Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 412
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 42(1): e111139, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36382711

RESUMO

Hirschsprung disease (HSCR), one of several neurocristopathies in children, is characterized by nerve loss in the large intestine and is mainly treated by surgery, which causes severe complications. Enteric neural crest-derived cell (ENCC) transplantation is a potential therapeutic strategy; however, so far with poor efficacy. Here, we assessed whether and how fecal microbiota transplantation (FMT) could improve ENCC transplantation in a rat model of hypoganglionosis; a condition similar to HSCR, with less intestinal innervation. We found that the hypoganglionosis intestinal microenvironment negatively influenced the ENCC functional phenotype in vitro and in vivo. Combining 16S rDNA sequencing and targeted mass spectrometry revealed microbial dysbiosis and reduced short-chain fatty acid (SCFA) production in the hypoganglionic gut. FMT increased the abundance of Bacteroides and Clostridium, SCFA production, and improved outcomes following ENCC transplantation. SCFAs alone stimulated ENCC proliferation, migration, and supported ENCC transplantation. Transcriptome-wide mRNA sequencing identified MAPK signaling as the top differentially regulated pathway in response to SCFA exposure, and inhibition of MEK1/2 signaling abrogated the SCFA-mediated effects on ENCC. This study demonstrates that FMT improves cell therapy for hypoganglionosis via short-chain fatty acid metabolism-induced MEK1/2 signaling.


Assuntos
Transplante de Microbiota Fecal , Doença de Hirschsprung , Ratos , Animais , Doença de Hirschsprung/terapia , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Transdução de Sinais , Ácidos Graxos Voláteis/metabolismo , Terapia Baseada em Transplante de Células e Tecidos
2.
Development ; 149(21)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36300492

RESUMO

The enteric nervous system is a vast intrinsic network of neurons and glia within the gastrointestinal tract and is largely derived from enteric neural crest cells (ENCCs) that emigrate into the gut during vertebrate embryonic development. Study of ENCC migration dynamics and their genetic regulators provides great insights into fundamentals of collective cell migration and nervous system formation, and these are pertinent subjects for study due to their relevance to the human congenital disease Hirschsprung disease (HSCR). For the first time, we performed in toto gut imaging and single-cell generation tracing of ENCC migration in wild type and a novel ret heterozygous background zebrafish (retwmr1/+) to gain insight into ENCC dynamics in vivo. We observed that retwmr1/+ zebrafish produced fewer ENCCs localized along the gut, and these ENCCs failed to reach the hindgut, resulting in HSCR-like phenotypes. Specifically, we observed a proliferation-dependent migration mechanism, where cell divisions were associated with inter-cell distances and migration speed. Lastly, we detected a premature neuronal differentiation gene expression signature in retwmr1/+ ENCCs. These results suggest that Ret signaling may regulate maintenance of a stem state in ENCCs.


Assuntos
Sistema Nervoso Entérico , Doença de Hirschsprung , Animais , Humanos , Divisão Celular , Movimento Celular/genética , Proliferação de Células , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Crista Neural , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Peixe-Zebra/genética , Intestinos
3.
Cell ; 139(4): 802-13, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19914172

RESUMO

The kinesin superfamily proteins (KIFs) are motor proteins that transport organelles and protein complexes in a microtubule- and ATP-dependent manner. We identified KIF26A as a new member of the murine KIFs. KIF26A is a rather atypical member as it lacks ATPase activity. Mice with a homozygous deletion of Kif26a developed a megacolon with enteric nerve hyperplasia. Kif26a-/- enteric neurons showed hypersensitivity for GDNF-Ret signaling, and we find that KIF26A suppressed GDNF-Ret signaling by direct binding and inhibition of Grb2, an essential component of GDNF/Akt/ERK signaling. We therefore propose that the unconventional kinesin KIF26A plays a key role in enteric nervous system development by repressing a cell growth signaling pathway.


Assuntos
Sistema Nervoso Entérico/embriologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Doença de Hirschsprung/metabolismo , Cinesinas/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais , Animais , Processos de Crescimento Celular , Linhagem Celular , Colo/citologia , Colo/embriologia , Colo/inervação , Proteína Adaptadora GRB2/metabolismo , Cinesinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neurônios/metabolismo
4.
BMC Pediatr ; 24(1): 189, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38493096

RESUMO

BACKGROUND: HSCR is a complex genetic disorder characterized by the absence of ganglion cells in the intestine, leading to a functional obstruction. It is due to a disruption of complex signaling pathways within the gene regulatory network (GRN) during the development of the enteric nervous system (ENS), including SRY-Box Transcription Factor 10 (SOX10) and REarranged during Transfection (RET). This study evaluated the expressions of SOX10 and RET in HSCR patients in Indonesia. METHODS: Total RNA of 19 HSCR ganglionic and aganglionic colons and 16 control colons were analyzed using quantitative real-time polymerase chain reaction for SOX10 and RET with GAPDH as the reference gene. Livak's method (2-ΔΔCT) was used to determine the expression levels of SOX10 and RET. RESULTS: Most patients were males (68.4%), in the short aganglionosis segment (78.9%), and had undergone transanal endorectal pull-through (36.6%). There were significant upregulated SOX10 expressions in both ganglionic (2.84-fold) and aganglionic (3.72-fold) colon of HSCR patients compared to controls' colon (ΔCT 5.21 ± 2.04 vs. 6.71 ± 1.90; p = 0.032; and ΔCT 4.82 ± 1.59 vs. 6.71 ± 1.90; p = 0.003; respectively). Interestingly, the RET expressions were significantly downregulated in both ganglionic (11.71-fold) and aganglionic (29.96-fold) colon of HSCR patients compared to controls' colon (ΔCT 12.54 ± 2.21 vs. 8.99 ± 3.13; p = 0.0004; and ΔCT 13.90 ± 2.64 vs. 8.99 ± 3.13; p = 0.0001; respectively). CONCLUSIONS: Our study shows aberrant SOX10 and RET expressions in HSCR patients, implying the critical role of SOX10 and RET in the pathogenesis of HSCR, particularly in the Indonesian population. Our study further confirms the involvement of SOX10-RET within the GNR during the ENS development.


Assuntos
Doença de Hirschsprung , Masculino , Humanos , Feminino , Doença de Hirschsprung/metabolismo , Transdução de Sinais , Indonésia , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Fatores de Transcrição SOXE/genética
5.
J Cell Mol Med ; 27(2): 287-298, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36606638

RESUMO

The aganglionic bowel in short-segment Hirschsprung's disease is characterized both by the absence of enteric ganglia and the presence of extrinsic thickened nerve bundles (TNBs). The relationship between the TNBs and the loss of enteric ganglia is unknown. Previous studies have described decreasing numbers of ganglia with increasing density of TNBs within the transition zone (TZ) between ganglionic and aganglionic gut, and there is some evidence of spatial contact between them in this region. To determine the cellular interactions involved, we have analysed the expression of perineurial markers of TNBs and enteric ganglionic markers for both neural cells and their ensheathing telocytes across four cranio-caudal segments consisting of most proximal ganglionic to most distal aganglionic from pull-through resected colon. We show that in the TZ, enteric ganglia are abnormal, being surrounded by perineurium cells characteristic of TNBs. Furthermore, short processes of ganglionic neurons extend caudally towards the aganglionic region, where telocytes in the TNB are located between the perineurium and nerve fibres into which they project telopodes. Thus, enteric ganglia within the TZ have abnormal structural characteristics, the cellular relationships of which are shared by the TNBs. These findings will help towards elucidation of the cellular mechanisms involved in the aetiology of Hirschsprung's disease.


Assuntos
Doença de Hirschsprung , Humanos , Lactente , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Colo/metabolismo , Gânglios/metabolismo , Fibras Nervosas , Nervos Periféricos/metabolismo
6.
Gastroenterology ; 162(1): 179-192.e11, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34425092

RESUMO

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.


Assuntos
Caderinas/metabolismo , Movimento Celular , Sistema Nervoso Entérico/metabolismo , Doença de Hirschsprung/metabolismo , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Fatores de Transcrição SOXE/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patologia , Animais , Caderinas/genética , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Cultura Embrionária , Sistema Nervoso Entérico/anormalidades , Regulação da Expressão Gênica no Desenvolvimento , Doença de Hirschsprung/genética , Doença de Hirschsprung/patologia , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Crista Neural/anormalidades , Células-Tronco Neurais/patologia , Ligação Proteica , Fatores de Transcrição SOXE/genética , Transdução de Sinais , Fatores de Tempo
7.
Development ; 147(21)2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32994173

RESUMO

Appropriately balanced RET signaling is of crucial importance during embryonic neural crest cell migration, proliferation and differentiation. RET deficiency, for example, leads to intestinal aganglionosis (Hirschsprung disease), whereas overactive RET can lead to multiple endocrine neoplasia (MEN) syndromes. Some RET mutations are associated with both intestinal aganglionosis and MEN-associated tumors. This seemingly paradoxical occurrence has led to speculation of a 'Janus mutation' in RET that causes overactivation or impairment of RET activity depending on the cellular context. Using an intestinal catenary culture system to test the effects of GDNF-mediated RET activation, we demonstrate the concurrent development of distal colonic aganglionosis and intestinal ganglioneuromas. Interestingly, the tumors induced by GDNF stimulation contain enteric neuronal progenitors capable of reconstituting an enteric nervous system when transplanted into a normal developmental environment. These results suggest that a Janus mutation may not be required to explain co-existing Hirschsprung disease and MEN-associated tumors, but rather that RET overstimulation alone is enough to cause both phenotypes. The results also suggest that reprogramming tumor cells toward non-pathological fates may represent a possible therapeutic avenue for MEN-associated neoplasms.


Assuntos
Ganglioneuroma/patologia , Doença de Hirschsprung/patologia , Intestinos/patologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Agregação Celular , Diferenciação Celular , Embrião de Galinha , Galinhas , Sistema Nervoso Entérico/patologia , Ganglioneuroma/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Doença de Hirschsprung/metabolismo , Camundongos Endogâmicos C57BL , Crista Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Nervo Vago/patologia
8.
Pediatr Res ; 94(6): 1935-1941, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37460708

RESUMO

BACKGROUND: Hirschsprung disease (HSCR) is a congenital intestinal malformation. Previous HSCR animal model needs invasive operation on adult animal. The aim of this study is to establish an early-onset animal model which is consistent with the clinical manifestation of HSCR patients. METHODS: The neonatal mice were randomly divided into the benzalkonium chloride (BAC) group, treated with BAC via enema, and the control group, treated with saline. Weight changes, excretion time of carmine, CT scan, hematoxylin-eosin staining and immunofluorescence staining were used to evaluate the effect of the model. Differentially expressed genes (DEGs) in the HSCR mice were analyzed by using DAVID 6.8 database and compared with DEGs from HSCR patients. RESULTS: The weight of mice was lower and the excretion time of carmine was longer in the BAC group. Moreover, distal colon stenosis and proximal colon enlargement appeared in the BAC group. Neurons in the distal colon decreased significantly after 4 weeks of BAC treatment and almost disappeared completely after 12 weeks. Transcriptome profiling of the mouse model and HSCR patients is similar in terms of altered gene expression. CONCLUSIONS: An economical and reliable HSCR animal model which has similar clinical characteristics to HSCR patients was successfully established. IMPACT: The animal model of Hirschsprung disease was first established in BALB/c mice. This model is an animal model of early-onset HSCR that is easy to operate and consistent with clinical manifestations. Transcriptome profiling of the mouse model and HSCR patients is similar in terms of altered gene expression.


Assuntos
Doença de Hirschsprung , Humanos , Camundongos , Animais , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Carmim , Intestinos , Modelos Animais de Doenças
9.
Mol Cell Proteomics ; 20: 100007, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33561610

RESUMO

Hirschsprung disease (HSCR) is a heterogeneous group of neurocristopathy characterized by the absence of the enteric ganglia along a variable length of the intestine. Genetic defects play a major role in the pathogenesis of HSCR, whereas family studies of pathogenic variants in all the known genes (loci) only demonstrate incomplete penetrance and variable expressivity for unknown reasons. Here, we applied large-scale, quantitative proteomics of human colon tissues from 21 patients using isobaric tags for relative and absolute quantification. method followed by bioinformatics analysis. Selected findings were confirmed by parallel reaction monitoring verification. At last, the interesting differentially expressed proteins were confirmed by Western blot. A total of 5341 proteins in human colon tissues were identified. Among them, 664 proteins with >1.2-fold difference were identified in six groups: groups A1 and A2 pooled protein from the ganglionic and aganglionic colon of male, long-segment HSCR patients (n = 7); groups B1 and B2 pooled protein from the ganglionic and aganglionic colon of male, short-segment HSCR patients (n = 7); and groups C1 and C2 pooled protein from the ganglionic and aganglionic colon of female, short-segment HSCR patients (n = 7). Based on these analyses, 49 proteins from five pathways were selected for parallel reaction monitoring verification, including ribosome, endocytosis, spliceosome, oxidative phosphorylation, and cell adhesion. The downregulation of three neuron projection development genes ARF4, KIF5B, and RAB8A in the aganglionic part of the colon was verified in 15 paired colon samples using Western blot. The findings of this study will shed new light on the pathogenesis of HSCR and facilitate the development of therapeutic targets.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Colo/metabolismo , Doença de Hirschsprung/metabolismo , Cinesinas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Fatores de Ribosilação do ADP/genética , Regulação para Baixo , Feminino , Doença de Hirschsprung/genética , Humanos , Lactente , Cinesinas/genética , Masculino , Neurônios , Proteômica , Proteínas rab de Ligação ao GTP/genética
10.
PLoS Genet ; 16(9): e1009008, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32898154

RESUMO

Hirschsprung disease (HSCR) is a complex genetic disorder of neural crest development resulting in incomplete formation of the enteric nervous system (ENS). This life-threatening neurocristopathy affects 1/5000 live births, with a currently unexplained male-biased ratio. To address this lack of knowledge, we took advantage of the TashT mutant mouse line, which is the only HSCR model to display a robust male bias. Our prior work revealed that the TashT insertional mutation perturbs a Chr.10 silencer-enriched non-coding region, leading to transcriptional dysregulation of hundreds of genes in neural crest-derived ENS progenitors of both sexes. Here, through sex-stratified transcriptome analyses and targeted overexpression in ENS progenitors, we show that male-biased ENS malformation in TashT embryos is not due to upregulation of Sry-the murine ortholog of a candidate gene for the HSCR male bias in humans-but instead involves upregulation of another Y-linked gene, Ddx3y. This discovery might be clinically relevant since we further found that the DDX3Y protein is also expressed in the ENS of a subset of male HSCR patients. Mechanistically, other data including chromosome conformation captured-based assays and CRISPR/Cas9-mediated deletions suggest that Ddx3y upregulation in male TashT ENS progenitors is due to increased transactivation by p53, which appears especially active in these cells yet without triggering apoptosis. Accordingly, in utero treatment of TashT embryos with the p53 inhibitor pifithrin-α decreased Ddx3y expression and abolished the otherwise more severe ENS defect in TashT males. Our data thus highlight novel pathogenic roles for p53 and DDX3Y during ENS formation in mice, a finding that might help to explain the intriguing male bias of HSCR in humans.


Assuntos
RNA Helicases DEAD-box/genética , Doença de Hirschsprung/genética , Antígenos de Histocompatibilidade Menor/genética , Animais , RNA Helicases DEAD-box/metabolismo , Modelos Animais de Doenças , Sistema Nervoso Entérico/metabolismo , Feminino , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Doença de Hirschsprung/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Camundongos , Antígenos de Histocompatibilidade Menor/metabolismo , Mutagênese Insercional , Mutação , Crista Neural/metabolismo , Fatores Sexuais , Ativação Transcricional/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
11.
Altern Ther Health Med ; 29(6): 364-369, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37384399

RESUMO

Objective: To investigate the expression and significance of intestinal Cathepsin D (CAD) and sex-determining region Y-frame protein 2 (SOX2) in children with Hirschsprung's disease (HD) after surgery. Methods: Immunohistochemistry and Western blot techniques were employed to examine the expression of CAD and SOX2 in colonic tissues obtained from 56 children with HD (HD group) and 23 colonic tissues obtained from fistulas for intestinal obstruction or perforation (control group). Pearson linear correlation analysis was conducted to analyze the relationship between CAD and SOX2 expression, the diameter of the intermuscular plexus, and the number of ganglion cells in the diseased intestinal segment. Results: The positive expression rates of CAD protein and SOX2 protein in the intestinal tissues of children with HD were lower than those in the control group (P < .05). Furthermore, the positive expression rates of CAD protein and SOX2 protein in the narrow intestinal tissue of HD children were lower than those in the transitional colon tissue (P < .05). The diameter of the intramuscular plexus and the number of ganglion cells in the intestinal tissue of the stenosis and transitional segments in HD children were lower than those in the control group (P < .05). There was a significant positive correlation between the diameter of the intermuscular plexus and the number of ganglion cells in the intestinal tissue of HD children and the expression intensity of CAD protein and SOX2 protein (P < .05). Conclusions: The down-regulated expression intensity of CAD protein and SOX2 protein in the diseased colon of children with HD may be associated with a decrease in the diameter of the intermuscular plexus and the number of ganglion cells.


Assuntos
Doença de Hirschsprung , Criança , Humanos , Lactente , Doença de Hirschsprung/cirurgia , Doença de Hirschsprung/metabolismo , Catepsina D , Imuno-Histoquímica , Fatores de Transcrição SOXB1
12.
Pediatr Surg Int ; 39(1): 126, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36790471

RESUMO

BACKGROUND: METTL3, an mRNA m6A methyltransferase, has been implicated in various steps of mRNA metabolism, such as stabilization, splicing, nuclear transportation, translation, and degradation. However, whether METTL3 dysregulation is involved in Hirschsprung disease (HSCR) development remains unclear. In this study, we preliminarily elucidated the role of METTL3 in HSCR and sought to identify the associated molecular mechanism. METHODS: The gene expression levels of YAP and several methyltransferases, demethylases, and effectors were evaluated by RT-qPCR. Protein levels were evaluated by western blot and immunohistochemistry. Cell proliferation and migration were detected by CCK-8 and Transwell assays, respectively. The overall levels of m6A modification were determined by colorimetry. RESULTS: We found that m6A levels were reduced in the stenotic intestinal tissue of patients with HSCR. When METTL3 was knocked down in SH-SY5Y and HEK-293T cells, the proliferative and migratory abilities of the cells were inhibited, m6A modification levels were reduced, and YAP expression was increased. Importantly, YAP and METTL3 expression displayed a negative correlation in both cell lines as well as in HSCR tissue. CONCLUSIONS: Our results provide evidence for an interaction between METTL3 and YAP in HSCR, and further suggest that METTL3 is involved in the pathogenesis of HSCR by regulating neural crest cell proliferation and migration upstream of YAP.


Assuntos
Doença de Hirschsprung , Neuroblastoma , Humanos , Proliferação de Células/genética , Doença de Hirschsprung/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , RNA Mensageiro/metabolismo
13.
Pediatr Surg Int ; 39(1): 214, 2023 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-37278766

RESUMO

BACKGROUND: Actin Alpha 2 (ACTA2) is expressed in intestinal smooth muscle cells (iSMCs) and is associated with contractility. Hirschsprung disease (HSCR), one of the most common digested tract malformations, shows peristaltic dysfunction and spasm smooth muscles. The arrangement of the circular and longitudinal smooth muscle (SM) of the aganglionic segments is disorganized. Does ACTA2, as a marker of iSMCs, exhibit abnormal expression in aganglionic segments? Does the ACTA2 expression level affect the contraction function of iSMCs? What are the spatiotemporal expression trends of ACTA2 during different developmental stages of the colon? METHODS: Immunohistochemical staining was used to detect the expression of ACTA2 in iSMCs of children with HSCR and Ednrb-/- mice, and the small interfering RNAs (siRNAs) knockdown technique was employed to investigate how Acta2 affected the systolic function of iSMCs. Additionally, Ednrb-/- mice were used to explore the changes in the expression level of iSMCs ACTA2 at different developmental stages. RESULTS: The expression of ACTA2 is higher in circular SM in the aganglionic segments of HSCR patients and Ednrb-/- mice than in normal control children and mice. Down regulation of Acta2 weakens the contraction ability of intestinal smooth muscle cells. Abnormally elevated expression of ACTA2 of circular smooth muscle occurs since embryonic day 15.5 (E15.5d) in aganglionic segments of Ednrb-/- mice. CONCLUSIONS: Abnormally elevated expression of ACTA2 in the circular SM leads to hyperactive contraction, which may cause the spasm of aganglionic segments in HSCR.


Assuntos
Actinas , Doença de Hirschsprung , Camundongos , Animais , Actinas/genética , Actinas/metabolismo , Doença de Hirschsprung/metabolismo , Colo/metabolismo , Músculo Liso/metabolismo , Regulação para Baixo
14.
Pediatr Surg Int ; 39(1): 188, 2023 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-37101012

RESUMO

Interstitial cells of Cajal (ICCs) are pacemaker cells of gastrointestinal motility that generate and transmit electrical slow waves to smooth muscle cells in the gut wall, thus inducing phasic contractions and coordinated peristalsis. Traditionally, tyrosine-protein kinase Kit (c-kit), also known as CD117 or mast/stem cell growth factor receptor, has been used as the primary marker of ICCs in pathology specimens. More recently, the Ca2+-activated chloride channel, anoctamin-1, has been introduced as a more specific marker of ICCs. Over the years, various gastrointestinal motility disorders have been described in infants and young children in which symptoms of functional bowel obstruction arise from ICC-related neuromuscular dysfunction of the colon and rectum. The current article provides a comprehensive overview of the embryonic origin, distribution, and functions of ICCs, while also illustrating the absence or deficiency of ICCs in pediatric patients with Hirschsprung disease intestinal neuronal dysplasia, isolated hypoganglionosis, internal anal sphincter achalasia, and congenital smooth muscle cell disorders such as megacystis microcolon intestinal hypoperistalsis syndrome.


Assuntos
Doença de Hirschsprung , Células Intersticiais de Cajal , Lactente , Criança , Humanos , Pré-Escolar , Células Intersticiais de Cajal/metabolismo , Relevância Clínica , Doença de Hirschsprung/metabolismo , Motilidade Gastrointestinal/fisiologia , Canal Anal/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
15.
Int J Mol Sci ; 24(7)2023 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37047673

RESUMO

Insights into the role of microRNAs (miRNAs) in disease pathogenesis have made them attractive therapeutic targets, and numerous miRNAs have been functionally linked to Hirschsprung disease (HSCR), a life-threatening genetic disorder due to defective migration, proliferation, and colonization of enteric neural crest cells (ENCCs) in the gut. Recent studies have demonstrated that miR-424 strongly inhibits migration in a variety of cell types and its potential target RICTOR is essential for neural crest cell development. We therefore sought to interrogate how miR-424 and RICTOR contribute to the pathogenesis of HSCR. We utilized HSCR cases and human neural cells to evaluate the miR-424-mediated regulation of RICTOR and the downstream AKT phosphorylation. We further developed an ex vivo model to assess the effects of miR-424 on ENCC migration and proliferation. Then, single-cell atlases of gene expression in both human and mouse fetal intestines were used to determine the characteristics of RICTOR and AKT expression in the developing gut. Our findings demonstrate that miR-424 levels are markedly increased in the colonic tissues of patients with HSCR and that it regulates human neural cell migration by directly targeting RICTOR. Up-regulation of miR-424 leads to decreased AKT phosphorylation levels in a RICTOR-dependent manner, and this, in turn, impairs ENCC proliferation and migration in the developing gut. Interestingly, we further identified prominent RICTOR and AKT expressions in the enteric neurons and other types of enteric neural cells in human and mouse fetal intestines. Our present study reveals the role of the miR-424/RICTOR axis in HSCR pathogenesis and indicates that miR-424 is a promising candidate for the development of targeted therapies against HSCR.


Assuntos
Sistema Nervoso Entérico , Doença de Hirschsprung , MicroRNAs , Camundongos , Animais , Humanos , Doença de Hirschsprung/metabolismo , Crista Neural/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima , Fosforilação , Movimento Celular/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fatores de Transcrição/metabolismo , Sistema Nervoso Entérico/metabolismo
16.
J Biol Chem ; 296: 100389, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33561442

RESUMO

The c-RET proto-oncogene encodes a receptor-tyrosine kinase. Loss-of-function mutations of RET have been shown to be associated with Hirschsprung disease and Down's syndrome (HSCR-DS) in humans. DS is known to involve cerebellar hypoplasia, which is characterized by reduced cerebellar size. Despite the fact that c-Ret has been shown to be associated with HSCR-DS in humans and to be expressed in Purkinje cells (PCs) in experimental animals, there is limited information about the role of activity of c-Ret/c-RET kinase in cerebellar hypoplasia. We found that a loss-of-function mutation of c-Ret Y1062 in PCs causes cerebellar hypoplasia in c-Ret mutant mice. Wild-type mice had increased phosphorylation of c-Ret in PCs during postnatal development, while c-Ret mutant mice had postnatal hypoplasia of the cerebellum with immature neurite outgrowth in PCs and granule cells (GCs). c-Ret mutant mice also showed decreased numbers of glial fibers and mitogenic sonic hedgehog (Shh)-positive vesicles in the external germinal layer of PCs. c-Ret-mediated cerebellar hypoplasia was rescued by subcutaneous injection of a smoothened agonist (SAG) as well as by reduced expression of Patched1, a negative regulator for Shh. Our results suggest that the loss-of-function mutation of c-Ret Y1062 results in the development of cerebellar hypoplasia via impairment of the Shh-mediated development of GCs and glial fibers in mice with HSCR-DS.


Assuntos
Cerebelo/anormalidades , Síndrome de Down/genética , Doença de Hirschsprung/genética , Mutação com Perda de Função , Malformações do Sistema Nervoso/genética , Proteínas Proto-Oncogênicas c-ret/genética , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Modelos Animais de Doenças , Síndrome de Down/complicações , Síndrome de Down/metabolismo , Síndrome de Down/patologia , Técnicas de Introdução de Genes/métodos , Proteínas Hedgehog/metabolismo , Doença de Hirschsprung/complicações , Doença de Hirschsprung/metabolismo , Doença de Hirschsprung/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/patologia , Neuroglia/metabolismo , Neuroglia/patologia , Fosforilação , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/patologia
17.
Stem Cells ; 39(9): 1236-1252, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33938072

RESUMO

Interplay between embryonic enteric neural stem cells (ENSCs) and enteric mesenchymal cells (EMCs) in the embryonic gut is essential for normal development of the enteric nervous system. Disruption of these interactions underlies the pathogenesis of intestinal aganglionosis in Hirschsprung disease (HSCR). ENSC therapy has been proposed as a possible treatment for HSCR, but whether the survival and development of postnatal-derived ENSCs similarly rely on signals from the mesenchymal environment is unknown and has important implications for developing protocols to expand ENSCs for cell transplantation therapy. Enteric neural crest-derived cells (ENCDCs) and EMCs were cultured from the small intestine of Wnt1-Rosa26-tdTomato mice. EMCs promoted the expansion of ENCDCs 9.5-fold by inducing ENSC properties, including expression of Nes, Sox10, Sox2, and Ngfr. EMCs enhanced the neurosphere-forming ability of ENCDCs, and this persisted after withdrawal of the EMCs. These effects were mediated by paracrine factors and several ligands known to support neural stem cells were identified in EMCs. Using the optimized expansion procedures, neurospheres were generated from small intestine of the Ednrb-/- mouse model of HSCR. These ENSCs had similar proliferative and migratory capacity to Ednrb+/+ ENSCs, albeit neurospheres contained fewer neurons. ENSCs derived from Ednrb-/- mice generated functional neurons with similar calcium responses to Ednrb+/+ ENSCs and survived after transplantation into the aganglionic colon of Ednrb-/- recipients. EMCs act as supporting cells to ENSCs postnatally via an array of synergistically acting paracrine signaling factors. These properties can be leveraged to expand autologous ENSCs from patients with HSCR mutations for therapeutic application.


Assuntos
Sistema Nervoso Entérico , Doença de Hirschsprung , Células-Tronco Neurais , Animais , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Doença de Hirschsprung/terapia , Humanos , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo
18.
Pediatr Res ; 92(3): 737-747, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34880446

RESUMO

BACKGROUND: This study aimed to identify key microRNAs (miRNAs), pathways, and target genes mediating Hirschsprung's disease (HSCR) pathogenesis and identify the diagnostic potential of miRNAs. METHODS: The Gene Expression Omnibus database and reverse transcription-quantitative PCR were used to compare miRNA expression between ganglionic and aganglionic colon tissues of children with HSCR, and the TAM 2.0 database was used to identify colon tissue-specific miRNAs. The StarBase database, TargetScan database, luciferase reporter, and western blot assays were used to analyze miRNA-messenger RNA interactions. OmicShare was used to perform functional and pathway enrichment analyses of the target genes. Migration assays were performed to validate the functions of the miRNAs. RESULTS: The TAM 2.0 database analysis and reverse transcription-quantitative PCR showed that hsa-miR-192-5p, hsa-miR-200a-3p, and hsa-miR-200b-3p were colon tissue-specific and upregulated in aganglionic colon tissue compared to paired ganglionic colon tissue. These three miRNAs effectively reduced cell viability and migration. Luciferase reporter and western blot assays verified the direct interaction between these three miRNAs and the target genes of ZEB2 and FNDC3B. Furthermore, the plasma levels of these miRNAs were higher in HSCR patients than in non-HSCR patients. CONCLUSIONS: Three plasma miRNAs (hsa-miR-192-5p, hsa-miR-200a-3p, and hsa-miR-200b-3p) are potential peripheral HSCR biomarkers. IMPACT: The molecular mechanisms underlying HSCR are unclear. HSCR is most accurately diagnosed using rectal biopsy samples, and no consensus has been reached on the use of blood-based tests for HSCR diagnosis. Circulating miRNAs may be candidate diagnostic HSCR biomarkers because they are typically easily detectable, stable, and tissue-specific. Three plasma miRNAs (miR-200a-3p, miR-192-5p, and miR-200b-3p) are potential peripheral HSCR biomarkers.


Assuntos
Doença de Hirschsprung , MicroRNAs , Biomarcadores , Criança , Doença de Hirschsprung/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro , Reação em Cadeia da Polimerase em Tempo Real
19.
BMC Pediatr ; 22(1): 216, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35443634

RESUMO

BACKGROUND: Hirschsprung disease (HSCR) is a complex genetic disorder characterized by the lack of ganglion cells in the intestines. A current study showed that the NRG1 rare variant frequency in Indonesian patients with HSCR is only 0.9%. Here, we investigated the impact of NRG1 expressions and methylation patterns on the pathogenesis of HSCR. METHODS: This cross-sectional study determined NRG1 type I (HRGα, HRGß1, HRGß2, HRGß3, HRGγ, and NDF43 isoforms), type II and type III expressions in both ganglionic and aganglionic colons of 20 patients with HSCR and 10 control colons by real-time polymerase chain reaction (qPCR). For methylation studies, we treated the extracted gDNA from 16 HSCR patients' and 17 control colons with sodium bisulfate and analyzed the methylation pattern of NRG1 exon 1 with methylation-specific PCR. The samples were collected and analyzed at our institution from December 2018 to December 2020. RESULTS: NRG1 types I, II and III expressions were upregulated (17.2-, 3.2-, and 7.2-fold, respectively) in the ganglionic colons compared with control colons (type I: 13.32 ± 1.65 vs. 17.42 ± 1.51, p < 0.01; type II: 13.73 ± 2.02 vs. 16.29 ± 2.19, p < 0.01; type III: 13.47 ± 3.01 vs. 16.32 ± 2.58, p = 0.03; respectively); while only type I (7.7-fold) and HRGß1/HRGß2 (3.3-fold) isoforms were significantly upregulated in the aganglionic colons compared to the controls (type I: 14.47 ± 1.66 vs. 17.42 ± 1.51, p < 0.01; HRGß1/HRGß2: 13.62 ± 3.42 vs 14.75 ± 1.26, p = 0.01). Moreover, the frequency of partially methylated NRG1 was higher in the ganglionic (81%) and aganglionic (75%) colons than in the controls (59%). CONCLUSIONS: Our study provides further insights into the aberrant NRG1 expression in the colons of patients with HSCR, both ganglionic and aganglionic bowel, which might contribute to the development of HSCR, particularly in Indonesia. Furthermore, these aberrant NRG1 expressions might be associated with its methylation pattern.


Assuntos
Doença de Hirschsprung , Estudos Transversais , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Humanos , Metilação , Neuregulina-1/genética , Reação em Cadeia da Polimerase em Tempo Real
20.
Pediatr Surg Int ; 38(9): 1273-1281, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35836014

RESUMO

BACKGROUND: Hirschsprung's disease (HSCR) is a common birth defect caused by dysplasia of neural crest cells in the gut. Long noncoding RNAs (lncRNAs) play an important role in cellular processes, including development and disease. Despite the known engagement of LINC00346 in several human diseases, its biological function in HSCR remains unknown. METHODS: The relative expression levels of LINC00346, miR-148a-3p and Dnmt1 in HSCR colon tissues were detected by quantitative real-time PCR. Western blot assays were conducted to investigate the Dnmt1 protein expression level. Knockdown of LINC00346 and overexpression of miR-148a-3p in SH-SY5Y and SK-N-BE(2) cell lines was conducted. Cell proliferation and migration were detected by cell counting Kit-8 assays, 5-ethynyl-2'-deoxyuridine assays and transwell assays. Cell apoptosis was verified by flow cytometric analysis. Furthermore, the competing endogenous RNA (ceRNA) activity of LINC00346 on miR-148a-5p was investigated via bioinformatics analysis and luciferase reporter assays. RESULT: Downregulation of LINC00346 and Dnmt1 was detected in HSCR tissues. Knockdown of LINC00346 and overexpression of miR-148a-3p in SK-N-BE(2) and SH-SY5Y cells inhibited cell migration and proliferation and promoted apoptosis. Moreover, the miR-148a-3p inhibitor rescued the downregulation of Dnmt1 in LINC00346 knockdown cell lines, which was evidence of the ceRNA regulatory mechanism of Dnmt1 by LINC00346. CONCLUSIONS: LINC00346 was downregulated in HSCR colon tissues and acted as a ceRNA to regulate the expression of Dnmt1 in vitro. Together, these findings indicate that LINC00346 could affect the occurrence of HSCR by participating in the development of enteric neural crest cells.


Assuntos
Doença de Hirschsprung , MicroRNAs , Neuroblastoma , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , DNA (Citosina-5-)-Metiltransferase 1 , Doença de Hirschsprung/genética , Doença de Hirschsprung/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA