Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Proc Natl Acad Sci U S A ; 107(50): 21830-5, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21098287

RESUMO

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.


Assuntos
Metformina/farmacologia , Emaranhados Neurofibrilares/metabolismo , Proteína Fosfatase 2/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas tau/metabolismo , Adenilato Quinase/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Epitopos , Células HeLa , Humanos , Hipoglicemiantes/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos , Emaranhados Neurofibrilares/patologia , Neurônios/citologia , Neurônios/metabolismo , Ácido Okadáico/farmacologia , Fosforilação , Proteína Fosfatase 2/genética , Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Proteínas tau/genética
2.
J Biol Chem ; 286(46): 39945-57, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21930711

RESUMO

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.


Assuntos
Proteínas dos Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/biossíntese , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Esôfago/anormalidades , Esôfago/metabolismo , Células HeLa , Humanos , Hipertelorismo/genética , Hipertelorismo/metabolismo , Hipospadia/genética , Hipospadia/metabolismo , Proteínas dos Microtúbulos/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética
3.
J Mol Med (Berl) ; 86(3): 281-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17987279

RESUMO

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.


Assuntos
Cardiomiopatia Dilatada/genética , Códon sem Sentido/genética , Lamina Tipo A/metabolismo , Proteínas Mutantes/metabolismo , Estabilidade de RNA , Taquicardia Ventricular/genética , Adulto , Alelos , Estudos de Casos e Controles , Núcleo Celular/metabolismo , Cromatina/metabolismo , Regulação para Baixo , Feminino , Fibroblastos/metabolismo , Saúde , Humanos , Masculino , Miocárdio/metabolismo , Proteínas Nucleares/metabolismo , Linhagem , Inibidores de Proteassoma , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Pele/metabolismo , Pele/patologia
4.
Eur J Hum Genet ; 22(1): 119-25, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23695275

RESUMO

So far, the role of mutations in the δ-sarcogylcan (Sgcd) gene in causing autosomal dominant dilated cardiomyopathy (DCM) remains inconclusive. A p.S151A missense mutation in exon 6 of the Sgcd gene was reported to cause severe isolated autosomal dominant DCM without affecting skeletal muscle. This is controversial to our previous findings in a large consanguineous family where this p.S151A mutation showed no relevance for cardiac disease. In this study, the potential of the p.S151A mutation to cause DCM was investigated by using two different approaches: (1) engineering and characterization of heterozygous knock-in (S151A-) mice carrying the p.S151A mutation and (2) evaluation of the potential of adeno-associated virus (AAV) 9-based cardiac-specific transfer of p.S151A-mutated Sgcd cDNA to rescue the cardiac phenotype in Sgcd-deficient (Sgcd-null) mice as it has been demonstrated for intact, wild-type Sgcd cDNA. Heterozygous S151A knock-in mice developed a rather mild phenotype of cardiomyopathy. Increased heart to body weight suggests cardiac enlargement in 1-year-old S151A knock-in mice. However, at this age cardiac function, assessed by echocardiography, is maintained and histopathology completely absent. Myocardial expression of p.S151A cDNA, similar to intact Sgcd cDNA, restores cardiac function, although not being able to prevent myocardial histopathology in Sgcd-null mice completely. Our results suggest that the p.S151A mutation causes a mild, subclinical phenotype of cardiomyopathy, which is prone to be overseen in patients carrying such sequence variants. Furthermore, this study shows the suitability of an AAV-mediated cardiac gene transfer approach to analyze whether a sequence variant is a disease-causing mutation.


Assuntos
Cardiomiopatias/genética , Mutação de Sentido Incorreto , Sarcoglicanas/genética , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Dependovirus , Técnicas de Introdução de Genes , Heterozigoto , Humanos , Camundongos , Miocárdio/patologia , Fenótipo
5.
Nat Commun ; 4: 1511, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23443539

RESUMO

Expansion of CAG repeats is a common feature of various neurodegenerative disorders, including Huntington's disease. Here we show that expanded CAG repeats bind to a translation regulatory protein complex containing MID1, protein phosphatase 2A and 40S ribosomal S6 kinase. Binding of the MID1-protein phosphatase 2A protein complex increases with CAG repeat size and stimulates translation of the CAG repeat expansion containing messenger RNA in a MID1-, protein phosphatase 2A- and mammalian target of rapamycin-dependent manner. Our data indicate that pathological CAG repeat expansions upregulate protein translation leading to an overproduction of aberrant protein and suggest that the MID1-complex may serve as a therapeutic target for the treatment of CAG repeat expansion disorders.


Assuntos
Proteínas dos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas/genética , Proteína Fosfatase 2/metabolismo , Fatores de Transcrição/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Animais , Western Blotting , Células HeLa , Humanos , Proteína Huntingtina , Luciferases/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Motivos de Nucleotídeos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ubiquitina-Proteína Ligases
6.
Hum Gene Ther ; 23(6): 566-75, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22248393

RESUMO

Dystrophin plays an important role in muscle contraction, linking the intracellular cytoskeleton to the extracellular matrix. Mutations of the dystrophin gene leading to a complete loss of the protein cause Duchenne muscular dystrophy (DMD), frequently associated with severe cardiomyopathy. Early clinical trials in DMD using gene transfer to skeletal muscle are underway, but gene transfer to dystrophic cardiac muscle has not yet been tested in humans. The aim of this study was to develop an optimized protocol for cardiac gene therapy in the mouse model of dystrophin deficiency (mdx), using a cardiac promoter for expression of a microdystrophin (µDys) transgene packaged into an adeno-associated virus serotype 9 vector (AAV9). In this study adult mdx mice were intravenously injected with 1×10(12) genomic particles of AAV9 vectors carrying a cDNA encoding µDys under the control of either a ubiquitously active cytomegalovirus (CMV) promoter or a cardiac-specific CMV-enhanced myosin light chain (MLC0.26) promoter. After 10 months, both AAV9 vectors led to sustained µDys expression in cardiac muscle, but the MLC promoter conferred about 4-fold higher protein levels. AAV9-CMV-MLC0.26-µDys resulted in significant protection of cardiac morphology and function as assessed by histopathology, echocardiography, and left ventricular catheterization. In conclusion, we established an AAV9-mediated gene transfer approach for efficient and specific long-term µDys expression in the hearts of mdx mice, resulting in a sustained therapeutic effect. Thus, this approach might be a basis for further translation into a treatment strategy for DMD-associated cardiomyopathy.


Assuntos
Dependovirus/genética , Distrofina/genética , Terapia Genética , Miocárdio/citologia , Animais , Western Blotting , Modelos Animais de Doenças , Distrofina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos mdx , Miocárdio/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas
7.
Cardiovasc Res ; 82(3): 404-10, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19218289

RESUMO

AIMS: Delta-sarcoglycan is a member of the dystrophin-associated glycoprotein complex linking the cytoskeleton to the extracellular matrix. Similar to patients with defects in the gene encoding delta-sarcoglycan (Sgcd), knockout mice develop cardiomyopathy and muscular dystrophy. The aim of our study was to develop an approach for preventing cardiomyopathy in Sgcd-deficient mice by cardiac expression of the intact cDNA upon systemic delivery of adeno-associated viral (AAV) vectors. METHODS AND RESULTS: We packaged the Sgcd cDNA under transcriptional control of a myosin light chain-promoter fused with a cytomegalovirus enhancer into AAV-9 capsids. Vectors carrying either the Sgcd cDNA or an enhanced green fluorescent protein (EGFP) reporter gene were intravenously injected into adult Sgcd knockout mice. After 6 months, immunohistochemistry revealed almost complete reconstitution of the sarcoglycan subcomplex in heart but not skeletal muscle of mice with the Sgcd vector. Furthermore, Sgcd gene transfer resulted in prevention of cardiac fibrosis and significantly increased running distance measured by voluntary wheel running. Left ventricular function remained stable in mice expressing Sgcd while it deteriorated in EGFP controls within 6 months, paralleled by increased expression of brain natriuretic peptide, a molecular marker of heart failure. CONCLUSION: Our study establishes an approach to specifically treat hereditary cardiomyopathies by targeting gene expression into the myocardium upon systemic application of AAV vectors.


Assuntos
Cardiomiopatias/prevenção & controle , Dependovirus , Terapia Genética , Miocárdio/metabolismo , Sarcoglicanas/genética , Animais , Teste de Esforço , Técnicas de Transferência de Genes , Vetores Genéticos , Insuficiência Cardíaca/prevenção & controle , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Miocárdio/patologia , Sarcoglicanas/metabolismo , Função Ventricular Esquerda
8.
Hum Genet ; 111(1): 16-25, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12136231

RESUMO

A supernumerary C-band-positive microchromosome was observed in 88% of lymphocyte metaphases from a healthy 24-year-old female. Traditional cytogenetic analyses failed to determine the microchromosome's origin and structure. However, hybridization experiments with a panel of chromosome-specific alpha-satellite probes demonstrated that this microchromosome was derived from chromosome 16 and consisted mainly of transcriptionally inactive alpha-satellite DNA. The microchromosome closely associated with the centromere of most chromosomes. An even more pronounced centromeric association pattern was observed in a further microchromosome that was found to contain chromosome 18-specific alpha-satellite DNA. The latter microchromosome was detected in a female newborn affected with fetal alcohol syndrome. The two microchromosomes described here did not appear to bear major phenotypic risks.


Assuntos
Centrômero/genética , Cromossomos Humanos Par 16/genética , Cromossomos Humanos Par 18/genética , Adulto , Aberrações Cromossômicas , Bandeamento Cromossômico , Sondas de DNA/genética , DNA Satélite , Feminino , Fibroblastos/fisiologia , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Cariotipagem , Linfócitos/fisiologia , Metáfase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA