Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Dev Biol ; 464(1): 11-23, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32450229

RESUMO

Development of the complex structure of the vertebrate limb requires carefully orchestrated interactions between multiple regulatory pathways and proteins. Among these, precise regulation of 5' Hox transcription factor expression is essential for proper limb bud patterning and elaboration of distinct limb skeletal elements. Here, we identified Geminin (Gmnn) as a novel regulator of this process. A conditional model of Gmnn deficiency resulted in loss or severe reduction of forelimb skeletal elements, while both the forelimb autopod and hindlimb were unaffected. 5' Hox gene expression expanded into more proximal and anterior regions of the embryonic forelimb buds in this Gmnn-deficient model. A second conditional model of Gmnn deficiency instead caused a similar but less severe reduction of hindlimb skeletal elements and hindlimb polydactyly, while not affecting the forelimb. An ectopic posterior SHH signaling center was evident in the anterior hindlimb bud of Gmnn-deficient embryos in this model. This center ectopically expressed Hoxd13, the HOXD13 target Shh, and the SHH target Ptch1, while these mutant hindlimb buds also had reduced levels of the cleaved, repressor form of GLI3, a SHH pathway antagonist. Together, this work delineates a new role for Gmnn in modulating Hox expression to pattern the vertebrate limb.


Assuntos
Embrião de Mamíferos/embriologia , Geminina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Membro Posterior/embriologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Embrião de Mamíferos/citologia , Geminina/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Membro Posterior/citologia , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Transgênicos , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Fatores de Transcrição/genética
2.
Dev Biol ; 393(1): 44-56, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24995796

RESUMO

Geminin is a nucleoprotein that can directly bind chromatin regulatory complexes to modulate gene expression during development. Geminin knockout mouse embryos are preimplantation lethal by the 32-cell stage, precluding in vivo study of Geminin's role in neural development. Therefore, here we used a conditional Geminin allele in combination with several Cre-driver lines to define an essential role for Geminin during mammalian neural tube (NT) formation and patterning. Geminin was required in the NT within a critical developmental time window (embryonic day 8.5-10.5), when NT patterning and closure occurs. Geminin excision at these stages resulted in strongly diminished expression of genes that mark and promote dorsal NT identities and decreased differentiation of ventral motor neurons, resulting in completely penetrant NT defects, while excision after embryonic day 10.5 did not result in NT defects. When Geminin was deleted specifically in the spinal NT, both NT defects and axial skeleton defects were observed, but neither defect occurred when Geminin was excised in paraxial mesenchyme, indicating a tissue autonomous requirement for Geminin in developing neuroectoderm. Despite a potential role for Geminin in cell cycle control, we found no evidence of proliferation defects or altered apoptosis. Comparisons of gene expression in the NT of Geminin mutant versus wild-type siblings at embryonic day 10.5 revealed decreased expression of key regulators of neurogenesis, including neurogenic bHLH transcription factors and dorsal interneuron progenitor markers. Together, these data demonstrate a requirement for Geminin for NT patterning and neuronal differentiation during mammalian neurulation in vivo.


Assuntos
Geminina/genética , Placa Neural/embriologia , Defeitos do Tubo Neural/genética , Tubo Neural/embriologia , Neurogênese/genética , Animais , Apoptose/genética , Proliferação de Células , Cromatina , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Tubo Neural/anormalidades , Neurulação/genética
3.
Sci Rep ; 10(1): 2864, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32071327

RESUMO

Exposure to thalidomide during a critical window of development results in limb defects in humans and non-human primates while mice and rats are refractory to these effects. Thalidomide-induced teratogenicity is dependent on its binding to cereblon (CRBN), the substrate receptor of the Cul4A-DDB1-CRBN-RBX1 E3 ubiquitin ligase complex. Thalidomide binding to CRBN elicits subsequent ubiquitination and proteasomal degradation of CRBN neosubstrates including SALL4, a transcription factor of which polymorphisms phenocopy thalidomide-induced limb defects in humans. Herein, thalidomide-induced degradation of SALL4 was examined in human induced pluripotent stem cells (hiPSCs) that were differentiated either to lateral plate mesoderm (LPM)-like cells, the developmental ontology of the limb bud, or definitive endoderm. Thalidomide and its immunomodulatory drug (IMiD) analogs, lenalidomide, and pomalidomide, dose-dependently inhibited hiPSC mesendoderm differentiation. Thalidomide- and IMiD-induced SALL4 degradation can be abrogated by CRBN V388I mutation or SALL4 G416A mutation in hiPSCs. Genetically modified hiPSCs expressing CRBN E377V/V388I mutant or SALL4 G416A mutant were insensitive to the inhibitory effects of thalidomide, lenalidomide, and pomalidomide on LPM differentiation while retaining sensitivity to another known limb teratogen, all-trans retinoic acid (atRA). Finally, disruption of LPM differentiation by atRA or thalidomide perturbed subsequent chondrogenic differentiation in vitro. The data here show that thalidomide, lenalidomide, and pomalidomide affect stem cell mesendoderm differentiation through CRBN-mediated degradation of SALL4 and highlight the utility of the LPM differentiation model for studying the teratogenicity of new CRBN modulating agents.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Deformidades Congênitas dos Membros/genética , Talidomida/farmacologia , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética , Animais , Proteínas de Transporte/genética , Proteínas Culina/genética , Proteínas de Ligação a DNA/genética , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Lenalidomida/farmacologia , Deformidades Congênitas dos Membros/induzido quimicamente , Deformidades Congênitas dos Membros/patologia , Camundongos , Complexos Multiproteicos/efeitos dos fármacos , Complexos Multiproteicos/genética , Mutação/genética , Proteólise/efeitos dos fármacos , Ratos , Talidomida/efeitos adversos
4.
Genes Cancer ; 8(9-10): 725-744, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29234490

RESUMO

Medulloblastoma is the most common malignant brain cancer of childhood. Further understanding of tumorigenic mechanisms may define new therapeutic targets. Geminin maintains genome fidelity by controlling re-initiation of DNA replication within a cell cycle. In some contexts, Geminin inhibition induces cancer-selective cell cycle arrest and apoptosis and/or sensitizes cancer cells to Topoisomerase IIα inhibitors such as etoposide, which is used in combination chemotherapies for medulloblastoma. However, Geminin's potential role in medulloblastoma tumorigenesis remained undefined. Here, we found that Geminin is highly expressed in human and mouse medulloblastomas and in murine granule neuron precursor (GNP) cells during cerebellar development. Conditional Geminin loss significantly enhanced survival in the SmoA1 mouse medulloblastoma model. Geminin loss in this model also reduced numbers of preneoplastic GNPs persisting at one postnatal month, while at two postnatal weeks these cells exhibited an elevated DNA damage response and apoptosis. Geminin knockdown likewise impaired human medulloblastoma cell growth, activating G2 checkpoint and DNA damage response pathways, triggering spontaneous apoptosis, and enhancing G2 accumulation of cells in response to etoposide treatment. Together, these data suggest preneoplastic and cancer cell-selective roles for Geminin in medulloblastoma, and suggest that targeting Geminin may impair tumor growth and enhance responsiveness to Topoisomerase IIα-directed chemotherapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA