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











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 9(1): e86470, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24466111

RESUMO

Cancer is a major public health problem worldwide. In the United States alone, 1 in 4 deaths is due to cancer and for 2013 a total of 1,660,290 new cancer cases and 580,350 cancer-related deaths are projected. Comprehensive profiling of multiple cancer genomes has revealed a highly complex genetic landscape in which a large number of altered genes, varying from tumor to tumor, impact core biological pathways and processes. This has implications for therapeutic targeting of signaling networks in the development of treatments for specific cancers. The NFκB transcription factor is constitutively active in a number of hematologic and solid tumors, and many signaling pathways implicated in cancer are likely connected to NFκB activation. A critical mediator of NFκB activity is TGFß-activated kinase 1 (TAK1). Here, we identify TAK1 as a novel interacting protein and target of fibroblast growth factor receptor 3 (FGFR3) tyrosine kinase activity. We further demonstrate that activating mutations in FGFR3 associated with both multiple myeloma and bladder cancer can modulate expression of genes that regulate NFκB signaling, and promote both NFκB transcriptional activity and cell adhesion in a manner dependent on TAK1 expression in both cancer cell types. Our findings suggest TAK1 as a potential therapeutic target for FGFR3-associated cancers, and other malignancies in which TAK1 contributes to constitutive NFκB activation.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Mieloma Múltiplo/metabolismo , NF-kappa B/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Tirosina/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Adesão Celular , Proliferação de Células , Perfilação da Expressão Gênica , Humanos , Imunoprecipitação , MAP Quinase Quinase Quinases/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , NF-kappa B/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fragmentos de Peptídeos , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia
2.
J Huntingtons Dis ; 1(1): 119-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-24086178

RESUMO

The Huntington's disease (HD) mutation leads to a complex process of Huntingtin (Htt) aggregation into multimeric species that eventually form visible inclusions in cytoplasm, nuclei and neuronal processes. One hypothesis is that smaller, soluble forms of amyloid proteins confer toxic effects and contribute to early cell dysfunction. However, analysis of mutant Htt aggregation intermediates to identify conformers that may represent toxic forms of the protein and represent potential drug targets remains difficult. We performed a detailed analysis of aggregation conformers in multiple in vitro, cell and ex vivo models of HD. Conformation-specific antibodies were used to identify and characterize aggregation species, allowing assessment of multiple conformers present during the aggregation process. Using a series of assays together with these antibodies, several forms could be identified. Fibrillar oligomers, defined as having a ß-sheet rich conformation, are observed in vitro using recombinant protein and in protein extracts from cells in culture or mouse brain and shown to be globular, soluble and non-sedimentable structures. Compounds previously described to modulate visible inclusion body formation and reduce toxicity in HD models were also tested and consistently found to alter the formation of fibrillar oligomers. Interestingly, these compounds did not alter the rate of visible inclusion formation, indicating that fibrillar oligomers are not necessarily the rate limiting step of inclusion body formation. Taken together, we provide insights into the structure and formation of mutant Htt fibrillar oligomers that are modulated by small molecules with protective potential in HD models.


Assuntos
Amiloide/química , Amiloide/genética , Doença de Huntington/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Animais , Química Encefálica , Linhagem Celular Tumoral , Humanos , Proteína Huntingtina , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutação/genética
3.
Neurodegener Dis ; 9(2): 104-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21912091

RESUMO

Huntingtin peptides with elongated polyglutamine domains, the root causes of Huntington's disease, hinder histone acetylation, which leads to transcriptional dysregulation. However, the range of acetyltransferases interacting with mutant Huntingtin has not been systematically evaluated. We used genetic interaction tests in Drosophila to determine whether specific acetyltransferases belonging to distinct protein families influence polyglutamine pathology. We found that flies expressing a mutant form of the Huntingtin protein (Httex1pQ93) exhibit reduced viability, which is further decreased by partial loss of Pcaf or nejire, while the tested MYST family acetyltransferases did not affect pathology. Reduced levels of Pcaf also led to the increased degeneration of photoreceptor neurons in the retina. Overexpression of Pcaf, however, was not sufficient to ameliorate these phenotypes, and the level of soluble Pcaf is unchanged in Httex1pQ93-expressing flies. Thus, our results indicate that while Pcaf has a significant impact on Huntington's disease pathology, therapeutic strategies aimed at elevating its levels are likely to be ineffective in ameliorating Huntington's disease pathology; however, strategies that aim to increase the specific activity of Pcaf remain to be tested.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Histona Acetiltransferases/genética , Doença de Huntington/genética , Peptídeos/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histona Acetiltransferases/metabolismo , Doença de Huntington/metabolismo , Immunoblotting , Degeneração Neural/genética , Degeneração Neural/metabolismo , Peptídeos/metabolismo
4.
J Neurosci ; 31(18): 6858-70, 2011 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-21543616

RESUMO

Oncogenic transformation of postmitotic neurons triggers cell death, but the identity of genes critical for degeneration remain unclear. The antitumor antibiotic mithramycin prolongs survival of mouse models of Huntington's disease in vivo and inhibits oxidative stress-induced death in cortical neurons in vitro. We had correlated protection by mithramycin with its ability to bind to GC-rich DNA and globally displace Sp1 family transcription factors. To understand how antitumor drugs prevent neurodegeneration, here we use structure-activity relationships of mithramycin analogs to discover that selective DNA-binding inhibition of the drug is necessary for its neuroprotective effect. We identify several genes (Myc, c-Src, Hif1α, and p21(waf1/cip1)) involved in neoplastic transformation, whose altered expression correlates with protective doses of mithramycin or its analogs. Most interestingly, inhibition of one these genes, Myc, is neuroprotective, whereas forced expression of Myc induces Rattus norvegicus neuronal cell death. These results support a model in which cancer cell transformation shares key genetic components with neurodegeneration.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neurônios/efeitos dos fármacos , Plicamicina/análogos & derivados , Plicamicina/farmacologia , Fator de Transcrição Sp1/metabolismo , Análise de Variância , Animais , Animais Geneticamente Modificados , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Imunoprecipitação da Cromatina , Drosophila , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Transcrição Sp1/genética , Relação Estrutura-Atividade
5.
J Biol Chem ; 284(43): 29427-36, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19710014

RESUMO

Huntingtin (Htt) is a widely expressed protein that causes tissue-specific degeneration when mutated to contain an expanded polyglutamine (poly(Q)) domain. Although Htt is large, 350 kDa, the appearance of amino-terminal fragments of Htt in extracts of postmortem brain tissue from patients with Huntington disease (HD), and the fact that an amino-terminal fragment, Htt exon 1 protein (Httex1p), is sufficient to cause disease in models of HD, points to the importance of the amino-terminal region of Htt in the disease process. The first exon of Htt encodes 17 amino acids followed by a poly(Q) repeat of variable length and culminating with a proline-rich domain of 50 amino acids. Because modifications to this fragment have the potential to directly affect pathogenesis in several ways, we have surveyed this fragment for potential post-translational modifications that might affect Htt behavior and detected several modifications of Httex1p. Here we report that the most prevalent modifications of Httex1p are NH(2)-terminal acetylation and phosphorylation of threonine 3 (pThr-3). We demonstrate that pThr-3 occurs on full-length Htt in vivo, and that this modification affects the aggregation and pathogenic properties of Htt. Thus, therapeutic strategies that modulate these events could in turn affect Htt pathogenesis.


Assuntos
Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Processamento de Proteína Pós-Traducional , Treonina/metabolismo , Acetilação , Células HeLa , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Treonina/química , Treonina/genética
6.
Hum Mol Genet ; 18(11): 1951-61, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19286672

RESUMO

Ectopic activation of fibroblast growth factor receptor 3 (FGFR3) is associated with several cancers, including multiple myeloma (MM). FGFR3 inhibition in these cells inhibits proliferation and induces apoptosis, validating FGFR3 signaling as a therapeutic target in t(4;14) MM cases. We have identified the PI3K regulatory subunit, p85alpha, as a novel interactor of FGFR3 by yeast two-hybrid, and confirmed an interaction with both p85alpha and p85beta in mammalian cells. The interaction of FGFR3 with p85 is dependent upon receptor activation. In contrast to the Gab1-mediated association of FGFRs with p85, the FGFR3-p85 interaction we observed requires FGFR3 Y760, previously identified as a PLCgamma binding site. The interaction of p85 with FGFR3 does not require PLCgamma, suggesting the p85 interaction is direct and independent of PLCgamma binding. FGFR3 and p85 proteins also interact in MM cell lines which consistently express p85alpha and p85beta, but not p50 or p55 subunits. siRNA knockdown of p85beta in MM cells caused an increased ERK response to FGF2. These data suggest that an endogenous negative regulatory role for the p85-FGFR3 interaction on the Ras/ERK/MAPK pathway may exist in response to FGFR3 activity and identifies a novel therapeutic target for MM.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mieloma Múltiplo/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Ativação Transcricional , Sítios de Ligação , MAP Quinases Reguladas por Sinal Extracelular/genética , Células HeLa , Humanos , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/genética , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/genética , Ligação Proteica , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética
7.
J Biol Chem ; 284(12): 7431-5, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-18957429

RESUMO

The dominant gain-of-function polyglutamine repeat diseases, in which the initiating mutation is known, allow development of models that recapitulate many aspects of human disease. To the extent that pathology is a consequence of disrupted fundamental cellular activities, one can effectively study strategies to ameliorate or protect against these cellular insults. Model organisms allow one to identify pathways that affect disease onset and progression, to test and screen for pharmacological agents that affect pathogenic processes, and to validate potential targets genetically as well as pharmacologically. Here, we describe polyglutamine repeat diseases that have been modeled in a variety of organisms, including worms, flies, mice, and non-human primates, and discuss examples of how they have broadened the therapeutic landscape.


Assuntos
Modelos Animais de Doenças , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Peptídeos/genética , Animais , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Transtornos Heredodegenerativos do Sistema Nervoso/terapia , Humanos , Peptídeos/metabolismo
8.
Cell Signal ; 21(1): 151-60, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18950705

RESUMO

Activation of fibroblast growth factor receptor 3 (FGFR3) leads to attenuation of cartilage growth. The members of the STAT family of transcription factors are believed to participate in FGFR3 signaling in cartilage, however the molecular mechanism of this action is poorly understood. Here, we demonstrate that a chronic FGF stimulus leads to accumulation of STAT1, 3, 5 and 6, evident in both in vitro chondrocyte model and murine limb explant cultures. Despite the accumulation, both endogenous and cytokine-induced activation of STAT1 and STAT3 is impaired by FGF, as demonstrated by imaging of active STAT nuclear translocation and analyses of STAT activatory phosphorylation and transcriptional activation. Further, we demonstrate that FGF induces expression of CIS, SOCS1 and SOCS3 inhibitors of gp130, a common receptor for the IL6-family of cytokines. Since cytokine-gp130 signaling represents an important positive regulator of cartilage, its inhibition may contribute to the growth-inhibitory effect of FGFR3 in cartilage.


Assuntos
Condrócitos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Interferon gama/farmacologia , Interleucina-6/farmacologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Sequência de Bases , Condrócitos/efeitos dos fármacos , Receptor gp130 de Citocina/metabolismo , Interferon gama/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Camundongos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo
9.
PLoS One ; 3(12): e3961, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19088846

RESUMO

Activating mutations in FGFR3 tyrosine kinase cause several forms of human skeletal dysplasia. Although the mechanisms of FGFR3 action in cartilage are not completely understood, it is believed that the STAT1 transcription factor plays a central role in pathogenic FGFR3 signaling. Here, we analyzed STAT1 activation by the N540K, G380R, R248C, Y373C, K650M and K650E-FGFR3 mutants associated with skeletal dysplasias. In a cell-free kinase assay, only K650M and K650E-FGFR3 caused activatory STAT1(Y701) phosphorylation. Similarly, in RCS chondrocytes, HeLa, and 293T cellular environments, only K650M and K650E-FGFR3 caused strong STAT1 activation. Other FGFR3 mutants caused weak (HeLa) or no activation (293T and RCS). This contrasted with ERK MAP kinase activation, which was strongly induced by all six mutants and correlated with the inhibition of proliferation in RCS chondrocytes. Thus the ability to activate STAT1 appears restricted to the K650M and K650E-FGFR3 mutants, which however account for only a small minority of the FGFR3-related skeletal dysplasia cases. Other pathways such as ERK should therefore be considered as central to pathological FGFR3 signaling in cartilage.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Cartilagem/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/fisiologia , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/fisiologia , Animais , Doenças do Desenvolvimento Ósseo/metabolismo , Doenças do Desenvolvimento Ósseo/patologia , Osso e Ossos/patologia , Células CHO , Sistema Livre de Células/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HeLa , Humanos , Modelos Biológicos , Proteínas Mutantes/fisiologia , Fosforilação , Ratos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Fator de Transcrição STAT1/análise , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
11.
J Cell Sci ; 121(Pt 3): 272-81, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18198189

RESUMO

Activating mutations in fibroblast growth factor receptor 3 (FGFR3) cause several human skeletal dysplasias as a result of attenuation of cartilage growth. It is believed that FGFR3 inhibits chondrocyte proliferation via activation of signal transducers and activators of transcription (STAT) proteins, although the exact mechanism of both STAT activation and STAT-mediated inhibition of chondrocyte growth is unclear. We show that FGFR3 interacts with STAT1 in cells and is capable of activating phosphorylation of STAT1 in a kinase assay, thus potentially serving as a STAT1 kinase in chondrocytes. However, as demonstrated by western blotting with phosphorylation-specific antibodies, imaging of STAT nuclear translocation, STAT transcription factor assays and STAT luciferase reporter assays, FGF does not activate STAT1 or STAT3 in RCS chondrocytes, which nevertheless respond to a FGF stimulus with potent growth arrest. Moreover, addition of active STAT1 and STAT3 to the FGF signal, by means of cytokine treatment, SRC-mediated STAT activation or expression of constitutively active STAT mutants does not sensitize RCS chondrocytes to FGF-mediated growth arrest. Since FGF-mediated growth arrest is rescued by siRNA-mediated downregulation of the MAP kinase ERK1/2 but not STAT1 or STAT3, our data support a model whereby the ERK arm but not STAT arm of FGF signaling in chondrocytes accounts for the growth arrest phenotype.


Assuntos
Condrócitos/citologia , Condrócitos/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Sequência de Bases , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Interferon gama/farmacologia , Interleucina-6/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transdução de Sinais , Transfecção
12.
Hum Mol Genet ; 16(1): 61-77, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17135277

RESUMO

A truncated form of the Huntington's disease (HD) protein that contains the polyglutamine repeat, Httex1p, causes HD-like phenotypes in multiple model organisms. Molecular signatures of pathogenesis appear to involve distinct domains within this polypeptide. We studied the contribution of each domain, singly or in combination, to sub-cellular localization, aggregation and intracellular Ca2+ ([Ca2+]i) dynamics in cells. We demonstrate that sub-cellular localization is most strongly influenced by the first 17 amino acids, with this sequence critically controlling Httex1p mitochondrial localization and also promoting association with the endoplasmic reticulum (ER) and Golgi. This domain also enhances the formation of visible aggregates and together with the expanded polyQ repeat acutely disrupts [Ca2+]i levels in glutamate-challenged PC12 cells. Isolated cortical mitochondria incubated with Httex1p resulted in uncoupling and depolarization of these organelles, further supporting the idea that Httex1p-dependent mitochondrial dysfunction could be instrumental in promoting acute Ca2+ dyshomeostasis. Interestingly, neither mitochondrial nor ER associations seem to be required to promote long-term [Ca2+]i dyshomeostasis.


Assuntos
Cálcio/metabolismo , Citoplasma/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/química , Animais , Membrana Celular/metabolismo , Córtex Cerebelar/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Homeostase , Proteína Huntingtina , Masculino , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Células PC12 , Peptídeos/genética , Prolina/química , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transfecção
13.
J Biol Chem ; 282(5): 2929-36, 2007 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-17145761

RESUMO

Fibroblast growth factors (FGFs) inhibit chondrocyte proliferation via the Erk MAP kinase pathway. Here, we explored the role of protein kinase C in FGF signaling in chondrocytes. Erk activity in FGF2-treated RCS (rat chondrosarcoma) chondrocytes or human primary chondrocytes was abolished by the protein kinase C inhibitor bisindolylmaleimide I (Bis I). Bis I inhibited FGF2-induced activation of MEK, Raf-1, and Ras members of Erk signaling module but not the FGF2-induced tyrosine phosphorylation of Frs2 or the kinase activity of FGFR3, demonstrating that it targets the Erk cascade immediately upstream of Ras. Indeed, Bis I abolished the FGF2-mediated association of Shp2 tyrosine phosphatase with Frs2 and Gab1 adaptor proteins necessary for proper Ras activation. We also determined which PKC isoform is involved in FGF2-mediated activation of Erk. When both conventional and novel PKCs expressed by RCS chondrocytes (PKCalpha, -gamma, -delta, and -epsilon) were down-regulated by phorbol ester, cells remained responsive to FGF2 with Erk activation, and this activation was sensitive to Bis I. Moreover, treatment with PKClambda/zeta pseudosubstrate lead to significant reduction of FGF2-mediated activation of Erk, suggesting involvement of an atypical PKC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Condrócitos/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Indóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Maleimidas/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Animais , Células CHO , Linhagem Celular Tumoral , Condrócitos/efeitos dos fármacos , Cricetinae , Cricetulus , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Ratos
14.
Proc Natl Acad Sci U S A ; 103(39): 14367-72, 2006 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16980414

RESUMO

The dependence of disease risk and age-of-onset on expanded CAG repeat length in diseases like Huntington's disease (HD) is well established and correlates with the repeat-length-dependent nucleation kinetics of polyglutamine (polyGln) aggregation. The wide variation in ages of onset among patients with the same repeat length, however, suggests a role for modifying factors. Here we describe the ability of normal-length polyGln repeat sequences to greatly accelerate the nucleation kinetics of an expanded polyGln peptide. We find that normal-length polyGln peptides enhance the in vitro nucleation kinetics of a Q(47) peptide in a concentration-dependent and repeat-length-dependent manner. In vivo, we show that coexpression of a Q(20) sequence in a Drosophila model of HD expressing Htt exon 1 protein with an Q(93) repeat accelerates both aggregate formation and neurotoxicity. The accelerating effect of short polyGln peptides is attributable to the promiscuity of polyGln aggregate elongation and reflects the intimate relationship between nucleus formation and early elongation events in establishing nucleation kinetics. The results suggest that the overall state of the polyGln protein network in a cellular environment may have a profound effect on the toxic consequences of polyGln expansion and thus may serve as a genetic modifier of age of onset in HD.


Assuntos
Peptídeos/química , Estrutura Quaternária de Proteína , Proteínas/química , Sequências Repetitivas de Aminoácidos , Animais , Morte Celular , Drosophila melanogaster/citologia , Olho/citologia , Olho/patologia , Humanos , Cinética , Modelos Animais , Doenças Neurodegenerativas/induzido quimicamente , Peptídeos/metabolismo , Peptídeos/toxicidade , Relação Estrutura-Atividade
15.
Hum Mol Genet ; 15(13): 2114-24, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16720620

RESUMO

Polyglutamine expansion in certain proteins causes neurodegeneration in inherited disorders such as Huntington disease and X-linked spinobulbar muscular atrophy. Polyglutamine tracts promote protein aggregation in vitro and in vivo with a strict length-dependence that strongly implicates alternative protein folding and/or aggregation as a proximal cause of cellular toxicity and neurodegeneration. We used an intracellular polyglutamine protein aggregation assay based on fluorescence resonance energy transfer (FRET) to identify inhibitors of androgen receptor (AR) aggregation in three libraries of biologically active small molecules: the Annotated Compound Library, the NINDS Custom Collection and a kinase inhibitor collection. In the primary screen 10 compounds reduced AR aggregation. While 10/10 also reduced huntingtin (Htt) exon 1 aggregation, only 2/10 reduced aggregation of pure polyglutamine peptides. In a PC-12 model 9/10 compounds reduced aggregation. Five out of nine compounds tested in an Htt exon 1 assay of neurodegeneration in Drosophila partially rescued the phenotype. Three of the five compounds effective in flies are FDA-approved drugs. These compounds provide new leads for therapeutic development for the polyglutamine diseases based on their efficacy in mammalian cells and a Drosophila model. The high predictive value of the primary screen suggests that the FRET-based screening assay may be useful for further primary and secondary screens for genes or small molecules that inhibit polyglutamine protein aggregation.


Assuntos
Antagonistas de Receptores de Andrógenos , Drogas em Investigação/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Peptídeos/antagonistas & inibidores , Animais , Bioensaio/métodos , Linhagem Celular , Relação Dose-Resposta a Droga , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/química , Drogas em Investigação/uso terapêutico , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Proteína Huntingtina , Doença de Huntington/tratamento farmacológico , Doença de Huntington/metabolismo , Estrutura Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Células PC12 , Peptídeos/química , Peptídeos/metabolismo , Dobramento de Proteína , Ratos , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Relação Estrutura-Atividade , Expansão das Repetições de Trinucleotídeos/efeitos dos fármacos
16.
Hum Mol Genet ; 14(11): 1529-38, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843401

RESUMO

Mutations in fibroblast growth factor receptor 3 (FGFR3) cause the most common genetic form of short-limbed dwarfism, achondroplasia (ACH), as well as neonatal lethal forms, thanatophoric dysplasia (TD) I and II. The causative mutations induce graded levels of constitutive activation of the receptor that correspond to the severity of the disorder, resulting in premature entry into hypertrophic differentiation and reduced proliferation of chondrocytes in developing cartilage. Although FGFR3 promotes growth in most tissues, it is a negative regulator of endochondral bone growth. Several signaling pathways have been implicated in these skeletal disorders including the Ras/MEK/ERK pathway and the JAK/STAT, the latter in the most severe phenotypes, however their functional relevance remains incompletely understood. Using PC12 cell lines stably expressing inducible mutant receptors containing the TDII mutation, K650E, sustained activation of ERK1/2 and activation of STAT1 and STAT3, but not STAT5, is observed in the absence of ligand. This activation leads to neurite outgrowth, a phenotypic readout of constitutive receptor activity, and sustained ERK1/2 activity is required for this ligand-independent differentiation. To assess the functional relevance of STAT activation induced by the mutant receptor, STATs were specifically downregulated using RNA-interference. Silencing of STAT1 or 3 independently or in combination had no significant effect on ligand-independent neurite outgrowth, ERK1/2 activation or p21(WAF1/CIP1) protein levels. These results support a model in which sustained activation of ERK1/2 is a key regulator of the increased transition to hypertrophic differentiation of the growth plate, whereas activation of STATs 1 and 3 is not required.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Displasia Tanatofórica/metabolismo , Animais , Sequência de Bases , Northern Blotting , Primers do DNA , Ativação Enzimática , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Células PC12 , Fenótipo , Fosforilação , Ratos , Displasia Tanatofórica/enzimologia , Displasia Tanatofórica/genética
17.
Proc Natl Acad Sci U S A ; 102(3): 892-7, 2005 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-15642944

RESUMO

Polyglutamine (polyQ) disorders, including Huntington's disease (HD), are caused by expansion of polyQ-encoding repeats within otherwise unrelated gene products. In polyQ diseases, the pathology and death of affected neurons are associated with the accumulation of mutant proteins in insoluble aggregates. Several studies implicate polyQ-dependent aggregation as a cause of neurodegeneration in HD, suggesting that inhibition of neuronal polyQ aggregation may be therapeutic in HD patients. We have used a yeast-based high-throughput screening assay to identify small-molecule inhibitors of polyQ aggregation. We validated the effects of four hit compounds in mammalian cell-based models of HD, optimized compound structures for potency, and then tested them in vitro in cultured brain slices from HD transgenic mice. These efforts identified a potent compound (IC50=10 nM) with long-term inhibitory effects on polyQ aggregation in HD neurons. Testing of this compound in a Drosophila HD model showed that it suppresses neurodegeneration in vivo, strongly suggesting an essential role for polyQ aggregation in HD pathology. The aggregation inhibitors identified in this screen represent four primary chemical scaffolds and are strong lead compounds for the development of therapeutics for human polyQ diseases.


Assuntos
Anilidas/farmacologia , Doença de Huntington/patologia , Doenças Neurodegenerativas/prevenção & controle , Neurônios/patologia , Peptídeos/antagonistas & inibidores , Hidrocarbonetos Policíclicos Aromáticos/farmacologia , Sulfonamidas/farmacologia , Animais , Encéfalo/patologia , Dimerização , Modelos Animais de Doenças , Drosophila , Doença de Huntington/metabolismo , Concentração Inibidora 50 , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Relação Estrutura-Atividade
18.
Neuron ; 40(4): 685-94, 2003 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-14622574

RESUMO

Many neurodegenerative diseases, including tauopathies, Parkinson's disease, amyotrophic lateral sclerosis, and the polyglutamine diseases, are characterized by intracellular aggregation of pathogenic proteins. It is difficult to study modifiers of this process in intact cells in a high-throughput and quantitative manner, although this could facilitate molecular insights into disease pathogenesis. Here we introduce a high-throughput assay to measure intracellular polyglutamine protein aggregation using fluorescence resonance energy transfer (FRET). We screened over 2800 biologically active small molecules for inhibitory activity and have characterized one lead compound in detail. Y-27632, an inhibitor of the Rho-associated kinase p160ROCK, diminished polyglutamine protein aggregation (EC(50) congruent with 5 microM) and reduced neurodegeneration in a Drosophila model of polyglutamine disease. This establishes a novel high-throughput approach to study protein misfolding and aggregation associated with neurodegenerative diseases and implicates a signaling pathway of previously unrecognized importance in polyglutamine protein processing.


Assuntos
Amidas/farmacologia , Bioensaio/métodos , Inibidores Enzimáticos/farmacologia , Transferência Ressonante de Energia de Fluorescência/métodos , Peptídeos/antagonistas & inibidores , Peptídeos/análise , Piridinas/farmacologia , Amidas/uso terapêutico , Animais , Animais Geneticamente Modificados , Células COS , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Drosophila melanogaster , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/uso terapêutico , Humanos , Proteína Huntingtina , Corpos de Inclusão/química , Corpos de Inclusão/efeitos dos fármacos , Corpos de Inclusão/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Peptídeos/metabolismo , Células Fotorreceptoras de Invertebrados/efeitos dos fármacos , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Invertebrados/patologia , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Expansão das Repetições de Trinucleotídeos/efeitos dos fármacos , Expansão das Repetições de Trinucleotídeos/genética , Quinases Associadas a rho
19.
Proc Natl Acad Sci U S A ; 100(10): 5950-5, 2003 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-12730384

RESUMO

The formation of polyglutamine-containing aggregates and inclusions are hallmarks of pathogenesis in Huntington's disease that can be recapitulated in model systems. Although the contribution of inclusions to pathogenesis is unclear, cell-based assays can be used to screen for chemical compounds that affect aggregation and may provide therapeutic benefit. We have developed inducible PC12 cell-culture models to screen for loss of visible aggregates. To test the validity of this approach, compounds that inhibit aggregation in the PC12 cell-based screen were tested in a Drosophila model of polyglutamine-repeat disease. The disruption of aggregation in PC12 cells strongly correlates with suppression of neuronal degeneration in Drosophila. Thus, the engineered PC12 cells coupled with the Drosophila model provide a rapid and effective method to screen and validate compounds.


Assuntos
Drosophila/genética , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Terapia Genética , Peptídeos/genética , Animais , Agregação Celular , Diferenciação Celular , Divisão Celular , Sobrevivência Celular , Cisteína Endopeptidases/metabolismo , Modelos Animais de Doenças , Éxons , Feminino , Humanos , Complexos Multienzimáticos/metabolismo , Neuritos/fisiologia , Neuritos/ultraestrutura , Células PC12 , Peptídeos/antagonistas & inibidores , Fenótipo , Complexo de Endopeptidases do Proteassoma , Ratos , Deleção de Sequência , Transfecção
20.
Expert Opin Ther Targets ; 7(2): 201-13, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12667098

RESUMO

Huntington's disease (HD) is one of a number of familial polyglutamine (polyQ) repeat diseases. These neurodegenerative disorders are caused by expression of otherwise unrelated proteins that contain an expansion of a polyQ tract, rendering them toxic to specific subsets of vulnerable neurons. These expanded repeats have an inherent propensity to aggregate; insoluble neuronal nuclear and cytoplasmic polyQ aggregates or inclusions are hallmarks of the disorders [1,2]. In HD, inclusions in diseased brains often precede onset of symptoms, and have been proposed to be involved in pathogenicity [3-5]. Various strategies to block the process of aggregation have been developed in an effort to create drugs that decrease neurotoxicity. A discussion of the effect of antibodies, caspase inhibitors, chemical inhibitors, heat-shock proteins, suppressor peptides and transglutaminase inhibitors upon aggregation and disease is presented.


Assuntos
Expansão das Repetições de DNA , Desenho de Fármacos , Doença de Huntington/tratamento farmacológico , Proteínas do Tecido Nervoso/efeitos dos fármacos , Nootrópicos/farmacologia , Proteínas Nucleares/efeitos dos fármacos , Peptídeos/genética , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/efeitos dos fármacos , Proteínas de Caenorhabditis elegans/fisiologia , Inibidores de Cisteína Proteinase/farmacologia , Inibidores de Cisteína Proteinase/uso terapêutico , Proteínas de Drosophila/química , Proteínas de Drosophila/efeitos dos fármacos , Proteínas de Drosophila/fisiologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Corpos de Inclusão Intranuclear/química , Corpos de Inclusão Intranuclear/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Chaperonas Moleculares/efeitos dos fármacos , Chaperonas Moleculares/fisiologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Nootrópicos/uso terapêutico , Proteínas Nucleares/química , Proteínas Nucleares/genética , Conformação Proteica , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Interferência de RNA , Sequências Repetitivas de Aminoácidos , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA