Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Biochem Biophys Res Commun ; 503(2): 882-887, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29928885

RESUMO

Bromodomain-containing protein 4 (Brd4) is known to play a key role in tumorigenesis. It binds acetylated histones to regulate the expression of numerous genes. Because of the importance of brd4 in tumorigenesis, much research has been undertaken to develop brd4 inhibitors with therapeutic potential. As a result, various scaffolds for bromodomain inhibitors have been identified. To discover new scaffolds, we performed mid-throughput screening using two different enzyme assays, alpha-screen and ELISA. We found a novel bromodomain inhibitor with a unique scaffold, aristoyagonine. This natural compound showed inhibitory activity in vitro and tumor growth inhibition in a Ty82-xenograft mouse model. In addition, we tested Brd4 inhibitors in gastric cancer cell lines, and found that aristoyagonine exerted cytotoxicity not only in I-BET-762-sensitive cancer cells, but also in I-BET-762-resistant cancer cells. This is the first paper to describe a natural compound as a Brd4 bromodomain inhibitor.


Assuntos
Produtos Biológicos/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Isoquinolinas/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Animais , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Neoplasias/prevenção & controle , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Angew Chem Int Ed Engl ; 54(41): 12020-4, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26315561

RESUMO

Targeted therapy based on protein-drug conjugates has attracted significant attention owing to its high efficacy and low side effects. However, efficient and stable drug conjugation to a protein binder remains a challenge. Herein, a chemoenzymatic method to generate highly stable and homogenous drug conjugates with high efficiency is presented. The approach comprises the insertion of the CaaX sequence at the C-terminal end of the protein binder, prenylation using farnesyltransferase, and drug conjugation through an oxime ligation reaction. MMAF and an EGFR-specific repebody are used as the antitumor agent and protein binder, respectively. The method enables the precisely controlled synthesis of repebody-drug conjugates with high yield and homogeneity. The utility of this approach is illustrated by the notable stability of the repebody-drug conjugates in human plasma, negligible off-target effects, and a remarkable antitumor activity in vivo. The present method can be widely used for generating highly homogeneous and stable PDCs for targeted therapy.


Assuntos
Antineoplásicos/química , Receptores ErbB/metabolismo , Oligopeptídeos/química , Oximas/química , Proteínas/química , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Farnesiltranstransferase/metabolismo , Humanos , Camundongos Nus , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Oligopeptídeos/metabolismo , Oligopeptídeos/uso terapêutico , Oximas/metabolismo , Ligação Proteica , Prenilação de Proteína , Proteínas/metabolismo , Proteínas/uso terapêutico
3.
Hum Mol Genet ; 21(1): 101-14, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21954231

RESUMO

Huntington's disease (HD), an inherited neurodegenerative disorder, is caused by an expansion of cytosine-adenine-guanine repeats in the huntingtin gene. The aggregation of mutant huntingtin (mtHTT) and striatal cell loss are representative features to cause uncontrolled movement and cognitive defect in HD. However, underlying mechanism of mtHTT aggregation and cell toxicity remains still elusive. Here, to find new genes modulating mtHTT aggregation, we performed cell-based functional screening using the cDNA expression library and isolated IRE1 gene, one of endoplasmic reticulum (ER) stress sensors. Ectopic expression of IRE1 led to its self-activation and accumulated detergent-resistant mtHTT aggregates. Treatment of neuronal cells with ER stress insults, tunicamycin and thapsigargin, increased mtHTT aggregation via IRE1 activation. The kinase activity of IRE1, but not the endoribonuclease activity, was necessary to stimulate mtHTT aggregation and increased death of neuronal cells, including SH-SY5Y and STHdhQ111/111 huntingtin knock-in striatal cells. Interestingly, ER stress impaired autophagy flux via IRE1-TRAF2 pathway, thus enhancing cellular accumulation of mtHTT. Atg5 deficiency in M5-7 cells increased mtHTT aggregation but blocked ER stress-induced mtHTT aggregation. Further, ER stress markers including p-IRE1 and autophagy markers such as p62 were up-regulated exclusively in the striatal tissues of HD mouse models and in HD patients. Moreover, down-regulation of IRE1 expression rescues the rough-eye phenotype by mtHTT in a HD fly model. These results suggest that IRE1 plays an essential role in ER stress-mediated aggregation of mtHTT via the inhibition of autophagy flux and thus neuronal toxicity of mtHTT aggregates in HD.


Assuntos
Autofagia , Regulação para Baixo , Estresse do Retículo Endoplasmático , Endorribonucleases/metabolismo , Doença de Huntington/enzimologia , Doença de Huntington/fisiopatologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Endorribonucleases/genética , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Proteínas de Membrana/genética , Camundongos , Mutação , Neurônios/enzimologia , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/genética , Ratos
4.
Proc Natl Acad Sci U S A ; 106(36): 15326-31, 2009 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-19706414

RESUMO

Osteopontin (OPN) is highly expressed in cancer patients and plays important roles in many stages of tumor progression, such as anti-apoptosis, proliferation, and metastasis. From functional screening of human cDNA library, we isolated OPN as a caspase-8 substrate that regulates cell death during hypoxia/reoxygenation (Hyp/RO). In vitro cleavage assays demonstrate that OPN is cleaved at Asp-135 and Asp-157 by caspase-8. Cellular cleavage of OPN is observed in apoptotic cells exposed to Hyp/RO among various apoptotic stimuli and its cleavage is blocked by zVAD or IETD caspase inhibitor. Further, over-expression of OPN, the form with secretion signal, inhibits Hyp/RO-induced cell death. Caspase cleavage-defective OPN mutant (OPN D135A/D157A) is more efficient to suppress Hyp/RO-induced cell death than wild-type OPN. OPN D135A/D157A sustains AKT activity to increase cell viability through inhibition of caspase-9 during Hyp/RO. In addition, OPN is highly induced in some tumor cells during Hyp/RO, such as HeLa and Huh-7 cells, which is associated with their resistance to Hyp/RO by sustaining AKT activity. Notably, OPN C-terminal cleavage fragment produced by caspase-8 is detected in the nucleus. Plasmid-encoded expression of OPN C-terminal cleavage fragment increases p53 protein level and induces apoptosis of wild-type mouse embryonic fibroblast cells, but not p53(-/-) mouse embryonic fibroblast cells. These observations suggest that the protective function of OPN during Hyp/RO is inactivated via the proteolytic cleavage by caspase-8 and its cleavage product subsequently induces cell death via p53, postulating caspase-8 as a negative regulator of tumorigenic activity of OPN.


Assuntos
Apoptose/fisiologia , Caspase 8/metabolismo , Hipóxia Celular/fisiologia , Osteopontina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Western Blotting , Densitometria , Células HeLa , Humanos , Osteopontina/genética
5.
Adv Sci (Weinh) ; 8(23): e2102414, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34664433

RESUMO

Human epidermal growth factor receptor 2 (HER2) is overexpressed in breast and gastric cancers and this causes poor clinical outcomes. Although both T-DM1 and Enhertu are approved as an HER2-targeting antibody-drug conjugate (ADC), the effects of these drugs are still not satisfactory to eradicate diverse tumors expressing HER2. To address this shortfall in HER2-targeted therapeutics, an elaborate cleavable linker is created and a novel HER2-targeting ADC composed with trastuzumab and monomethyl auristatin F, which is being investigated in a phase 1 clinical trial and is referred to as LegoChem Bisciences-ADC (LCB-ADC). LCB-ADC displays a higher cytotoxic potency than T-DM1 and it also has a higher G2/M arrest ratio. In animal studies, LCB-ADC produces noticeable tumor growth inhibition compared with trastuzumab or T-DM1 in an HER2 high-expressing N87 xenograft tumor. Especially, LCB-ADC shows good efficacy in terms of suppressing tumor growth in a patient-derived xenograft (PDX) model of HER2-positive gastric cancer as well as in T-DM1-resistant models such as HER2 low-expressing HER2 low expressing JIMT-1 xenograft tumor and PDX. Collectively, the results demonstrate that LCB-ADC with the elaborate linker has a higher efficacy and greater biostability than its ADC counterparts and may successfully treat cancers that are nonresponsive to previous therapeutics.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoconjugados/uso terapêutico , Oligopeptídeos/uso terapêutico , Receptor ErbB-2/genética , Neoplasias Gástricas/tratamento farmacológico , Trastuzumab/uso terapêutico , Animais , Modelos Animais de Doenças , Haplorrinos , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Ratos , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia
6.
Oncogene ; 24(4): 688-96, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15592525

RESUMO

Caspase-8 is the most receptor-proximal, upstream caspase in the caspase cascade and plays a key role in cell death triggered by various death receptors. Here, we addressed the role of endogenous caspase-8 in tumor necrosis factor (TNF)-alpha-induced activation of NF-kappaB. Direct targeting of caspase-8 with siRNA and antisense (AS) approaches abolished TNF-alpha-induced activation of NF-kappaB in NIH3T3, HeLa, and HEK293 cells as determined with luciferase reporter gene and cell fractionation assays. Reconstitution of caspase-8-deficient C33A cells with processing-defective (P/D) mutant of caspase-8 sensitized the cells to TNF-alpha for NF-kappaB activation. In contrast to wild-type caspase-8, death effector domain mutant replacing Asp73 with Ala (caspase-8 (D73A)) failed to activate NF-kappaB and to bind FLICE-associated huge protein (FLASH) in vitro and in vivo. Instead, caspase-8 (D73A) mutant bound to caspase-8 and blocked NF-kappaB activation triggered by TNF-alpha and caspase-8. In addition, expression of an NF-kappaB-activating domain-deletion mutant of FLASH or transfection of FLASH AS oligonucleotides abolished TNF-alpha and caspase-8, but not phorbol 12-myristate 13-acetate, -induced activation of NF-kappaB. Further, immunoprecipitation assays showed that caspase-8 formed triple complex with TRAF2 and FLASH. Taken together, these results suggest that endogenous caspase-8 mediates TNF-alpha-induced activation of NF-kappaB via FLASH.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Caspases/metabolismo , Mutação/genética , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Proteínas Reguladoras de Apoptose , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Caspase 8 , Inibidores de Caspase , Caspases/deficiência , Caspases/genética , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
7.
FEBS Lett ; 578(3): 239-44, 2004 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-15637807

RESUMO

We have identified a novel CARD-containing protein from EST database. BinCARD (Bcl10-interacting protein with CARD). BinCARD was ubiquitously expressed. Co-immunoprecipitation, In vitro binding, mammalian two-hybrid, and immunostaining assays revealed that BinCARD interacted with Bcl10 through CARD. BinCARD potently suppressed NF-kappa B activation induced by Bcl10 and decreased the amounts of phosphorylated Bcl10. Mutations at the residue Leu17 or Leu65, which is highly conserved in CARD, abolished the inhibitory effects of BinCARD on both Bcl10-induced activation of NF-kappa B and phosphorylation of Bcl10. Further, expression of BinCARD inhibited Bcl10 phosphorylation induced by T cell activation signal. These results suggest that BinCARD interacts with Bcl10 to inhibit Bcl10-mediated activation of NF-kappa B and to suppress Bcl10 phosphorylation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Transporte/metabolismo , NF-kappa B/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteína 10 de Linfoma CCL de Células B , Western Blotting , Proteínas Adaptadoras de Sinalização CARD/química , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Sequência Conservada , Análise Mutacional de DNA , Regulação para Baixo , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica , Células Jurkat , Leucina/química , Leucina/genética , Luciferases/metabolismo , Ativação Linfocitária , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Testes de Precipitina , Ligação Proteica , Estrutura Secundária de Proteína , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Linfócitos T/metabolismo , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido
8.
Arch Pharm Res ; 33(11): 1813-23, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21116785

RESUMO

Mitochondrial reactive oxygen species and reactive nitrogen species are proven to be major sources of oxidative stress in the cell; they play a prominent role in a wide range of human disorders resulting from nonapoptotic cell death. The aim of this study is to examine the cytoprotective effect of the NecroX series against harmful stresses, including pro-oxidant (tertiarybutylhydroperoxide), doxorubicin, CCl4, and hypoxic injury. In this study, these novel chemical molecules inhibited caspase-independent cell death with necrotic morphology, which is distinctly different from apoptosis, autophagy, and necroptosis. In addition, they displayed strong mitochondrial reactive oxygen species and ONOO⁻ scavenging activity. Further, oral administration of these molecules in C57BL/6 mice attenuated streptozotocin-induced pancreatic islet ß-cell destruction as well as CCl4-induced hepatotoxicity in vivo. Taken together, these results demonstrate that the NecroX series are involved in the blockade of nonapoptotic cell death against mitochondrial oxidative stresses. Thus, these chemical molecules are potential therapeutic agents in mitochondria-related human diseases involving necrotic tissue injury.


Assuntos
Antioxidantes/farmacologia , Sequestradores de Radicais Livres/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Citoproteção , Humanos , Células LLC-PK1 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Necrose , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Suínos
9.
J Biol Chem ; 284(17): 11318-25, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19240033

RESUMO

Accumulation of expanded polyglutamine proteins is considered to be a major pathogenic biomarker of Huntington disease. We isolated SCAMP5 as a novel regulator of cellular accumulation of expanded polyglutamine track protein using cell-based aggregation assays. Ectopic expression of SCAMP5 augments the formation of ubiquitin-positive and detergent-resistant aggregates of mutant huntingtin (mtHTT). Expression of SCAMP5 is markedly increased in the striatum of Huntington disease patients and is induced in cultured striatal neurons by endoplasmic reticulum (ER) stress or by mtHTT. The increase of SCAMP5 impairs endocytosis, which in turn enhances mtHTT aggregation. On the contrary, down-regulation of SCAMP5 alleviates ER stress-induced mtHTT aggregation and endocytosis inhibition. Moreover, stereotactic injection into the striatum and intraperitoneal injection of tunicamycin significantly increase mtHTT aggregation in the striatum of R6/2 mice and in the cortex of N171-82Q mice, respectively. Taken together, these results suggest that exposure to ER stress increases SCAMP5 in the striatum, which positively regulates mtHTT aggregation via the endocytosis pathway.


Assuntos
Proteínas de Transporte/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Peptídeos/metabolismo , Regulação para Cima , Animais , Encéfalo/embriologia , Endocitose , Humanos , Proteína Huntingtina , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Ratos , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
10.
J Cell Biol ; 182(4): 675-84, 2008 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-18710920

RESUMO

Amyloid-beta (Abeta) neurotoxicity is believed to contribute to the pathogenesis of Alzheimer's disease (AD). Previously we found that E2-25K/Hip-2, an E2 ubiquitin-conjugating enzyme, mediates Abeta neurotoxicity. Here, we report that E2-25K/Hip-2 modulates caspase-12 activity via the ubiquitin/proteasome system. Levels of endoplasmic reticulum (ER)-resident caspase-12 are strongly up-regulated in the brains of AD model mice, where the enzyme colocalizes with E2-25K/Hip-2. Abeta increases expression of E2-25K/Hip-2, which then stabilizes caspase-12 protein by inhibiting proteasome activity. This increase in E2-25K/Hip-2 also induces proteolytic activation of caspase-12 through its ability to induce calpainlike activity. Knockdown of E2-25K/Hip-2 expression suppresses neuronal cell death triggered by ER stress, and thus caspase-12 is required for the E2-25K/Hip-2-mediated cell death. Finally, we find that E2-25K/Hip-2-deficient cortical neurons are resistant to Abeta toxicity and to the induction of ER stress and caspase-12 expression by Abeta. E2-25K/Hip-2 is thus an essential upstream regulator of the expression and activation of caspase-12 in ER stress-mediated Abeta neurotoxicity.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Caspase 12/metabolismo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/patologia , Neurotoxinas/toxicidade , Enzimas de Conjugação de Ubiquitina/metabolismo , Animais , Calpaína/metabolismo , Caspase 12/biossíntese , Caspase 12/química , Morte Celular/efeitos dos fármacos , Linhagem Celular , Córtex Cerebral/citologia , Córtex Cerebral/enzimologia , Regulação para Baixo/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Estabilidade Enzimática/efeitos dos fármacos , Humanos , Camundongos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Dobramento de Proteína , Ratos , Espécies Reativas de Oxigênio/farmacologia
11.
Cytokine ; 20(6): 283-8, 2002 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-12633570

RESUMO

Epithelial cell apoptosis triggered cooperatively by multiple cytokines contributes to the injury induced by inflammatory responses in the lung and elsewhere. Here we show that interferon-gamma (IFN-gamma) sensitizes A549 cells, human lung epithelial cells, to cytokine-mediated apoptosis by upregulating caspase-8 expression. Pretreating the cells with IFN-gamma potentiated Fas- and TNF-related apoptosis inducing ligand (TRAIL)-induced cell death, but other forms of apoptosis, not mediated via receptors, were unaffected. Western blotting and inhibitor assays showed that IFN-gamma selectively increased expression of caspases-7 and -8, but not caspases-2, -3, -9, or -10, as a necessary step leading to apoptosis. Assaying promoter activity using a luciferase reporter gene indicated that an IFN-gamma response element was located in the 5'-flanking region of the caspase-8 gene, spanning positions -227 to -219. Taken together, these findings suggest that IFN-gamma potentiates Fas- and TRAIL-mediated apoptosis by increasing caspase-8 expression via an IFN-gamma response element in A549 cells.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Interferon gama/metabolismo , Glicoproteínas de Membrana/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Receptor fas/metabolismo , Região 5'-Flanqueadora/fisiologia , Proteínas Reguladoras de Apoptose , Caspase 8 , Caspase 9 , Epitélio/metabolismo , Humanos , Pulmão/metabolismo , Regiões Promotoras Genéticas , Ligante Indutor de Apoptose Relacionado a TNF
12.
Neurobiol Dis ; 14(3): 557-66, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14678771

RESUMO

Abnormal Tau protein is known to be closely associated with several neurodegenerative diseases. Previously, we showed that Tau was cleaved by caspase-3 to generate the cleavage product lacking the C-terminus (DeltaTau-1) during neuronal cell death. Here we characterized caspase-8-dependent neurotoxicity of the truncated Tau. Introduction of DeltaTau-1 into primary hippocampal neurons induced loss of neurites in a caspase-dependent manner. Caspase-8 and -6 were proteolytically activated during DeltaTau-1-triggered neuronal cell death, which was suppressed by IETD-fmk, caspase-8 inhibitor. Direct targeting of caspase-8 and its associated FADD with antisense approaches and transient expression of their dominant-negative mutants reduced DeltaTau-1-induced apopotosis. Cells deficient in caspase-8, but not caspase-3, became sensitized to DeltaTau-1-mediated toxicity upon reconstitution with caspase-8. In addition, ectopic expression of mitochondrial antiapoptotic Bcl-2, Bcl-X(L), or inactive caspase-9 short form suppressed DeltaTau-1 toxicity. These results suggest that the truncated Tau protein activates proximal caspase-8 through FADD as a necessary step leading to neuronal cell death and neurite regression, contributing to the progression of abnormal Tau-associated neurodegeneracy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Caspases/metabolismo , Degeneração Neural/metabolismo , Neuritos/metabolismo , Proteínas tau/metabolismo , Animais , Elementos Antissenso (Genética)/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 8 , Inibidores de Caspase , Caspases/genética , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Proteína de Domínio de Morte Associada a Fas , Células HeLa , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Peso Molecular , Mutação/genética , Degeneração Neural/fisiopatologia , Neuritos/efeitos dos fármacos , Neuritos/patologia , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas tau/farmacologia
13.
J Cell Biochem ; 85(2): 334-45, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11948689

RESUMO

In response to a diverse array of signals, IkappaBalpha is targeted for phosphorylation-dependent degradation by the proteasome, thereby activating NF-kappaB. Here we demonstrate a role of the cleavage product of IkappaBalpha in various death signals. During apoptosis of NIH3T3, Jurkat, Rat-1, and L929 cells exposed to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), Fas, serum deprivation, or TNF-alpha, respectively, IkappaBalpha was cleaved in a caspase-dependent manner. In vitro and in vivo cleavage assays and site-directed mutagenesis showed that caspase-3 cleaved IkappaBalpha between Asp31 and Ser32. Expression of the cleavage product lacking amino-terminus (1-31), DeltaIkappaBalpha, sensitized otherwise resistant NIH3T3 fibroblast cells to apoptosis induced by TNF-alpha or TRAIL, and HeLa tumor cells to TNF-alpha. DeltaIkappaBalpha was more pro-apoptotic compared to wild type or cleavage-resistant (D31E)IkappaBalpha mutant and the sensitization elicited by DeltaIkappaBalpha was as effective as that by the dominant negative mutant, (S32,36A)IkappaBalpha, in NIH3T3 cells. DeltaIkappaBalpha suppressed the transactivation of NF-kappaB induced by TNF-alpha or TRAIL, as reflected by luciferase-reporter activity. Conversely, expression of the p65 subunit of NF-kappaB suppressed TNF-alpha-, TRAIL-, and serum deprivation-induced cell death. On the contrary, DeltaIkappaBalpha was less effective at increasing the death rate of HeLa cells that were already sensitive to death signals including TRAIL, etoposide, or taxol. These results suggest that DeltaIkappaBalpha generated by various death signals sensitizes cells to apoptosis by suppressing NF-kappaB activity.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , Glicoproteínas de Membrana/farmacologia , NF-kappa B/antagonistas & inibidores , Células Tumorais Cultivadas/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose , Ácido Aspártico/metabolismo , Western Blotting , Primers do DNA/química , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Feminino , Fibroblastos/enzimologia , Humanos , Luciferases/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/fisiologia , Paclitaxel/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , beta-Galactosidase/metabolismo
14.
Mol Cell ; 12(3): 553-63, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14527403

RESUMO

The ubiquitin/proteasome system has been proposed to play an important role in Alzheimer's disease (AD) pathogenesis. However, the critical factor(s) modulating both amyloid-beta peptide (Abeta) neurotoxicity and ubiquitin/proteasome system in AD are not known. We report the isolation of an unusual ubiquitin-conjugating enzyme, E2-25K/Hip-2, as a mediator of Abeta toxicity. The expression of E2-25K/Hip-2 was upregulated in the neurons exposed to Abeta(1-42) in vivo and in culture. Enzymatic activity of E2-25K/Hip-2 was required for both Abeta(1-42) neurotoxicity and inhibition of proteasome activity. E2-25K/Hip-2 functioned upstream of apoptosis signal-regulating kinase 1 (ASK1) and c-Jun N-terminal kinase (JNK) in Abeta(1-42) toxicity. Further, the ubiquitin mutant, UBB+1, a potent inhibitor of the proteasome which is found in Alzheimer's brains, was colocalized and functionally interacted with E2-25K/Hip-2 in mediating neurotoxicity. These results suggest that E2-25K/Hip-2 is a crucial factor in regulating Abeta neurotoxicity and could play a role in the pathogenesis of Alzheimer's disease.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/enzimologia , Ligases/metabolismo , Neurônios/enzimologia , Enzimas de Conjugação de Ubiquitina , Regulação para Cima/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/toxicidade , Animais , Apoptose/genética , Encéfalo/patologia , Encéfalo/fisiopatologia , Células Cultivadas , Cisteína Endopeptidases/metabolismo , Feminino , Feto , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Ligases/genética , MAP Quinase Quinase Quinase 5 , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/metabolismo , Mutação/genética , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Complexo de Endopeptidases do Proteassoma , Ratos , Ubiquitina/genética , Ubiquitina/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA