Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Trends Immunol ; 41(5): 359-362, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32249062

RESUMEN

There is a major gap in our understanding of how the intestinal immune and nervous systems are integrated to regulate protective adaptations to enteric infections while maintaining tissue homeostasis. Three recent complementary reports published in Cell (2020) provide new mechanistic insights into how this enteric neuro-immune crosstalk may occur.


Asunto(s)
Homeostasis , Enfermedades Intestinales , Intestinos , Sistema Nervioso , Animales , Homeostasis/inmunología , Humanos , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/microbiología , Intestinos/inmunología , Sistema Nervioso/inmunología
2.
Perfusion ; : 2676591231202719, 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37772723

RESUMEN

INTRODUCTION: del Nido cardioplegia was developed for immature myocardium to prevent myocardial damage by Ca+2 in traditional blood cardioplegia. But due to increased hemodilution and decreased colloid oncotic pressure it may cause myocardial edema and increased cardiac morbidity. Microplegia may have better cardioprotection in comparison to del Nido as there is less hemodilution. MATERIAL AND METHODS: 60 patients from the age group of 1 to 14 years were divided into two groups i.e. del Nido based microplegia group and conventional del Nido group for studying two different cardioplegia technique. Data were collected and compared for intraoperative Hb, CPK-MB and Trop-I levels changes and requirement for defibrillation in intraoperative period. Demographic data, CPB time and ACC time were also collected. RESULTS: Marked elevation in CPK-MB and Trop-I levels were seen in both groups. Statistically significant difference was seen in CPK-MB levels after 6 h of surgery where del Nido group has higher value in comparison to microplegia group. No statistical difference was seen in Trop-I levels in both groups. Strength of correlation (r) was also stronger for CPK-MB rise in association with CPB time and ACC time, in del Nido group but not for Trop-I. Significantly higher hemodilution was also seen in del Nido group after delivering cardioplegia. None of the patients required defibrillation in any group. CONCLUSION: Lesser hemodilution was seen in microplegia group. Significant cardioprotection is associated with use of microplegia solution in pediatric age group.

3.
Gastroenterology ; 159(1): 200-213.e8, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32234538

RESUMEN

BACKGROUND & AIMS: The enteric nervous system (ENS) exists in close proximity to luminal bacteria. Intestinal microbes regulate ENS development, but little is known about their effects on adult enteric neurons. We investigated whether intestinal bacteria or their products affect the adult ENS via toll-like receptors (TLRs) in mice. METHODS: We performed studies with conventional C57/BL6, germ-free C57/BL6, Nestin-creERT2:tdTomato, Nestin-GFP, and ChAT-cre:tdTomato. Mice were given drinking water with ampicillin or without (controls). Germ-free mice were given drinking water with TLR2 agonist or without (controls). Some mice were given a blocking antibody against TLR2 or a TLR4 inhibitor. We performed whole gut transit, bead latency, and geometric center studies. Feces were collected and analyzed by 16S ribosomal RNA gene sequencing. Longitudinal muscle myenteric plexus (LMMP) tissues were collected, analyzed by immunohistochemistry, and levels of nitric oxide were measured. Cells were isolated from colonic LMMP of Nestin-creERT2:tdTomato mice and incubated with agonists of TLR2 (receptor for gram-positive bacteria), TLR4 (receptor for gram-negative bacteria), or distilled water (control) and analyzed by flow cytometry. RESULTS: Stool from mice given ampicillin had altered composition of gut microbiota with reduced abundance of gram-positive bacteria and increased abundance of gram-negative bacteria, compared with mice given only water. Mice given ampicillin had reduced colon motility compared with mice given only water, and their colonic LMMP had reduced numbers of nitrergic neurons, reduced neuronal nitric oxide synthase production, and reduced colonic neurogenesis. Numbers of colonic myenteric neurons increased after mice were switched from ampicillin to plain water, with increased markers of neurogenesis. Nestin-positive enteric neural precursor cells expressed TLR2 and TLR4. In cells isolated from the colonic LMMP, incubation with the TLR2 agonist increased the percentage of neurons originating from enteric neural precursor cells to approximately 10%, compared with approximately 0.01% in cells incubated with the TLR4 agonist or distilled water. Mice given an antibody against TLR2 had prolonged whole gut transit times; their colonic LMMP had reduced total neurons and a smaller proportion of nitrergic neurons per ganglion, and reduced markers of neurogenesis compared with mice given saline. Colonic LMMP of mice given the TLR4 inhibitor did not have reduced markers of neurogenesis. Colonic LMMP of germ-free mice given TLR2 agonist had increased neuronal numbers compared with control germ-free mice. CONCLUSIONS: In the adult mouse colon, TLR2 promotes colonic neurogenesis, regulated by intestinal bacteria. Our findings indicate that colonic microbiota help maintain the adult ENS via a specific signaling pathway. Pharmacologic and probiotic approaches directed towards specific TLR2 signaling processes might be developed for treatment of colonic motility disorders related to use of antibiotics or other factors.


Asunto(s)
Disbiosis/fisiopatología , Sistema Nervioso Entérico/fisiología , Microbioma Gastrointestinal/fisiología , Neurogénesis/fisiología , Receptor Toll-Like 2/metabolismo , Adulto , Ampicilina/administración & dosificación , Ampicilina/efectos adversos , Animales , Células Cultivadas , Colon/inervación , Colon/microbiología , Colon/fisiología , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Ratones Transgénicos , Plexo Mientérico/citología , Plexo Mientérico/fisiología , Nestina/genética , Neurogénesis/efectos de los fármacos , Neuronas Nitrérgicas/fisiología , Óxido Nítrico/metabolismo , Cultivo Primario de Células , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo
4.
Am J Physiol Cell Physiol ; 319(2): C321-C330, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32551856

RESUMEN

Acetylcholine induces robust electrogenic anion secretion in mammalian intestine and it has long been hypothesized that it mediates the epithelial response through the M3 and, to a lesser extent, the M1 muscarinic receptors in the mouse. However, nicotinic receptors have recently been identified in intestinal enterocytes by quantitative real-time (qRT)-PCR/RNAseq, although any direct influence on intestinal transport has not been identified. We tested the hypothesis that cholinergic-induced anion secretion in the intestine is a result of both muscarinic and nicotinic pathways that are intrinsic to the intestinal epithelia. We developed a method to generate mouse jejunal enteroid monolayers which were used to measure active electrogenic anion secretion by the Ussing chamber/voltage-clamp technique. Here, we show that the cholinergic agonist carbachol (CCh) and the muscarinic agonist bethanechol (BCh) stimulate short-lived, concentration-dependent anion secretion in the epithelial cell-only enteroid monolayers. The muscarinic antagonist atropine completely inhibited CCh- and BCh-induced secretion, while the nicotinic antagonist hexamethonium reduced the CCh response by ~45%. While nicotine alone did not alter anion secretion, it increased the BCh-induced increase in short-circuit current in a concentration-dependent manner; this synergy was prevented by pretreatment with hexamethonium. In addition to being sensitive to hexamethonium, monolayers express both classes of cholinergic receptor by qRT-PCR, including 13 of 16 nicotinic receptor subunits. Our findings indicate that an interaction between muscarinic and nicotinic agonists synergistically stimulates anion secretion in mouse jejunal epithelial cells and identify a role for epithelial nicotinic receptors in anion secretion.


Asunto(s)
Agonistas Muscarínicos/farmacología , Sistema Colinérgico no Neuronal/genética , Receptores Muscarínicos/genética , Receptores Nicotínicos/genética , Acetilcolina/farmacología , Animales , Aniones/metabolismo , Atropina/farmacología , Agonistas Colinérgicos/farmacología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Hexametonio/farmacología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Ratones , Sistema Colinérgico no Neuronal/efectos de los fármacos , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(18): E3709-E3718, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28420791

RESUMEN

According to current dogma, there is little or no ongoing neurogenesis in the fully developed adult enteric nervous system. This lack of neurogenesis leaves unanswered the question of how enteric neuronal populations are maintained in adult guts, given previous reports of ongoing neuronal death. Here, we confirm that despite ongoing neuronal cell loss because of apoptosis in the myenteric ganglia of the adult small intestine, total myenteric neuronal numbers remain constant. This observed neuronal homeostasis is maintained by new neurons formed in vivo from dividing precursor cells that are located within myenteric ganglia and express both Nestin and p75NTR, but not the pan-glial marker Sox10. Mutation of the phosphatase and tensin homolog gene in this pool of adult precursors leads to an increase in enteric neuronal number, resulting in ganglioneuromatosis, modeling the corresponding disorder in humans. Taken together, our results show significant turnover and neurogenesis of adult enteric neurons and provide a paradigm for understanding the enteric nervous system in health and disease.


Asunto(s)
Apoptosis , Sistema Nervioso Entérico/metabolismo , Nestina/metabolismo , Neurogénesis , Receptores de Factor de Crecimiento Nervioso/metabolismo , Factores de Transcripción SOXE/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos , Nestina/genética , Receptores de Factor de Crecimiento Nervioso/genética , Factores de Transcripción SOXE/genética
6.
J Neurosci ; 38(44): 9346-9354, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30381426

RESUMEN

The enteric nervous system (ENS) is a large, complex division of the peripheral nervous system that regulates many digestive, immune, hormonal, and metabolic functions. Recent advances have elucidated the dynamic nature of the mature ENS, as well as the complex, bidirectional interactions among enteric neurons, glia, and the many other cell types that are important for mediating gut behaviors. Here, we provide an overview of ENS development and maintenance, and focus on the latest insights gained from the use of novel model systems and live-imaging techniques. We discuss major advances in the understanding of enteric glia, and the functional interactions among enteric neurons, glia, and enteroendocrine cells, a large class of sensory epithelial cells. We conclude by highlighting recent work on muscularis macrophages, a group of immune cells that closely interact with the ENS in the gut wall, and the importance of neurological-immune system communication in digestive health and disease.


Asunto(s)
Encéfalo/metabolismo , Sistema Nervioso Entérico/metabolismo , Enfermedades Gastrointestinales/metabolismo , Tracto Gastrointestinal/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/patología , Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/patología , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Humanos , Neurobiología
7.
Gut ; 67(5): 827-836, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28228489

RESUMEN

OBJECTIVE: The enteric nervous system (ENS) undergoes neuronal loss and degenerative changes with age. The cause of this neurodegeneration is poorly understood. Muscularis macrophages residing in close proximity to enteric ganglia maintain neuromuscular function via direct crosstalk with enteric neurons and have been implicated in the pathogenesis of GI motility disorders like gastroparesis and postoperative ileus. The aim of this study was to assess whether ageing causes alterations in macrophage phenotype that contributes to age-related degeneration of the ENS. DESIGN: Longitudinal muscle and myenteric plexus from small intestine of young, mid-aged and old mice were dissected and prepared for whole mount immunostaining, flow cytometry, Luminex immunoassays, western blot analysis, enteric neural stem cell (ENSC) isolation or conditioned media. Bone marrow derived macrophages were prepared and polarised to classic (M1) or alternative (M2) activation states. Markers for macrophage phenotype were measured using quantitative RT-PCR. RESULTS: Ageing causes a shift in macrophage polarisation from anti-inflammatory 'M2' to proinflammatory 'M1' that is associated with a rise in cytokines and immune cells in the ENS. This phenotypic shift is associated with a neural response to inflammatory signals, increase in apoptosis and loss of enteric neurons and ENSCs, and delayed intestinal transit. An age-dependent decrease in expression of the transcription factor FoxO3, a known longevity gene, contributes to the loss of anti-inflammatory behaviour in macrophages of old mice, and FoxO3-deficient mice demonstrate signs of premature ageing of the ENS. CONCLUSIONS: A shift by macrophages towards a proinflammatory phenotype with ageing causes inflammation-mediated degeneration of the ENS.


Asunto(s)
Envejecimiento/patología , Sistema Nervioso Entérico/patología , Macrófagos/metabolismo , Envejecimiento/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Células Cultivadas , Citocinas/metabolismo , Sistema Nervioso Entérico/metabolismo , Citometría de Flujo , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica , Inflamación/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
8.
J Physiol ; 600(19): 4251-4252, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35980322
10.
Proc Natl Acad Sci U S A ; 110(31): 12643-8, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858471

RESUMEN

Organs are composites of tissue types with diverse developmental origins, and they rely on distinct stem and progenitor cells to meet physiological demands for cellular production and homeostasis. How diverse stem cell activity is coordinated within organs is not well understood. Here we describe a lineage-restricted, self-renewing common skeletal progenitor (bone, cartilage, stromal progenitor; BCSP) isolated from limb bones and bone marrow tissue of fetal, neonatal, and adult mice. The BCSP clonally produces chondrocytes (cartilage-forming) and osteogenic (bone-forming) cells and at least three subsets of stromal cells that exhibit differential expression of cell surface markers, including CD105 (or endoglin), Thy1 [or CD90 (cluster of differentiation 90)], and 6C3 [ENPEP glutamyl aminopeptidase (aminopeptidase A)]. These three stromal subsets exhibit differential capacities to support hematopoietic (blood-forming) stem and progenitor cells. Although the 6C3-expressing subset demonstrates functional stem cell niche activity by maintaining primitive hematopoietic stem cell (HSC) renewal in vitro, the other stromal populations promote HSC differentiation to more committed lines of hematopoiesis, such as the B-cell lineage. Gene expression analysis and microscopic studies further reveal a microenvironment in which CD105-, Thy1-, and 6C3-expressing marrow stroma collaborate to provide cytokine signaling to HSCs and more committed hematopoietic progenitors. As a result, within the context of bone as a blood-forming organ, the BCSP plays a critical role in supporting hematopoiesis through its generation of diverse osteogenic and hematopoietic-promoting stroma, including HSC supportive 6C3(+) niche cells.


Asunto(s)
Huesos/metabolismo , Cartílago/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal/fisiología , Nicho de Células Madre/fisiología , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Huesos/citología , Cartílago/citología , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/citología , Ratones , Ratones Transgénicos , Células del Estroma/citología , Células del Estroma/metabolismo
11.
Arthritis Rheumatol ; 76(1): 92-99, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37530745

RESUMEN

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.


Asunto(s)
Autoanticuerpos , Proteínas de la Membrana , Esclerodermia Sistémica , Proteínas de la Membrana/metabolismo , Autoantígenos/metabolismo , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/fisiopatología , Autoanticuerpos/análisis , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiopatología , Humanos , Animales , Ratones , Neuronas/metabolismo , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/fisiopatología
12.
Gastroenterology ; 142(5): 1195-1205.e6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22333952

RESUMEN

BACKGROUND & AIMS: Paneth cells contribute to the small intestinal niche of Lgr5(+) stem cells. Although the colon also contains Lgr5(+) stem cells, it does not contain Paneth cells. We investigated the existence of colonic Paneth-like cells that have a distinct transcriptional signature and support Lgr5(+) stem cells. METHODS: We used multicolor fluorescence-activated cell sorting to isolate different subregions of colon crypts, based on known markers, from dissociated colonic epithelium of mice. We performed multiplexed single-cell gene expression analysis with quantitative reverse transcriptase polymerase chain reaction followed by hierarchical clustering analysis to characterize distinct cell types. We used immunostaining and fluorescence-activated cell sorting analyses with in vivo administration of a Notch inhibitor and in vitro organoid cultures to characterize different cell types. RESULTS: Multicolor fluorescence-activated cell sorting could isolate distinct regions of colonic crypts. Four major epithelial subtypes or transcriptional states were revealed by gene expression analysis of selected populations of single cells. One of these, the goblet cells, contained a distinct cKit/CD117(+) crypt base subpopulation that expressed Dll1, Dll4, and epidermal growth factor, similar to Paneth cells, which were also marked by cKit. In the colon, cKit(+) goblet cells were interdigitated with Lgr5(+) stem cells. In vivo, this colonic cKit(+) population was regulated by Notch signaling; administration of a γ-secretase inhibitor to mice increased the number of cKit(+) cells. When isolated from mouse colon, cKit(+) cells promoted formation of organoids from Lgr5(+) stem cells, which expressed Kitl/stem cell factor, the ligand for cKit. When organoids were depleted of cKit(+) cells using a toxin-conjugated antibody, organoid formation decreased. CONCLUSIONS: cKit marks small intestinal Paneth cells and a subset of colonic goblet cells that are regulated by Notch signaling and support Lgr5(+) stem cells.


Asunto(s)
Colon/citología , Células de Paneth/química , Células de Paneth/fisiología , Proteínas Proto-Oncogénicas c-kit/análisis , Receptores Acoplados a Proteínas G/análisis , Células Madre/fisiología , Animales , Antígenos CD/análisis , Moléculas de Adhesión Celular/análisis , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Células Caliciformes/fisiología , Receptores de Hialuranos/análisis , Ratones , Ratones Endogámicos C57BL , Receptores Notch/fisiología , Análisis de la Célula Individual , Células Madre/química
13.
Gut ; 61(4): 613-21, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21816959

RESUMEN

Current advances in our understanding of stem and precursor cell biology and in the protocols of stem cell isolation and transplantation have opened up the possibility of transplanting neural stem cells for the treatment of gastrointestinal motility disorders. This review summarises the current status of research in this field, identifies the major gaps in our knowledge and discusses the potential opportunities and hurdles for clinical application.


Asunto(s)
Enfermedades Gastrointestinales/terapia , Enfermedades Neurodegenerativas/terapia , Trasplante de Células Madre/métodos , Células Madre Embrionarias/trasplante , Motilidad Gastrointestinal/fisiología , Humanos , Células-Madre Neurales/trasplante , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre/tendencias
14.
Nat Rev Rheumatol ; 19(3): 166-181, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36747090

RESUMEN

Nearly all patients with systemic sclerosis (SSc) are negatively affected by dysfunction in the gastrointestinal tract, and the severity of gastrointestinal disease in SSc correlates with high mortality. The clinical complications of this dysfunction are heterogeneous and include gastro-oesophageal reflux disease, gastroparesis, small intestinal bacterial overgrowth, intestinal pseudo-obstruction, malabsorption and the requirement for total parenteral nutrition. The abnormal gastrointestinal physiology that promotes the clinical manifestations of SSc gastrointestinal disease throughout the gastrointestinal tract are diverse and present a range of therapeutic targets. Furthermore, the armamentarium of medications and non-pharmacological interventions that can benefit affected patients has substantially expanded in the past 10 years, and research is increasingly focused in this area. Here, we review the details of the gastrointestinal complications in SSc, tie physiological abnormalities to clinical manifestations, detail the roles of standard and novel therapies and lay a foundation for future investigative work.


Asunto(s)
Enfermedades Gastrointestinales , Esclerodermia Sistémica , Humanos , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/terapia , Esclerodermia Sistémica/complicaciones , Factores de Riesgo
15.
Elife ; 122023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38108810

RESUMEN

The enteric nervous system (ENS), a collection of neural cells contained in the wall of the gut, is of fundamental importance to gastrointestinal and systemic health. According to the prevailing paradigm, the ENS arises from progenitor cells migrating from the neural crest and remains largely unchanged thereafter. Here, we show that the lineage composition of maturing ENS changes with time, with a decline in the canonical lineage of neural-crest derived neurons and their replacement by a newly identified lineage of mesoderm-derived neurons. Single cell transcriptomics and immunochemical approaches establish a distinct expression profile of mesoderm-derived neurons. The dynamic balance between the proportions of neurons from these two different lineages in the post-natal gut is dependent on the availability of their respective trophic signals, GDNF-RET and HGF-MET. With increasing age, the mesoderm-derived neurons become the dominant form of neurons in the ENS, a change associated with significant functional effects on intestinal motility which can be reversed by GDNF supplementation. Transcriptomic analyses of human gut tissues show reduced GDNF-RET signaling in patients with intestinal dysmotility which is associated with reduction in neural crest-derived neuronal markers and concomitant increase in transcriptional patterns specific to mesoderm-derived neurons. Normal intestinal function in the adult gastrointestinal tract therefore appears to require an optimal balance between these two distinct lineages within the ENS.


Asunto(s)
Sistema Nervioso Entérico , Factor Neurotrófico Derivado de la Línea Celular Glial , Adulto , Humanos , Motilidad Gastrointestinal , Perfilación de la Expresión Génica , Mesodermo
16.
Am J Physiol Gastrointest Liver Physiol ; 302(9): G958-65, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22361728

RESUMEN

Enteric neural stem cells (ENSCs) are a population of neural crest-derived multipotent stem cells present in postnatal gut that may play an important role in regeneration of the enteric nervous system. In most studies, these cells have been isolated from the layer of the gut containing the myenteric plexus. However, a recent report demonstrated that neurosphere-like bodies (NLBs) containing ENSCs could be isolated from mucosal biopsy specimens from children, suggesting that ENSCs are present in multiple layers of the gut. The aim of our study was to assess whether NLBs isolated from layers of gut containing either myenteric or submucosal plexus are equivalent. We divided the mouse small intestine into two layers, one containing myenteric plexus and the other submucosal plexus, and assessed for NLB formation. Differences in NLB density, proliferation, apoptosis, neural crest origin, and phenotype were investigated. NLBs isolated from the myenteric plexus layer were present at a higher density and demonstrated greater proliferation, lower apoptosis, and higher expression of nestin, p75, Sox10, and Ret than those from submucosal plexus. Additionally, they contained a higher percentage of neural crest-derived cells (99.4 ± 1.5 vs. 0.7 ± 1.19% of Wnt1-cre:tdTomato cells; P < 0.0001) and produced more neurons and glial cells than those from submucosal plexus. NLBs from the submucosal plexus layer expressed higher CD34 and produced more smooth muscle-like cells. NLBs from the myenteric plexus layer contain more neural crest-derived ENSCs while those from submucosal plexus appear more heterogeneous, likely containing a population of mesenchymal stem cells.


Asunto(s)
Intestino Delgado/citología , Células-Madre Neurales/citología , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL
17.
Arthritis Care Res (Hoboken) ; 74(3): 442-450, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33064934

RESUMEN

OBJECTIVE: Systemic sclerosis (SSc)-associated gastrointestinal (GI) complications are attributed to a variety of factors, including diet, microbiota dysbiosis, or GI transit abnormalities. Our objective was to examine the contribution of abnormal GI transit to SSc Medsger GI severity scores and/or University of California Los Angeles Scleroderma Clinical Trial Consortium Gastrointestinal Tract (UCLA GIT) 2.0 symptoms. METHODS: Patients with SSc and GI symptoms (n = 71) and healthy controls (n = 18) underwent whole gut transit (WGT) scintigraphy to assess transit from the esophagus to the colon. The presence of delayed transit and percent emptying in each GI region were measured. We compared the WGT measurements between categories of the Medsger GI severity score (0-4) and across UCLA GIT 2.0 domains and total score (0-3). RESULTS: A total of 88% of patients had >1 abnormal region of the gut on WGT scintigraphy. All patients requiring total parenteral nutrition had delayed small bowel transit, compared to only approximately 11% of patients in other Medsger GI severity groups (P ≤ 0.01). Severe colonic transit delays were more likely in patients with Medsger GI scores of 3 (pseudo-obstruction and/or malabsorption) compared to other Medsger GI groups (P = 0.02). Seventy-percent of these patients had ≤30% colonic emptying at 72 hours. Modest associations were noted between gastroesophageal reflux disease symptoms and delayed esophageal (r = -0.31, P = 0.05) and gastric emptying (r = -0.32, P = 0.05). CONCLUSION: These data are important in providing evidence that SSc bowel disease affects transit of GI content and that delay in transit accounts in part for both bowel symptoms and Medsger GI severity. Prospective studies examining the benefit of early therapeutic intervention targeting GI transit abnormalities in patients at high risk for severe GI complications are needed.


Asunto(s)
Enfermedades Gastrointestinales/etiología , Tránsito Gastrointestinal , Esclerodermia Sistémica/complicaciones , Adulto , Estudios de Casos y Controles , Femenino , Enfermedades Gastrointestinales/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Cintigrafía , Índice de Severidad de la Enfermedad
18.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G644-55, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21817062

RESUMEN

Recent studies have explored the potential of central nervous system-derived neural stem cells (CNS-NSC) to repopulate the enteric nervous system. However, the exact phenotypic fate of gut-transplanted CNS-NSC has not been characterized. The aim of this study was to investigate the effect of the gut microenvironment on phenotypic fate of CNS-NSC in vitro. With the use of Transwell culture, differentiation of mouse embryonic CNS-NSC was studied when cocultured without direct contact with mouse intestinal longitudinal muscle-myenteric plexus preparations (LM-MP) compared with control noncocultured cells, in a differentiating medium. Differentiated cells were analyzed by immunocytochemistry and quantitative RT-PCR to assess the expression of specific markers and by whole cell patch-clamp studies for functional characterization of their phenotype. We found that LM-MP cocultured cells had a significant increase in the numbers of cells that were immune reactive against the panneuronal marker ß-tubulin, neurotransmitters neuronal nitric oxide synthase (nNOS), choline acetyltransferase (ChAT), and neuropeptide vasoactive intestinal peptide (VIP) and showed an increase in expression of these genes, compared with control cells. Whole cell patch-clamp analysis showed that coculture with LM-MP decreases cell excitability and reduces voltage-gated Na(+) currents but significantly enhances A-current and late afterhyperpolarization (AHP) and increases the expression of the four AHP-generating Ca(2+)-dependent K(+) channel genes (KCNN), compared with control cells. In a separate experiment, differentiation of LM-MP cocultured CNS-NSC produced a significant increase in the numbers of cells that were immune reactive against the neurotransmitters nNOS, ChAT, and the neuropeptide VIP compared with CNS-NSC differentiated similarly in the presence of neonatal brain tissue. Our results show that the gut microenvironment induces CNS-NSC to produce neurons that share some of the characteristics of classical enteric neurons, further supporting the therapeutic use of these cells for gastrointestinal disorders.


Asunto(s)
Sistema Nervioso Entérico/crecimiento & desarrollo , Intestinos/fisiología , Células-Madre Neurales/fisiología , Potenciales de Acción/fisiología , Animales , Encéfalo/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Sistema Nervioso Entérico/fisiología , Femenino , Masculino , Ratones , Plexo Mientérico/fisiología , Neurogénesis/efectos de los fármacos , Neuronas/citología , Técnicas de Placa-Clamp
19.
J Clin Invest ; 131(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34196307

RESUMEN

Neurodegenerative disorders (NDs) affect essential functions not only in the CNS, but also cause persistent gut dysfunctions, suggesting that they have an impact on both CNS and gut-innervating neurons. Although the CNS biology of NDs continues to be well studied, how gut-innervating neurons, including those that connect the gut to the brain, are affected by or involved in the etiology of these debilitating and progressive disorders has been understudied. Studies in recent years have shown how CNS and gut biology, aided by the gut-brain connecting neurons, modulate each other's functions. These studies underscore the importance of exploring the gut-innervating and gut-brain connecting neurons of the CNS and gut function in health, as well as the etiology and progression of dysfunction in NDs. In this Review, we discuss our current understanding of how the various gut-innervating neurons and gut physiology are involved in the etiology of NDs, including Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, to cause progressive CNS and persistent gut dysfunction.


Asunto(s)
Sistema Nervioso Entérico/fisiopatología , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/fisiopatología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/fisiopatología , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Encéfalo/fisiopatología , Sistema Digestivo/inervación , Sistema Digestivo/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Disbiosis/fisiopatología , Microbioma Gastrointestinal/fisiología , Humanos , Enfermedad de Huntington/etiología , Enfermedad de Huntington/fisiopatología , Modelos Neurológicos , Mutación , Enfermedades Neurodegenerativas/microbiología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/fisiopatología
20.
Curr Opin Immunol ; 68: 64-71, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33130386

RESUMEN

The gastrointestinal (GI) tract performs a set of vital physiological functions related to food and water consumption. To help regulate these complex physiological processes, the GI tract is innervated by extensive neural networks. The GI tract also serves as the largest immune organ aimed to protect hosts from harmful microbes and toxins ingested with food. It emerges that the enteric nervous and immune systems are highly integrated to optimize digestion while reinforcing immune protection. In this review, we will discuss key cellular players involved in the neuro-immune interactions within the GI mucosa with the focus on the recently uncovered neural pathways that regulate mucosal immunity in a context relevant to GI health and disease.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Neuroinmunomodulación/inmunología , Animales , Tracto Gastrointestinal/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA