Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
FASEB J ; 36(3): e22176, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35129866

RESUMO

To gain further insight into chromatin-mediated regulation of mammalian sex determination, we analyzed the role of the CHARGE syndrome-associated proteins FAM172A and CHD7. This study is based on our prior discoveries that a subset of corresponding mutant mice display complete male-to-female sex reversal, and that both of these proteins regulate co-transcriptional alternative splicing in neural crest cells. Here, we report that FAM172A and CHD7 are present in the developing gonads when sex determination normally occurs in mice. The interactome of FAM172A in pre-Sertoli cells again suggests a role at the chromatin-spliceosome interface, like in neural crest cells. Accordingly, analysis of Fam172a-mutant pre-Sertoli cells revealed transcriptional and splicing dysregulation of hundreds of genes. Many of these genes are similarly affected in Chd7-mutant pre-Sertoli cells, including several known key regulators of sex determination and subsequent formation of testis cords. Among them, we notably identified Sry as a direct transcriptional target and WNT pathway-associated Lef1 and Tcf7l2 as direct splicing targets. The identified molecular defects are also associated with the abnormal morphology of seminiferous tubules in mutant postnatal testes. Altogether, our results thus identify FAM172A and CHD7 as new players in the regulation of male sex determination and differentiation in mice, and further highlight the importance of chromatin-mediated regulatory mechanisms in these processes.


Assuntos
Processamento Alternativo , Síndrome CHARGE/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas/metabolismo , Processos de Determinação Sexual , Transcriptoma , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas/genética , Células de Sertoli/metabolismo , Espermatogênese , Suínos
2.
EMBO Rep ; 22(6): e50958, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33900016

RESUMO

Mutations in the chromatin remodeller-coding gene CHD7 cause CHARGE syndrome (CS). CS features include moderate to severe neurological and behavioural problems, clinically characterized by intellectual disability, attention-deficit/hyperactivity disorder and autism spectrum disorder. To investigate the poorly characterized neurobiological role of CHD7, we here generate a zebrafish chd7-/- model. chd7-/- mutants have less GABAergic neurons and exhibit a hyperactivity behavioural phenotype. The GABAergic neuron defect is at least in part due to downregulation of the CHD7 direct target gene paqr3b, and subsequent upregulation of MAPK/ERK signalling, which is also dysregulated in CHD7 mutant human cells. Through a phenotype-based screen in chd7-/- zebrafish and Caenorhabditis elegans, we show that the small molecule ephedrine restores normal levels of MAPK/ERK signalling and improves both GABAergic defects and behavioural anomalies. We conclude that chd7 promotes paqr3b expression and that this is required for normal GABAergic network development. This work provides insight into the neuropathogenesis associated with CHD7 deficiency and identifies a promising compound for further preclinical studies.


Assuntos
Transtorno do Espectro Autista , Animais , Caenorhabditis elegans , Cromatina , DNA Helicases , Proteínas de Ligação a DNA/genética , Neurônios GABAérgicos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Mutação , Peixe-Zebra
3.
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
4.
PLoS Genet ; 11(3): e1005093, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25786024

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Doença de Hirschsprung/genética , Doença de Hirschsprung/patologia , Camundongos , Mutagênese Insercional , Animais , Cromossomos de Mamíferos , Sistema Nervoso Entérico/anormalidades , Doença de Hirschsprung/embriologia , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/metabolismo , Elementos Silenciadores Transcricionais , Transcriptoma
5.
Dis Model Mech ; 9(11): 1283-1293, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27585883

RESUMO

Waardenburg syndrome is a neurocristopathy characterized by a combination of skin and hair depigmentation, and inner ear defects. In the type 4 form, these defects show comorbidity with Hirschsprung disease, a disorder marked by an absence of neural ganglia in the distal colon, triggering functional intestinal obstruction. Here, we report that the Spot mouse line - obtained through an insertional mutagenesis screen for genes involved in neural crest cell (NCC) development - is a model for Waardenburg syndrome type 4. We found that the Spot insertional mutation causes overexpression of an overlapping gene pair composed of the transcription-factor-encoding Nr2f1 and the antisense long non-coding RNA A830082K12Rik in NCCs through a mechanism involving relief of repression of these genes. Consistent with the previously described role of Nr2f1 in promoting gliogenesis in the central nervous system, we further found that NCC-derived progenitors of the enteric nervous system fail to fully colonize Spot embryonic guts owing to their premature differentiation in glial cells. Taken together, our data thus identify silencer elements of the Nr2f1-A830082K12Rik gene pair as new candidate loci for Waardenburg syndrome type 4.


Assuntos
Fator I de Transcrição COUP/metabolismo , Doença de Hirschsprung/genética , Crista Neural/metabolismo , Crista Neural/patologia , RNA Longo não Codificante/metabolismo , Regulação para Cima/genética , Síndrome de Waardenburg/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Diferenciação Celular/genética , Endolinfa/metabolismo , Sistema Nervoso Entérico/patologia , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos , Camundongos Mutantes , Mutagênese Insercional , Neuroglia/metabolismo , Neuroglia/patologia , Fenótipo , Pigmentação/genética , RNA Longo não Codificante/genética , Transgenes
6.
J Vis Exp ; (79): e50709, 2013 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-24084298

RESUMO

Neural crest cells (NCC) are a transient and multipotent cell population that originates from the dorsal neural tube and migrates extensively throughout the developing vertebrate embryo. In addition to providing peripheral glia and neurons, NCC generate melanocytes as well as most of the cranio-facial skeleton. NCC migration and differentiation is controlled by a combination of their axial origin along the neural tube and their exposure to regionally distinct extracellular cues. Such contribution of extracellular ligands is especially evident during the formation of the enteric nervous system (ENS), a complex interconnected network of neural ganglia that locally controls (among other things) gut muscle movement and intestinal motility. Most of the ENS is derived from a small initial pool of NCC that undertake a long journey in order to colonize - in a rostral to caudal fashion - the entire length of the prospective gut. Among several signaling pathways known to influence enteric NCC colonization, GDNF/RET signaling is recognized as the most important. Indeed, spatiotemporally controlled secretion of the RET ligand GDNF by the gut mesenchyme is chiefly responsible for the attraction and guidance of RET-expressing enteric NCC to and within the embryonic gut. Here, we describe an ex vivo cell migration assay, making use of a transgenic mouse line possessing fluorescently labeled NCC, which allows precise quantification of enteric NCC migration potential in the presence of various growth factors, including GDNF.


Assuntos
Movimento Celular/fisiologia , Sistema Nervoso Entérico/citologia , Crista Neural/citologia , Células-Tronco Neurais/citologia , Animais , Sistema Nervoso Entérico/embriologia , Feminino , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Crista Neural/embriologia , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA