Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 476: 173-188, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33839113

RESUMO

Mouse models of Spina bifida (SB) have been instrumental for identifying genes, developmental processes, and environmental factors that influence neurulation and neural tube closure. Beyond the prominent neural tube defects, other aspects of the nervous system can be affected in SB with significant changes in essential bodily functions such as urination. SB patients frequently experience bladder dysfunction and SB fetuses exhibit reduced density of bladder nerves and smooth muscle although the developmental origins of these deficits have not been determined. The Pax3 Splotch-delayed (Pax3Sp-d) mouse model of SB is one of a very few mouse SB models that survives to late stages of gestation. Through analysis of Pax3Sp-d mutants we sought to define how altered bladder innervation in SB might arise by tracing sacral neural crest (NC) development, pelvic ganglia neuronal differentiation, and assessing bladder nerve fiber density. In Pax3Sp-d/Sp-d fetal mice we observed delayed migration of Sox10+ NC-derived progenitors (NCPs), deficient pelvic ganglia neurogenesis, and reduced density of bladder wall innervation. We further combined NC-specific deletion of Pax3 with the constitutive Pax3Sp-d allele in an effort to generate viable Pax3 mutants to examine later stages of bladder innervation and postnatal bladder function. Neural crest specific deletion of a Pax3 flox allele, using a Sox10-cre driver, in combination with a constitutive Pax3Sp-d mutation produced postnatal viable offspring that exhibited altered bladder function as well as reduced bladder wall innervation and altered connectivity between accessory ganglia at the bladder neck. Combined, the results show that Pax3 plays critical roles within sacral NC that are essential for initiation of neurogenesis and differentiation of autonomic neurons within pelvic ganglia.


Assuntos
Crista Neural/inervação , Fator de Transcrição PAX3/genética , Bexiga Urinária/inervação , Animais , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Feminino , Gânglios , Masculino , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Sistema Nervoso/embriologia , Crista Neural/fisiologia , Defeitos do Tubo Neural/genética , Neurogênese , Fator de Transcrição PAX3/fisiologia , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição SOXE , Região Sacrococcígea/inervação , Disrafismo Espinal/complicações , Disrafismo Espinal/genética , Bexiga Urinária/embriologia
2.
Cell Biol Int ; 44(10): 2131-2139, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32672875

RESUMO

Multiple studies have confirmed the pro-oncogenic effects of PAX3 in an array of cancers, but its role in prostate cancer (PCa) remains largely undefined. The aim of this study is to investigate the role of PAX3 in PCa. PAX3 expression was compared between PCa tumor tissue and nontumor tissues and PCa cell lines and normal prostate epithelial cells (PNT2) by western blot analysis and immunohistochemistry staining. MTT and immunofluorescence assays were used to detect PCa cell proliferation. Flow cytometry was used to evaluate cell apoptosis in PCa. Transwell assays were used for the determination of cell migration and PCa cell invasion. PAX3 expression was higher in PCa tissues and human PCa cell lines. Moreover, PAX3 silencing inhibited the proliferation, metastasis, and epithelial-mesenchymal transition (EMT) of PCa cells, and increased the rates of apoptosis. PAX3 silencing inhibited transforming growth factor-ß (TGF-ß)/Smad signaling in PCa cells. The effects of si-PAX3 on the proliferation, apoptosis, metastasis, and EMT of PCa cells were alleviated by TGF-ß1 treatment. PAX3 silencing inhibits PCa progression through the inhibition of TGF-ß/Smad signaling. This reveals PAX3 as a novel biomarker and therapeutic target for future PCa treatments.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fator de Transcrição PAX3/fisiologia , Neoplasias da Próstata , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Idoso , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Células PC-3 , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais
3.
Dis Model Mech ; 12(11)2019 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-31636139

RESUMO

Neural tube defects (NTDs), including spina bifida and anencephaly, are among the most common birth defects worldwide, but their underlying genetic and cellular causes are not well understood. Some NTDs are preventable by supplemental folic acid. However, despite widespread use of folic acid supplements and implementation of food fortification in many countries, the protective mechanism is unclear. Pax3 mutant (splotch; Sp2H ) mice provide a model in which NTDs are preventable by folic acid and exacerbated by maternal folate deficiency. Here, we found that cell proliferation was diminished in the dorsal neuroepithelium of mutant embryos, corresponding to the region of abolished Pax3 function. This was accompanied by premature neuronal differentiation in the prospective midbrain. Contrary to previous reports, we did not find evidence that increased apoptosis could underlie failed neural tube closure in Pax3 mutant embryos, nor that inhibition of apoptosis could prevent NTDs. These findings suggest that Pax3 functions to maintain the neuroepithelium in a proliferative, undifferentiated state, allowing neurulation to proceed. NTDs in Pax3 mutants were not associated with abnormal abundance of specific folates and were not prevented by formate, a one-carbon donor to folate metabolism. Supplemental folic acid restored proliferation in the cranial neuroepithelium. This effect was mediated by enhanced progression of the cell cycle from S to G2 phase, specifically in the Pax3 mutant dorsal neuroepithelium. We propose that the cell-cycle-promoting effect of folic acid compensates for the loss of Pax3 and thereby prevents cranial NTDs.


Assuntos
Ácido Fólico/administração & dosagem , Mutação , Defeitos do Tubo Neural/etiologia , Fator de Transcrição PAX3/genética , Animais , Apoptose , Ciclo Celular/efeitos dos fármacos , Suplementos Nutricionais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos CBA , Defeitos do Tubo Neural/prevenção & controle , Fator de Transcrição PAX3/fisiologia
4.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30561639

RESUMO

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Assuntos
Cóclea/embriologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Adulto , Animais , Criança , Cóclea/metabolismo , Cóclea/fisiologia , Desenvolvimento Embrionário , Feminino , Células Ciliadas Auditivas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Organogênese , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/fisiologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia , Transdução de Sinais/genética , Sequenciamento Completo do Genoma
5.
Dev Biol ; 444 Suppl 1: S202-S208, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29522707

RESUMO

The neural crest is a multipotent population of cells that originates a variety of cell types. Many animal models are used to study neural crest induction, migration and differentiation, with amphibians and birds being the most widely used systems. A major technological advance to study neural crest development in mouse, chick and zebrafish has been the generation of transgenic animals in which neural crest specific enhancers/promoters drive the expression of either fluorescent proteins for use as lineage tracers, or modified genes for use in functional studies. Unfortunately, no such transgenic animals currently exist for the amphibians Xenopus laevis and tropicalis, key model systems for studying neural crest development. Here we describe the generation and characterization of two transgenic Xenopus laevis lines, Pax3-GFP and Sox10-GFP, in which GFP is expressed in the pre-migratory and migratory neural crest, respectively. We show that Pax3-GFP could be a powerful tool to study neural crest induction, whereas Sox10-GFP could be used in the study of neural crest migration in living embryos.


Assuntos
Crista Neural/metabolismo , Fator de Transcrição PAX3/metabolismo , Fatores de Transcrição SOXE/metabolismo , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento/genética , Engenharia Genética/métodos , Proteínas de Fluorescência Verde , Humanos , Crista Neural/embriologia , Crista Neural/fisiologia , Neurogênese , Fator de Transcrição PAX3/fisiologia , Fatores de Transcrição SOXE/fisiologia , Xenopus laevis/embriologia
6.
Sci Rep ; 8(1): 3325, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29463853

RESUMO

Fractures are a common comorbidity in children with the neural tube defect (NTD) spina bifida. Mutations in the Wnt/planar cell polarity (PCP) pathway contribute to NTDs in humans and mice, but whether this pathway independently determines bone mass is poorly understood. Here, we first confirmed that core Wnt/PCP components are expressed in osteoblasts and osteoclasts in vitro. In vivo, we performed detailed µCT comparisons of bone structure in tibiae from young male mice heterozygous for NTD-associated mutations versus WT littermates. PCP signalling disruption caused by Vangl2 (Vangl2Lp/+) or Celsr1 (Celsr1Crsh/+) mutations significantly reduced trabecular bone mass and distal tibial cortical thickness. NTD-associated mutations in non-PCP transcription factors were also investigated. Pax3 mutation (Pax3Sp2H/+) had minimal effects on bone mass. Zic2 mutation (Zic2Ku/+) significantly altered the position of the tibia/fibula junction and diminished cortical bone in the proximal tibia. Beyond these genes, we bioinformatically documented the known extent of shared genetic networks between NTDs and bone properties. 46 genes involved in neural tube closure are annotated with bone-related ontologies. These findings document shared genetic networks between spina bifida risk and bone structure, including PCP components and Zic2. Genetic variants which predispose to spina bifida may therefore independently diminish bone mass.


Assuntos
Osso e Ossos/patologia , Polaridade Celular , Mutação , Proteínas do Tecido Nervoso/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Disrafismo Espinal/patologia , Fatores de Transcrição/fisiologia , Animais , Osso e Ossos/metabolismo , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Defeitos do Tubo Neural/genética , Fator de Transcrição PAX3/fisiologia , Disrafismo Espinal/genética , Disrafismo Espinal/metabolismo
7.
Dev Biol ; 432(1): 24-33, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28625870

RESUMO

Transcription factors are key orchestrators of the emergence of neuronal diversity within the developing spinal cord. As such, the two paralogous proteins Pax3 and Pax7 regulate the specification of progenitor cells within the intermediate neural tube, by defining a neat segregation between those fated to form motor circuits and those involved in the integration of sensory inputs. To attain insights into the molecular means by which they control this process, we have performed detailed phenotypic analyses of the intermediate spinal interneurons (IN), namely the dI6, V0D, V0VCG and V1 populations in compound null mutants for Pax3 and Pax7. This has revealed that the levels of Pax3/7 proteins determine both the dorso-ventral extent and the number of cells produced in each subpopulation; with increasing levels leading to the dorsalisation of their fate. Furthermore, thanks to the examination of mutants in which Pax3 transcriptional activity is skewed either towards repression or activation, we demonstrate that this cell diversification process is mainly dictated by Pax3/7 ability to repress gene expression. Consistently, we show that Pax3 and Pax7 inhibit the expression of Dbx1 and of its repressor Prdm12, fate determinants of the V0 and V1 interneurons, respectively. Notably, we provide evidence for the activity of several cis-regulatory modules of Dbx1 to be sensitive to Pax3 and Pax7 transcriptional activity levels. Altogether, our study provides insights into how the redundancy within a TF family, together with discrete dynamics of expression profiles of each member, are exploited to generate cellular diversity. Furthermore, our data supports the model whereby cell fate choices in the neural tube do not rely on binary decisions but rather on inhibition of multiple alternative fates.


Assuntos
Proteínas de Homeodomínio/fisiologia , Interneurônios/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fator de Transcrição PAX3/fisiologia , Fator de Transcrição PAX7/fisiologia , Medula Espinal/citologia , Animais , Diferenciação Celular/fisiologia , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Camundongos , Tubo Neural/fisiologia , Medula Espinal/embriologia , Células-Tronco/citologia , Células-Tronco/fisiologia
8.
Exp Anim ; 66(3): 245-250, 2017 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-28381738

RESUMO

Mice with dominant white spotting occurred spontaneously in the C3.NSY-(D11Mit74-D11Mit229) strain. Linkage analysis indicated that the locus for white spotting was located in the vicinity of the Pax3 gene on chromosome 1. Crosses of white-spotted mice showed that homozygosity for the mutation caused tail and limb abnormalities and embryonic lethality as a result of exencephaly; these phenotypes were analogous to those found in other Pax3 mutants. Sequence analysis identified a missense point mutation (c.101G>A) in exon 2 of Pax3 that resulted in a methionine to isoleucine conversion at amino acid 62 of the PAX3 protein. This mutation site was located in the N-terminal HTH (helix-turn-helix) motif of the paired domain of Pax3, which is necessary for binding to DNA and is highly conserved in vertebrate species. Alteration of DNA binding affinity was responsible for embryonic lethality in homozygotes and white spotting in heterozygotes. We named the mutant allele as Pax3Sp-Nag. The C3H/HeN-Pax3Sp-Nag strain may be useful for analyzing the function of Pax3 as a new model of the human disease, Waardenburg Syndrome.


Assuntos
Mutação de Sentido Incorreto , Fator de Transcrição PAX3/genética , Mutação Puntual , Domínios Proteicos/genética , Síndrome de Waardenburg/genética , Alelos , Sequência de Aminoácidos/genética , Animais , DNA/metabolismo , Modelos Animais de Doenças , Sequências Hélice-Volta-Hélice/genética , Humanos , Isoleucina , Metionina , Camundongos Endogâmicos , Fator de Transcrição PAX3/química , Fator de Transcrição PAX3/metabolismo , Fator de Transcrição PAX3/fisiologia , Ligação Proteica
9.
Proc Natl Acad Sci U S A ; 113(32): 9015-20, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27450084

RESUMO

Cytosolic and organelle-based heat-shock protein (HSP) chaperones ensure proper folding and function of nascent and injured polypeptides to support cell growth. Under conditions of cellular stress, including oncogenic transformation, proteostasis components maintain homeostasis and prevent apoptosis. Although this cancer-relevant function has provided a rationale for therapeutically targeting proteostasis regulators (e.g., HSP90), cancer-subtype dependencies upon particular proteostasis components are relatively undefined. Here, we show that human rhabdomyosarcoma (RMS) cells, but not several other cancer cell types, depend upon heat-shock protein 70 kDA (HSP70) for survival. HSP70-targeted therapy (but not chemotherapeutic agents) promoted apoptosis in RMS cells by triggering an unfolded protein response (UPR) that induced PRKR-like endoplasmic reticulum kinase (PERK)-eukaryotic translation initiation factor α (eIF2α)-CEBP homologous protein (CHOP) signaling and CHOP-mediated cell death. Intriguingly, inhibition of only cytosolic HSP70 induced the UPR, suggesting that the essential activity of HSP70 in RMS cells lies at the endoplasmic reticulum-cytosol interface. We also found that increased CHOP mRNA in clinical specimens was a biomarker for poor outcomes in chemotherapy-treated RMS patients. The data suggest that, like human epidermal growth factor receptor 2 (HER2) amplification in breast cancer, increased CHOP in RMS is a biomarker of decreased response to chemotherapy but enhanced response to targeted therapy. Our findings identify the cytosolic HSP70-UPR axis as an unexpected regulator of RMS pathogenesis, revealing HSP70-targeted therapy as a promising strategy to engage CHOP-mediated apoptosis and improve RMS treatment. Our study highlights the utility of dissecting cancer subtype-specific dependencies on proteostasis networks to uncover unanticipated cancer vulnerabilities.


Assuntos
Proteínas de Choque Térmico HSP70/fisiologia , Rabdomiossarcoma/etiologia , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Humanos , Fator de Transcrição PAX3/fisiologia , Rabdomiossarcoma/tratamento farmacológico , Rabdomiossarcoma/patologia , Fator de Transcrição CHOP/fisiologia , Resposta a Proteínas não Dobradas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...