Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Gene Ther ; 15(9): 616-24, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18535619

RESUMO

Vaccinia virus has recently been used as an expression vector for gene delivery and an oncolytic agent for cancer therapy. Although it has been established that interferon-induced double-stranded RNA (dsRNA)-activated protein kinase (PKR) and RNase L interfere with viral replication, little else is known about the other host factors that might affect viral replication and virus-mediated host cell killing. In this study, we evaluated the roles of c-Jun NH2-terminal kinase (JNK) in oncolytic vaccinia virus replication and vaccinia virus-mediated host cell killing. We found that JNK knockout mouse embryonic fibroblasts (MEFs) were more susceptible to oncolytic vaccinia virus infection than wild-type MEFs. Moreover, viral replication and the production of infectious viral progeny were up to 100-fold greater in JNK-deficient MEFs than in wild-type MEFs. A similar result was observed for wild-type vaccinia virus. The increased killing of infected cells and the production of viral progeny was also observed in wild-type MEFs that had been treated with JNK inhibitors and in human colon cancer cells that had been transfected with dominant-negative JNK constructs. Moreover, testing on several human lung cancer cell lines and HeLa cells showed an inverse correlation between levels of JNK expression and susceptibility to oncolytic vaccinia virus. Our study also revealed that oncolytic virus infection-mediated PKR activation was blocked or diminished in JNK-deficient MEFs. The adenovirus-mediated ectopic expression of human PKR in JNK-deficient MEFs reduced vaccinia virus replication to the levels observed in wild-type MEFs, indicating that JNK is required for vaccinia virus to efficiently activate PKR. Our results demonstrated that the cellular status of JNK function can dramatically affect oncolytic vaccinia virus replication and vaccinia virus-mediated host cell killing. This finding may enable further improvements in oncolytic virotherapy using vaccinia virus.


Assuntos
Proteína Quinase 9 Ativada por Mitógeno/genética , Vírus Oncolíticos/fisiologia , Vaccinia virus/fisiologia , Replicação Viral , eIF-2 Quinase/metabolismo , Animais , Linhagem Celular Tumoral , Ativação Enzimática/genética , Fatores de Iniciação em Eucariotos/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/genética , Vírus Oncolíticos/genética , Fosforilação , RNA de Cadeia Dupla/metabolismo , Vaccinia virus/genética , eIF-2 Quinase/genética
2.
Oncogene ; 26(8): 1201-12, 2007 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-16924232

RESUMO

Double-stranded RNA-dependent protein kinase (PKR), a ubiquitously expressed serine/threonine kinase, has been implicated in the regulation or modulation of cell growth through multiple signaling pathways, but how PKR regulates tumor necrosis factor (TNF)-induced signaling pathways is poorly understood. In the present study, we used fibroblasts derived from PKR gene-deleted mice to investigate the role of PKR in TNF-induced activation of nuclear factor-kappaB (NF-kappaB), mitogen-activated protein kinases (MAPKs) and growth modulation. We found that in wild-type mouse embryonic fibroblast (MEF), TNF induced NF-kappaB activation as measured by DNA binding but deletion of PKR abolished this activation. This inhibition was associated with suppression of inhibitory subunit of NF-kappaB (IkappaB)alpha kinase (IKK) activation, IkappaBalpha phosphorylation and degradation, p65 phosphorylation and nuclear translocation, and NF-kappaB-dependent reporter gene transcription. TNF-induced Akt activation needed for IKK activation was also abolished by deletion of PKR. NF-kappaB activation was diminished in PKR-deleted cells transfected with TNF receptor (TNFR) 1, TNFR-associated death domain and TRAF2 plasmids; NF-kappaB activated by NF-kappaB-inducing kinase, IKK or p65, however, was minimally affected. Among the MAPKs, it was interesting that whereas TNF-induced c-Jun N-terminal kinase (JNK) activation was abolished, activation of p44/p42 MAPK and p38 MAPK was potentiated in PKR-deleted cells. TNF induced the expression of NF-kappaB-regulated gene products cyclin D1, c-Myc, matrix metalloproteinase-9, survivin, X-linked inhibitor-of-apoptosis protein (IAP), IAP1, Bcl-x(L), A1/Bfl-1 and Fas-associated death domain protein-like IL-1beta-converting enzyme-inhibitory protein in wild-type MEF but not in PKR-/- cells. Similarly, TNF induced the proliferation of wild-type cells, but this proliferation was completely suppressed in PKR-deleted cells. Overall, our results indicate that PKR differentially regulates TNF signaling; IKK, Akt and JNK were positively regulated, whereas p44/p42 MAPK and p38 MAPK were negatively regulated.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , eIF-2 Quinase/fisiologia , Animais , Apoptose , Ativação Enzimática , Deleção de Genes , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Quinase I-kappa B/metabolismo , MAP Quinase Quinase 4/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , eIF-2 Quinase/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Cancer Gene Ther ; 14(1): 12-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17024233

RESUMO

Our previous studies demonstrated that adenovirus-mediated overexpression of melanoma differentiation-associated gene-7 (Ad-mda7) leads to rapid induction of double-stranded RNA-dependent protein kinase (PKR) and activation of its downstream targets, resulting in apoptosis induction in human lung cancer cells. Here, we report that Ad-mda7 and the benzoquinone ansamycin geldanamycin (GA) interact in a highly synergistic manner to induce cell death in human lung cancer cells. Co-administration of Ad-mda7 and GA did not modify expression of MDA-7, and was not associated with further PKR induction and activation; instead the enhanced cytotoxicity of this combination was associated with inactivation of AKT by GA. By surface staining using anti-E-cadherin monoclonal antibody and flow cytometry, we found that treatment with the combination of Ad-mda7 and GA increased E-cadherin levels in these cancer cells. Ad-mda7 and GA cotreatment also inhibited lung cancer cell motility by increasing the beta-catenin/E-cadherin association. Moreover, combination of GA derivative 17-allyl-amino, 17-demethoxygeldanamycin (17AAG), with Ad-mda7 resulted in enhancement of cell death in A549 and H460 human lung cancer cells.


Assuntos
Adenoviridae/genética , Benzoquinonas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Lactamas Macrocíclicas/farmacologia , Neoplasias Pulmonares/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Benzoquinonas/química , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Imunofluorescência , Vetores Genéticos , Humanos , Lactamas Macrocíclicas/química
4.
Cancer Gene Ther ; 13(8): 806-14, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16628227

RESUMO

Adenoviral delivery of the p53 gene is a potential therapeutic approach for the treatment of lung cancer. Furthermore, amifostine is a cytoprotective agent and recent reports have described its potentiation of chemotherapy's antitumor activity in lung cancer. Therefore, we wished to investigate the ability of amifostine both alone and in combination with p53-based therapy to induce apoptosis, and to understand the mechanisms by which this apoptosis occurs. Using p53 null and wild-type p53 human lung cancer cells and normal human bronchial epithelial cells, we evaluated the effects of amifostine on proliferation and apoptosis. We then analyzed Adp53 in combination with amifostine and performed isobologram analysis. Expression of p53, p21(WAF1), Bax, Bak, bcl-2, as well as total and phosphorylated Cdc2 in the absence and presence of olomoucine, a phosphorylated Cdc2 kinase inhibitor, was then determined. Amifostine-induced apoptosis in human lung cancer cells in a dose-dependent fashion. The combination of amifostine and Adp53 significantly enhanced, with a supra-additive effect, the inhibition of proliferation of lung cancer cells. This enhancement of apoptosis by amifostine was associated with activation of p53 and dephosphorylation of Cdc2 proteins. Notably, olomoucine effectively prevented amifostine and/or Adp53-induced Cdc2 kinase activation and subsequent apoptosis. Our data shows that amifostine alone can induce apoptosis of human lung cancer cells, and that the combination of Adp53 with amifostine resulted in significantly higher levels of apoptosis. In addition, it appears that Cdc2 kinase plays an important role in the induction of apoptosis by amifostine and Adp53.


Assuntos
Amifostina/uso terapêutico , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Genes p53/genética , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , Adenoviridae , Apoptose/genética , Proteína Quinase CDC2/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Inibidor de Quinase Dependente de Ciclina p21/genética , Expressão Gênica , Genes bcl-2/genética , Vetores Genéticos , Humanos , Neoplasias Pulmonares/genética , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína X Associada a bcl-2/genética
5.
Cancer Gene Ther ; 13(5): 490-502, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16282987

RESUMO

Current therapies used in the treatment of breast cancer are limited by systemic toxicity, rapid drug metabolism and intrinsic and acquired drug resistance. We have previously shown that adenoviral-mediated transfer of the melanoma differentiation-associated gene-7 (mda-7) elicits growth inhibition and apoptosis in various tumor types. Here, we evaluate the effects of Ad-mda7, alone and in combination with other therapies, against a panel of nine breast tumor cell lines and their normal counterparts; we report selective Ad-mda7-mediated p53-independent growth inhibition, G2/M cell cycle arrest, and apoptosis. In vivo, Ad-mda7 induced p53-independent tumor growth inhibition (P<0.004) in multiple xenograft models. We then evaluated the combination of Ad-mda7 with agents commonly used to treat breast cancer: radiotherapy (XRT), Tamoxifen, Taxotere, Adriamycin, and Herceptin. These agents exhibit diverse modes of action, including formation of bulky adducts, inhibition of DNA replication (Adriamycin, XRT), damage to microtubules (Taxotere), nonsteroidal estrogen antagonists (Tamoxifen), or Her2/neu receptor blockade (Herceptin). Treated with conventional anticancer drugs or radiation, MDA-7-expressing cells display additive or synergistic cytotoxicity and apoptosis that correlates with decreased BCL-2 expression and BAX upregulation. In vivo, animals that received Ad-mda7 and XRT underwent significant reduction of tumor growth (P<0.002). This is the first report of the synergistic effects of Ad-mda7 combined with chemotherapy or radiotherapy on human breast carcinoma cells.


Assuntos
Neoplasias da Mama/terapia , Carcinoma/terapia , Terapia Genética , Interleucinas/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Adenoviridae/genética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose , Terapia Biológica , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Carcinoma/tratamento farmacológico , Carcinoma/radioterapia , Terapia Combinada , Feminino , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-bcl-2/análise
6.
J Thorac Cardiovasc Surg ; 121(1): 61-7, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11135160

RESUMO

OBJECTIVE: Conventional treatment for mesothelioma is largely ineffective. We therefore evaluated the novel approach of adenoviral gene transfer of the proapoptotic Bcl-2 family member Bak in mesothelioma cancer cell lines, which are sensitive and resistant to adenoviral p53. METHODS: Binary adenoviral Bak (Ad/GT-Bak and Ad/GV16) and LacZ (Ad/GT-LacZ and Ad/GV16) vectors were used for transduction of the mesothelioma cell lines I-45 (p53 resistant) and REN (p53 sensitive). Protein levels were determined by Western blotting. Apoptosis was assessed by morphologic changes, caspase-3 cleavage, and fluorescence-activated cell sorter analysis of subdiploid populations. Cell viability was determined with the XTT assay. Statistical analysis was performed with analysis of variance and the Student t test. RESULTS: High levels of Bak gene transfer were seen after coadministration of Ad/GT-Bak and Ad/GV16 in both mesothelioma cell lines. Apoptosis was induced 24 hours after Bak but not LacZ gene transfer ([Bak: I-45, 36%; REN, 25%] vs [LacZ: I-45, 1%; REN, 3%], P <.05]) in p53-sensitive (REN) and p53-resistant (I-45) cell lines. Cellular viability was significantly decreased 48 to 72 hours after Bak gene transfer compared with control vector in both cell lines (72 hours: Bak I-45, 1.4% +/- 1.0%, and Bak REN, 4.7% +/- 1%, vs Lac-Z I-45, 83% +/- 3%, and Lac-Z REN, 100% +/- 1%; P <.05). CONCLUSIONS: Adenovirus-mediated overexpression of the Bak gene induces apoptosis and decreased cellular viability in p53-sensitive and p53-resistant mesothelioma cells. These data suggest that the gene transfer of proapoptotic Bcl-2 family members may represent a novel gene therapy strategy to treat mesothelioma.


Assuntos
Adenoviridae/genética , Apoptose , Proteínas de Membrana/genética , Mesotelioma/patologia , Neoplasias Pleurais/patologia , Transdução Genética/métodos , Apoptose/genética , Western Blotting , Sobrevivência Celular , Citometria de Fluxo , Expressão Gênica , Vetores Genéticos , Humanos , Proteínas de Membrana/metabolismo , Mesotelioma/genética , Mesotelioma/virologia , Mutação , Neoplasias Pleurais/genética , Neoplasias Pleurais/virologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Killer-Antagonista Homóloga a bcl-2
7.
Cancer Res ; 60(4): 788-92, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10706081

RESUMO

One of the most promising strategies in cancer gene therapy is adenoviral transfer of proapoptotic genes. We therefore evaluated the novel strategy of adenovirally overexpressing the proapoptotic Bak gene. Our results showed marked apoptosis in cancer cells in vivo and in vitro after Bak gene transfer via a binary adenoviral vector system. This effect was not seen in a caspase 3-defective cell line (MCF-7) and was abrogated in Bak-sensitive tumors after administration of the caspase inhibitor z-DEVD-fmk. Our results suggest that adenoviral-mediated overexpression of Bak provides a novel therapeutic strategy for cancer therapy, but this process appears to be caspase dependent.


Assuntos
Adenoviridae/genética , Caspases/fisiologia , Terapia Genética , Proteínas de Membrana/genética , Neoplasias/terapia , Animais , Apoptose , Feminino , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Células Tumorais Cultivadas , Proteína Killer-Antagonista Homóloga a bcl-2
8.
Cancer Res ; 57(14): 2904-8, 1997 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9230199

RESUMO

Development of pulmonary adenomas (PAs) in mice is under the genetic control of multiple host genes. We have established a new set of SMXA recombinant inbred strains from PA-susceptible A/J and PA-resistant SM/J mice. The number of urethan-induced PAs was variable among substrains of the SMXA recombinant inbred strains, indicating the involvement of multiple genes. SMXA24 mice were highly resistant to PA, although they had susceptible alleles at all four known susceptibility genes, including kras2 and MHC. To identify the resistance gene in SMXA24, progeny of reciprocal F1 crosses and progeny of backcrosses to A/J were given urethan at 4 weeks of age and examined for induced PA at the age of 5 months. In reciprocal F1 cross progeny, the incidence of PA was very low, indicating that the resistance was a semidominant trait. Quantitative trait analysis of the backcross generation revealed significant linkages to loci on chromosome 12 (logarithm of odds score, 6.47) and chromosome 11 (logarithm of odds score, 4.35). To date, two PA resistance (PAR) genes, Par1 (located on chromosome 11) and Par2 (located on chromosome 18), have been reported. From the map position, one of the resistance genes on chromosome 11 was indistinguishable from Par1. However, another resistance gene on chromosome 12 was new, and we named this gene Par3. A likely candidate gene for Par3 is nPKCn, which is expressed exclusively in skin and lung and is down-regulated in PA. Par1 and Par3 seemed to act synergistically.


Assuntos
Adenoma/genética , Neoplasias Pulmonares/genética , Uretana/toxicidade , Adenoma/induzido quimicamente , Animais , Neoplasias Pulmonares/induzido quimicamente , Camundongos , Camundongos Endogâmicos , Proteína Quinase C/fisiologia , Recombinação Genética
9.
Leuk Res ; 21(4): 337-42, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9150351

RESUMO

The pre-B lymphoma-prone inbred strain SL/Kh mice showed a polyclonal expansion of BP-1+ pre-B cells in bone marrow early in life. Preneoplastic pre-B cells did not express adhesion molecules LECAM-1 and LFA-1, whereas neoplastic pre-B cells consistently expressed both molecules. There were two types of pre-B lymphomas in SL/Kh with distinct in vivo behavior. One infiltrated lymph nodes and spleen and another, predominantly bone marrow. However, lymphoma cells of both types expressed BP-1, LECAM-1 and LFA-1. Expression of these adhesion molecules on BP-1+ cells, therefore, may represent an important consequence of pre-B lymphomagenesis in SL/Kh strain, but is not sufficient to explain the in vivo behavior of the pre-B lymphoma cells.


Assuntos
Linfócitos B/metabolismo , Selectina L/biossíntese , Antígeno-1 Associado à Função Linfocitária/biossíntese , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Animais , Linfócitos B/patologia , Adesão Celular , Diferenciação Celular , Transplante de Células , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia
10.
Cancer Res ; 56(16): 3716-20, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8706013

RESUMO

To explore possible host genes suppressing spontaneous B-lymphomagenesis in the mouse, expression of ecotropic murine leukemia virus (E-MuLV) and lymphoma development were observed in crosses between the pre-B lymphoma-prone SL/Kh and low-lymphoma strains of mice. E-MuLV expression was intensely inhibited in F1 hybrids with the strains either with the Fv-1b allele (BALB/C, C57BL/10, and A/J) or with the Fv-1nr allele (NZB). In these F1 mice, no lymphoma developed by 18 months of age. On the other hand, F1 hybrids with the strains with the Fv-1n allele [C3H/He, CBA/N, SJL, DBA/2, and MSM/Ms (hereafter referred to as MSM)], high or intermediate levels of E-MuLV expression were observed. Lymphoma incidence in these F1 hybrids, however, was low. This observation suggests the presence of non-Fv-1 dominant resistance genes in these strains. In an attempt to characterize such host genes, we analyzed crosses between SL/Kh mice and a wild mouse-derived inbred strain, MSM/Ms. The latter was susceptible to N-tropic virus expression, but (SL/Kh x MSM)F1 hybrids, did not develop and lymphomas. Of 60 SL/Kh x (SL/Kh x MSM)F1 hybrids, 14 B-lineage lymphomas, including 13 pre-B and 1 follicular center cell lymphoma, developed by 18 months of age. This was compatible with the hypothesis of two independently segregating dominant genes of MSM suppressing lymphomagenesis. By scanning all chromosomes for linkage of lymphoma susceptibility with polymorphic microsatellite loci, one significant linkage disequilibrium was found in the proximal segment of chromosome 17, containing D17MIT44 (map position 15.0) to D17MIT150 (position 33.3), and another linkage disequilibrium, in the midproximal segment of chromosome 18, containing D18MIT90 (map position 28.0) and D18MIT140 (37.0). All 13 pre-B lymphoma-bearing backcross mice were homozygous for SL/Kh-derived alleles at these loci. We named the gene on chromosome 17 Msmr1 (for MSM resistance 1) and that on chromosome 18 Msmr 2 (for MSM resistance 2).


Assuntos
Genes Dominantes , Linfoma de Células B/genética , Animais , Mapeamento Cromossômico , Feminino , Vírus da Leucemia Murina/isolamento & purificação , Linfoma de Células B/imunologia , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos
11.
Jpn J Cancer Res ; 87(4): 401-4, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8641972

RESUMO

The effects of two host genes on retrovirus-induced murine lymphoma were evaluated by studying 114 F2 intercross mice between SL/Kh and AKR/Ms mice. Out of 47 T-lymphoma-bearing F2 mice, 45 had the AKR-derived dominant allele at Tism-1. The length of the lymphoma latent period was not related to type of tumor. Instead, it was significantly shortened by a recessive SL/Kh-derived allele at a major histocompatibility complex (MHC)-linked locus on Chr. 17. A quantitative trait analysis of the latent period yielded a maximal logarithm of likelihood ratio for linkage (LOD) score of 7.06 at a class II gene within MHC. The SL/Kh-derived recessive gene was named lla (lymphoma latency acceleration).


Assuntos
Antígenos de Histocompatibilidade Classe II/genética , Linfoma de Células B/genética , Linfoma de Células B/imunologia , Linfoma de Células T/genética , Linfoma de Células T/imunologia , Alelos , Animais , DNA Satélite/genética , Ligação Genética , Genótipo , Camundongos , Camundongos Endogâmicos , Polimorfismo Genético
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...