Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cancer Immunol Immunother ; 71(2): 473-489, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34247273

RESUMO

Cluster of differentiation 47 (CD47) is a transmembrane protein ubiquitously expressed on human cells but overexpressed on many different tumor cells. The interaction of CD47 with signal-regulatory protein alpha (SIRPα) triggers a "don't eat me" signal to the macrophage, inhibiting phagocytosis. Thus, overexpression of CD47 enables tumor cells to escape from immune surveillance via the blockade of phagocytic mechanisms. We report here the development and characterization of CC-90002, a humanized anti-CD47 antibody. CC-90002 is unique among previously reported anti-CD47 bivalent antibodies that it does not promote hemagglutination while maintaining high-affinity binding to CD47 and inhibition of the CD47-SIRPα interaction. Studies in a panel of hematological cancer cell lines showed concentration-dependent CC-90002-mediated phagocytosis in acute lymphoblastic leukemia, acute myeloid leukemia (AML), lenalidomide-resistant multiple myeloma (MM) cell lines and AML cells from patients. In vivo studies with MM cell line-derived xenograft models established in immunodeficient mice demonstrated significant dose-dependent antitumor activity of CC-90002. Treatment with CC-90002 significantly prolonged survival in an HL-60-disseminated AML model. Mechanistic studies confirmed the binding of CC-90002 to tumor cells and concomitant recruitment of F4-80 positive macrophages into the tumor and an increase in expression of select chemokines and cytokines of murine origin. Furthermore, the role of macrophages in the CC-90002-mediated antitumor activity was demonstrated by transient depletion of macrophages with liposome-clodronate treatment. In non-human primates, CC-90002 displayed acceptable pharmacokinetic properties and a favorable toxicity profile. These data demonstrate the potential activity of CC-90002 across hematological malignancies and provided basis for clinical studies CC-90002-ST-001 (NCT02367196) and CC-90002-AML-001 (NCT02641002).


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Diferenciação/imunologia , Antígeno CD47/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Leucemia Promielocítica Aguda/tratamento farmacológico , Macrófagos/imunologia , Receptores Imunológicos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos de Diferenciação/metabolismo , Antineoplásicos Imunológicos/imunologia , Antineoplásicos Imunológicos/farmacologia , Apoptose , Antígeno CD47/metabolismo , Diferenciação Celular , Proliferação de Células , Feminino , Humanos , Leucemia Promielocítica Aguda/imunologia , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fagocitose , Prognóstico , Receptores Imunológicos/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Nat Chem Biol ; 6(5): 359-68, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20383151

RESUMO

Mps1, a dual-specificity kinase, is required for the proper functioning of the spindle assembly checkpoint and for the maintenance of chromosomal stability. As Mps1 function has been implicated in numerous phases of the cell cycle, the development of a potent, selective small-molecule inhibitor of Mps1 should facilitate dissection of Mps1-related biology. We describe the cellular effects and Mps1 cocrystal structures of new, selective small-molecule inhibitors of Mps1. Consistent with RNAi studies, chemical inhibition of Mps1 leads to defects in Mad1 and Mad2 establishment at unattached kinetochores, decreased Aurora B kinase activity, premature mitotic exit and gross aneuploidy, without any evidence of centrosome duplication defects. However, in U2OS cells having extra centrosomes (an abnormality found in some cancers), Mps1 inhibition increases the frequency of multipolar mitoses. Lastly, Mps1 inhibitor treatment resulted in a decrease in cancer cell viability.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Ciclo Celular , Inibidores Enzimáticos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Modelos Moleculares , Inibidores de Proteínas Quinases/química , Proteínas Tirosina Quinases
3.
Cancer Cell ; 5(5): 501-12, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15144957

RESUMO

The genetic concept of synthetic lethality provides a framework for identifying genotype-selective anticancer agents. In this approach, changes in cellular physiology that arise as a consequence of oncogene activation or tumor suppressor gene loss, rather than oncoproteins themselves, are targeted to achieve tumor selectivity. Here we show that agonists of the TRAIL death receptor DR5 potently induce apoptosis in human cells overexpressing the MYC oncogene, both in vitro and as tumor xenografts in vivo. MYC sensitizes cells to DR5 in a p53-independent manner by upregulating DR5 cell surface levels and stimulating autocatalytic processing of procaspase-8. These results identify a novel mechanism by which MYC sensitizes cells to apoptosis and validate DR5 agonists as potential MYC-selective cancer therapeutics.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Mutação/genética , Proteínas Proto-Oncogênicas c-myc/fisiologia , RNA Interferente Pequeno/farmacologia , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Animais , Caspase 8 , Caspases/metabolismo , Fibroblastos/metabolismo , Marcação de Genes , Humanos , Masculino , Camundongos , Camundongos Nus , Camundongos SCID , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Retroviridae/genética , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
4.
Mol Cell Biol ; 22(6): 1754-66, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11865055

RESUMO

The antiapoptotic properties of the inhibitor of apoptosis (IAP) family of proteins have been linked to caspase inhibition. We have previously described an alternative mechanism of XIAP inhibition of apoptosis that depends on the selective activation of JNK1. Here we report that two other members of the IAP family, NAIP and ML-IAP, both activate JNK1. Expression of catalytically inactive JNK1 blocks NAIP and ML-IAP protection against ICE- and TNF-alpha-induced apoptosis, indicating that JNK1 activation is necessary for the antiapoptotic effect of these proteins. The MAP3 kinase, TAK1, appears to be an essential component of this antiapoptotic pathway since IAP-mediated activation of JNK1, as well as protection against TNF-alpha- and ICE-induced apoptosis, is inhibited when catalytically inactive TAK1 is expressed. In addition, XIAP, NAIP, and JNK1 bind to TAK1. Importantly, expression of catalytically inactive TAK1 did not affect XIAP inhibition of caspase activity. These data suggest that XIAP's antiapoptotic activity is achieved by two separate mechanisms: one requiring TAK1-dependent JNK1 activation and the second involving caspase inhibition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose/fisiologia , Inibidores de Caspase , Proteínas de Insetos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase 4 , MAP Quinase Quinase Quinases/metabolismo , Proteínas Associadas aos Microtúbulos , Proteínas Proto-Oncogênicas c-bcl-2 , Transdução de Sinais/fisiologia , Apoptose/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Caspase 1/farmacologia , Caspases/metabolismo , Linhagem Celular , Proteínas Cromossômicas não Histona/metabolismo , Ativação Enzimática/efeitos dos fármacos , Proteína gp120 do Envelope de HIV/metabolismo , Humanos , Proteínas Inibidoras de Apoptose , Proteínas de Insetos/farmacologia , Rim/citologia , Rim/metabolismo , MAP Quinase Quinase 7 , Proteína Quinase 8 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Inibidora de Apoptose Neuronal , Ligação Proteica , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Survivina , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X , Proteína X Associada a bcl-2
5.
Mol Biol Cell ; 15(11): 5064-74, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15356269

RESUMO

TRAIL (TNF-related apoptosis-inducing ligand) death receptors DR4 and DR5 facilitate the selective elimination of malignant cells through the induction of apoptosis. From previous studies the regulation of the DR4 and DR5 cell-death pathways appeared similar; nevertheless in this study we screened a library of small interfering RNA (siRNA) for genes, which when silenced, differentially affect DR4- vs. DR5-mediated apoptosis. These experiments revealed that expression of the signal recognition particle (SRP) complex is essential for apoptosis mediated by DR4, but not DR5. Selective diminution of SRP subunits by RNA interference resulted in a dramatic decrease in cell surface DR4 receptors that correlated with inhibition of DR4-dependent cell death. Conversely, SRP silencing had little influence on cell surface DR5 levels or DR5-mediated apoptosis. Although loss of SRP function in bacteria, yeast and protozoan parasites causes lethality or severe growth defects, we observed no overt phenotypes in the human cancer cells studied--even in stable cell lines with diminished expression of SRP components. The lack of severe phenotype after SRP depletion allowed us to delineate, for the first time, a mechanism for the differential regulation of the TRAIL death receptors DR4 and DR5--implicating the SRP complex as an essential component of the DR4 cell-death pathway.


Assuntos
Regulação da Expressão Gênica , Receptores do Fator de Necrose Tumoral/metabolismo , Partícula de Reconhecimento de Sinal , Animais , Apoptose , Western Blotting , Morte Celular , Membrana Celular/metabolismo , Separação Celular , Clonagem Molecular , Citometria de Fluxo , Biblioteca Gênica , Inativação Gênica , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Luciferases/metabolismo , Microscopia de Fluorescência , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Partícula de Reconhecimento de Sinal/metabolismo , Frações Subcelulares , Fatores de Tempo , Transfecção
6.
Oncogene ; 23(51): 8392-400, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15517021

RESUMO

Cancer develops through the successive accumulation and selection of genetic and epigenetic alterations, enabling cells to survive, replicate and evade homeostatic control mechanisms such as apoptosis and antiproliferative signals. This transformation process, however, may create vulnerabilities since the accumulation of mutations can expose synthetic lethal gene interactions and oncogene-driven cellular reprogramming ('addiction'), giving rise to new therapeutic avenues. With the completion of the human genome project, it is anticipated that the identification and characterization of genetic networks that regulate cell growth, differentiation, apoptosis and transformation will be fundamental to decoding the complexity of these processes, and ultimately, cancer itself. Genomic methodologies, such as large-scale mRNA profiling using microarrays, have already begun to reveal the molecular basis of cancer heterogeneity and the clinical behavior of tumors. The combination of traditional cell culture techniques with high-throughput screening approaches has given rise to new cellular-genomics methodologies that enable the simultaneous interrogation of thousands of genes in live cells, facilitating true functional profiling of biological processes. Among these, RNA interference (RNAi) has the potential to enable rapid genome-wide loss-of-function (LOF) screens in mammalian systems, which until recently has been the sole domain of lower organisms. Here, we present a broad overview of this maturing technology and explore how, within current technical constraints, large-scale LOF use of RNAi can be exploited to uncover the molecular basis of cancer--from the genetics of synthetic lethality and oncogene-dependent cellular addiction to the acquisition of cancer-associated cellular phenotypes.


Assuntos
Neoplasias/genética , Oncogenes , Interferência de RNA/fisiologia , Reações Falso-Negativas , Reações Falso-Positivas , Humanos , Neoplasias/metabolismo
7.
Oncogene ; 23(39): 6621-9, 2004 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-15208666

RESUMO

Gene expression profiling of anatomically diverse carcinomas and their corresponding normal tissues was used to identify genes with cancer-associated expression. We show here that the ubiquitin conjugase, UbcH10, is significantly overexpressed in many different types of cancers and is associated with the degree of tumor differentiation in carcinomas of the breast, lung, ovary and bladder, as well as in glioblastomas. We also show that UbcH10 overexpression in gastro-esophageal, and probably other carcinomas may be a direct consequence of chromosomal amplification at the UbcH10 locus, 20q13.1, a region known to be amplified in diverse tumors. To evaluate whether inhibition of UbcH10 function may be therapeutically relevant in cancer, we used small interfering RNAs (siRNAs) to silence UbcH10 transcription selectively. Diminution of UbcH10 expression significantly inhibited both tumor and normal cell proliferation without inducing cell death. However, when combined with agonists of the DR5/TRAIL receptor, siRNAs directed against the UbcH10 transcript dramatically enhanced killing of cancer cells, but not of proliferating primary human epithelial cells or fibroblasts. Together, these data demonstrate that UbcH10 plays an important role in tumor development and that its inhibition in combination with agonists of the TRAIL receptor may provide an enhanced therapeutic index.


Assuntos
Amplificação de Genes , Neoplasias/genética , Enzimas de Conjugação de Ubiquitina/antagonistas & inibidores , Apoptose/fisiologia , Sequência de Bases , Western Blotting , Primers do DNA , Humanos , Imuno-Histoquímica , Neoplasias/classificação , Neoplasias/patologia , Neoplasias/terapia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/fisiologia
8.
Chem Biol ; 10(8): 759-67, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12954335

RESUMO

Elevated expression of inhibitor of apoptosis protein (IAP) family members in various types of cancers is thought to provide a survival advantage to these cells. Thus, antiapoptotic functions of IAPs, and their potential as novel anticancer targets have attracted considerable interest. Among the IAPs, the X chromosome-linked inhibitor of apoptosis protein (XIAP) is regarded as the most potent suppressor of mammalian apoptosis through direct binding and inhibition of caspases. A high-throughput biochemical screen of a combinatorial chemical library led to the discovery of a novel nonpeptidic small molecule that has the ability to disrupt the XIAP/caspase-3 interaction. The activity of this nonpeptidic small molecule inhibitor of the XIAP/caspase-3 interaction has been characterized both in vitro and in cells. Molecules of this type can be used to conditionally inhibit the cellular function of XIAP and may provide insights into the development of therapeutic agents that act by modulating apoptotic pathways.


Assuntos
Inibidores de Caspase , Inibidores Enzimáticos/farmacologia , Proteínas/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Caspase 3 , Caspases/genética , Caspases/metabolismo , Linhagem Celular , Técnicas de Química Combinatória , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Células HCT116 , Humanos , Estrutura Molecular , Piperidinas/farmacologia , Proteínas/genética , Proteínas/metabolismo , Sulfonamidas/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
9.
Vet Immunol Immunopathol ; 164(3-4): 148-59, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25764941

RESUMO

B-cell lymphoma is one of the most frequently observed non-cutaneous neoplasms in dogs. For both human and canine BCL, the standard of care treatment typically involves a combination chemotherapy, e.g. "CHOP" therapy. Treatment for human lymphoma greatly benefited from the addition of anti-CD20 targeted biological therapeutics to these chemotherapy protocols; this type of therapeutic has not been available to the veterinary oncologist. Here, we describe the generation and characterization of a rituximab-like anti-CD20 antibody intended as a candidate treatment for canine B-cell lymphoma. A panel of anti-canine CD20 monoclonal antibodies was generated using a mouse hybridoma approach. Mouse monoclonal antibody 1E4 was selected for construction of a canine chimeric molecule based on its rank ordering in a flow cytometry-based affinity assay. 1E4 binds to approximately the same location in the extracellular domain of CD20 as rituximab, and 1E4-based chimeric antibodies co-stain canine B cells in flow cytometric analysis of canine leukocytes using an anti-canine CD21 antibody. We show that two of the four reported canine IgG subclasses (cIgGB and cIgGC) can bind to canine CD16a, a receptor involved in antibody-dependent cellular cytotoxicity (ADCC). Chimeric monoclonal antibodies were assembled using canine heavy chain constant regions that incorporated the appropriate effector function along with the mouse monoclonal 1E4 anti-canine CD20 variable regions, and expressed in CHO cells. We observed that 1E4-cIgGB and 1E4-cIgGC significantly deplete B-cell levels in healthy beagle dogs. The in vivo half-life of 1E4-cIgGB in a healthy dog was ∼14 days. The antibody 1E4-cIgGB has been selected for further testing and development as an agent for the treatment of canine B-cell lymphoma.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD20/imunologia , Doenças do Cão/tratamento farmacológico , Linfoma de Células B/veterinária , Animais , Citotoxicidade Celular Dependente de Anticorpos , Doenças do Cão/imunologia , Cães , Humanos , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/imunologia , Rituximab/uso terapêutico
10.
Cancer Biol Ther ; 2(5): 534-40, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14614322

RESUMO

Aberrant expression of the apoptosis inhibitor bcl-2 provides a survival advantage throughout oncogenesis and can facilitate chemotherapeutic resistance in a variety of human cancers. Follicular lymphoma (FL) for example, is characterized by the chromosomal translocation t(14;18), which results in bcl-2 overexpression and initiates lymphomagenesis. Although FL cells possess ample amounts of bcl-2, they respond remarkably well to standard first-round chemotherapy. However, the vast majority of patients relapses and becomes progressively resistant to therapy. We obtained cell lines derived from chemosensitive and chemoresistant FL patients, that are characterized by the chromosomal translocation t(14;18) and expression of bcl-2, to investigate how chemotherapeutic drugs can circumvent bcl-2 anti-apoptotic function and to identify alterations in those pathways that may facilitate resistance to DNA damaging drugs. In chemosensitive FL cells, we found that DNA damaging drugs promote apoptosis through p53-dependent upregulation of the TRAIL-DR5 receptor, resulting in activation of caspase-8 and downstream executioner caspases, thereby evading bcl-2 mediated suppression of apoptosis. Examination of drug resistant FL cell lines revealed that at least two defects in this pathway can contribute to chemotherapeutic resistance: 1. p53 gene mutations that disable the transcriptional response to DNA damaging drugs, including expression of the TRAIL-DR5 receptor, and 2. transcriptional repression of the cell-death executioner enzyme caspase-3.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Resistencia a Medicamentos Antineoplásicos , Linfoma Folicular/patologia , Receptores do Fator de Necrose Tumoral/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Antineoplásicos Fitogênicos/farmacologia , Inibidores de Caspase , Dano ao DNA , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Humanos , Ligantes , Linfoma Folicular/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia
11.
Proc Natl Acad Sci U S A ; 102(42): 15195-200, 2005 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-16210249

RESUMO

The MYC protooncogene is frequently deregulated in human cancers. Here, by screening a kinase-directed library of small inhibitory RNAs, we identify glycogen synthase kinase 3beta (GSK3beta) as a gene whose inactivation potentiates TNF-related apoptosis-inducing ligand death receptor-mediated apoptosis specifically in MYC-overexpressing cells. Small inhibitory RNA-induced silencing of GSK3beta prevents phosphorylation of MYC on T58, thereby inhibiting recognition of MYC by the E3 ubiquitin ligase component FBW7. Attenuating the GSK3beta-FBW7 axis results in stabilization of MYC, up-regulation of surface levels of the TNF-related apoptosis-inducing ligand death receptor 5, and potentiation of death receptor 5-induced apoptosis in vitro and in vivo. These results identify GSK3beta and FBW7 as potential cancer therapeutic targets and MYC as a critical substrate in the GSK3beta survival-signaling pathway. The results also demonstrate paradoxically that MYC-expressing tumors might be treatable by drug combinations that increase rather than decrease MYC oncoprotein function.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/genética , Caspase 8 , Caspases/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Biblioteca Gênica , Inativação Gênica , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Glicoproteínas de Membrana/genética , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/genética , Ubiquitina-Proteína Ligases/genética
12.
J Biol Chem ; 279(33): 35047-52, 2004 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-15173176

RESUMO

In many mammalian cell types, engagement of the TRAIL/Apo2L death receptors DR4 and DR5 alters mitochondrial physiology, thereby promoting the release of pro-apoptotic proteins normally contained within this organelle. A contemporary view of this process is that in so-called type II cells death receptor-activated caspase-8 cleaves the Bcl-2 family member Bid, which generates a truncated Bid fragment that collaborates with Bax, another Bcl-2 relative, to promote the release of mitochondrial factors necessary for activation of executioner caspases and apoptosis. Here we show that in some type II cells caspase-2 is necessary for optimal TRAIL-mediated cleavage of Bid. Down-regulation of caspase-2 using RNA interference significantly inhibited TRAIL-induced apoptosis. Analysis of the TRAIL proteolytic cascade following gene silencing of specific pathway components revealed that caspase-2 is necessary for efficient cleavage of Bid; however, caspase-2 proteolytic processing, which occurs downstream of Bax, is not necessary for its role in Bid cleavage.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Caspases/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Western Blotting , Caspase 2 , Caspase 3 , Caspase 7 , Caspase 8 , Caspases/metabolismo , Morte Celular , Linhagem Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Regulação para Baixo , Ativação Enzimática , Etoposídeo/farmacologia , Inativação Gênica , Humanos , Mitocôndrias/metabolismo , Modelos Biológicos , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estaurosporina/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Fatores de Tempo , Transfecção , Proteína X Associada a bcl-2
13.
Biol Chem ; 385(5): 363-72, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15195995

RESUMO

Our study was aimed at examinating whether or not the human genome encodes for previously unreported cysteine cathepsins. To this end, we used analyses of the genome sequence and mRNA expression levels. The program TBLASTN was employed to scan the draft sequence of the human genome for the 11 known cysteine cathepsins. The cathepsin-like segments in the genome were inspected, filtered, and annotated. In addition to the known cysteine cathepsins, the scan identified three pseudogenes, closely related to cathepsin L, on chromosome 10, as well as two remote homologs, tubulointerstitial protein antigen and tubulointerstitial protein antigen-related protein. No new members of the family were identified. mRNA expression profiles for 10 known human cysteine cathepsins showed varying expression levels in 46 different human tissues and cell lines. No expression of any of the three cathepsin L-like pseudogenes was found. Based on these results, it is likely that to date all human cysteine cathepsins are known.


Assuntos
Catepsinas/genética , Cisteína Endopeptidases/genética , Genoma Humano , Catepsinas/metabolismo , Cisteína/análise , Cisteína/metabolismo , Cisteína Endopeptidases/metabolismo , Bases de Dados de Proteínas , Projeto Genoma Humano , Humanos , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Homologia de Sequência
14.
Proc Natl Acad Sci U S A ; 100(8): 4568-73, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12676992

RESUMO

The genesis of carcinoma cells often involves epithelial-to-mesenchymal transitions and the acquisition of apoptosis resistance, but it is unclear whether these alterations are controlled coordinately or independently. Our previously reported effects of adenovirus E1a in human tumor cells raised the possibility that the E1a-interacting corepressor protein C-terminal-binding protein (CtBP) might selectively repress epithelial cell adhesion and proapoptotic genes. Here, we report that CtBP-knockout cells were hypersensitive to apoptosis. Correspondingly, microarray analysis of CtBP-knockout vs. CtBP-rescued mouse embryo fibroblasts revealed that many epithelial-specific and proapoptotic genes were indeed regulated by CtBP. Neither the apoptosis nor the repression activities of CtBP required histidine-315, suggesting that the proposed dehydrogenase activity is not essential for CtBP function. The results presented herein establish two functional roles of CtBP: to corepress epithelial genes, thus permitting epithelial-to-mesenchymal transitions, and to modulate the cellular threshold for apoptotic responses.


Assuntos
Apoptose/genética , Proteínas de Ligação a DNA/metabolismo , Fosfoproteínas/metabolismo , Proteínas Repressoras/metabolismo , Oxirredutases do Álcool , Animais , Sítios de Ligação/genética , Linhagem Celular , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Expressão Gênica , Humanos , Técnicas In Vitro , Camundongos , Camundongos Knockout , Oxirredutases/química , Oxirredutases/genética , Oxirredutases/metabolismo , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Proteínas Repressoras/química , Proteínas Repressoras/genética
15.
Mol Cell ; 12(3): 627-37, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14527409

RESUMO

New opportunities in mammalian functional genomics are emerging through the combination of high throughput technology and methods that allow manipulation of gene expression in living cells. Here we describe the application of an RNAi-based forward genomics approach toward understanding the biology and mechanism of TRAIL-induced apoptosis. TRAIL is a TNF superfamily member that induces selective cytotoxicity of tumor cells when bound to its cognate receptors. In addition to detecting well-characterized genes in the apoptosis pathway, we uncover several modulators including DOBI, a gene required for progression of the apoptotic signal through the intrinsic mitochondrial cell death pathway, and MIRSA, a gene that acts to limit TRAIL-induced apoptosis. Moreover, our data suggest a role for MYC and the WNT pathway in maintaining susceptibility to TRAIL. Collectively, these observations offer several insights on how TRAIL mediates the selective killing of tumor cells and demonstrate the utility of large-scale RNAi screens in mammalian cells.


Assuntos
Apoptose/genética , Apoptose/imunologia , Testes Genéticos/métodos , Vigilância Imunológica/imunologia , Glicoproteínas de Membrana/imunologia , Neoplasias/imunologia , Interferência de RNA/imunologia , Fator de Necrose Tumoral alfa/imunologia , Proteínas de Peixe-Zebra , Proteínas Reguladoras de Apoptose , Regulação da Expressão Gênica/genética , Células HeLa , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/genética , Ligante Indutor de Apoptose Relacionado a TNF , Proteínas Wnt
16.
Semin Cancer Biol ; 13(4): 293-300, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14563124

RESUMO

Oncogenesis occurs through the acquisition and selection of multiple somatic mutations--each contributing to the growth, survival and spread of the cancer. Key attributes of the malignant phenotype, such as unchecked proliferation and cell survival, can often be "reversed" by the selective diminution of dominant oncogenes by chemical or genetic means (e.g. beta-catenin in colorectal carcinomas; bcr-abl in chronic myelogenous leukemias (CMLs)). These observations suggest that the products of oncogenes, or of secondary genes that mediate and maintain tumor phenotypes, might be revealed through the systematic disruption of each and every gene in tumor-derived cells. Some of these genes may encode proteins amenable to therapeutic intervention, thus fueling the cancer drug discovery process. However, a functional assessment of each known or predicted gene in mammalian cells is a daunting task and represents the rate-limiting step in drug target identification and validation. In this regard, RNA interference (RNAi) by small interfering RNAs (siRNA) holds great promise as the "tool of choice" to mediate the selective attenuation of mammalian gene expression and protein function. Here, we review strategies by which RNAi might be used to determine the genetic alterations that contribute to malignant transformation via large-scale cell-based screens, and propose how this information can be used in conjunction with small molecule screens to identify pathways critical to cancer cell survival.


Assuntos
Carcinógenos , Desenho de Fármacos , Neoplasias/terapia , Interferência de RNA , RNA Interferente Pequeno/fisiologia , Transdução de Sinais/genética , Animais , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Terapia Genética , Genoma , Humanos , Neoplasias/genética
17.
J Biol Chem ; 277(34): 30454-62, 2002 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-12048196

RESUMO

We have previously described a new aspect of the Inhibitor of Apoptosis (IAP) family of proteins anti-apoptotic activity that involves the TAK1/JNK1 signal transduction pathway (1,2). Our findings suggest the existence of a novel mechanism that regulates the anti-apoptotic activity of IAPs that is separate from caspase inhibition but instead involves TAK1-mediated activation of JNK1. In a search for proteins involved in the XIAP/TAK1/JNK1 signaling pathway we isolated by yeast two-hybrid screening a novel X chromosome-linked IAP (XIAP)-interacting protein that we called ILPIP (hILP-Interacting Protein). Whereas ILPIP moderately activates JNK family members when expressed alone, it strongly enhances XIAP-mediated activation of JNK1, JNK2, and JNK3. The expression of a catalytically inactive mutant of TAK1 blocked XIAP/ILPIP synergistic activation of JNK1 thereby implicating TAK1 in this signaling pathway. ILPIP moderately protects against interleukin-1beta converting enzyme- or Fas-induced apoptosis and significantly potentiates the anti-apoptotic activity of XIAP. In vivo co-precipitation experiments show that both ILPIP and XIAP interact with TAK1 and tumor necrosis factor receptor-associated factor 6. Finally, expression of ILPIP did not affect the ability of XIAP to inhibit caspase activation, further supporting the idea that XIAP protection against apoptosis is achieved by two separate mechanisms: one requiring JNK1 activation and a second involving caspase inhibition.


Assuntos
Apoptose , MAP Quinase Quinase Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Caspases/metabolismo , Células Cultivadas , Ativação Enzimática , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Dados de Sequência Molecular , Proteínas Quinases/fisiologia , Transdução de Sinais , Fator 6 Associado a Receptor de TNF , Técnicas do Sistema de Duplo-Híbrido , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA