Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
DNA Cell Biol ; 42(6): 274-288, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36576491

RESUMO

Together with an anti-tumor immune response, oncolysis using a recombinant viral vector promises to eliminate cancer cells by both gene transfer and host-mediated functions. In this study we explore oncolysis induced by nonreplicating adenoviral vectors used for p14ARF and interferon-ß (hIFNß) gene transfer in human melanoma cell lines, revealing an unexpected role for p14ARF in promoting cellular responses predictive of immune stimulation. Oncolysis was confirmed when UACC-62 (p53 wild-type) cells succumbed upon p14ARF gene transfer in vitro, whereas SK-Mel-29 (p53-mutant) benefitted from its combination with hIFNß. In the case of UACC-62, in situ gene therapy in nude mice yielded reduced tumor progression in response to the p14ARF and hIFNß combination. Potential for immune stimulation was revealed where p14ARF gene transfer in vitro was sufficient to induce emission of immunogenic cell death factors in UACC-62 and upregulate pro-immune genes, including IRF1, IRF7, IRF9, ISG15, TAP-1, and B2M. In SK-Mel-29, p14ARF gene transfer induced a subset of these factors. hIFNß was, as expected, sufficient to induce these immune-stimulating genes in both cell lines. This work is a significant advancement for our melanoma gene therapy strategy because we revealed not only the induction of oncolysis, but also the potential contribution of p14ARF to immune stimulation.


Assuntos
Melanoma , Proteína Supressora de Tumor p14ARF , Camundongos , Animais , Humanos , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/genética , Camundongos Nus , Apoptose/fisiologia , Linhagem Celular , Melanoma/genética , Melanoma/terapia
2.
Gene Ther ; 27(1-2): 15-26, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-30926960

RESUMO

For patients with metastatic prostate cancer, the 5-year survival rate of 31% points to a need for novel therapies and improvement of existing modalities. We propose that p53 gene therapy and chemotherapy, when combined, will provide superior tumor cell killing for the treatment of prostate carcinoma. To this end, we have developed the AdRGD-PGp53 vector which offers autoregulated expression of p53, resulting in enhanced tumor cell killing in vitro and in vivo. Here, we combined AdRGD-PGp53 along with the chemotherapy drugs used in the clinical treatment of prostate carcinoma, mitoxantrone, docetaxel, or cabazitaxel. Our results indicate that all drugs increase phosphorylation of p53, leading to improved induction of p53 targets. In vitro experiments reveal that AdRGD-PGp53 sensitizes prostate cancer cells to each of the drugs tested, conferring increased levels of cell death. In a xenograft mouse model of in situ gene therapy, AdRGD-PGp53 treatment, when combined with cabazitaxel, drastically reduced tumor progression and increased survival rates to 100%. Strikingly, we used a sub-therapeutic dose of cabazitaxel thus avoiding leukopenia, yet still showed potent anti-tumor effects when combined with AdRGD-PGp53 in this mouse model. The AdRGD-PGp53 approach warrants further development for its application in gene therapy of prostate carcinoma.


Assuntos
Genes p53/genética , Neoplasias da Próstata/terapia , Taxoides/farmacologia , Animais , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Intervalo Livre de Doença , Quimioterapia Combinada/métodos , Regulação Neoplásica da Expressão Gênica/genética , Genes p53/imunologia , Terapia Genética/métodos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Taxoides/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
Gene Ther ; 27(1-2): 51-61, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31439890

RESUMO

Cancer therapies that target a single protein or pathway may be limited by their specificity, thus missing key players that control cellular proliferation and contributing to the failure of the treatment. We propose that approaches to cancer therapy that hit multiple targets would limit the chances of escape. To this end, we have developed a bicistronic adenoviral vector encoding both the CDKN2A and p53 tumor suppressor genes. The bicistronic vector, AdCDKN2A-I-p53, supports the translation of both gene products from a single transcript, assuring that all transduced cells will express both proteins. We show that combined, but not single, gene transfer results in markedly reduced proliferation and increased cell death correlated with reduced levels of phosphorylated pRB, induction of CDKN1A and caspase 3 activity, yet avoiding the induction of senescence. Using isogenic cell lines, we show that these effects were not impeded by the presence of mutant p53. In a mouse model of in situ gene therapy, a single intratumoral treatment with the bicistronic vector conferred markedly inhibited tumor progression while the treatment with either CDKN2A or p53 alone only partially controlled tumor growth. Histologic analysis revealed widespread transduction, yet reduced proliferation and increased cell death was associated only with the simultaneous transfer of CDKN2A and p53. We propose that restoration of two of the most frequently altered genes in human cancer, mediated by AdCDKN2A-I-p53, is beneficial since multiple targets are reached, thus increasing the efficacy of the treatment.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/administração & dosagem , Inibidor p16 de Quinase Dependente de Ciclina/genética , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , Proteína Supressora de Tumor p53/administração & dosagem , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Feminino , Genes p53 , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução Genética/métodos , Proteína Supressora de Tumor p53/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Hum Gene Ther ; 28(8): 639-653, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28181816

RESUMO

Previously, the authors developed an adenoviral vector, Ad-PG, where transgene expression is regulated by a p53-responsive promoter. When used to transfer the p53 cDNA, a positive feedback mechanism is established. In the present study, a critical comparison is performed between Ad-PGp53 and AdRGD-PGp53, where the RGD motif was incorporated in the adenoviral fiber protein. AdRGD-PGp53 provided superior transgene expression levels and resulted in the killing of prostate carcinoma cell lines DU145 and PC3. In vitro, this effect was associated with increased production of cytoplasmic and mitochondrial oxidants, DNA damage as revealed by detection of phosphorylated H2AX, as well as cell death consistent with apoptosis. Differential gene expression of key mediators of reactive oxygen species pathways was also observed. Specifically, it was noted that induction of known p53-target genes Sestrin2 and PIG3, as well as a novel target, NOX1, occurred in PC3 cells only when transduced with the improved vector, AdRGD-PGp53. The participation of NOX1 was confirmed upon its inhibition using a specific peptide, resulting in reduced cell death. In situ gene therapy also resulted in significantly improved inhibition of tumor progression consistent with oxidant-induced DNA damage only when treated with the novel AdRGD-PGp53 vector. The study shows that the improved adenovirus overcomes limitations associated with other p53-expressing vectors and induces oxidant-mediating killing, thus supporting its further development for cancer gene therapy.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Oxidantes/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Dano ao DNA , Modelos Animais de Doenças , Expressão Gênica , Genes Reporter , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Masculino , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Espécies Reativas de Oxigênio/metabolismo , Transdução Genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Transl Oncol ; 9(6): 565-574, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27916291

RESUMO

Therapeutic strategies that act by eliciting and enhancing antitumor immunity have been clinically validated as an effective treatment modality but may benefit from the induction of both cell death and immune activation as primary stimuli. Using our AdRGD-PG adenovector platform, we show here for the first time that in situ gene transfer of p19Arf and interferon-ß (IFNß) in the LLC1 mouse model of lung carcinoma acts as an immunotherapy. Although p19Arf is sufficient to induce cell death, only its pairing with IFNß significantly induced markers of immunogenic cell death. In situ gene therapy with IFNß, either alone or in combination with p19Arf, could retard tumor progression, but only the combined treatment was associated with a protective immune response. Specifically in the case of combined intratumoral gene transfer, we identified 167 differentially expressed genes when using microarray to evaluate tumors that were treated in vivo and confirmed the activation of CCL3, CXCL3, IL1α, IL1ß, CD274, and OSM, involved in immune response and chemotaxis. Histologic evaluation revealed significant tumor infiltration by neutrophils, whereas functional depletion of granulocytes ablated the antitumor effect of our approach. The association of in situ gene therapy with cisplatin resulted in synergistic elimination of tumor progression. In all, in situ gene transfer with p19Arf and IFNß acts as an immunotherapy involving recruitment of neutrophils, a desirable but previously untested outcome, and this approach may be allied with chemotherapy, thus providing significant antitumor activity and warranting further development for the treatment of lung carcinoma.

7.
Cancer Biol Ther ; 17(12): 1221-1230, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27646031

RESUMO

Alternative treatments for cancer using gene therapy approaches have shown promising results and some have even reached the marketplace. Even so, additional improvements are needed, such as employing a strategically chosen promoter to drive expression of the transgene in the target cell. Previously, we described viral vectors where high-level transgene expression was achieved using a p53-responsive promoter. Here we present an adenoviral vector (AdPGp53) where p53 is employed to regulate its own expression and which outperforms a traditional vector when tested in a model of gene therapy for prostate cancer. The functionality of AdPGp53 and AdCMVp53 were compared in human prostate carcinoma cell lines. AdPGp53 conferred greatly enhanced levels of p53 protein and induction of the p53 target gene, p21, as well as superior cell killing by a mechanism consistent with apoptosis. DU145 cells were susceptible to induction of death with AdPGp53, yet PC3 cells were quite resistant. Though AdCMVp53 was shown to be reliable, extremely high-level expression of p53 offered by AdPGp53 was necessary for tumor suppressor activity in PC3 and DU145. In situ gene therapy experiments revealed tumor inhibition and increased overall survival in response to AdPGp53, but not AdCMVp53. Upon histologic examination, only AdPGp53 treatment was correlated with the detection of both p53 and TUNEL-positive cells. This study points to the importance of improved vector performance for gene therapy of prostate cancer.


Assuntos
Adenoviridae/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Vetores Genéticos/administração & dosagem , Neoplasias da Próstata/terapia , Proteína Supressora de Tumor p53/genética , Animais , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Terapia Genética , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Regiões Promotoras Genéticas , Próstata , Neoplasias da Próstata/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Immunol Immunother ; 65(4): 371-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26887933

RESUMO

Previously, we combined p19(Arf) (Cdkn2a, tumor suppressor protein) and interferon beta (IFN-ß, immunomodulatory cytokine) gene transfer in order to enhance cell death in a murine model of melanoma. Here, we present evidence of the immune response induced when B16 cells succumbing to death due to treatment with p19(Arf) and IFN-ß are applied in vaccine models. Use of dying cells for prophylactic vaccination was investigated, identifying conditions for tumor-free survival. After combined p19(Arf) and IFN-ß treatment, we observed immune rejection at the vaccine site in immune competent and nude mice with normal NK activity, but not in NOD-SCID and dexamethasone immunosuppressed mice (NK deficient). Combined treatment induced IL-15, ULBP1, FAS/APO1 and KILLER/DR5 expression, providing a mechanism for NK activation. Prophylactic vaccination protected against tumor challenge, where markedly delayed progression and leukocyte infiltration were observed. Analysis of primed lymphocytes revealed secretion of TH1-related cytokines and depletion protocols showed that both CD4(+) and CD8(+) T lymphocytes are necessary for immune protection. However, application of this prophylactic vaccine where cells were treated either with IFN-ß alone or combined with p19(Arf) conferred similar immune protection and cytokine activation, yet only the combination was associated with increased overall survival. In a therapeutic vaccine protocol, only the combination was associated with reduced tumor progression. Our results indicate that by harnessing cell death in an immunogenic context, our p19(Arf) and IFN-ß combination offers a clear advantage when both genes are included in the vaccine and warrants further development as a novel immunotherapy for melanoma.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/imunologia , Interferon beta/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Vacinação/métodos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Terapia Genética/métodos , Imunoterapia/métodos , Interferon beta/genética , Interleucina-15/imunologia , Interleucina-15/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Melanoma Experimental/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Carga Tumoral/genética , Carga Tumoral/imunologia
9.
Circ Res ; 107(2): 204-16, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20522805

RESUMO

RATIONALE: Major coronary vessels derive from the proepicardium, the cellular progenitor of the epicardium, coronary endothelium, and coronary smooth muscle cells (CoSMCs). CoSMCs are delayed in their differentiation relative to coronary endothelial cells (CoEs), such that CoSMCs mature only after CoEs have assembled into tubes. The mechanisms underlying this sequential CoE/CoSMC differentiation are unknown. Retinoic acid (RA) is crucial for vascular development and the main RA-synthesizing enzyme is progressively lost from epicardially derived cells as they differentiate into blood vessel types. In parallel, myocardial vascular endothelial growth factor (VEGF) expression also decreases along coronary vessel muscularization. OBJECTIVE: We hypothesized that RA and VEGF act coordinately as physiological brakes to CoSMC differentiation. METHODS AND RESULTS: In vitro assays (proepicardial cultures, cocultures, and RALDH2 [retinaldehyde dehydrogenase-2]/VEGF adenoviral overexpression) and in vivo inhibition of RA synthesis show that RA and VEGF act as repressors of CoSMC differentiation, whereas VEGF biases epicardially derived cell differentiation toward the endothelial phenotype. CONCLUSION: Experiments support a model in which early high levels of RA and VEGF prevent CoSMC differentiation from epicardially derived cells before RA and VEGF levels decline as an extensive endothelial network is established. We suggest this physiological delay guarantees the formation of a complex, hierarchical, tree of coronary vessels.


Assuntos
Diferenciação Celular , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Miócitos de Músculo Liso/metabolismo , Pericárdio/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Apoptose , Comunicação Autócrina , Diferenciação Celular/genética , Células Cultivadas , Embrião de Galinha , Técnicas de Cocultura , Vasos Coronários/embriologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Morfogênese , Miócitos Cardíacos/metabolismo , Comunicação Parácrina , Pericárdio/embriologia , Codorniz , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/genética , Técnicas de Cultura de Tecidos , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/genética
10.
BMC Cancer ; 10: 316, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20569441

RESUMO

BACKGROUND: Reactivation of p53 by either gene transfer or pharmacologic approaches may compensate for loss of p19Arf or excess mdm2 expression, common events in melanoma and glioma. In our previous work, we constructed the pCLPG retroviral vector where transgene expression is controlled by p53 through a p53-responsive promoter. The use of this vector to introduce p19Arf into tumor cells that harbor p53wt should yield viral expression of p19Arf which, in turn, would activate the endogenous p53 and result in enhanced vector expression and tumor suppression. Since nutlin-3 can activate p53 by blocking its interaction with mdm2, we explored the possibility that the combination of p19Arf gene transfer and nutlin-3 drug treatment may provide an additive benefit in stimulating p53 function. METHODS: B16 (mouse melanoma) and C6 (rat glioma) cell lines, which harbor p53wt, were transduced with pCLPGp19 and these were additionally treated with nutlin-3 or the DNA damaging agent, doxorubicin. Viral expression was confirmed by Western, Northern and immunofluorescence assays. p53 function was assessed by reporter gene activity provided by a p53-responsive construct. Alterations in proliferation and viability were measured by colony formation, growth curve, cell cycle and MTT assays. In an animal model, B16 cells were treated with the pCLPGp19 virus and/or drugs before subcutaneous injection in C57BL/6 mice, observation of tumor progression and histopathologic analyses. RESULTS: Here we show that the functional activation of endogenous p53wt in B16 was particularly challenging, but accomplished when combined gene transfer and drug treatments were applied, resulting in increased transactivation by p53, marked cell cycle alteration and reduced viability in culture. In an animal model, B16 cells treated with both p19Arf and nutlin-3 yielded increased necrosis and decreased BrdU marking. In comparison, C6 cells were quite susceptible to either treatment, yet p53 was further activated by the combination of p19Arf and nutlin-3. CONCLUSIONS: To the best of our knowledge, this is the first study to apply both p19Arf and nutlin-3 for the stimulation of p53 activity. These results support the notion that a p53 responsive vector may prove to be an interesting gene transfer tool, especially when combined with p53-activating agents, for the treatment of tumors that retain wild-type p53.


Assuntos
Antineoplásicos/farmacologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Terapia Genética , Glioma/terapia , Imidazóis/farmacologia , Melanoma Experimental/terapia , Piperazinas/farmacologia , Transdução Genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Terapia Combinada , Inibidor p16 de Quinase Dependente de Ciclina/genética , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Imunofluorescência , Vetores Genéticos , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Retroviridae/genética , Fatores de Tempo , Ativação Transcricional , Transfecção , Carga Tumoral , Proteína Supressora de Tumor p53/genética
11.
Cancer Gene Ther ; 12(12): 935-46, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15905860

RESUMO

Typically, gene transfer strategies utilize a promoter/transgene arrangement that treat these elements independently and do not offer any interplay between them. Our goal was to establish a promoter/transgene combination that would result in improvement in both expression and therapeutic effect by utilizing the transcriptional properties of p53 to drive its own expression as well as act as a tumor suppressor. The pCL retroviral system was modified in the U3 region of the 3' LTR by the addition of a p53-responsive sequence (the PG element), creating the pCLPG system. Upon reverse transcription, the 5' LTR is converted, as shown here, to a p53-dependent promoter. We also show, using a temperature-sensitive model, that the pCLPG system could be driven by p53 encoded within the virus construct and expression was modulated depending on the p53 phenotype, demonstrating a regulatory feedback loop. Moreover, the pCLPG system was shown to express the transgene at a higher level and to inhibit tumor cell proliferation more robustly than the original pCL system. This novel system employs the transgene to serve two purposes, drive viral expression and inhibit tumor cell proliferation. The pCLPG vectors represent a new gene transfer strategy of synergizing the promoter and transgene activities.


Assuntos
Técnicas de Transferência de Genes , Neoplasias/genética , Neoplasias/patologia , Regiões Promotoras Genéticas/genética , Transgenes/genética , Linhagem Celular , Proliferação de Células , Humanos , Plasmídeos/genética , Retroviridae/genética , Temperatura , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Genome Res ; 14(7): 1413-23, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15197164

RESUMO

We report the results of a transcript finishing initiative, undertaken for the purpose of identifying and characterizing novel human transcripts, in which RT-PCR was used to bridge gaps between paired EST clusters, mapped against the genomic sequence. Each pair of EST clusters selected for experimental validation was designated a transcript finishing unit (TFU). A total of 489 TFUs were selected for validation, and an overall efficiency of 43.1% was achieved. We generated a total of 59,975 bp of transcribed sequences organized into 432 exons, contributing to the definition of the structure of 211 human transcripts. The structure of several transcripts reported here was confirmed during the course of this project, through the generation of their corresponding full-length cDNA sequences. Nevertheless, for 21% of the validated TFUs, a full-length cDNA sequence is not yet available in public databases, and the structure of 69.2% of these TFUs was not correctly predicted by computer programs. The TF strategy provides a significant contribution to the definition of the complete catalog of human genes and transcripts, because it appears to be particularly useful for identification of low abundance transcripts expressed in a restricted set of tissues as well as for the delineation of gene boundaries and alternatively spliced isoforms.


Assuntos
Software , Transcrição Gênica/genética , Processamento Alternativo/genética , Linhagem Celular , Linhagem Celular Tumoral , Biologia Computacional/métodos , Biologia Computacional/estatística & dados numéricos , Sequência Consenso/genética , DNA de Neoplasias , Bases de Dados Genéticas/classificação , Etiquetas de Sequências Expressas , Genes/genética , Genoma Humano , Células HeLa/patologia , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Design de Software , Validação de Programas de Computador , Células U937/patologia
13.
Virology ; 321(2): 165-72, 2004 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-15051377

RESUMO

We have modified the pCL retroviral system by the insertion of a p53-responsive element, called PG, in the U3 region of the 3'-LTR, either in addition to or in place of the native negative control region/enhancer sequence. We show here that either endogenous or exogenous wild-type p53 may be used to drive expression from the pCLPG system in transduced cells. Upon genotoxic induction of endogenous p53, pCLPG expression surpassed that of the parental, nonmodified virus, specifically when the native promoter was removed and substituted by the p53-responsive element. We propose that the novel pCLPG system will prove to be a valuable tool whether used as a reporter system of p53 function or as an in vitro and in vivo gene transfer vehicle.


Assuntos
Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Retroviridae/genética , Retroviridae/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Linhagem Celular , Elementos Facilitadores Genéticos , Vetores Genéticos , Humanos , Regiões Promotoras Genéticas , Sequências Repetidas Terminais , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Replicação Viral
14.
J Biotechnol ; 103(2): 97-106, 2003 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-12814868

RESUMO

The pCL system was developed to aid in the production of retrovirus that encodes cytotoxic or cytostatic cDNA's. A principal feature of this system is the transient production of virus after co-transfection of the viral and packaging vectors in the 293T cell line. This approach obviates the need for selection of the producer cells, thus minimizing potential affects of the encoded genes. However, the transient nature of this system also creates a number of experimental variables. In this study we have examined and optimized elements related to the production of the pCL retrovirus. For example, co-transfection of the packaging sequence along with the viral vector has been optimized in terms of both the total amount of DNA transfected and the relative proportion of each plasmid. We have also tested the affect of increased synthesis of viral proteins in the producer cells and the kinetics of virus accumulation in the supernatant. These findings may be of interest to those who use pCL or any transient packaging system in their gene transfer studies. In addition, these studies may aid in the validation and development of transient retrovirus production systems for clinical applications.


Assuntos
Biotecnologia/métodos , Vetores Genéticos , Retroviridae/crescimento & desenvolvimento , Retroviridae/genética , Animais , Humanos , Rim/citologia , Cinética , Camundongos , Células NIH 3T3 , Plasmídeos/genética , Transfecção , Proteínas Virais/genética , beta-Galactosidase/genética
15.
Cancer Cell Int ; 2(1): 2, 2002 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-11983028

RESUMO

BACKGROUND: The p16INK4A gene product halts cell proliferation by preventing phosphorylation of the Rb protein. The p16INK4a gene is often deleted in human glioblastoma multiforme, contributing to unchecked Rb phosphorylation and rapid cell division. We show here that transduction of the human p16INK4a cDNA using the pCL retroviral system is an efficient means of stopping the proliferation of the rat-derrived glioma cell line, C6, both in tissue culture and in an animal model. C6 cells were transduced with pCL retrovirus encoding the p16INK4a, p53, or Rb genes. These cells were analyzed by a colony formation assay. Expression of p16INK4a was confirmed by immunohistochemistry and Western blot analysis. The altered morphology of the p16-expressing cells was further characterized by the senescence-associated beta-galactosidase assay. C6 cells infected ex vivo were implanted by stereotaxic injection in order to assess tumor formation. RESULTS: The p16INK4a gene arrested C6 cells more efficiently than either p53 or Rb. Continued studies with the p16INK4a gene revealed that a large portion of infected cells expressed the p16INK4a protein and the morphology of these cells was altered. The enlarged, flat, and bi-polar shape indicated a senescence-like state, confirmed by the senescence-associated beta-galactosidase assay. The animal model revealed that cells infected with the pCLp16 virus did not form tumors. CONCLUSION: Our results show that retrovirus mediated transfer of p16INK4a halts glioma formation in a rat model. These results corroborate the idea that retrovirus-mediated transfer of the p16INK4a gene may be an effective means to arrest human glioma and glioblastoma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA