Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol (Mosk) ; 50(1): 27-33, 2016.
Artigo em Russo | MEDLINE | ID: mdl-27028808

RESUMO

Tyrosyl phosphorylation participates in various pathological and physiological processes, which are regulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). The Src homology-2 domain containing phosphatase SHP2 (encoded by PTPN11) is an important phosphatase, which was found to be implicated in the regulation of genetic disease, development, metabolic, neurological, muscle, skeletal disease and cancer. Germline mutations in PTPN11 cause the Noonan Syndrome, LEOPARD syndrome and metachondromatosis. Somatic PTPN11 mutations occur in hematologic malignancies and in solid tumors. SHP2 is also an important component in oncogenic signaling pathways. It may play different roles in different stages and positions of human cancers. Whether SHP2 is an oncogene or cancer suppressor gene remains to be elucidated. Elucidation of the regulatory mechanisms of SHP2 in human disease will provide new insights into disease and new targets for therapy. Here, we summarized the structural basis and recent research progression on SHP2 in various human disease, including genetic and cancer diseases.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Humanos , Síndrome LEOPARD/enzimologia , Mutação , Neoplasias/enzimologia , Síndrome de Noonan/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Transdução de Sinais
2.
PLoS One ; 9(9): e106682, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25184253

RESUMO

Noonan syndrome (NS) and LEOPARD syndrome (LS) cause congenital afflictions such as short stature, hypertelorism and heart defects. More than 50% of NS and almost all of LS cases are caused by activating and inactivating mutations of the phosphatase Shp2, respectively. How these biochemically opposing mutations lead to similar clinical outcomes is not clear. Using zebrafish models of NS and LS and mass spectrometry-based phosphotyrosine proteomics, we identified a down-regulated peptide of Fer kinase in both NS and LS. Further investigation showed a role for Fer during development, where morpholino-based knockdown caused craniofacial defects, heart edema and short stature. During gastrulation, loss of Fer caused convergence and extension defects without affecting cell fate. Moreover, Fer knockdown cooperated with NS and LS, but not wild type Shp2 to induce developmental defects, suggesting a role for Fer in the pathogenesis of both NS and LS.


Assuntos
Síndrome LEOPARD/enzimologia , Mutação , Síndrome de Noonan/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Técnicas de Silenciamento de Genes , Síndrome LEOPARD/genética , Síndrome de Noonan/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Tirosina Quinases/genética , Proteômica , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
3.
Curr Cancer Drug Targets ; 14(6): 567-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25039348

RESUMO

Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2), encoded by the human PTPN11 gene, is a ubiquitously expressed protein tyrosine phosphatase (PTP) that consists of two tandem Src homology (SH2) domains (N-SH2 and C-SH2), a PTP catalytic domain, and a C-terminal tail with tyrosyl phosphorylation sites. It plays critical roles in numerous cellular processes through the regulation of various signaling pathways in PTP catalytic activity-dependent and -independent manners. Dysfunction of SHP2 resulting from pathogenic mutations and aberrant expression leads to the dysregulation of multiple signaling pathways, thus contributing to different human disorders. Germline and somatic mutations in PTPN11 are involved in Noonan syndrome (NS), LEOPARD syndrome (LS), and hematological malignancies, as well as several solid tumors. In this report, we provide an overview of the current knowledge of the structure and function of SHP2, and further discuss the molecular and pathogenic mechanism of SHP2 in human diseases, with a special focus on tumorigenesis. Furthermore, we summarize that SHP2 might itself represent a potential drug target for cancer prevention and treatment. Ongoing research and development of SHP2-specific inhibitors would enhance this potential.


Assuntos
Transformação Celular Neoplásica/metabolismo , Síndrome LEOPARD/enzimologia , Neoplasias/enzimologia , Síndrome de Noonan/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Antineoplásicos/uso terapêutico , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Desenho de Fármacos , Inibidores Enzimáticos/uso terapêutico , Predisposição Genética para Doença , Humanos , Síndrome LEOPARD/genética , Modelos Moleculares , Terapia de Alvo Molecular , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Síndrome de Noonan/genética , Fenótipo , Conformação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/química , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Transdução de Sinais , Relação Estrutura-Atividade
4.
Am J Med Genet A ; 164A(4): 934-42, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24458522

RESUMO

Here, we describe neurobehavioral features in patients with RASopathies (i.e., Noonan syndrome, LEOPARD syndrome, Costello syndrome, and cardiofaciocutaneous syndrome), developmental disorders caused by mutations in genes coding transducers participating in the RAS-MAPK signaling cascade. Parents of 70 individuals with a RASopathy were asked to fill out the following questionnaires: Child Behavior Checklist (CBCL), Social Communication Questionnaire version lifetime (SCQ-L), and Modified Checklist for Autism in toddlers (M-CHAT). Data analysis indicated high rates of internalizing (37%) and externalizing problems (31%) on CBCL. Scores over the cut-off were documented in 64% of patients with cardiofaciocutaneous syndrome, 44% with Costello syndrome, and 12% with Noonan syndrome on SCQ-L/M-CHAT. Our findings indicate that mutations promoting dysregulation of the RAS-MAPK cascade mark an increased psychopathological risk and highlight that autistic-like behavior could be underdiagnosed in patients with RASopathies.


Assuntos
Sistema de Sinalização das MAP Quinases/genética , Transtornos Mentais/enzimologia , Transtornos Mentais/genética , Proteínas ras/genética , Adolescente , Adulto , Transtorno Autístico/enzimologia , Transtorno Autístico/genética , Criança , Pré-Escolar , Síndrome de Costello/enzimologia , Síndrome de Costello/genética , Deficiências do Desenvolvimento/enzimologia , Deficiências do Desenvolvimento/genética , Displasia Ectodérmica/enzimologia , Displasia Ectodérmica/genética , Fácies , Insuficiência de Crescimento/enzimologia , Insuficiência de Crescimento/genética , Feminino , Cardiopatias Congênitas/enzimologia , Cardiopatias Congênitas/genética , Humanos , Síndrome LEOPARD/enzimologia , Síndrome LEOPARD/genética , Masculino , Mutação/genética , Síndrome de Noonan/enzimologia , Síndrome de Noonan/genética , Adulto Jovem
5.
J Biol Chem ; 288(25): 18335-44, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23673659

RESUMO

In LEOPARD syndrome (LS) patients, mutations in the protein tyrosine phosphatase Shp2 cause hypertrophic cardiomyopathy. The prohypertrophic effects of mutant Shp2 are mediated downstream by hyperactivation of mammalian target of rapamycin. Our goal was to further define the signaling cascade that is essential for the underlying pathomechanism, thus expanding the list of potential future therapeutic targets. Using cultured neonatal rat cardiomyocytes with adenoviral gene delivery and pharmacological inhibitors, we found that hypertrophy induced by a particularly aggressive LS mutation in Shp2 depends on hyperactivation of Akt and focal adhesion kinase as well as mammalian target of rapamycin. Dissecting domain-specific functions of Shp2 using double and truncation mutants, we determined that the hypertrophic effects of mutant Shp2 depend on the two SH2 domains and on an intact catalytic center. The latter finding prompted us to test the efficacy of a Shp2 inhibitor targeted directly at the catalytic pocket. This compound, PHPS1, effectively prevented mutant Shp2-induced hypertrophy. In summary, we identified three novel targets for pharmacological therapy of LS-associated cardiac hypertrophy. Of particular importance is the finding that intervention directly at the mutant Shp2 protein is effective because this would facilitate custom-tailored therapeutic approaches for patients carrying LS mutations in Shp2.


Assuntos
Cardiomegalia/enzimologia , Síndrome LEOPARD/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Benzenossulfonatos/farmacologia , Western Blotting , Cardiomegalia/genética , Cardiomegalia/prevenção & controle , Células Cultivadas , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Hidrazonas/farmacologia , Síndrome LEOPARD/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
6.
PLoS Genet ; 3(12): e225, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18159945

RESUMO

Shp2 is a cytoplasmic protein-tyrosine phosphatase that is essential for normal development. Activating and inactivating mutations have been identified in humans to cause the related Noonan and LEOPARD syndromes, respectively. The cell biological cause of these syndromes remains to be determined. We have used the zebrafish to assess the role of Shp2 in early development. Here, we report that morpholino-mediated knockdown of Shp2 in zebrafish resulted in defects during gastrulation. Cell tracing experiments demonstrated that Shp2 knockdown induced defects in convergence and extension cell movements. In situ hybridization using a panel of markers indicated that cell fate was not affected by Shp2 knock down. The Shp2 knockdown-induced defects were rescued by active Fyn and Yes and by active RhoA. We generated mutants of Shp2 with mutations that were identified in human patients with Noonan or LEOPARD Syndrome and established that Noonan Shp2 was activated and LEOPARD Shp2 lacked catalytic protein-tyrosine phosphatase activity. Expression of Noonan or LEOPARD mutant Shp2 in zebrafish embryos induced convergence and extension cell movement defects without affecting cell fate. Moreover, these embryos displayed craniofacial and cardiac defects, reminiscent of human symptoms. Noonan and LEOPARD mutant Shp2s were not additive nor synergistic, consistent with the mutant Shp2s having activating and inactivating roles in the same signaling pathway. Our results demonstrate that Shp2 is required for normal convergence and extension cell movements during gastrulation and that Src family kinases and RhoA were downstream of Shp2. Expression of Noonan or LEOPARD Shp2 phenocopied the craniofacial and cardiac defects of human patients. The finding that defective Shp2 signaling induced cell movement defects as early as gastrulation may have implications for the monitoring and diagnosis of Noonan and LEOPARD syndrome.


Assuntos
Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Diferenciação Celular , Movimento Celular , Modelos Animais de Doenças , Gastrulação , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Marcação de Genes , Humanos , Síndrome LEOPARD/enzimologia , Síndrome LEOPARD/genética , Mutação , Síndrome de Noonan/enzimologia , Síndrome de Noonan/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Proteínas Proto-Oncogênicas c-yes/fisiologia , Transdução de Sinais , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA