1
|
Tasnády KR, Jehoul R, de Ravé MG, Gijbels MJ, Brône B, Dewachter I, Melotte V, Boesmans W. Gastrointestinal Dysfunction and Low-Grade Inflammation Associate With Enteric Neuronal Amyloid-β in a Model for Amyloid Pathology. Neurogastroenterol Motil 2025; 37:e15016. [PMID: 40051115 PMCID: PMC11996054 DOI: 10.1111/nmo.15016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Patients suffering from Alzheimer's disease, a progressive neurodegenerative disorder involving cognitive decline and memory impairment, often present with gastrointestinal comorbidities. Accumulating data also indicate that alterations in the gut can modulate Alzheimer's disease pathology, highlighting the need to better understand the link between gastrointestinal abnormalities and neurodegeneration in the brain. METHODS To disentangle the pathophysiology of gastrointestinal dysfunction in Alzheimer's disease, we conducted a detailed pathological characterization of the gastrointestinal tract of 5xFAD mice by performing histological analyses, gene expression studies, immunofluorescence labeling and gut function assays. RESULTS We found that 5xFAD mice have elevated levels of intestinal amyloid precursor protein and accumulate amyloid-β in enteric neurons. Histopathology revealed that this is associated with mild intestinal inflammation and fibrosis and accompanied by increased expression of proinflammatory cytokines. While overall enteric nervous system composition and organization appeared unaffected, 5xFAD mice have faster gastrointestinal transit. CONCLUSION Our findings indicate that amyloid-β accumulation in enteric neurons is associated with low-grade intestinal inflammation and altered motility and suggest that peripheral pathology may cause gastrointestinal dysfunction in Alzheimer's disease patients.
Collapse
Affiliation(s)
- Kinga Réka Tasnády
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
- Department of Pathology, GROW‐Research Institute for Oncology and ReproductionMaastricht University Medical CentreMaastrichtthe Netherlands
| | - Reindert Jehoul
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
| | | | - Marion J. Gijbels
- Department of Pathology, NUTRIM Institute of Nutrition and Translational Research in MetabolismMaastricht University Medical CentreMaastrichtthe Netherlands
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular SciencesAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Bert Brône
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
| | - Ilse Dewachter
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
| | - Veerle Melotte
- Department of Pathology, GROW‐Research Institute for Oncology and ReproductionMaastricht University Medical CentreMaastrichtthe Netherlands
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamthe Netherlands
| | - Werend Boesmans
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
- Department of Pathology, GROW‐Research Institute for Oncology and ReproductionMaastricht University Medical CentreMaastrichtthe Netherlands
| |
Collapse
|
2
|
Sorensen L, Humenick A, Poon SSB, Han MN, Mahdavian NS, Rowe MC, Hamnett R, Gómez-de-Mariscal E, Neckel PH, Saito A, Mutunduwe K, Glennan C, Haase R, McQuade RM, Foong JPP, Brookes SJH, Kaltschmidt JA, Muñoz-Barrutia A, King SK, Veldhuis NA, Carbone SE, Poole DP, Rajasekhar P. Gut Analysis Toolbox - automating quantitative analysis of enteric neurons. J Cell Sci 2024; 137:jcs261950. [PMID: 39219476 PMCID: PMC11698042 DOI: 10.1242/jcs.261950] [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: 01/18/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
The enteric nervous system (ENS) consists of an extensive network of neurons and glial cells embedded within the wall of the gastrointestinal (GI) tract. Alterations in neuronal distribution and function are strongly associated with GI dysfunction. Current methods for assessing neuronal distribution suffer from undersampling, partly due to challenges associated with imaging and analyzing large tissue areas, and operator bias due to manual analysis. We present the Gut Analysis Toolbox (GAT), an image analysis tool designed for characterization of enteric neurons and their neurochemical coding using two-dimensional images of GI wholemount preparations. GAT is developed in Fiji, has a user-friendly interface, and offers rapid and accurate segmentation via custom deep learning (DL)-based cell segmentation models developed using StarDist, as well as a ganglia segmentation model in deepImageJ. We apply proximal neighbor-based spatial analysis to reveal differences in cellular distribution across gut regions using a public dataset. In summary, GAT provides an easy-to-use toolbox to streamline routine image analysis tasks in ENS research. GAT enhances throughput, allowing rapid unbiased analysis of larger tissue areas, multiple neuronal markers and numerous samples.
Collapse
Affiliation(s)
- Luke Sorensen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Adam Humenick
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Sabrina S. B. Poon
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Myat Noe Han
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Narges S. Mahdavian
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Matthew C. Rowe
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Ryan Hamnett
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Peter H. Neckel
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen 72076, Germany
| | - Ayame Saito
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Keith Mutunduwe
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Christie Glennan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Robert Haase
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI) Dresden/Leipzig, Universität Leipzig, Humboldtstraße 25, Leipzig 04105, Germany
| | - Rachel M. McQuade
- Gut Barrier and Disease Laboratory, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Medicine, Western Health, The University of Melbourne, Melbourne, VIC 3021, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne, Melbourne, VIC 3021, Australia
| | - Jaime P. P. Foong
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Simon J. H. Brookes
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arrate Muñoz-Barrutia
- Bioengineering Department, Universidad Carlos III de Madrid, ES 28911, Leganés, Spain
- Bioengineering Division, Instituto de Investigación Sanitaria Gregorio Marañon, ES 28007, Madrid, Spain
| | - Sebastian K. King
- Department of Paediatric Surgery, The Royal Children's Hospital, Parkville, VIC 3052, Australia
- Surgical Research, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicholas A. Veldhuis
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Simona E. Carbone
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Daniel P. Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Pradeep Rajasekhar
- Centre for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
3
|
Andriolo IRL, Longo B, de Melo DM, de Souza MM, Prediger RD, da Silva LM. Gastrointestinal Issues in Depression, Anxiety, and Neurodegenerative Diseases: A Systematic Review on Pathways and Clinical Targets Implications. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1371-1391. [PMID: 38500273 DOI: 10.2174/0118715273289138240306050532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Multiple illnesses commonly involve both the Central Nervous System (CNS) and the Gastrointestinal Tract (GI) simultaneously. Consistent evidence suggests that neurological disorders impair GI tract function and worsen the symptomatology and pathophysiology of digestive disorders. On the other hand, it has been proposed that early functional changes in the GI tract contribute to the genesis of several CNS illnesses. Additionally, the role played by the gut in these diseases can be seen as a paradigm for how the gut and the brain interact. METHODS We mentioned significant GI symptoms and discussed how the GI tract affects central nervous system illnesses, including depression, anxiety, Alzheimer's disease, and Parkinson's disease in this study. We also explored potential pathophysiological underpinnings and novel targets for the creation of future therapies targeted at gut-brain connections. RESULTS & DISCUSSION In this situation, modulating the gut microbiota through the administration of fecal microbiota transplants or probiotics may represent a new therapeutic option for this population, not only to treat GI problems but also behavioral problems, given the role that dysbiosis and leaky gut play in many neurological disorders. CONCLUSION Accurate diagnosis and treatment of co-existing illnesses also require coordination between psychiatrists, neurologists, gastroenterologists, and other specialties, as well as a thorough history and thorough physical examination.
Collapse
Affiliation(s)
| | - Bruna Longo
- Graduate Program in Pharmaceutical Sciences, University of Vale do Itajaí, Itajaí, Santa Catarina, Brazil
| | - Dayse Machado de Melo
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Márcia Maria de Souza
- Graduate Program in Pharmaceutical Sciences, University of Vale do Itajaí, Itajaí, Santa Catarina, Brazil
| | - Rui Daniel Prediger
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Luisa Mota da Silva
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
4
|
López-Pingarrón L, Almeida H, Soria-Aznar M, Reyes-Gonzales MC, Rodríguez-Moratinos AB, Muñoz-Hoyos A, García JJ. Interstitial Cells of Cajal and Enteric Nervous System in Gastrointestinal and Neurological Pathology, Relation to Oxidative Stress. Curr Issues Mol Biol 2023; 45:3552-3572. [PMID: 37185756 PMCID: PMC10136929 DOI: 10.3390/cimb45040232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The enteric nervous system (ENS) is organized into two plexuses-submucosal and myenteric-which regulate smooth muscle contraction, secretion, and blood flow along the gastrointestinal tract under the influence of the rest of the autonomic nervous system (ANS). Interstitial cells of Cajal (ICCs) are mainly located in the submucosa between the two muscle layers and at the intramuscular level. They communicate with neurons of the enteric nerve plexuses and smooth muscle fibers and generate slow waves that contribute to the control of gastrointestinal motility. They are also involved in enteric neurotransmission and exhibit mechanoreceptor activity. A close relationship appears to exist between oxidative stress and gastrointestinal diseases, in which ICCs can play a prominent role. Thus, gastrointestinal motility disorders in patients with neurological diseases may have a common ENS and central nervous system (CNS) nexus. In fact, the deleterious effects of free radicals could affect the fine interactions between ICCs and the ENS, as well as between the ENS and the CNS. In this review, we discuss possible disturbances in enteric neurotransmission and ICC function that may cause anomalous motility in the gut.
Collapse
Affiliation(s)
- Laura López-Pingarrón
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Henrique Almeida
- i3S-Instituto de Investigação e Inovação em Saúde, Porto University, 4200-135 Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
- Department of Obstetrics and Gynecology, Hospital-CUF Porto, 4100-180 Porto, Portugal
| | - Marisol Soria-Aznar
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Marcos C Reyes-Gonzales
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | | | - Antonio Muñoz-Hoyos
- Department of Pediatrics, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Joaquín J García
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
5
|
Makdissi S, Parsons BD, Di Cara F. Towards early detection of neurodegenerative diseases: A gut feeling. Front Cell Dev Biol 2023; 11:1087091. [PMID: 36824371 PMCID: PMC9941184 DOI: 10.3389/fcell.2023.1087091] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
The gastrointestinal tract communicates with the nervous system through a bidirectional network of signaling pathways called the gut-brain axis, which consists of multiple connections, including the enteric nervous system, the vagus nerve, the immune system, endocrine signals, the microbiota, and its metabolites. Alteration of communications in the gut-brain axis is emerging as an overlooked cause of neuroinflammation. Neuroinflammation is a common feature of the pathogenic mechanisms involved in various neurodegenerative diseases (NDs) that are incurable and debilitating conditions resulting in progressive degeneration and death of neurons, such as in Alzheimer and Parkinson diseases. NDs are a leading cause of global death and disability, and the incidences are expected to increase in the following decades if prevention strategies and successful treatment remain elusive. To date, the etiology of NDs is unclear due to the complexity of the mechanisms of diseases involving genetic and environmental factors, including diet and microbiota. Emerging evidence suggests that changes in diet, alteration of the microbiota, and deregulation of metabolism in the intestinal epithelium influence the inflammatory status of the neurons linked to disease insurgence and progression. This review will describe the leading players of the so-called diet-microbiota-gut-brain (DMGB) axis in the context of NDs. We will report recent findings from studies in model organisms such as rodents and fruit flies that support the role of diets, commensals, and intestinal epithelial functions as an overlooked primary regulator of brain health. We will finish discussing the pivotal role of metabolisms of cellular organelles such as mitochondria and peroxisomes in maintaining the DMGB axis and how alteration of the latter can be used as early disease makers and novel therapeutic targets.
Collapse
Affiliation(s)
- Stephanie Makdissi
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS, Canada
- IWK Health Centre, Department of Pediatrics, Halifax, Canada
| | - Brendon D. Parsons
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS, Canada
| | - Francesca Di Cara
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS, Canada
- IWK Health Centre, Department of Pediatrics, Halifax, Canada
| |
Collapse
|
6
|
Baidoo N, Sanger GJ, Belai A. Effect of old age on the subpopulations of enteric glial cells in human descending colon. Glia 2023; 71:305-316. [PMID: 36128665 PMCID: PMC10087700 DOI: 10.1002/glia.24272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/06/2022]
Abstract
Old age is associated with a higher incidence of lower bowel conditions such as constipation. Recent evidence suggest that colonic motility may be influenced by enteric glial cells (EGCs). Little is known about the effect of aging on the subpopulation of EGCs in the human colon. We assessed and compared the pattern of distribution of EGCs in adult and elderly human colon. Human descending colon were obtained from 23 cancer patients comprising of adults (23-63 years; 6 male, 7 female) and elderly (66-81 year; 6 male, 4 female). Specimens were serially-sectioned and immunolabeled with anti-Sox-10, anti-S100 and anti-GFAP for morphometric analysis. Standardized procedures were utilized to ensure unbiased counting and densitometric evaluation of EGCs. The number of Sox-10 immunoreactive (IR) EGCs were unaltered with age in both the myenteric plexus (MP) (respectively, in adult and elderly patients, 1939 ± 82 and 1760 ± 44/mm length; p > .05) and submucosal plexus; there were no apparent differences between adult males and females. The density of S100-IR EGCs declined among the elderly in the circular muscle and within the MP per ganglionic area. In the adult colon, there were more S100-IR EGCs distributed in the circular muscle per unit area than the Taenia coli. There was little or no GFAP-IR EGCs in both adult and elderly colon. We concluded that aging of the human descending colon does not result in a loss of Sox-10-IR EGCs in the MP and SMP but reduces S100-IR EGCs density within the musculature. This alteration in myenteric EGCs density with age may contribute to colonic dysfunction.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Gareth J Sanger
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Abi Belai
- School of Life and Health Sciences, University of Roehampton, London, UK
| |
Collapse
|
7
|
Al-Nakkash L, Mason D, Ismail N, Bowman T, Ahlert J, Rubin M, Smith E, Rosander A, Broderick TL. Exercise Training Prevents the Loss of Wall Thickness and Lowers Expression of Alzheimer's Related Proteins in 3xTg Mouse Jejunum. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14164. [PMID: 36361051 PMCID: PMC9653708 DOI: 10.3390/ijerph192114164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Growing evidence has demonstrated the benefits of regular exercise on cardiovascular, neural, and cognitive function in humans with Alzheimer's disease (AD). However, the consequences of AD on gastrointestinal morphology and the effects of regular exercise, which plays an important role against the development of certain gastrointestinal-related diseases, are still poorly understood. Therefore, to assess the changes in intestinal structure in a mouse model of AD and the impact of exercise, 2-month-old 3xTg-AD male mice were subjected to treadmill running 5 days per week for a period of 5 months. Jejunum from 3xTg-AD mice analyzed by histochemical methods revealed significant alterations in morphology. Compared to age-matched wild-type (WT) mice, villi length and crypt depth were increased, and collagen content of jejunum was elevated in 3xTg-AD mice. Jejunum wall dimensions, expressed as total wall thickness, outer longitudinal thickness, and inner circular thickness were decreased in 3xTg-AD compared to WT. Smooth muscle actin expression in jejunal wall was decreased in 3xTg-AD. Most of these aberrations were improved with exercise. Western blot expression of cyclin dependent kinase 5 (CDK5, involved in neural cell death and hyperphosphorylation of tau), was elevated in 3xTg-AD jejunum. This was associated with a 4-fold increase in tau5 expression. Exercise prevented the increase in expression of CDK5 and tau5. Expression of caspase 3 (an apoptotic marker) was elevated in 3xTg-AD jejunum and exercise prevented this. The results of our study indicate that the abnormalities in jejunum of the 3xTg mouse model of AD were prevented with exercise training.
Collapse
Affiliation(s)
- Layla Al-Nakkash
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Daniel Mason
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Niamatullah Ismail
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Taylor Bowman
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - John Ahlert
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Maxwell Rubin
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Emma Smith
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Abigail Rosander
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Tom L. Broderick
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| |
Collapse
|
8
|
Sorboni SG, Moghaddam HS, Jafarzadeh-Esfehani R, Soleimanpour S. A Comprehensive Review on the Role of the Gut Microbiome in Human Neurological Disorders. Clin Microbiol Rev 2022; 35:e0033820. [PMID: 34985325 PMCID: PMC8729913 DOI: 10.1128/cmr.00338-20] [Citation(s) in RCA: 234] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The human body is full of an extensive number of commensal microbes, consisting of bacteria, viruses, and fungi, collectively termed the human microbiome. The initial acquisition of microbiota occurs from both the external and maternal environments, and the vast majority of them colonize the gastrointestinal tract (GIT). These microbial communities play a central role in the maturation and development of the immune system, the central nervous system, and the GIT system and are also responsible for essential metabolic pathways. Various factors, including host genetic predisposition, environmental factors, lifestyle, diet, antibiotic or nonantibiotic drug use, etc., affect the composition of the gut microbiota. Recent publications have highlighted that an imbalance in the gut microflora, known as dysbiosis, is associated with the onset and progression of neurological disorders. Moreover, characterization of the microbiome-host cross talk pathways provides insight into novel therapeutic strategies. Novel preclinical and clinical research on interventions related to the gut microbiome for treating neurological conditions, including autism spectrum disorders, Parkinson's disease, schizophrenia, multiple sclerosis, Alzheimer's disease, epilepsy, and stroke, hold significant promise. This review aims to present a comprehensive overview of the potential involvement of the human gut microbiome in the pathogenesis of neurological disorders, with a particular emphasis on the potential of microbe-based therapies and/or diagnostic microbial biomarkers. This review also discusses the potential health benefits of the administration of probiotics, prebiotics, postbiotics, and synbiotics and fecal microbiota transplantation in neurological disorders.
Collapse
Affiliation(s)
| | | | - Reza Jafarzadeh-Esfehani
- Blood Borne Infectious Research Center, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Centre, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Interactions between the microbiota and enteric nervous system during gut-brain disorders. Neuropharmacology 2021; 197:108721. [PMID: 34274348 DOI: 10.1016/j.neuropharm.2021.108721] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 02/08/2023]
Abstract
For the last 20 years, researchers have focused their intention on the impact of gut microbiota in healthy and pathological conditions. This year (2021), more than 25,000 articles can be retrieved from PubMed with the keywords "gut microbiota and physiology", showing the constant progress and impact of gut microbes in scientific life. As a result, numerous therapeutic perspectives have been proposed to modulate the gut microbiota composition and/or bioactive factors released from microbes to restore our body functions. Currently, the gut is considered a primary site for the development of pathologies that modify brain functions such as neurodegenerative (Parkinson's, Alzheimer's, etc.) and metabolic (type 2 diabetes, obesity, etc.) disorders. Deciphering the mode of interaction between microbiota and the brain is a real original option to prevent (and maybe treat in the future) the establishment of gut-brain pathologies. The objective of this review is to describe recent scientific elements that explore the communication between gut microbiota and the brain by focusing our interest on the enteric nervous system (ENS) as an intermediate partner. The ENS, which is known as the "second brain", could be under the direct or indirect influence of the gut microbiota and its released factors (short-chain fatty acids, neurotransmitters, gaseous factors, etc.). Thus, in addition to their actions on tissue (adipose tissue, liver, brain, etc.), microbes can have an impact on local ENS activity. This potential modification of ENS function has global repercussions in the whole body via the gut-brain axis and represents a new therapeutic strategy.
Collapse
|
10
|
Abstract
The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.
Collapse
|
11
|
Chiocchetti R, Hitrec T, Giancola F, Sadeghinezhad J, Squarcio F, Galiazzo G, Piscitiello E, De Silva M, Cerri M, Amici R, Luppi M. Phosphorylated Tau protein in the myenteric plexus of the ileum and colon of normothermic rats and during synthetic torpor. Cell Tissue Res 2021; 384:287-299. [PMID: 33511469 PMCID: PMC8141491 DOI: 10.1007/s00441-020-03328-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
Tau protein is of primary importance for neuronal homeostasis and when hyperphosphorylated (PP-Tau), it tends to aggregate in neurofibrillary tangles, as is the case with tauopathies, a class of neurodegenerative disorders. Reversible PP-Tau accumulation occurs in the brain of hibernating rodents and it was recently observed in rats (a non-hibernator) during synthetic torpor (ST), a pharmacological-induced torpor-like condition. To date, the expression of PP-Tau in the rat enteric nervous system (ENS) is still unknown. The present study immunohistochemically investigates the PP-Tau expression in the myenteric plexus of the ileum and colon of normothermic rats (CTRL) and during ST, focusing on the two major subclasses of enteric neurons, i.e., cholinergic and nitrergic.Results showed that both groups of rats expressed PP-Tau, with a significantly increased percentage of PP-Tau immunoreactive (IR) neurons in ST vs. CTRL. In all rats, the majority of PP-Tau-IR neurons were cholinergic. In ST rats, the percentage of PP-Tau-IR neurons expressing a nitrergic phenotype increased, although with no significant differences between groups. In addition, the ileum of ST rats showed a significant decrease in the percentage of nitrergic neurons. In conclusion, our findings suggest an adaptive response of ENS to very low core body temperatures, with changes involving PP-tau expression in enteric neurons, especially the ileal nitrergic subpopulation. In addition, the high presence of PP-Tau in cholinergic neurons, specifically, is very interesting and deserves further investigation. Altogether, these data strengthen the hypothesis of a common cellular mechanism triggered by ST, natural hibernation and tauopathies occurring in ENS neurons.
Collapse
Affiliation(s)
- R Chiocchetti
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
| | - T Hitrec
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - F Giancola
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - J Sadeghinezhad
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - F Squarcio
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - G Galiazzo
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - E Piscitiello
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - M De Silva
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - M Cerri
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - R Amici
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - M Luppi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Miller AL, Bessho S, Grando K, Tükel Ç. Microbiome or Infections: Amyloid-Containing Biofilms as a Trigger for Complex Human Diseases. Front Immunol 2021; 12:638867. [PMID: 33717189 PMCID: PMC7952436 DOI: 10.3389/fimmu.2021.638867] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The human microbiota is the community of microorganisms that live upon or within their human host. The microbiota consists of various microorganisms including bacteria, fungi, viruses, and archaea; the gut microbiota is comprised mostly of bacteria. Many bacterial species within the gut microbiome grow as biofilms, which are multicellular communities embedded in an extracellular matrix. Studies have shown that the relative abundances of bacterial species, and therefore biofilms and bacterial byproducts, change during progression of a variety of human diseases including gastrointestinal, autoimmune, neurodegenerative, and cancer. Studies have shown the location and proximity of the biofilms within the gastrointestinal tract might impact disease outcome. Gram-negative enteric bacteria secrete the amyloid curli, which makes up as much as 85% of the extracellular matrix of enteric biofilms. Curli mediates cell-cell attachment and attachment to various surfaces including extracellular matrix components such as fibronectin and laminin. Structurally, curli is strikingly similar to pathological and immunomodulatory human amyloids such as amyloid-β, which has been implicated in Alzheimer's disease, α-synuclein, which is involved in Parkinson's disease, and serum amyloid A, which is secreted during the acute phase of inflammation. The immune system recognizes both bacterial amyloid curli and human amyloids utilizing the same receptors, so curli also induces inflammation. Moreover, recent work indicates that curli can participate in the self-assembly process of pathological human amyloids. Curli is found within biofilms of commensal enteric bacteria as well as invasive pathogens; therefore, evidence suggests that curli contributes to complex human diseases. In this review, we summarize the recent findings on how bacterial biofilms containing curli participate in the pathological and immunological processes in gastrointestinal diseases, systemic autoimmune diseases, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Amanda L Miller
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Shingo Bessho
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Kaitlyn Grando
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Çagla Tükel
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Xu W, Wong G, Hwang YY, Larbi A. The untwining of immunosenescence and aging. Semin Immunopathol 2020; 42:559-572. [PMID: 33165716 PMCID: PMC7665974 DOI: 10.1007/s00281-020-00824-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023]
Abstract
From a holistic point of view, aging results from the cumulative erosion of the various systems. Among these, the immune system is interconnected to the rest as immune cells are present in all organs and recirculate through bloodstream. Immunosenescence is the term used to define the remodelling of immune changes during aging. Because immune cells-and particularly lymphocytes-can further differentiate after their maturation in response to pathogen recognition, it is therefore unclear when senescence is induced in these cells. Additionally, it is also unclear which signals triggers senescence in immune cells (i) aging per se, (ii) specific response to pathogens, (iii) underlying conditions, or (iv) inflammaging. In this review, we will cover the current knowledge and concepts linked to immunosenescence and we focus this review on lymphocytes and T cells, which represent the typical model for replicative senescence. With the evidence presented, we propose to disentangle the senescence of immune cells from chronological aging.
Collapse
Affiliation(s)
- Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore
| | - Glenn Wong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos, Singapore, Singapore.
- Department of Geriatrics, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
14
|
Ismail MM, Alotaibi BS, EL-Sheekh MM. Therapeutic Uses of Red Macroalgae. Molecules 2020; 25:4411. [PMID: 32992919 PMCID: PMC7583832 DOI: 10.3390/molecules25194411] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Red Seaweed "Rhodophyta" are an important group of macroalgae that include approximately 7000 species. They are a rich source of structurally diverse bioactive constituents, including protein, sulfated polysaccharides, pigments, polyunsaturated fatty acids, vitamins, minerals, and phenolic compounds with nutritional, medical, and industrial importance. Polysaccharides are the main components in the cell wall of red algae and represent about 40-50% of the dry weight, which are extensively utilized in industry and pharmaceutical compounds, due to their thickening and gelling properties. The hydrocolloids galactans carrageenans and agars are the main red seaweed cell wall polysaccharides, which had broad-spectrum therapeutic characters. Generally, the chemical contents of seaweed are different according to the algal species, growth stage, environment, and external conditions, e.g., the temperature of the water, light intensity, nutrient concentrations in the ecosystem. Economically, they can be recommended as a substitute source for natural ingredients that contribute to a broad range of bioactivities like cancer therapy, anti-inflammatory agents, and acetylcholinesterase inhibitory. This review touches on the main points of the pharmaceutical applications of red seaweed, as well as the exploitation of their specific compounds and secondary metabolites with vital roles.
Collapse
Affiliation(s)
- Mona M. Ismail
- National Institute of Oceanography and Fisheries, NIOF, Alexandria 21556, Egypt;
| | - Badriyah S. Alotaibi
- Pharmaceutical Sciences Department, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | | |
Collapse
|
15
|
Endres K. Retinoic Acid and the Gut Microbiota in Alzheimer's Disease: Fighting Back-to-Back? Curr Alzheimer Res 2020; 16:405-417. [PMID: 30907321 DOI: 10.2174/1567205016666190321163705] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND There is growing evidence that the gut microbiota may play an important role in neurodegenerative diseases such as Alzheimer's disease. However, how these commensals influence disease risk and progression still has to be deciphered. OBJECTIVE The objective of this review was to summarize current knowledge on the interplay between gut microbiota and retinoic acid. The latter one represents one of the important micronutrients, which have been correlated to Alzheimer's disease and are used in initial therapeutic intervention studies. METHODS A selective overview of the literature is given with the focus on the function of retinoic acid in the healthy and diseased brain, its metabolism in the gut, and the potential influence that the bioactive ligand may have on microbiota, gut physiology and, Alzheimer's disease. RESULTS Retinoic acid can influence neuronal functionality by means of plasticity but also by neurogenesis and modulating proteostasis. Impaired retinoid-signaling, therefore, might contribute to the development of diseases in the brain. Despite its rather direct impact, retinoic acid also influences other organ systems such as gut by regulating the residing immune cells but also factors such as permeability or commensal microbiota. These in turn can also interfere with retinoid-metabolism and via the gutbrain- axis furthermore with Alzheimer's disease pathology within the brain. CONCLUSION Potentially, it is yet too early to conclude from the few reports on changed microbiota in Alzheimer's disease to a dysfunctional role in retinoid-signaling. However, there are several routes how microbial commensals might affect and might be affected by vitamin A and its derivatives.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
16
|
Manocha GD, Floden AM, Miller NM, Smith AJ, Nagamoto-Combs K, Saito T, Saido TC, Combs CK. Temporal progression of Alzheimer's disease in brains and intestines of transgenic mice. Neurobiol Aging 2019; 81:166-176. [PMID: 31284126 DOI: 10.1016/j.neurobiolaging.2019.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/21/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022]
Abstract
The amyloid beta (Aβ) peptide is associated with the neurodegenerative and inflammatory changes in brains affected by Alzheimer's disease (AD). We hypothesized that the enteric nervous system also produces Aβ in an intestinal component of disease. To test this idea, we compared C57BL/6 wild-type (WT) male and female mice to two models of Alzheimer's disease, amyloid precursor protein (APP)/presenilin 1 (PS1) mice and amyloid precursor protein NL-G-F (AppNL-G-F) mice, at 3, 6, and 12 months of age. Brain Aβ plaque deposition in AppNL-G-F mice preceded that in the APP/PS1 mice, observable by 3 months. Three-month-old female AppNL-G-F mice had decreased intestinal motility compared with WT and APP/PS1 mice. However, 3-month-old female APP/PS1 mice demonstrated increased intestinal permeability compared with WT and AppNL-G-F mice. Both sexes of APP/PS1 and AppNL-G-F mice demonstrated increased colon lipocalin 2 mRNA and insoluble Aβ 1-42 levels at 3 months. These data demonstrate an unrecognized enteric aspect of disease in 2 different mouse models correlating with the earliest brain changes.
Collapse
Affiliation(s)
- Gunjan D Manocha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Angela M Floden
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicole M Miller
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Abbie J Smith
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Kumi Nagamoto-Combs
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA.
| |
Collapse
|
17
|
Canadas RF, Ren T, Tocchio A, Marques AP, Oliveira JM, Reis RL, Demirci U. Tunable anisotropic networks for 3-D oriented neural tissue models. Biomaterials 2018; 181:402-414. [DOI: 10.1016/j.biomaterials.2018.07.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/24/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
|
18
|
Chalazonitis A, Rao M. Enteric nervous system manifestations of neurodegenerative disease. Brain Res 2018; 1693:207-213. [PMID: 29360466 PMCID: PMC6003851 DOI: 10.1016/j.brainres.2018.01.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/26/2022]
Abstract
Neurological disorders cause gastrointestinal (GI) symptoms that are debilitating and markedly diminish quality of life in patients. The enteric nervous system (ENS), the intrinsic nervous system of the GI tract that is often referred to as "the second brain", shares many features with the central nervous system. The ENS plays an essential role in regulating many GI functions including motility and fluid secretion. Enteric neuronal degeneration could therefore be responsible for the GI symptoms commonly observed in neurological conditions. Here we describe the organization and functions of the ENS and then review the evidence for ENS involvement in two common neurodegenerative disorders, Parkinson's disease (PD) and Alzheimer's disease (AD). Data from patients as well as animal models suggest that PD affects distinct subsets of neurons and glia in the ENS, and that the ENS may participate in the pathogenesis of this disorder. While there has been great enthusiasm for the possibility of sampling the ENS for diagnosis or therapeutic monitoring of PD, further work is needed to determine which enteric neurons are most affected and how ENS function could be modulated to ameliorate GI symptoms in patients. Although AD is far more common than PD and AD patients also experience GI symptoms, understanding of ENS dysfunction in AD is in its infancy. Much work remains to be done in both of these fields to determine how the ENS contributes to and/or is altered by these disorders, and how to target the ENS for more effective treatment of GI comorbidities.
Collapse
Affiliation(s)
- Alcmène Chalazonitis
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, P&S 11-511, New York, NY, USA.
| | - Meenakshi Rao
- Department of Pediatrics, Columbia University, 622 West 168th Street, PH 17, New York, NY, USA.
| |
Collapse
|
19
|
Nagpal R, Mainali R, Ahmadi S, Wang S, Singh R, Kavanagh K, Kitzman DW, Kushugulova A, Marotta F, Yadav H. Gut microbiome and aging: Physiological and mechanistic insights. NUTRITION AND HEALTHY AGING 2018; 4:267-285. [PMID: 29951588 PMCID: PMC6004897 DOI: 10.3233/nha-170030] [Citation(s) in RCA: 418] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The development of human gut microbiota begins as soon as the neonate leaves the protective environment of the uterus (or maybe in-utero) and is exposed to innumerable microorganisms from the mother as well as the surrounding environment. Concurrently, the host responses to these microbes during early life manifest during the development of an otherwise hitherto immature immune system. The human gut microbiome, which comprises an extremely diverse and complex community of microorganisms inhabiting the intestinal tract, keeps on fluctuating during different stages of life. While these deviations are largely natural, inevitable and benign, recent studies show that unsolicited perturbations in gut microbiota configuration could have strong impact on several features of host health and disease. Our microbiota undergoes the most prominent deviations during infancy and old age and, interestingly, our immune health is also in its weakest and most unstable state during these two critical stages of life, indicating that our microbiota and health develop and age hand-in-hand. However, the mechanisms underlying these interactions are only now beginning to be revealed. The present review summarizes the evidences related to the age-associated changes in intestinal microbiota and vice-versa, mechanisms involved in this bi-directional relationship, and the prospective for development of microbiota-based interventions such as probiotics for healthy aging.
Collapse
Affiliation(s)
- Ravinder Nagpal
- Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Rabina Mainali
- Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shokouh Ahmadi
- Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Shaohua Wang
- Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ria Singh
- Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kylie Kavanagh
- Department of Pathology (Comparative Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dalane W. Kitzman
- Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Almagul Kushugulova
- Center for Life Sciences, NLA, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Francesco Marotta
- ReGenera Research and Development for Aging Interventions, and San Babila Clinic, Corso Matteotti 1/A, Milano, Italy
| | - Hariom Yadav
- Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, NC, USA
- Corresponding author: Hariom Yadav, PhD., Center for Diabetes, Obesity and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Department of Microbiology and Immunology, Biotech Place, Suite 2E-034, 575 Patterson Ave., Winston-Salem, NC 27101, USA. Tel.: +1 336 713 5049; Fax: +1 336 716 9928; E-mail:
| |
Collapse
|
20
|
Feng J, Dong L, Zhang J, Han X, Tang S, Song L, Cong L, Wang X, Wang Y, Du Y. Unique expression pattern of KIBRA in the enteric nervous system of APP/PS1 mice. Neurosci Lett 2018. [DOI: 10.1016/j.neulet.2018.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
21
|
Tzeng NS, Chung CH, Lin FH, Huang CF, Yeh CB, Huang SY, Lu RB, Chang HA, Kao YC, Yeh HW, Chiang WS, Chou YC, Tsao CH, Wu YF, Chien WC. Magnesium oxide use and reduced risk of dementia: a retrospective, nationwide cohort study in Taiwan. Curr Med Res Opin 2018; 34:163-169. [PMID: 28952385 DOI: 10.1080/03007995.2017.1385449] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Dietary magnesium may be associated with a lower risk of dementia; however, the impact of magnesium oxide (MgO), a common laxative, on dementia has yet to be elucidated. This study aimed to investigate the association between the usage of MgO and the risk of developing dementia. METHODS We used a dataset from the National Health Research Institute Database (NHRID) of Taiwan containing one million randomly sampled subjects to identify patients aged ≥50 years with no history of MgO usage. A total of 1547 patients who had used MgO were enrolled, along with 4641 controls who had not used the MgO propensity score matched by age, gender and comorbidity, at a ratio of 1:3. After adjusting for confounding risk factors, a Cox proportional hazards model was used to compare the risk of developing dementia during a 10 year follow-up period. RESULTS Of the enrolled patients, 44 (2.84%) developed dementia, when compared to 199 (4.28%) in the control group. The Cox proportional hazards regression analysis revealed that the patients who had used MgO were less likely to develop dementia with a crude hazard ratio of 0.617 (95% CI, 0.445-0.856, p = .004). After adjusting for age, gender, comorbidity, geographical area and urbanization level of residence, and monthly income, the adjusted hazard ratio was 0.517 (95% CI, 0.412-0.793, p = .001). CONCLUSIONS The patients who used MgO had a decreased risk of developing dementia. Further studies on the effects of MgO in reducing the risk of dementia are therefore warranted.
Collapse
Affiliation(s)
- Nian-Sheng Tzeng
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- b Student Counseling Center , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Chi-Hsiang Chung
- c Taiwanese Injury Prevention and Safety Promotion Association , Taipei , Taiwan , ROC
- d School of Public Health , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Fu-Huang Lin
- d School of Public Health , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Ching-Feng Huang
- e Division of Gastroenterology, Children's Medical Center, Taipei Veterans General Hospital
- f School of Medicine, National Yang-Ming University
- g Department of Pediatrics , Tri-Service General Hospital, National Defense Medical Center , Taipei , Taiwan , ROC
| | - Chin-Bin Yeh
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- h Graduate Institute of Medical Sciences , National Defense Medical Center , Taipei , Taiwan , ROC
| | - San-Yuan Huang
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- h Graduate Institute of Medical Sciences , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Ru-Band Lu
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- h Graduate Institute of Medical Sciences , National Defense Medical Center , Taipei , Taiwan , ROC
- i Division of Clinical Psychology , Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University , Tainan , Taiwan , ROC
- j Department of Psychiatry, College of Medicine , National Cheng Kung University , Tainan , Taiwan , ROC
- k Institute of Behavioral Medicine , College of Medicine, National Cheng Kung University , Tainan , Taiwan , ROC
- l Department of Psychiatry , National Cheng Kung University Hospital , Tainan , Taiwan , ROC
- m Center for Neuropsychiatric Research , National Health Research Institute , Zhunan, Miaoli County , Taiwan , ROC
| | - Hsin-An Chang
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- b Student Counseling Center , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Yu-Chen Kao
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- n Department of Psychiatry , Tri-Service General Hospital, Song-Shan Branch, National Defense Medical Center , Taipei , Taiwan , ROC
| | - Hui-Wen Yeh
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- o Institute of Bioinformatics and Systems Biology , National Chiao Tung University , Hsin-Chu , Taiwan , ROC
- p Department of Nursing , Tri-Service General Hospital, and School of Nursing, National Defense Medical Center , Taipei , Taiwan , ROC
| | - Wei-Shan Chiang
- a Department of Psychiatry , Tri-Service General Hospital, School of Medicine, National Defense Medical Center , Taipei , Taiwan , ROC
- q Department and Institute of Mathematics , Tamkang University , New Taipei City , Taiwan , ROC
| | - Yu-Ching Chou
- d School of Public Health , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Chang-Huei Tsao
- r Department of Medical Research , Tri-Service General Hospital, National Defense Medical Center , Taipei , Taiwan , ROC
- s Department of Microbiology & Immunology , National Defense Medical Center , Taipei , Taiwan , ROC
| | - Yung-Fu Wu
- r Department of Medical Research , Tri-Service General Hospital, National Defense Medical Center , Taipei , Taiwan , ROC
| | - Wu-Chien Chien
- d School of Public Health , National Defense Medical Center , Taipei , Taiwan , ROC
- r Department of Medical Research , Tri-Service General Hospital, National Defense Medical Center , Taipei , Taiwan , ROC
| |
Collapse
|
22
|
Melo CGDS, Perles JVCM, Zanoni JN, Souza SRGD, Santos EX, Leite ADL, Heubel AD, E Souza CO, Souza JGD, Buzalaf MAR. Enteric innervation combined with proteomics for the evaluation of the effects of chronic fluoride exposure on the duodenum of rats. Sci Rep 2017; 7:1070. [PMID: 28432311 PMCID: PMC5430799 DOI: 10.1038/s41598-017-01090-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/24/2017] [Indexed: 02/07/2023] Open
Abstract
Ingested fluoride (F) is absorbed mainly in the small intestine, which is controlled by the Enteric Nervous System (ENS). Although important intestinal symptomatology has been described after excessive F exposure, there have been no studies reporting the effects of F on the ENS. In this study, the effects of chronic F exposure were evaluated on the duodenums of rats through proteomic and morphological analyses. Concentrations of 0, 10, or 50 ppm of F were applied to the drinking water for 30 days. Immunofluorescence techniques were performed in the myenteric plexus of the duodenum to detect HuC/D, neuronal nitric oxide (nNOS), vasoactive intestinal peptide (VIP), calcitonin gene related peptide (CGRP), and substance P (SP). The 50 ppm F group presented a significant decrease in the density of nNOS-IR neurons. Significant morphological alterations were also observed in HUC/D-IR and nNOS-IR neurons; VIP-IR, CGRP-IR, and SP-IR varicosities for both groups (10 and 50 ppm F). Proteomic analysis of the duodenum demonstrated alterations in the expression of several proteins, especially those related to important biological processes, such as protein polymerization, which helps to explain the downregulation of many proteins upon exposure to 50 ppm of F.
Collapse
Affiliation(s)
| | | | | | | | - Erika Xavier Santos
- Department of Morphophysiological Sciences, State University of Maringá, Paraná, Brazil
| | - Aline de Lima Leite
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | - Camila Oliveira E Souza
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Juliana Gadelha de Souza
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | |
Collapse
|
23
|
Gui H, Schriemer D, Cheng WW, Chauhan RK, Antiňolo G, Berrios C, Bleda M, Brooks AS, Brouwer RWW, Burns AJ, Cherny SS, Dopazo J, Eggen BJL, Griseri P, Jalloh B, Le TL, Lui VCH, Luzón-Toro B, Matera I, Ngan ESW, Pelet A, Ruiz-Ferrer M, Sham PC, Shepherd IT, So MT, Sribudiani Y, Tang CSM, van den Hout MCGN, van der Linde HC, van Ham TJ, van IJcken WFJ, Verheij JBGM, Amiel J, Borrego S, Ceccherini I, Chakravarti A, Lyonnet S, Tam PKH, Garcia-Barceló MM, Hofstra RMW. Whole exome sequencing coupled with unbiased functional analysis reveals new Hirschsprung disease genes. Genome Biol 2017; 18:48. [PMID: 28274275 PMCID: PMC5343413 DOI: 10.1186/s13059-017-1174-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/17/2017] [Indexed: 12/17/2022] Open
Abstract
Background Hirschsprung disease (HSCR), which is congenital obstruction of the bowel, results from a failure of enteric nervous system (ENS) progenitors to migrate, proliferate, differentiate, or survive within the distal intestine. Previous studies that have searched for genes underlying HSCR have focused on ENS-related pathways and genes not fitting the current knowledge have thus often been ignored. We identify and validate novel HSCR genes using whole exome sequencing (WES), burden tests, in silico prediction, unbiased in vivo analyses of the mutated genes in zebrafish, and expression analyses in zebrafish, mouse, and human. Results We performed de novo mutation (DNM) screening on 24 HSCR trios. We identify 28 DNMs in 21 different genes. Eight of the DNMs we identified occur in RET, the main HSCR gene, and the remaining 20 DNMs reside in genes not reported in the ENS. Knockdown of all 12 genes with missense or loss-of-function DNMs showed that the orthologs of four genes (DENND3, NCLN, NUP98, and TBATA) are indispensable for ENS development in zebrafish, and these results were confirmed by CRISPR knockout. These genes are also expressed in human and mouse gut and/or ENS progenitors. Importantly, the encoded proteins are linked to neuronal processes shared by the central nervous system and the ENS. Conclusions Our data open new fields of investigation into HSCR pathology and provide novel insights into the development of the ENS. Moreover, the study demonstrates that functional analyses of genes carrying DNMs are warranted to delineate the full genetic architecture of rare complex diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1174-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongsheng Gui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.,Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Duco Schriemer
- Department of Neuroscience, section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - William W Cheng
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.,Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Rajendra K Chauhan
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Guillermo Antiňolo
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Courtney Berrios
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Marta Bleda
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain.,Department of Medicine, School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Alice S Brooks
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Rutger W W Brouwer
- Erasmus Center for Biomics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alan J Burns
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands.,Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Institute of Child Health, London, UK
| | - Stacey S Cherny
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Joaquin Dopazo
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Bart J L Eggen
- Department of Neuroscience, section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Binta Jalloh
- Department of Biology, Emory University, Atlanta, USA
| | - Thuy-Linh Le
- Laboratory of embryology and genetics of human malformations, INSERM UMR 1163, Institut Imagine, Paris, France.,Department of Genetics, Paris Descartes-Sorbonne Paris Cité University, Hôpital Necker-Enfants Malades (APHP), Paris, France
| | - Vincent C H Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Berta Luzón-Toro
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Ivana Matera
- UOC Genetica Medica, Istituto Gaslini, Genoa, Italy
| | - Elly S W Ngan
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Anna Pelet
- Laboratory of embryology and genetics of human malformations, INSERM UMR 1163, Institut Imagine, Paris, France.,Department of Genetics, Paris Descartes-Sorbonne Paris Cité University, Hôpital Necker-Enfants Malades (APHP), Paris, France
| | - Macarena Ruiz-Ferrer
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Pak C Sham
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | | | - Man-Ting So
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Yunia Sribudiani
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Clara S M Tang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | | | - Herma C van der Linde
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | | | - Joke B G M Verheij
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jeanne Amiel
- Laboratory of embryology and genetics of human malformations, INSERM UMR 1163, Institut Imagine, Paris, France.,Department of Genetics, Paris Descartes-Sorbonne Paris Cité University, Hôpital Necker-Enfants Malades (APHP), Paris, France
| | - Salud Borrego
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío/CSIC/University of Seville, Seville, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | | | - Aravinda Chakravarti
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Stanislas Lyonnet
- Laboratory of embryology and genetics of human malformations, INSERM UMR 1163, Institut Imagine, Paris, France.,Department of Genetics, Paris Descartes-Sorbonne Paris Cité University, Hôpital Necker-Enfants Malades (APHP), Paris, France
| | - Paul K H Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Maria-Mercè Garcia-Barceló
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands. .,Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Institute of Child Health, London, UK.
| |
Collapse
|
24
|
Han X, Tang S, Dong L, Song L, Dong Y, Wang Y, Du Y. Loss of nitrergic and cholinergic neurons in the enteric nervous system of APP/PS1 transgenic mouse model. Neurosci Lett 2017; 642:59-65. [DOI: 10.1016/j.neulet.2017.01.061] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 02/06/2023]
|
25
|
Wu J, Fu B, Lei H, Tang H, Wang Y. Gender differences of peripheral plasma and liver metabolic profiling in APP/PS1 transgenic AD mice. Neuroscience 2016; 332:160-9. [PMID: 27393253 DOI: 10.1016/j.neuroscience.2016.06.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/25/2016] [Accepted: 06/28/2016] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive impairment. Currently, there is less knowledge of the involvement of the peripheral biofluid/organ in AD, compared with the central nervous system. In addition, with reported high morbidity in women in particular, it has become very important to explore whether gender difference in the peripheral metabolome is associated with AD. Here, we investigated metabolic responses of both plasma and liver tissues using an APP/PS1 double mutant transgenic mouse model with NMR spectroscopy, as well as analysis from serum biochemistry and histological staining. Fatty acid composition from plasma and liver extracts was analyzed using GC-FID/MS. We found clear gender differences in AD transgenic mice when compared with their wild-type counterparts. Female AD mice displayed more intensive responses, which were highlighted by higher levels of lipids, 3-hydroxybutyrate and nucleotide-related metabolites, together with lower levels of glucose. These observations indicate that AD induces oxidative stress and impairs cellular energy metabolism in peripheral organs. Disturbances in AD male mice were milder with depletion of monounsaturated fatty acids. We also observed a higher activity of delta-6-desaturate and suppressed activity of delta-5-desaturate in female mice, whereas inhibited stearoyl-CoA-desaturase in male mice suggested that AD induced by the double mutant genes results in different fatty acids catabolism depending on gender. Our results provide metabolic clues into the peripheral biofluid/organs involved in AD, and we propose that a gender-specific scheme for AD treatment in men and women may be required.
Collapse
Affiliation(s)
- Junfang Wu
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bin Fu
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese & Western Medicine, Wuhan 430015, China
| | - Hehua Lei
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Huiru Tang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Ministry of Education Key Laboratory of Contemporary Anthropology, Metabonomics and Systems Biology Laboratory, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Yulan Wang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
26
|
Morphometric and biomechanical remodeling of the small intestine during aging in rats. J Biomech 2015; 48:4271-8. [DOI: 10.1016/j.jbiomech.2015.10.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 01/27/2023]
|
27
|
Puig KL, Lutz BM, Urquhart SA, Rebel AA, Zhou X, Manocha GD, Sens M, Tuteja AK, Foster NL, Combs CK. Overexpression of mutant amyloid-β protein precursor and presenilin 1 modulates enteric nervous system. J Alzheimers Dis 2015; 44:1263-78. [PMID: 25408221 PMCID: PMC6295343 DOI: 10.3233/jad-142259] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder histologically characterized by amyloid-β (Aβ) protein accumulation and activation of associated microglia. Although these features are well described in the central nervous system, the process and consequences of Aβ accumulation in the enteric nervous system have not been extensively studied. We hypothesized that Aβ also may accumulate in the enteric nervous system and lead to immune cell activation and neuronal dysfunction in the digestive tract not unlike that observed in diseased brain. To test this hypothesis, ileums of the small intestine of thirteen month old AβPP/PS1 and C57BL/6 (wild type) mice were collected and analyzed using immunohistochemistry, western blot analysis, cytokine arrays, and ELISA. AβPP/PS1 mice demonstrated no differences in intestinal motility or water absorption but elevated luminal IgA levels compared to wild type mice. They also had increased protein levels of AβPP and the proteolytic enzyme, BACE, corresponding to an increase in Aβ1-40 in the intestinal lysate as well as an increase in both Aβ1-40 and Aβ1-42 in the stool. This correlated with increased protein markers of proinflammatory and immune cell activation. Histologic analysis localized AβPP within enteric neurons but also intestinal epithelial cells with elevated Aβ immunoreactivity in the AβPP/PS1 mice. The presence of AβPP, Aβ, and CD68 immunoreactivity in the intestines of some patients with neuropathologically-confirmed AD are consistent with the findings in this mouse model. These data support the hypothesis that in AD the intestine, much like the brain, may develop proinflammatory and immune changes related to AβPP and Aβ.
Collapse
Affiliation(s)
- Kendra L. Puig
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Brianna M. Lutz
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Siri A. Urquhart
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Andrew A. Rebel
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xudong Zhou
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Gunjan D. Manocha
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - MaryAnn Sens
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Ashok K. Tuteja
- Division of Gastroenterology, University of Utah, Salt Lake City, UT, USA
| | - Norman L. Foster
- Center for Alzheimer’s Care, Imaging and Research, Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Colin K. Combs
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
28
|
Saffrey MJ. Aging of the mammalian gastrointestinal tract: a complex organ system. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9603. [PMID: 24352567 PMCID: PMC4082571 DOI: 10.1007/s11357-013-9603-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 11/25/2013] [Indexed: 05/23/2023]
Abstract
Gastrointestinal disorders are a major cause of morbidity in the elderly population. The gastrointestinal tract is the most complex organ system; its diverse cells perform a range of functions essential to life, not only secretion, digestion, absorption and excretion, but also, very importantly, defence. The gastrointestinal tract acts not only as a barrier to harmful materials and pathogens but also contains the vast number of beneficial bacterial populations that make up the microbiota. Communication between the cells of the gastrointestinal tract and the central nervous and endocrine systems modifies behaviour; the organisms of the microbiota also contribute to this brain-gut-enteric microbiota axis. Age-related physiological changes in the gut are not only common, but also variable, and likely to be influenced by external factors as well as intrinsic aging of the cells involved. The cellular and molecular changes exhibited by the aging gut cells also vary. Aging intestinal smooth muscle cells exhibit a number of changes in the signalling pathways that regulate contraction. There is some evidence for age-associated degeneration of neurons and glia of the enteric nervous system, although enteric neuronal losses are likely not to be nearly as extensive as previously believed. Aging enteric neurons have been shown to exhibit a senescence-associated phenotype. Epithelial stem cells exhibit increased mitochondrial mutation in aging that affects their progeny in the mucosal epithelium. Changes to the microbiota and intestinal immune system during aging are likely to contribute to wider aging of the organism and are increasingly important areas of analysis. How changes of the different cell types of the gut during aging affect the numerous cellular interactions that are essential for normal gut functions will be important areas for future aging research.
Collapse
Affiliation(s)
- M Jill Saffrey
- Department of Life Health and Chemical Sciences, Biomedical Research Network, The Open University, Milton Keynes, MK7 6AA, UK,
| |
Collapse
|
29
|
McClain J, Grubišić V, Fried D, Gomez-Suarez RA, Leinninger GM, Sévigny J, Parpura V, Gulbransen BD. Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice. Gastroenterology 2014; 146:497-507.e1. [PMID: 24211490 PMCID: PMC3935238 DOI: 10.1053/j.gastro.2013.10.061] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 10/18/2013] [Accepted: 10/30/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS In the enteric nervous system, neurotransmitters initiate changes in calcium (Ca(2+) responses) in glia, but it is not clear how this process affects intestinal function. We investigated whether Ca(2+)-mediated responses in enteric glia are required to maintain gastrointestinal function. METHODS We used in situ Ca(2+) imaging to monitor glial Ca(2+) responses, which were manipulated with pharmacologic agents or via glia-specific disruption of the gene encoding connexin-43 (Cx43) (hGFAP::CreER(T2+/-)/Cx43(f/f) mice). Gastrointestinal function was assessed based on pellet output, total gut transit, colonic bead expulsion, and muscle tension recordings. Proteins were localized and quantified by immunohistochemistry, immunoblot, and reverse transcription polymerase chain reaction analyses. RESULTS Ca(2+) responses in enteric glia of mice were mediated by Cx43 hemichannels. Cx43 immunoreactivity was confined to enteric glia within the myenteric plexus of the mouse colon; the Cx43 inhibitors carbenoxolone and 43Gap26 inhibited the ability of enteric glia to propagate Ca(2+) responses. In vivo attenuation of Ca(2+) responses in the enteric glial network slowed gut transit overall and delayed colonic transit--these changes are also observed during normal aging. Altered motility with increasing age was associated with reduced glial Ca(2+)-mediated responses and changes in glial expression of Cx43 messenger RNA and protein. CONCLUSIONS Ca(2+)-mediated responses in enteric glia regulate gastrointestinal function in mice. Altered intercellular signaling between enteric glia and neurons might contribute to motility disorders.
Collapse
Affiliation(s)
- Jonathon McClain
- Neuroscience Program and Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Vladimir Grubišić
- Department of Neurobiology, Center for Glial Biology in Medicine, Atomic Force Microscopy and Nanotechnology Laboratories, Civitan International Research Center, Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL 35294, USA
| | - David Fried
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824
| | - Roberto A Gomez-Suarez
- Department of Neurobiology, Center for Glial Biology in Medicine, Atomic Force Microscopy and Nanotechnology Laboratories, Civitan International Research Center, Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL 35294, USA.,Department of Pediatrics Division of Pediatric Gastroenterology Hepatology And Nutrition at Nemours Chlidren's Hospital. Orlando, FL 32827, USA
| | - Gina M Leinninger
- Neuroscience Program and Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Centre de recherche du CHU de Québec, Québec, QC, G1V 4G2 Canada
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Atomic Force Microscopy and Nanotechnology Laboratories, Civitan International Research Center, Evelyn F. McKnight Brain Institute, University of Alabama, Birmingham, AL 35294, USA.,Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Brian D Gulbransen
- Neuroscience Program and Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| |
Collapse
|
30
|
Janssens J, Etienne H, Idriss S, Azmi A, Martin B, Maudsley S. Systems-Level G Protein-Coupled Receptor Therapy Across a Neurodegenerative Continuum by the GLP-1 Receptor System. Front Endocrinol (Lausanne) 2014; 5:142. [PMID: 25225492 PMCID: PMC4150252 DOI: 10.3389/fendo.2014.00142] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/14/2014] [Indexed: 12/31/2022] Open
Abstract
With our increasing appreciation of the true complexity of diseases and pathophysiologies, it is clear that this knowledge needs to inform the future development of pharmacotherapeutics. For many disorders, the disease mechanism itself is a complex process spanning multiple signaling networks, tissues, and organ systems. Identifying the precise nature and locations of the pathophysiology is crucial for the creation of systemically effective drugs. Diseases once considered constrained to a limited range of organ systems, e.g., central neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington' disease (HD), the role of multiple central and peripheral organ systems in the etiology of such diseases is now widely accepted. With this knowledge, it is increasingly clear that these seemingly distinct neurodegenerative disorders (AD, PD, and HD) possess multiple pathophysiological similarities thereby demonstrating an inter-related continuum of disease-related molecular alterations. With this systems-level appreciation of neurodegenerative diseases, it is now imperative to consider that pharmacotherapeutics should be developed specifically to address the systemic imbalances that create the disorders. Identification of potential systems-level signaling axes may facilitate the generation of therapeutic agents with synergistic remedial activity across multiple tissues, organ systems, and even diseases. Here, we discuss the potentially therapeutic systems-level interaction of the glucagon-like peptide 1 (GLP-1) ligand-receptor axis with multiple aspects of the AD, PD, and HD neurodegenerative continuum.
Collapse
Affiliation(s)
- Jonathan Janssens
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Antwerp, Belgium
| | - Harmonie Etienne
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Antwerp, Belgium
| | - Sherif Idriss
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Antwerp, Belgium
| | - Abdelkrim Azmi
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Antwerp, Belgium
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Stuart Maudsley
- Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Antwerp, Belgium
- *Correspondence: Stuart Maudsley, Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, Universiteitsplein 1, Building V, Antwerpen B2610, Belgium e-mail:
| |
Collapse
|
31
|
Gamage PPKM, Ranson RN, Patel BA, Yeoman MS, Saffrey MJ. Myenteric neuron numbers are maintained in aging mouse distal colon. Neurogastroenterol Motil 2013; 25:e495-e505. [PMID: 23517051 DOI: 10.1111/nmo.12114] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/15/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Age-associated myenteric neuronal loss has been described in several species. In some studies,cholinergic neurons have been reported to be selectively vulnerable, whereas nitrergic neurons are spared. Aging of the mouse enteric nervous system(ENS) and the subtypes of mouse myenteric neurons that may be lost have been little studied. We therefore investigated changes in the numbers of total neurons and two neuronal subpopulations in the mouse distal colon during aging. METHODS Wholemount preparations from 3–4-, 12–13-, 18–19-, and 24–25-month-old C57BL/6 mice were double immunolabeled with HuC/D antibody to identify the total neuronal population and antisera to either calbindin or neuronal nitric oxide synthase (nNOS) to identify myenteric neuronal subpopulations. Samples were analyzed by confocal microscopy. New procedures were employed to ensure unbiased counting and to correct for changes in gut dimensions with age and stretch during sample preparation. The density of nerve fibers in the tertiary plexus was also studied. KEY RESULTS No significant change in numbers of total neurons or of either subpopulation with age was measured, but because of gut growth, the density of myenteric neurons decreased between 3–4 and 12–13 months. The density of nNOS-immunoreactive nerve fibers in the tertiary plexus increased significantly with age, up to 18–19 months. Numerous swollen processes of CB and nNOS-immunoreactive neurons were observed in 18–19- and 24–25-month-old animals. Conclusions &Inferences These results indicate that aging does not result in a loss of myenteric neurons in mouse distal colon at the ages studied, although neurodegenerative changes, which may impact on neuronal function, do occur.
Collapse
Affiliation(s)
- P. P. K. M. Gamage
- Department of Life, Health and Chemical Sciences; Biomedical Research Network; Open University; Walton Hall; Milton Keynes; MK7 6AA; UK
| | - R. N. Ranson
- Faculty of Health and Life Sciences; Northumbria University; Ellison Building; Newcastle upon Tyne; NE1 8ST; UK
| | - B. A. Patel
- School of Pharmacy and Biomolecular Sciences, Huxley Building; University of Brighton; Lewes Road; Brighton; BN2 4GJ; UK
| | - M. S. Yeoman
- School of Pharmacy and Biomolecular Sciences, Huxley Building; University of Brighton; Lewes Road; Brighton; BN2 4GJ; UK
| | - M. J. Saffrey
- Department of Life, Health and Chemical Sciences; Biomedical Research Network; Open University; Walton Hall; Milton Keynes; MK7 6AA; UK
| |
Collapse
|
32
|
Saffrey MJ. Cellular changes in the enteric nervous system during ageing. Dev Biol 2013; 382:344-55. [PMID: 23537898 DOI: 10.1016/j.ydbio.2013.03.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/22/2013] [Accepted: 03/19/2013] [Indexed: 02/06/2023]
Abstract
The intrinsic neurons of the gut, enteric neurons, have an essential role in gastrointestinal functions. The enteric nervous system is plastic and continues to undergo changes throughout life, as the gut grows and responds to dietary and other environmental changes. Detailed analysis of changes in the ENS during ageing suggests that enteric neurons are more vulnerable to age-related degeneration and cell death than neurons in other parts of the nervous system, although there is considerable variation in the extent and time course of age-related enteric neuronal loss reported in different studies. Specific neuronal subpopulations, particularly cholinergic myenteric neurons, may be more vulnerable than others to age-associated loss or damage. Enteric degeneration and other age-related neuronal changes may contribute to gastrointestinal dysfunction that is common in the elderly population. Evidence suggests that caloric restriction protects against age-associated loss of enteric neurons, but recent advances in the understanding of the effects of the microbiota and the complex interactions between enteric ganglion cells, mucosal immune system and intestinal epithelium indicate that other factors may well influence ageing of enteric neurons. Much remains to be understood about the mechanisms of neuronal loss and damage in the gut, although there is evidence that reactive oxygen species, neurotrophic factor dysregulation and/or activation of a senescence associated phenotype may be involved. To date, there is no evidence for ongoing neurogenesis that might replace dying neurons in the ageing gut, although small local sites of neurogenesis would be difficult to detect. Finally, despite the considerable evidence for enteric neurodegeneration during ageing, and evidence for some physiological changes in animal models, the ageing gut appears to maintain its function remarkably well in animals that exhibit major neuronal loss, indicating that the ENS has considerable functional reserve.
Collapse
Affiliation(s)
- M Jill Saffrey
- Department Life, Health & Chemical Sciences, Open University, Walton Hall, Milton Keynes MK7 6AA, United Kingdom.
| |
Collapse
|
33
|
Natale G, Pasquali L, Paparelli A, Fornai F. Parallel manifestations of neuropathologies in the enteric and central nervous systems. Neurogastroenterol Motil 2011; 23:1056-65. [PMID: 21951862 DOI: 10.1111/j.1365-2982.2011.01794.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Neurodegenerative diseases may extend outside the central nervous system (CNS) and involve the gastrointestinal (GI) tract. The gut would appear to be a pathological marker for neurodegeneration, as well as a site for studying the pathophysiology of neurodegeneration. In fact, both in the ENS and CNS, misfolded proteins are likely to initiate a process of neurodegeneration. For example, the very same protein aggregates can be detected both in the ENS and CNS. In both systems, misfolded proteins are likely to share common cell-to-cell diffusion mechanisms, which may occur through a parallel prion-like diffusion process. Independently from the enteric or central origin, misfolded proteins may proceed along the following steps, they: (i) form aggregates; (ii) are expressed on plasma membrane; (iii) are secreted extracellularly; (iv) are glycated to form advanced glycation end-products (AGEs); (v) are internalized through specific receptors placed on neighboring cells (RAGEs); (vi) are cleared by autophagy; and (vii) are neurotoxic. These features are common for a-synuclein (in Parkinson's disease and other synucleinopathies), β-amyloid and tau (in degenerative dementia), SOD-1 and TDP43 (in amyotrophic lateral sclerosis), and PrPsc (in prion diseases). While in some diseases these features are common to both ENS and CNS, in others this remains a working hypothesis. PURPOSE This review analyzes GI alterations from a pathological perspective to assess whether the enteric nervous system (ENS) mirrors the neuropathology described in the CNS. We discuss the potential mechanisms that lead to the onset and spread of neurodegeneration within the gut, from the gut to the brain, and vice versa.
Collapse
Affiliation(s)
- G Natale
- Department of Human Morphology and Applied Biology, University of Pisa, Italy
| | | | | | | |
Collapse
|
34
|
Amyloid precursor protein expression modulates intestine immune phenotype. J Neuroimmune Pharmacol 2011; 7:215-30. [PMID: 22124967 DOI: 10.1007/s11481-011-9327-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 11/14/2011] [Indexed: 12/16/2022]
Abstract
Amyloid precursor protein (APP) is widely expressed across many tissue and cell types. Proteolytic processing of the protein gives rise to a plethora of protein fragments with varied biological activities. Although a large amount of data has been generated describing the metabolism of the protein in neurons, its role in regulating the phenotype of other cells remains unclear. Based upon prior work demonstrating that APP regulates the activation phenotype of monocytic lineage cells, we hypothesized that APP can regulate macrophage activation phenotype in tissues other than brain. Ileums of the small intestines from C57BL6/J wild type and APP(-/-) mice were compared as a representative tissue normally associated with abundant macrophage infiltration. APP(-/-) intestines demonstrated diminished CD68 immunoreactivity compared to wild type mice. This correlated with significantly less cyclooxygenase-2 (cox-2), CD68, CD40, CD11c, and βIII-tubulin protein levels. Peritoneal macrophages from APP(-/-) mice demonstrated decreased in vitro migratory ability compared to wild type cells and diminished basal KC cytokine secretion. Whereas, APP(-/-) intestinal macrophages had an increase in basal KC cytokine secretion compared to wild type cells. Conversely, there was a significant decrease in multiple cytokine levels in APP(-/-) compared to wild type ileums. Finally, APP(-/-) mice demonstrated impaired absorption and increased motility compared to wild type mice. These data demonstrate the APP expression regulates immune cell secretions and phenotype and intestinal function. This data set describes a novel function for this protein or its metabolites that may be relevant not only for Alzheimer's disease but a range of immune-related disorders.
Collapse
|
35
|
Kleinschmidt S, Nolte I, Hewicker-Trautwein M. Structural and Functional Components of the Feline Enteric Nervous System. Anat Histol Embryol 2011; 40:450-6. [DOI: 10.1111/j.1439-0264.2011.01091.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|