Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Biol (Mosk) ; 50(1): 27-33, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-27028808

RESUMEN

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.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Humanos , Síndrome LEOPARD/enzimología , Mutación , Neoplasias/enzimología , Síndrome de Noonan/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Transducción de Señal
2.
PLoS One ; 9(9): e106682, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25184253

RESUMEN

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.


Asunto(s)
Síndrome LEOPARD/enzimología , Mutación , Síndrome de Noonan/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Técnicas de Silenciamiento del Gen , Síndrome LEOPARD/genética , Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Tirosina Quinasas/genética , Proteómica , Pez Cebra/genética , Proteínas de Pez Cebra/genética
3.
Curr Cancer Drug Targets ; 14(6): 567-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25039348

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Síndrome LEOPARD/enzimología , Neoplasias/enzimología , Síndrome de Noonan/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Animales , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Diseño de Fármacos , Inhibidores Enzimáticos/uso terapéutico , Predisposición Genética a la Enfermedad , Humanos , Síndrome LEOPARD/genética , Modelos Moleculares , Terapia Molecular Dirigida , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Síndrome de Noonan/genética , Fenotipo , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/química , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Transducción de Señal , Relación Estructura-Actividad
4.
Biochemistry ; 53(25): 4136-51, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24935154

RESUMEN

The Src homology 2 (SH2) domain-containing protein tyrosine phosphatase 2 (SHP2) is a critical signal transducer downstream of growth factors that promotes the activation of the RAS-ERK1/2 cascade. In its basal state, SHP2 exists in an autoinhibited closed conformation because of an intramolecular interaction between its N-SH2 and protein tyrosine phosphatase (PTP) domains. Binding to pTyr ligands present on growth factor receptors and adaptor proteins with its N-SH2 domain localizes SHP2 to its substrates and frees the active site from allosteric inhibition. Germline mutations in SHP2 are known to cause both Noonan syndrome (NS) and LEOPARD syndrome (LS), two clinically similar autosomal dominant developmental disorders. NS-associated SHP2 mutants display elevated phosphatase activity, while LS-associated SHP2 mutants exhibit reduced catalytic activity. A conundrum in how clinically similar diseases result from mutations to SHP2 that have opposite effects on this enzyme's catalytic functionality exists. Here we report a comprehensive investigation of the kinetic, structural, dynamic, and biochemical signaling properties of the wild type as well as all reported LS-associated SHP2 mutants. The results reveal that LS-causing mutations not only affect SHP2 phosphatase activity but also induce a weakening of the intramolecular interaction between the N-SH2 and PTP domains, leading to mutants that are more readily activated by competing pTyr ligands. Our data also indicate that the residual phosphatase activity associated with the LS SHP2 mutant is required for enhanced ERK1/2 activation. Consequently, catalytically impaired SHP2 mutants could display gain-of-function properties because of their ability to localize to the vicinity of substrates for longer periods of time, thereby affording the opportunity for prolonged substrate turnover and sustained RAS-ERK1/2 activation.


Asunto(s)
Síndrome LEOPARD/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/química , Activación Enzimática , Células HEK293 , Humanos , Cinética , Síndrome LEOPARD/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Moleculares , Mutación , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Dominios Homologos src
5.
Development ; 141(9): 1961-70, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24718990

RESUMEN

Germline mutations in PTPN11, encoding Shp2, cause Noonan syndrome (NS) and LEOPARD syndrome (LS), two developmental disorders that are characterized by multiple overlapping symptoms. Interestingly, Shp2 catalytic activity is enhanced by NS mutations and reduced by LS mutations. Defective cardiac development is a prominent symptom of both NS and LS, but how the Shp2 variants affect cardiac development is unclear. Here, we have expressed the most common NS and LS Shp2-variants in zebrafish embryos to investigate their role in cardiac development in vivo. Heart function was impaired in embryos expressing NS and LS variants of Shp2. The cardiac anomalies first occurred during elongation of the heart tube and consisted of reduced cardiomyocyte migration, coupled with impaired leftward heart displacement. Expression of specific laterality markers was randomized in embryos expressing NS and LS variants of Shp2. Ciliogenesis and cilia function in Kupffer's vesicle was impaired, likely accounting for the left/right asymmetry defects. Mitogen-activated protein kinase (MAPK) signaling was activated to a similar extent in embryos expressing NS and LS Shp2 variants. Interestingly, inhibition of MAPK signaling prior to gastrulation rescued cilia length and heart laterality defects. These results suggest that NS and LS Shp2 variant-mediated hyperactivation of MAPK signaling leads to impaired cilia function in Kupffer's vesicle, causing left-right asymmetry defects and defective early cardiac development.


Asunto(s)
Cardiopatías Congénitas/genética , Síndrome LEOPARD/genética , Mutación/genética , Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Benzamidas/farmacología , Tipificación del Cuerpo/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Cilios/efectos de los fármacos , Cilios/metabolismo , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/enzimología , Embrión no Mamífero/patología , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/fisiopatología , Pruebas de Función Cardíaca , Humanos , Síndrome LEOPARD/enzimología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Síndrome de Noonan/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
6.
Am J Med Genet A ; 164A(4): 934-42, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24458522

RESUMEN

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.


Asunto(s)
Sistema de Señalización de MAP Quinasas/genética , Trastornos Mentales/enzimología , Trastornos Mentales/genética , Proteínas ras/genética , Adolescente , Adulto , Trastorno Autístico/enzimología , Trastorno Autístico/genética , Niño , Preescolar , Síndrome de Costello/enzimología , Síndrome de Costello/genética , Discapacidades del Desarrollo/enzimología , Discapacidades del Desarrollo/genética , Displasia Ectodérmica/enzimología , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/enzimología , Insuficiencia de Crecimiento/genética , Femenino , Cardiopatías Congénitas/enzimología , Cardiopatías Congénitas/genética , Humanos , Síndrome LEOPARD/enzimología , Síndrome LEOPARD/genética , Masculino , Mutación/genética , Síndrome de Noonan/enzimología , Síndrome de Noonan/genética , Adulto Joven
7.
J Biol Chem ; 288(25): 18335-44, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23673659

RESUMEN

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.


Asunto(s)
Cardiomegalia/enzimología , Síndrome LEOPARD/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Bencenosulfonatos/farmacología , Western Blotting , Cardiomegalia/genética , Cardiomegalia/prevención & control , Células Cultivadas , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Hidrazonas/farmacología , Síndrome LEOPARD/genética , Ratones , Ratones Endogámicos C57BL , Mutación , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
8.
J Biol Chem ; 288(15): 10472-82, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23457302

RESUMEN

SHP2 is an allosteric phosphatase essential for growth factor-mediated Ras activation. Germ-line mutations in SHP2 cause clinically similar LEOPARD and Noonan syndromes, two of several autosomal-dominant conditions characterized by gain-of-function mutations in the Ras pathway. Interestingly, Noonan syndrome SHP2 mutants are constitutively active, whereas LEOPARD syndrome SHP2 mutants exhibit reduced phosphatase activity. How do catalytically impaired LEOPARD syndrome mutants engender gain-of-function phenotypes? Our study reveals that LEOPARD syndrome mutations weaken the intramolecular interaction between the N-SH2 and phosphatase domains, leading to a change in SHP2 molecular switching mechanism. Consequently, LEOPARD syndrome SHP2 mutants bind upstream activators preferentially and are hypersensitive to growth factor stimulation. They also stay longer with scaffolding adapters, thus prolonging substrate turnover, which compensates for the reduced phosphatase activity. The study provides a solid framework for understanding how individual SHP2 mutations cause diseases.


Asunto(s)
Síndrome LEOPARD/enzimología , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/química , Cristalografía por Rayos X , Humanos , Síndrome LEOPARD/genética , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Relación Estructura-Actividad
9.
Hum Mol Genet ; 18(1): 193-201, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18849586

RESUMEN

Missense mutations in the PTPN11 gene, which encodes the protein tyrosine phosphatase SHP-2, cause clinically similar but distinctive disorders, LEOPARD (LS) and Noonan (NS) syndromes. The LS is an autosomal dominant disorder with pleomorphic developmental abnormalities including lentigines, cardiac defects, short stature and deafness. Biochemical analyses indicated that LS alleles engender loss-of-function (LOF) effects, while NS mutations result in gain-of-function (GOF). These biochemical findings lead to an enigma that how PTPN11 mutations with opposite effects on function result in disorders that are so similar. To study the developmental effects of the commonest LS PTPN11 alleles (Y279C and T468M), we generated LS transgenic fruitflies using corkscrew (csw), the Drosophila orthologue of PTPN11. Ubiquitous expression of the LS csw mutant alleles resulted in ectopic wing veins and, for the Y279C allele, rough eyes with increased R7 photoreceptor numbers. These were GOF phenotypes mediated by increased RAS/MAPK signaling and requiring the LS mutant's residual phosphatase activity. Our findings provide the first evidence that LS mutant alleles have GOF developmental effects despite reduced phosphatase activity, providing a rationale for how PTPN11 mutations with GOF and LOF produce similar but distinctive syndromes.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/crecimiento & desarrollo , Drosophila/genética , Síndrome LEOPARD/enzimología , Mutación Missense , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Animales Modificados Genéticamente , Drosophila/anatomía & histología , Drosophila/enzimología , Proteínas de Drosophila/metabolismo , Ojo/anatomía & histología , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Humanos , Síndrome LEOPARD/genética , Síndrome de Noonan/enzimología , Síndrome de Noonan/genética , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Transducción de Señal , Alas de Animales/anatomía & histología , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo
10.
Circulation ; 117(11): 1423-35, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18316486

RESUMEN

BACKGROUND: Heart failure is the leading cause of death in the United States. By delineating the pathways that regulate cardiomyocyte function, we can better understand the pathogenesis of cardiac disease. Many cardiomyocyte signaling pathways activate protein tyrosine kinases. However, the role of specific protein tyrosine phosphatases (PTPs) in these pathways is unknown. METHODS AND RESULTS: Here, we show that mice with muscle-specific deletion of Ptpn11, the gene encoding the SH2 domain-containing PTP Shp2, rapidly develop a compensated dilated cardiomyopathy without an intervening hypertrophic phase, with signs of cardiac dysfunction appearing by the second postnatal month. Shp2-deficient primary cardiomyocytes are defective in extracellular signal-regulated kinase/mitogen-activated protein kinase (Erk/MAPK) activation in response to a variety of soluble agonists and pressure overload but show hyperactivation of the RhoA signaling pathway. Treatment of primary cardiomyocytes with Erk1/2- and RhoA pathway-specific inhibitors suggests that both abnormal Erk/MAPK and RhoA activities contribute to the dilated phenotype of Shp2-deficient hearts. CONCLUSIONS: Our results identify Shp2 as the first PTP with a critical role in adult cardiac function, indicate that in the absence of Shp2 cardiac hypertrophy does not occur in response to pressure overload, and demonstrate that the cardioprotective role of Shp2 is mediated via control of both the Erk/MAPK and RhoA signaling pathways.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Miocitos Cardíacos/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/fisiología , Animales , Cardiomegalia/enzimología , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Síndrome LEOPARD/enzimología , Síndrome LEOPARD/genética , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Modelos Animales , Síndrome de Noonan/enzimología , Síndrome de Noonan/genética , Especificidad de Órganos , Fenotipo , Presión , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA
11.
PLoS Genet ; 3(12): e225, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18159945

RESUMEN

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.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Diferenciación Celular , Movimiento Celular , Modelos Animales de Enfermedad , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Marcación de Gen , Humanos , Síndrome LEOPARD/enzimología , Síndrome LEOPARD/genética , Mutación , Síndrome de Noonan/enzimología , Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Proteínas Proto-Oncogénicas c-fyn/fisiología , Proteínas Proto-Oncogénicas c-yes/fisiología , Transducción de Señal , Pez Cebra/fisiología , Proteínas de Pez Cebra/fisiología , Proteína de Unión al GTP rhoA/fisiología
12.
FEBS Lett ; 580(10): 2477-82, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16638574

RESUMEN

LEOPARD (LS) and Noonan (NS) are overlapping syndromes associated with distinct mutations of SHP-2. Whereas NS mutations enhance SHP-2 catalytic activity, we show that the activity of three representative LS mutants is undetectable when assayed using a standard protein tyrosine phosphatase (PTP) substrate. A different assay using a specific SHP-2 substrate confirms their decreased PTP activity, but also reveals a significant activity of the T468M mutant. In transfected cells stimulated with epidermal growth factor, the least active LS mutants promote Gab1/PI3K binding, validating our in vitro data. LS mutants thus display a reduced PTP activity both in vitro and in transfected cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Síndrome LEOPARD/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Catálisis , Chlorocebus aethiops , Factor de Crecimiento Epidérmico/farmacología , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Modelos Moleculares , Mutación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/genética , Células Vero
13.
J Biol Chem ; 281(10): 6785-92, 2006 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-16377799

RESUMEN

Multiple lentigines/LEOPARD syndrome (LS) is a rare, autosomal dominant disorder characterized by Lentigines, Electrocardiogram abnormalities, Ocular hypertelorism, Pulmonic valvular stenosis, Abnormalities of genitalia, Retardation of growth, and Deafness. Like the more common Noonan syndrome (NS), LS is caused by germ line missense mutations in PTPN11, encoding the protein-tyrosine phosphatase Shp2. Enzymologic, structural, cell biological, and mouse genetic studies indicate that NS is caused by gain-of-function PTPN11 mutations. Because NS and LS share several features, LS has been viewed as an NS variant. We examined a panel of LS mutants, including the two most common alleles. Surprisingly, we found that in marked contrast to NS, LS mutants are catalytically defective and act as dominant negative mutations that interfere with growth factor/Erk-mitogen-activated protein kinase-mediated signaling. Molecular modeling and biochemical studies suggest that LS mutations contort the Shp2 catalytic domain and result in open, inactive forms of Shp2. Our results establish that the pathogenesis of LS and NS is distinct and suggest that these disorders should be distinguished by mutational analysis rather than clinical presentation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Síndrome LEOPARD/genética , Proteínas Tirosina Fosfatasas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Dominio Catalítico/genética , Línea Celular , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Síndrome LEOPARD/enzimología , Mutación Missense , Síndrome de Noonan/enzimología , Síndrome de Noonan/genética , Fosfoproteínas/metabolismo , Estructura Terciaria de Proteína/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo
14.
J Hum Genet ; 50(1): 21-25, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15690106

RESUMEN

LEOPARD syndrome (lentigines, electrocardiographic conduction abnormalities, ocular hypertelorism, pulmonary stenosis, abnormal genitalia, retardation of growth, and sensorineural deafness) is an autosomal dominant condition. The main clinical features include multiple lentigines, cardiovascular defects, and facial anomalies, some of which are shared with Noonan syndrome (NS). Recent reports have shown that LEOPARD syndrome can be caused by mutations in PTPN11, the gene in which mutations can produce NS. Here we report the findings of mutation screening and linkage analysis of PTPN11 in three families with LEOPARD syndrome. We identified a novel mutation in one family. The mutation (1529A>C) substitutes proline for glutamine at amino acid 510 (Gln510Pro). No variations in sequence were observed in the other two families, and negative LOD scores excluded linkage to the PTPN11 locus, showing that LEOPARD syndrome is genetically heterogeneous.


Asunto(s)
Síndrome LEOPARD/enzimología , Síndrome LEOPARD/genética , Proteínas Tirosina Fosfatasas/genética , Adolescente , Sustitución de Aminoácidos , Secuencia de Bases , Niño , Preescolar , ADN/genética , Femenino , Ligamiento Genético , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Mutación , Síndrome de Noonan/enzimología , Síndrome de Noonan/genética , Linaje , Mutación Puntual , Proteína Tirosina Fosfatasa no Receptora Tipo 11
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...