Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Med ; 12(1): 128-32, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16327803

RESUMO

Many heat-shock proteins (Hsp) are members of evolutionarily conserved families of chaperone proteins that inhibit the aggregation of unfolded polypeptides and refold denatured proteins, thereby remedying phenotypic effects that may result from protein aggregation or protein instability. Here we report that the mitochondrial chaperone Hsp40, also known as Dnaja3 or Tid1, is differentially expressed during cardiac development and pathological hypertrophy. Mice deficient in Dnaja3 developed dilated cardiomyopathy (DCM) and died before 10 weeks of age. Progressive respiratory chain deficiency and decreased copy number of mitochondrial DNA were evident in cardiomyocytes lacking Dnaja3. Profiling of Dnaja3-interacting proteins identified the alpha-subunit of DNA polymerase gamma (Polga) as a client protein. These findings suggest that Dnaja3 is crucial for mitochondrial biogenesis, at least in part, through its chaperone activity on Polga and provide genetic evidence of the necessity for mitochondrial Hsp40 in preventing DCM.


Assuntos
Cardiomiopatia Dilatada/patologia , DNA Polimerase Dirigida por DNA/metabolismo , Proteínas de Choque Térmico HSP40/química , Proteínas de Choque Térmico HSP40/fisiologia , Animais , Aorta/patologia , Cardiomiopatia Dilatada/prevenção & controle , Linhagem Celular , DNA/metabolismo , DNA Polimerase gama , DNA Mitocondrial/metabolismo , Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Evolução Molecular , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP40/metabolismo , Humanos , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Ligação Proteica , Desnaturação Proteica , Dobramento de Proteína , Renaturação Proteica , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção
2.
Mol Ther ; 20(4): 849-59, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22273578

RESUMO

HIV-1-infected individuals can harbor viral isolates that can use CCR5, as well as CXCR4, for viral entry. To genetically engineer HIV-1 resistance in CD4(+) T cells, we assessed whether transient, adenovirus delivered zinc-finger nuclease (ZFN) disruption of genomic cxcr4 or stable lentiviral expression of short hairpin RNAs (shRNAs) targeting CXCR4 mRNAs provides durable resistance to HIV-1 challenge. ZFN-modification of cxcr4 in CD4(+) T cells was found to be superior to cell integrated lentivirus-expressing CXCR4 targeting shRNAs when CD4(+) T cells were challenged with HIV-1s that utilizes CXCR4 for entry. Cxcr4 disruption in CD4(+) T cells was found to be stable, conferred resistance, and provided for continued cell enrichment during HIV-1 infection in tissue culture and, in vivo, in peripheral blood mononuclear cell transplanted NSG mice. Moreover, HIV-1-infected mice with engrafted cxcr4 ZFN-modified CD4(+) T cells demonstrated lower viral levels in contrast to mice engrafted with unmodified CD4(+) T cells. These findings provide evidence that ZFN-mediated disruption of cxcr4 provides a selective advantage to CD4(+) T cells during HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Endonucleases/metabolismo , HIV-1/imunologia , RNA Interferente Pequeno/genética , Receptores CXCR4/genética , Adenoviridae/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Endonucleases/genética , Humanos , Camundongos , Dedos de Zinco/genética
3.
J Biol Chem ; 284(25): 16752-16758, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19406746

RESUMO

Hypoxia-inducible factor-1 (HIF-1) plays a central role in tumor progression by regulating genes involved in proliferation, glycolysis, angiogenesis, and metastasis. To improve our understanding of HIF-1 regulation by kinome, we screened a kinase-specific small interference RNA library using a hypoxia-response element (HRE) luciferase reporter assay under hypoxic conditions. This screen determined that depletion of cellular SMG-1 kinase most significantly modified cellular HIF-1 activity in hypoxia. SMG-1 is the newest and least studied member of the phosphoinositide 3-kinase-related kinase family, which consists of ATM, ATR, DNA-PKcs, mTOR, and SMG-1. We individually depleted members of the phosphoinositide 3-kinase-related kinase family, and only SMG-1 deficiency significantly augmented HIF-1 activity in hypoxia. We subsequently discovered that SMG-1 kinase activity was activated by hypoxia, and depletion of SMG-1 up-regulated MAPK activity under low oxygen. Suppressing cellular MAPK by silencing ERK1/2 or by treatment with U0126, a MAPK inhibitor, partially blocked the escalation of HIF-1 activity resulting from SMG-1 deficiency in hypoxic cells. Increased expression of SMG-1 but not kinase-dead SMG-1 effectively inhibited the activity of HIF-1alpha. In addition, cellular SMG-1 deficiency increased secretion of the HIF-1alpha-regulated angiogenic factor, vascular epidermal growth factor, and survival factor, carbonic anhydrase IX (CA9), as well as promoted the hypoxic cell motility. Taken together, we discovered that SMG-1 negatively regulated HIF-1alpha activity in hypoxia, in part through blocking MAPK activation.


Assuntos
Hipóxia Celular/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , RNA Interferente Pequeno/genética , Antígenos de Neoplasias/biossíntese , Anidrase Carbônica IX , Anidrases Carbônicas/biossíntese , Hipóxia Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Biblioteca Gênica , Células HeLa , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Inibidores de Fosfoinositídeo-3 Quinase , Análise Serial de Proteínas , Proteínas Serina-Treonina Quinases , Proteoma , Fator A de Crescimento do Endotélio Vascular/biossíntese
4.
Biochem J ; 421(2): 301-10, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19426145

RESUMO

TLR7 (Toll-like receptor 7) mediates anti-viral immunity by recognizing ssRNA (single-stranded RNA) viruses. Small-molecular-mass TLR7 agonists have been approved, or are being evaluated, for treatment of cancers or infectious diseases. Although TLR7 is predominantly expressed in a restricted set of immune cell types, including pDCs (plasmacytoid dendritic cells), it is also expressed in non-native expressing cells (e.g. hepatocytes) under certain circumstances. To elucidate the molecular basis of TLR7 induction by pro-inflammatory stimulation and the subsequent cellular responses in these non-native TLR7-expressing cell types, we first cloned and characterized the 5'-promoter region of TLR7. The proximal region of this promoter drives the transcription of the TLR7 gene. Pro-inflammatory stimuli activated TLR 7 transcription via a NF-kappaB (nuclear factor kappaB)-binding motif in this region, and this activation could be blocked by mutation of the NF-kappaB binding site or addition of NF-kappaB inhibitors. Further studies showed that pretreatment of the Hep3B hepatocytes with TNF-alpha (tumour necrosis factor-alpha) or IL-1 (interleukin-1) rendered them responsive to TLR7 activation by a TLR7 agonist. However, distinct from TLR7 activation in pDCs, which respond to stimulation with Th1 polarized cytokine production, TLR7 induction by pro-inflammatory signals in hepatocytes reconstitutes the NF-kappaB-dependent cascade but not the IRF7 (interferon regulatory factor 7)-dependent cascade, resulting in a pro-inflammatory polarized response rather than a Th1 polarized response. These results indicate that inflammatory stimulation is capable of priming cells to respond to TLR7 agonist with an immune response that differs from that in native TLR7-expressing cells.


Assuntos
Mediadores da Inflamação/farmacologia , NF-kappa B/metabolismo , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/genética , Ativação Transcricional , Sequência de Bases , Humanos , Interleucina-1/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Receptor 7 Toll-Like/biossíntese , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
5.
J Leukoc Biol ; 82(1): 177-83, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17403772

RESUMO

Nucleotide-binding oligomerization domain (Nod)2 is a sensor of muramyl dipeptides (MDP) derived from bacterial peptidoglycan. Nod2 also plays a role in some autoinflammatory diseases. Cold-induced autoinflammatory syndrome 1 (CIAS1)/NACHT domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NALP3) has been suggested to be sufficient for MDP-dependent release of mature IL-1beta, but the role of Nod2 in this process is unclear. Using mice bearing selective gene deletions, we provide in vitro and in vivo data showing that MDP-induced IL-1beta release requires Nod2 and CIAS1/NALP3 as well as receptor-interacting protein-2 (Rip2), apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), and caspase-1. In contrast, MDP-dependent IL-6 production only requires Nod2 and Rip2. Together, our data provide a new understanding of this important pathway of IL-1beta production and allow for further studies of the role of these proteins within the broader context of inflammatory disease.


Assuntos
Acetilmuramil-Alanil-Isoglutamina/farmacologia , Proteínas de Transporte/fisiologia , Interleucina-1beta/biossíntese , Proteína Adaptadora de Sinalização NOD2/fisiologia , Adjuvantes Imunológicos/farmacologia , Animais , Inflamação , Interleucina-6/biossíntese , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteína Serina-Treonina Quinase 2 de Interação com Receptor , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia
6.
Mol Cell Biol ; 24(6): 2226-36, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14993262

RESUMO

Tid1 is the mammalian counterpart of the Drosophila tumor suppressor Tid56 and is also a DnaJ protein containing a conserved J domain through which it interacts with the heat shock protein 70 (Hsp70) family of chaperone proteins. We generated a Tid1 conditional mutation in mice, and the subsequent global removal of the Tid1 protein was achieved by crossing these conditional knockout mice with general deletor mice. No Tid1(-/-) embryos were detected as early as embryonic day 7.5 (E7.5). Nonetheless, Tid1-deficient blastocysts were viable, hatched, formed an inner cell mass and trophectoderm, and implanted (E4.5), suggesting that the homozygous mutant embryos die between E4.5 and E7.5. To assess the function of Tid1 in embryonic cells, mouse embryonic fibroblasts with the homologous Tid1 floxed allele were produced. Tid1 removal in these cells led to massive cell death. The death of Tid1-deficient cells could be rescued by ectopic expression of wild-type Tid1 but not by expression of the Tid1 protein that had a mutated J domain and was thus incapable of binding to Hsp70. We propose that Tid1 is critical for early mammalian development, most likely for its function in sustaining embryonic-cell survival, which requires its association with Hsp70.


Assuntos
Sobrevivência Celular/fisiologia , Desenvolvimento Embrionário e Fetal/fisiologia , Proteínas de Choque Térmico HSP70/fisiologia , Chaperonas Moleculares/fisiologia , Animais , Sequência de Bases , Sobrevivência Celular/genética , DNA Complementar/genética , Proteínas de Ligação a DNA , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Desenvolvimento Embrionário e Fetal/genética , Feminino , Marcação de Genes , Idade Gestacional , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiologia , MAP Quinase Quinase 7 , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutação , Gravidez , Fatores de Transcrição
7.
Cancer Res ; 65(17): 7699-706, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140937

RESUMO

Although big mitogen-activated protein kinase 1 (BMK1) has been shown to be critical for embryonic angiogenesis, the role of BMK1 in tumor-associated neovascularization is poorly understood. Exogenous tumors were established in BMK1+/+, BMK1flox/+, or BMK1flox/flox mice carrying the Mx1-Cre transgene. Induced deletion of host BMK1 gene significantly reduced the volumes of B16F10 and LL/2 tumor xenografts in BMK1flox/flox mice by 63% and 72%, respectively. Examining the tumors in these induced BMK1-knockout animals showed a significant decrease in vascular density. Localized reexpression of BMK1 in BMK1-knockout mice by administration of adenovirus encoding BMK1 restored tumor growth and angiogenesis to the levels observed in wild-type mice. These observations were further supported by in vivo Matrigel plug assays in which vascular endothelial growth factor- and basic fibroblast growth factor-induced neovacularization was impaired by removing BMK1. Through screening with the Pepchip microarray, we discovered that in BMK1-knockout endothelial cells, phosphorylation of ribosomal protein S6 (rpS6) at Ser235/236 was mostly abrogated, and this BMK1-dependent phosphorylation required the activity of p90 ribosomal S6 kinase (RSK). Immunofluorescent analysis of tumor vasculature from BMK1-knockout and control animals revealed a strong correlation between the presence of BMK1 and the phosphorylation of rpS6 in tumor-associated endothelial cells of blood vessels. As both RSK and rpS6 are known to be important for cell proliferation and survival, which are critical endothelial cell functions during neovascularization, these findings suggest that the BMK1 pathway is crucial for tumor-associated angiogenesis through its role in the regulation of the RSK-rpS6 signaling module.


Assuntos
Carcinoma Pulmonar de Lewis/irrigação sanguínea , Sistema de Sinalização das MAP Quinases/fisiologia , Melanoma Experimental/irrigação sanguínea , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neovascularização Patológica/enzimologia , Sequência de Aminoácidos , Animais , Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Proteína Quinase 7 Ativada por Mitógeno/deficiência , Dados de Sequência Molecular , Fosforilação , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Proteínas Ribossômicas/metabolismo , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
Cancer Res ; 65(19): 8784-91, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16204048

RESUMO

Tid1 is the human homologue of the Drosophila tumor suppressor, Tid56. Reducing the expression of Tid1 in MDA-MB231 breast cancer cells enhanced their migration without affecting their survival or growth rate. From microarray screening, we discovered that after Tid1 depletion, the mRNA level of interleukin-8 (IL-8) was significantly increased in these cancer cells, which consequently increased secretion of IL-8 protein by 3.5-fold. The enhanced migration of these Tid1-knockdown cells was blocked by reducing the IL-8 expression or by adding an IL-8 neutralizing antibody to the culture medium, suggesting that enhancement of cell motility in these Tid1-deficient cells is dependent on the de novo synthesis of IL-8. Subsequently, we found that abrogating the nuclear factor kappaB binding site in the IL-8 promoter completely blocked the Tid1 depletion-induced IL-8 expression in the breast cancer cells. As increased IL-8 levels are known to promote tumor metastasis, we tested the effect of Tid1 knockdown on tumor metastasis and found that Tid1 depletion enhanced the metastasis of breast cancer cells in animals. Together, these results indicate that Tid1 negatively regulates the motility and metastasis of breast cancer cells, most likely through attenuation of nuclear factor kappaB activity on the promoter of the IL8 gene.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular/fisiologia , Proteínas de Choque Térmico HSP40/fisiologia , Interleucina-8/antagonistas & inibidores , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Fator VII/farmacologia , Fator VIIa , Proteínas de Choque Térmico HSP40/deficiência , Proteínas de Choque Térmico HSP40/genética , Humanos , Interleucina-8/biossíntese , Interleucina-8/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , NF-kappa B/metabolismo , Metástase Neoplásica , RNA Interferente Pequeno/genética , Proteínas Recombinantes/farmacologia , Transfecção , Regulação para Cima
9.
Cancer Res ; 64(21): 7732-9, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15520177

RESUMO

The ErbB-2/HER-2 receptor tyrosine kinase is overexpressed in a wide range of solid human tumors. The ErbB-2 gene product is a transmembrane glycoprotein belonging to the epidermal growth factor receptor family, and its cytoplasmic domain is responsible for sending the mitogenic signals into cells. We discovered that this domain of ErbB-2 interacts with Tid1 protein, the human counterpart of the Drosophila tumor suppressor Tid56, whose null mutation causes lethal tumorigenesis during the larval stage. Tid1 also is known as a cochaperone of heat shock protein 70 (HSP70) and binds to HSP70 through its conserved DnaJ domain. We found that increased expression of Tid1 in human mammary carcinomas overexpressing ErbB-2 suppresses the expression level of ErbB-2 and attenuates the resultant ErbB-2-dependent oncogenic extracellular signal-regulated kinase 1/2 and big mitogen-activated protein kinase 1 signaling pathways leading to programmed cell death (PCD). A functional DnaJ domain of Tid1 also is required for its inhibition of ErbB-2 expression and the consequent PCD of carcinoma cells resulting from increased Tid1 expression. Importantly, ErbB-2-dependent tumor progression in animals is inhibited by increased expression of Tid1 in tumor cells. Collectively, these results suggest that Tid1 modulates the uncontrolled proliferation of ErbB-2-overexpressing carcinoma cells by reducing ErbB-2 expression and as a result suppresses the ErbB-2-dependent cancerous signaling and tumor progression. Moreover, the cochaperonic and regulatory functions of Tid1 on HSP70 most likely play an essential role in this antitumor function of Tid1 in carcinoma cells.


Assuntos
Proteínas de Choque Térmico/fisiologia , Neoplasias Experimentais/prevenção & controle , Receptor ErbB-2/antagonistas & inibidores , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Feminino , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Humanos , Camundongos , Receptor ErbB-2/fisiologia
10.
Nat Rev Drug Discov ; 14(11): 759-80, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26338154

RESUMO

The aggregation of specific proteins is hypothesized to underlie several degenerative diseases, which are collectively known as amyloid disorders. However, the mechanistic connection between the process of protein aggregation and tissue degeneration is not yet fully understood. Here, we review current and emerging strategies to ameliorate aggregation-associated degenerative disorders, with a focus on disease-modifying strategies that prevent the formation of and/or eliminate protein aggregates. Persuasive pharmacological and genetic evidence now supports protein aggregation as the cause of postmitotic tissue dysfunction or loss. However, a more detailed understanding of the factors that trigger and sustain aggregate formation and of the structure-activity relationships underlying proteotoxicity is needed to develop future disease-modifying therapies.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Agregação Patológica de Proteínas/tratamento farmacológico , Transporte Proteico/efeitos dos fármacos , Animais , Fatores Biológicos/farmacologia , Fatores Biológicos/uso terapêutico , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Pré-Albumina/antagonistas & inibidores , Pré-Albumina/química , Pré-Albumina/metabolismo , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Dobramento de Proteína/efeitos dos fármacos , Relação Estrutura-Atividade , Resultado do Tratamento
11.
J Mol Biol ; 421(2-3): 185-203, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22244854

RESUMO

Transthyretin (TTR) is one of the many proteins that are known to misfold and aggregate (i.e., undergo amyloidogenesis) in vivo. The process of TTR amyloidogenesis causes nervous system and/or heart pathology. While several of these maladies are associated with mutations that destabilize the native TTR quaternary and/or tertiary structure, wild-type TTR amyloidogenesis also leads to the degeneration of postmitotic tissue. Over the past 20 years, much has been learned about the factors that influence the propensity of TTR to aggregate. This biophysical information led to the development of a therapeutic strategy, termed "kinetic stabilization," to prevent TTR amyloidogenesis. This strategy afforded the drug tafamidis which was recently approved by the European Medicines Agency for the treatment of TTR familial amyloid polyneuropathy, the most common familial TTR amyloid disease. Tafamidis is the first and currently the only medication approved to treat TTR familial amyloid polyneuropathy. Here we review the biophysical basis for the kinetic stabilization strategy and the structure-based drug design effort that led to this first-in-class pharmacologic agent.


Assuntos
Amiloidose/metabolismo , Aprovação de Drogas , Pré-Albumina/metabolismo , Animais , Humanos , Cinética , Modelos Moleculares , Pré-Albumina/química , Conformação Proteica
12.
Cancer Cell ; 18(3): 258-67, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20832753

RESUMO

BMK1 is activated by mitogens and oncogenic signals and, thus, is strongly implicated in tumorigenesis. We found that BMK1 interacted with promyelocytic leukemia protein (PML), and inhibited its tumor-suppressor function through phosphorylation. Furthermore, activated BMK1 notably inhibited PML-dependent activation of p21. To further investigate the BMK-mediated inhibition of the tumor suppressor activity of PML in tumor cells, we developed a small-molecule inhibitor of the kinase activity of BMK1, XMD8-92. Inhibition of BMK1 by XMD8-92 blocked tumor cell proliferation in vitro and significantly inhibited tumor growth in vivo by 95%, demonstrating the efficacy and tolerability of BMK1-targeted cancer treatment in animals.


Assuntos
Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Proteínas Supressoras de Tumor/antagonistas & inibidores , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Citosol/enzimologia , Citosol/metabolismo , Genes Supressores de Tumor , Células HeLa , Humanos , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Proteínas Nucleares/metabolismo , Fosforilação , Proteína da Leucemia Promielocítica , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
13.
Cancer Res ; 69(8): 3713-20, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19351860

RESUMO

Integrins interact with extracellular matrix (ECM) and deliver intracellular signaling for cell proliferation, survival, and motility. During tumor metastasis, integrin-mediated cell adhesion to and migration on the ECM proteins are required for cancer cell survival and adaptation to the new microenvironment. Using stable isotope labeling by amino acids in cell culture-mass spectrometry, we profiled the phosphoproteomic changes induced by the interactions of cell integrins with type I collagen, the most common ECM substratum. Integrin-ECM interactions modulate phosphorylation of 517 serine, threonine, or tyrosine residues in 513 peptides, corresponding to 357 proteins. Among these proteins, 33 key signaling mediators with kinase or phosphatase activity were subjected to small interfering RNA-based functional screening. Three integrin-regulated kinases, DBF4, PAK2, and GRK6, were identified for their critical role in cell adhesion and migration possibly through their regulation of actin cytoskeleton arrangement. Altogether, we not only depict an integrin-modulated phosphorylation network during cell-ECM protein interactions but also reveal novel regulators for cell adhesion and migration.


Assuntos
Integrinas/metabolismo , Neoplasias/metabolismo , Proteômica/métodos , RNA Interferente Pequeno/genética , Actinas/metabolismo , Adesão Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Movimento Celular/fisiologia , Colágeno Tipo I , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Quinases de Receptores Acoplados a Proteína G/metabolismo , Células HeLa , Humanos , Espectrometria de Massas/métodos , Neoplasias/genética , Neoplasias/patologia , Transfecção , Quinases Ativadas por p21/metabolismo
14.
Cancer Res ; 69(6): 2663-8, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19276368

RESUMO

Because the mammalian target of rapamycin (mTOR) pathway is commonly deregulated in human cancer, mTOR inhibitors, rapamycin and its derivatives, are being actively tested in cancer clinical trials. Clinical updates indicate that the anticancer effect of these drugs is limited, perhaps due to rapamycin-dependent induction of oncogenic cascades by an as yet unclear mechanism. As such, we investigated rapamycin-dependent phosphoproteomics and discovered that 250 phosphosites in 161 cellular proteins were sensitive to rapamycin. Among these, rapamycin regulated four kinases and four phosphatases. A siRNA-dependent screen of these proteins showed that AKT induction by rapamycin was attenuated by depleting cellular CDC25B phosphatase. Rapamycin induces the phosphorylation of CDC25B at Serine375, and mutating this site to Alanine substantially reduced CDC25B phosphatase activity. Additionally, expression of CDC25B (S375A) inhibited the AKT activation by rapamycin, indicating that phosphorylation of CDC25B is critical for CDC25B activity and its ability to transduce rapamycin-induced oncogenic AKT activity. Importantly, we also found that CDC25B depletion in various cancer cell lines enhanced the anticancer effect of rapamycin. Together, using rapamycin phosphoproteomics, we not only advance the global mechanistic understanding of the action of rapamycin but also show that CDC25B may serve as a drug target for improving mTOR-targeted cancer therapies.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Próstata/metabolismo , Sirolimo/farmacologia , Fosfatases cdc25/metabolismo , Antibióticos Antineoplásicos/antagonistas & inibidores , Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Masculino , Fosforilação , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Quinases/metabolismo , Proteômica , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/antagonistas & inibidores , Serina-Treonina Quinases TOR
15.
J Biol Chem ; 281(45): 34592-600, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16968706

RESUMO

Toll-like receptors (TLRs) play a crucial role in innate immunity by recognizing microbial pathogens. Triad3A is an E3 ubiquitin-protein ligase that interacts with the Toll/interleukin-1 receptor domain of TLRs and promotes their proteolytic degradation. In the present study, we further investigated its activity on signaling molecules downstream of TLRs and tumor necrosis factor (TNF) receptor 1. Triad3A promoted down-regulation of two TIR domain-containing adapter proteins, TIRAP and TRIF, as well as a RIP1 but had no effect on other adapter molecules in either the TLRs or TNF-alpha signaling pathways. Multiple sequence alignment analysis suggested that RIP1 contains a TIR homologous domain, and mutation of amino acid residues in this domain identified three residues critical for its interaction with Triad3A. Moreover, Triad3A acted as a negative regulator in TNF-alpha signaling. Reduction of Triad3A expression by small interference RNAs rendered cells hyperresponsive to TNF-alpha stimulation. Conversely, overexpression of Triad3A in cells blocked TNF-alpha-induced cell activation. This negative regulation was effected independently of changes in the cellular protein level of RIP1. Further studies indicated that RIP1 formed a complex with Triad3A and heat shock protein 90 (Hsp90), which is a chaperone protein capable of maintaining the stability of its client proteins. Treatment of cells with geldanamycin to disrupt the Hsp90 complex led to proteasomal degradation of RIP1. Depletion of Triad3A by small interference RNA treatment inhibited geldanamycin-activated ubiquitination and proteolytic degradation of RIP1. These results suggest that Triad3A is an E3 ubiquitin-protein ligase to RIP1 and that Hsp90 and Triad3A cooperatively maintain the homeostasis of RIP1.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Benzoquinonas/farmacologia , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Proteínas de Choque Térmico HSP90/genética , Humanos , Immunoblotting , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Rim/citologia , Rim/metabolismo , Lactamas Macrocíclicas/farmacologia , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , NF-kappa B/genética , NF-kappa B/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Plasmídeos , Ligação Proteica , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a RNA/genética , Receptores de Interleucina-1/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitina-Proteína Ligases
16.
J Biol Chem ; 281(39): 28822-30, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16893899

RESUMO

Innate immune system receptors function as sensors of infection and trigger the immune responses through ligand-specific signaling pathways. These ligands are pathogen-associated products, such as components of bacterial walls and viral nuclear acids. A common response to such ligands is the activation of mitogen-activated protein kinase p38, whereas double-stranded viral RNA additionally induces the phosphorylation of eukaryotic translation initiation factor 2alpha (eIF2alpha). Here we have shown that p38 and eIF2alpha phosphorylation represent two biochemical markers of the effects induced by N-(3-oxo-acyl)homoserine lactones, the secreted products of a number of Gram-negative bacteria, including the human opportunistic pathogen Pseudomonas aeruginosa. Furthermore, N-(3-oxo-dodecanoyl)homoserine lactone induced distension of mitochondria and the endoplasmic reticulum as well as c-jun gene transcription. These effects occurred in a wide variety of cell types including alveolar macrophages and bronchial epithelial cells, requiring the structural integrity of the lactone ring motif and its natural stereochemistry. These findings suggest that N-(3-oxo-acyl)homoserine lactones might be recognized by receptors of the innate immune system. However, we provide evidence that N-(3-oxo-dodecanoyl)homoserine lactone-mediated signaling does not require the presence of the canonical innate immune system receptors, Toll-like receptors, or two members of the NLR/Nod/Caterpillar family, Nod1 and Nod2. These data offer a new understanding of the effects of N-(3-oxo-dodecanoyl)homoserine lactone on host cells and its role in persistent airway infections caused by P. aeruginosa.


Assuntos
4-Butirolactona/análogos & derivados , Células da Medula Óssea/microbiologia , Regulação da Expressão Gênica , Macrófagos/microbiologia , 4-Butirolactona/química , 4-Butirolactona/fisiologia , Motivos de Aminoácidos , Animais , Células da Medula Óssea/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Pseudomonas aeruginosa/metabolismo , RNA Viral/metabolismo , Transdução de Sinais
17.
J Lipid Res ; 47(5): 1097-111, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16479018

RESUMO

The LIPID MAPS Consortium (www.lipidmaps.org) is developing comprehensive procedures for identifying all lipids of the macrophage, following activation by endotoxin. The goal is to quantify temporal and spatial changes in lipids that occur with cellular metabolism and to develop bioinformatic approaches that establish dynamic lipid networks. To achieve these aims, an endotoxin of the highest possible analytical specification is crucial. We now report a large-scale preparation of 3-deoxy-D-manno-octulosonic acid (Kdo)(2)-Lipid A, a nearly homogeneous Re lipopolysaccharide (LPS) sub-structure with endotoxin activity equal to LPS. Kdo(2)-Lipid A was extracted from 2 kg cell paste of a heptose-deficient Escherichia coli mutant. It was purified by chromatography on silica, DEAE-cellulose, and C18 reverse-phase resin. Structure and purity were evaluated by electrospray ionization/mass spectrometry, liquid chromatography/mass spectrometry and (1)H-NMR. Its bioactivity was compared with LPS in RAW 264.7 cells and bone marrow macrophages from wild-type and toll-like receptor 4 (TLR-4)-deficient mice. Cytokine and eicosanoid production, in conjunction with gene expression profiling, were employed as readouts. Kdo(2)-Lipid A is comparable to LPS by these criteria. Its activity is reduced by >10(3) in cells from TLR-4-deficient mice. The purity of Kdo(2)-Lipid A should facilitate structural analysis of complexes with receptors like TLR-4/MD2.


Assuntos
Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Receptor 4 Toll-Like/fisiologia , Animais , Cromatografia Líquida de Alta Pressão/métodos , Escherichia coli/metabolismo , Lipopolissacarídeos/isolamento & purificação , Camundongos , Ressonância Magnética Nuclear Biomolecular , Prostaglandina D2/metabolismo , Espectrometria de Massas por Ionização por Electrospray
18.
J Immunol ; 174(10): 6105-12, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15879105

RESUMO

Tid1, a DnaJ cochaperone protein, is the mammalian homologue of the Drosophila tumor suppressor Tid56 whose antitumor function is most likely mediated through its capacity to regulate cell differentiation in imaginal discs. We suspected that the mammalian counterpart, tid1, may also be involved in regulating cell differentiation. To investigate this, we exploited the system of T cell development to examine whether tid1 plays a role in this well-defined process. Mice with tid1 specifically deleted in T cells developed thymic atrophy, with dramatic reduction of double-positive and single-positive thymocytes in the tid1(-/-) thymus. Although the subpopulations of tid1(-/-) double-negative (DN) 1-3 thymocytes were normal, the subpopulation of DN4 thymocytes was measurably smaller because of reduced proliferation and significant cell death. Immature tid1(-/-) thymocytes show normal VDJ beta-chain rearrangement and pre-TCR and CD3 expression in both DN3 and DN4 thymocytes, but in DN4 thymocytes, there was significantly reduced expression of the antiapoptotic bcl-2 gene. Restoring the expression level of Bcl-2 protein in tid1(-/-) thymus by introduction of a transgenic human bcl-2 gene resulted in reversal of the developmental defects in tid1(-/-) thymus. Together, these results demonstrate that tid1 is critical in early thymocyte development, especially during transition from the DN3 to double-positive stages, possibly through its regulation of bcl-2 expression, which provides survival signals.


Assuntos
Diferenciação Celular/imunologia , Proteínas de Choque Térmico/fisiologia , Subpopulações de Linfócitos T/citologia , Adjuvantes Imunológicos/deficiência , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/fisiologia , Animais , Morte Celular/genética , Morte Celular/imunologia , Diferenciação Celular/genética , Citocinas/biossíntese , Citocinas/fisiologia , Epitopos de Linfócito T/imunologia , Perfilação da Expressão Gênica , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Inativação Gênica , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/deficiência , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Timo/citologia , Timo/metabolismo
19.
Eur J Immunol ; 32(3): 761-72, 2002 03.
Artigo em Inglês | MEDLINE | ID: mdl-11870620

RESUMO

Propionibacterium acnes-primed mice develop an IFN-gamma-dependent hypersensitivity towards LPS. Since CD14 plays a key role in LPS-induced cell activation the regulation and function of CD14 in this sensitization process were studied in IFN-gamma R-/- and the respective wild-type (wt) mice. In unprimed mice, CD14 (mRNA and protein) was either absent (liver) or only weakly expressed in organs (spleen, lung) and in plasma. Priming with P. acnes led to a moderate, mainly IFN-gamma-dependent up-regulation of CD14. LPS challenge of unprimed mice induced an IFN-gamma-independent increase in CD14 mRNA and CD14 protein. LPS challenge of P. acnes-primed mice induced a strong CD14 overexpression. This response was completely absent in IFN-gamma R-/- mice and is therefore strictly IFN-gamma-dependent. The requirement for CD14 in LPS hyper-responsiveness was assessed by comparing CD14-/- and the respective wt mice with respect to their ability to produce TNF and IFN-gamma, two recognized indices of LPS activity. LPS challenge without priming led to a weaker cytokine reaction in CD14-/- than in wt mice. However, priming with P. acnes enhanced the cytokine response to LPS in both wt and CD14-/- mice, although in the latter absolute levels of cytokines were lower. Therefore, hyperreactivity to LPS is characterized by an up-regulation of CD14, but the sensitization by P. acnes is not CD14 dependent.


Assuntos
Proteínas de Drosophila , Receptores de Lipopolissacarídeos/imunologia , Lipopolissacarídeos/imunologia , Tecido Linfoide/imunologia , Propionibacterium acnes/imunologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Imunização , Interferon gama/biossíntese , Interferon gama/fisiologia , Receptores de Lipopolissacarídeos/biossíntese , Receptores de Lipopolissacarídeos/sangue , Receptores de Lipopolissacarídeos/genética , Fígado/metabolismo , Pulmão/metabolismo , Tecido Linfoide/metabolismo , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/fisiologia , Solubilidade , Organismos Livres de Patógenos Específicos , Baço/metabolismo , Receptores Toll-Like , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Receptor de Interferon gama
20.
Am J Pathol ; 163(6): 2555-63, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14633627

RESUMO

The stress-inducible gene heme oxygenase (HO-1) has previously been shown to provide cytoprotection against oxidative stress. The mechanism(s) by which HO-1 provides this cytoprotection is poorly understood. We demonstrate here that carbon monoxide (CO), a byproduct released during the degradation of heme by HO, plays a major role in mediating the cytoprotection against oxidant-induced lung injury. We show in vitro that CO protects cultured epithelial cells from hyperoxic damage. By using dominant negative mutants and mice deficient in the genes for the various MAP kinases, we demonstrate that the cytoprotective effects of CO are mediated by selective activation of the MKK3/p38 beta protein MAP kinase pathway. In vivo, our experiments demonstrate that CO at a low concentration protects the lungs, extends the survival of the animals, and exerts potent anti-inflammatory effects with reduced inflammatory cell influx into the lungs and marked attenuation in the expression of pro-inflammatory cytokines.


Assuntos
Monóxido de Carbono/farmacologia , Citoproteção , Pneumopatias/induzido quimicamente , Pneumopatias/prevenção & controle , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oxidantes , Proteínas Tirosina Quinases/metabolismo , Animais , Carboxihemoglobina/metabolismo , Células Cultivadas , Citocinas/antagonistas & inibidores , Ativação Enzimática , Hiperóxia/mortalidade , Hiperóxia/patologia , Hiperóxia/fisiopatologia , Hiperóxia/prevenção & controle , Mediadores da Inflamação/antagonistas & inibidores , Pulmão/patologia , MAP Quinase Quinase 3 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Infiltração de Neutrófilos/efeitos dos fármacos , Pneumonia/prevenção & controle , Análise de Sobrevida , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA