 8 weeks old. The animals were randomly divided into 3 study groups (n = 5/group). The control group received empty gelatin capsules once a day for 28 days, the first research group received daily gelatin capsules with polyethylene terephthalate (PET) particles as a mixture of particles of various sizes (maximum particle size 300 µm) at a dose of 0.1 g/animal/day. The second study group received a dose ten times higher-1 g/animal/day. RESULTS A dose of 1 g/day/animal causes more changes in the enteric nervous system and in the histological structure of duodenum. Statistically significant differences in the expression of cocaine and amphetamine regulated transcript, galanin, neuronal nitric oxide synthase, substance P, vesicular acetylcholine transporter and vasoactive intestinal peptide between control and high dose group was noted. The histopathological changes were more frequently observed in the pigs receiving higher dose of PET. CONCLUSION Based on this study it may be assumed, that oral intake of microplastic might have potential negative influence on digestive tract, but it is dose-dependent.
Collapse
Affiliation(s)
- Ismena Gałęcka
- Department of Epizootiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland.
- Deparment of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland.
| | - Natalia Szyryńska
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland
| | - Jarosław Całka
- Deparment of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719, Olsztyn, Poland
| |
Collapse
|
5
|
Masaoka T. Current Management of Chronic Constipation in Japan. Keio J Med 2023; 72:95-101. [PMID: 37612093 DOI: 10.2302/kjm.2022-0036-ir] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Constipation is a complicated condition. Chronic constipation is diagnosed when constipation occurs for more than 3 months. Chronic constipation is classified using patient symptoms and the pathophysiology. New therapeutic agents to treat chronic constipation have recently been approved in Japan. However, treatments for constipation that is refractory to traditional laxatives have been approved, an algorithm for the treatment of chronic constipation has not yet been developed. The accumulation of knowledge and data is necessary to develop a new algorithm.
Collapse
Affiliation(s)
- Tatsuhiro Masaoka
- Department of Gastroenterology and Hepatology, International University of Health and Welfare Mita Hospital, Tokyo, Japan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Lefèvre C, Le Roy C, Bessard A, Le Berre-Scoul C, Marchix J, Coron E, Le Rhun M, Brochard C, Perrouin-Verbe B, Neunlist M. Region-specific remodeling of the enteric nervous system and enteroendocrine cells in the colon of spinal cord injury patients. Sci Rep 2023; 13:16902. [PMID: 37803037 PMCID: PMC10558436 DOI: 10.1038/s41598-023-44057-y] [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: 02/01/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023] Open
Abstract
Patients with spinal cord injury (SCI) suffer from major bowel dysfunction, whose exact pathophysiology, particularly the involvement of the enteric nervous system or epithelial dysfunction is poorly understood. Herein, we aimed to characterize the mucosal biopsies of the right and left colon in SCI patients vs controls (CT): (1) remodeling of key enteric neurotransmitters, (2) remodeling of enteroendocrine cells, and (3) mucosal inflammation compared to those in controls. In SCI, mucosal ACh concentration was lower in the right colon as compared to CT, but no change was observed in the left colon, and AChE expression was lower in both the right and left colons than in CT. While the VIP concentration was similar in the right and left colons, VIP mRNA expression was increased in the right colon and decreased in the left colon, in SCI patients as compared to CT. Interestingly, 5-HT concentration was reduced in the left colon but not in the right colon in SCI patients. Moreover, in SCI patients, as compared to CT, SERT mRNA expression was selectively increased in the left colon while TPH1 mRNA expression was increased in the right and left colons. Although mucosal TNFα and IL-1β mRNA expression did not significantly differ between SCI and CT groups, we identified a significant positive correlation between TNFα and IL-1β mRNA expression and left colon transit time in the SCI group. In conclusion, region-specific changes occur in the enteric neurotransmitter, serotonergic, and inflammatory pathways in the colon of SCI patients. The significant correlations between these pathways and clinical parameters in the left colon further set a scientific basis for designing therapeutic targets to improve colonic motor dysfunction in patients.Biobank information: Spinal cord injury patients: PHRC ConstiCAPE-clinical trial NCT02566746. Controls: Anosain-clinical trial NCT03054415 and biobank of the "Institut des Maladies de l'Appareil Digestif (IMAD)" registered under number DC-2008-402.
Collapse
Affiliation(s)
- Chloë Lefèvre
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
- Service de Médecine Physique et Réadaptation Neurologique, Nantes Université, CHU Nantes, 44000, Nantes, France
| | - Camille Le Roy
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
- Service de Médecine Physique et Réadaptation Neurologique, Nantes Université, CHU Nantes, 44000, Nantes, France
| | - Anne Bessard
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
| | - Catherine Le Berre-Scoul
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
| | - Justine Marchix
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
| | - Emmanuel Coron
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
- Service de Gastroentérologie, Nantes Université, CHU Nantes, IMAD, 44000, Nantes, France
| | - Marc Le Rhun
- Service de Gastroentérologie, Nantes Université, CHU Nantes, IMAD, 44000, Nantes, France
| | - Charlène Brochard
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
- CHU Rennes, Explorations Fonctionnelles Digestives, 35000, Rennes, France
| | - Brigitte Perrouin-Verbe
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France
- Service de Médecine Physique et Réadaptation Neurologique, Nantes Université, CHU Nantes, 44000, Nantes, France
| | - Michel Neunlist
- Nantes Université, INSERM, CHU Nantes, IMAD, "The Enteric Nervous System in Gut and Brain Disorders", 44000, Nantes, France.
| |
Collapse
|
7
|
Brusciano L, Gambardella C, Falato A, Ronchi A, Tolone S, Lucido FS, Del Genio G, Gualtieri G, Terracciano G, Docimo L. Rectal Prolapse Pathological Features: Findings in Patients With Outlet Obstruction Treated With Stapled Transanal Rectal Resection. Dis Colon Rectum 2023; 66:e826-e833. [PMID: 35239529 DOI: 10.1097/dcr.0000000000002269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Obstructed defecation syndrome is a common multifactorial disease for which treatment is based primarily on clinic presentation for the lack of reliable instrumental and anatomopathological criteria. OBJECTIVE The study aimed to analyze the pathological findings of the resected rectal specimens after stapled transanal rectal resection in patients affected by outlet obstruction. DESIGN Retrospective cohort study. SETTINGS University hospital. PATIENTS Patients who underwent rectal resection for obstructed defecation syndrome due to internal rectal prolapse were included. INTERVENTIONS Specimens of patients with obstructed defecation syndrome were analyzed through conventional histology and morphometric image analysis and compared with those of rectal specimens excised for oncological diseases. MAIN OUTCOME MEASURES Analysis of the anatomopathological impairments underlying rectal prolapse. RESULTS From January 2017 to December 2020, 46 specimens from the stapled transanal rectal resection group were compared with 40 specimens from the control group. In the stapled transanal rectal resection group, conventional histology revealed 34 samples (73.9%) had moderate- to severe-grade fibrosis with moderate-grade nerve degeneration in 33 cases (71.7%). In the control group, conventional histology revealed the absence of fibrosis in 31 specimens (77.5%), whereas the absence of nerve degeneration was detected in 37 specimens (92.5%). In the stapled transanal rectal resection group, morphometric image analysis showed severe-grade fibrosis in 25 cases (54.4%) compared to only low-grade fibrosis in 11 cases (27.5%). LIMITATIONS The small sample size and the retrospective design of the study were limitations. Moreover, there was no chance to use specimens from healthy volunteers as the control group. CONCLUSIONS Stapled transanal rectal resection specimens showed a higher rate of fibrosis and nerve dysplasia, an important parameter that is typically not considered preoperatively and could lead patients with rectal prolapse to the best treatment approach. See Video Abstract at http://links.lww.com/DCR/B928 . CARACTERSTICAS ANATOMOPATOLGICAS EN EL PROLAPSO DE RECTO HALLAZGOS EN PACIENTES CON OBSTRUCCIN DEL TRACTO DE SALIDA TRATADOS CON RESECCIN RECTAL TRANSANAL CON GRAPAS ANTECEDENTES:El síndrome de obstrucción del tracto de salida, es una afección multifactorial común, cuyo tratamiento está basado principalmente en la presentación clínica, ésto, debido a la falta de criterios confiables tanto instrumentales como anatomopatológicos.OBJETIVO:Analizamos los hallazgos histopatológicos de las piezas de resección rectal obtenidas por vía transanal mediante grapas, realizadas en pacientes que presentaban obstrucción del tracto de salida.DISEÑO:Este fue un estudio de cohorte retrospectivo.AJUSTE:El escenario fue un hospital universitario.PACIENTES:Aquellos sometidos a resección rectal por síndrome obstructivo del tracto de salida causada por un prolapso rectal interno.INTERVENCIONES:Los especímenes de pacientes con síndrome obstructivo defecatorio fueron analizados mediante histología convencional y análisis de imágenes morfométricas, comparados con especímenes rectales resecados por enfermedad oncológica.PRINCIPALES MEDIDAS DE RESULTADO:El resultado primario se concentró en la investigación de las deficiencias anatomopatológicas subyacentes del prolapso rectal interno.RESULTADOS:Desde enero de 2017 hasta diciembre de 2020, se compararon 46 especímenes del grupo de resección rectal transanal con grapas con 40 especímenes del grupo de control. En histología convencional, el grupo de resección rectal transanal con grapas, 34 muestras (73,9%) presentaron un grado de fibrosis moderada-severa con un grado moderado de degeneración neurógena en 33 casos (71,7%). En el grupo control, la histología convencional reveló ausencia de fibrosis en 31 especímenes (77,5 %), mientras que la ausencia de degeneración neurógena se detectó en 37 controles (92,5 %). En el grupo de resección rectal transanal con grapas, el análisis de imágenes morfométricas mostró fibrosis moderada y fibrosis severa en 19 (41,3%) y 25 (54,4%) casos, respectivamente. En el grupo control, el análisis de imágenes morfométricas mostró solo un bajo grado de fibrosis en 11 casos (27,5%). Se encontró una diferencia significativa en todos los grados de fibrosis y displasia neurógena entre los grupos en las evaluaciones de análisis de imagen morfométrica e histología convencional (p < 0,001).LIMITACIONES:El pequeño tamaño de la muestra y el diseño retrospectivo del estudio. Además, no existe la posibilidad de utilizar un grupo de control con muestras de voluntarios sanos.CONCLUSIONES:Los especímenes de resección rectal transanal con grapas mostraron una mayor tasa de fibrosis y displasia neurógena, parámetro importante que actualmente no está considerado antes de la operación y en un futuro muy cercano podría orientar a los pacientes con prolapso rectal interno hacia un mejor enfoque de tratamiento. Consulte Video Resumen en http://links.lww.com/DCR/B928 . (Traducción-Dr. Xavier Delgadillo ).
Collapse
Affiliation(s)
- Luigi Brusciano
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Claudio Gambardella
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Armando Falato
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Andrea Ronchi
- Pathology Unit, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Salvatore Tolone
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Francesco S Lucido
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Gianmattia Del Genio
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Giorgia Gualtieri
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Gianmattia Terracciano
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Ludovico Docimo
- Division of General, Oncologic, Mininvasive and Bariatric Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
8
|
Chen BN, Humenick A, Yew WP, Peterson RA, Wiklendt L, Dinning PG, Spencer NJ, Wattchow DA, Costa M, Brookes SJH. Types of Neurons in the Human Colonic Myenteric Plexus Identified by Multilayer Immunohistochemical Coding. Cell Mol Gastroenterol Hepatol 2023; 16:573-605. [PMID: 37355216 PMCID: PMC10469081 DOI: 10.1016/j.jcmgh.2023.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND AIMS Gut functions including motility, secretion, and blood flow are largely controlled by the enteric nervous system. Characterizing the different classes of enteric neurons in the human gut is an important step to understand how its circuitry is organized and how it is affected by disease. METHODS Using multiplexed immunohistochemistry, 12 discriminating antisera were applied to distinguish different classes of myenteric neurons in the human colon (2596 neurons, 12 patients) according to their chemical coding. All antisera were applied to every neuron, in multiple layers, separated by elutions. RESULTS A total of 164 combinations of immunohistochemical markers were present among the 2596 neurons, which could be divided into 20 classes, with statistical validation. Putative functions were ascribed for 4 classes of putative excitatory motor neurons (EMN1-4), 4 inhibitory motor neurons (IMN1-4), 3 ascending interneurons (AIN1-3), 6 descending interneurons (DIN1-6), 2 classes of multiaxonal sensory neurons (SN1-2), and a small, miscellaneous group (1.8% of total). Soma-dendritic morphology was analyzed, revealing 5 common shapes distributed differentially between the 20 classes. Distinctive baskets of axonal varicosities surrounded 45% of myenteric nerve cell bodies and were associated with close appositions, suggesting possible connectivity. Baskets of cholinergic terminals and several other types of baskets selectively targeted ascending interneurons and excitatory motor neurons but were significantly sparser around inhibitory motor neurons. CONCLUSIONS Using a simple immunohistochemical method, human myenteric neurons were shown to comprise multiple classes based on chemical coding and morphology and dense clusters of axonal varicosities were selectively associated with some classes.
Collapse
Affiliation(s)
- Bao Nan Chen
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Adam Humenick
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Rochelle A Peterson
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Lukasz Wiklendt
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia; Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Nick J Spencer
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - David A Wattchow
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Marcello Costa
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Simon J H Brookes
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
9
|
Michel K, Kuch B, Dengler S, Demir IE, Zeller F, Schemann M. How big is the little brain in the gut? Neuronal numbers in the enteric nervous system of mice, Guinea pig, and human. Neurogastroenterol Motil 2022; 34:e14440. [PMID: 35929768 DOI: 10.1111/nmo.14440] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/23/2022] [Accepted: 07/21/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Despite numerous studies on the enteric nervous system (ENS), we lack fundamental knowledge on neuronal densities or total neuron numbers in different species. There are more anecdotal than actual figures on nerve counts. METHODS We used standardized preparation techniques and immunohistochemistry with validated panneuronal markers (human or mouse anti-HuD/C) to determine neuronal densities in specimen from the entire gastrointestinal tract of mice, guinea pig, and humans. In parallel, we measured the dimensions of the gastrointestinal regions in mouse and guinea pig. For humans, we had to rely on literature data. KEY RESULTS The average neuronal densities along the gastrointestinal tract were 35,011 ± 25,017 1/cm2 for the myenteric and 16,685 ± 9098 1/cm2 for the submucous plexus in mice, 24,315 ± 16,627 and 11,850 ± 6122 1/cm2 for guinea pig myenteric and submucous plexus, respectively, and 21,698 ± 9492 and 16,367 ± 5655 1/cm2 for human myenteric and submucous plexus, respectively. The total number of neurons in the ENS was 2.6 million for mice, 14.6 million for guinea pig, and 168 million for human. CONCLUSIONS & INFERENCES This study reports the first comprehensive nerve cell count in mice, guinea pig, and human ENS. Neuronal densities were comparable between the three species and the differences in the total numbers of enteric neurons are likely due to body size and intestinal length. The number of enteric neurons is comparable to the number of neurons in the spinal cord for all three species.
Collapse
Affiliation(s)
- Klaus Michel
- Human Biology, Technical University of Munich, Freising, Germany
| | - Birgit Kuch
- Human Biology, Technical University of Munich, Freising, Germany
| | - Sophie Dengler
- Human Biology, Technical University of Munich, Freising, Germany
| | - Ihsan Ekin Demir
- Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Michael Schemann
- Human Biology, Technical University of Munich, Freising, Germany
| |
Collapse
|
10
|
Khanna L, Zeydan B, Kantarci OH, Camilleri M. Gastrointestinal motility disorders in patients with multiple sclerosis: A single-center study. Neurogastroenterol Motil 2022; 34:e14326. [PMID: 35112759 PMCID: PMC9338921 DOI: 10.1111/nmo.14326] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/12/2022] [Accepted: 01/22/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Most prevalent gastrointestinal symptoms in multiple sclerosis (MS) relate to lower bowel dysfunction, often in association with bladder manifestations. OBJECTIVE To assess clinical and objective gastrointestinal motor dysfunctions in patients with MS. METHODS This was a single-center, retrospective study of 166 patients evaluated between 1996 and 2020. We reviewed characterization of the MS, gastrointestinal and neurological symptoms, measurements of gastrointestinal and colonic transit, and anorectal manometry. KEY RESULTS At the time of the gastrointestinal evaluations of the 166 patients with MS (138 women; 83%), 111 were in the relapsing-remitting phase and 52 were in the progressive phase. In 3 patients, disease phase was not assigned due to insufficient data. Constipation was identified in 82% (136/166) of patients. Most [103/116 (88%)] patients with bladder symptoms also had constipation or fecal incontinence. Delayed gastric emptying at 4 h and colonic transit at 24 h was identified in 16% and 7% of the cohort, respectively; 22% had accelerated gastric emptying. On anorectal manometry, resting anal sphincter pressure >90 mm Hg and rectoanal pressure differential below -50mm Hg suggested evacuation disorder in patients with constipation. CONCLUSIONS AND INFERENCES In addition to slow colonic transit and anorectal dysfunction leading to constipation in MS, 22% of patients had accelerated gastric emptying.
Collapse
Affiliation(s)
- Lehar Khanna
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER) Division of Gastroenterology and Hepatology Rochester MN USA
| | - Burcu Zeydan
- Department of Neurology and Center for Multiple Sclerosis and Autoimmune Neurology Mayo Clinic Rochester MN USA
| | - Orhun H. Kantarci
- Department of Neurology and Center for Multiple Sclerosis and Autoimmune Neurology Mayo Clinic Rochester MN USA
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER) Division of Gastroenterology and Hepatology Rochester MN USA
| |
Collapse
|
11
|
Characterization of Neurochemical Signature Alterations in the Enteric Nervous System in Autoimmune Encephalomyelitis. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To date, it has remained unclear whether gastrointestinal symptoms, which are frequently observed in patients with multiple sclerosis (MS), are accompanied by pathology of the enteric nervous system (ENS). Here, the neurotransmitter signature of ENS neurons and morphological alterations of interstitial cells of Cajal (ICCs) were studied in patients with MS and mice with experimental autoimmune encephalomyelitis (EAE), which is an animal model of MS. Immunohistochemical analysis was performed on colonic whole mounts from mice with EAE and on paraffin-embedded sections of intestinal tissue from patients with MS. Antibodies against neurotransmitters or their enzymes (including vasoactive intestinal peptide (VIP), neuronal nitric oxide synthase (nNOS), and choline acetyltransferase (ChAT)) were used in conjunction with pan-neuronal markers. In addition, the presence of anoctamin 1 (ANO1)-expressing ICCs was studied. ENS changes were observed in the myenteric plexus, but they were absent in the submucosal plexus of both EAE mice and patients with MS. There was a significant decrease in the percentage of ChAT-positive neurons in EAE mice as opposed to a trend toward an increase in patients with MS. Moreover, while ANO1 expression was decreased in EAE mice, patients with MS displayed a significant increase. Although additional studies are necessary to accomplish an in-depth characterization of ENS alterations in MS, our results imply that such alterations exist and may reveal novel insights into the pathophysiology of MS.
Collapse
|
12
|
Nguyen VTT, Taheri N, Chandra A, Hayashi Y. Aging of enteric neuromuscular systems in gastrointestinal tract. Neurogastroenterol Motil 2022; 34:e14352. [PMID: 35279912 PMCID: PMC9308104 DOI: 10.1111/nmo.14352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/12/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Aging is a complex biological process and associated with a progressive decline in functions of most organs including the gastrointestinal (GI) tract. Age-related GI motor disorders/dysfunctions include esophageal reflux, dysphagia, constipation, fecal incontinence, reduced compliance, and accommodation. Although the incidence and severity of these diseases and conditions increase with age, they are often underestimated due in part to nonspecific and variable symptoms and lack of sufficient medical attention. They negatively affect quality of life and predispose the elderly to other diseases, sarcopenia, and frailty. The mechanisms underlying aging-associated GI dysfunctions remain unclear, and there is limited data examining the effect of aging on GI motor functions. Many studies on aging-associated changes to cells within the tunica muscularis including enteric neurons, smooth muscles, and interstitial cells have proposed that cell loss and/or molecular changes may be involved in the pathogenesis of age-related GI motor disorders/dysfunctions. There is also evidence that the aging contributes to phenotypic changes in innate immune cells, which are physically and functionally linked to other cells in the tunica muscularis and can alter GI (patho) physiology. However, various patterns of changes have been reported, some of which are contradictory, indicating a need for additional work in this area. PURPOSE Although GI infection due to intestinal bacterial overgrowth, bleeding, and cancers are also important and common problems in the elderly patients, this mini-review focuses on data obtained from enteric neuromuscular aging research with the goal of better understanding the cellular and molecular mechanisms of enteric neuromuscular aging to enhance future therapy.
Collapse
Affiliation(s)
- Vy Truong Thuy Nguyen
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA,Gastroenterology Research Unit, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Negar Taheri
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA,Gastroenterology Research Unit, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Abhishek Chandra
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA,Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota, USA,Gastroenterology Research Unit, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
Wang F, Fei M, Hu WZ, Wang XD, Liu S, Zeng Y, Zhang JH, Lv Y, Niu JP, Meng XL, Cai P, Li Y, Gang BZ, You Y, Lv Y, Ji Y. Prevalence of Constipation in Elderly and Its Association With Dementia and Mild Cognitive Impairment: A Cross-Sectional Study. Front Neurosci 2022; 15:821654. [PMID: 35140587 PMCID: PMC8819140 DOI: 10.3389/fnins.2021.821654] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
Background Constipation and dementia have similar epidemiological characteristics. Changes in intestinal flora and characteristics of the brain-gut axis play roles in the pathogeneses of the two diseases, suggesting that there may be a close connection between the two. Most of the studies on constipation in dementia patients have focused on the population with α-synucleinopathies [Parkinson’s disease dementia (PDD), dementia with Lewy bodies (DLB)]. Few studies have reported the prevalence of constipation in all-cause dementia and mild cognitive impairment (MCI) populations. Objective To assess the prevalence of constipation in patients with all-cause dementia and MCI subtypes and to explore the association between constipation with dementia and MCI subtypes. Methods From May 2019 to December 2019, we conducted a population-based cross-sectional survey. A total of 11,743 participants aged 65 or older from nine cities in China were surveyed. Participants underwent a series of clinical examinations and neuropsychological measurements. Constipation, dementia, MCI and MCI subtype were diagnosed according to established criteria through standard diagnostic procedures. Results The overall age- and sex-adjusted prevalence of constipation in individuals aged 65 years and older was 14.8% (95% CI, 14.6–15.0). The prevalence rates of constipation were19.2% (95% CI, 17.3–21.0), 19.1% (95% CI, 16.8–21.5), 14.4% (95% CI, 12.8–15.9), and 13.8% (95% CI, 13.0–14.6) in the dementia, non-amnestic (na)-MCI, amnestic (a)-MCI and normal cognition populations, respectively. Multivariate logistic regression analysis showed that higher prevalence of constipation was associated with dementia (p = 0.0.032, OR = 1.18, 95% CI: 1.02–1.38) and na-MCI (p = 0.003, OR = 1.30, 95% CI: 1.09–1.54). Conclusion The present study found a high prevalence of constipation in elderly individuals in China, and higher in patients with dementia and na-MCI.
Collapse
Affiliation(s)
- Fei Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Yuncheng Central Hospital, Shanxi Medical University, Yuncheng, China
| | - Min Fei
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Yuncheng Central Hospital, Shanxi Medical University, Yuncheng, China
| | - Wen-Zheng Hu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiao-Dan Wang
- Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Department of Neurology, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Shuai Liu
- Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Department of Neurology, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, China
| | - Jin-Hong Zhang
- Department of Neurology, Cangzhou People’s Hospital, Cangzhou, China
| | - Yang Lv
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-ping Niu
- Department of Neurology, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, China
| | - Xin-ling Meng
- Department of Neurology, Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Urumqi, China
| | - Pan Cai
- Dementia Clinic, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yang Li
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bao-zhi Gang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong You
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yan Lv
- Department of Neurology, Hainan General Hospital, Haikou, China
| | - Yong Ji
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
- Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Department of Neurology, Tianjin Dementia Institute, Tianjin Huanhu Hospital, Tianjin, China
- *Correspondence: Yong Ji,
| |
Collapse
|
15
|
Wang Q, Qi Y, Shen W, Xu J, Wang L, Chen S, Hou T, Si J. The Aged Intestine: Performance and Rejuvenation. Aging Dis 2021; 12:1693-1712. [PMID: 34631215 PMCID: PMC8460310 DOI: 10.14336/ad.2021.0202] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Owing to the growing elderly population, age-related problems are gaining increasing attention from the scientific community. With senescence, the intestine undergoes a spectrum of changes and infirmities that are likely the causes of overall aging. Therefore, identification of the aged intestine and the search for novel strategies to rescue it, are required. Although progress has been made in research on some components of the aged intestine, such as intestinal stem cells, the comprehensive understanding of intestinal aging is still limited, and this restricts the in-depth search for efficient strategies. In this concise review, we discuss several aspects of intestinal aging. More emphasis is placed on the appraisal of current and potential strategies to alleviate intestinal aging, as well as future targets to rejuvenate the aged intestine.
Collapse
Affiliation(s)
- Qiwen Wang
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Yadong Qi
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Weiyi Shen
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Jilei Xu
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Lan Wang
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Shujie Chen
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Tongyao Hou
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Jianmin Si
- 1Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.,2Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| |
Collapse
|
16
|
Lee S, Jo K, Jeong HG, Choi YS, Yong HI, Jung S. Understanding protein digestion in infants and the elderly: Current in vitro digestion models. Crit Rev Food Sci Nutr 2021; 63:975-992. [PMID: 34346822 DOI: 10.1080/10408398.2021.1957765] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The last decades have witnessed a surge of interest in the fate of dietary proteins during gastrointestinal (GI) digestion. Although several in vitro digestion models are available as alternatives to clinical experiments, most of them focus on the digestive conditions of healthy young adults. This study investigates the static/dynamic models used to simulate digestion in infants and the elderly and considers the related in vivo conditions. The in vitro digestive protocols targeting these two groups are summarized, and the challenges associated with the further development of in vitro digestion models are discussed. Static models rely on several factors (e.g., enzyme concentration, pH, reaction time, and rotation speed) to differentiate digestive conditions depending on age. Dynamic models can more accurately simulate the complex digestion process and allow the inclusion of further parameters (sequential secretion of digestive fluids, gradual changes in pH, peristaltic mixing, GI emptying, and the inoculation of luminal microbiota). In the case of infants, age or growth stage clarification and the differentiation of digestive protocols between full-term and preterm infants are required, whereas protocols dealing with various health statuses are required in the case of the elderly, as this group is prone to oral cavity and GI function deterioration.
Collapse
Affiliation(s)
- Seonmin Lee
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| | - Kyung Jo
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun Gyung Jeong
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| | - Yun-Sang Choi
- Research Group of Food Processing, Korea Food Research Institute, Wanju, Republic of Korea
| | - Hae In Yong
- Research Group of Food Processing, Korea Food Research Institute, Wanju, Republic of Korea
| | - Samooel Jung
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
17
|
Madison AA, Kiecolt-Glaser JK. The gut microbiota and nervous system: Age-defined and age-defying. Semin Cell Dev Biol 2021; 116:98-107. [PMID: 33422403 PMCID: PMC8257779 DOI: 10.1016/j.semcdb.2020.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/01/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Even healthy older adults experience gastrointestinal (GI) and neurological changes. In fact, the aging process of these two systems are interrelated due the extensive, multifaceted communication network connecting them, termed the gut-brain axis. Age-related modification of the GI environment can influence the bacterial species that survive and thrive there. Additionally, the lifestyle common to older adults in the West, including sedentariness, polypharmacy, and a poor diet, can compound the effect of aging on the GI tract, gut microbiota, and nervous system. Emerging animal and human findings suggest that GI organisms play a major role in gut-brain communication, ultimately shaping neurological aging trajectories by either helping to maintain nervous system function into late life or promoting pathology. Aging and age-related behaviors help to define the gut microbiota's composition and function, but, conversely, the gut microbiota may help to determine late-life functionality and may be harnessed to limit the prevalence of steep neurological decline and diseases. Focusing primarily on clinical research, this review first defines the gut-brain axis, then details age-related GI and nervous system changes, and discusses the impact of age-related lifestyle factors on the GI and nervous systems. The remainder of this review describes cutting-edge research that positions the gut microbiota as an arbiter of age-related neurological decline.
Collapse
Affiliation(s)
- Annelise A Madison
- The Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, USA; Department of Psychology, The Ohio State University, USA
| | - Janice K Kiecolt-Glaser
- The Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, USA; Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, USA.
| |
Collapse
|
18
|
Joshi V, Strege PR, Farrugia G, Beyder A. Mechanotransduction in gastrointestinal smooth muscle cells: role of mechanosensitive ion channels. Am J Physiol Gastrointest Liver Physiol 2021; 320:G897-G906. [PMID: 33729004 PMCID: PMC8202201 DOI: 10.1152/ajpgi.00481.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mechanosensation, the ability to properly sense mechanical stimuli and transduce them into physiologic responses, is an essential determinant of gastrointestinal (GI) function. Abnormalities in this process result in highly prevalent GI functional and motility disorders. In the GI tract, several cell types sense mechanical forces and transduce them into electrical signals, which elicit specific cellular responses. Some mechanosensitive cells like sensory neurons act as specialized mechanosensitive cells that detect forces and transduce signals into tissue-level physiological reactions. Nonspecialized mechanosensitive cells like smooth muscle cells (SMCs) adjust their function in response to forces. Mechanosensitive cells use various mechanoreceptors and mechanotransducers. Mechanoreceptors detect and convert force into electrical and biochemical signals, and mechanotransducers amplify and direct mechanoreceptor responses. Mechanoreceptors and mechanotransducers include ion channels, specialized cytoskeletal proteins, cell junction molecules, and G protein-coupled receptors. SMCs are particularly important due to their role as final effectors for motor function. Myogenic reflex-the ability of smooth muscle to contract in response to stretch rapidly-is a critical smooth muscle function. Such rapid mechanotransduction responses rely on mechano-gated and mechanosensitive ion channels, which alter their ion pores' opening in response to force, allowing fast electrical and Ca2+ responses. Although GI SMCs express a variety of such ion channels, their identities remain unknown. Recent advancements in electrophysiological, genetic, in vivo imaging, and multi-omic technologies broaden our understanding of how SMC mechano-gated and mechanosensitive ion channels regulate GI functions. This review discusses GI SMC mechanosensitivity's current developments with a particular emphasis on mechano-gated and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Vikram Joshi
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota
| | - Peter R. Strege
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota,2Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota,2Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
19
|
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: 8] [Impact Index Per Article: 2.7] [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
|
20
|
Severity of constipation related to palonosetron during first-line chemotherapy: a retrospective observational study. Support Care Cancer 2021; 29:4723-4732. [PMID: 33515108 DOI: 10.1007/s00520-021-06023-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE Palonosetron, a long-acting 5-HT3 receptor antagonist, is an effective antiemetic agent for chemotherapy-induced nausea and vomiting; however, it sometimes causes severe constipation. The aim of the present study was to evaluate the severity of palonosetron-related constipation. METHODS We retrospectively analyzed the incidence and severity of constipation after intravenous administration of 0.75-mg palonosetron in 150 chemotherapy-naïve patients who received first-line chemotherapy at Saga University Hospital. Constipation was classified into grades 1-5 according to the Common Terminology Criteria for Adverse Events version 5.0. Multiple logistic regression analysis was performed to identify factors associated with palonosetron-related worsening of constipation to grade 2 or higher. RESULTS Palonosetron significantly increased the incidence and severity of constipation (incidence: before vs. after palonosetron, 35.4% vs. 74.0%, p < 0.0001, and severity: before vs. after palonosetron, 26.7% and 8.7% in grades 1 and 2, respectively, vs. 46.7%, 23.3%, and 4.0% in grades 1, 2, and 3, respectively, p < 0.0001). Despite the use of laxatives, 4.0% of patients had grade 3 constipation requiring manual evacuation. Combination treatment with aprepitant (odds ratio (OR), 10.9; 95% confidence interval (CI), 1.3-90.0; p = 0.026) and older age (OR, 1.25; 95% CI, 1.01-1.57; p = 0.039) were factors associated with the severity of constipation. CONCLUSION Constipation was more severe in patients receiving combination treatment with aprepitant than in those treated with palonosetron alone. Older age was also associated with increased risk of severe palonosetron-related constipation. Identification of risk factors can help target risk-based laxative therapy.
Collapse
|
21
|
Ha TS. Geriatric Physiology for Surgical Intensivists: Part I. JOURNAL OF ACUTE CARE SURGERY 2020. [DOI: 10.17479/jacs.2020.10.3.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
22
|
Walrath T, Dyamenahalli KU, Hulsebus HJ, McCullough RL, Idrovo JP, Boe DM, McMahan RH, Kovacs EJ. Age-related changes in intestinal immunity and the microbiome. J Leukoc Biol 2020; 109:1045-1061. [PMID: 33020981 DOI: 10.1002/jlb.3ri0620-405rr] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/19/2022] Open
Abstract
The gastrointestinal (GI) tract is a vitally important site for the adsorption of nutrients as well as the education of immune cells. Homeostasis of the gut is maintained by the interplay of the intestinal epithelium, immune cells, luminal Ags, and the intestinal microbiota. The well-being of the gut is intrinsically linked to the overall health of the host, and perturbations to this homeostasis can have severe impacts on local and systemic health. One factor that causes disruptions in gut homeostasis is age, and recent research has elucidated how critical systems within the gut are altered during the aging process. Intestinal stem cell proliferation, epithelial barrier function, the gut microbiota, and the composition of innate and adaptive immune responses are all altered in advanced age. The aging population continues to expand worldwide, a phenomenon referred to as the "Silver Tsunami," and every effort must be made to understand how best to prevent and treat age-related maladies. Here, recent research about changes observed in the intestinal epithelium, the intestinal immune system, the microbiota, and how the aging gut interacts with and influences other organs such as the liver, lung, and brain are reviewed. Better understanding of these age-related changes and their impact on multi-organ interactions will aid the development of therapies to increase the quality of life for all aged individuals.
Collapse
Affiliation(s)
- Travis Walrath
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Kiran U Dyamenahalli
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Holly J Hulsebus
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA.,Immunology Graduate Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Rebecca L McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, USA.,GI and Liver Innate Immune Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Juan-Pablo Idrovo
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Devin M Boe
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA.,Immunology Graduate Program, University of Colorado Denver, Aurora, Colorado, USA.,Medical Scientist Training Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Rachel H McMahan
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Elizabeth J Kovacs
- Department of Surgery, Division of GI, Trauma and Endocrine Surgery, and Burn Research Program, University of Colorado Denver, Aurora, Colorado, USA.,Immunology Graduate Program, University of Colorado Denver, Aurora, Colorado, USA.,Medical Scientist Training Program, University of Colorado Denver, Aurora, Colorado, USA.,GI and Liver Innate Immune Program, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
23
|
Becher KF, Rupp S. Darmmotilitätsstörungen bei Multimorbidität: zwischen Obstipation und Diarrhö. Dtsch Med Wochenschr 2020; 145:1456-1459. [DOI: 10.1055/a-1165-1351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Was ist Neu?
Diagnostische Strategie Die Rom-IV-Kriterien für die primäre chronische Obstipation helfen, zwischen kurzfristigen Beschwerden und einem chronischen Beschwerdebild zu unterscheiden 1. Im Allgemeinen finden sich in der Altersgruppe der Hochbetagten mehrere Ursachen für eine Störung der Defäkation. Anamnese und Beachtung von Red Flags neben einem geriatrischen Basisassessment ermöglichen es, die Ursachen einzugrenzen. Die Unterscheidung zwischen einer Low-Transit-Obstipation und normaler Darmfunktion kann die therapeutische Konsequenz entscheidend beeinflussen.
Therapeutische Möglichkeiten Bei der medikamentösen Intervention werden osmotische Abführmittel, stimulierende Abführmittel, Zäpfchen und Einläufe mit unterschiedlichen Zusätzen in erster Linie weiterhin sehr häufig verwendet. In zweiter Linie können das Prokinetikum Prucaloprid und seit einigen Jahren Methylnaltrexoniumbromid als ein Vertreter der peripher wirkenden µ-Opioid-Rezeptor-Antagonisten Anwendung finden 2
3. Faserreiche Ernährung vermehrt die Stuhlmasse und präbiotische Zusatzkost kann das Mikrobiom günstig beeinflussen. Zusätzliche Ballaststoffe haben wahrscheinlich keinen Nutzen bei schwerer Verstopfung, insbesondere bei Patienten mit Low-Transit-Obstipation oder Defäkationsstörungen. Bei Vorbereitungen auf Koloskopien scheint eine zusätzliche Bewegungsaktivierung positive Effekte auf die Qualität der Darmreinigung zu haben 4.
Collapse
Affiliation(s)
| | - Saskia Rupp
- Klinik Wartenberg, Prof. Dr. Selmair GmbH & Co.KG, Wartenberg
| |
Collapse
|
24
|
Drokhlyansky E, Smillie CS, Van Wittenberghe N, Ericsson M, Griffin GK, Eraslan G, Dionne D, Cuoco MS, Goder-Reiser MN, Sharova T, Kuksenko O, Aguirre AJ, Boland GM, Graham D, Rozenblatt-Rosen O, Xavier RJ, Regev A. The Human and Mouse Enteric Nervous System at Single-Cell Resolution. Cell 2020; 182:1606-1622.e23. [PMID: 32888429 PMCID: PMC8358727 DOI: 10.1016/j.cell.2020.08.003] [Citation(s) in RCA: 291] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/15/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
The enteric nervous system (ENS) coordinates diverse functions in the intestine but has eluded comprehensive molecular characterization because of the rarity and diversity of cells. Here we develop two methods to profile the ENS of adult mice and humans at single-cell resolution: RAISIN RNA-seq for profiling intact nuclei with ribosome-bound mRNA and MIRACL-seq for label-free enrichment of rare cell types by droplet-based profiling. The 1,187,535 nuclei in our mouse atlas include 5,068 neurons from the ileum and colon, revealing extraordinary neuron diversity. We highlight circadian expression changes in enteric neurons, show that disease-related genes are dysregulated with aging, and identify differences between the ileum and proximal/distal colon. In humans, we profile 436,202 nuclei, recovering 1,445 neurons, and identify conserved and species-specific transcriptional programs and putative neuro-epithelial, neuro-stromal, and neuro-immune interactions. The human ENS expresses risk genes for neuropathic, inflammatory, and extra-intestinal diseases, suggesting neuronal contributions to disease.
Collapse
MESH Headings
- Aging/genetics
- Aging/metabolism
- Animals
- Circadian Clocks/genetics
- Colon/cytology
- Colon/metabolism
- Endoplasmic Reticulum, Rough/genetics
- Endoplasmic Reticulum, Rough/metabolism
- Endoplasmic Reticulum, Rough/ultrastructure
- Enteric Nervous System/cytology
- Enteric Nervous System/metabolism
- Epithelial Cells/metabolism
- Female
- Gene Expression Regulation, Developmental/genetics
- Genetic Predisposition to Disease/genetics
- Humans
- Ileum/cytology
- Ileum/metabolism
- Inflammation/genetics
- Inflammation/metabolism
- Intestinal Diseases/genetics
- Intestinal Diseases/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Nervous System Diseases/genetics
- Nervous System Diseases/metabolism
- Neuroglia/cytology
- Neuroglia/metabolism
- Neurons/cytology
- Neurons/metabolism
- Nissl Bodies/genetics
- Nissl Bodies/metabolism
- Nissl Bodies/ultrastructure
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Seq
- Ribosomes/metabolism
- Ribosomes/ultrastructure
- Single-Cell Analysis/methods
- Stromal Cells/metabolism
Collapse
Affiliation(s)
- Eugene Drokhlyansky
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Maria Ericsson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gabriel K Griffin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Gokcen Eraslan
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael S Cuoco
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Olena Kuksenko
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew J Aguirre
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Genevieve M Boland
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA, USA
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
25
|
Hayashi Y, Asuzu DT, Bardsley MR, Gajdos GB, Kvasha SM, Linden DR, Nagy RA, Saravanaperumal SA, Syed SA, Toyomasu Y, Yan H, Chini EN, Gibbons SJ, Kellogg TA, Khazaie K, Kuro-o M, Machado Espindola Netto J, Singh MP, Tidball JG, Wehling-Henricks M, Farrugia G, Ordog T. Wnt-induced, TRP53-mediated Cell Cycle Arrest of Precursors Underlies Interstitial Cell of Cajal Depletion During Aging. Cell Mol Gastroenterol Hepatol 2020; 11:117-145. [PMID: 32771388 PMCID: PMC7672319 DOI: 10.1016/j.jcmgh.2020.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Gastric dysfunction in the elderly may cause reduced food intake, frailty, and increased mortality. The pacemaker and neuromodulator cells interstitial cells of Cajal (ICC) decline with age in humans, and their loss contributes to gastric dysfunction in progeric klotho mice hypomorphic for the anti-aging Klotho protein. The mechanisms of ICC depletion remain unclear. Klotho attenuates Wnt (wingless-type MMTV integration site) signaling. Here, we examined whether unopposed Wnt signaling could underlie aging-associated ICC loss by up-regulating transformation related protein TRP53 in ICC stem cells (ICC-SC). METHODS Mice aged 1-107 weeks, klotho mice, APCΔ468 mice with overactive Wnt signaling, mouse ICC-SC, and human gastric smooth muscles were studied by RNA sequencing, reverse transcription-polymerase chain reaction, immunoblots, immunofluorescence, histochemistry, flow cytometry, and methyltetrazolium, ethynyl/bromodeoxyuridine incorporation, and ex-vivo gastric compliance assays. Cells were manipulated pharmacologically and by gene overexpression and RNA interference. RESULTS The klotho and aged mice showed similar ICC loss and impaired gastric compliance. ICC-SC decline preceded ICC depletion. Canonical Wnt signaling and TRP53 increased in gastric muscles of klotho and aged mice and middle-aged humans. Overstimulated canonical Wnt signaling increased DNA damage response and TRP53 and reduced ICC-SC self-renewal and gastric ICC. TRP53 induction persistently inhibited G1/S and G2/M cell cycle phase transitions without activating apoptosis, autophagy, cellular quiescence, or canonical markers/mediators of senescence. G1/S block reflected increased cyclin-dependent kinase inhibitor 1B and reduced cyclin D1 from reduced extracellular signal-regulated kinase activity. CONCLUSIONS Increased Wnt signaling causes age-related ICC loss by up-regulating TRP53, which induces persistent ICC-SC cell cycle arrest without up-regulating canonical senescence markers.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota,Yujiro Hayashi, PhD, Mayo Clinic, Guggenheim 10, 200 First Street SW, Rochester, Minnesota 55906. fax: (507) 255-6318.
| | - David T. Asuzu
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Bardsley
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gabriella B. Gajdos
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sergiy M. Kvasha
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Rea A. Nagy
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Siva Arumugam Saravanaperumal
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sabriya A. Syed
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Toyomasu
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Huihuang Yan
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Eduardo N. Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center and Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | | | - Makoto Kuro-o
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas,Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Jair Machado Espindola Netto
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center and Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - James G. Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | | | - Gianrico Farrugia
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Tamas Ordog
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota,Correspondence Address correspondence to: Tamas Ordog, MD, Mayo Clinic, Guggenheim 10, 200 First Street SW, Rochester, Minnesota 55906. fax: (507) 255-6318.
| |
Collapse
|
26
|
Graham KD, López SH, Sengupta R, Shenoy A, Schneider S, Wright CM, Feldman M, Furth E, Valdivieso F, Lemke A, Wilkins BJ, Naji A, Doolin E, Howard MJ, Heuckeroth RO. Robust, 3-Dimensional Visualization of Human Colon Enteric Nervous System Without Tissue Sectioning. Gastroenterology 2020; 158:2221-2235.e5. [PMID: 32113825 PMCID: PMC7392351 DOI: 10.1053/j.gastro.2020.02.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Small, 2-dimensional sections routinely used for human pathology analysis provide limited information about bowel innervation. We developed a technique to image human enteric nervous system (ENS) and other intramural cells in 3 dimensions. METHODS Using mouse and human colon tissues, we developed a method that combines tissue clearing, immunohistochemistry, confocal microscopy, and quantitative analysis of full-thickness bowel without sectioning to quantify ENS and other intramural cells in 3 dimensions. RESULTS We provided 280 adult human colon confocal Z-stacks from persons without known bowel motility disorders. Most of our images were of myenteric ganglia, captured using a 20× objective lens. Full-thickness colon images, viewed with a 10× objective lens, were as large as 4 × 5 mm2. Colon from 2 pediatric patients with Hirschsprung disease was used to show distal colon without enteric ganglia, as well as a transition zone and proximal pull-through resection margin where ENS was present. After testing a panel of antibodies with our method, we identified 16 antibodies that bind to molecules in neurons, glia, interstitial cells of Cajal, and muscularis macrophages. Quantitative analyses demonstrated myenteric plexus in 24.5% ± 2.4% of flattened colon Z-stack area. Myenteric ganglia occupied 34% ± 4% of myenteric plexus. Single myenteric ganglion volume averaged 3,527,678 ± 573,832 mm3 with 38,706 ± 5763 neuron/mm3 and 129,321 ± 25,356 glia/mm3. Images of large areas provided insight into why published values of ENS density vary up to 150-fold-ENS density varies greatly, across millimeters, so analyses of small numbers of thin sections from the same bowel region can produce varying results. Neuron subtype analysis revealed that approximately 56% of myenteric neurons stained with neuronal nitric oxide synthase antibody and approximately 33% of neurons produce and store acetylcholine. Transition zone regions from colon tissues of patients with Hirschsprung disease had ganglia in multiple layers and thick nerve fiber bundles without neurons. Submucosal neuron distribution varied among imaged colon regions. CONCLUSIONS We developed a 3-dimensional imaging method for colon that provides more information about ENS structure than tissue sectioning. This approach could improve diagnosis for human bowel motility disorders and may be useful for other bowel diseases as well.
Collapse
Affiliation(s)
- Kahleb D. Graham
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Cincinnati Children’s Hospital Medical Center and the Department of Pediatrics at University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Silvia Huerta López
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318
| | - Rajarshi Sengupta
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,American Association for Cancer Research, 615 Chestnut Street, 17th Floor, Philadelphia, PA 19106-4404
| | - Archana Shenoy
- Department of Pathology, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A., 19104-4318
| | - Sabine Schneider
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| | - Christina M. Wright
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Emma Furth
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Federico Valdivieso
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Amanda Lemke
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318
| | - Benjamin J. Wilkins
- Department of Pathology, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A., 19104-4318
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104-4318
| | - Edward Doolin
- Pediatric General, Thoracic and Fetal Surgery, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A. 19104-4318
| | - Marthe J. Howard
- Department of Neurosciences, University of Toledo, Mail Stop # 1007, 3000 Arlington Avenue, Toledo, OH, U.S.A, 43614-2598
| | - Robert O. Heuckeroth
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| |
Collapse
|
27
|
Baker C, Ahmed M, Cheng K, Arciero E, Bhave S, Natalie Ho WL, Goldstein AM, Hotta R. Hypoganglionosis in the gastric antrum causes delayed gastric emptying. Neurogastroenterol Motil 2020; 32:e13766. [PMID: 31773831 PMCID: PMC7182502 DOI: 10.1111/nmo.13766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/03/2019] [Accepted: 10/24/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Enteric nervous system (ENS) abnormalities have been implicated in delayed gastric emptying but studies exploring potential treatment options are limited by the lack of an experimental animal model. We examined the ENS abnormalities in the mouse stomach associated with aging, developed a novel model of gastroparesis, and established a new approach to measure gastric emptying. METHODS A modified gastric emptying assay was developed, validated in nNOS -/- mice, and tested in mice at multiple ages. Age-related changes in ENS structure were analyzed by immunohistochemistry. Gastric aganglionosis was generated in Wnt1-iDTR mice using focal administration of diphtheria toxin (DT) into the anterior antral wall. KEY RESULTS Older mice (>5 months) exhibit hypoganglionosis in the gastric antrum and a decreased proportion of nNOS neurons as compared to younger mice (age 5-7 weeks). This was associated with a significant age-dependent decrease in liquid and solid gastric emptying. A novel model of gastric antrum hypoganglionosis was established using neural crest-specific expression of diphtheria toxin receptor. In this model, a significant reduction in liquid and solid gastric emptying is observed. CONCLUSIONS & INFERENCES Older mice exhibit delayed gastric emptying associated with hypoganglionosis and a reduction in nNOS-expressing neurons in the antrum. The causal relationship between antral hypoganglionosis and delayed gastric emptying was verified using a novel experimental model of ENS ablation. This study provides new information regarding the pathogenesis of delayed gastric emptying and provides a robust model system to study this disease and develop novel treatments.
Collapse
Affiliation(s)
- Corey Baker
- Department of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Minhal Ahmed
- Department of Bioengineering, Northeastern University, Boston MA 02115
| | - Katarina Cheng
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Emily Arciero
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Sukhada Bhave
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Wing Lam Natalie Ho
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| |
Collapse
|
28
|
Mari A, Mahamid M, Amara H, Baker FA, Yaccob A. Chronic Constipation in the Elderly Patient: Updates in Evaluation and Management. Korean J Fam Med 2020; 41:139-145. [PMID: 32062960 PMCID: PMC7272371 DOI: 10.4082/kjfm.18.0182] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/19/2018] [Indexed: 01/09/2023] Open
Abstract
Chronic constipation (CC) is a common disorder in the elderly population globally and is associated with comorbidities and negative implications on the quality of life. Constipation prevalence varies in different studies, primarily owing to the nonuniformity of the diagnostic criteria. However, 15%-30% of individuals aged >60 years are diagnosed with CC. Primary care physicians are the main healthcare providers that manage constipation in elderly patients in parallel with increased population aging and increased prevalence of constipation. Physical inactivity, polypharmacy, chronic medical conditions, rectal hyposensitivity, and defecatory disorders all play a role in the pathogenesis of CC in elderly patients. Detailed anamnesis, particularly history related to chronic medication use, with digital rectal examination may assist in identifying constipation causes. Additionally, blood tests and colonoscopy may identify organic causes of CC. Physiologic tests (i.e., anorectal manometry, colonic transit time with radiopaque markers, and defecography) can evaluate the physiologic function of the colon, rectum, and anus. However, generally, there are several causes of constipation in older patients, and an individualized approach is recommended. Treatment of chronic idiopathic constipation is empiric, based on the stepwise approach. Lifestyle advice, adjustment of chronic medications, and prescription of laxatives are the first steps of management. Several laxatives are available, and the treatment is evolving in the last decade. Biofeedback is an effective therapy especially for defecatory disorders. This review aimed to summarize the most updated knowledge for primary care physicians in the approach and management of CC in elderly patients.
Collapse
Affiliation(s)
- Amir Mari
- Gastroenterology Institute, Nazareth EMMS Hospital, Nazareth, Israel.,The Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Mahmud Mahamid
- Gastroenterology Institute, Nazareth EMMS Hospital, Nazareth, Israel.,The Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Hana Amara
- Gastroenterology Institute, Nazareth EMMS Hospital, Nazareth, Israel.,The Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Fadi Abu Baker
- Gastroenterology Institute, Hillel Yaffe Medical Center, Rappaport Faculty of Medicine, Haifa, Israel
| | - Afif Yaccob
- Gastroenterology and Liver Disease Department, Rambam Healthcare Campus, Haifa, Israel
| |
Collapse
|
29
|
Schemann M, Frieling T, Enck P. To learn, to remember, to forget-How smart is the gut? Acta Physiol (Oxf) 2020; 228:e13296. [PMID: 31063665 DOI: 10.1111/apha.13296] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022]
Abstract
The enteric nervous system (ENS) resides within the gut wall and autonomously controls gut functions through coordinated activation of sensory, inter and motor neurons. Its activity is modulated by the enteric immune and endocrine system as well as by afferent and efferent nerves of the parasympathetic and sympathetic nervous system. The ENS is often referred to as the second brain and hence is able to perform sophisticated tasks. We review the evidence that the "smartness" of the ENS may even extend to its ability to learn and to memorize. Examples for habituation, sensitization, conditioned behaviour and long-term facilitation are evidence for various forms of implicit learning. Moreover, we discuss how this may change not only basic Neurogastroenterology but also our understanding of development of gut diseases and chronic disorders in gut functions. At the same time, we identify open questions and future challenges to confirm learning, memory and memory deficits in the gut. Despite some remaining experimental challenges, we are convinced that the gut is able to learn and are tempted to answer the question with: Yes, the gut is smart.
Collapse
Affiliation(s)
| | | | - Paul Enck
- Department of Internal Medicine VI, Psychosomatic Medicine and Psychotherapy University Hospital Tübingen Tübingen Germany
| |
Collapse
|
30
|
Sun T, Li D, Hu S, Huang L, Sun H, Yang S, Wu B, Ji F, Zhou D. Aging-dependent decrease in the numbers of enteric neurons, interstitial cells of Cajal and expression of connexin43 in various regions of gastrointestinal tract. Aging (Albany NY) 2019; 10:3851-3865. [PMID: 30530917 PMCID: PMC6326649 DOI: 10.18632/aging.101677] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/18/2018] [Indexed: 12/20/2022]
Abstract
Aging is a significant risk factor for gastrointestinal dysmotility, but aging-associated differences between different organs and the exact time to start degenerating have remained obscure. Here we evaluated alterations of interstitial cells of Cajal, enteric neurons and connexin43 expression in the stomach, jejunum and colon in 2-, 12-, 16-, 20- and 24-month-old mice, as well as in aged human colon. Interstitial cells of Cajal, cholinergic and nitrergic neurons within the whole digestive tract were reduced over time, but their loss first appeared in stomach, then in intestine, helping to understand that gastric function was first impaired during aging. The decrease of connexin43 expression occurred before interstitial cells of Cajal and neurons loss, suggesting that connexin43 might be the major target influenced during senescence. Furthermore, changes in expressions of pro-inflammatory cytokines (tumour necrosis factor-α, interleukin-1β, interleukin-6) and apoptosis-related proteins (B-cell lymphoma-2, caspase-3) which indicated “inflammaging”, might contribute to the loss of enteric neurons and interstitial cells of Cajal in aged gastrointestinal tract. Our results provide possible therapeutic time window for beneficial intervention for geriatric patients with gastrointestinal motility disorders.
Collapse
Affiliation(s)
- Tingyi Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.,Cancer Institute of Capital Medical University, Beijing, 100069, China
| | - Dandan Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Shilong Hu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Li Huang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Haimei Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.,Cancer Institute of Capital Medical University, Beijing, 100069, China
| | - Shu Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.,Cancer Institute of Capital Medical University, Beijing, 100069, China
| | - Bo Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.,Cancer Institute of Capital Medical University, Beijing, 100069, China
| | - Fengqing Ji
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.,Cancer Institute of Capital Medical University, Beijing, 100069, China
| | - Deshan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.,Cancer Institute of Capital Medical University, Beijing, 100069, China
| |
Collapse
|
31
|
Boschetti E, Malagelada C, Accarino A, Malagelada JR, Cogliandro RF, Gori A, Bonora E, Giancola F, Bianco F, Tugnoli V, Clavenzani P, Azpiroz F, Stanghellini V, Sternini C, De Giorgio R. Enteric neuron density correlates with clinical features of severe gut dysmotility. Am J Physiol Gastrointest Liver Physiol 2019; 317:G793-G801. [PMID: 31545923 PMCID: PMC6962493 DOI: 10.1152/ajpgi.00199.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) symptoms can originate from severe dysmotility due to enteric neuropathies. Current methods used to demonstrate enteric neuropathies are based mainly on classic qualitative histopathological/immunohistochemical evaluation. This study was designed to identify an objective morphometric method for paraffin-embedded tissue samples to quantify the interganglionic distance between neighboring myenteric ganglia immunoreactive for neuron-specific enolase, as well as the number of myenteric and submucosal neuronal cell bodies/ganglion in jejunal specimens of patients with severe GI dysmotility. Jejunal full-thickness biopsies were collected from 32 patients (22 females; 16-77 yr) with well-characterized severe dysmotility and 8 controls (4 females; 47-73 yr). A symptom questionnaire was filled before surgery. Mann-Whitney U test, Kruskal-Wallis coupled with Dunn's posttest and nonparametric linear regression tests were used for analyzing morphometric data and clinical correlations, respectively. Compared with controls, patients with severe dysmotility exhibited a significant increase in myenteric interganglionic distance (P = 0.0005) along with a decrease in the number of myenteric (P < 0.00001) and submucosal (P < 0.0004) neurons. A 50% reduction in the number of submucosal and myenteric neurons correlated with an increased interganglionic distance and severity of dysmotility. Our study proposes a relatively simple tool that can be applied for quantitative evaluation of paraffin sections from patients with severe dysmotility. The finding of an increased interganglionic distance may aid diagnosis and limit the direct quantitative analysis of neurons per ganglion in patients with an interganglionic distance within the control range.NEW & NOTEWORTHY Enteric neuropathies are challenging conditions characterized by a severe impairment of gut physiology, including motility. An accurate, unambiguous assessment of enteric neurons provided by quantitative analysis of routine paraffin sections may help to define neuropathy-related gut dysmotility. We showed that patients with severe gut dysmotility exhibited an increased interganglionic distance associated with a decreased number of myenteric and submucosal neurons, which correlated with symptoms and clinical manifestations of deranged intestinal motility.
Collapse
Affiliation(s)
- Elisa Boschetti
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Carolina Malagelada
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | - Anna Accarino
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | - Juan R. Malagelada
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | | | - Alessandra Gori
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elena Bonora
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fiorella Giancola
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bianco
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vitaliano Tugnoli
- 3Department of Biomedical and Neuro Motor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Clavenzani
- 4Department of Veterinary Medicine, University of Bologna, Ozzano, Italy
| | - Fernando Azpiroz
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | | | - Catia Sternini
- 5Digestive Disease Division, Departments of Medicine and Neurobiology, University of California, Los Angeles, California
| | | |
Collapse
|
32
|
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: 1.0] [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
|
33
|
Broad J, Kung VWS, Palmer A, Elahi S, Karami A, Darreh-Shori T, Ahmed S, Thaha MA, Carroll R, Chin-Aleong J, Martin JE, Saffrey MJ, Knowles CH, Sanger GJ. Changes in neuromuscular structure and functions of human colon during ageing are region-dependent. Gut 2019; 68:1210-1223. [PMID: 30228216 PMCID: PMC6594449 DOI: 10.1136/gutjnl-2018-316279] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To determine if human colonic neuromuscular functions decline with increasing age. DESIGN Looking for non-specific changes in neuromuscular function, a standard burst of electrical field stimulation (EFS) was used to evoke neuronally mediated (cholinergic/nitrergic) contractions/relaxations in ex vivomuscle strips of human ascending and descending colon, aged 35-91 years (macroscopically normal tissue; 239 patients undergoing cancer resection). Then, to understand mechanisms of change, numbers and phenotype of myenteric neurons (30 306 neurons stained with different markers), densities of intramuscular nerve fibres (51 patients in total) and pathways involved in functional changes were systematically investigated (by immunohistochemistry and use of pharmacological tools) in elderly (≥70 years) and adult (35-60 years) groups. RESULTS With increasing age, EFS was more likely to evoke muscle relaxation in ascending colon instead of contraction (linear regression: n=109, slope 0.49%±0.21%/year, 95% CI), generally uninfluenced by comorbidity or use of medications. Similar changes were absent in descending colon. In the elderly, overall numbers of myenteric and neuronal nitric oxide synthase-immunoreactive neurons and intramuscular nerve densities were unchanged in ascending and descending colon, compared with adults. In elderly ascending, not descending, colon numbers of cell bodies exhibiting choline acetyltransferase immunoreactivity increased compared with adults (5.0±0.6 vs 2.4±0.3 neurons/mm myenteric plexus, p=0.04). Cholinergically mediated contractions were smaller in elderly ascending colon compared with adults (2.1±0.4 and 4.1±1.1 g-tension/g-tissue during EFS; n=25/14; p=0.04); there were no changes in nitrergic function or in ability of the muscle to contract/relax. Similar changes were absent in descending colon. CONCLUSION In ascending not descending colon, ageing impairs cholinergic function.
Collapse
Affiliation(s)
- John Broad
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Victor W S Kung
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexandra Palmer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shezan Elahi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Azadeh Karami
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, NEO, Stockholm, Sweden
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, NEO, Stockholm, Sweden
| | - Shafi Ahmed
- Department of Surgery, Barts Health NHS Trust, The Royal London Hospital, London, UK,Department of Pathology, Barts Health NHS Trust, The Royal London Hospital, London, UK
| | - Mohamed Adhnan Thaha
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK,Department of Surgery, Barts Health NHS Trust, The Royal London Hospital, London, UK,Department of Pathology, Barts Health NHS Trust, The Royal London Hospital, London, UK
| | - Rebecca Carroll
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joanne Chin-Aleong
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joanne E Martin
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - M Jill Saffrey
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes, UK
| | - Charles H Knowles
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK,Department of Surgery, Barts Health NHS Trust, The Royal London Hospital, London, UK,Department of Pathology, Barts Health NHS Trust, The Royal London Hospital, London, UK
| | - Gareth John Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
34
|
Age and Sex-Dependent Differences in the Neurochemical Characterization of Calcitonin Gene-Related Peptide-Like Immunoreactive (CGRP-LI) Nervous Structures in the Porcine Descending Colon. Int J Mol Sci 2019; 20:ijms20051024. [PMID: 30818742 PMCID: PMC6429317 DOI: 10.3390/ijms20051024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 12/26/2022] Open
Abstract
Neurons of the enteric nervous system (ENS) may undergo changes during maturation and aging, but knowledge of physiological stimuli-dependent changes in the ENS is still fragmentary. On the other hand, the frequency of many ENS-related intestinal illnesses depends on age and/or sex. The double immunofluorescence technique was used to study the influence of both of these factors on calcitonin gene-related peptide (CGRP)—positive enteric nervous structures—in the descending colon in young and adult female and castrated male pigs. The influence of age and gender on the number and neurochemical characterization (i.e., co-localization of CGRP with substance P, nitric oxide synthase, galanin, cocaine- and amphetamine-regulated transcript peptide and vesicular acetylcholine transporter) of CGRP-positive nerve structures in the colonic wall has been shown. These observations strongly suggest the participation of CGRP in adaptive processes in the ENS during GI tract maturation. Moreover, although the castration of males may mask some aspects of sex-dependent influences on the ENS, the sex-specific differences in CGRP-positive nervous structures were mainly visible in adult animals. This may suggest that the distribution and exact role of this substance in the ENS depend on the sex hormones.
Collapse
|
35
|
Dumic I, Nordin T, Jecmenica M, Stojkovic Lalosevic M, Milosavljevic T, Milovanovic T. Gastrointestinal Tract Disorders in Older Age. Can J Gastroenterol Hepatol 2019; 2019:6757524. [PMID: 30792972 PMCID: PMC6354172 DOI: 10.1155/2019/6757524] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023] Open
Abstract
Considering an increase in the life expectancy leading to a rise in the elderly population, it is important to recognize the changes that occur along the process of aging. Gastrointestinal (GI) changes in the elderly are common, and despite some GI disorders being more prevalent in the elderly, there is no GI disease that is limited to this age group. While some changes associated with aging GI system are physiologic, others are pathological and particularly more prevalent among those above age 65 years. This article reviews the most important GI disorders in the elderly that clinicians encounter on a daily basis. We highlight age-related changes of the oral cavity, esophagus, stomach, small and large bowels, and the clinical implications of these changes. We review epidemiology and pathophysiology of common diseases, especially as they relate to clinical manifestation in elderly. Details regarding management of specific disease are discussed in detail if they significantly differ from the management for younger groups or if they are associated with significant challenges due to side effects or polypharmacy. Cancers of GI tract are not included in the scope of this article.
Collapse
Affiliation(s)
- Igor Dumic
- 1Division of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI, USA
- 2Mayo Clinic College of Medicine and Sciences, Rochester, MN, USA
| | - Terri Nordin
- 2Mayo Clinic College of Medicine and Sciences, Rochester, MN, USA
- 3Department of Family Medicine, Mayo Clinic Health System, Eau Claire WI, USA
| | - Mladen Jecmenica
- 4Gastroenterology Fellowship Program, The Wright Center for Graduate Medical Education, Scranton, PA, USA
| | | | - Tomica Milosavljevic
- 5Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Belgrade, Serbia
- 6School of Medicine, Belgrade University, Belgrade, Serbia
| | - Tamara Milovanovic
- 5Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, Belgrade, Serbia
- 6School of Medicine, Belgrade University, Belgrade, Serbia
| |
Collapse
|
36
|
Bianco F, Eisenman ST, Colmenares Aguilar MG, Bonora E, Clavenzani P, Linden DR, De Giorgio R, Farrugia G, Gibbons SJ. Expression of RAD21 immunoreactivity in myenteric neurons of the human and mouse small intestine. Neurogastroenterol Motil 2018; 30:e13429. [PMID: 30069982 PMCID: PMC6150808 DOI: 10.1111/nmo.13429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND RAD21 is a double-strand-break repair protein and component of the cohesin complex with key roles in cellular functions. A RAD21 loss-of-function mutation was found in cases of chronic intestinal pseudo-obstruction (CIPO) with associated enteric neuronal loss. Analysis of RAD21 expression in the enteric nervous system is lacking, thus we aimed to characterize RAD21 immunoreactivity (IR) in myenteric ganglia. METHODS Double labeling immunofluorescence in mouse and human jejunum was used to determine colocalization of RAD21 with HuC/D, PGP9.5, neuronal nitric oxide synthase (nNOS), neuropeptide Y (NPY), choline acetyl transferase (ChAT), Kit, platelet-derived growth factor receptor-α (PDGFRα), and glial fibrillary acid protein (GFAP) IRs. RESULTS A subset of PGP9.5- and HuC/D-IR neuronal cell bodies and nerve fibers in the myenteric plexus of human and mouse small intestine also displayed cytoplasmic RAD21-IR Cytoplasmic RAD21-IR was found in 43% of HuC/D-IR neurons in adult and neonatal mice but did not colocalize with nNOS. A subset of ChAT-positive neurons had cytoplasmic RAD21-IR Punctate RAD21-IR was restricted to the nucleus in most cell types consistent with labeling of the cohesin complex. Cytoplasmic RAD21-IR was not detected in interstitial cells of Cajal, fibroblast-like cells or glia. Subsets of neurons in primary culture exhibited cytoplasmic RAD21-IR Suppression of RAD21 expression by shRNA knockdown abolished RAD21-IR in cultured neurons. CONCLUSIONS Our data showing cytoplasmic RAD21 expression in enteric neurons provide a basis toward understanding how mutations of this gene may contribute to altered neuronal function/survival thus leading to gut-motor abnormalities.
Collapse
Affiliation(s)
- F Bianco
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | - S T Eisenman
- Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - M G Colmenares Aguilar
- Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - E Bonora
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - P Clavenzani
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | - D R Linden
- Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - R De Giorgio
- Department of Medical Sciences, Nuovo Arcispedale S.Anna, University of Ferrara, Ferrara, Italy
| | - G Farrugia
- Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - S J Gibbons
- Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
37
|
Aubert P, Oleynikova E, Rizvi H, Ndjim M, Le Berre-Scoul C, Grohard PA, Chevalier J, Segain JP, Le Drean G, Neunlist M, Boudin H. Maternal protein restriction induces gastrointestinal dysfunction and enteric nervous system remodeling in rat offspring. FASEB J 2018; 33:770-781. [DOI: 10.1096/fj.201800079r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Philippe Aubert
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Elena Oleynikova
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Hina Rizvi
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Marième Ndjim
- Institute National de la Recherche Agronomique (INRA) Unité Mixte de Recherche 1280Physiologie des Adaptations Nutritionnelles (PhAN)Institut des Maladies de l'Appareil Digestif Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Catherine Le Berre-Scoul
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Pierre Antoine Grohard
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Julien Chevalier
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Jean-Pierre Segain
- Institute National de la Recherche Agronomique (INRA) Unité Mixte de Recherche 1280Physiologie des Adaptations Nutritionnelles (PhAN)Institut des Maladies de l'Appareil Digestif Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Gwenola Le Drean
- Institute National de la Recherche Agronomique (INRA) Unité Mixte de Recherche 1280Physiologie des Adaptations Nutritionnelles (PhAN)Institut des Maladies de l'Appareil Digestif Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| | - Helene Boudin
- The Enteric Nervous System in Gut and Brain DisordersINSERMUniversité de Nantes Nantes France
- Centre de Recherche en Nutrition Humaine Grand Ouest Nantes France
| |
Collapse
|
38
|
Muraoka K, Morizane S, Hieda K, Honda M, Sejima T, Murakami G, Abe SI, Takenaka A. Site-dependent differences in the composite fibers of male pelvic plexus branches: an immunohistochemical analysis of donated elderly cadavers. BMC Urol 2018; 18:47. [PMID: 29789007 PMCID: PMC5964883 DOI: 10.1186/s12894-018-0369-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/16/2018] [Indexed: 11/25/2022] Open
Abstract
Background Although the pelvic autonomic plexus branches are considered to be a mixture of sympathetic and parasympathetic nerves, little is known regarding the composite fibers of the pelvic plexus branches. This study aimed to investigate the immunohistochemical features of sympathetic and parasympathetic nerves in the pelvic autonomic plexus branches. Methods Using 10 donated elderly male cadavers, the detailed topohistology of nerve fibers at and around the bladder, seminal vesicle, prostate, and rectum was examined. Neuronal nitric oxide synthase (nNOS) and vasoactive intestinal polypeptide (VIP) were used as parasympathetic nerve markers; tyrosine hydroxylase (TH) was used as a sympathetic nerve marker. The myenteric plexus of the colon was utilized as a positive control. Results Most nerve fibers in the bladder, seminal vesicle, prostate, and rectum were both nNOS- and TH-positive. Thus, pelvic plexus branches were classified into two types: 1) triple-positive mixed nerves (nNOS+, VIP+, TH+, thick myelinated fibers + or -) and 2) double-positive mixed nerves (nNOS+, VIP-, TH+, thick myelinated fibers + or -). Notably, triple-positive nerves were localized within the posterosuperior part of the plexus (near the rectum) and travelled anteroinferiorly toward the posterolateral corner of the prostate. The posteriorly and inferiorly located nerves were predominantly composed of parasympathetic, rather than sympathetic, fibers. In contrast, nerve fibers within and along the bladder and seminal vesicle contained either no or few VIP-positive nerves. These superiorly located nerves were characterized by clear sympathetic nerve dominance. Conclusions The nerves of the pelvic plexus branches were clearly classified into nerves around the bladder and seminal vesicle (VIP-negative) and nerves around the prostate (VIP-positive). Although nNOS- and VIP-positive nerve fibers are candidate cavernous nerves, cavernous nerve identity cannot be definitively concluded for these nerves in the periprostatic region.
Collapse
Affiliation(s)
- Kuniyasu Muraoka
- Department of Urology, Tottori University Faculty of Medicine, Yonago, Japan. .,Division of Urology, Department of Surgery, Tottori University Faculty of Medicine, 36-1 Nishi-cho, Yonago, 683-8504, Japan.
| | - Shuichi Morizane
- Department of Urology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Keisuke Hieda
- Department of Urology, Hiroshima University Faculty of Medicine, Hiroshima, Japan
| | - Masashi Honda
- Department of Urology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Takehiro Sejima
- Department of Urology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Gen Murakami
- Division of Internal Medicine, Iwamizawa Kojin-kai Hospital, Iwamizawa, Japan
| | - Shin-Ichi Abe
- Department of Anatomy, Tokyo Dental College, Tokyo, Japan
| | - Atsushi Takenaka
- Department of Urology, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
39
|
Ng KS, Montes-Adrian NA, Mahns DA, Gladman MA. Quantification and neurochemical coding of the myenteric plexus in humans: No regional variation between the distal colon and rectum. Neurogastroenterol Motil 2018; 30. [PMID: 28836741 DOI: 10.1111/nmo.13193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND It remains unclear whether regional variation exists in the human enteric nervous system (ENS) ie, whether intrinsic innervation varies along the gut. Recent classification of gastrointestinal neuropathies has highlighted inadequacies in the quantification of the human ENS. This study used paired wholemounts to accurately quantify and neurochemically code the hindgut myenteric plexus, comparing human distal colon and rectum. METHODS Paired human descending colonic/rectal specimens were procured from 15 patients undergoing anterior resection. Wholemounts of myenteric plexi were triple-immunostained with anti-Hu/NOS/ChAT antibodies. Images were acquired by motorized epifluorescence microscopy, allowing assessment of ganglionic density/size, ganglionic area density, and neuronal density. 'Stretch-corrected' values were calculated using stretched/relaxed tissue dimensions. KEY RESULTS Tile-stitching created a collage with average area 99 300 000 μm2 . Stretch-corrected ganglionic densities were similar (colon: median 510 ganglia/100 mm2 [range 386-1170], rectum: 585 [307-923]; P = .99), as were average ganglionic sizes (colon: 57 593 μm2 [40 301-126 579], rectum: 54 901 [38 701-90 211], P = .36). Ganglionic area density (colon: 11.92 mm2 per 100 mm2 [7.53-18.64], rectum: 9.84 [5.80-17.19], P = .10) and stretch-corrected neuronal densities (colon: 189 neurons/mm2 [117-388], rectum: 182 [89-361], P = .31) were also similar, as were the neurochemical profiles of myenteric ganglia, with comparable proportions of NOS+ and ChAT+ neurons (P > .10). CONCLUSIONS AND INFERENCES This study has revealed similar neuronal and ganglionic densities and neurochemical profiles in human distal colon and rectum. Further investigation of other components of the ENS, incorporating additional immunohistochemical markers are required to confirm that there is no regional variation in the human hindgut ENS.
Collapse
Affiliation(s)
- K-S Ng
- Academic Colorectal Unit, Sydney Medical School - Concord, University of Sydney, Concord, New South Wales, Australia
| | - N A Montes-Adrian
- Academic Colorectal Unit, Sydney Medical School - Concord, University of Sydney, Concord, New South Wales, Australia
| | - D A Mahns
- Department of Integrative Physiology, School of Medicine, University of Western Sydney, Campbelltown, New South Wales, Australia
| | - M A Gladman
- Academic Colorectal Unit, Sydney Medical School - Concord, University of Sydney, Concord, New South Wales, Australia.,Enteric Neuroscience and Gastrointestinal Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
40
|
Kim HJ, Kim N, Kim YS, Nam RH, Lee SM, Park JH, Choi D, Hwang YJ, Lee J, Lee HS, Kim MS, Lee MY, Lee DH. Changes in the interstitial cells of Cajal and neuronal nitric oxide synthase positive neuronal cells with aging in the esophagus of F344 rats. PLoS One 2017; 12:e0186322. [PMID: 29182640 PMCID: PMC5705109 DOI: 10.1371/journal.pone.0186322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 09/28/2017] [Indexed: 01/15/2023] Open
Abstract
The aging-associated cellular and molecular changes in esophagus have not been established, yet. Thus we evaluated histological structure, interstitial cells of Cajal (ICCs), neuronal nitric oxide synthase (nNOS)-positive cells, and contractility in the esophagus of Fischer 344 rat at different ages (6-, 31-, 74-weeks, and 2-years). The lamina propria thickness and endomysial area were calculated. The immunoreactivity of c-Kit, nNOS and protein gene product (PGP) 9.5 was counted after immunohistochemistry. Expression of c-Kit, stem cell factor (SCF), nNOS and PGP 9.5 mRNA was measured by real-time PCR, and expression of c-Kit and nNOS protein was detected by Western blot. Isovolumetric contractile force measurement and electrical field stimulation (EFS) were conducted. The lamina propria thickness increased (6 week vs 2 year, P = 0.005) and the endomysial area of longitudinal muscle decreased with aging (6 week vs 2 year, P<0.001), while endomysial area of circular muscle did not significantly decrease. The proportions of NOS-immunoreactive cells and c-Kit-immunoreactive areas declined with aging (6 week vs 2 year; P<0.001 and P = 0.004, respectively), but there was no significant change of PGP 9.5-immunopositiviy. The expressions of nNOS, c-Kit and SCF mRNA also reduced with aging (6 week vs 2 year; P = 0.006, P = 0.001 and P = 0.006, respectively), while the change of PGP 9.5 mRNA expression was not significant. Western blot showed the significant decreases of nNOS and c-Kit protein expression with aging (6 week vs 2 year; P = 0.008 and P = 0.012, respectively). The EFS-induced esophageal contractions significantly decreased in 2-yr-old rat compared with 6-wk-old rats, however, L-NG-Nitroarginine methylester did not significantly increase the spontaneous and EFS-induced contractions in the 6-wk- and 2-yr-old rat esophagus. In conclusion, an increase of lamina propria thickness, a decrease of endomysial area, c-Kit, SCF and NOS expression with preserved total enteric neurons, and contractility in aged rat esophagus may explain the aging-associated esophageal dysmotility.
Collapse
Affiliation(s)
- Hee Jin Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
- Department of Internal Medicine, Myongji Hospital, Goyang, S. Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, S. Korea
- * E-mail:
| | - Yong Sung Kim
- Department of Gastroenterology and Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, S. Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Sun Min Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Ji Hyun Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Daeun Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Young-Jae Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Jongchan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, S. Korea
| | - Min-Seob Kim
- Department of Physiology and Institute of Wonkwang Medical Science, Wonkwang University College of Medicine, Iksan, S. Korea
| | - Moon Young Lee
- Department of Physiology and Institute of Wonkwang Medical Science, Wonkwang University College of Medicine, Iksan, S. Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, S. Korea
| |
Collapse
|
41
|
Kaneko T, Nemoto T, Funahashi K, Koike J, Shibuya K, Kaneko H. Differences in innervated neurons of the internal anal sphincter based on age and sex: A histological study. Geriatr Gerontol Int 2017; 18:495-500. [PMID: 29143476 DOI: 10.1111/ggi.13193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/02/2017] [Accepted: 08/27/2017] [Indexed: 01/19/2023]
Abstract
AIM Previous studies have shown sex and age differences in anal sphincter function, but few morphological studies have focused on the quality and quantity of the nerves that control the sphincter muscles. The present study aimed to determine whether there are morphological and quantitative sex and age differences in the nerves in the conjoined longitudinal muscle. METHODS This was a single-center, retrospective study using surgical specimens from 44 patients who underwent abdominoperineal resection between 2003 and 2012. Hematoxylin-eosin- and S-100-stained peripheral nerves (nerve fibers and ganglion cells) in the conjoined longitudinal muscle beneath the dentate line were observed microscopically. A qualitative examination assessed the degeneration score, which was based on the presence or absence of karyopyknosis, vacuolar degeneration, acidophilic degeneration of the cytoplasm, denucleation and adventitial neuronal changes. For quantitative examinations, each neuronal and muscular area was traced to calculate the neuronal area ratio in S-100-immunostained photomicrographs at the observation site. RESULTS Women had a significantly lower quantity of nerves than men. Older individuals (aged ≥80 years) had a significantly lower quantity of nerves than younger individuals. Furthermore, older individuals tended to show greater morphological changes that appeared to be a result of degeneration. CONCLUSIONS The present findings suggest that anal hypofunction in women and older individuals might result from differences in the quantity and quality of the neurons controlling the anal sphincter muscle. Geriatr Gerontol Int 2018; 18: 495-500.
Collapse
Affiliation(s)
- Tomoaki Kaneko
- Department of Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Tetsuo Nemoto
- Department of Surgical Pathology, Toho University School of Medicine, Tokyo, Japan
| | | | - Junichi Koike
- Department of Surgery, Toho University School of Medicine, Tokyo, Japan
| | - Kazutoshi Shibuya
- Department of Surgical Pathology, Toho University School of Medicine, Tokyo, Japan
| | - Hironori Kaneko
- Department of Surgery, Toho University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Lee SM, Kim N, Jo HJ, Park JH, Nam RH, Lee HS, Kim HJ, Lee MY, Kim YS, Lee DH. Comparison of Changes in the Interstitial Cells of Cajal and Neuronal Nitric Oxide Synthase-positive Neuronal Cells With Aging Between the Ascending and Descending Colon of F344 Rats. J Neurogastroenterol Motil 2017; 23:592-605. [PMID: 28774159 PMCID: PMC5628993 DOI: 10.5056/jnm17061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/20/2017] [Accepted: 07/09/2017] [Indexed: 12/22/2022] Open
Abstract
Background/Aims Neuronal degeneration and changes in interstitial cells of Cajal (ICCs) are important mechanisms of age-related constipation. This study aims to compare the distribution of ICCs and neuronal nitric oxide synthase (nNOS) with regard to age-related changes between the ascending colon (AC) and descending colon (DC) in 6-, 31-, and 74-week old and 2-year old male Fischer-344 rats. Methods The amount of fecal pellet and the bead expulsion times were measured. Fat proportion in the muscle layer of the colon was analyzed by hematoxylin and eosin staining. Proto-oncogene receptor tyrosine kinase (KIT) and neuronal nitric oxide synthase (nNOS) expression were analyzed with Western blotting and immunohistochemistry. Isovolumetric contractile measurements and electrical field stimulation were used to assess smooth muscle contractility. Results Colon transit and bead expulsion slowed with senescence. Fat in the muscle layer accumulated with age in the AC, but not in the DC. The proportion of KIT-immunoreactive ICCs in the submucosal and myenteric plexus was higher in the DC than in the AC, and it declined with age, especially in the AC. In contrast, the proportion of NOS-immunoreactive neurons in the myenteric plexus was higher in the AC than in the DC, and both decreased in older rats. Nitric oxide levels declined with age in the DC. Muscle strip experiments showed that the inhibitory response mediated by nitric oxide in the circular direction of the DC was reduced in 2-year old rats. Conclusion The AC and DC differ in their distribution of ICCs and nNOS, and age-related loss of nitrergic neurons more severely affects the DC than the AC.
Collapse
Affiliation(s)
- Sun Min Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jin Jo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Jinju, Gyeongsangnam-do, Korea
| | - Moon Young Lee
- Department of Physiology and Institute of Wonkwang Medical Science, Wonkwang University College of Medicine, Iksan, Jeollabuk-do, Korea
| | - Yong Sung Kim
- Division of Gastroenterology and Wonkwang Digestive Disease Research Institute, Department of Internal Medicine, Wonkwang University Sanbon Hospital, Gunpo, Gyeonggi-do, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| |
Collapse
|
43
|
Change in the Interstitial Cells of Cajal and nNOS Positive Neuronal Cells with Aging in the Stomach of F344 Rats. PLoS One 2017; 12:e0169113. [PMID: 28045993 PMCID: PMC5207530 DOI: 10.1371/journal.pone.0169113] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/11/2016] [Indexed: 12/22/2022] Open
Abstract
The gastric accommodation reflex is an important mechanism in gastric physiology. However, the aging-associated structural and functional changes in gastric relaxation have not yet been established. Thus, we evaluated the molecular changes of interstitial cell of Cajal (ICC) and neuronal nitric oxide synthase (nNOS) and the function changes in the corpus of F344 rats at different ages (6-, 31-, 74-wk and 2-yr). The proportion of the c-Kit-positive area in the submucosal border (SMB) and myenteric plexus (MP) layer was significantly lower in the older rats, as indicated by immunohistochemistry. The density of the nNOS-positive immunoreactive area also decreased with age in the SMB, circular muscle (CM), and MP. Similarly, the percent of nNOS-positive neuronal cells per total neuronal cells and the proportion of nNOS immunoreactive area of MP also decreased in aged rats. In addition, the mRNA and protein expression of c-Kit and nNOS significantly decreased with age. Expression of stem cell factor (SCF) and the pan-neuronal marker PGP 9.5 mRNA was significantly lower in the older rats than in the younger rats. Barostat studies showed no difference depending on age. Instead, the change of volume was significantly decreased by L-NG63-nitroarginine methyl ester in the 2-yr-old rats compared with the 6-wk-old rats (P = 0.003). Taken together, the quantitative and molecular nNOS changes in the stomach might play a role in the decrease of gastric accommodation with age.
Collapse
|
44
|
Abstract
The number of persons 60 years and older has increased 3-fold between 1950 and 2000. Aging alone does not greatly impact the gastrointestinal (GI) tract. Digestive dysfunction, including esophageal reflux, achalasia, dysphagia, dyspepsia, delayed gastric emptying, constipation, fecal incontinence, and fecal impaction, is a result of the highly prevalent comorbid conditions and the medications with which those conditions are treated. A multidisciplinary approach with the expertise of a geriatrician, gastroenterologist, neurologist, speech pathologist, and physical therapist ensures a comprehensive functional and neurological assessment of the older patient. Radiographic and endoscopic evaluation may be warranted in the evaluation of the symptomatic older patient with consideration given to the risks and benefits of the test being used. Treatment of the digestive dysfunction is aimed at improving health-related quality of life if cure cannot be achieved. Promotion of healthy aging, treatment of comorbid conditions, and avoidance of polypharmacy may prevent some of these digestive disorders. The age-related changes in GI motility, clinical presentation of GI dysmotility, and therapeutic principles in the symptomatic older patient are reviewed here.
Collapse
|
45
|
Yu Y, Daly DM, Adam IJ, Kitsanta P, Hill CJ, Wild J, Shorthouse A, Grundy D, Jiang W. Interplay between mast cells, enterochromaffin cells, and sensory signaling in the aging human bowel. Neurogastroenterol Motil 2016; 28:1465-79. [PMID: 27206689 PMCID: PMC5053273 DOI: 10.1111/nmo.12842] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Advanced age is associated with a reduction in clinical visceral pain perception. However, the underlying mechanisms remain largely unknown. Previous studies have suggested that an abnormal interplay between mast cells, enterochromaffin (EC) cells, and afferent nerves contribute to nociception in gastrointestinal disorders. The aim of this study was to investigate how aging affects afferent sensitivity and neuro-immune association in the human bowel. METHODS Mechanical and chemical sensitivity of human bowel afferents were examined by ex vivo afferent nerve recordings. Age-related changes in the density of mast cells, EC cells, sensory nerve terminals, and mast cell-nerve micro-anatomical association were investigated by histological and immune staining. KEY RESULTS Human afferents could be broadly classified into subpopulations displaying mechanical and chemical sensitivity, adaptation, chemo-sensitization, and recruitment. Interestingly human bowel afferent nerve sensitivity was attenuated with age. The density of substance P-immunoreactive (SP-IR) nerve varicosities was also reduced with age. In contrast, the density of ileal and colonic mucosal mast cells was increased with age, as was ileal EC cell number. An increased proportion of mast cells was found in close apposition to SP-IR nerves. CONCLUSIONS & INFERENCES Afferent sensitivity in human bowel was reduced with advancing age. Augmentation of mast cells and EC cell numbers and the mast cell-nerve association suggest a compensatory mechanism for sensory neurodegeneration.
Collapse
Affiliation(s)
- Y. Yu
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - D. M. Daly
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - I. J. Adam
- Department of Colorectal Surgical UnitNorthern General HospitalSheffield Teaching HospitalSheffieldUK
| | - P. Kitsanta
- Department of HistopathologyNorthern General HospitalSheffield Teaching HospitalSheffieldUK
| | - C. J. Hill
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - J. Wild
- Department of Colorectal Surgical UnitNorthern General HospitalSheffield Teaching HospitalSheffieldUK
| | - A. Shorthouse
- Department of Colorectal Surgical UnitNorthern General HospitalSheffield Teaching HospitalSheffieldUK
| | - D. Grundy
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - W. Jiang
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| |
Collapse
|
46
|
Rahman AA, Robinson AM, Brookes SJH, Eri R, Nurgali K. Rectal prolapse in Winnie mice with spontaneous chronic colitis: changes in intrinsic and extrinsic innervation of the rectum. Cell Tissue Res 2016; 366:285-299. [DOI: 10.1007/s00441-016-2465-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/29/2016] [Indexed: 12/19/2022]
|
47
|
Dalziel JE, Young W, Bercik P, Spencer NJ, Ryan LJ, Dunstan KE, Lloyd-West CM, Gopal PK, Haggarty NW, Roy NC. Tracking gastrointestinal transit of solids in aged rats as pharmacological models of chronic dysmotility. Neurogastroenterol Motil 2016; 28:1241-51. [PMID: 27028044 DOI: 10.1111/nmo.12824] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/24/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dysmotility in the gastrointestinal (GI) tract often leads to impaired transit of luminal contents leading to symptoms of diarrhea or constipation. The aim of this research was to develop a technique using high resolution X-ray imaging to study pharmacologically induced aged rat models of chronic GI dysmotility that mimic accelerated transit (diarrhea) or constipation. The 5-hydroxytryptamine type 4 (5-HT4 ) receptor agonist prucalopride was used to accelerate transit, and the opioid agonist loperamide was used to delay transit. METHODS Male rats (18 months) were given 0, 1, 2, or 4 mg/kg/day prucalopride or loperamide (in dimethyl sulfoxide, DMSO) for 7 days by continuous 7-day dosing. To determine the GI region-specific effect, transit of six metallic beads was tracked over 12 h using high resolution X-ray imaging. An established rating scale was used to classify GI bead location in vivo and the distance beads had propagated from the caecum was confirmed postmortem. KEY RESULTS Loperamide (1 mg/kg) slowed stomach emptying and GI transit at 9 and 12 h. Prucalopride (4 mg/kg) did not significantly alter GI transit scores, but at a dose of 4 mg/kg beads had moved significantly more distal than the caecum in 12 h compared to controls. CONCLUSIONS & INFERENCES We report a novel high-resolution, non-invasive, X-ray imaging technique that provides new insights into GI transit rates in live rats. The results demonstrate that loperamide slowed overall transit in aged rats, while prucalopride increased stomach emptying and accelerates colonic transit.
Collapse
Affiliation(s)
- J E Dalziel
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - W Young
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - P Bercik
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - N J Spencer
- School of Medicine, Flinders University, Adelaide, SA, Australia
| | - L J Ryan
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - K E Dunstan
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - C M Lloyd-West
- Bioinformatics Mathematics and Statistics, AgResearch, Palmerston North, New Zealand
| | - P K Gopal
- Fonterra Co-operative Group, Palmerston North, New Zealand
| | - N W Haggarty
- Fonterra Co-operative Group, Palmerston North, New Zealand
| | - N C Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch, Palmerston North, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,Gravida: National Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
48
|
Xuan Q, Zhang YX, Liu DG, Chan P, Xu SL, Cui YQ. Post-translational modifications of α-synuclein contribute to neurodegeneration in the colon of elderly individuals. Mol Med Rep 2016; 13:5077-83. [PMID: 27109489 PMCID: PMC4878562 DOI: 10.3892/mmr.2016.5166] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 02/23/2016] [Indexed: 01/24/2023] Open
Abstract
Synucleinopathies and abnormalities in the nerves of the enteric nervous system are hypothesized to be involved in age-associated motility disorders. The aim of the present study was to investigate the expression of various antigens, including α‑synuclein (Syn) and its post‑translational modified forms, in the human colon at various ages. In addition, the study aimed to correlate the expression of Syn with neurodegeneration. Immunohistochemistry was used to detect the expression of neurofilament (NF), Syn, as well as its nitrated (N) form in the healthy colonic tissue of 12 young (34.08±5.12 years), 10 middle‑aged (51.80±3.52 years), and 11 elderly (75.82±7.70 years) individuals. To the best of our knowledge, the current study is the first to demonstrate the presence of N‑Syn in the colonic tissue. N‑Syn was identified in the upper layer of the mucosa and submucosa layer. Furthermore, Syn (wild‑type) was present in the mucosa and submucosa. The number of NF‑positive neurons in the submucosal layer declined significantly with age (P<0.01). In addition, Syn and N‑Syn significantly increased during aging (P<0.01). Furthermore, a negative correlation was identified between neuron number and synucleinopathies, indicating the abnormal accumulation of both wild-type Syn and N‑Syn in the mucosa, submucosa, muscle layer and myenteric plexus. The present study demonstrates that the Syn pathology may be linked to colic neuronal degeneration during normal aging, and this link may cause functional deficits.
Collapse
Affiliation(s)
- Qi Xuan
- Department of Nutrition, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Yu-Xian Zhang
- Department of Internal Medicine, Beijing Moslem Hospital, Beijing 100054, P.R. China
| | - Dian-Gang Liu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Piu Chan
- Department of Neurobiology, Institute of Geriatrics of Beijing, Xuanwu Hospital, Capital Medical University, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing 100053, P.R. China
| | - Sheng-Li Xu
- Department of Neurobiology, Institute of Geriatrics of Beijing, Xuanwu Hospital, Capital Medical University, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing 100053, P.R. China
| | - Ye-Qing Cui
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
49
|
Burns AJ, Goldstein AM, Newgreen DF, Stamp L, Schäfer KH, Metzger M, Hotta R, Young HM, Andrews PW, Thapar N, Belkind-Gerson J, Bondurand N, Bornstein JC, Chan WY, Cheah K, Gershon MD, Heuckeroth RO, Hofstra RMW, Just L, Kapur RP, King SK, McCann CJ, Nagy N, Ngan E, Obermayr F, Pachnis V, Pasricha PJ, Sham MH, Tam P, Vanden Berghe P. White paper on guidelines concerning enteric nervous system stem cell therapy for enteric neuropathies. Dev Biol 2016; 417:229-51. [PMID: 27059883 DOI: 10.1016/j.ydbio.2016.04.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/29/2016] [Accepted: 04/02/2016] [Indexed: 12/22/2022]
Abstract
Over the last 20 years, there has been increasing focus on the development of novel stem cell based therapies for the treatment of disorders and diseases affecting the enteric nervous system (ENS) of the gastrointestinal tract (so-called enteric neuropathies). Here, the idea is that ENS progenitor/stem cells could be transplanted into the gut wall to replace the damaged or absent neurons and glia of the ENS. This White Paper sets out experts' views on the commonly used methods and approaches to identify, isolate, purify, expand and optimize ENS stem cells, transplant them into the bowel, and assess transplant success, including restoration of gut function. We also highlight obstacles that must be overcome in order to progress from successful preclinical studies in animal models to ENS stem cell therapies in the clinic.
Collapse
Affiliation(s)
- Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donald F Newgreen
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville 3052, Victoria, Australia
| | - Lincon Stamp
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Karl-Herbert Schäfer
- University of Applied Sciences, Kaiserlautern, Germany; Clinic of Pediatric Surgery, University Hospital Mannheim, University Heidelberg, Germany
| | - Marco Metzger
- Fraunhofer-Institute Interfacial Engineering and Biotechnology IGB Translational Centre - Würzburg branch and University Hospital Würzburg - Tissue Engineering and Regenerative Medicine (TERM), Würzburg, Germany
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter W Andrews
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Jaime Belkind-Gerson
- Division of Gastroenterology, Hepatology and Nutrition, Massachusetts General Hospital for Children, Harvard Medical School, Boston, USA
| | - Nadege Bondurand
- INSERM U955, 51 Avenue du Maréchal de Lattre de Tassigny, F-94000 Créteil, France; Université Paris-Est, UPEC, F-94000 Créteil, France
| | - Joel C Bornstein
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Wood Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Kathryn Cheah
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University, New York 10032, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Robert M W Hofstra
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lothar Just
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Germany
| | - Raj P Kapur
- Department of Pathology, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Sebastian K King
- Department of Paediatric and Neonatal Surgery, The Royal Children's Hospital, Melbourne, Australia
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Elly Ngan
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Florian Obermayr
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tübingen, D-72076 Tübingen, Germany
| | | | | | - Mai Har Sham
- Department of Biochemistry, The University of Hong Kong, Hong Kong
| | - Paul Tam
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), TARGID, University of Leuven, Belgium
| |
Collapse
|
50
|
Beyder A, Gibbons SJ, Mazzone A, Strege PR, Saravanaperumal SA, Sha L, Higgins S, Eisenman ST, Bernard CE, Geurts A, Kline CF, Mohler PJ, Farrugia G. Expression and function of the Scn5a-encoded voltage-gated sodium channel NaV 1.5 in the rat jejunum. Neurogastroenterol Motil 2016; 28:64-73. [PMID: 26459913 PMCID: PMC4688076 DOI: 10.1111/nmo.12697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/05/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND The SCN5A-encoded voltage-gated sodium channel NaV 1.5 is expressed in human jejunum and colon. Mutations in NaV 1.5 are associated with gastrointestinal motility disorders. The rat gastrointestinal tract expresses voltage-gated sodium channels, but their molecular identity and role in rat gastrointestinal electrophysiology are unknown. METHODS The presence and distribution of Scn5a-encoded NaV 1.5 was examined by PCR, Western blotting and immunohistochemistry in rat jejunum. Freshly dissociated smooth muscle cells were examined by whole cell electrophysiology. Zinc finger nuclease was used to target Scn5a in rats. Lentiviral-mediated transduction with shRNA was used to target Scn5a in rat jejunum smooth muscle organotypic cultures. Organotypic cultures were examined by sharp electrode electrophysiology and RT-PCR. KEY RESULTS We found NaV 1.5 in rat jejunum and colon smooth muscle by Western blot. Immunohistochemistry using two other antibodies of different portions of NaV 1.5 revealed the presence of the ion channel in rat jejunum. Whole cell voltage-clamp in dissociated smooth muscle cells from rat jejunum showed fast activating and inactivating voltage-dependent inward current that was eliminated by Na(+) replacement by NMDG(+) . Constitutive rat Scn5a knockout resulted in death in utero. NaV 1.5 shRNA delivered by lentivirus into rat jejunum smooth muscle organotypic culture resulted in 57% loss of Scn5a mRNA and several significant changes in slow waves, namely 40% decrease in peak amplitude, 30% decrease in half-width, and 7 mV hyperpolarization of the membrane potential at peak amplitude. CONCLUSIONS & INFERENCES Scn5a-encoded NaV 1.5 is expressed in rat gastrointestinal smooth muscle and it contributes to smooth muscle electrophysiology.
Collapse
Affiliation(s)
- A Beyder
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S J Gibbons
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A Mazzone
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - P R Strege
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S A Saravanaperumal
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - L Sha
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S Higgins
- College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - S T Eisenman
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - C E Bernard
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A Geurts
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - C F Kline
- The Dorothy M. Davis Heart and Lung Research Institute and Departments of Physiology & Cell Biology and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - P J Mohler
- The Dorothy M. Davis Heart and Lung Research Institute and Departments of Physiology & Cell Biology and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - G Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|