Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Gastroenterology ; 162(1): 179-192.e11, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34425092

RESUMEN

BACKGROUND AND AIMS: The enteric nervous system, which regulates many gastrointestinal functions, is derived from neural crest cells (NCCs). Defective NCC migration during embryonic development may lead to enteric neuropathies such as Hirschsprung's disease (hindgut aganglionosis). Sox10 is known to be essential for cell migration but downstream molecular events regulating early NCC migration have not been fully elucidated. This study aimed to determine how Sox10 regulates migration of sacral NCCs toward the hindgut using Dominant megacolon mice, an animal model of Hirschsprung's disease with a Sox10 mutation. METHODS: We used the following: time-lapse live cell imaging to determine the migration defects of mutant sacral NCCs; genome-wide microarrays, site-directed mutagenesis, and whole embryo culture to identify Sox10 targets; and liquid chromatography and tandem mass spectrometry to ascertain downstream effectors of Sox10. RESULTS: Sacral NCCs exhibited retarded migration to the distal hindgut in Sox10-null embryos with simultaneous down-regulated expression of cadherin-19 (Cdh19). Sox10 was found to bind directly to the Cdh19 promoter. Cdh19 knockdown resulted in retarded sacral NCC migration in vitro and ex vivo, whereas re-expression of Cdh19 partially rescued the retarded migration of mutant sacral NCCs in vitro. Cdh19 formed cadherin-catenin complexes, which then bound to filamentous actin of the cytoskeleton during cell migration. CONCLUSIONS: Cdh19 is a direct target of Sox10 during early sacral NCC migration toward the hindgut and forms cadherin-catenin complexes which interact with the cytoskeleton in migrating cells. Elucidation of this novel molecular pathway helps to provide insights into the pathogenesis of enteric nervous system developmental defects.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Sistema Nervioso Entérico/metabolismo , Enfermedad de Hirschsprung/metabolismo , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Factores de Transcripción SOXE/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Cadherinas/genética , Células Cultivadas , Modelos Animales de Enfermedad , Técnicas de Cultivo de Embriones , Sistema Nervioso Entérico/anomalías , Regulación del Desarrollo de la Expresión Génica , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Cresta Neural/anomalías , Células-Madre Neurales/patología , Unión Proteica , Factores de Transcripción SOXE/genética , Transducción de Señal , Factores de Tiempo
2.
Rev. cient. Esc. Univ. Cienc. Salud ; 7(1): 35-41, ene.-jun. 2020. ilus
Artículo en Español | LILACS | ID: biblio-1224619

RESUMEN

La enfermedad de Hirschsprung es una malformación del sistema nervioso entérico, caracterizada por falta de células ganglionares en plexo submucoso y mientérico en pared distal del colon. En el 80% solo existe afectación del recto-sigma, sin embargo, pueden encontrarse casos de aganglionosis total. Reporte de Caso: Se presenta pre-escolar masculino de 4 años de edad, procedente de La Iguala, Lempira, Honduras, con historia de constipación y distensión abdominal de dos años de evolución, que ha empeorado progresivamente, y se atenuaba con el uso de enemas cada dos días para poder defecar. En mayo de 2016 llega al Hospital Mario Catarino Rivas, donde se le realizó una laparotomía exploratoria de emergencia debido a la presencia de deterioro clínico por constipación y distensión abdominal, acompañada de vómitos con restos alimenticios y fiebre alta de una semana de evolución. En dicha operación se realizó colectomía parcial izquierda, además se realizó una colostomía terapéutica y se diagnósticó enfermedad de Hirs-chsprung mediante biopsia, además del hallazgo incidental de apendicitis, se realizó apendicectomía obteniendo en general, una evolución satisfactoria.El estudio histopatológico de la biopsia del recto es el estándar de oro para realizar el diagnóstico. La ausencia de células ganglionares en el plexo submucoso con latinción hematoxilina y eosina establece el diagnóstico. El tratamiento de la enfermedad de Hirschsprung es de tipo quirúrgico y se busca la eliminación del segmento colónico afectado, de manera que se pueda lograr una anastomosis del colon proximal y distal al área agangliónica...(AU)


Asunto(s)
Humanos , Masculino , Preescolar , Apendicitis/diagnóstico , Enfermedad de Hirschsprung/diagnóstico , Sistema Nervioso Entérico/anomalías , Estreñimiento
3.
Clin Transl Gastroenterol ; 10(12): e00093, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31789936

RESUMEN

OBJECTIVES: Intestinal neuronal dysplasia (IND) is a common malformation of the enteric nervous system. Diagnosis requires a full-thickness colonic specimen and an experienced pathologist, emphasizing the need for noninvasive analytical methods. Recently, the methylation level of the Sox10 promoter has been found to be critical for enteric nervous system development. However, whether it can be used for diagnostic purposes in IND is unclear. METHODS: Blood and colon specimens were collected from 32 patients with IND, 60 patients with Hirschsprung disease (HD), and 60 controls. Sox10 promoter methylation in the blood and the Sox10 expression level in the colon were determined, and their correlation was analyzed. The diagnostic efficacy of blood Sox10 promoter methylation was analyzed by receiver operating characteristic curve. RESULTS: The blood level of Sox10 promoter methylation at the 32nd locus was 100% (90%-100%; 95% confidence interval [CI], 92.29%-96.37%) in control, 90% (80%-90%; 95% CI, 82.84%-87.83%) in HD, and 60% (50%-80%; 95% CI, 57.12%-69.76%) in IND specimens. Sox10 promoter methylation in the peripheral blood was negatively correlated with Sox10 expression in the colon, which was low in control, moderate in HD, and high in IND specimens (r = -0.89). The area under the curve of Sox10 promoter methylation in the diagnosis of IND was 0.94 (95% CI, 0.874-1.000, P = 0.000), with a cutoff value of 85% (sensitivity, 90.6%; specificity, 95.0%). By applying a cutoff value of 65%, promoter methylation was more indicative of IND than HD. DISCUSSION: The analysis of Sox10 promoter methylation in the peripheral blood can be used as a noninvasive method for IND diagnosis.


Asunto(s)
Metilación de ADN , Sistema Nervioso Entérico/anomalías , Enfermedades Intestinales/diagnóstico , Enfermedades del Sistema Nervioso/diagnóstico , Factores de Transcripción SOXE/genética , Biomarcadores/sangre , Niño , Colon/patología , Colon/cirugía , Islas de CpG/genética , Diagnóstico Diferencial , Estudios de Factibilidad , Femenino , Enfermedad de Hirschsprung/sangre , Enfermedad de Hirschsprung/diagnóstico , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Humanos , Enfermedades Intestinales/sangre , Enfermedades Intestinales/genética , Enfermedades Intestinales/patología , Mucosa Intestinal/inervación , Masculino , Enfermedades del Sistema Nervioso/sangre , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología , Valor Predictivo de las Pruebas , Regiones Promotoras Genéticas/genética , Curva ROC , Factores de Transcripción SOXE/metabolismo , Análisis de Secuencia de ADN
4.
Asian J Surg ; 40(1): 29-34, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26216257

RESUMEN

BACKGROUND: Allied disorders of Hirschsprung's disease (ADHD) have been proposed to be the concept of the functional obstruction of the intestine with the presence of ganglion cells in the terminal rectum. They are classified into two categories based on pathology: (1) abnormal ganglia, including immaturity of ganglia, hypoganglionosis (HG), and intestinal neuronal dysplasia; (2) normal ganglia, including megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS), segmental dilatation (SD), internal anal sphincter achalasia (IASA), and chronic idiopathic intestinal pseudo-obstruction (CIIP). Some of these show poor prognosis, therefore, the establishment of criteria and appropriate treatment strategies is required. METHODS: The questionnaires were sent to the 161 major institutes of pediatric surgery or gastroenterology in Japan, in order to collect the cases of ADHD during 10 years from 2001 and 2010. RESULTS: In total, 355 cases were collected. They included 28 immaturity of ganglia, 130 HG (121 congenital, 9 acquired), 18 intestinal neuronal dysplasia, 33 MMIHS, 43 SD, three IASA, and 100 CIIP. Of the 95 institutes, 69 (72.6%) had their own criteria for ADHD. Criteria were based on clinical symptoms and signs, and conventional pathological examinations. Prognosis was poor in congenital HG, MMIHS, and CIIP, while the others showed good survival rates. CONCLUSION: Almost all Japanese cases of ADHD in the past 10 years were collected. Congenital HG and CIIP showed relatively high incidence, whereas acquired HG and IASA were extremely rare in Japan. The criteria of each disorder were also collected and summarized. Prognosis was poor in congenital HG, MMIHS, and CIIP.


Asunto(s)
Anomalías Múltiples/epidemiología , Colon/anomalías , Sistema Nervioso Entérico/anomalías , Ganglios/anomalías , Enfermedad de Hirschsprung/epidemiología , Seudoobstrucción Intestinal/epidemiología , Vejiga Urinaria/anomalías , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Colon/patología , Diagnóstico Diferencial , Estudios de Seguimiento , Encuestas Epidemiológicas , Enfermedad de Hirschsprung/diagnóstico , Enfermedad de Hirschsprung/patología , Humanos , Incidencia , Seudoobstrucción Intestinal/diagnóstico , Seudoobstrucción Intestinal/patología , Japón/epidemiología , Pronóstico , Estudios Retrospectivos , Vejiga Urinaria/patología
6.
Berl Munch Tierarztl Wochenschr ; 129(7-8): 269-81, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27529988

RESUMEN

Leucism is characterized by a complete or partial white skin and hair in combination with pigmented irides, which can be vivid blue or heterochromatic. This is due to a complete or partial lack of melanocytes. The underlying pathogenesis is a disturbed emigration or differentiation of neural crest-derived cells. Therefore, leucistic phenotypes can be associated with defects, which mainly impair sensory organs and nerves. In humans, a well-known example is the Waardenburg syndrome. Leucism-associated disorders were also described in mouse, rat, hamster, rabbit, mink, cat, dog, pig, sheep, llama, alpaca, cattle and horse. In some of these species already identified causal mutations affect the genes EDN3, EDNRB, KIT, MITF, PAX3, SILV and SOX10. Defect alleles represent different types of genetic variation, ranging from single nucleotide substitutions up to larger chromosomal deletions. Some of the defect alleles produce desired coat color patterns. In some but not all cases, available genetic tests enable breeders to avoid production of animals affected by a leucism-associated disorder.


Asunto(s)
Deleción Cromosómica , Mutación Puntual , Síndrome de Waardenburg/veterinaria , Animales , Gatos , Bovinos , Perros , Sistema Nervioso Entérico/anomalías , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/veterinaria , Caballos , Humanos , Ratones , Cresta Neural/embriología , Conejos , Ratas , Ovinos , Síndrome de Waardenburg/genética , Síndrome de Waardenburg/prevención & control
7.
J Clin Invest ; 126(6): 2221-35, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27111230

RESUMEN

Autism spectrum disorder (ASD) is an increasingly common behavioral condition that frequently presents with gastrointestinal (GI) disturbances. It is not clear, however, how gut dysfunction relates to core ASD features. Multiple, rare hyperfunctional coding variants of the serotonin (5-HT) transporter (SERT, encoded by SLC6A4) have been identified in ASD. Expression of the most common SERT variant (Ala56) in mice increases 5-HT clearance and causes ASD-like behaviors. Here, we demonstrated that Ala56-expressing mice display GI defects that resemble those seen in mice lacking neuronal 5-HT. These defects included enteric nervous system hypoplasia, slow GI transit, diminished peristaltic reflex activity, and proliferation of crypt epithelial cells. An opposite phenotype was seen in SERT-deficient mice and in progeny of WT dams given the SERT antagonist fluoxetine. The reciprocal phenotypes that resulted from increased or decreased SERT activity support the idea that 5-HT signaling regulates enteric neuronal development and can, when disturbed, cause long-lasting abnormalities of GI function. Administration of a 5-HT4 agonist to Ala56 mice during development prevented Ala56-associated GI perturbations, suggesting that excessive SERT activity leads to inadequate 5-HT4-mediated neurogenesis. We propose that deficient 5-HT signaling during development may contribute to GI and behavioral features of ASD. The consequences of therapies targeting SERT during pregnancy warrant further evaluation.


Asunto(s)
Tracto Gastrointestinal/anomalías , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Sistema Nervioso Entérico/anomalías , Sistema Nervioso Entérico/crecimiento & desarrollo , Sistema Nervioso Entérico/fisiopatología , Femenino , Motilidad Gastrointestinal/genética , Motilidad Gastrointestinal/fisiología , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/fisiopatología , Variación Genética , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Modelos Animales , Neurogénesis , Embarazo , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia
9.
Gastroenterology ; 149(7): 1837-1848.e5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26261006

RESUMEN

BACKGROUND & AIMS: Hirschsprung disease is characterized by a deficit in enteric neurons, which are derived from neural crest cells (NCCs). Aberrant hedgehog signaling disrupts NCC differentiation and might cause Hirschsprung disease. We performed genetic analyses to determine whether hedgehog signaling is involved in pathogenesis. METHODS: We performed deep-target sequencing of DNA from 20 patients with Hirschsprung disease (16 men, 4 women), and 20 individuals without (controls), and searched for mutation(s) in GLI1, GLI2, GLI3, SUFU, and SOX10. Biological effects of GLI mutations were tested in luciferase reporter assays using HeLa or neuroblastoma cell lines. Development of the enteric nervous system was studied in Sufu(f/f), Gli3(Δ699), Wnt1-Cre, and Sox10(NGFP) mice using immunohistochemical and whole-mount staining procedures to quantify enteric neurons and glia and analyze axon fasciculation, respectively. NCC migration was studied using time-lapse imaging. RESULTS: We identified 3 mutations in GLI in 5 patients with Hirschsprung disease but no controls; all lead to increased transcription of SOX10 in cell lines. SUFU, GLI, and SOX10 form a regulatory loop that controls the neuronal vs glial lineages and migration of NCCs. Sufu mutants mice had high Gli activity, due to loss of Sufu, disrupting the regulatory loop and migration of enteric NCCs, leading to defective axonal fasciculation, delayed gut colonization, or intestinal hypoganglionosis. The ratio of enteric neurons to glia correlated inversely with Gli activity. CONCLUSIONS: We identified mutations that increase GLI activity in patients with Hirschsprung disease. Disruption of the SUFU-GLI-SOX10 regulatory loop disrupts migration of NCCs and development of the enteric nervous system in mice.


Asunto(s)
Sistema Nervioso Entérico/anomalías , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/patología , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Estudios de Casos y Controles , Linaje de la Célula , Movimiento Celular , Análisis Mutacional de ADN/métodos , Modelos Animales de Enfermedad , Sistema Nervioso Entérico/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedad de Hirschsprung/diagnóstico , Enfermedad de Hirschsprung/metabolismo , Humanos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Cresta Neural/metabolismo , Neurogénesis , Proteínas Nucleares/metabolismo , Fenotipo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Factores de Transcripción/metabolismo , Transfección , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
10.
Neurogastroenterol Motil ; 27(10): 1371-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26172388

RESUMEN

BACKGROUND: Hirschsprung disease (HSCR) is a congenital and heterogeneous disorder characterized by the absence of enteric ganglia during enteric nervous system (ENS) development. Our recent genome-wide association study has identified a variant (rs6509940) of interleukin-11 (IL-11) as a potential susceptible locus for HSCR. As interleukins play important roles in the ENS, we further studied associations with HSCR of nine common single nucleotide polymorphisms (SNPs) on IL-11. METHODS: Biopsy specimens or surgical materials from all patients that were tested for histological examination based on the absence of the enteric ganglia were collected. A total of nine SNPs on IL-11 were genotyped in 187 HSCR patients and 283 unaffected controls using TaqMan genotyping assay. KEY RESULTS: Combined analysis revealed that several SNPs (minimum p = 1.57 × 10(-7) ) showed statistically significant associations with HSCR, even after Bonferroni correction (pcorr  = 1.73 × 10(-6) for the SNP). Moreover, the most common haplotype was strongly associated with HSCR (pcorr  = 2.20 × 10(-6) ). In further analysis among three HSCR subtypes (short segment, S-HSCR; long segment, L-HSCR; total colonic aganglionosis, TCA) based on the extent of aganglionic segment, the result showed a different association pattern depending on the subtypes (minimum pcorr  = 6.12 × 10(-5) for rs6509940 in S-HSCR; but no significant SNP in L-HSCR and TCA). CONCLUSIONS & INFERENCES: Although further replication in a larger cohort and functional evaluations are needed, our findings suggest that genetic variations of IL-11 may be associated with the risk of HSCR and/or the mechanisms related to ENS development.


Asunto(s)
Sistema Nervioso Entérico/anomalías , Enfermedad de Hirschsprung/genética , Interleucina-11/genética , Femenino , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple
11.
PLoS Genet ; 11(3): e1005093, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25786024

RESUMEN

Neural crest cells (NCC) are a transient migratory cell population that generates diverse cell types such as neurons and glia of the enteric nervous system (ENS). Via an insertional mutation screen for loci affecting NCC development in mice, we identified one line-named TashT-that displays a partially penetrant aganglionic megacolon phenotype in a strong male-biased manner. Interestingly, this phenotype is highly reminiscent of human Hirschsprung's disease, a neurocristopathy with a still unexplained male sex bias. In contrast to the megacolon phenotype, colonic aganglionosis is almost fully penetrant in homozygous TashT animals. The sex bias in megacolon expressivity can be explained by the fact that the male ENS ends, on average, around a "tipping point" of minimal colonic ganglionosis while the female ENS ends, on average, just beyond it. Detailed analysis of embryonic intestines revealed that aganglionosis in homozygous TashT animals is due to slower migration of enteric NCC. The TashT insertional mutation is localized in a gene desert containing multiple highly conserved elements that exhibit repressive activity in reporter assays. RNAseq analyses and 3C assays revealed that the TashT insertion results, at least in part, in NCC-specific relief of repression of the uncharacterized gene Fam162b; an outcome independently confirmed via transient transgenesis. The transcriptional signature of enteric NCC from homozygous TashT embryos is also characterized by the deregulation of genes encoding members of the most important signaling pathways for ENS formation-Gdnf/Ret and Edn3/Ednrb-and, intriguingly, the downregulation of specific subsets of X-linked genes. In conclusion, this study not only allowed the identification of Fam162b coding and regulatory sequences as novel candidate loci for Hirschsprung's disease but also provides important new insights into its male sex bias.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Ratones , Mutagénesis Insercional , Animales , Cromosomas de los Mamíferos , Sistema Nervioso Entérico/anomalías , Enfermedad de Hirschsprung/embriología , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/metabolismo , Elementos Silenciadores Transcripcionales , Transcriptoma
12.
J Clin Invest ; 123(11): 4875-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24216510

RESUMEN

Hirschsprung disease (HSCR) is a partially penetrant oligogenic birth defect that occurs when enteric nervous system (ENS) precursors fail to colonize the distal bowel during early pregnancy. Genetic defects underlie HSCR, but much of the variability in the occurrence and severity of the birth defect remain unexplained. We hypothesized that nongenetic factors might contribute to disease development. Here we found that mycophenolate, an inhibitor of de novo guanine nucleotide biosynthesis, and 8 other drugs identified in a zebrafish screen impaired ENS development. In mice, mycophenolate treatment selectively impaired ENS precursor proliferation, delayed precursor migration, and induced bowel aganglionosis. In 2 different mouse models of HSCR, addition of mycophenolate increased the penetrance and severity of Hirschsprung-like pathology. Mycophenolate treatment also reduced ENS precursor migration as well as lamellipodia formation, proliferation, and survival in cultured enteric neural crest­derived cells. Using X-inactivation mosaicism for the purine salvage gene Hprt, we found that reduced ENS precursor proliferation most likely causes mycophenolate-induced migration defects and aganglionosis. To the best of our knowledge, mycophenolate is the first medicine identified that causes major ENS malformations and Hirschsprung-like pathology in a mammalian model. These studies demonstrate a critical role for de novo guanine nucleotide biosynthesis in ENS development and suggest that some cases of HSCR may be preventable.


Asunto(s)
Guanosina Monofosfato/biosíntesis , Enfermedad de Hirschsprung/etiología , Enfermedad de Hirschsprung/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , ADN/biosíntesis , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sistema Nervioso Entérico/anomalías , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/metabolismo , Femenino , Enfermedad de Hirschsprung/embriología , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ácido Micofenólico/toxicidad , Embarazo , Pez Cebra
13.
J Pediatr Gastroenterol Nutr ; 57(5): 677-86, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24177787

RESUMEN

OBJECTIVE: Primary gastrointestinal neuropathies are a heterogeneous group of enteric nervous system (ENS) disorders that continue to cause difficulties in diagnosis and histological interpretation. Recently, an international working group published guidelines for histological techniques and reporting, along with a classification of gastrointestinal neuromuscular pathology. The aim of this article was to review and summarize the key issues for pediatric gastroenterologists on the diagnostic workup of congenital ENS disorders. In addition, we provide further commentary on the continuing controversies in the field. RESULTS: Although the diagnostic criteria for Hirschsprung disease are well established, those for other forms of dysganglionosis remain ill-defined. Appropriate tissue sampling, handling, and expert interpretation are crucial to maximize diagnostic accuracy and reduce interobserver variability. The absence of validated age-related normal values for neuronal density, along with the lack of correlation between clinical and histological findings, result in significant diagnostic uncertainties while diagnosing quantitative aberrations such as hypoganglionosis or ultrashort Hirschsprung disease. Intestinal neuronal dysplasia remains a histological description of unclear significance. CONCLUSIONS: The evaluation of cellular quantitative or qualitative abnormalities of the ENS for clinical diagnosis remains complex. Such analysis should be carried out in laboratories that have the necessary expertise and access to their own validated reference values.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo/diagnóstico , Anomalías del Sistema Digestivo/diagnóstico , Sistema Nervioso Entérico/fisiopatología , Enfermedades Gastrointestinales/diagnóstico , Tracto Gastrointestinal/inervación , Guías de Práctica Clínica como Asunto , Adolescente , Adulto , Enfermedades del Sistema Nervioso Autónomo/congénito , Enfermedades del Sistema Nervioso Autónomo/patología , Enfermedades del Sistema Nervioso Autónomo/fisiopatología , Niño , Consenso , Anomalías del Sistema Digestivo/patología , Anomalías del Sistema Digestivo/fisiopatología , Neoplasias del Sistema Digestivo/diagnóstico , Neoplasias del Sistema Digestivo/patología , Neoplasias del Sistema Digestivo/fisiopatología , Sistema Nervioso Entérico/anomalías , Sistema Nervioso Entérico/patología , Ganglioneuroma/diagnóstico , Ganglioneuroma/patología , Ganglioneuroma/fisiopatología , Gastroenterología/métodos , Enfermedades Gastrointestinales/congénito , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/fisiopatología , Tracto Gastrointestinal/anomalías , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/fisiopatología , Humanos , Lactante , Seudoobstrucción Intestinal/diagnóstico , Seudoobstrucción Intestinal/patología , Seudoobstrucción Intestinal/fisiopatología , Neoplasia Endocrina Múltiple Tipo 2b/diagnóstico , Neoplasia Endocrina Múltiple Tipo 2b/patología , Neoplasia Endocrina Múltiple Tipo 2b/fisiopatología , Pediatría/métodos
14.
Dev Biol ; 382(1): 305-19, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23838398

RESUMEN

We review morphogenesis of the enteric nervous system from migratory neural crest cells, and defects of this process such as Hirschsprung disease, centering on cell motility and assembly, and cell adhesion and extracellular matrix molecules, along with cell proliferation and growth factors. We then review continuum and agent-based (cellular automata) models with rules of cell movement and logistical proliferation. Both movement and proliferation at the individual cell level are modeled with stochastic components from which stereotyped outcomes emerge at the population level. These models reproduced the wave-like colonization of the intestine by enteric neural crest cells, and several new properties emerged, such as colonization by frontal expansion, which were later confirmed biologically. These models predict a surprising level of clonal heterogeneity both in terms of number and distribution of daughter cells. Biologically, migrating cells form stable chains made up of unstable cells, but this is not seen in the initial model. We outline additional rules for cell differentiation into neurons, axon extension, cell-axon and cell-cell adhesions, chemotaxis and repulsion which can reproduce chain migration. After the migration stage, the cells re-arrange as a network of ganglia. Changes in cell adhesion molecules parallel this, and we describe additional rules based on Steinberg's Differential Adhesion Hypothesis, reflecting changing levels of adhesion in neural crest cells and neurons. This was able to reproduce enteric ganglionation in a model. Mouse mutants with disturbances of enteric nervous system morphogenesis are discussed, and these suggest future refinement of the models. The modeling suggests a relatively simple set of cell behavioral rules could account for complex patterns of morphogenesis. The model has allowed the proposal that Hirschsprung disease is mostly an enteric neural crest cell proliferation defect, not a defect of cell migration. In addition, the model suggests an explanations for zonal and skip segment variants of Hirschsprung disease, and also gives a novel stochastic explanation for the observed discordancy of Hirschsprung disease in identical twins.


Asunto(s)
Sistema Nervioso Entérico/anomalías , Sistema Nervioso Entérico/embriología , Modelos Biológicos , Animales , Sistema Nervioso Entérico/patología , Ganglios/embriología , Ganglios/metabolismo , Ganglios/patología , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/inervación , Humanos , Morfogénesis , Cresta Neural/embriología , Cresta Neural/patología
15.
Transl Res ; 162(1): 1-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23528997

RESUMEN

The enteric nervous system is the part of the autonomic nervous system that directly controls the gastrointestinal tract. Derived from a multipotent, migratory cell population called the neural crest, a complete enteric nervous system is necessary for proper gut function. Disorders that arise as a consequence of defective neural crest cell development are termed neurocristopathies. One such disorder is Hirschsprung disease (HSCR), also known as congenital megacolon or intestinal aganglionosis. HSCR occurs in 1/5000 live births and typically presents with the inability to pass meconium, along with abdominal distension and discomfort that usually requires surgical resection of the aganglionic bowel. This disorder is characterized by a congenital absence of neurons in a portion of the intestinal tract, usually the distal colon, because of a disruption of normal neural crest cell migration, proliferation, differentiation, survival, and/or apoptosis. The inheritance of HSCR disease is complex, often non-Mendelian, and characterized by variable penetrance. Extensive research has identified a number of key genes that regulate neural crest cell development in the pathogenesis of HSCR including RET, GDNF, GFRα1, NRTN, EDNRB, ET3, ZFHX1B, PHOX2b, SOX10, and SHH. However, mutations in these genes account for only ∼50% of the known cases of HSCR. Thus, other genetic mutations and combinations of genetic mutations and modifiers likely contribute to the etiology and pathogenesis of HSCR. The aims of this review are to summarize the HSCR phenotype, diagnosis, and treatment options; to discuss the major genetic causes and the mechanisms by which they disrupt normal enteric neural crest cell development; and to explore new pathways that may contribute to HSCR pathogenesis.


Asunto(s)
Enfermedad de Hirschsprung , Animales , Movimiento Celular , Sistema Nervioso Entérico/anomalías , Tracto Gastrointestinal/anomalías , Enfermedad de Hirschsprung/diagnóstico , Enfermedad de Hirschsprung/embriología , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Enfermedad de Hirschsprung/terapia , Humanos , Mutación , Cresta Neural/citología , Cresta Neural/patología
16.
Prog Neurobiol ; 96(2): 176-89, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22266104

RESUMEN

Knowledge of molecular mechanisms that underlie development of the enteric nervous system has greatly expanded in recent decades. Enteric neuropathies related to aberrant genetic development are thus becoming increasingly recognized. There has been no recent review of these often highly morbid disorders. This review highlights advances in knowledge of the molecular pathogenesis of these disorders from a clinical perspective. It includes diseases characterized by an infantile aganglionic Hirschsprung phenotype and those in which structural abnormalities are less pronounced. The implications for diagnosis, screening and possible reparative approaches are presented.


Asunto(s)
Sistema Nervioso Entérico/anomalías , Sistema Nervioso Entérico/fisiología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Diagnóstico Diferencial , Sistema Nervioso Entérico/patología , Sistema Nervioso Entérico/fisiopatología , Asesoramiento Genético , Pruebas Genéticas , Terapia Genética , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Enfermedad de Hirschsprung/fisiopatología , Humanos
17.
Am J Med Genet A ; 155A(8): 1798-802, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21739599

RESUMEN

Chronic intestinal pseudo-obstruction (CIPO) can occur as a consequence of neuropathies including diffuse Intestinal Neuronal Dysplasia (IND), a relatively rare enteric nervous system (ENS) abnormality. Although various authors reported of diffuse IND associated either with intestinal malrotation or megacystis, the co-existence of these three entities in the same patient has never been described before. The aim of this paper is to report for the first time in literature a series of patient with such association, focusing on one who carries a de novo duplication of chromosome 12, suggesting a new syndromic association (megacolon, megacystis, malrotation).


Asunto(s)
Anomalías Múltiples/genética , Sistema Nervioso Entérico/anomalías , Enfermedades Fetales/diagnóstico , Tracto Gastrointestinal/anomalías , Megacolon/diagnóstico , Anomalía Torsional/diagnóstico , Preescolar , Duplicación Cromosómica , Cromosomas Humanos Par 12/genética , Hibridación Genómica Comparativa , Duodeno/anomalías , Resultado Fatal , Femenino , Enfermedades Fetales/genética , Enfermedades Fetales/terapia , Tracto Gastrointestinal/cirugía , Humanos , Ileostomía , Megacolon/genética , Megacolon/cirugía , Síndrome , Anomalía Torsional/genética , Anomalía Torsional/cirugía , Vejiga Urinaria/anomalías
18.
J Pediatr Surg ; 46(7): 1400-5, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21763842

RESUMEN

OBJECTIVE: The aim of this study was to study the effect of vitamin A deficiency (VAD) on the embryological development of anorectal malformations (ARMs) and the enteric nervous system. MATERIALS AND METHODS: Female Sprague-Dawley rats were divided into 3 groups: VAD group, normal group (negative control), and ethylene thiourea (ETU) group (positive control) with a normal diet. On day 20 of pregnancy, cesarean section was performed on all rats. The incidence of ARMs in the fetal rats and Protein gene product 9.5 (PGP9.5) and S-100 protein expression by immunohistochemistry were determined. RESULTS: The incidence of ARMs in VAD and ETU groups was 64.8% (59/91) and 45.9% (61/133), respectively (P > .05). Anorectal malformations were not found in the normal group. Protein gene product 9.5 and S-100 protein expression in the non-ARM rectums of the VAD group was lower than the ETU (P = .0156 vs P = .0105) and normal groups (P = .0091 vs P = .0024). There was no significant difference in PGP9.5 and S-100 protein expression between ETU and normal groups. In the ARM rectums, PGP9.5 and S-100 protein expression in the VAD group was lower than the ETU group (P < .0001). Protein gene product 9.5 and S-100 protein expression was also lower in ARM than non-ARM rectums in the VAD and ETU groups (P < .0001, P = .0203, and P = .0122, respectively). CONCLUSION: Vitamin A deficiency during pregnancy may result in the embryological development of ARMs. Enteric nervous system development may be related to ARMs.


Asunto(s)
Anomalías Múltiples/etiología , Canal Anal/anomalías , Sistema Nervioso Entérico/anomalías , Complicaciones del Embarazo/fisiopatología , Recto/anomalías , Deficiencia de Vitamina A/fisiopatología , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/etiología , Anomalías Múltiples/inducido químicamente , Anomalías Múltiples/embriología , Canal Anal/embriología , Animales , Ano Imperforado/embriología , Ano Imperforado/etiología , Dieta , Sistema Nervioso Entérico/embriología , Etilenotiourea/toxicidad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Recto/embriología , Proteínas S100/biosíntesis , Proteínas S100/genética , Cola (estructura animal)/anomalías , Cola (estructura animal)/embriología , Teratógenos/toxicidad , Ubiquitina Tiolesterasa/biosíntesis , Ubiquitina Tiolesterasa/genética , Vitamina A/sangre , Deficiencia de Vitamina A/metabolismo
19.
Neurogastroenterol Motil ; 23(11): e510-22, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21395909

RESUMEN

BACKGROUND: The enteric nervous system originates from neural crest cells that migrate into the embryonic foregut and then sequentially colonize the midgut and hindgut. Defects in neural crest migration result in regions of the gut that lack enteric ganglia, a condition in humans called Hirschsprung's disease. The high degree of phenotypic variability reported in Hirschsprung's disease suggests the involvement of modifier genes. METHODS: We used a two-locus complementation approach to screen for genetic interactions between L1cam and members of the endothelin signalling pathway. Immunohistochemistry was used to label PGP9.5(+) enteric neurons and Sox10(+) neural crest-derived cells in wholemount preparations of embryonic gut. Key Results Loss or haploinsufficiency of L1cam significantly increased the severity of aganglionosis in Et-3 and Ednrb null mutant embryos. Furthermore, the colonization of the developing gut by neural crest-derived cells was significantly delayed in L1cam(-/y) ; Et-3(-/-) and L1cam(-/y) ;Ednrb(sl/sl) embryos. CONCLUSIONS & INFERENCES: We have identified the X-linked gene, L1cam, as the first modifier gene for members of the endothelin signalling pathway during development of the enteric nervous system. Mutations in L1CAM may act to modulate the severity of aganglionosis in some cases of Hirschsprung's disease.


Asunto(s)
Endotelinas/metabolismo , Sistema Nervioso Entérico/embriología , Genes Modificadores , Genes Ligados a X , Molécula L1 de Adhesión de Célula Nerviosa/genética , Receptores de Endotelina/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Endotelinas/genética , Sistema Nervioso Entérico/anomalías , Prueba de Complementación Genética , Enfermedad de Hirschsprung/embriología , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Humanos , Intestinos/embriología , Intestinos/inervación , Ratones , Ratones Noqueados , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Receptores de Endotelina/genética
20.
Neurosci Lett ; 495(2): 88-92, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21440597

RESUMEN

To investigate whether anorectal malformations (ARMs) were associated with a global neuromuscular maldevelopment of the lower gastrointestinal (GI) tract and anorectum, the distribution of neuronal markers protein gene product (PGP9.5), nitric oxide synthases (NOs), neuromuscular junction markers (synaptophysin, SYP), interstitial cells of Cajal (ICC) marker (c-kit) within the terminal rectum were analyzed by immunohistochemistry and Western blot in rat embryos with ethylenethiourea (ETU) induced ARMs. From Gestational day16 (Gd16) to Gd21, neural crest-derived cells (NCC) migrated from the proximal gut into the terminal colon, colonising it along its entire length, gradually proliferated and differentiated to innervate the distal gut. From Gd19 to Gd21, significant gross-morphological differences of the anorectum of normal (n=90) and ARMs (n=90) embryos were found. Different myenteric plexus (MPs) development of the anorectum suggested that ARMs were associated with a global abnormal innervation patterns in the anorectum in gestational course and might have some postoperative effect.


Asunto(s)
Canal Anal/inervación , Sistema Nervioso Entérico/anomalías , Recto/inervación , Canal Anal/anomalías , Canal Anal/embriología , Animales , Malformaciones Anorrectales , Ano Imperforado/inducido químicamente , Ano Imperforado/patología , Ano Imperforado/fisiopatología , Biomarcadores/metabolismo , Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/metabolismo , Femenino , Edad Gestacional , Plexo Mientérico/anomalías , Plexo Mientérico/embriología , Plexo Mientérico/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Unión Neuromuscular/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ratas , Ratas Wistar , Recto/anomalías , Recto/embriología , Sinaptofisina/metabolismo , Ubiquitina Tiolesterasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA