Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cancer Res ; 58(11): 2331-4, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9622068

RESUMO

Mutated in multiple advanced cancers 1/phosphatase and tensin homologue (MMAC1/PTEN) is a novel tumor suppressor gene candidate located on chromosome 10 that is commonly mutated in human glioblastoma multiforme and several other cancer types. To evaluate the function of this gene as a tumor suppressor, we constructed a replication-defective adenovirus (MMCB) for efficient, transient transduction of MMAC1 into tumor cells. Infection of MMAC1-mutated U87MG glioblastoma cells with MMCB resulted in dose-dependent exogenous MMAC1 protein expression as detected by Western blotting of cell lysates. In vitro proliferation of U87MG cells was inhibited by MMCB in comparison to several control adenoviruses at equal viral doses, implying a specific effect of MMAC1 expression. Anchorage-independent growth in soft agar was also inhibited by MMCB compared to control adenovirus. Tumorigenicity in nude mice of transiently transduced mass cell cultures was then assessed. MMCB-infected U87MG cells were almost completely nontumorigenic compared to untreated and several control adenovirus-treated cells at equal viral doses. These data support an in vivo tumor suppression activity of MMAC1/PTEN and suggest that in vivo gene transfer with this recombinant adenoviral vector has a potential use in cancer gene therapy.


Assuntos
Técnicas de Transferência de Genes , Genes Supressores de Tumor , Terapia Genética , Glioblastoma/terapia , Monoéster Fosfórico Hidrolases , Proteínas Tirosina Fosfatases/genética , Proteínas Supressoras de Tumor , Adenoviridae/genética , Animais , Cromossomos Humanos Par 10 , Citometria de Fluxo , Mutação em Linhagem Germinativa , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , PTEN Fosfo-Hidrolase , Células Tumorais Cultivadas
2.
Cancer Gene Ther ; 8(12): 982-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11781661

RESUMO

Progressive growth and metastasis of solid tumors require angiogenesis, or the formation of new blood vessels. Endostatin is a 20-kDa carboxy-terminal fragment of collagen XVIII that has been shown to inhibit endothelial cell proliferation and tumor angiogenesis. Replication-deficient recombinant adenovirus (rAd) vectors were constructed, which encoded secreted forms of human and mouse endostatin (HECB and MECB, respectively), and, as a control, human alkaline phosphatase (APCB). Accumulation of endostatin was demonstrated in supernatants of cultured cells infected with the endostatin rAds. These supernatants disrupted tubule formation, inhibited migration and proliferation, and induced apoptosis in human dermal vascular endothelial cells or human vascular endothelial cells. Endostatin-containing supernatants had no effect on the proliferation of MidT2-1 mouse mammary tumor cells in vitro. A pharmacokinetic study of MECB in immunocompetent FVB mice demonstrated a 10-fold increase of serum endostatin concentrations 3 days after intravenous administration of 1x10(10) particles of this rAd (215-257 ng/mL compared to 12-38 ng/mL in control rAd-treated mice). Intravenous administration of MECB reduced b-FGF stimulated angiogenesis into Matrigel plugs by 38%. Intratumoral MECB inhibited growth of MidT2-1 syngeneic mammary tumors in FVB mice, but had minimal impact on the growth of MDA-MB-231 human breast tumors in SCID mice. Intravenous therapy with MECB also initially inhibited growth of MidT2-1 tumors, but this activity was subsequently blocked by induced anti-rAd antibodies. In summary, endostatin gene therapy effectively suppressed angiogenic processes in vitro and in vivo in several model systems.


Assuntos
Inibidores da Angiogênese/genética , Colágeno/genética , Terapia Genética , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Fragmentos de Peptídeos/genética , Adenoviridae , Inibidores da Angiogênese/uso terapêutico , Animais , Células Cultivadas , Colágeno/uso terapêutico , Colágeno Tipo XVIII , Endostatinas , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Vetores Genéticos , Humanos , Camundongos , Fragmentos de Peptídeos/uso terapêutico , Vírus Reordenados
3.
Gene Ther ; 8(24): 1847-54, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11821938

RESUMO

The retinoblastoma protein (Rb), a key regulator of cell cycle progression, can bind the transcription factor E2F converting it from a positive transcriptional factor capable of driving cells into S phase into a negative complex which arrests cells in G1. We have created a potent transcriptional repressor of E2F-dependent transcription by fusing the C-terminal fragment of Rb (p56) to the DNA and DP1-binding domains of E2F. Because the expression of E2F/56 fusion protein from a constitutive promoter was incompatible with virus growth, adenovirus constructs were prepared where transgenes were expressed from a fragment of the smooth muscle alpha-actin (SMA) promoter. Immunoblot and beta-galactosidase staining demonstrated smooth muscle-specific expression of this transcriptional element in vitro. The SMA-p56 and SMA-E2F/p56 adenoviral constructs also induced G0/G1 cell cycle arrest specifically in smooth muscle cells. Following administration to rat tissues, the SMA-beta-galactosidase construct exhibited expression in balloon-injured carotid arteries, but not in liver, bladder or skeletal muscle. Local delivery of the SMA-E2F/p56 adenoviral construct to balloon-injured carotid arteries inhibited intimal hyperplasia. Our results demonstrate that local delivery of the SMA-E2F/p56 adenoviral construct can limit intimal hyperplasia in balloon-injured vessels, while avoiding toxicity that could occur from the dissemination and expression of the viral transgene.


Assuntos
Actinas/genética , Adenoviridae/genética , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Músculo Liso Vascular/patologia , beta-Galactosidase/genética , Angioplastia com Balão/efeitos adversos , Animais , Fusão Gênica Artificial/métodos , Artérias Carótidas/patologia , Ciclo Celular , Fatores de Transcrição E2F , Humanos , Hiperplasia/prevenção & controle , Ratos , Proteína do Retinoblastoma/genética , Fatores de Transcrição/genética , Transgenes , Túnica Íntima/patologia
4.
J Virol ; 75(18): 8733-41, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507218

RESUMO

In vitro experiments have demonstrated intercellular trafficking of the VP22 tegument protein of herpes simplex virus type 1 from infected cells to neighboring cells, which internalize VP22 and transport it to the nucleus. VP22 also can mediate intercellular transport of fusion proteins, providing a strategy for increasing the distribution of therapeutic proteins in gene therapy. Intercellular trafficking of the p53 tumor suppressor protein was demonstrated in vitro using a plasmid expressing full-length p53 fused in-frame to full-length VP22. The p53-VP22 chimeric protein induced apoptosis both in transfected tumor cells and in neighboring cells, resulting in a widespread cytotoxic effect. To evaluate the anti-tumor activity of p53-VP22 in vivo, we constructed recombinant adenoviruses expressing either wild-type p53 (FTCB) or a p53-VP22 fusion protein (FVCB) and compared their effects in p53-resistant tumor cells. In vitro, treatment of tumor cells with FVCB resulted in enhanced p53-specific apoptosis compared to treatment with equivalent doses of FTCB. However, in normal cells there was no difference in the dose-related cytotoxicity of FVCB compared to that of FTCB. In vivo, treatment of established tumors with FVCB was more effective than equivalent doses of FTCB. The dose-response curve to FVCB was flatter than that to FTCB; maximal antitumor responses could be achieved using FVCB at doses 1 log lower than those obtained with FTCB. Increased antitumor efficacy was correlated with increased distribution of p53 protein in FVCB-treated tumors. This study is the first demonstration that VP22 can enhance the in vivo distribution of therapeutic proteins and improve efficacy in gene therapy.


Assuntos
Adenovírus Humanos , Vetores Genéticos , Herpesvirus Humano 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Apoptose , Células COS , Caspase 9 , Caspases/metabolismo , Chlorocebus aethiops , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ativação Enzimática , Expressão Gênica , Humanos , Neoplasias Experimentais/fisiopatologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteínas Estruturais Virais/genética
5.
Mol Ther ; 4(1): 5-12, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11472100

RESUMO

TP53 is the most commonly altered tumor-suppressor gene in cancer and is currently being tested in Phase II/III gene replacement trials. Many tumors contain wild-type TP53 sequence with elevated MDM2 protein levels, targeting p53 for degradation. These tumors are more refractory to treatment with exogenous wild-type p53. Here we generate a recombinant adenovirus expressing a p53 variant, rAd-p53 (d 13-19), that is deleted for the amino acid sequence necessary for MDM2 binding (amino acids 13-19). We compared the apoptotic activity of rAd-p53 (d 13-19) with that of a recombinant adenovirus expressing wild-type p53 (rAd-p53) in cell lines that differ in endogenous p53 status. rAd-p53 (d 13-19) caused higher levels of apoptosis in p53 wild-type tumor lines compared with wild-type p53 treatment, as measured by annexin V-FITC staining. In p53-altered tumor lines, rAd-p53 (d 13-19) showed apoptotic activity similar to that seen with wild-type p53 treatment. In normal cells, no increase in cytopathicity was detected with rAd-p53 (d 13-19) compared with wild-type p53 treatment. This variant protein displayed synergy with chemotherapeutic agents to inhibit proliferation of ovarian and breast cell lines. The p53 variant showed greater antitumor activity in an established p53 wild-type tumor compared with treatment with wild-type p53. The p53 variant represents a means of expanding TP53 gene therapy to tumors that are resistant to p53 treatment due to the cellular responses to wild-type p53.


Assuntos
Apoptose , Neoplasias da Mama/terapia , Genes p53 , Neoplasias Ovarianas/terapia , Proteína Supressora de Tumor p53/genética , Adenoviridae/genética , Animais , Caspase 9 , Caspases/metabolismo , Divisão Celular/fisiologia , Cisplatino/administração & dosagem , Terapia Combinada , Feminino , Variação Genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/administração & dosagem , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA