Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Leukemia ; 28(7): 1486-93, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24406841

RESUMO

Pulmonary hypertension (PH) is a frequently under recognized complication of myelofibrosis (MF). The pathophysiology of PH in MF is unknown and no definitive therapies have been established. We studied 15 patients with MF-associated PH and compared their echocardiographic and PH relevant biomarkers (nitric oxide (NO), N-terminal pro-hormone of brain natriuretic peptide (NT-pro BNP), von Willebrand antigen (vWB), ristocetin-cofactor activity (RCA) and uric acid (UA)) pre- and post-ruxolitinib treatment. Ruxolitinib decreased the plasma levels of NT-pro BNP (73%; P=0.043), UA (60%), vWB (86%) and RCA (73%; P=0.036). Improvements in echocardiographic findings were also seen in 66% of patients (P=0.022). Furthermore, marked increase in NO compared with baseline (69.75 vs 40.1 picomolar (pM); P=0.001) was observed post-ruxolitinib therapy, whereas no changes were noted with conventional therapies. Treatment with ruxolitinib also resulted in the reduction of key cytokines (tumor necrosis factor alpha, interleukin-4 (IL-4), IL-6 and IL-8) and induction of interferon-gamma. Animal studies further supported the role of ruxolitinib in the induction of NO levels. In conclusion, aberrant Janus kinase (JAK)-signal transducer and activator of transcription signaling in MF may mediate PH through dysregulation of NO and cytokine levels, which can be restored by therapy with JAK inhibitors suggesting that inhibition of this pathway is a novel target for the management of patients with PH.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Mielofibrose Primária/complicações , Pirazóis/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Animais , Biomarcadores/sangue , Citocinas/sangue , Modelos Animais de Doenças , Ecocardiografia , Feminino , Ferritinas/sangue , Humanos , Hipertensão Pulmonar/diagnóstico , Janus Quinases/antagonistas & inibidores , Masculino , Camundongos Knockout , Pessoa de Meia-Idade , Óxido Nítrico/sangue , Nitrilas , Mielofibrose Primária/sangue , Mielofibrose Primária/genética , Pirimidinas
2.
Oncogene ; 33(24): 3195-204, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-23851499

RESUMO

The non-receptor tyrosine kinase Src is a major player in multiple physiological responses including growth, survival and differentiation. Overexpression and/or oncogenic mutation in the Src gene have been documented in human tumors. The v-Src protein is an oncogenic mutant of Src, which promotes cell survival, migration, invasion and division. GRIM-19 is an antioncogene isolated using a genome-wide knockdown screen. Genes associated with Retinoid-IFN-induced Mortality (GRIM)-19 binds to transcription factor STAT3 and ablates its pro-oncogenic effects while v-Src activates STAT3 to promote its oncogenic effects. However, we found that GRIM-19 inhibits the pro-oncogenic effects of v-Src independently of STAT3. Here, we report the identification of functionally inactivating GRIM-19 mutations in a set of head and neck cancer patients. While wild-type GRIM-19 strongly ablated v-Src-induced cell migration, cytoskeletal remodeling and tumor metastasis, the tumor-derived mutants (L(71)P, L(91)P and A(95)T) did not. These mutants were also incapable of inhibiting the drug resistance of v-Src-transformed cells. v-Src downregulated the expression of Pag1, a lipid raft-associated inhibitor of Src, which was restored by wild-type GRIM-19. The tumor-derived mutant GRIM-19 proteins failed to upregulate Pag1. These studies show a novel mechanism that deregulates Src activity in cancer cells.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica/patologia , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Mutação/genética , NADH NADPH Oxirredutases/genética , Proteína Oncogênica pp60(v-src)/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Movimento Celular , Proliferação de Células , Imunofluorescência , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Oncogênica pp60(v-src)/genética , Fosforilação , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Oncogene ; 31(6): 776-86, 2012 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21725364

RESUMO

In clear-cell renal cell carcinoma (ccRCC), inactivation of the tumor suppressor von Hippel-Lindau (VHL) occurs in the majority of the tumors and is causal for the pathogenesis of ccRCC. Recently, a large-scale genomic sequencing study of ccRCC tumors revealed that enzymes that regulate histone H3 lysine 4 trimethylation (H3K4Me3), such as JARID1C/KDM5C/SMCX and MLL2, were mutated in ccRCC tumors, suggesting that H3K4Me3 might have an important role in regulating gene expression and tumorigenesis. In this study we report that in VHL-deficient ccRCC cells, the overall H3K4Me3 levels were significantly lower than that of VHL+/+ counterparts. Furthermore, this was hypoxia-inducible factor (HIF) dependent, as depletion of HIF subunits by small hairpin RNA in VHL-deficient ccRCC cells restored H3K4Me3 levels. In addition, we demonstrated that only loss of JARID1C, not JARID1A or JARID1B, abolished the difference of H3K4Me3 levels between VHL-/- and VHL+/+ cells, and JARID1C displayed HIF-dependent expression pattern. JARID1C in VHL-/- cells was responsible for the suppression of HIF-responsive genes insulin-like growth factor-binding protein 3 (IGFBP3), DNAJC12, COL6A1, growth and differentiation factor 15 (GDF15) and density-enhanced phosphatase 1. Consistent with these findings, the H3K4Me3 levels at the promoters of IGFBP3, DNAJC12, COL6A1 and GDF15 were lower in VHL-/- cells than in VHL+/+ cells, and the differences disappeared after JARID1C depletion. Although HIF2α is an oncogene in ccRCC, some of its targets might have tumor suppressive activity. Consistent with this, knockdown of JARID1C in 786-O VHL-/- ccRCC cells significantly enhanced tumor growth in a xenograft model, suggesting that JARID1C is tumor suppressive and its mutations are tumor promoting in ccRCC. Thus, VHL inactivation decreases H3K4Me3 levels through JARID1C, which alters gene expression and suppresses tumor growth.


Assuntos
Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Histona Desmetilases , Histonas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Neoplasias Renais/genética , Neoplasias Renais/patologia , Lisina/metabolismo , Metilação , Camundongos , Camundongos Nus , Mutação , Oxirredutases N-Desmetilantes/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Carga Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/genética
4.
Oncogene ; 28(25): 2383-92, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19430495

RESUMO

Inositol hexakisphosphate kinase 2 (IP6K2), a member of the inositol hexakisphosphate kinase family, functions as a growth suppressive and apoptosis-enhancing kinase during cell stress. We created mice with a targeted deletion of IP6K2; these mice display normal embryogenesis, development, growth and fertility. Chronic exposure to the carcinogen 4-nitroquinoline 1-oxide (4-NQO, a UV-mimetic compound) in drinking water resulted in fourfold increased incidence of invasive squamous cell carcinoma (SCC) formation in the oral cavity and esophagus of the knockout (KO) mice compared to the wild-type (WT) littermates. Paradoxically, KO mice displayed relative resistance to ionizing radiation and exhibit enhanced survival following 8-10 Gy total body irradiation. Primary KO fibroblasts displayed resistance to antiproliferative effects of interferon-beta and increased colony forming units following ionizing radiation. Radioresistance of KO fibroblasts was associated with accelerated DNA repair measured by comet assay. Direct microinjection of 5-PP-Ins(1,2,3,4,6)P(5) (the enzymatic product of IP6K2), but not InsP(6) (the substrate of IP6K2) induced cell death in SCC22A squamous carcinoma cells.


Assuntos
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Deleção de Genes , Predisposição Genética para Doença/genética , Neoplasias Bucais/genética , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , 4-Nitroquinolina-1-Óxido/toxicidade , Animais , Apoptose , Carcinógenos/toxicidade , Carcinoma de Células Escamosas/radioterapia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Raios gama , Perfilação da Expressão Gênica , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microinjeções , Tolerância a Radiação , Radiação Ionizante , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Oncogene ; 28(10): 1339-47, 2009 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-19151760

RESUMO

GRIM-19 (Gene associated with Retinoid-Interferon-induced Mortality 19) is a novel tumor suppressor regulated by interferon/retinoid combination. We have recently shown that GRIM-19 inhibits v-Src-induced oncogenic transformation and metastatic behavior of cells. Oncogenic v-Src induces cell motility by cytoskeletal remodeling, especially the formation of podosomes and. Here, we show that GRIM-19 inhibited the v-Src-induced cell motility by inhibiting cytoskeletal remodeling, that is, podosome formation. We also show that the N terminus of GRIM-19 played a major role in this process and identified critical residues in this region. More importantly, we show that tumor-associated GRIM-19 mutations disrupted its ability to inhibit v-Src-induced cell motility. These actions appear to occur independently of STAT3, a known target of GRIM-19-mediated inhibition. Lastly, tumor-associated GRIM-19 mutants significantly lost their ability to control v-Src-induced metastases in vivo, indicating the biological and pathological significance of these observations.


Assuntos
Movimento Celular , Citoesqueleto/química , NADH NADPH Oxirredutases/fisiologia , Proteína Oncogênica pp60(v-src)/antagonistas & inibidores , Animais , Transformação Celular Neoplásica , Cortactina/metabolismo , Camundongos , NADH NADPH Oxirredutases/química , NADH NADPH Oxirredutases/genética , Fosforilação , Relação Estrutura-Atividade
6.
Oncogene ; 26(20): 2894-901, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17130843

RESUMO

Ovarian cancer is a highly metastatic disease. Lysophosphatidic acid (LPA) levels are elevated in ascites from ovarian cancer patients, but its potential role in ovarian cancer metastasis has just begun to be revealed. In this work, we show that LPA stimulates invasion of primary ovarian cancer cells, but not ovarian epithelial or borderline ovarian tumor cells, although these benign cells indeed respond to LPA in cell migration. We have found that LPA downregulates tissue inhibitor of metalloproteinases (TIMPs). TIMP2 and TIMP3 play functional role in LPA-induced invasion as negative regulators. G(i) protein, phosphatidylinositol-3 kinase (PI3K), p38 mitogen-activated protein kinase (MAPK), cytosolic phospholipase A(2) and urokinase type plasminogen activator (uPA) are required for LPA-induced cells invasion. TIMP3 may affect two independent downstream targets, vascular endothelial growth factor receptor and p38 MAPK. In vivo, LPA stimulates tumor metastasis in an orthotopic ovarian tumor model, which can be inhibited by a PI3K inhibitor, LY294002. In summary, LPA is likely a key component for promoting ovarian metastasis in vivo. LPA downregulates TIMP3, which may have targets other than metalloproteinases. Our in vivo metastasis mouse model is useful for studying the efficacy of therapeutic regimes of ovarian cancer.


Assuntos
Movimento Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/genética , Inibidores Teciduais de Metaloproteinases/genética , Animais , Células Epiteliais/efeitos dos fármacos , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Humanos , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Ovarianas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores Teciduais de Metaloproteinases/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Cell Death Differ ; 11(8): 915-23, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15118763

RESUMO

Melanoma cells are relatively resistant to Apo2L/TRAIL (TNF-related apoptosis-inducing ligand). We postulated that resistance might result from higher expression of inhibitors of apoptosis including Bcl-2, FLIP (FLICE-like inhibitory protein) or IAPs such as XIAP (X-linked inhibitor of apoptosis) or survivin. Compared to scrambled or mismatch controls, targeting individual inhibitors with siRNA (si-Bcl-2, si-XIAP, si-FLIP or si-Surv), followed by Apo2L/TRAIL resulted in marked increase in apoptosis in melanoma cells. Compared to Bcl-2 or FLIP, siRNAs against XIAP and survivin were most potent in sensitizing melanoma cells. A similar substantial increase in apoptosis was seen in renal carcinoma cells (SKRC-45, Caki-2), following the inhibition of either XIAP or survivin by siRNAs. Apo2L/TRAIL treatment in IAP-targeted cells resulted in cleavage of Bid, activation of caspase-9 and cleavage of PARP (poly ADP-ribose polymerase). Thus, Apo2L/TRAIL resistance can be overcome by interfering with expression of inhibitors of apoptosis regulating both extrinsic (death receptor) or intrinsic (mitochondrial) pathways of apoptosis in melanoma cells.


Assuntos
Apoptose/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana/farmacologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/metabolismo , Caspase 9 , Caspases/metabolismo , Genes bcl-2/fisiologia , Humanos , Proteínas Inibidoras de Apoptose , Melanoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias , RNA Interferente Pequeno/genética , Survivina , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
8.
Apoptosis ; 8(3): 237-49, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12766484

RESUMO

IFNs are a family of cytokines with pleiotropic biological effects mediated by scores of responsive genes. IFNs were the first human proteins to be effective in cancer therapy and were among the first recombinant DNA products to be used clinically. Both quality and quantity of life has been improved in response to IFNs in various malignancies. Despite its beneficial effects, unraveling the mechanisms of the anti-tumor effects of IFN has proven to be a complex task. IFNs may mediate anti-tumor effects either indirectly by modulating immunomodulatory and anti-angiogenic responses or by directly affecting proliferation or cellular differentiation of tumor cells. Both direct or indirect effects of IFNs result from induction of a subset of genes, called IFN stimulated genes (ISGs). In addition to the ISGs implicated in anti-viral, anti-angiogenic, immunomodulatory and cell cycle inhibitory effects, oligonucleotide microarray studies have identified ISGs with apoptotic functions. These include TNF-alpha related apoptosis inducing ligand (TRAIL/Apo2L), Fas/FasL, XIAP associated factor-1 (XAF-1), caspase-4, caspase-8, dsRNA activated protein kinase (PKR), 2'5'A oligoadenylate synthetase (OAS), death activating protein kinases (DAP kinase), phospholipid scramblase, galectin 9, IFN regulatory factors (IRFs), promyelocytic leukemia gene (PML) and regulators of IFN induced death (RIDs). In vitro IFN-alpha, IFN-beta and IFN-gamma induced apoptosis in multiple cell lines of varied histologies. This review will emphasize possible mechanisms and the role of ISGs involved in mediating apoptotic function of IFNs.


Assuntos
Apoptose/genética , Regulação da Expressão Gênica/genética , Interferons/genética , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Regulação da Expressão Gênica/imunologia , Humanos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/genética
9.
Oncogene ; 20(28): 3703-15, 2001 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-11439333

RESUMO

Interferons (IFNs) and retinoids are potent tumor growth suppressors. We have shown earlier that the IFN-beta and all-trans retinoic acid combination, but not the single agents, induces death in several tumor cell lines. Employing a genetic approach we have recently identified several Genes associated with Retinoid-IFN induced Mortality (GRIM) that mediate the cell death effect of IFN/RA combination. One of the GRIMs, GRIM-12, was identical to human thioredoxin reductase (TR), an enzyme that controls intracellular redox state. To define the participants of TR mediated death pathway we have examined the role of thioredoxin (Trx), its downstream substrate, and its influence on IFN/RA-induced death regulation. Inhibition of the thioredoxin expression by antisense RNA suppressed cell death. Similarly, a mutant Trx1 lacking the critical cysteine residues blocked cell death. In contrast, overexpression of wildtype thioredoxin augmented cell death. This effect of Trx1 was in part due to its ability to augment cell death via caspase-8. The redox inactive Trx1 mutant inhibits the cell death induced by caspase-8 but not caspase-3. These studies identify a novel mechanism of cell death regulation by IFN/RA combination involving redox enzymes.


Assuntos
Antineoplásicos/metabolismo , Caspases/metabolismo , Morte Celular , Interferon beta/metabolismo , Transdução de Sinais , Tiorredoxinas/metabolismo , Tretinoína/metabolismo , Antineoplásicos/farmacologia , Caspase 8 , Caspase 9 , Regulação para Baixo , Ativação Enzimática , Expressão Gênica , Humanos , Interferon beta/farmacologia , Tiorredoxinas/genética , Transfecção , Tretinoína/farmacologia , Células Tumorais Cultivadas
10.
Oncogene ; 20(31): 4235-48, 2001 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-11464290

RESUMO

We have shown earlier that the IFN-beta and all-trans retinoic acid (RA) combination, but not the single agents, induces death in several tumor cell lines. Employing a genetic technique we have identified several Genes associated with Retinoid-IFN induced Mortality (GRIM). One of the GRIMs was human thioredoxin reductase (TR), a redox enzyme. Since the overexpressed TR augments IFN/RA stimulated cell death, we explored the mechanisms of TR-mediated death. Here we show that TR augments cell death by upregulating the transcriptional activity of p53 tumor suppressor. This process does not involve a physical increase in levels of p53. Using redox inactive mutants of TR and its substrate, thioredoxin (Trx), we demonstrate that IFN/RA-induced regulation of p53 dependent gene expression requires TR and Trx. In contrast-over-expression of wildtype TR or Trx augment the p53 dependent gene expression in response to IFN/RA treatment. Consistent with these results an increased DNA binding activity of p53 was noted in the presence of TR. These studies identify a novel mechanism of p53 mediated cell death regulation involving redox enzymes.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/fisiologia , Proteínas de Bactérias , Regulação da Expressão Gênica/fisiologia , Interferon beta/administração & dosagem , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/metabolismo , Tretinoína/administração & dosagem , Proteína Supressora de Tumor p53/fisiologia , Sequência de Bases , Primers do DNA , Humanos , Tiorredoxina Dissulfeto Redutase/genética , Tiorredoxinas/genética , Transcrição Gênica , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
11.
J Biol Chem ; 276(27): 24965-70, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11337497

RESUMO

Interferons (IFNs) regulate the expression of genes that mediate their antiviral, antitumor, and immunomodulatory actions. We have previously shown that IFN-beta suppresses growth of human ovarian carcinoma xenografts in vivo and induces apoptosis of ovarian carcinoma cells in vitro. To investigate mechanisms of IFN-beta-induced apoptosis we employed an antisense technical knockout approach to identify gene products that mediate cell death and have isolated several regulators of interferon-induced death (RIDs). In this investigation, we have characterized one of the RIDs, RID-2. Sequence analysis revealed that RID-2 was identical to human inositol hexakisphosphate kinase 2 (IP6K2). IP6K2 is post-transcriptionally induced by IFN-beta in ovarian carcinoma cells. A mutant IP6K2 with substitutions in the putative inositol phosphate binding domain abrogates IFN-beta-induced apoptosis. These studies identify a novel function for IP6K2 in cell growth regulation and apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Interferon beta/farmacologia , Isoenzimas/metabolismo , Neoplasias Ovarianas/patologia , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sequência Consenso , Feminino , Humanos , Oligonucleotídeos Antissenso/farmacologia , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
12.
J Biol Chem ; 276(27): 24843-54, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11331281

RESUMO

Interferons (IFNs) and retinoids are potent biological response modifiers. The IFN-beta and all-trans-retinoic acid combination, but not these single agents individually, induces death in several tumor cell lines. To elucidate the molecular basis for these actions, we have employed an antisense knockout approach to identify the gene products that mediate cell death and isolated several genes associated with retinoid-IFN-induced mortality (GRIMs). One of the GRIM cDNAs, GRIM-12, was identical to human thioredoxin reductase (TR). To define the functional relevance of TR to cell death and to define its mechanism of death-modulating functions, we generated mutants of TR and studied their influence on the IFN/RA-induced death regulatory functions of caspases. Wild-type TR activates cell death that was inhibited in the presence of caspase inhibitors or catalytically inactive caspases. A mutant TR, lacking the active site cysteines, inhibits the cell death induced by caspase 8. IFN/all-trans-retinoic acid-induced cytochrome c release from the mitochondrion was promoted in the presence of wild type and was inhibited in the presence of mutant TR. We find that TR modulates the activity of caspase 8 to promote death. This effect is in part caused by the stimulation of death receptor gene expression. These studies identify a new mechanism of cell death regulation by the IFN/all-trans-retinoic acid combination involving redox enzymes.


Assuntos
Morte Celular/efeitos dos fármacos , Morte Celular/genética , Proteínas de Drosophila , Interferons/farmacologia , Tiorredoxina Dissulfeto Redutase/metabolismo , Tretinoína/farmacologia , Animais , Caspase 8 , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Galinhas , Inibidores de Cisteína Proteinase/farmacologia , Grupo dos Citocromos c/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Mitocôndrias/enzimologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Oxirredução , Coelhos , Receptores do Fator de Necrose Tumoral/metabolismo , Tiorredoxina Dissulfeto Redutase/genética , Transfecção , Células Tumorais Cultivadas
13.
J Biol Chem ; 275(43): 33416-26, 2000 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-10924506

RESUMO

We show here that the combination of interferon-beta (IFN-beta) and all-trans-retinoic acid (RA) induces the death of tumor cells. To understand the molecular basis for synergistic growth-suppressive action and to identify the gene products that participate in this process, we have employed an antisense knock-out technique. This approach permits the isolation of cell death-associated genes based on their selective inactivation by overexpression of antisense cDNAs. Because the antisense mRNA inactivates gene expression of death-specific genes, transfected cells survive in the presence death inducers. Several Genes associated with Retinoid-IFN-induced Mortality (GRIM) were identified using this approach. Here we report the isolation of a novel GRIM gene, GRIM-19. This 552-base pair cDNA encodes a 16-kDa protein. Antisense expression of GRIM-19 confers a strong resistance against IFN/RA-induced death by reducing the intracellular levels of GRIM-19 protein. Overexpression of GRIM-19 enhances cell death in response to IFN/RA. GRIM-19 is primarily a nuclear protein whose expression is induced by the IFN/RA combination. Together, our studies identify a novel cell death-regulatory molecule.


Assuntos
Apoptose , Proteínas de Drosophila , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon beta/farmacologia , Neuropeptídeos/genética , Tretinoína/farmacologia , Sequência de Aminoácidos , Neoplasias da Mama/patologia , DNA Antissenso/farmacologia , Feminino , Humanos , Dados de Sequência Molecular , Neuropeptídeos/química , Neuropeptídeos/imunologia , RNA Mensageiro/análise , Relação Estrutura-Atividade , Células Tumorais Cultivadas
14.
Biochim Biophys Acta ; 1496(2-3): 196-206, 2000 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-10771088

RESUMO

Interferons (IFNs) suppress cell growth by inducing cellular genes. The anti-estrogen tamoxifen (Tam), binds to estrogen receptor and inhibits transcription of estrogen stimulated genes. In cells resistant to IFN-induced growth suppression, IFN/Tam combination causes cell death. We previously reported that the combination of IFN-beta and Tam was a more potent growth suppressor of human tumor xenografts than either agent alone. The IFN/Tam combination acts in a manner similar to the IFN/retinoic acid combination. Using a genetic technique, we have recently identified several genes associated with retinoid-IFN-induced mortality (GRIM). One such gene, GRIM-12, was identical to human thioredoxin reductase (TR). In the present study we have examined whether the IFN/Tam combination also requires GRIM-12 for inducing cell death. We report here that GRIM-12 is necessary for mediating the cell death effects of IFN/Tam, and its expression is induced by IFN/Tam at a post-transcriptional stage. Repression of GRIM-12 levels either by antisense expression or by dominant negative inhibitors caused resistance to IFN/Tam induced death and promoted cell growth. Overexpression of GRIM-12 increased IFN/Tam induced apoptosis. Thus, these studies have identified a critical role for GRIM-12 (TR) in apoptosis.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Interferon beta/farmacologia , Tamoxifeno/farmacologia , Tiorredoxina Dissulfeto Redutase/metabolismo , Animais , Neoplasias da Mama , Divisão Celular/efeitos dos fármacos , DNA Antissenso , Sinergismo Farmacológico , Endorribonucleases/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Neoplasias Ovarianas , Tiorredoxina Dissulfeto Redutase/biossíntese , Tiorredoxina Dissulfeto Redutase/genética , Células Tumorais Cultivadas
15.
Nucl Med Commun ; 20(11): 1055-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10572916

RESUMO

We studied the biodistribution and tumour localization of 99Tcm-labelled-5-thio-D-glucose (99Tcm-TG). 5-Thio-D-glucose was labelled with 99Tcm by direct stannous ion reduction. The biodistribution of 99Tcm-TG was investigated in normal rabbits and in mice bearing experimental tumours. In rabbits, the plasma and clearance of 99Tcm-TG was 14.5 +/- 2.0 and 11.3 +/- 3.0 ml.min-1 respectively. Urinary excretion at 1 h was 53 +/- 5%. 99Tcm-TG was injected intravenously in mice bearing MC26 colon carcinoma and tissue samples were analysed by gamma scintillation counting at various times. Uptake of 99Tcm-TG in tumour at 1 and 3 h was 1.6 +/- 0.3% and 1.2 +/- 0.3%; the tumour to muscle ratios were 2.7:1 and 4:1 respectively. The autoradiographic biodistribution of 99Tcm-TG in MX-1 human breast xenografted nude mice showed more persistent tumour uptake of 99Tcm-TG than 14C-2-deoxyglucose (14C-DG). 99Tcm-TG accumulated in the centre of the tumours; 14C-DG was decreased in this central region probably because of zones of infarction on necrosis. The discordance between the tumour uptake of 99Tcm-TG and 14C-DG indicates that 99Tcm-TG does not act like a glucose analog, suggesting 99Tcm-TG avidity for zones of infarction or necrosis. The further study of 99Tcm-TG in tumours and ischaemic injury is warranted.


Assuntos
Glucose/análogos & derivados , Compostos de Organotecnécio/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Animais , Antimetabólitos , Autorradiografia , Neoplasias da Mama/diagnóstico por imagem , Desoxiglucose , Glucose/farmacocinética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante de Neoplasias/diagnóstico por imagem , Neoplasias Experimentais/diagnóstico por imagem , Coelhos , Cintilografia , Distribuição Tecidual , Transplante Heterólogo
16.
Mol Cell Biol ; 18(11): 6493-504, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9774665

RESUMO

Interferons (IFNs) and retinoids are potent biological response modifiers. By using JAK-STAT pathways, IFNs regulate the expression of genes involved in antiviral, antitumor, and immunomodulatory actions. Retinoids exert their cell growth-regulatory effects via nuclear receptors, which also function as transcription factors. Although these ligands act through distinct mechanisms, several studies have shown that the combination of IFNs and retinoids synergistically inhibits cell growth. We have previously reported that IFN-beta-all-trans-retinoic acid (RA) combination is a more potent growth suppressor of human tumor xenografts in vivo than either agent alone. Furthermore, the IFN-RA combination causes cell death in several tumor cell lines in vitro. However, the molecular basis for these growth-suppressive actions is unknown. It has been suggested that certain gene products, which mediate the antiviral actions of IFNs, are also responsible for the antitumor actions of the IFN-RA combination. However, we did not find a correlation between their activities and cell death. Therefore, we have used an antisense knockout approach to directly identify the gene products that mediate cell death and have isolated several genes associated with retinoid-IFN-induced mortality (GRIM). In this investigation, we characterized one of the GRIM cDNAs, GRIM-12. Sequence analysis suggests that the GRIM-12 product is identical to human thioredoxin reductase (TR). TR is posttranscriptionally induced by the IFN-RA combination in human breast carcinoma cells. Overexpression of GRIM-12 causes a small amount of cell death and further enhances the susceptibility of cells to IFN-RA-induced death. Dominant negative inhibitors directed against TR inhibit its cell death-inducing functions. Interference with TR enzymatic activity led to growth promotion in the presence of the IFN-RA combination. Thus, these studies identify a novel function for TR in cell growth regulation.


Assuntos
Apoptose/efeitos dos fármacos , Interferons/farmacologia , Tiorredoxina Dissulfeto Redutase/fisiologia , Tretinoína/farmacologia , Sequência de Aminoácidos , Neoplasias da Mama/enzimologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Oligonucleotídeos Antissenso/farmacologia , Análise de Sequência , Células Tumorais Cultivadas
17.
Proc Natl Acad Sci U S A ; 95(3): 1085-90, 1998 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-9448289

RESUMO

Murine polyoma virus (MPyV) is a small DNA virus that induces tumors in multiple tissues of infected host. In this investigation, we show that cell lines derived from wild type virus-induced breast tumors are resistant to the growth inhibitory action of interferon beta (IFN-beta). Furthermore, replication of heterologous viruses such as vesicular stomatitis virus and encephalomyocarditis virus was not inhibited by IFN-beta in these cells. This effect was due to inhibition of IFN-stimulated gene expression by viral T antigen. Activation of IFN-stimulated gene factor 3 was inhibited in cells derived from a tumor induced by wild-type MPyV but not those from a mutant that lacks the pRB binding site of the large T antigen. Similarly IFN-gamma-inducible gene expression was also inhibited in cells transformed by wild-type virus. The levels of components of IFN-stimulated gene factor 3 and signal transducing Janus tyrosine kinases were comparable between the cells transformed by the wild-type and mutant viruses. The viral large T antigen bound to Janus tyrosine kinase 1 and inactivated signaling through IFN receptors. Thus, these studies identify a mechanism of viral resistance to IFN action.


Assuntos
Antígenos Transformantes de Poliomavirus/fisiologia , Transformação Celular Viral , Regulação Viral da Expressão Gênica , Interferon beta/fisiologia , Interferon gama/fisiologia , Polyomavirus/fisiologia , Animais , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Vírus da Encefalomiocardite/fisiologia , Humanos , Fator Regulador 3 de Interferon , Janus Quinase 1 , Camundongos , Polyomavirus/imunologia , Ligação Proteica , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Vírus da Estomatite Vesicular Indiana/fisiologia , Replicação Viral
18.
J Interferon Cytokine Res ; 17(11): 681-93, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9402106

RESUMO

Solid tumors are relatively resistant to growth inhibition by interferons (IFNs). To enhance sensitivity, we assessed combinations of IFNs with tamoxifen in estrogen receptor-positive (ER-positive) and ER-negative human tumor xenografts. In nude mice, the growth of MCF-7 human breast tumors (ER-positive) and NIH-OVCAR-3 ovarian tumors (functionally ER-negative) was suppressed completely when tamoxifen and IFN-alpha or IFN-beta was started 2 days after tumor inoculation. Established, 6-week-old MCF-7 and NIH-OVCAR-3 tumors regressed when treated with the combination of IFN-beta and tamoxifen but not with single-agent therapy. Treatment with the combination also resulted in an augmented antitumor response in vivo in an ER-negative breast tumor (MDA-MB-231), a colon carcinoma (HT-29), and a melanoma (SK-MEL-1). Antiproliferative studies in vitro suggested that growth of both MCF-7 and NIH-OVCAR-3 cells was inhibited to a greater degree by combination treatment with human IFN-alpha and tamoxifen or IFN-beta and tamoxifen compared with single agents. Median effect analysis defined synergy. Four ER-negative carcinomas (MDA-MB-231, MDA-MB-468, BT-20, and HT-29) also exhibited synergistic growth inhibition in response to the drug combination. The response of these four cell lines was particularly striking. Tamoxifen as a single agent had little effect (up to 2.0 microM) but caused enhanced antiproliferative activity when added to IFN-beta. Sequential treatment of MCF-7 cells in vitro with tamoxifen followed by IFN-beta was more effective at inhibiting growth than treatment with IFN-beta followed by tamoxifen, suggesting that tamoxifen modulated the anticellular response to IFN-beta rather than the converse. Similar results were obtained with IFN-alpha. Cell cycle analysis indicated that 7 days of exposure to the combination resulted in MCF-7 cell fragmentation and death. Together with our recent studies demonstrating enhancement of IFN-stimulated gene expression (ISG) by tamoxifen pretreatment in IFN-resistant cells, these data suggest that combination treatment with tamoxifen and IFNs may increase ISG expression in IFN-resistant tumors, leading to augmented antitumor effects. These effects appear to be independent of ER expression.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Receptores de Estrogênio/análise , Tamoxifeno/administração & dosagem , Animais , Divisão Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Interferon Tipo I/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células Tumorais Cultivadas
19.
Proc Natl Acad Sci U S A ; 94(14): 7227-32, 1997 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-9207073

RESUMO

p48 protein is an integral component of the multimeric interferon (IFN)-regulated transcription factor, ISGF3. We have shown earlier that this gene is regulated by a novel IFN-gamma-regulated element. In addition to the IFN-regulated element, a myc-max binding site is also present in this promoter. In this investigation we have studied the role of this site in the regulation of the p48 gene. In serum-induced quiescent cells Myc up-regulated the expression of p48 mRNA. We show that the protooncogene Myc regulates the expression of p48 through the element CACGTG. Mutations in this motif abolish Myc-inducibility of the reporter genes carrying p48 promoter elements. Purified Myc and Max proteins interact with the Myc-stimulated element of the p48 promoter. We also show that cells lacking p48 expression are highly susceptible to the cytocidal action of anticancer drugs. Taken together these data suggest that p48 may function as an anti-stress cell survival factor.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Genes myc , Fatores de Transcrição/genética , Animais , Sequência de Bases , Linhagem Celular , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferons/genética , Camundongos , Dados de Sequência Molecular
20.
Semin Oncol ; 24(3 Suppl 9): S9-99-S9-104, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9208879

RESUMO

Alpha-interferons (IFN-alphas) have been shown to be effective agents in inducing the regression of various malignancies, including leukemias and lymphomas as well as solid tumors, and are the first human therapeutic proteins to result in increased survival in cancer patients. Based on their pleiotropic activities, IFNs have the potential for interacting synergistically with other anticancer agents. For example, preclinical in vitro and animal data suggest a synergistic antitumor interaction between IFN-alpha2 and the antiestrogens toremifene and tamoxifen. Further studies are required to elucidate the molecular basis for such synergistic interactions, particularly with respect to IFN-stimulated genes.


Assuntos
Interferon Tipo I/uso terapêutico , Neoplasias/tratamento farmacológico , Terapia Combinada , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Proteínas Recombinantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA