Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Gene Ther ; 8(10): 788-95, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11687902

RESUMEN

A recombinant adenovirus expressing human interferon alpha2b driven by the cytomegalovirus promoter, IACB, was shown to produce and secrete biologically active protein in vitro and in vivo. Intravenous administration of IACB in Buffalo rats resulted in circulating levels of biologically active human interferon at 70,000 international units/mL for up to 15 days. Distribution of interferon protein after IACB administration was different from that seen with the subcutaneous delivery of interferon protein. Higher levels of interferon protein were observed in liver and spleen after IACB delivery compared to protein delivery. The antitumor efficacy of IACB, as measured by suppression of tumor growth, was tested in athymic nude mice bearing established human tumor xenografts from different types of human cancer. Subcutaneous tumors most responsive to the intratumoral administration of IACB ranked as U87MG (glioblastoma) and K562 (chronic myelogenous leukemia), followed by Hep 3B (hepatocellular carcinoma) and LN229 cells (glioblastoma). Intravenous administration of IACB in animals bearing U87MG or Hep 3B xenografts was also effective in suppressing tumor growth, although to a lesser extent than the intratumoral administration. IACB was also tested in a metastatic model in beige/SCID mice generated with H69 (small cell lung carcinoma) cells and was found to prolong survival in tumor-bearing animals. This suggested that interferon gene delivery can be effective in suppressing tumor growth in a wide variety of cells.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos , Interferón-alfa/genética , Neoplasias Experimentales/terapia , Animales , Western Blotting , División Celular , Citomegalovirus/genética , Cartilla de ADN/química , Femenino , Humanos , Interferón alfa-2 , Interferón-alfa/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neoplasias Experimentales/mortalidad , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Trasplante Heterólogo , Células Tumorales Cultivadas
2.
J Virol ; 75(18): 8733-41, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11507218

RESUMEN

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.


Asunto(s)
Adenovirus Humanos , Vectores Genéticos , Herpesvirus Humano 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Apoptosis , Células COS , Caspasa 9 , Caspasas/metabolismo , Chlorocebus aethiops , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Activación Enzimática , Expresión Génica , Humanos , Neoplasias Experimentales/fisiopatología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteínas Estructurales Virales/genética
3.
J Interferon Cytokine Res ; 21(6): 399-408, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11440637

RESUMEN

Replication-deficient adenoviruses expressing human interferon-alpha2b (HuIFN-alpha2b) or the hybrid IFN-alpha2alpha1 or those with the secretory signal deleted, whose express is driven by the alpha-fetoprotein (AFP) promoter, were constructed and characterized. Synthesis of IFN protein and secretion or intracellular retention were tested by Western blotting and immunoassay. Expression of IFN by the recombinant adenoviruses was restricted to cells that constitutively express AFP. In these cells, expression of both secreted and nonsecreted recombinant IFN resulted in inhibition of cell proliferation, resistance to viral infection, induction of major histocompatibility complex (MHC) class I expression, increased apoptosis, and activation of an IFN-stimulated response element (ISRE)-containing promoter. Also, the induction of protein kinase R (PKR), increased phosphorylation of Stat1, and accumulation of hypophosphorylated pRb were observed for both the secreted and nonsecreted IFN, suggesting that the nonsecreted IFN may act through a similar pathway. Hep3B cells, an AFP-positive line derived from a patient with hepatocellular carcinoma (HCC), were injected subcutaneously (s.c.) into athymic nude mice to generate established tumors. Intratumoral injection of recombinant adenoviruses expressing secreted as well as the nonsecreted IFN caused suppression of tumor growth. As the AFP promoter is activated in many HCC cells but is silent in normal cells, these constructs may be useful in restricting IFN effects to the tumor cells while reducing toxicity to the neighboring tissues.


Asunto(s)
Adenoviridae/genética , Interferón-alfa/genética , Animales , Apoptosis , Secuencia de Bases , División Celular , Línea Celular , Cartilla de ADN/genética , Virus de la Encefalomiocarditis/inmunología , Femenino , Expresión Génica , Vectores Genéticos , Humanos , Interferón alfa-2 , Interferón-alfa/metabolismo , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Proteínas Recombinantes , Transducción de Señal , Trasplante Heterólogo , Células Tumorales Cultivadas , alfa-Fetoproteínas/genética
4.
Mol Ther ; 4(1): 5-12, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11472100

RESUMEN

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.


Asunto(s)
Apoptosis , Neoplasias de la Mama/terapia , Genes p53 , Neoplasias Ováricas/terapia , Proteína p53 Supresora de Tumor/genética , Adenoviridae/genética , Animales , Caspasa 9 , Caspasas/metabolismo , División Celular/fisiología , Cisplatino/administración & dosificación , Terapia Combinada , Femenino , Variación Genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Paclitaxel/administración & dosificación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis
5.
Gene Ther ; 8(24): 1847-54, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11821938

RESUMEN

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.


Asunto(s)
Actinas/genética , Adenoviridae/genética , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Músculo Liso Vascular/patología , beta-Galactosidasa/genética , Angioplastia de Balón/efectos adversos , Animales , Fusión Artificial Génica/métodos , Arterias Carótidas/patología , Ciclo Celular , Factores de Transcripción E2F , Humanos , Hiperplasia/prevención & control , Ratas , Proteína de Retinoblastoma/genética , Factores de Transcripción/genética , Transgenes , Túnica Íntima/patología
6.
Hum Gene Ther ; 10(1): 77-84, 1999 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10022532

RESUMEN

A replication-deficient adenovirus encoding human interferon alpha2b, driven by the human cytomegalovirus (CMV) promoter, was constructed and characterized. This construct was used to infect human cells derived from different types of cancer. The production of protein and its secretion into the culture medium were tested by Western blotting and immunoassay. Inhibition of cell proliferation and antiviral activity, two of the most important biological activities of interferon, were observed with this construct. PC-3 cells, derived from human prostatic cancer, or Hep3B cells, derived from human hepatocellular carcinoma, were injected subcutaneously to generate and establish in vivo tumors in athymic nude mice. Intratumoral injection with the recombinant adenovirus expressing interferon alpha2b resulted in complete regression of tumor growth. Our results demonstrate that interferon gene delivery using recombinant adenoviral vectors may be a useful approach to treat a variety of cancers.


Asunto(s)
Terapia Genética/métodos , Interferón-alfa/genética , Adenoviridae/genética , Animales , Western Blotting , División Celular , Citomegalovirus/genética , Femenino , Vectores Genéticos , Humanos , Inmunoensayo , Interferón alfa-2 , Interferón-alfa/biosíntesis , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Recombinantes , Factores de Tiempo , Células Tumorales Cultivadas
7.
Clin Cancer Res ; 4(7): 1649-59, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9676839

RESUMEN

p53 tumor suppressor gene therapy has been proposed for cancers characterized by inactivation of p53 function, and successful therapy will require efficient strategies for gene delivery. To maximize transgene expression in tumors, a clinical strategy has been proposed to treat neoplasms in the liver via hepatic artery administration of a recombinant adenovirus encoding wild-type p53 (rAd-p53). We have developed a syngeneic rat model using a p53mut hepatocellular carcinoma cell line (McA-RH7777) that results in multifocal liver tumor nodules to provide experimental support for this strategy. Treatment of McA-RH7777 cells with rAd-p53 in vitro resulted in efficient transgene expression, growth suppression, and apoptosis. Intrahepatic artery dosing with rAd-p53 or an adenovirus encoding beta-galactosidase (rAd-betagal) increased transgene expression in tumor tissue and decreased systemic exposure when compared with i.v. dosing. Daily hepatic artery dosing of rAd-p53 suppressed tumor growth when compared with untreated rats or animals treated with rAd-betagal. These data demonstrate the potential for arterial gene delivery to tumors using recombinant adenoviruses, and support continued investigation of rAd-p53 gene therapy for liver malignancies.


Asunto(s)
Adenoviridae , Carcinoma Hepatocelular/terapia , Genes p53 , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Neoplasias Hepáticas Experimentales/terapia , Proteínas de Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Carcinoma Hepatocelular/metabolismo , Virus Defectuosos , Femenino , Expresión Génica , Arteria Hepática , Humanos , Infusiones Intraarteriales , Infusiones Intravenosas , Neoplasias Hepáticas Experimentales/metabolismo , Proteínas de Neoplasias/genética , Ratas , Transgenes , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
8.
Cancer Res ; 58(11): 2331-4, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9622068

RESUMEN

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.


Asunto(s)
Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Terapia Genética , Glioblastoma/terapia , Monoéster Fosfórico Hidrolasas , Proteínas Tirosina Fosfatasas/genética , Proteínas Supresoras de Tumor , Adenoviridae/genética , Animales , Cromosomas Humanos Par 10 , Citometría de Flujo , Mutación de Línea Germinal , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosfohidrolasa PTEN , Células Tumorales Cultivadas
9.
Circulation ; 96(6): 1899-905, 1997 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-9323079

RESUMEN

BACKGROUND: The retinoblastoma (Rb) protein is a key cell-cycle regulator that controls entry into the S phase by modulating the activity of the E2F transcription factor. We analyzed the effects of full-length phosphorylation-competent and a mutant truncated form of human Rb for their effects on vascular smooth muscle cell (VSMC) proliferation and neointima formation. METHODS AND RESULTS: A number of mutant forms, both phosphorylation competent and incompetent, of human Rb protein were evaluated for their ability to inhibit E2F activity. The results of these assays indicated that a phosphorylation competent, amino-terminal-truncated Rb protein (Rb56) was a particularly potent inhibitor of E2F-mediated transcription relative to the full-length Rb construct (Rb110). Adenoviral constructs containing Rb56 or Rb110 expressed their respective Rb forms in VSMCs, as determined by Western immunoblot analysis, and were similar in their abilities to arrest the cell cycle, as determined by assays of 3H-thymidine incorporation and by flow cytometric analyses. When examined for their effect on neointima formation after balloon injury of the rat carotid artery, both full-length and truncated forms of Rb inhibited formation of this VSMC-derived lesion. CONCLUSIONS: These analyses revealed that the maintenance of high levels of phosphorylation-competent human Rb, either full-length or truncated forms, in VSMCs is an effective method of modulating the extent of intimal hyperplasia that occurs after balloon-induced vascular injury.


Asunto(s)
Adenoviridae , Proteínas Portadoras , Proteínas de Unión al ADN , Vectores Genéticos , Músculo Liso Vascular/citología , Proteína de Retinoblastoma/genética , Túnica Íntima/patología , Angioplastia de Balón/efectos adversos , Animales , Western Blotting , Estenosis Carotídea/etiología , Estenosis Carotídea/patología , Proteínas de Ciclo Celular/genética , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Factores de Transcripción E2F , Citometría de Flujo , Regulación Viral de la Expresión Génica , Humanos , Hiperplasia , Músculo Liso Vascular/lesiones , Mutación/fisiología , Fosforilación , Ratas , Proteínas Recombinantes/farmacología , Recurrencia , Proteína de Retinoblastoma/análisis , Proteína 1 de Unión a Retinoblastoma , Vena Safena/citología , Factor de Transcripción DP1 , Factores de Transcripción/genética , Transcripción Genética/fisiología , Transfección , Túnica Íntima/lesiones
10.
Cancer Res ; 57(3): 351-4, 1997 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9012453

RESUMEN

During in vitro myogenesis, a portion of myoblasts undergo apoptosis, whereas others continue with their differentiation program and form myotubes that are resistant to cell death. Previous work has shown that the expression of the Cdk inhibitor p21 correlates with enhanced resistance to apoptosis and that forced expression of p21 will confer this phenotype on differentiating myocytes. Here we examine the role of the retinoblastoma gene (Rb) in myocyte survival. Compared with wild-type myocytes, CC42 (Rb-/-) myocytes undergo higher frequencies of apoptosis during mitogen deprivation-induced myogenesis. Despite these features, Rb-/- myocytes display normal up-regulation of p21 and down-regulation of Cdk activities upon differentiation. Adenoviral constructs expressing the Cdk inhibitors p21 or p16 inhibit apoptosis in wild-type but not Rb-/- myocyte cultures. On the other hand, a Rb-expressing adenoviral construct inhibited apoptosis in both cell types. These data demonstrate that Rb functions downstream from the Cdk inhibitors to coordinate cell cycle withdrawal with programmed cell death during myocyte differentiation.


Asunto(s)
Apoptosis , Músculo Esquelético/citología , Proteína de Retinoblastoma/fisiología , Animales , Proteínas Portadoras/fisiología , Diferenciación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/fisiología , Ratones
11.
Int J Cancer ; 67(6): 808-15, 1996 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-8824552

RESUMEN

Alterations in the p53 tumor-suppressor gene occur in 35-60% of human glioblastomas, and re-introduction of p53 can suppress neoplastic growth. To evaluate the potential for p53 gene therapy of glioblastoma, we have analyzed the response of human glioblastoma cell lines in vitro and in vivo to experimental therapy with replication-deficient recombinant adenoviruses encoding wild-type p53 (rAd-p53). Western blot analyses showed high-level expression of p53 protein after treatment with rAd-p53, and transgene expression was dependent on promoter strength. A p53-specific dose-dependent inhibition of in vitro cellular proliferation was observed in 5 of 6 cell lines, and growth inhibition corresponded to adenovirus-mediated gene transfer and expression. p53-specific cell death was quantitated by release of the lactate dehydrogenase enzyme. Fragmentation of DNA into nucleosomal oligomers and the occurrence of a hypodiploid cell population detected by flow cytometry provided evidence for apoptosis. Studies in nude mice demonstrated that ex vivo infection with rAd-p53 suppressed the tumorigenic potential of human glioblastoma cells. Furthermore, direct injection of rAd-p53 into established s.c. xenografts inhibited tumor growth. Our observations suggest that re-introduction of wild-type p53 may have potential clinical utility for gene therapy of glioblastoma.


Asunto(s)
Técnicas de Transferencia de Gen , Genes p53/genética , Terapia Genética , Glioblastoma/terapia , Adenoviridae/genética , Animales , Apoptosis , División Celular/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , L-Lactato Deshidrogenasa/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratas , Células Tumorales Cultivadas
12.
Cancer Gene Ther ; 3(2): 121-30, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8729911

RESUMEN

Human malignancies are often characterized by mutations of the p53 tumor suppressor gene. In a large proportion of cases, the mutation results in production of an altered protein that can bind and inactivate the wild-type gene product. This "dominant-negative" activity of mutant p53 molecules may limit the utility of p53 gene therapy of cancer. Using replication-deficient recombinant adenoviruses (rAd-p53) as a p53 gene delivery system, we evaluated the effects of p53 reintroduction on a series of 45 human cell lines containing wild-type, mutated, or no p53 protein. Results indicate a p53-specific, dose-dependent, and promoter-specific growth inhibition of a majority of p53-altered cell lines that correlates with the degree of adenovirus transgene expression. Similar effects were not observed on cells containing wild-type p53. rAd-p53 inhibited the growth of cells expressing various mutant p53 proteins including those characterized as "dominant negative mutants", and the antiproliferative effects were not abrogated by high levels of endogenous mutated p53 protein. In vivo, rAd-p53 also suppressed tumor growth and increased survival of nude mice bearing tumors that express mutant p53. These results support a role for p53 gene therapy of cancer, including malignancies harboring mutations in this tumor suppressor gene.


Asunto(s)
Adenoviridae/genética , Neoplasias Colorrectales/terapia , Genes p53/genética , Terapia Genética/métodos , Proteína p53 Supresora de Tumor/biosíntesis , Animales , División Celular/efectos de los fármacos , División Celular/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Recombinación Genética , Tasa de Supervivencia , Transducción Genética , Células Tumorales Cultivadas
13.
Cancer Gene Ther ; 2(3): 191-7, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8528962

RESUMEN

We have investigated whether adenovirally mediated gene transfer of the herpes simplex thymidine kinase gene to human hepatocellular carcinoma (HCC) cell lines can sensitize these cells to the prodrug ganciclovir and thereby provide a therapeutic option for this intractable cancer. Two replication-deficient adenoviruses encoding for the herpes simplex virus type-1 (HSV) thymidine kinase (TK) gene were generated in which expression of TK is under the control of either the human cytomegalovirus immediate early promoter (CMV) or the human alpha-fetoprotein (AFP) promoter/enhancer. We demonstrate that the combination of adenovirally mediated TK gene transfer and ganciclovir treatment effectively inhibits proliferation and causes cell death of HCC cells in vitro and that in vivo TK gene transfer and ganciclovir treatment inhibits hepatocellular tumor growth in a mouse model of this cancer. Furthermore, we show that expression of the TK gene can be restricted to those HCCs that express the tumor marker AFP through the incorporation of the AFP enhancer/promoter within an adenoviral vector.


Asunto(s)
Carcinoma Hepatocelular/terapia , Resistencia a Antineoplásicos/genética , Terapia Genética , Herpesvirus Humano 1/genética , Timidina Quinasa/genética , Animales , Carcinoma Hepatocelular/genética , División Celular/efectos de los fármacos , Ganciclovir/farmacología , Vectores Genéticos/farmacología , Humanos , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Timidina Quinasa/farmacología , alfa-Fetoproteínas/biosíntesis , alfa-Fetoproteínas/genética
14.
Hum Gene Ther ; 5(9): 1079-88, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7833367

RESUMEN

We have constructed recombinant human adenoviruses that express wild-type human p53 under the control of either the Ad 2 major late promoter (MLP) or the human cytomegalovirus (CMV) immediate early gene promoter. Each construct replaces the Ad 5 E1a and E1b coding sequences necessary for viral replication with the p53 cDNA and MLP or CMV promoter. These p53/Ad recombinants are able to express p53 protein in a dose-dependent manner in infected human cancer cells. Tumor suppressor activity of the expressed p53 protein was assayed by several methods. [3H]Thymidine incorporation assays showed that the recombinant adenoviruses were capable of inhibiting DNA synthesis in a p53-specific, dose-dependent fashion. Ex vivo treatment of Saos-2 tumor cells, followed by injection of the treated cells into nude mice, led to complete tumor suppression using the MLP/p53 recombinant. Following a single injection of CMV/p53 recombinant adenovirus into the peritumoral space surrounding an in vivo established tumor derived from a human small cell lung carcinoma cell line (NIH-H69), we were able to detect p53 mRNA in the tumors at 2 and 7 days post-injection. Continued treatment of established H69 tumors with MLP/p53 recombinant led to reduced tumor growth and increased survival time compared to control treated animals. These results indicate that recombinant adenoviruses expressing wild-type p53 may be useful vectors for gene therapy of human cancer.


Asunto(s)
Adenovirus Humanos/genética , Carcinoma de Células Pequeñas/terapia , Virus Defectuosos/genética , Genes p53 , Terapia Genética , Vectores Genéticos , Neoplasias/terapia , Animales , Secuencia de Bases , Carcinoma de Células Pequeñas/patología , Citomegalovirus/genética , Replicación del ADN , ADN Complementario/genética , ADN Recombinante/genética , ADN Viral/genética , Femenino , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Neoplasias/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
15.
Mol Cell Biol ; 11(12): 6016-25, 1991 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1944274

RESUMEN

Many essential biological pathways, including cell growth, development, and metabolism, are regulated by thyroid hormones (THs). TH action is mediated by intracellular receptors that belong to a large family of ligand-dependent transcription factors, including the steroid hormone and retinoic acid receptors. So far it has been assumed that TH receptors (TRs) regulate gene transcription only through the classical protein-DNA interaction mechanism. Here we provide evidence for a regulatory pathway that allows cross-talk between TRs and the signal transduction pathway used by many growth factors, oncogenes, and tumor promoters. In transient transfection studies, we observed that the oncogenes c-jun and c-fos inhibit TR activities, while TRs inhibit induction of the c-fos promoter and repress AP-1 site-dependent gene activation. A truncated TR that lacks only 17 amino acids from the carboxy terminus can no longer antagonize AP-1 activity. The cross-regulation between TRs and the signal transduction pathway appears to be based on the ability of TRs to inhibit DNA binding of the transcription factor AP-1 in the presence of THs. The constituents of AP-1, c-Jun, and c-Fos, vice versa, can inhibit TR-induced gene activation in vivo, and c-Jun inhibits TR DNA binding in vitro. This novel regulatory pathway is likely to play a major role in growth control and differentiation by THs.


Asunto(s)
Genes fos , Genes jun , Receptores de Hormona Tiroidea/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular , ADN/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células HeLa , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Activación Transcripcional
16.
Mol Endocrinol ; 5(8): 1109-19, 1991 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1944307

RESUMEN

Thyroid hormone receptors (TRs) are nuclear proteins that regulate gene expression through interactions with specific DNA sequences. It is well known that thyroid hormones have critical functions in the control of normal brain development. In the rat brain, at least three mRNA species are generated by differential processing of the TR alpha transcript. Only one of the isoforms, TR alpha-1, is a transcriptional activator, while the regulatory roles of the carboxy-terminal variants TR alpha-2 and TR alpha-2v remain unclear. In this study we have used polymerase chain reaction amplification of total RNA to compare TR alpha-1, TR alpha-2, and TR alpha-2v mRNA levels in the brainstem, cerebellum, cerebrum, midbrain, and olfactory bulbs of developing neonatal brains in rats. RNA was collected 5, 10, 15, 20, and 25 days after birth from both normal and hypothyroid animals. Coordinate expression of all three isoforms was observed in most tissues during development, with TR alpha-2 generally maintaining the highest level of expression, and TR alpha-1 the lowest. In hypothyroid tissues, TR alpha-1 message was generally increased, while TR alpha-2 was not. To explore the possible roles of the TR alpha isoforms, we have compared their DNA-binding activities. We report that compared to TR alpha-1, the carboxy-terminal variants TR alpha-2 and TR alpha-2v show different binding patterns with a thyroid hormone response element, suggesting that they bind only poorly as monomers. The varying ratios of the TR alpha isoform expression together with their distinct binding patterns and reported repressor functions suggest that TR alpha isoforms have important roles during brain development and function, and may serve to fine-tune the biological responses to thyroid hormone.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Encéfalo/crecimiento & desarrollo , Expresión Génica , Receptores de Hormona Tiroidea/genética , Animales , Animales Recién Nacidos/metabolismo , Encéfalo/metabolismo , ADN/metabolismo , Hipotiroidismo/metabolismo , Sustancias Macromoleculares , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Ratas , Receptores de Hormona Tiroidea/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos
17.
Cell Regul ; 2(7): 565-74, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1782215

RESUMEN

Gene regulation by thyroid hormones is mediated through multiple nuclear receptors. Only some of these thyroid hormone receptor (TR) isoforms become transcriptional enhancers in the presence of the thyroid hormone T3. Here we analyze the regulatory function of the human TR alpha 2 isoform. This protein does not bind T3 and is not a transcriptional activator of thyroid hormone-responsive elements (TRE). Transfected TR alpha 2 functions as a constitutive repressor of the transcriptional activators TR alpha 1 and TR beta 1 but also represses heterologous receptors, including the retinoic acid receptor and the estrogen receptor, which can activate TRE-controlled genes. TR alpha 2 protein showed strongly reduced DNA binding to a palindromic TRE when compared with the active TRs. Hybrid receptor analysis revealed that the special properties of the TR alpha 2 protein, including its repressor function and DNA binding characteristics, are intrinsic properties of its carboxyterminus and can be transferred to other receptors. Although it has been shown that the active TRs can act as repressors and silencers due to their strong DNA binding in the absence of hormone, our data show that TR alpha 2 is unlikely to inhibit TRs and other receptors through a competitive DNA binding mechanism. Antibody gel shift experiments suggest that repression by TR alpha 2 might result from interaction with active receptors. Thus, the receptor-like TR alpha 2 isoform differs from typical nuclear receptors in its DNA-binding and ligand-binding properties and appears to regulate the activity of other receptors via protein-protein interaction.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Receptores de Hormona Tiroidea/fisiología , Secuencia de Bases , Sitios de Unión/fisiología , ADN/metabolismo , Vectores Genéticos , Humanos , Ligandos , Masculino , Datos de Secuencia Molecular , Receptores de Hormona Tiroidea/genética , Transcripción Genética
18.
New Biol ; 3(2): 169-81, 1991 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1648384

RESUMEN

We report here that the thyroid hormone receptors TR alpha and TR beta, and the retinoic acid receptor, RAR, can bind cooperatively to the thyroid hormone response elements (TRE) in both the presence and absence of ligand. Although the transcriptional synergism induced by such cooperative DNA binding could also be influenced by the position of the DNA-binding site on the promoter, the strength of the receptor-DNA interaction in the absence of the cognate ligand of each receptor was in general correlated with the repression activity. The strong-binding TRs, but not the weaker-binding RAR, allowed repression of a constitutive promoter. In addition, strong-binding receptors could repress transcriptional activation of weaker-binding receptors on the TRE. We also show here that the presence of thyroid hormone affects the cooperative DNA binding of TR beta to a TRE dimer by increasing the dissociation rate and decreasing the association rate of TR beta with the DNA. Hybrid receptor analysis revealed that receptor-DNA interaction and repressor activity are largely influenced by the ligand-binding domain of the receptor. We used deletion analysis to localize the sequences conferring a negative effect of thyroid hormone on TR beta binding to DNA and on receptor dimerization or oligomerization. Our data indicate that the ligand-binding domain of thyroid hormone receptors has an essential role in DNA binding and repressor functions, and that this domain exerts its effects by controlling receptor dimerization and oligomerization in the absence and presence of ligand.


Asunto(s)
ADN/metabolismo , Receptores de Hormona Tiroidea/fisiología , Animales , Secuencia de Bases , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/fisiología , Células Cultivadas , Mapeo Cromosómico , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Plásmidos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Receptores de Estrógenos/genética , Receptores de Estrógenos/fisiología , Receptores de Ácido Retinoico , Receptores de Hormona Tiroidea/efectos de los fármacos , Receptores de Hormona Tiroidea/metabolismo , Transcripción Genética , Activación Transcripcional , Triyodotironina/fisiología
19.
New Biol ; 2(7): 613-20, 1990 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2083252

RESUMEN

Most studies on the cloned human estrogen receptor (hER) have been conducted with a mutant receptor in which Gly400 is changed to Val. Here we describe two novel regulatory functions of wild-type hER that are hormone independent: (i) a constitutive activator function and (ii) a repressor activity. Mutations in the hormone-binding domain, including the Val400 mutation, impair both of these functions. In addition, DNA binding is strongly reduced in the mutant receptors. The hormone-binding domain of the hER thus controls DNA binding (and thereby the repressor function) of the hER as well as its constitutive activator function. Moreover, we find that the antiestrogen tamoxifen restores the constitutive activator function, the DNA binding, and the repressor function of the Val400 mutant, but has no effect on the constitutive activator function or DNA binding of the wild-type hER.


Asunto(s)
Receptores de Estrógenos/metabolismo , Secuencia de Bases , Sitios de Unión , ADN/genética , ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Mutación , Plásmidos , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Tamoxifeno/farmacología , Transcripción Genética/efectos de los fármacos
20.
Arch Biochem Biophys ; 278(1): 81-7, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2138870

RESUMEN

The regulation of phosphofructokinase during development of C2C12 myoblasts to myotubes was investigated. Enzyme activity was markedly increased during myogenic development. The increase was observed when enzyme activity was measured under optimal conditions and was not due to changes in the allosteric kinetic properties of the enzyme. Immunoprecipitation of phosphofructokinase from [35S]methionine-labeled myogenic cells revealed that equal amounts of liver and muscle isozymes are present in myoblasts, while in myotubes there was a much higher level of the muscle isozyme. These results were confirmed using an immunoblotting technique. The increase in the level of muscle isozyme in myotubes is due to an increase in the rate of synthesis of the muscle isozyme and occurs in spite of a measurably small increase in its degradation rate. Northern blot analysis using a synthetic oligonucleotide probe showed a 25-fold increase in the level of muscle phosphofructokinase mRNA in myotubes. The conclusion is drawn that the increase in muscle isozyme in myotubes during myogenesis is due to an increase in its mRNA level.


Asunto(s)
Isoenzimas/metabolismo , Músculos/enzimología , Fosfofructoquinasa-1/metabolismo , Animales , Diferenciación Celular , Línea Celular , Electroforesis en Gel de Poliacrilamida , Immunoblotting , Isoenzimas/biosíntesis , Isoenzimas/aislamiento & purificación , Cinética , Peso Molecular , Músculos/citología , Fosfofructoquinasa-1/biosíntesis , Fosfofructoquinasa-1/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA