Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.485
Filter
1.
J Vis Exp ; (207)2024 May 17.
Article in English | MEDLINE | ID: mdl-38829111

ABSTRACT

The human enteric nervous system, ENS, is a large network of glial and neuronal cell types with remarkable neurotransmitter diversity. The ENS controls bowel motility, enzyme secretion, and nutrient absorption and interacts with the immune system and the gut microbiome. Consequently, developmental and acquired defects of the ENS are responsible for many human diseases and may contribute to symptoms of Parkinson's disease. Limitations in animal model systems and access to primary tissue pose significant experimental challenges in studies of the human ENS. Here, a detailed protocol is presented for effective in vitro derivation of the ENS lineages from human pluripotent stem cells, hPSC, using defined culture conditions. Our protocol begins with directed differentiation of hPSCs to enteric neural crest cells within 15 days and yields diverse subtypes of functional enteric neurons within 30 days. This platform provides a scalable resource for developmental studies, disease modeling, drug discovery, and regenerative applications.


Subject(s)
Cell Differentiation , Enteric Nervous System , Neural Crest , Pluripotent Stem Cells , Humans , Enteric Nervous System/cytology , Pluripotent Stem Cells/cytology , Cell Differentiation/physiology , Neural Crest/cytology , Cytological Techniques/methods , Neurons/cytology
3.
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
4.
PLoS One ; 19(5): e0303914, 2024.
Article in English | MEDLINE | ID: mdl-38809858

ABSTRACT

The vertebrate enteric nervous system (ENS) is a crucial network of enteric neurons and glia resident within the entire gastrointestinal tract (GI). Overseeing essential GI functions such as gut motility and water balance, the ENS serves as a pivotal bidirectional link in the gut-brain axis. During early development, the ENS is primarily derived from enteric neural crest cells (ENCCs). Disruptions to ENCC development, as seen in conditions like Hirschsprung disease (HSCR), lead to the absence of ENS in the GI, particularly in the colon. In this study, using zebrafish, we devised an in vivo F0 CRISPR-based screen employing a robust, rapid pipeline integrating single-cell RNA sequencing, CRISPR reverse genetics, and high-content imaging. Our findings unveil various genes, including those encoding opioid receptors, as possible regulators of ENS establishment. In addition, we present evidence that suggests opioid receptor involvement in the neurochemical coding of the larval ENS. In summary, our work presents a novel, efficient CRISPR screen targeting ENS development, facilitating the discovery of previously unknown genes, and increasing knowledge of nervous system construction.


Subject(s)
CRISPR-Cas Systems , Enteric Nervous System , Zebrafish , Animals , Enteric Nervous System/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Neural Crest/metabolism , Hirschsprung Disease/genetics
5.
Cells ; 13(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38786042

ABSTRACT

This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.


Subject(s)
Coculture Techniques , Enteric Nervous System , Gastrointestinal Diseases , Organoids , Humans , Coculture Techniques/methods , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/therapy , Animals , Models, Biological , Neurons/metabolism , Intestines
6.
J Clin Invest ; 134(9)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38690732

ABSTRACT

Epigenetic regulatory mechanisms are underappreciated, yet are critical for enteric nervous system (ENS) development and maintenance. We discovered that fetal loss of the epigenetic regulator Bap1 in the ENS lineage caused severe postnatal bowel dysfunction and early death in Tyrosinase-Cre Bap1fl/fl mice. Bap1-depleted ENS appeared normal in neonates; however, by P15, Bap1-deficient enteric neurons were largely absent from the small and large intestine of Tyrosinase-Cre Bap1fl/fl mice. Bowel motility became markedly abnormal with disproportionate loss of cholinergic neurons. Single-cell RNA sequencing at P5 showed that fetal Bap1 loss in Tyrosinase-Cre Bap1fl/fl mice markedly altered the composition and relative proportions of enteric neuron subtypes. In contrast, postnatal deletion of Bap1 did not cause enteric neuron loss or impaired bowel motility. These findings suggest that BAP1 is critical for postnatal enteric neuron differentiation and for early enteric neuron survival, a finding that may be relevant to the recently described human BAP1-associated neurodevelopmental disorder.


Subject(s)
Cell Differentiation , Enteric Nervous System , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Animals , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Mice , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice, Knockout , Female , Gastrointestinal Motility/genetics , Humans
7.
Biosens Bioelectron ; 258: 116298, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38701537

ABSTRACT

Wireless activation of the enteric nervous system (ENS) in freely moving animals with implantable optogenetic devices offers a unique and exciting opportunity to selectively control gastrointestinal (GI) transit in vivo, including the gut-brain axis. Programmed delivery of light to targeted locations in the GI-tract, however, poses many challenges not encountered within the central nervous system (CNS). We report here the development of a fully implantable, battery-free wireless device specifically designed for optogenetic control of the GI-tract, capable of generating sufficient light over large areas to robustly activate the ENS, potently inducing colonic motility ex vivo and increased propulsion in vivo. Use in in vivo studies reveals unique stimulation patterns that increase expulsion of colonic content, likely mediated in part by activation of an extrinsic brain-gut motor pathway, via pelvic nerves. This technology overcomes major limitations of conventional wireless optogenetic hardware designed for the CNS, providing targeted control of specific neurochemical classes of neurons in the ENS and brain-gut axis, for direct modulation of GI-transit and associated behaviours in freely moving animals.


Subject(s)
Enteric Nervous System , Optogenetics , Wireless Technology , Animals , Optogenetics/instrumentation , Enteric Nervous System/physiology , Mice , Wireless Technology/instrumentation , Brain-Gut Axis/physiology , Biosensing Techniques/instrumentation , Equipment Design , Brain/physiology , Mice, Inbred C57BL
8.
Nat Commun ; 15(1): 4400, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782898

ABSTRACT

Digestive Chagas disease (DCD) is an enteric neuropathy caused by Trypanosoma cruzi infection. There is a lack of evidence on the mechanism of pathogenesis and rationales for treatment. We used a female C3H/HeN mouse model that recapitulates key clinical manifestations to study how infection dynamics shape DCD pathology and the impact of treatment with the front-line, anti-parasitic drug benznidazole. Curative treatment 6 weeks post-infection resulted in sustained recovery of gastrointestinal transit function, whereas treatment failure led to infection relapse and gradual return of DCD symptoms. Neuro/immune gene expression patterns shifted from chronic inflammation to a tissue repair profile after cure, accompanied by increased cellular proliferation, glial cell marker expression and recovery of neuronal density in the myenteric plexus. Delaying treatment until 24 weeks post-infection led to partial reversal of DCD, suggesting the accumulation of permanent tissue damage over the course of chronic infection. Our study shows that murine DCD pathogenesis is sustained by chronic T. cruzi infection and is not an inevitable consequence of acute stage denervation. The risk of irreversible enteric neuromuscular tissue damage and dysfunction developing highlights the importance of prompt diagnosis and treatment. These findings support the concept of treating asymptomatic, T. cruzi-infected individuals with benznidazole to prevent DCD development.


Subject(s)
Chagas Disease , Disease Models, Animal , Enteric Nervous System , Mice, Inbred C3H , Nitroimidazoles , Trypanocidal Agents , Trypanosoma cruzi , Animals , Chagas Disease/drug therapy , Chagas Disease/parasitology , Female , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Nitroimidazoles/pharmacology , Nitroimidazoles/therapeutic use , Trypanosoma cruzi/drug effects , Mice , Enteric Nervous System/drug effects , Nerve Regeneration/drug effects
9.
Eur J Histochem ; 68(1)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38568200

ABSTRACT

The paper presents a summary of immunohistochemical (IHC) and biochemical investigations on the presence of galanin (Gal), one of the neuropeptides abundant in the enteric nervous systems, and three types of its receptors (GalR1-3) in colorectal cancer (CRC) tissue and non-involved colon wall and their associations with clinical-pathological data of the CRC patients. We were the first to morphologically demonstrate the presence of endogenous Gal in CRC sections and measure its content in homogenates of tumor tissue and dissected compartments of unchanged colon wall. The prominent atrophy of myenteric plexuses displaying Gal immunoreactivity (Gal-Ir) located close to the tumor invasion was found to be accompanied by higher Gal content in the tumor-adjacent muscularis externa than in tumor-distant tissue. In further studies for the first time, we demonstrated by the IHC technique the presence of the GalR1-3 receptors in the CRC tumors and the colon mucosa and found that higher GalR3-Ir in the tumor tissue correlated with longer overall survival of CRC patients. Furthermore, we discovered that lower GalR1 expression in submucosal plexuses located near the tumor correlated with a better prognosis in patients with CRC. These findings suggest that GalR1 could be considered as a novel therapeutic target in CRC. In conclusion, our morphological investigations provided novel data documenting the involvement of Gal and its receptors in the progression of CRC and showed the usefulness of the IHC technique for the prognosis of CRC patients.


Subject(s)
Colorectal Neoplasms , Enteric Nervous System , Humans , Galanin , Prognosis , Colorectal Neoplasms/diagnosis
10.
Biochem Biophys Res Commun ; 710: 149861, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38581949

ABSTRACT

During early development, the enteric nervous system forms from the migration of enteric neural crest cells (ENCCs) from the foregut to the hindgut, where they undergo proliferation and differentiation facilitated by interactions with enteric mesenchymal cells (EMCs). This study investigates the impact on ENCC migration of EMC-ENCC communication mediated by GFRA1b expressed in EMCs. GFRA1-expressing cells in day 11-12 (E11-12) mouse embryos differentiated into smooth muscle cells from E12 onwards. Observations at E12-13.5 revealed high levels of GFRA1 expression on the anti-mesenteric side of the hindgut, correlating with enhanced ENCC migration. This indicates that GFRA1 in EMCs plays a role in ENCC migration during development. Examining GFRA1 isoforms, we found high levels of GFRA1b, which lacks amino acids 140-144, in EMCs. To assess the impact of GFRA1 isoforms on EMC-ENCC communication, we conducted neurosphere drop assays. This revealed that GFRA1b-expressing cells promoted GDNF-dependent extension and increased neurite density in ENCC neurospheres. Co-culture of ENCC mimetic cells expressing RET and GFRA1a with EMC mimetic cells expressing GFRA1a, GFRA1b, or vector alone showed that only GFRA1b-expressing co-cultured cells sustained RET phosphorylation in ENCC-mimetic cells for over 120 min upon GDNF stimulation. Our study provides evidence that GFRA1b-mediated cell-to-cell communication plays a critical role in ENCC motility in enteric nervous system development. These findings contribute to understanding the cellular interactions and signaling mechanisms that underlie enteric nervous system formation and highlight potential therapeutic targets for gastrointestinal motility disorders.


Subject(s)
Enteric Nervous System , Neural Crest , Animals , Mice , Cell Differentiation/physiology , Cell Movement/physiology , Enteric Nervous System/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neural Crest/metabolism , Protein Isoforms/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G712-G725, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38626403

ABSTRACT

Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Tract , Humans , Animals , Gastrointestinal Tract/immunology , Gastrointestinal Tract/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Microbiome/physiology , Neuroimmunomodulation/physiology , Enteric Nervous System/physiology , Enteric Nervous System/immunology
12.
Nat Metab ; 6(5): 837-846, 2024 May.
Article in English | MEDLINE | ID: mdl-38570627

ABSTRACT

Sodium is essential for all living organisms1. Animals including insects and mammals detect sodium primarily through peripheral taste cells2-7. It is not known, however, whether animals can detect this essential micronutrient independently of the taste system. Here, we report that Drosophila Ir76b mutants that were unable to detect sodium2 became capable of responding to sodium following a period of salt deprivation. From a screen for cells required for the deprivation-induced sodium preference, we identified a population of anterior enteric neurons, which we named internal sodium-sensing (INSO) neurons, that are essential for directing a behavioural preference for sodium. Enteric INSO neurons innervate the gut epithelia mainly through their dendritic processes and send their axonal projections along the oesophagus to the brain and to the crop duct. Through calcium imaging and CaLexA experiments, we found that INSO neurons respond immediately and specifically to sodium ions. Notably, the sodium-evoked responses were observed only after a period of sodium deprivation. Taken together, we have identified a taste-independent sodium sensor that is essential for the maintenance of sodium homeostasis.


Subject(s)
Drosophila Proteins , Neurons , Sodium , Animals , Sodium/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Neurons/metabolism , Postprandial Period , Drosophila melanogaster , Enteric Nervous System/metabolism , Taste/physiology , Mutation , Drosophila , Sodium Channels , Receptors, Ionotropic Glutamate
13.
Auton Neurosci ; 253: 103176, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669866

ABSTRACT

Tributyltin (TBT) is a biocide used in the formulation of antifouling paints and it is highly harmful. Despite the ban, the compound persists in the environment, contaminating marine foodstuffs and household products. Therefore, considering the route of exposure to the contaminant, the gastrointestinal tract (GIT) acts as an important barrier against harmful substances and is a potential biomarker for understanding the consequences of these agents. This work aimed to evaluate histological and neuronal alterations in the duodenum of male Wistar rats that received 20 ng/g TBT and 600 ng/g via gavage for 30 consecutive days. After the experimental period, the animals were euthanized, and the duodenum was intended for neuronal histochemistry (total and metabolically active populations) and histological routine (morphometry and histopathology). The results showed more severe changes in neuronal density and intestinal morphometry in rats exposed to 20 ng/g, such as total neuronal density decrease and reduction of intestinal layers. In rats exposed to 600 ng/g of TBT, it was possible to observe only an increase in intraepithelial lymphocytes. We conclude that TBT can be more harmful to intestinal homeostasis when consumed in lower concentrations.


Subject(s)
Duodenum , Neuronal Plasticity , Rats, Wistar , Trialkyltin Compounds , Animals , Trialkyltin Compounds/toxicity , Male , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Duodenum/drug effects , Duodenum/pathology , Neurons/drug effects , Neurons/pathology , Rats , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Enteric Nervous System/drug effects , Enteric Nervous System/pathology
14.
J Smooth Muscle Res ; 60: 1-9, 2024.
Article in English | MEDLINE | ID: mdl-38462479

ABSTRACT

Macrophages are the originators of inflammatory compounds, phagocytic purifiers in their local environment, and wound healing protectors in oxidative environments. They are molded by the tissue milieu they inhabit, with gastrointestinal (GI) muscularis macrophages (MMs) being a prime example. MMs are located in the muscular layer of the GI tract and contribute to muscle repair and maintenance of GI motility. MMs are often in close proximity to the enteric nervous system, specifically near the enteric neurons and interstitial cells of Cajal (ICCs). Consequently, the anti-inflammatory function of MMs corresponds to the development and maintenance of neural networks in the GI tract. The capacity of MMs to shift from anti-inflammatory to proinflammatory states may contribute to the inflammatory aspects of various GI diseases and disorders such as diabetic gastroparesis or postoperative ileus, functional disorders such as irritable bowel syndrome, and organic diseases such as inflammatory bowel disease. We reviewed the current knowledge of MMs and their influence on neighboring cells due to their important role in the GI tract.


Subject(s)
Enteric Nervous System , Anti-Inflammatory Agents , Gastrointestinal Motility , Gastrointestinal Tract , Macrophages , Muscles , Humans
15.
Nat Commun ; 15(1): 2479, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509106

ABSTRACT

Neurointestinal diseases cause significant morbidity and effective treatments are lacking. This study aimes to test the feasibility of transplanting autologous enteric neural stem cells (ENSCs) to rescue the enteric nervous system (ENS) in a model of colonic aganglionosis. ENSCs are isolated from a segment of small intestine from Wnt1::Cre;R26iDTR mice in which focal colonic aganglionosis is simultaneously created by diphtheria toxin injection. Autologous ENSCs are isolated, expanded, labeled with lentiviral-GFP, and transplanted into the aganglionic segment in vivo. ENSCs differentiate into neurons and glia, cluster to form neo-ganglia, and restore colonic contractile activity as shown by electrical field stimulation and optogenetics. Using a non-lethal model of colonic aganglionosis, our results demonstrate the potential of autologous ENSC therapy to improve functional outcomes in neurointestinal disease, laying the groundwork for clinical application of this regenerative cell-based approach.


Subject(s)
Colorectal Neoplasms , Enteric Nervous System , Hirschsprung Disease , Neural Stem Cells , Mice , Animals , Hirschsprung Disease/therapy , Stem Cell Transplantation/methods , Neural Stem Cells/transplantation , Neurons
16.
Biomolecules ; 14(3)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38540765

ABSTRACT

Phosphatase and tensin homolog (Pten) is a key regulator of cell proliferation and a potential target to stimulate postnatal enteric neuro- and/or gliogenesis. To investigate this, we generated two tamoxifen-inducible Cre recombinase murine models in which Pten was conditionally ablated, (1) in glia (Plp1-expressing cells) and (2) in neurons (Calb2-expressing cells). Tamoxifen-treated adult (7-12 weeks of age; n = 4-15) mice were given DSS to induce colitis, EdU to monitor cell proliferation, and were evaluated at two timepoints: (1) early (3-4 days post-DSS) and (2) late (3-4 weeks post-DSS). We investigated gut motility and evaluated the enteric nervous system. Pten inhibition in Plp1-expressing cells elicited gliogenesis at baseline and post-DSS (early and late) in the colon, and neurogenesis post-DSS late in the proximal colon. They also exhibited an increased frequency of colonic migrating motor complexes (CMMC) and slower whole gut transit times. Pten inhibition in Calb2-expressing cells did not induce enteric neuro- or gliogenesis, and no alterations were detected in CMMC or whole gut transit times when compared to the control at baseline or post-DSS (early and late). Our results merit further research into Pten modulation where increased glia and/or slower intestinal transit times are desired (e.g., short-bowel syndrome and rapid-transit disorders).


Subject(s)
Enteric Nervous System , Animals , Mice , Enteric Nervous System/metabolism , Neurogenesis/physiology , Proteolipids/metabolism , Tamoxifen/pharmacology , Tensins/metabolism
17.
J Surg Res ; 297: 9-17, 2024 May.
Article in English | MEDLINE | ID: mdl-38428262

ABSTRACT

Postoperative ileus (POI) is caused by enteric neural dysfunction and inflammatory response to the stress of surgery as well as the effect of anesthetics and opioid pain medications. POI results in prolonged hospital stays, increased medical costs, and diminished enteral nutrition, rendering it a problem worth tackling. Many cellular pathways are implicated in this disease process, creating numerous opportunities for targeted management strategies. There is a gap in the literature in studies exploring neonatal POI pathophysiology and treatment options. It is well known that neonatal immune and enteric nervous systems are immature, and this results in gut physiology which is distinct from adults. Neonates undergoing abdominal surgery face similar surgical stressors and exposure to medications that cause POI in adults. In this review, we aim to summarize the existing adult and neonatal literature on POI pathophysiology and management and explore applications in the neonatal population.


Subject(s)
Enteric Nervous System , Ileus , Adult , Infant, Newborn , Humans , Postoperative Complications/etiology , Ileus/epidemiology , Analgesics, Opioid , Enteral Nutrition/adverse effects
18.
Gastroenterology ; 166(6): 1085-1099, 2024 06.
Article in English | MEDLINE | ID: mdl-38452824

ABSTRACT

BACKGROUND & AIMS: The enteric nervous system (ENS), the gut's intrinsic nervous system critical for gastrointestinal function and gut-brain communication, is believed to mainly originate from vagal neural crest cells (vNCCs) and partially from sacral NCCs (sNCCs). Resolving the exact origins of the ENS is critical for understanding congenital ENS diseases but has been confounded by the inability to distinguish between both NCC populations in situ. Here, we aimed to resolve the exact origins of the mammalian ENS. METHODS: We genetically engineered mouse embryos facilitating comparative lineage-tracing of either all (pan-) NCCs including vNCCs or caudal trunk and sNCCs (s/tNCCs) excluding vNCCs. This was combined with dual-lineage tracing and 3-dimensional reconstruction of pelvic plexus and hindgut to precisely pinpoint sNCC and vNCC contributions. We further used coculture assays to determine the specificity of cell migration from different neural tissues into the hindgut. RESULTS: Both pan-NCCs and s/tNCCs contributed to established NCC derivatives but only pan-NCCs contributed to the ENS. Dual-lineage tracing combined with 3-dimensional reconstruction revealed that s/tNCCs settle in complex patterns in pelvic plexus and hindgut-surrounding tissues, explaining previous confusion regarding their contributions. Coculture experiments revealed unspecific cell migration from autonomic, sensory, and neural tube explants into the hindgut. Lineage tracing of ENS precursors lastly provided complimentary evidence for an exclusive vNCC origin of the murine ENS. CONCLUSIONS: sNCCs do not contribute to the murine ENS, suggesting that the mammalian ENS exclusively originates from vNCCs. These results have immediate implications for comprehending (and devising treatments for) congenital ENS disorders, including Hirschsprung's disease.


Subject(s)
Cell Lineage , Cell Movement , Enteric Nervous System , Neural Crest , Animals , Neural Crest/cytology , Neural Crest/embryology , Enteric Nervous System/embryology , Mice , Coculture Techniques , Mice, Transgenic , Vagus Nerve/embryology , Sacrum/innervation
19.
Cell Rep ; 43(4): 113953, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38517896

ABSTRACT

The gastrointestinal (GI) tract is innervated by intrinsic neurons of the enteric nervous system (ENS) and extrinsic neurons of the central nervous system and peripheral ganglia. The GI tract also harbors a diverse microbiome, but interactions between the ENS and the microbiome remain poorly understood. Here, we activate choline acetyltransferase (ChAT)-expressing or tyrosine hydroxylase (TH)-expressing gut-associated neurons in mice to determine effects on intestinal microbial communities and their metabolites as well as on host physiology. The resulting multi-omics datasets support broad roles for discrete peripheral neuronal subtypes in shaping microbiome structure, including modulating bile acid profiles and fungal colonization. Physiologically, activation of either ChAT+ or TH+ neurons increases fecal output, while only ChAT+ activation results in increased colonic contractility and diarrhea-like fluid secretion. These findings suggest that specific subsets of peripherally activated neurons differentially regulate the gut microbiome and GI physiology in mice without involvement of signals from the brain.


Subject(s)
Gastrointestinal Microbiome , Neurons , Animals , Gastrointestinal Microbiome/physiology , Mice , Neurons/metabolism , Choline O-Acetyltransferase/metabolism , Enteric Nervous System/physiology , Mice, Inbred C57BL , Tyrosine 3-Monooxygenase/metabolism , Male , Gastrointestinal Tract/microbiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...