Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 545
Filter
1.
Proc Natl Acad Sci U S A ; 121(39): e2406479121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39284050

ABSTRACT

Parkinson's disease (PD) is typically a sporadic late-onset disorder, which has made it difficult to model in mice. Several transgenic mouse models bearing mutations in SNCA, which encodes alpha-Synuclein (α-Syn), have been made, but these lines do not express SNCA in a physiologically accurate spatiotemporal pattern, which limits the ability of the mice to recapitulate the features of human PD. Here, we generated knock-in mice bearing the G51D SNCA mutation. After establishing that their motor symptoms begin at 9 mo of age, we then sought earlier pathologies. We assessed the phosphorylation at Serine 129 of α-Syn in different tissues and detected phospho-α-Syn in the olfactory bulb and enteric nervous system at 3 mo of age. Olfactory deficit and impaired gut transit followed at 6 mo, preceding motor symptoms. The SncaG51D mice thus parallel the progression of human PD and will enable us to study PD pathogenesis and test future therapies.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Parkinson Disease , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Parkinson Disease/pathology , Mice, Transgenic , Phosphorylation , Olfaction Disorders/genetics , Olfaction Disorders/metabolism , Olfaction Disorders/physiopathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology , Humans , Male
3.
JCI Insight ; 9(17)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042470

ABSTRACT

The goal of this study was to determine if transplantation of enteric neural stem cells (ENSCs) can rescue the enteric nervous system, restore gut motility, reduce colonic inflammation, and improve survival in the Ednrb-KO mouse model of Hirschsprung disease (HSCR). ENSCs were isolated from mouse intestine, expanded to form neurospheres, and microinjected into the colons of recipient Ednrb-KO mice. Transplanted ENSCs were identified in recipient colons as cell clusters in "neo-ganglia." Immunohistochemical evaluation demonstrated extensive cell migration away from the sites of cell delivery and across the muscle layers. Electrical field stimulation and optogenetics showed significantly enhanced contractile activity of aganglionic colonic smooth muscle following ENSC transplantation and confirmed functional neuromuscular integration of the transplanted ENSC-derived neurons. ENSC injection also partially restored the colonic migrating motor complex. Histological examination revealed a significant reduction in inflammation in ENSC-transplanted aganglionic recipient colon compared with that of sham-operated mice. Interestingly, mice that received cell transplant also had prolonged survival compared with controls. This study demonstrates that ENSC transplantation can improve outcomes in HSCR by restoring gut motility and reducing the severity of Hirschsprung-associated enterocolitis, the leading cause of death in human HSCR.


Subject(s)
Disease Models, Animal , Enteric Nervous System , Gastrointestinal Motility , Hirschsprung Disease , Mice, Knockout , Neural Stem Cells , Animals , Hirschsprung Disease/therapy , Hirschsprung Disease/pathology , Neural Stem Cells/transplantation , Gastrointestinal Motility/physiology , Mice , Enteric Nervous System/physiopathology , Colon/pathology , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Stem Cell Transplantation/methods , Cell Movement , Female , Humans , Male , Muscle, Smooth
4.
Obes Surg ; 34(9): 3147-3160, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39046627

ABSTRACT

PURPOSE: Changes in autonomic (ANS) and enteric nervous systems (ENS) may be involved in pathogenesis of obesity. We hypothesized that baseline autonomic and enteric parameters may predict outcomes of diverse obesity therapies. MATERIAL AND METHODS: We studied ANS and ENS physiology in 37 patients (8 male, 29 female, age 45 years, weight 129.7 kg) at 4 centers in patients undergoing medical (9: low-calorie diet) versus invasive (22: 16 sleeve, 6 bypass) and semi-invasive (6: 2 band, 2 high energy stimulation, 2 aspiration) weight loss therapies. Weight loss was reported as percent weight loss from baseline to latest values at 1 year and in some up to 5 years; classified as < or > /= 20% for each group. ANS testing included sympathetic adrenergic function by measuring reflex vasoconstriction and postural adjustment ratio. ENS was measured non-invasively using cutaneous low-resolution electrogastrogram. RESULTS: Percent weight loss was greater with the invasive (28.5%) than semi-invasive (9.1%) or non-invasive low-calorie diet (4.4%) (p < .001). Percent weight loss at 1 year (and up to 5 years) corresponded to the adrenergic measure of postural adjustment ratio (r = .42, p = .012), total pulse amplitude at rest (r = .56, p < .001), and electrogastrogram standing-to-rest difference (r = .33, p = .056). CONCLUSION: Baseline autonomic and enteric function measures correspond to percentage with loss in this pilot study using diverse weight loss methods. Autonomic and enteric profiling has potential clinical use for evaluation and treatment of obesity but needed larger controlled trials.


Subject(s)
Autonomic Nervous System , Obesity, Morbid , Weight Loss , Humans , Female , Male , Middle Aged , Weight Loss/physiology , Autonomic Nervous System/physiopathology , Obesity, Morbid/therapy , Obesity, Morbid/physiopathology , Adult , Enteric Nervous System/physiopathology , Treatment Outcome , Bariatric Surgery , Obesity/therapy , Obesity/physiopathology , Caloric Restriction , Predictive Value of Tests , Diet, Reducing
5.
Med Sci (Paris) ; 40(6-7): 544-549, 2024.
Article in French | MEDLINE | ID: mdl-38986099

ABSTRACT

The enteric nervous system (ENS), often called the "second brain", plays a crucial role in regulating digestive functions. Dysfunctions of the ENS are associated with several diseases such as Parkinson's disease. Recent studies suggest that early digestive disorders, notably chronic constipation, may be early signs of this neurodegenerative disease. Three-dimensional imaging of the ENS offers new insights into early diagnosis, in particular through the analysis of intestinal biopsies. This new research axis raises questions about the intestinal cause of Parkinson's disease, and opens the door to a better understanding and earlier treatment of this disease.


Title: L'intestin, lanceur d'alerte, dans les prémices de la maladie de Parkinson. Abstract: Le système nerveux entérique (SNE), souvent qualifié de « deuxième cerveau ¼, joue un rôle crucial dans la régulation des fonctions digestives. Des dysfonctionnements du SNE sont associés à diverses maladies telles que la maladie de Parkinson. Des études récentes suggèrent que les troubles digestifs précoces, notamment la constipation chronique, pourraient être des signes avant-coureurs de cette maladie neurodégénérative. L'imagerie tridimensionnelle du SNE offre de nouvelles perspectives pour un diagnostic précoce via notamment l'analyse de biopsies intestinales. Ce nouvel axe de recherche soulève des questions sur l'origine intestinale de la maladie de Parkinson et ouvre la porte à une meilleure compréhension et une prise en charge anticipée de cette maladie.


Subject(s)
Enteric Nervous System , Parkinson Disease , Humans , Parkinson Disease/pathology , Parkinson Disease/diagnosis , Enteric Nervous System/pathology , Enteric Nervous System/physiopathology , Enteric Nervous System/physiology , Early Diagnosis , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Gastrointestinal Tract/physiology , Animals , Intestines/pathology , Intestines/physiology
6.
World J Gastroenterol ; 30(22): 2852-2865, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38947292

ABSTRACT

Diabetes, commonly known for its metabolic effects, also critically affects the enteric nervous system (ENS), which is essential in regulating gastrointestinal (GI) motility, secretion, and absorption. The development of diabetes-induced enteric neuropathy can lead to various GI dysfunctions, such as gastroparesis and irregular bowel habits, primarily due to disruptions in the function of neuronal and glial cells within the ENS, as well as oxidative stress and inflammation. This editorial explores the pathophysiological mechanisms underlying the development of enteric neuropathy in diabetic patients. Additionally, it discusses the latest advances in diagnostic approaches, emphasizing the need for early detection and intervention to mitigate GI complications in diabetic individuals. The editorial also reviews current and emerging therapeutic strategies, focusing on pharmacological treatments, dietary management, and potential neuromodulatory interventions. Ultimately, this editorial highlights the necessity of a multidisciplinary approach in managing enteric neuropathy in diabetes, aiming to enhance patient quality of life and address a frequently overlooked complication of this widespread disease.


Subject(s)
Diabetic Neuropathies , Enteric Nervous System , Gastrointestinal Motility , Humans , Diabetic Neuropathies/etiology , Diabetic Neuropathies/therapy , Diabetic Neuropathies/diagnosis , Diabetic Neuropathies/physiopathology , Enteric Nervous System/physiopathology , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/etiology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiopathology , Gastroparesis/therapy , Gastroparesis/physiopathology , Gastroparesis/diagnosis , Gastroparesis/etiology , Oxidative Stress , Quality of Life
7.
Life Sci ; 351: 122793, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38848938

ABSTRACT

The enteric nervous system (ENS) regulates numerous functional and immunological attributes of the gastrointestinal tract. Alterations in ENS cell function have been linked to intestinal outcomes in various metabolic, intestinal, and neurological disorders. Chronic kidney disease (CKD) is associated with a challenging intestinal environment due to gut dysbiosis, which further affects patient quality of life. Although the gut-related repercussions of CKD have been thoroughly investigated, the involvement of the ENS in this puzzle remains unclear. ENS cell dysfunction, such as glial reactivity and alterations in cholinergic signaling in the small intestine and colon, in CKD are associated with a wide range of intestinal pathways and responses in affected patients. This review discusses how the ENS is affected in CKD and how it is involved in gut-related outcomes, including intestinal permeability, inflammation, oxidative stress, and dysmotility.


Subject(s)
Enteric Nervous System , Renal Insufficiency, Chronic , Humans , Enteric Nervous System/physiopathology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/metabolism , Animals , Kidney/physiopathology , Gastrointestinal Microbiome , Oxidative Stress , Dysbiosis/complications , Gastrointestinal Tract/physiopathology , Gastrointestinal Tract/metabolism , Inflammation
8.
Brain Behav Immun ; 119: 867-877, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750700

ABSTRACT

The gastrointestinal tract is one of the main organs affected during systemic inflammation and disrupted gastrointestinal motility is a major clinical manifestation. Many studies have investigated the involvement of neuroimmune interactions in regulating colonic motility during localized colonic inflammation, i.e., colitis. However, little is known about how the enteric nervous system and intestinal macrophages contribute to dysregulated motility during systemic inflammation. Given that systemic inflammation commonly results from the innate immune response against bacterial infection, we mimicked bacterial infection by administering lipopolysaccharide (LPS) to rats and assessed colonic motility using ex vivo video imaging techniques. We utilized the Cx3cr1-Dtr rat model of transient depletion of macrophages to investigate the role of intestinal macrophages in regulating colonic motility during LPS infection. To investigate the role of inhibitory enteric neurotransmission on colonic motility following LPS, we applied the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (NOLA). Our results confirmed an increase in colonic contraction frequency during LPS-induced systemic inflammation. However, neither the depletion of intestinal macrophages, nor the suppression of inhibitory enteric nervous system activity impacted colonic motility disruption during inflammation. This implies that the interplay between the enteric nervous system and intestinal macrophages is nuanced, and complex, and further investigation is needed to clarify their joint roles in colonic motility.


Subject(s)
Enteric Nervous System , Gastrointestinal Motility , Inflammation , Lipopolysaccharides , Macrophages , Animals , Lipopolysaccharides/pharmacology , Rats , Gastrointestinal Motility/physiology , Macrophages/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Enteric Nervous System/physiopathology , Enteric Nervous System/metabolism , Male , Brain-Gut Axis/physiology , Colon/metabolism , Gastrointestinal Tract/metabolism , Colitis/physiopathology , Colitis/metabolism , Colitis/chemically induced , Brain/metabolism , Rats, Sprague-Dawley , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/metabolism
9.
Cell Mol Gastroenterol Hepatol ; 18(2): 101332, 2024.
Article in English | MEDLINE | ID: mdl-38479486

ABSTRACT

The enteric nervous system (ENS) controls gastrointestinal (GI) motility, and defects in ENS development underlie pediatric GI motility disorders. In disorders such as Hirschsprung's disease (HSCR), pediatric intestinal pseudo-obstruction (PIPO), and intestinal neuronal dysplasia type B (INDB), ENS structure is altered with noted decreased neuronal density in HSCR and reports of increased neuronal density in PIPO and INDB. The developmental origin of these structural deficits is not fully understood. Here, we review the current understanding of ENS development and pediatric GI motility disorders incorporating new data on ENS structure. In particular, emerging evidence demonstrates that enteric neurons are patterned into circumferential stripes along the longitudinal axis of the intestine during mouse and human development. This novel understanding of ENS structure proposes new questions about the pathophysiology of pediatric GI motility disorders. If the ENS is organized into stripes, could the observed changes in enteric neuron density in HSCR, PIPO, and INDB represent differences in the distribution of enteric neuronal stripes? We review mechanisms of striped patterning from other biological systems and propose how defects in striped ENS patterning could explain structural deficits observed in pediatric GI motility disorders.


Subject(s)
Enteric Nervous System , Gastrointestinal Motility , Hirschsprung Disease , Enteric Nervous System/physiopathology , Enteric Nervous System/pathology , Humans , Animals , Hirschsprung Disease/pathology , Hirschsprung Disease/physiopathology , Mice , Neurons/pathology , Neurons/metabolism , Intestinal Pseudo-Obstruction/pathology , Intestinal Pseudo-Obstruction/physiopathology , Body Patterning
11.
Parkinsonism Relat Disord ; 122: 106101, 2024 May.
Article in English | MEDLINE | ID: mdl-38519273

ABSTRACT

We recently proposed a new disease model of Parkinson's disease - the a-Synuclein Origin site and Connectome model. The model posits that the initial pathology starts either in the olfactory bulb or amygdala leading to a brain-first subtype, or in the enteric nervous system leading to a body-first subtype. These subtypes should be distinguishable early in the disease course on a range of imaging, clinical, and neuropathological markers. Here, we review recent original human studies, which tested the predictions of the model. Molecular imaging studies were generally in agreement with the model, whereas structural imaging studies, such as MRI volumetry, showed conflicting findings. Most large-scale clinical studies were supportive, reporting clustering of relevant markers of the body-first subtype, including REM-sleep behavior disorder, constipation, autonomic dysfunction, neuropsychiatric symptoms, and cognitive impairment. Finally, studies of a-synuclein deposition in antemortem and postmortem tissues revealed distribution of pathology, which generally supports the model.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Parkinson Disease/diagnostic imaging , alpha-Synuclein/metabolism , Brain/diagnostic imaging , Brain/pathology , Connectome , Enteric Nervous System/pathology , Enteric Nervous System/physiopathology
12.
Arthritis Rheumatol ; 76(1): 92-99, 2024 01.
Article in English | MEDLINE | ID: mdl-37530745

ABSTRACT

OBJECTIVE: Autoantibodies are clinically useful in phenotyping patients with systemic sclerosis (SSc). Gastrointestinal (GI) function is regulated by the enteric nervous system (ENS) and commonly impaired in SSc, suggesting that the SSc autoimmune response may target ENS antigens. We sought to identify novel anti-ENS autoantibodies with an aim to clinically phenotype SSc GI dysfunction. METHODS: Serum from a patient with SSc with GI dysfunction but without defined SSc-associated autoantibodies was used for autoantibody discovery. Immunoprecipitations performed with murine myenteric plexus lysates were on-bead digested, and autoantigens were identified by mass spectrometry. Prevalence was determined, and clinical features associated with novel autoantibodies were evaluated in a SSc cohort using regression analyses. The expression of gephyrin in human GI tract tissue was examined by immunohistochemistry. RESULTS: We identified gephyrin as a novel SSc autoantigen. Anti-gephyrin antibodies were present in 9% of patients with SSc (16/188) and absent in healthy controls (0/46). Anti-gephyrin antibody-positive patients had higher constipation scores (1.00 vs 0.50, P = 0.02) and were more likely to have severe constipation and severe distention/bloating (46% vs 15%, P = 0.005; 54% vs 25%, P = 0.023, respectively). Anti-gephyrin antibody levels were significantly higher among patients with severe constipation (0.04 vs 0.00; P = 0.001) and severe distention and bloating (0.03 vs 0.004; P = 0.010). Severe constipation was associated with anti-gephyrin antibodies even in the adjusted model. Importantly, gephyrin was expressed in the ENS, which regulates gut motility. CONCLUSION: Gephyrin is a novel ENS autoantigen that is expressed in human myenteric ganglia. Anti-gephyrin autoantibodies are associated with the presence and severity of constipation in patients with SSc.


Subject(s)
Autoantibodies , Membrane Proteins , Scleroderma, Systemic , Membrane Proteins/metabolism , Autoantigens/metabolism , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Autoantibodies/analysis , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiopathology , Humans , Animals , Mice , Neurons/metabolism , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology
13.
Neurosci Lett ; 806: 137221, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37031943

ABSTRACT

Enteric glia are a unique population of peripheral neuroglia associated with the enteric nervous system (ENS) throughout the digestive tract. The emerging data from the latest glial biology studies unveiled enteric glia as a heterogenic population with plastic and adaptative abilities that display phenotypic and functional changes upon distinct extrinsic cues. This aspect is essential in the dynamic signaling that enteric glia engage with neurons and other neighboring cells within the intestinal wall, such as epithelial, endocrine, and immune cells to maintain local homeostasis. Likewise, enteric glia sense signals from luminal microbes, although the extent of this active communication is still unclear. In this minireview, we discuss the recent findings that support glia-microbes crosstalk in the intestine in health and disease, pointing out the critical aspects that require further investigation.


Subject(s)
Disease , Enteric Nervous System , Gastrointestinal Microbiome , Health , Neuroglia , Humans , Biodiversity , Enteric Nervous System/cytology , Enteric Nervous System/physiology , Enteric Nervous System/physiopathology , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Inflammation/microbiology , Neuroglia/physiology , Probiotics , Animals
14.
Drugs ; 82(2): 169-197, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35076890

ABSTRACT

There has been exponential growth in the awareness and understanding of gastrointestinal (GI) dysfunction in Parkinson's disease (PD) over the past 3 decades. The clinical features of GI dysfunction in PD have been clearly identified and innovative research has demonstrated the presence of pathology within the enteric nervous system (ENS) in individuals with PD, leading to suggestions that the GI system may be ground zero for the genesis and the portal of entry of PD pathology, which then ascends via the vagus nerve to the central nervous system (CNS). This theory, as well as the more recent recognition of the association of PD with dysbiosis within the gut microbiota, has been the object of intense study and scrutiny. Since most PD medications are absorbed through the GI system, the need for better understanding of changes within the GI tract that may potentially affect the pattern of response to medications has become evident. In this review, current knowledge of the pathophysiology of changes within the GI tract and the gut microbiome of individuals with PD, including changes that occur with progression of the disease, will be addressed. We focus on common clinical GI problems in PD that can arise from different segments of the GI tract. Relevant diagnostic evaluations and treatment options for each of these problems will be reviewed.


Subject(s)
Antiparkinson Agents/therapeutic use , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/therapy , Gastrointestinal Motility/physiology , Parkinson Disease/physiopathology , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Deglutition Disorders/physiopathology , Diet , Enteric Nervous System/physiopathology , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Transit/physiology , Humans , Oral Health , Weight Loss/physiology
15.
Gastroenterology ; 162(1): 68-87.e1, 2022 01.
Article in English | MEDLINE | ID: mdl-34717924

ABSTRACT

Gastroparesis is characterized by symptoms suggestive of, and objective evidence of, delayed gastric emptying in the absence of mechanical obstruction. This review addresses the normal emptying of solids and liquids from the stomach and details the myogenic and neuromuscular control mechanisms, including the specialized function of the pyloric sphincter, that result in normal emptying, based predominantly on animal research. A clear understanding of fundamental mechanisms is necessary to comprehend derangements leading to gastroparesis, and additional research on human gastric muscles is needed. The section on pathophysiology of gastroparesis considers neuromuscular diseases that affect nonsphincteric gastric muscle, disorders of the extrinsic neural control, and pyloric dysfunction that lead to gastroparesis. The potential cellular basis for gastroparesis is attributed to the effects of oxidative stress and inflammation, with increased pro-inflammatory and decreased resident macrophages, as observed in full-thickness biopsies from patients with gastroparesis. Predominant diagnostic tests involving measurements of gastric emptying, the use of a functional luminal imaging probe, and high-resolution antral duodenal manometry in characterizing the abnormal motor functions at the gastroduodenal junction are discussed. Management is based on supporting nutrition; dietary interventions, including the physical reduction in particle size of solid foods; pharmacological agents, including prokinetics and anti-emetics; and interventions such as gastric electrical stimulation and pyloromyotomy. These are discussed briefly, and comment is added on the potential for individualized treatments in the future, based on optimal gastric emptying measurement and objective documentation of the underlying pathophysiology causing the gastroparesis.


Subject(s)
Enteric Nervous System/physiopathology , Gastric Emptying , Gastroparesis/physiopathology , Pylorus/innervation , Animals , Gastroparesis/diagnosis , Gastroparesis/therapy , Humans , Predictive Value of Tests , Treatment Outcome
16.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34755545

ABSTRACT

Allosteric modulators (AMs) are molecules that can fine-tune signaling by G protein-coupled receptors (GPCRs). Although they are a promising therapeutic approach for treating a range of disorders, allosteric modulation of GPCRs in the context of the enteric nervous system (ENS) and digestive dysfunction remains largely unexplored. This study examined allosteric modulation of the delta opioid receptor (DOR) in the ENS and assessed the suitability of DOR AMs for the treatment of irritable bowel syndrome (IBS) symptoms using mouse models. The effects of the positive allosteric modulator (PAM) of DOR, BMS-986187, on neurogenic contractions of the mouse colon and on DOR internalization in enteric neurons were quantified. The ability of BMS-986187 to influence colonic motility was assessed both in vitro and in vivo. BMS-986187 displayed DOR-selective PAM-agonist activity and orthosteric agonist probe dependence in the mouse colon. BMS-986187 augmented the inhibitory effects of DOR agonists on neurogenic contractions and enhanced reflex-evoked DOR internalization in myenteric neurons. BMS-986187 significantly increased DOR endocytosis in myenteric neurons in response to the weakly internalizing agonist ARM390. BMS-986187 reduced the generation of complex motor patterns in the isolated intact colon. BMS-986187 reduced fecal output and diarrhea onset in the novel environment stress and castor oil models of IBS symptoms, respectively. DOR PAMs enhance DOR-mediated signaling in the ENS and have potential benefit for the treatment of dysmotility. This study provides proof of concept to support the use of GPCR AMs for the treatment of gastrointestinal motility disorders.NEW & NOTEWORTHY This study assesses the use of positive allosteric modulation as a pharmacological approach to enhance opioid receptor signaling in the enteric nervous system. We demonstrate that selective modulation of endogenous delta opioid receptor signaling can suppress colonic motility without causing constipation. We propose that allosteric modulation of opioid receptor signaling may be a therapeutic strategy to normalize gastrointestinal motility in conditions such as irritable bowel syndrome.


Subject(s)
Enteric Nervous System/drug effects , Gastrointestinal Motility/drug effects , Receptors, Opioid, delta/drug effects , Xanthones/pharmacology , Analgesics, Opioid/pharmacology , Benzamides/pharmacology , Colon/drug effects , Enteric Nervous System/physiopathology , Gastrointestinal Motility/physiology , Humans , Receptors, Opioid/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects
18.
Gut Microbes ; 13(1): 1996848, 2021.
Article in English | MEDLINE | ID: mdl-34812107

ABSTRACT

Amyotrophic Lateral Sclerosis is a neuromuscular disease characterized by the progressive death of motor neurons and muscle atrophy. The gastrointestinal symptoms in ALS patients were largely ignored or underestimated. The relationship between the enteric neuromuscular system and microbiome in ALS progression is unknown. We performed longitudinal studies on the enteric neuron system (ENS) and microbiome in the ALS human-SOD1G93A (Superoxide Dismutase 1) transgenic mice. We treated age-matched wild-type and ALS mice with butyrate or antibiotics to investigate the microbiome and neuromuscular functions. We examined intestinal mobility, microbiome, an ENS marker GFAP (Glial Fibrillary Acidic Protein), a smooth muscle marker (SMMHC, Smooth Muscle Myosin Heavy Chain), and human colonoids. The distribution of human-G93A-SOD1 protein was tested as an indicator of ALS progression. At 2-month-old before ALS onset, SOD1G93A mice had significantly lower intestinal mobility, decreased grip strength, and reduced time in the rotarod. We observed increased GFAP and decreased SMMHC expression. These changes correlated with consistent increased aggregation of mutated SOD1G93A in the colon, small intestine, and spinal cord. Butyrate or antibiotics treated SOD1G93A mice had a significantly longer latency to fall in the rotarod test, reduced SOD1G93A aggregation, and enhanced enteric neuromuscular function. Feces from 2-month-old SOD1G93A mice significantly enhanced SOD1G93A aggregation in human colonoids transfected with a SOD1G93A-GFP plasmid. Longitudinal studies of microbiome data further showed the altered bacterial community related to autoimmunity (e.g., Clostridium sp. ASF502, Lachnospiraceae bacterium A4), inflammation (e.g., Enterohabdus Muris,), and metabolism (e.g., Desulfovibrio fairfieldensis) at 1- and 2-month-old SOD1G93A mice, suggesting the early microbial contribution to the pathological changes. We have demonstrated a novel link between the microbiome, hSOD1G93A aggregation, and intestinal mobility. Dysbiosis occurred at the early stage of the ALS mice before observed mutated-SOD1 aggregation and dysfunction of ENS. Manipulating the microbiome improves the muscle performance of SOD1G93A mice. We provide insights into the fundamentals of intestinal neuromuscular function and microbiome in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/microbiology , Dysbiosis/microbiology , Enteric Nervous System/physiopathology , Muscle, Smooth/physiopathology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Anti-Bacterial Agents/therapeutic use , Butyrates/therapeutic use , Disease Models, Animal , Dysbiosis/drug therapy , Dysbiosis/physiopathology , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Motility/drug effects , Humans , Intestine, Small/innervation , Intestine, Small/metabolism , Intestine, Small/pathology , Intestine, Small/physiopathology , Longitudinal Studies , Mice , Mice, Transgenic , Muscle Strength/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/microbiology , Protein Aggregation, Pathological/physiopathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
19.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G552-G575, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34612070

ABSTRACT

Our understanding of human colonic motility, and autonomic reflexes that generate motor patterns, has increased markedly through high-resolution manometry. Details of the motor patterns are emerging related to frequency and propagation characteristics that allow linkage to interstitial cells of Cajal (ICC) networks. In studies on colonic motor dysfunction requiring surgery, ICC are almost always abnormal or significantly reduced. However, there are still gaps in our knowledge about the role of ICC in the control of colonic motility and there is little understanding of a mechanistic link between ICC abnormalities and colonic motor dysfunction. This review will outline the various ICC networks in the human colon and their proven and likely associations with the enteric and extrinsic autonomic nervous systems. Based on our extensive knowledge of the role of ICC in the control of gastrointestinal motility of animal models and the human stomach and small intestine, we propose how ICC networks are underlying the motor patterns of the human colon. The role of ICC will be reviewed in the autonomic neural reflexes that evoke essential motor patterns for transit and defecation. Mechanisms underlying ICC injury, maintenance, and repair will be discussed. Hypotheses are formulated as to how ICC dysfunction can lead to motor abnormalities in slow transit constipation, chronic idiopathic pseudo-obstruction, Hirschsprung's disease, fecal incontinence, diverticular disease, and inflammatory conditions. Recent studies on ICC repair after injury hold promise for future therapies.


Subject(s)
Colon/pathology , Colonic Diseases/pathology , Defecation , Gastrointestinal Motility , Interstitial Cells of Cajal/pathology , Animals , Autonomic Nervous System/physiopathology , Colon/innervation , Colon/metabolism , Colonic Diseases/metabolism , Colonic Diseases/physiopathology , Colonic Pseudo-Obstruction/metabolism , Colonic Pseudo-Obstruction/pathology , Colonic Pseudo-Obstruction/physiopathology , Constipation/metabolism , Constipation/pathology , Constipation/physiopathology , Enteric Nervous System/physiopathology , Fecal Incontinence/metabolism , Fecal Incontinence/pathology , Fecal Incontinence/physiopathology , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Hirschsprung Disease/physiopathology , Humans , Interstitial Cells of Cajal/metabolism , Manometry
20.
Mil Med Res ; 8(1): 50, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34503577

ABSTRACT

By its nature, Gulf war illness (GWI) is multisymptomatic and affects several organ systems in the body. Along with other symptoms, veterans who suffer from GWI commonly report chronic gastrointestinal issues such as constipation, pain, indigestion, etc. However, until recently, most attention has been focused on neurological disturbances such as cognitive impairments, chronic fatigue, and chronic pain among affected veterans. With such high prevalence of gastrointestinal problems among Gulf war (GW) veterans, it is surprising that there is little research to investigate the mechanisms behind these issues. This review summarizes all the available works on the mechanisms behind gastrointestinal problems in GWI that have been published to date in various databases. Generally, these studies, which were done in rodent models, in vitro and human cohorts propose that an altered microbiome, a reactive enteric nervous system or a leaky gut among other possible mechanisms are the major drivers of gastrointestinal problems reported in GWI. This review aims to draw attention to the gastrointestinal tract as an important player in GWI disease pathology and a potential therapeutic target.


Subject(s)
Gastrointestinal Diseases/etiology , Persian Gulf Syndrome/complications , Enteric Nervous System/drug effects , Enteric Nervous System/physiopathology , Gastrointestinal Diseases/physiopathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Humans , Persian Gulf Syndrome/physiopathology , Veterans/statistics & numerical data
SELECTION OF CITATIONS
SEARCH DETAIL