Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(13): 2823-2838.e20, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37236193

RESUMEN

Mental health profoundly impacts inflammatory responses in the body. This is particularly apparent in inflammatory bowel disease (IBD), in which psychological stress is associated with exacerbated disease flares. Here, we discover a critical role for the enteric nervous system (ENS) in mediating the aggravating effect of chronic stress on intestinal inflammation. We find that chronically elevated levels of glucocorticoids drive the generation of an inflammatory subset of enteric glia that promotes monocyte- and TNF-mediated inflammation via CSF1. Additionally, glucocorticoids cause transcriptional immaturity in enteric neurons, acetylcholine deficiency, and dysmotility via TGF-ß2. We verify the connection between the psychological state, intestinal inflammation, and dysmotility in three cohorts of IBD patients. Together, these findings offer a mechanistic explanation for the impact of the brain on peripheral inflammation, define the ENS as a relay between psychological stress and gut inflammation, and suggest that stress management could serve as a valuable component of IBD care.


Asunto(s)
Sistema Nervioso Entérico , Enfermedades Inflamatorias del Intestino , Humanos , Glucocorticoides/farmacología , Inflamación , Sistema Nervioso Entérico/fisiología , Estrés Psicológico
2.
Cell ; 175(5): 1198-1212.e12, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30293866

RESUMEN

Although chronic gastrointestinal dysmotility syndromes are a common worldwide health problem, underlying causes for these disorders are poorly understood. We show that flavivirus infection of enteric neurons leads to acute neuronal injury and cell death, inflammation, bowel dilation, and slowing of intestinal transit in mice. Flavivirus-primed CD8+ T cells promote these phenotypes, as their absence diminished enteric neuron injury and intestinal transit delays, and their adoptive transfer reestablished dysmotility after flavivirus infection. Remarkably, mice surviving acute flavivirus infection developed chronic gastrointestinal dysmotility that was exacerbated by immunization with an unrelated alphavirus vaccine or exposure to a non-infectious inflammatory stimulus. This model of chronic post-infectious gastrointestinal dysmotility in mice suggests that viral infections with tropism for enteric neurons and the ensuing immune response might contribute to the development of bowel motility disorders in humans. These results suggest an opportunity for unique approaches to diagnosis and therapy of gastrointestinal dysmotility syndromes.


Asunto(s)
Infecciones por Flavivirus/patología , Flavivirus/patogenicidad , Motilidad Gastrointestinal , Intestinos/patología , Animales , Linfocitos T CD8-positivos/inmunología , Flavivirus/genética , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Intestinos/virología , Leucocitos/citología , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Neuronas/ultraestructura , ARN Viral/aislamiento & purificación , ARN Viral/metabolismo , Síndrome
3.
Dev Biol ; 505: 42-57, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37827362

RESUMEN

BAZ1B is one of 25-27 coding genes deleted in canonical Williams syndrome, a multi-system disorder causing slow growth, vascular stenosis, and gastrointestinal complaints, including constipation. BAZ1B is involved in (among other processes) chromatin organization, DNA damage repair, and mitosis, suggesting reduced BAZ1B may contribute to Williams syndrome symptoms. In mice, loss of Baz1b causes early neonatal death. 89.6% of Baz1b-/- mice die within 24 h of birth without vascular anomalies or congenital heart disease (except for patent ductus arteriosus). Some (<50%) Baz1b-/- were noted to have prolonged neonatal cyanosis, patent ductus arteriosus, or reduced lung aeration, and none developed a milk spot. Meanwhile, 35.5% of Baz1b+/- mice die over the first three weeks after birth. Surviving Baz1b heterozygotes grow slowly (with variable severity). 66.7% of Baz1b+/- mice develop bowel dilation, compared to 37.8% of wild-type mice, but small bowel and colon transit studies were normal. Additionally, enteric neuron density appeared normal in Baz1b-/- mice except in distal colon myenteric plexus, where neuron density was modestly elevated. Combined with several rare phenotypes (agnathia, microphthalmia, bowel dilation) recovered, our work confirms the importance of BAZ1B in survival and growth and suggests that reduced copy number of BAZ1B may contribute to the variability in Williams syndrome phenotypes.


Asunto(s)
Conducto Arterioso Permeable , Síndrome de Williams , Animales , Ratones , Colon , Reparación del ADN , Neuronas , Síndrome de Williams/genética
4.
Gastroenterology ; 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38494035

RESUMEN

BACKGROUND & AIMS: Hirschsprung's disease is defined by the absence of the enteric nervous system (ENS) from the distal bowel. Primary treatment is "pull-through" surgery to remove bowel that lacks ENS, with reanastomosis of "normal" bowel near the anal verge. Problems after pull-through are common, and some may be due to retained hypoganglionic bowel (ie, low ENS density). Testing this hypothesis has been difficult because counting enteric neurons in tissue sections is unreliable, even for experts. Tissue clearing and 3-dimensional imaging provide better data about ENS structure than sectioning. METHODS: Regions from 11 human colons and 1 ileal specimen resected during Hirschsprung's disease pull-through surgery were cleared, stained with antibodies to visualize the ENS, and imaged by confocal microscopy. Control distal colon from people with no known bowel problems were similarly cleared, stained, and imaged. RESULTS: Quantitative analyses of human colon, ranging from 3 days to 60 years old, suggest age-dependent changes in the myenteric plexus area, ENS ganglion area, percentage of myenteric plexus occupied by ganglia, neurons/mm2, and neuron Feret's diameter. Neuron counting using 3-dimensional images was highly reproducible. High ENS density in neonatal colon allowed reliable neuron counts using 500-µm2 × 500-µm2 regions (36-fold smaller than in adults). Hirschsprung's samples varied 8-fold in proximal margin enteric neuron density and had diverse ENS architecture in resected bowel. CONCLUSIONS: Tissue clearing and 3-dimensional imaging provide more reliable information about ENS structure than tissue sections. ENS structure changes during childhood. Three-dimensional ENS anatomy may provide new insight into human bowel motility disorders, including Hirschsprung's disease.

5.
Proc Natl Acad Sci U S A ; 119(41): e2209150119, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36197995

RESUMEN

Actin is the most abundant protein in the cytoplasm of eukaryotic cells and interacts with hundreds of proteins to perform essential functions, including cell motility and cytokinesis. Numerous diseases are caused by mutations in actin, but studying the biochemistry of actin mutants is difficult without a reliable method to obtain recombinant actin. Moreover, biochemical studies have typically used tissue-purified α-actin, whereas humans express six isoforms that are nearly identical but perform specialized functions and are difficult to obtain in isolation from natural sources. Here, we describe a solution to the problem of actin expression and purification. We obtain high yields of actin isoforms in human Expi293F cells. Experiments along the multistep purification protocol demonstrate the removal of endogenous actin and the functional integrity of recombinant actin isoforms. Proteomics analysis of endogenous vs. recombinant actin isoforms confirms the presence of native posttranslational modifications, including N-terminal acetylation achieved after affinity-tag removal using the actin-specific enzyme Naa80. The method described facilitates studies of actin under fully native conditions to determine differences among isoforms and the effects of disease-causing mutations that occur in all six isoforms.


Asunto(s)
Actinas , Procesamiento Proteico-Postraduccional , Acetilación , Actinas/genética , Actinas/metabolismo , Movimiento Celular , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
6.
FASEB J ; 36 Suppl 12022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35723872

RESUMEN

BAZ1B is one of several genes deleted in Williams-Beuren Syndrome (WBS), a complex, multisystem genetic condition that occurs in ~1 in 8000 live births. Also known as Williams Syndrome Transcription Factor (WSTF), BAZ1B is thought to be essential for neural crest migration. To evaluate the impact of Baz1b loss of function, we evaluated the "knockout first" allele of Baz1btm2a(KOMP)Wtsi . Quantitative PCR revealed markedly reduced, but not absent, expression of Baz1b, suggesting that Baz1btm2a(KOMP)Wtsi mutants are knockdowns rather than knockouts. Homozygous Baz1btm2a(KOMP)Wtsi mutant mice die just hours after birth, and both homozygous mutants and heterozygotes are smaller than age-matched wildtype littermates. Survival analyses conducted on 388 Baz1btm2a(KOMP)Wtsi mice revealed that heterozygotes and homozygous mutants are approximately three and sixteen times more likely to die than wildtype mice, respectively [hazard ratio for death in Baz1b+/- : 3.04 (95% CI, 1.83-5.06), p<0.0001; hazard ratio for death in Baz1b-/- : 15.83 (95% CI, 8.54-29.37); p<0.0001]. Furthermore, a linear mixed effects model for the weights of wildtype and heterozygous mice over a 29-day period showed a significant difference in size based on genotype (mean: WT 7.97 g, Baz1b+/- 6.56 g, p<0.0001). Because neural crest lineages contribute to cardiac development, structure, and function, we hypothesized that early sudden death and failure to thrive in mutant mice may be at least partially attributable to cardiac abnormalities. To evaluate any morphologic and functional abnormalities, we performed microCT and echocardiography. MicroCT analysis of the hearts from P0 pups did not reveal congenital heart disease typical of neural crest defects (e.g. tetralogy of Fallot, truncus arteriosus, double outlet right ventricle, or interrupted aortic arch). Echocardiograms, performed at 1-month to align with the growth analysis timeline, revealed mildly decreased ejection fraction (EF, median: WT 64%, Baz1b+/- 56%, p<0.01) and fractional shortening (FS, median: WT 34%, Baz1b+/- 29%, p<0.01), increased left ventricular internal dimension at diastole (LViDd) normalized to animal size (median: WT 0.22 mm/g, Baz1b+/- 0.27 mm/g, p<0.05), and unchanged left ventricular posterior wall dimension at diastole (LVPWd) normalized to body size (median: WT 0.041 mm/g, Baz1b+/- 0.048 mm/g, p=0.19) in Baz1b+/- when compared to wildtype. However, Baz1b+/- LVPWd is significantly smaller than WT when body size is not considered (median: WT 0.63 mm, Baz1b+/- 0.62 mm, p<0.01), suggesting a relationship between cardiac function and mutant animal growth (all tests for genotype in n=14 WT and n=14 Baz1b+/- by Mann-Whitney U Test). Taken together, our data suggest that Baz1b+/- mice exhibit a dilated cardiomyopathy and that dosage for this gene may contribute to early death, decreased somatic growth, and cardiac abnormalities in Baz1b mutant mice. Additional analyses in older mice and with mutants generated using the conditional Baz1btm2a(KOMP)Wtsi allele will allow us to better explore the mechanisms of both the growth failure and cardiomyopathy phenotypes in this model.


Asunto(s)
Cardiomiopatía Dilatada , Cardiopatías Congénitas , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Corazón , Cardiopatías Congénitas/genética , Ratones , Cresta Neural/metabolismo , Fenotipo
7.
Dig Dis Sci ; 68(10): 3857-3871, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37650948

RESUMEN

Visceral myopathy is a rare, life-threatening disease linked to identified genetic mutations in 60% of cases. Mostly due to the dearth of knowledge regarding its pathogenesis, effective treatments are lacking. The disease is most commonly diagnosed in children with recurrent or persistent disabling episodes of functional intestinal obstruction, which can be life threatening, often requiring long-term parenteral or specialized enteral nutritional support. Although these interventions are undisputedly life-saving as they allow affected individuals to avoid malnutrition and related complications, they also seriously compromise their quality of life and can carry the risk of sepsis and thrombosis. Animal models for visceral myopathy, which could be crucial for advancing the scientific knowledge of this condition, are scarce. Clearly, a collaborative network is needed to develop research plans to clarify genotype-phenotype correlations and unravel molecular mechanisms to provide targeted therapeutic strategies. This paper represents a summary report of the first 'European Forum on Visceral Myopathy'. This forum was attended by an international interdisciplinary working group that met to better understand visceral myopathy and foster interaction among scientists actively involved in the field and clinicians who specialize in care of people with visceral myopathy.


Asunto(s)
Seudoobstrucción Intestinal , Desnutrición , Animales , Niño , Humanos , Calidad de Vida , Modelos Animales , Mutación , Enfermedades Raras
8.
Annu Rev Physiol ; 81: 235-259, 2019 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-30379617

RESUMEN

At the most fundamental level, the bowel facilitates absorption of small molecules, regulates fluid and electrolyte flux, and eliminates waste. To successfully coordinate this complex array of functions, the bowel relies on the enteric nervous system (ENS), an intricate network of more than 500 million neurons and supporting glia that are organized into distinct layers or plexi within the bowel wall. Neuron and glial diversity, as well as neurotransmitter and receptor expression in the ENS, resembles that of the central nervous system. The most carefully studied ENS functions include control of bowel motility, epithelial secretion, and blood flow, but the ENS also interacts with enteroendocrine cells, influences epithelial proliferation and repair, modulates the intestinal immune system, and mediates extrinsic nerve input. Here, we review the many different cell types that communicate with the ENS, integrating data about ENS function into a broader view of human health and disease. In particular, we focus on exciting new literature highlighting relationships between the ENS and its lesser-known interacting partners.


Asunto(s)
Encéfalo/fisiología , Sistema Nervioso Entérico/fisiología , Tracto Gastrointestinal/fisiología , Animales , Motilidad Gastrointestinal/fisiología , Humanos , Neuronas/fisiología
9.
Gastroenterology ; 160(4): 1208-1223.e4, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32980343

RESUMEN

BACKGROUND & AIMS: The colon is innervated by intrinsic and extrinsic neurons that coordinate functions necessary for digestive health. Sympathetic input suppresses colon motility by acting on intrinsic myenteric neurons, but the extent of sympathetic-induced changes on large-scale network activity in myenteric circuits has not been determined. Compounding the complexity of sympathetic function, there is evidence that sympathetic transmitters can regulate activity in non-neuronal cells (such as enteric glia and innate immune cells). METHODS: We performed anatomical tracing, immunohistochemistry, optogenetic (GCaMP calcium imaging, channelrhodopsin), and colon motility studies in mice and single-cell RNA sequencing in human colon to investigate how sympathetic postganglionic neurons modulate colon function. RESULTS: Individual neurons in each sympathetic prevertebral ganglion innervated the proximal or distal colon, with processes closely opposed to multiple cell types. Calcium imaging in semi-intact mouse colon preparations revealed changes in spontaneous and evoked neural activity, as well as activation of non-neuronal cells, induced by sympathetic nerve stimulation. The overall pattern of response to sympathetic stimulation was unique to the proximal or distal colon. Region-specific changes in cellular activity correlated with motility patterns produced by electrical and optogenetic stimulation of sympathetic pathways. Pharmacology experiments (mouse) and RNA sequencing (human) indicated that appropriate receptors were expressed on different cell types to account for the responses to sympathetic stimulation. Regional differences in expression of α-1 adrenoceptors in human colon emphasize the translational relevance of our mouse findings. CONCLUSIONS: Sympathetic neurons differentially regulate activity of neurons and non-neuronal cells in proximal and distal colon to promote distinct changes in motility patterns, likely reflecting the distinct roles played by these 2 regions.


Asunto(s)
Colon/inervación , Ganglios Simpáticos/fisiología , Motilidad Gastrointestinal/fisiología , Plexo Mientérico/fisiología , Animales , Colon/citología , Colon/efectos de los fármacos , Colon/fisiología , Femenino , Ganglios Simpáticos/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Guanetidina/farmacología , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inervación , Mucosa Intestinal/fisiología , Masculino , Ratones , Modelos Animales , Plexo Mientérico/citología , Plexo Mientérico/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Optogenética , Prazosina/farmacología , RNA-Seq , Análisis de la Célula Individual , Yohimbina/farmacología
10.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G919-G935, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33729000

RESUMEN

Visceral smooth muscle is a crucial component of the walls of hollow organs like the gut, bladder, and uterus. This specialized smooth muscle has unique properties that distinguish it from other muscle types and facilitate robust dilation and contraction. Visceral myopathies are diseases where severe visceral smooth muscle dysfunction prevents efficient movement of air and nutrients through the bowel, impairs bladder emptying, and affects normal uterine contraction and relaxation, particularly during pregnancy. Disease severity exists along a spectrum. The most debilitating defects cause highly dysfunctional bowel, reduced intrauterine colon growth (microcolon), and bladder-emptying defects requiring catheterization, a condition called megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS). People with MMIHS often die early in childhood. When the bowel is the main organ affected and microcolon is absent, the condition is known as myopathic chronic intestinal pseudo-obstruction (CIPO). Visceral myopathies like MMIHS and myopathic CIPO are most commonly caused by mutations in contractile apparatus cytoskeletal proteins. Here, we review visceral myopathy-causing mutations and normal functions of these disease-associated proteins. We propose molecular, cellular, and tissue-level models that may explain clinical and histopathological features of visceral myopathy and hope these observations prompt new mechanistic studies.


Asunto(s)
Citoesqueleto/genética , Seudoobstrucción Intestinal/diagnóstico , Músculo Liso/patología , Citoesqueleto/patología , Humanos , Seudoobstrucción Intestinal/genética , Seudoobstrucción Intestinal/patología , Mutación
11.
Gastroenterology ; 159(5): 1824-1838.e17, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32687927

RESUMEN

BACKGROUND & AIMS: Hirschsprung disease (HSCR) is a life-threatening birth defect in which the distal colon is devoid of enteric neural ganglia. HSCR is treated by surgical removal of aganglionic bowel, but many children continue to have severe problems after surgery. We studied whether administration of glial cell derived neurotrophic factor (GDNF) induces enteric nervous system regeneration in mouse models of HSCR. METHODS: We performed studies with four mouse models of HSCR: Holstein (HolTg/Tg, a model for trisomy 21-associated HSCR), TashT (TashTTg/Tg, a model for male-biased HSCR), Piebald-lethal (Ednrbs-l//s-l, a model for EDNRB mutation-associated HSCR), and Ret9/- (with aganglionosis induced by mycophenolate). Mice were given rectal enemas containing GDNF or saline (control) from postnatal days 4 through 8. We measured survival times of mice, and colon tissues were analyzed by histology, immunofluorescence, and immunoblots. Neural ganglia regeneration and structure, bowel motility, epithelial permeability, muscle thickness, and neutrophil infiltration were studied in colon tissues and in mice. Stool samples were collected, and microbiomes were analyzed by 16S rRNA gene sequencing. Time-lapse imaging and genetic cell-lineage tracing were used to identify a source of GDNF-targeted neural progenitors. Human aganglionic colon explants from children with HSCR were cultured with GDNF and evaluated for neurogenesis. RESULTS: GDNF significantly prolonged mean survival times of HolTg/Tg mice, Ednrbs-l//s-l mice, and male TashTTg/Tg mice, compared with control mice, but not Ret9/- mice (which had mycophenolate toxicity). Mice given GDNF developed neurons and glia in distal bowel tissues that were aganglionic in control mice, had a significant increase in colon motility, and had significant decreases in epithelial permeability, muscle thickness, and neutrophil density. We observed dysbiosis in fecal samples from HolTg/Tg mice compared with feces from wild-type mice; fecal microbiomes of mice given GDNF were similar to those of wild-type mice except for Bacteroides. Exogenous luminal GDNF penetrated aganglionic colon epithelium of HolTg/Tg mice, inducing production of endogenous GDNF, and new enteric neurons and glia appeared to arise from Schwann cells within extrinsic nerves. GDNF application to cultured explants of human aganglionic bowel induced proliferation of Schwann cells and formation of new neurons. CONCLUSIONS: GDNF prolonged survival, induced enteric neurogenesis, and improved colon structure and function in 3 mouse models of HSCR. Application of GDNF to cultured explants of aganglionic bowel from children with HSCR induced proliferation of Schwann cells and formation of new neurons. GDNF might be developed for treatment of HSCR.


Asunto(s)
Colon/efectos de los fármacos , Colon/inervación , Sistema Nervioso Entérico/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Enfermedad de Hirschsprung/tratamiento farmacológico , Regeneración Nerviosa/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Animales , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Disbiosis , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/patología , Sistema Nervioso Entérico/fisiopatología , Microbioma Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Enfermedad de Hirschsprung/fisiopatología , Humanos , Absorción Intestinal/efectos de los fármacos , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Permeabilidad , Recuperación de la Función , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Células de Schwann/patología , Técnicas de Cultivo de Tejidos
12.
Gastroenterology ; 158(8): 2221-2235.e5, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32113825

RESUMEN

BACKGROUND & AIMS: Small, 2-dimensional sections routinely used for human pathology analysis provide limited information about bowel innervation. We developed a technique to image human enteric nervous system (ENS) and other intramural cells in 3 dimensions. METHODS: Using mouse and human colon tissues, we developed a method that combines tissue clearing, immunohistochemistry, confocal microscopy, and quantitative analysis of full-thickness bowel without sectioning to quantify ENS and other intramural cells in 3 dimensions. RESULTS: We provided 280 adult human colon confocal Z-stacks from persons without known bowel motility disorders. Most of our images were of myenteric ganglia, captured using a 20× objective lens. Full-thickness colon images, viewed with a 10× objective lens, were as large as 4 × 5 mm2. Colon from 2 pediatric patients with Hirschsprung disease was used to show distal colon without enteric ganglia, as well as a transition zone and proximal pull-through resection margin where ENS was present. After testing a panel of antibodies with our method, we identified 16 antibodies that bind to molecules in neurons, glia, interstitial cells of Cajal, and muscularis macrophages. Quantitative analyses demonstrated myenteric plexus in 24.5% ± 2.4% of flattened colon Z-stack area. Myenteric ganglia occupied 34% ± 4% of myenteric plexus. Single myenteric ganglion volume averaged 3,527,678 ± 573,832 mm3 with 38,706 ± 5763 neuron/mm3 and 129,321 ± 25,356 glia/mm3. Images of large areas provided insight into why published values of ENS density vary up to 150-fold-ENS density varies greatly, across millimeters, so analyses of small numbers of thin sections from the same bowel region can produce varying results. Neuron subtype analysis revealed that approximately 56% of myenteric neurons stained with neuronal nitric oxide synthase antibody and approximately 33% of neurons produce and store acetylcholine. Transition zone regions from colon tissues of patients with Hirschsprung disease had ganglia in multiple layers and thick nerve fiber bundles without neurons. Submucosal neuron distribution varied among imaged colon regions. CONCLUSIONS: We developed a 3-dimensional imaging method for colon that provides more information about ENS structure than tissue sectioning. This approach could improve diagnosis for human bowel motility disorders and may be useful for other bowel diseases as well.


Asunto(s)
Colon/inervación , Ganglios Autónomos/patología , Enfermedad de Hirschsprung/patología , Interpretación de Imagen Asistida por Computador , Imagenología Tridimensional , Microscopía Confocal , Plexo Mientérico/patología , Plexo Submucoso/patología , Animales , Automatización , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Nitrérgicas/patología , Valor Predictivo de las Pruebas , Fijación del Tejido
13.
Proc Natl Acad Sci U S A ; 115(18): 4696-4701, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29666241

RESUMEN

The nervous system of the bowel regulates the inflammatory phenotype of tissue resident muscularis macrophages (MM), and in adult mice, enteric neurons are the main local source of colony stimulating factor 1 (CSF1), a protein required for MM survival. Surprisingly, we find that during development MM colonize the bowel before enteric neurons. This calls into question the requirement for neuron-derived CSF1 for MM colonization of the bowel. To determine if intestinal innervation is required for MM development, we analyzed MM of neonatal Ret-/- (Ret KO) mice that have no enteric nervous system in small bowel or colon. We found normal numbers of well-patterned MM in Ret KO bowel. Similarly, the abundance and distribution of MM in aganglionic human colon obtained from Hirschsprung disease patients was normal. We also identify endothelial cells and interstitial cells of Cajal as the main sources of CSF1 in the developing bowel. Additionally, MM from neonatal Ret KOs do not differ from controls in baseline activation status or cytokine-production in response to lipopolysaccharide. Unexpectedly, these data demonstrate that the enteric nervous system is dispensable for MM colonization and patterning in the bowel, and suggest that modulatory interactions between MM and the bowel nervous system are established postnatally.


Asunto(s)
Comunicación Celular/fisiología , Sistema Nervioso Entérico/embriología , Feto/embriología , Intestinos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Neuronas/metabolismo , Animales , Sistema Nervioso Entérico/citología , Feto/citología , Intestinos/citología , Intestinos/embriología , Intestinos/inervación , Macrófagos/citología , Ratones , Ratones Noqueados , Neuronas/citología
14.
J Pediatr Gastroenterol Nutr ; 71(2): e59-e67, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32287151

RESUMEN

OBJECTIVES: Motility and functional disorders are common in children and often debilitating, yet these disorders remain challenging to treat effectively. At the 2018 Annual North American Society for Pediatric Gastroenterology, Hepatology and Nutrition meeting, the Neurogastroenterology and Motility Committee held a full day symposium entitled, 2018 Advances In Motility and In NeuroGastroenterology - AIMING for the future. The symposium aimed to explore clinical paradigms in pediatric gastrointestinal motility disorders and provided a foundation for advancing new scientific and therapeutic research strategies. METHODS: The symposium brought together leading experts throughout North America to review the state of the art in the diagnosis and management of motility and functional disorders in children. Presentations were divided into esophageal, antral duodenal, and colorectal modules. Each module included oral presentations by experts in the respective fields, leading to thought-provoking discussions. There were 2 breakout sessions with small group discussions on select topics, focusing on defining scientific insights into the diagnosis and management of pediatric functional gastrointestinal and motility disorders in a systematic, segment-based approach. CONCLUSIONS: The field of neurogastroenterology has made remarkable progress in the last decade. The current report summarizes the major learning points from the symposium highlighting the diagnosis and promising therapies on the horizon for pediatric neurogastrointestinal and motility disorders.


Asunto(s)
Gastroenterología , Enfermedades Gastrointestinales , Niño , Esófago , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/terapia , Motilidad Gastrointestinal , Humanos , América del Norte
15.
Dev Biol ; 444 Suppl 1: S337-S351, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30292786

RESUMEN

Transcription factors that coordinate migration, differentiation or proliferation of enteric nervous system (ENS) precursors are not well defined. To identify novel transcriptional regulators of ENS development, we performed microarray analysis at embryonic day (E) 17.5 and identified many genes that were enriched in the ENS compared to other bowel cells. We decided to investigate the T-box transcription factor Tbx3, which is prominently expressed in developing and mature ENS. Haploinsufficiency for TBX3 causes ulnar-mammary syndrome (UMS) in humans, a multi-organ system disorder. TBX3 also regulates several genes known to be important for ENS development. To test the hypothesis that Tbx3 is important for ENS development or function, we inactivated Tbx3 in all neural crest derivatives, including ENS progenitors using Wnt1-Cre and a floxed Tbx3 allele. Tbx3 fl/fl; Wnt1-Cre conditional mutant mice die shortly after birth with cleft palate and difficulty feeding. The ENS of mutants was well-organized with a normal density of enteric neurons and nerve fiber bundles, but small bowel glial cell density was reduced. Despite this, bowel motility appeared normal. Furthermore, although Tbx3 is expressed in cardiac neural crest, Tbx3 fl/fl; Wnt1-Cre mice had structurally normal hearts. Thus, loss of Tbx3 within neural crest has selective effects on Tbx3-expressing neural crest derivatives.


Asunto(s)
Sistema Nervioso Entérico/embriología , Cresta Neural/embriología , Proteínas de Dominio T Box/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Fisura del Paladar/embriología , Fisura del Paladar/genética , Corazón/embriología , Intestinos/embriología , Ratones , Ratones Endogámicos C57BL , Cresta Neural/metabolismo , Cresta Neural/fisiología , Neurogénesis , Neuroglía/fisiología , Neuronas , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Proteína Wnt1
17.
Dev Biol ; 417(2): 188-97, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-26997034

RESUMEN

Intestinal function is primarily controlled by an intrinsic nervous system of the bowel called the enteric nervous system (ENS). The cells of the ENS are neural crest derivatives that migrate into and through the bowel during early stages of organogenesis before differentiating into a wide variety of neurons and glia. Although genetic factors critically underlie ENS development, it is now clear that many non-genetic factors may influence the number of enteric neurons, types of enteric neurons, and ratio of neurons to glia. These non-genetic influences include dietary nutrients and medicines that may impact ENS structure and function before or after birth. This review summarizes current data about gene-environment interactions that affect ENS development and suggests that these factors may contribute to human intestinal motility disorders like Hirschsprung disease or irritable bowel syndrome.


Asunto(s)
Sistema Nervioso Entérico/crecimiento & desarrollo , Sistema Nervioso Entérico/fisiopatología , Tracto Gastrointestinal/inervación , Interacción Gen-Ambiente , Enfermedad de Hirschsprung/patología , Plasticidad Neuronal/fisiología , Animales , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Motilidad Gastrointestinal/fisiología , Enfermedad de Hirschsprung/genética , Humanos , Síndrome del Colon Irritable/patología , Cresta Neural/citología , Neuroglía/citología
18.
Dev Biol ; 409(1): 152-165, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26546974

RESUMEN

Mutations that impair the proliferation of enteric neural crest-derived cells (ENCDC) cause Hirschsprung disease, a potentially lethal birth defect where the enteric nervous system (ENS) is absent from distal bowel. Inosine 5' monophosphate dehydrogenase (IMPDH) activity is essential for de novo GMP synthesis, and chemical inhibition of IMPDH induces Hirschsprung disease-like pathology in mouse models by reducing ENCDC proliferation. Two IMPDH isoforms are ubiquitously expressed in the embryo, but only IMPDH2 is required for life. To further understand the role of IMPDH2 in ENS and neural crest development, we characterized a conditional Impdh2 mutant mouse. Deletion of Impdh2 in the early neural crest using the Wnt1-Cre transgene produced defects in multiple neural crest derivatives including highly penetrant intestinal aganglionosis, agenesis of the craniofacial skeleton, and cardiac outflow tract and great vessel malformations. Analysis using a Rosa26 reporter mouse suggested that some or all of the remaining ENS in Impdh2 conditional-knockout animals was derived from cells that escaped Wnt1-Cre mediated DNA recombination. These data suggest that IMPDH2 mediated guanine nucleotide synthesis is essential for normal development of the ENS and other neural crest derivatives.


Asunto(s)
Sistema Nervioso Entérico/irrigación sanguínea , Sistema Nervioso Entérico/embriología , Cara/embriología , IMP Deshidrogenasa/metabolismo , Cresta Neural/embriología , Cresta Neural/enzimología , Cráneo/embriología , Alelos , Animales , Bromodesoxiuridina/metabolismo , Sistema Nervioso Entérico/enzimología , Sistema Nervioso Entérico/patología , Femenino , Feto/anomalías , Feto/embriología , Eliminación de Gen , Genes Reporteros , Enfermedad de Hirschsprung/patología , IMP Deshidrogenasa/deficiencia , Etiquetado Corte-Fin in Situ , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Especificidad de Órganos , ARN no Traducido/metabolismo , Recombinación Genética/genética , Cráneo/metabolismo , Proteína Wnt1/metabolismo
19.
Dev Biol ; 409(2): 473-88, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26586201

RESUMEN

Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Sistema Nervioso Entérico/citología , Ibuprofeno/farmacología , Intestinos/citología , Células-Madre Neurales/citología , Citoesqueleto de Actina/metabolismo , Animales , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Pollos , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/metabolismo , Activación Enzimática/efectos de los fármacos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Ratones , Modelos Biológicos , Células 3T3 NIH , Células-Madre Neurales/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , PPAR gamma/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pez Cebra , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
20.
Dev Biol ; 417(2): 229-51, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27059883

RESUMEN

Over the last 20 years, there has been increasing focus on the development of novel stem cell based therapies for the treatment of disorders and diseases affecting the enteric nervous system (ENS) of the gastrointestinal tract (so-called enteric neuropathies). Here, the idea is that ENS progenitor/stem cells could be transplanted into the gut wall to replace the damaged or absent neurons and glia of the ENS. This White Paper sets out experts' views on the commonly used methods and approaches to identify, isolate, purify, expand and optimize ENS stem cells, transplant them into the bowel, and assess transplant success, including restoration of gut function. We also highlight obstacles that must be overcome in order to progress from successful preclinical studies in animal models to ENS stem cell therapies in the clinic.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistema Nervioso Entérico/patología , Tracto Gastrointestinal/patología , Enfermedad de Hirschsprung/terapia , Seudoobstrucción Intestinal/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Tracto Gastrointestinal/inervación , Guías como Asunto , Enfermedad de Hirschsprung/patología , Humanos , Seudoobstrucción Intestinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA