Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Biol Chem ; 286(46): 39945-57, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21930711

RESUMEN

We have shown previously that the ubiquitin ligase MID1, mutations of which cause the midline malformation Opitz BBB/G syndrome (OS), serves as scaffold for a microtubule-associated protein complex that regulates protein phosphatase 2A (PP2A) activity in a ubiquitin-dependent manner. Here, we show that the MID1 protein complex associates with mRNAs via a purine-rich sequence motif called MIDAS (MID1 association sequence) and thereby increases stability and translational efficiency of these mRNAs. Strikingly, inclusion of multiple copies of the MIDAS motif into mammalian mRNAs increases production of the encoded proteins up to 20-fold. Mutated MID1, as found in OS patients, loses its influence on MIDAS-containing mRNAs, suggesting that the malformations in OS patients could be caused by failures in the regulation of cytoskeleton-bound protein translation. This is supported by the observation that the majority of mRNAs that carry MIDAS motifs is involved in developmental processes and/or energy homeostasis. Further analysis of one of the proteins encoded by a MIDAS-containing mRNA, namely PDPK-1 (3-phosphoinositide dependent protein kinase-1), which is an important regulator of mammalian target of rapamycin/PP2A signaling, showed that PDPK-1 protein synthesis is significantly reduced in cells from an OS patient compared with an age-matched control and can be rescued by functional MID1. Together, our data uncover a novel messenger ribonucleoprotein complex that regulates microtubule-associated protein translation. They suggest a novel mechanism underlying OS and point at an enormous potential of the MIDAS motif to increase the efficiency of biotechnological protein production in mammalian cells.


Asunto(s)
Proteínas de Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/biosíntesis , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Esófago/anomalías , Esófago/metabolismo , Células HeLa , Humanos , Hipertelorismo/genética , Hipertelorismo/metabolismo , Hipospadias/genética , Hipospadias/metabolismo , Proteínas de Microtúbulos/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética
2.
Proc Natl Acad Sci U S A ; 107(50): 21830-5, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21098287

RESUMEN

Hyperphosphorylated tau plays an important role in the formation of neurofibrillary tangles in brains of patients with Alzheimer's disease (AD) and related tauopathies and is a crucial factor in the pathogenesis of these disorders. Though diverse kinases have been implicated in tau phosphorylation, protein phosphatase 2A (PP2A) seems to be the major tau phosphatase. Using murine primary neurons from wild-type and human tau transgenic mice, we show that the antidiabetic drug metformin induces PP2A activity and reduces tau phosphorylation at PP2A-dependent epitopes in vitro and in vivo. This tau dephosphorylating potency can be blocked entirely by the PP2A inhibitors okadaic acid and fostriecin, confirming that PP2A is an important mediator of the observed effects. Surprisingly, metformin effects on PP2A activity and tau phosphorylation seem to be independent of AMPK activation, because in our experiments (i) metformin induces PP2A activity before and at lower levels than AMPK activity and (ii) the AMPK activator AICAR does not influence the phosphorylation of tau at the sites analyzed. Affinity chromatography and immunoprecipitation experiments together with PP2A activity assays indicate that metformin interferes with the association of the catalytic subunit of PP2A (PP2Ac) to the so-called MID1-α4 protein complex, which regulates the degradation of PP2Ac and thereby influences PP2A activity. In summary, our data suggest a potential beneficial role of biguanides such as metformin in the prophylaxis and/or therapy of AD.


Asunto(s)
Metformina/farmacología , Ovillos Neurofibrilares/metabolismo , Proteína Fosfatasa 2/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas tau/metabolismo , Adenilato Quinasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Epítopos , Células HeLa , Humanos , Hipoglucemiantes/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos , Ovillos Neurofibrilares/patología , Neuronas/citología , Neuronas/metabolismo , Ácido Ocadaico/farmacología , Fosforilación , Proteína Fosfatasa 2/genética , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Proteínas tau/genética
3.
Cardiovasc Res ; 86(3): 452-60, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20083571

RESUMEN

AIMS: Left ventricular non-compaction (LVNC) is caused by mutations in multiple genes. It is still unclear whether LVNC is the primary determinant of cardiomyopathy or rather a secondary phenomenon with intrinsic cardiomyocyte dysfunction being the actual cause of the disease. Here, we describe a family with LVNC due to a novel missense mutation, pE96K, in the cardiac troponin T gene (TNNT2). METHODS AND RESULTS: The novel mutation was identified in the index patient and all affected relatives, but not in 430 healthy control individuals. Mutations in known LVNC-associated genes were excluded. To investigate the pathophysiological implications of the mutation, we generated transgenic mice expressing human wild-type cTNT (hcTNT) or a human troponin T harbouring the pE96K mutation (mut cTNT). Animals were characterized by echocardiography, histology, and gene expression analysis. Mut cTNT mice displayed an impaired left ventricular function and induction of marker genes of heart failure. Remarkably, left ventricular non-compaction was not observed. CONCLUSION: Familial co-segregation and the cardiomyopathy phenotype of mut cTNT mice strongly support a causal relationship of the pE96K mutation and disease in our index patient. In addition, our data suggest that a non-compaction phenotype is not required for the development of cardiomyopathy in this specific TNNT2 mutation leading to LVNC.


Asunto(s)
No Compactación Aislada del Miocardio Ventricular/genética , Mutación Missense , Troponina T/genética , Animales , Biopsia , Estudios de Casos y Controles , Análisis Mutacional de ADN , Ecocardiografía Doppler en Color , Exones , Femenino , Predisposición Genética a la Enfermedad , Humanos , Lactante , No Compactación Aislada del Miocardio Ventricular/diagnóstico , No Compactación Aislada del Miocardio Ventricular/fisiopatología , Imagen por Resonancia Cinemagnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Contracción Miocárdica/genética , Linaje , Fenotipo , Índice de Severidad de la Enfermedad , Volumen Sistólico/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Adulto Joven
4.
BMC Med Genet ; 9: 95, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18957093

RESUMEN

BACKGROUND: Mutations in MYBPC3 encoding myosin binding protein C belong to the most frequent causes of hypertrophic cardiomyopathy (HCM) and may also lead to dilated cardiomyopathy (DCM). MYBPC3 mutations initially were considered to cause a benign form of HCM. The aim of this study was to examine the clinical outcome of patients and their relatives with 18 different MYBPC3 mutations. METHODS: 87 patients with HCM and 71 patients with DCM were screened for MYBPC3 mutations by denaturing gradient gel electrophoresis and sequencing. Close relatives of mutation carriers were genotyped for the respective mutation. Relatives with mutation were then evaluated by echocardiography and magnetic resonance imaging. A detailed family history regarding adverse clinical events was recorded. RESULTS: In 16 HCM (18.4%) and two DCM (2.8%) index patients a mutation was detected. Seven mutations were novel. Mutation carriers exhibited no additional mutations in genes MYH7, TNNT2, TNNI3, ACTC and TPM1. Including relatives of twelve families, a total number of 42 mutation carriers was identified of which eleven (26.2%) had at least one adverse event. Considering the twelve families and six single patients with mutations, 45 individuals with cardiomyopathy and nine with borderline phenotype were identified. Among the 45 patients, 23 (51.1%) suffered from an adverse event. In eleven patients of seven families an unexplained sudden death was reported at the age between 13 and 67 years. Stroke or a transient ischemic attack occurred in six patients of five families. At least one adverse event occurred in eleven of twelve families. CONCLUSION: MYBPC3 mutations can be associated with cardiac events such as progressive heart failure, stroke and sudden death even at younger age. Therefore, patients with MYBPC3 mutations require thorough clinical risk assessment.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Proteínas Portadoras/genética , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Hipertrófica Familiar/diagnóstico , Ecocardiografía , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Linaje , Pronóstico , Factores de Riesgo
5.
J Mol Med (Berl) ; 86(3): 281-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17987279

RESUMEN

We have identified a family in which several members died of sudden cardiac death or suffer from dilated cardiomyopathy (DCM) and rhythm disturbances. Mutation screening revealed co-segregation of a novel nonsense mutation (pR321X) in the lamin A gene, LMNA, with the disease. Lamin A, and its smaller splice form lamin C are nuclear intermediate filament proteins forming a major part of the lamina, which is underlying the inner nuclear membrane. They are involved in the organization of heterochromatin and both in DNA replication and transcription. Recently, an increasing number of missense mutations in LMNA have been discovered to cause various types of rare diseases. Here, we investigated the causal role of the new nonsense mutation for the disease. Quantification of wild type and mutant lamin A mRNA from explanted myocardial tissue and cultured fibroblasts revealed an up to 30-fold reduction in the relative amount of the mutant transcript indicating that its synthesis was massively down-regulated by nonsense-mediated mRNA decay (NMD). Correspondingly, we did not detect the mutant truncated lamin A by Western blot analysis in extracts of patient fibroblasts and cardiac muscle tissue. Both wild type lamin A and C were present, however, in normal quantities. The immunohistochemical analyses of patient tissues revealed a normal distribution of lamin A/C and of major inner nuclear membrane proteins such as emerin and the lamin B receptor. Moreover, both chromatin distribution and nuclear shape were normal. However, over-expression of truncated lamin A in HeLa cells by transient transfection caused major disturbances of lamin A organization within both the nucleoplasm and the cytoplasm. In addition, after treatment of patient fibroblasts with the proteasome inhibitor epoxomicin, mutant truncated lamin A was detected in relatively high levels by Western blotting demonstrating that it is synthesized in these cells. Therefore, we conclude that NMD is not sufficient to completely prevent the expression of truncated lamin A and that even trace amounts of it may negatively interfere with structural and/or regulatory functions of lamin A/C eventually leading to the development of DCM and rhythm disturbances.


Asunto(s)
Cardiomiopatía Dilatada/genética , Codón sin Sentido/genética , Lamina Tipo A/metabolismo , Proteínas Mutantes/metabolismo , Estabilidad del ARN , Taquicardia Ventricular/genética , Adulto , Alelos , Estudios de Casos y Controles , Núcleo Celular/metabolismo , Cromatina/metabolismo , Regulación hacia Abajo , Femenino , Fibroblastos/metabolismo , Salud , Humanos , Masculino , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Linaje , Inhibidores de Proteasoma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/metabolismo , Piel/patología
6.
J Mol Med (Berl) ; 84(8): 682-91, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16715312

RESUMEN

Cardiomyopathies are complex myocardial diseases characterized by inappropriate ventricular hypertrophy (HCM) or dilation (DCM). Both disorders may lead to sudden death or progressive heart failure and exhibit familial aggregation with marked genetic heterogeneity. Many candidate genes were identified by linkage analysis, experimental animal studies, and expression analysis. A systematic assessment of the prevalence of different mutations in these genes requires high-throughput analyses. In this paper, we present a simple and reliable protocol for mutation screening by heteroduplex analysis which reduced costs and workload of sequencing. Employing denaturing gradient gel electrophoresis (DGGE), 11 known and 14 potential candidate genes for HCM and DCM were analyzed. DGGE assays allowed analysis of 286 of the 312 protein coding exons, performing only four alternative polymerase chain reaction protocols and only two different DGGE analysis conditions. Sensitivity for the detection of heteroduplexes proved excellent, even for GC-rich DNA fragments, which were analyzed by a combination of DGGE and constant denaturant gel electrophoresis. To confirm DGGE sensitivity in cases where no variants in our human DNA samples could be observed, we generated heteroduplexes from homologous human and chimpanzee DNA. The platform proved a valuable contribution to elucidating the genetic causes of DCM and HCM as demonstrated by the identification of 17 different known and novel mutations and 98 different polymorphisms in our setting.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Adolescente , Adulto , Secuencia de Bases , Niño , Preescolar , Electroforesis en Gel de Poliacrilamida , Femenino , Perfilación de la Expresión Génica , Humanos , Lactante , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Linaje , Proyectos Piloto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA