Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Genes Dev ; 32(23-24): 1537-1549, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30463901

RESUMO

Human globin gene production transcriptionally "switches" from fetal to adult synthesis shortly after birth and is controlled by macromolecular complexes that enhance or suppress transcription by cis elements scattered throughout the locus. The DRED (direct repeat erythroid-definitive) repressor is recruited to the ε-globin and γ-globin promoters by the orphan nuclear receptors TR2 (NR2C1) and TR4 (NR2C2) to engender their silencing in adult erythroid cells. Here we found that nuclear receptor corepressor-1 (NCoR1) is a critical component of DRED that acts as a scaffold to unite the DNA-binding and epigenetic enzyme components (e.g., DNA methyltransferase 1 [DNMT1] and lysine-specific demethylase 1 [LSD1]) that elicit DRED function. We also describe a potent new regulator of γ-globin repression: The deubiquitinase BRCA1-associated protein-1 (BAP1) is a component of the repressor complex whose activity maintains NCoR1 at sites in the ß-globin locus, and BAP1 inhibition in erythroid cells massively induces γ-globin synthesis. These data provide new mechanistic insights through the discovery of novel epigenetic enzymes that mediate γ-globin gene repression.


Assuntos
Regulação da Expressão Gênica/genética , Correpressor 1 de Receptor Nuclear/genética , Correpressor 1 de Receptor Nuclear/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , gama-Globinas/genética , Sítios de Ligação , Linhagem Celular , Ativação Enzimática/genética , Epigênese Genética/genética , Células Eritroides/metabolismo , Inativação Gênica , Células HEK293 , Humanos , Células K562 , Membro 1 do Grupo C da Subfamília 2 de Receptores Nucleares/metabolismo , Domínios Proteicos , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo
2.
BMC Neurosci ; 16: 34, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26077244

RESUMO

BACKGROUND: Inhibitory molecules in the adult central nervous system, including NogoA, impede neural repair by blocking axon outgrowth. The actin-myosin regulatory protein Shroom3 directly interacts with Rho kinase and conveys axon outgrowth inhibitory signals from Nogo66, a C-terminal inhibitory domain of NogoA. The purpose of this study was to identify small molecules that block the Shroom3-Rho kinase protein-protein interaction as a means to modulate NogoA signaling and, in the longer term, enhance axon outgrowth during neural repair. RESULTS: A high throughput screen for inhibitors of the Shroom3-Rho kinase protein-protein interaction identified CCG-17444 (Chem ID: 2816053). CCG-17444 inhibits the Shroom3-Rho kinase interaction in vitro with micromolar potency. This compound acts through an irreversible, covalent mechanism of action, targeting Shroom3 Cys1816 to inhibit the Shroom3-Rho kinase protein-protein interaction. Inhibition of the Shroom3-Rho kinase protein-protein interaction with CCG-17444 counteracts the inhibitory action of Nogo66 and enhances neurite outgrowth. CONCLUSIONS: This study identifies a small molecule inhibitor of the Shroom3-Rho kinase protein-protein interaction that circumvents the inhibitory action of Nogo66 in neurons. Identification of a small molecule compound that blocks the Shroom3-Rho kinase protein-protein interaction provides a first step towards a potential new strategy for enhancing neural repair.


Assuntos
Axônios/efeitos dos fármacos , Crescimento Celular/efeitos dos fármacos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas da Mielina/metabolismo , Pirimidinonas/farmacologia , Quinolonas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Animais , Axônios/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/fisiologia , Relação Dose-Resposta a Droga , Escherichia coli , Ensaios de Triagem em Larga Escala , Camundongos , Proteínas dos Microfilamentos/metabolismo , Proteínas Nogo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Recombinantes/metabolismo , Quinases Associadas a rho/metabolismo
3.
J Neurosci ; 30(40): 13319-25, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20926658

RESUMO

Myelin-derived inhibitors limit axon outgrowth and plasticity during development and in the adult mammalian CNS. Nogo66, a functional domain of the myelin-derived inhibitor NogoA, signals through the PirB receptor to inhibit axon outgrowth. The signaling pathway mobilized by Nogo66 engagement of PirB is not well understood. We identify a critical role for the scaffold protein Plenty of SH3s (POSH) in relaying process outgrowth inhibition downstream of Nogo66 and PirB. Blocking the function of POSH, or two POSH-associated proteins, leucine zipper kinase (LZK) and Shroom3, with RNAi in cortical neurons leads to release from myelin and Nogo66 inhibition. We also observed autocrine inhibition of process outgrowth by NogoA, and suppression analysis with the POSH-associated kinase LZK demonstrated that LZK operates downstream of NogoA and PirB in a POSH-dependent manner. In addition, cerebellar granule neurons with an RNAi-mediated knockdown in POSH function were refractory to the inhibitory action of Nogo66, indicating that a POSH-dependent mechanism operates to inhibit axon outgrowth in different types of CNS neurons. These studies delineate an intracellular signaling pathway for process outgrowth inhibition by Nogo66, comprised of NogoA, PirB, POSH, LZK, and Shroom3, and implicate the POSH complex as a potential therapeutic target to enhance axon outgrowth and plasticity in the injured CNS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas do Citoesqueleto/fisiologia , Cones de Crescimento/metabolismo , Líquido Intracelular/fisiologia , Proteínas da Mielina/metabolismo , Inibição Neural/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/genética , Cones de Crescimento/fisiologia , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Líquido Intracelular/metabolismo , MAP Quinase Quinase Quinases/fisiologia , Camundongos , Proteínas dos Microfilamentos/fisiologia , Proteínas da Mielina/genética , Proteínas da Mielina/fisiologia , Inibição Neural/genética , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Proteínas Nogo , Ratos , Receptores Imunológicos/fisiologia , Transdução de Sinais/genética , Frações Subcelulares/química , Frações Subcelulares/metabolismo , Frações Subcelulares/fisiologia
4.
Cell Signal ; 19(1): 177-84, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16887332

RESUMO

UNC-51-like kinases (ULK) are members of an evolutionarily conserved sub-family of ubiquitously expressed serine/threonine-specific protein kinases. Here we report that fibroblast growth factor receptor substrate (FRS) 2/3 are novel ULK2 carboxy-terminal domain interacting proteins. FRS2/3 are homologs that function as adaptor proteins to mediate signaling of multiple receptor tyrosine kinases. ULK2 interacts with the phospho-tyrosine binding (PTB) domain of FRS2/3. We demonstrate that siRNA targeting ULK2 in mouse P19 cells results in elevated FGFR1 mediated FRS3 and SHP2 tyrosyl phosphorylation. In addition, RNAi-mediated decrease in ULK2 causes increased interaction between FGFR1 and FRS3. ULK2 phosphorylates FRS2/3 in vitro, suggesting that ULK2 mediated phosphorylation may be a mechanism of FRS2/3 regulation. The data presented support a model in which ULK2, by interaction with FRS2/3 and inhibition of SynGAP, functions to negatively regulate tyrosyl phosphorylation of signaling proteins downstream of FGFR1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Sequência de Aminoácidos , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido , Tirosina/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
5.
Nucleic Acids Res ; 34(7): e53, 2006 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-16614444

RESUMO

Vector-based RNA interference (RNAi) has emerged as a valuable tool for analysis of gene function. We have developed new RNA polymerase II expression vectors for RNAi, designated SIBR vectors, based upon the non-coding RNA BIC. BIC contains the miR-155 microRNA (miRNA) precursor, and we find that expression of a short region of the third exon of mouse BIC is sufficient to produce miR-155 in mammalian cells. The SIBR vectors use a modified miR-155 precursor stem-loop and flanking BIC sequences to express synthetic miRNAs complementary to target RNAs. Like RNA polymerase III driven short hairpin RNA vectors, the SIBR vectors efficiently reduce target mRNA and protein expression. The synthetic miRNAs can be expressed from an intron, allowing coexpression of a marker or other protein with the miRNAs. In addition, intronic expression of a synthetic miRNA from a two intron vector enhances RNAi. A SIBR vector can express two different miRNAs from a single transcript for effective inhibition of two different target mRNAs. Furthermore, at least eight tandem copies of a synthetic miRNA can be expressed in a polycistronic transcript to increase the inhibition of a target RNA. The SIBR vectors are flexible tools for a variety of RNAi applications.


Assuntos
Vetores Genéticos , MicroRNAs/genética , Interferência de RNA , RNA Polimerase II/metabolismo , Animais , Genes Reporter , Humanos , Íntrons , Luciferases/análise , Luciferases/genética , Camundongos , MicroRNAs/biossíntese , Transcrição Gênica
6.
Mol Cell Biol ; 23(13): 4417-27, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12808085

RESUMO

Neural basic helix-loop-helix (bHLH) transcription factors regulate neurogenesis in vertebrates. Signaling by peptide growth factors also plays critical roles in regulating neuronal differentiation and survival. Many peptide growth factors activate phosphatidylinositol 3-kinase (PI3K) and subsequently the Akt kinases, raising the possibility that Akt may impact bHLH protein function during neurogenesis. Here we demonstrate that reducing expression of endogenous Akt1 and Akt2 by RNA interference (RNAi) reduces neuron generation in P19 cells transfected with a neural bHLH expression vector. The reduction in neuron generation from decreased Akt expression is not solely due to decreased cell survival, since addition of the caspase inhibitor z-VAD-FMK rescues cell death associated with loss of Akt function but does not restore neuron formation. This result indicates that Akt1 and Akt2 have additional functions during neuronal differentiation that are separable from neuronal survival. We show that activated Akt1 enhances complex formation between bHLH proteins and the transcriptional coactivator p300. Activated Akt1 also significantly augments the transcriptional activity of the bHLH protein neurogenin 3 in complex with the coactivators p300 or CBP. In addition, inhibition of endogenous Akt activity by the PI3K/Akt inhibitor LY294002 abolishes transcriptional cooperativity between the bHLH proteins and p300. We propose that Akt regulates the assembly and activity of bHLH-coactivator complexes to promote neuronal differentiation.


Assuntos
Neurônios/citologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Sequência de Bases , Sítios de Ligação , Western Blotting , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Cromonas/farmacologia , Proteína p300 Associada a E1A , Inibidores Enzimáticos/farmacologia , Epitopos , Vetores Genéticos , Sequências Hélice-Alça-Hélice , Humanos , Luciferases/metabolismo , Camundongos , Dados de Sequência Molecular , Morfolinas/farmacologia , Mutação , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/metabolismo , Ativação Transcricional , Transfecção
7.
Oncogene ; 22(36): 5554-61, 2003 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-12944902

RESUMO

Cross-talk between signaling pathways plays an important role in regulation of cell growth, differentiation, survival, and death. Here, we show that Akt regulates the Elk-1 transcription factor, independent of its negative regulation of Raf kinases. Using a constitutively active Mek1 to bypass the regulation of Raf by Akt, we find that the Elk-1 and Sap1a proteins are dramatically decreased in the presence of activated Akt. Akt catalytic activity is required. Also, Mek-dependent activation of a TCF (Elk-1/Sap-1a)-dependent c-fos reporter is decreased by activated Akt. Neither the level of Elk-1 mRNA nor the stability of the Elk-1 protein is altered by activated Akt. Instead, the rate of incorporation of labeled methionine into Elk-1 protein is decreased in the presence of Akt. In addition, the level of the Elk-1 protein but not GFP is significantly decreased in the presence of activated Akt, when GFP is expressed from an IRES element in a bicistronic message with Elk-1. We conclude that Akt negatively regulates translation of the Elk-1 mRNA. A coding region determinant that maps within the first 279 nts of the Elk-1 message is necessary and sufficient for Akt-mediated regulation of Elk-1.


Assuntos
Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição , Animais , Células COS , Proteínas de Ligação a DNA/genética , Genes Reporter , Genes fos , Humanos , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-akt , Transcrição Gênica , Proteínas Elk-1 do Domínio ets , Proteínas Elk-4 do Domínio ets
8.
Methods Enzymol ; 392: 186-99, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15644182

RESUMO

The use of RNA interference (RNAi) in mammalian cells has become a powerful tool for the analysis of gene function. Here we discuss the use of DNA vectors to produce short hairpin RNAs (shRNAs) and inhibit gene expression in mammalian neural progenitors and neurons. Protocols are presented for introducing shRNA vectors into mouse P19 cells differentiated as neurons in vitro and for electroporation of shRNA vectors into primary neural progenitors from the embryonic mouse dorsal telencephalon (prospective cerebral cortex). Transfected primary cortical progenitors can be differentiated in vitro either in dissociated culture or organotypic slice culture. The use of shRNA vectors for RNAi provides a versatile approach to understand gene function during mammalian neural development.


Assuntos
Encéfalo/embriologia , Vetores Genéticos , RNA/genética , Animais , Sequência de Bases , Eletroporação , Feminino , Camundongos , Gravidez
9.
Mol Biol Cell ; 19(12): 5181-92, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18829867

RESUMO

How scaffold proteins integrate signaling pathways with cytoskeletal components to drive axon outgrowth is not well understood. We report here that the multidomain scaffold protein Plenty of SH3s (POSH) regulates axon outgrowth. Reduction of POSH function by RNA interference (RNAi) enhances axon outgrowth in differentiating mouse primary cortical neurons and in neurons derived from mouse P19 cells, suggesting POSH negatively regulates axon outgrowth. Complementation analysis reveals a requirement for the third Src homology (SH) 3 domain of POSH, and we find that the actomyosin regulatory protein Shroom3 interacts with this domain of POSH. Inhibition of Shroom3 expression by RNAi leads to increased process lengths, as observed for POSH RNAi, suggesting that POSH and Shroom function together to inhibit process outgrowth. Complementation analysis and interference of protein function by dominant-negative approaches suggest that Shroom3 recruits Rho kinase to inhibit process outgrowth. Furthermore, inhibition of myosin II function reverses the POSH or Shroom3 RNAi phenotype, indicating a role for myosin II regulation as a target of the POSH-Shroom complex. Collectively, these results suggest that the molecular scaffold protein POSH assembles an inhibitory complex that links to the actin-myosin network to regulate neuronal process outgrowth.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Axônios/metabolismo , Proteínas do Citoesqueleto/metabolismo , Neurônios/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Axônios/ultraestrutura , Células Cultivadas , Córtex Cerebral/citologia , Proteínas do Citoesqueleto/genética , Teste de Complementação Genética , Humanos , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Neurônios/citologia , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIA/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Interferência de RNA , Técnicas do Sistema de Duplo-Híbrido , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
10.
J Biol Chem ; 278(48): 47922-7, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-14504284

RESUMO

We demonstrate that POSH, a scaffold for the JNK signaling pathway, binds to Akt2. A POSH mutant that is unable to bind Akt2 (POSH W489A) exhibits enhanced-binding to MLK3, and this increase in binding is accompanied by increased activation of the JNK signaling pathway. In addition, we show that the association of MLK3 with POSH is increased upon inhibition of the endogenous phosphatidylinositol 3-kinase/Akt signaling pathway. Thus, the assembly of an active JNK signaling complex by POSH is negatively regulated by Akt2. Further, the level of Akt-phosphorylated MLK3 is reduced in cells expressing the Akt2 binding domain of POSH, which acts as a dominant interfering protein. Taken together, our results support a model in which Akt2 binds to a POSH-MLK-MKK-JNK complex and phosphorylates MLK3; phosphorylation of MLK3 by Akt2 results in the disassembly of the JNK complex bound to POSH and down-regulation of the JNK signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase Quinases/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular , DNA Complementar/metabolismo , Regulação para Baixo , Técnica Indireta de Fluorescência para Anticorpo , Regulação Enzimológica da Expressão Gênica , Biblioteca Gênica , Genes Dominantes , Genes Reporter , Glutationa Transferase/metabolismo , Humanos , MAP Quinase Quinase 4 , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido , Domínios de Homologia de src , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
11.
Mol Ther ; 7(2): 228-36, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12597911

RESUMO

Short interfering RNAs (siRNAs) can mediate sequence-specific inhibition of gene expression in mammalian cells. We and others have recently developed expression vector-based systems for synthesizing siRNAs or hairpin siRNAs in mammalian cells. Expression vector-based RNA interference (RNAi) effectively suppresses expression of target genes and is likely to be a powerful tool for analysis of gene function. Here we compare inhibition by vectors expressing hairpin siRNA designs either with different loop sequences connecting the two siRNA strands, or with duplex regions of different lengths. Our results suggest that lengthening the 19-nucleotide duplex region of a relatively ineffective hairpin siRNA can increase inhibition, but increasing the length of an effective 19-nt hairpin siRNA does not increase inhibition. We also demonstrate that hairpin siRNA vectors can be used to inhibit two target genes simultaneously. We have targeted glycogen synthase kinase-3alpha (GSK-3alpha) and GSK-3beta, two related kinases involved in the regulation of a variety of cellular processes and also implicated in the pathogenesis of several human diseases. Inhibition of either GSK-3alpha or GSK-3beta by transfection of hairpin siRNA vectors leads to elevated expression of the GSK-3 target beta-catenin, whereas inhibition of both kinases further increases beta-catenin expression. Our results suggest that vector-based siRNA inhibition may be useful for dissecting the functional roles of GSK-3alpha and GSK-3beta in somatic cells. The ability to inhibit two or more genes simultaneously with hairpin siRNA expression vectors should facilitate studies of gene function in mammalian cells.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , RNA Interferente Pequeno/genética , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Proteínas do Citoesqueleto/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Camundongos , Dados de Sequência Molecular , Plasmídeos/metabolismo , Interferência de RNA , Transativadores/metabolismo , Transfecção , Regulação para Cima , beta Catenina
12.
Hum Mol Genet ; 12(16): 1981-93, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12913069

RESUMO

FGD1 mutations result in Faciogenital Dysplasia (FGDY), an X-linked human disease that affects skeletal formation and embryonic morphogenesis. FGD1 and Fgd1, the mouse FGD1 ortholog, encode guanine nucleotide exchange factors (GEF) that specifically activate Cdc42, a Rho GTPase that controls the organization of the actin cytoskeleton. To further understand FGD1/Fgd1 signaling and begin to elucidate the molecular pathophysiology of FGDY, we demonstrate that Fgd1 directly interacts with cortactin and mouse actin-binding protein 1 (mAbp1), actin-binding proteins that regulate actin polymerization through the Arp2/3 complex. In yeast two-hybrid studies, cortactin and mAbp1 Src homology 3 (SH3) domains interact with a single Fgd1 SH3-binding domain (SH3-BD), and biochemical studies show that the Fgd1 SH3-BD directly binds to cortactin and mAbp1 in vitro. Immunoprecipitation studies show that Fgd1 interacts with cortactin and mAbp1 in vivo and that Fgd1 SH3-BD mutations disrupt binding. Immunocytochemical studies show that Fgd1 colocalizes with cortactin and mAbp1 in lamellipodia and membrane ruffles, and that Fgd1 subcellular targeting is dynamic. By using truncated cortactin proteins, immunocytochemical studies show that the cortactin SH3 domain targets Fgd1 to the subcortical actin cytoskeleton, and that abnormal Fgd1 localization results in actin cytoskeletal abnormalities and significant changes in cell shape and viability. Thus, this study provides novel in vitro and in vivo evidence that Fgd1 specifically and directly interacts with cortactin and mAbp1, and that these interactions play an important role in regulating the actin cytoskeleton and, subsequently, cell shape.


Assuntos
Citoesqueleto de Actina/metabolismo , Ossos Faciais/anormalidades , Ossos Faciais/embriologia , Displasia Fibrosa Poliostótica/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas/metabolismo , Anormalidades Urogenitais/genética , Proteína 2 Relacionada a Actina , Proteína 3 Relacionada a Actina , Sequência de Aminoácidos , Animais , Linhagem Celular , Tamanho Celular , Cortactina , Proteínas do Citoesqueleto/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Camundongos , Dados de Sequência Molecular , Proteínas/genética , Homologia de Sequência de Aminoácidos , Síndrome , Proteína cdc42 de Ligação ao GTP/genética , Domínios de Homologia de src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA