Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 4: e887, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24176845

RESUMEN

Epithelial ovarian carcinoma is characterized by high frequency of recurrence (70% of patients) and carboplatin resistance acquisition. Carcinoma-associated mesenchymal stem cells (CA-MSC) have been shown to induce ovarian cancer chemoresistance through trogocytosis. Here we examined CA-MSC properties to protect ovarian cancer cells from carboplatin-induced apoptosis. Apoptosis was determined by Propidium Iodide and Annexin-V-FITC labelling and poly-ADP-ribose polymerase cleavage analysis. We showed a significant increase of inhibitory concentration 50 and a 30% decrease of carboplatin-induced apoptosis in ovarian cancer cells incubated in the presence of CA-MSC-conditioned medium (CM). A molecular analysis of apoptosis signalling pathway in response to carboplatin revealed that the presence of CA-MSC CM induced a 30% decrease of effector caspases-3 and -7 activation and proteolysis activity. CA-MSC secretions promoted Akt and X-linked inhibitor of apoptosis protein (XIAP; caspase inhibitor from inhibitor of apoptosis protein (IAP) family) phosphorylation. XIAP depletion by siRNA strategy permitted to restore apoptosis in ovarian cancer cells stimulated by CA-MSC CM. The factors secreted by CA-MSC are able to confer chemoresistance to carboplatin in ovarian cancer cells through the inhibition of effector caspases activation and apoptosis blockade. Activation of the phosphatidylinositol 3-kinase (PI3K)/Akt signalling pathway and the phosphorylation of its downstream target XIAP underlined the implication of this signalling pathway in ovarian cancer chemoresistance. This study reveals the potentialities of targeting XIAP in ovarian cancer therapy.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neoplasias Ováricas/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Carboplatino/farmacología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Línea Celular Tumoral , Cisplatino/farmacología , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Concentración 50 Inhibidora , Transfección , Proteína Inhibidora de la Apoptosis Ligada a X/genética
2.
Br J Cancer ; 103(1): 61-72, 2010 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-20588279

RESUMEN

BACKGROUND: The incidence of ovarian cancer has been increasing worldwide and it is currently the leading cause of death from gynaecological malignancy. Unlike breast cancer, the prognostic role of the human epidermal growth factor receptor-2 (HER-2) in ovarian carcinoma remains controversial. METHODS: The aim of this preclinical study was to further characterise the biological, molecular and cellular effects of trastuzumab (Herceptin) using NIH-OVCAR-3 and derived cell lines both in vitro and in vivo. RESULTS: In vitro assessments have shown that trastuzumab treatment inhibited total and phosphorylated HER-2. This was associated with inhibition of the phosphorylated form of phosphatase and tensin homologue (PTEN), mitogen-activated protein kinase and AKT, and the total level of p27(kip). Inhibition of PTEN is associated with phosphorylated MEK1/2 upregulation, suggesting a specific inhibition of the protein phosphatase function of PTEN. Moreover, trastuzumab induced the upregulation of RhoB. These molecular modifications promote inhibition of cell migration and potentially restoration of tumour cell contact inhibition. RhoB induction in NIH-OVCAR-3 control cell lines mimics the molecular and cellular trastuzumab long-time exposition effects. RhoB inhibition in NIH-OVCAR-3 long-time exposed to trastuzumab cell line reverses the cellular and molecular effects observed in this model. In vivo examinations have shown that these changes are also associated with the restoration of structural, morphological and normal functions of the peritoneum of an ovarian carcinoma mouse model. CONCLUSION: These results provide an indication of the mechanisms underlying the anti-tumour activity of trastuzumab that strongly implicate RhoB in an ovarian carcinoma model that does not show HER-2 amplification or overexpression. These findings highlight that trastuzumab effects involve a possible cross-talk between RhoB and PTEN in the early stages of tumour re-growth in a model of micrometastatic ovarian cancer.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Fosfohidrolasa PTEN/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteína de Unión al GTP rhoB/fisiología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/análisis , Citoesqueleto/química , Citoesqueleto/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfohidrolasa PTEN/fisiología , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Permeabilidad , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor ErbB-2/análisis , Trastuzumab
3.
Cancer Gene Ther ; 16(4): 329-37, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18989351

RESUMEN

Preclinical studies in several animal models as well as clinical trials have shown a reduction in tumor growth following immunotherapy with interleukin-12 (IL-12). This cytokine is appropriate to test in therapeutic clinical trials to treat hepatocarcinoma (HC), a pathology often associated with hepatitis B or C-induced cirrhosis. The local delivery into the liver would be achieved through ex vivo gene transfer using retroviral (rv) vectors in autologous fibroblast carriers. In support of this clinical trial, a rv vector has been constructed to express coordinately both chains p35 and p40 of human IL-12. Here, we have tested good manufacturing practices (GMP) clinical lots of viral vectors derived from the transfected packaging cell line, PG13rvIL-12. We have also devised methods to facilitate the isolation of fibroblasts from freshly harvested skin specimens, enhance their outgrowth in large-scale cultures and assay IL-12 production following transduction, without any selection and irradiation. Twenty-four human skin specimens were processed to obtain fibroblast suspensions that were typically maintained for up to 8 or 12 passages. The mean +/-s.d. overall time for obtaining the required number of transduced cells for the highest IL-12 need was 40 days. The procedure, in accordance with the French medical agency for gene therapy clinical trials, is now ready to begin a clinical trial.


Asunto(s)
Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Neoplasias Hepáticas/terapia , Carcinoma Hepatocelular/genética , Técnicas de Cultivo de Célula , Evaluación Preclínica de Medicamentos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Rayos gamma , Vectores Genéticos , Humanos , Subunidad p35 de la Interleucina-12/biosíntesis , Subunidad p35 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/biosíntesis , Subunidad p40 de la Interleucina-12/genética , Neoplasias Hepáticas/genética , Retroviridae/genética , Transducción Genética
4.
Cancer Gene Ther ; 15(7): 456-64, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18340357

RESUMEN

Ovarian cancers are very aggressive cancers most often diagnosed when metastasis has already occurred in the entire peritoneal cavity. Ovarian adenocarcinoma cells present an undetectable level of RhoB GTPase. Using preclinical ovarian cancer models, we aimed to evaluate the potential use of RhoB cDNA as a tumor suppressor gene in gene therapy. RhoB restoration in vitro, through recombinant adenovirus transduction, resulted in the apoptosis of endogenous RhoB protein low-expressing cell lines (OVCAR-3 and IGROV-1) through the activation of the intrinsic apoptotic caspase cascade. We showed that a single injection of 10(8) p.f.u. of adenoviral vector encoding a reporter gene into the peritoneal cavity of ovarian tumor bearing mice can induce the gene modification of a large quantity of cells throughout the cavity. We thereby tested the effect of AdRhoB injections to treat ovarian cancer-bearing mice. The ectopic expression of RhoB, following its introduction via viral transduction into nude mice in vivo, was highly effective in suppressing tumor growth of ovarian cancer xenografts. Therapeutic agents designed to correct defects of RhoB at the molecular level may thereby provide innovative treatment options for patients not responding to standard therapies.


Asunto(s)
Adenocarcinoma/terapia , Regulación Neoplásica de la Expresión Génica/genética , Terapia Genética/métodos , Neoplasias Ováricas/terapia , Proteína de Unión al GTP rhoB/metabolismo , Adenocarcinoma/enzimología , Adenoviridae , Animales , Línea Celular Tumoral , Femenino , Vectores Genéticos/genética , Humanos , Immunoblotting , Ratones , Microscopía Fluorescente , Neoplasias Ováricas/enzimología , Proteína de Unión al GTP rhoB/genética
5.
Gene Ther ; 14(9): 752-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17344906

RESUMEN

RNA interference (RNAi)-mediated gene silencing approaches appear very promising for therapies based on the targeted inhibition of disease-relevant genes. The major hurdle to the therapeutic development of RNAi strategies remains, however, the efficient delivery of the RNAi-inducing molecules, the short interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs), to the target tissue. With respect to cancer treatment the development of efficient delivery methods into solid tumors appears as a critical issue. However, very few studies have addressed this problem. In this study we have investigated the contribution of electrically mediated delivery of siRNA into murine tumors stably expressing an enhanced green fluorescent protein (EGFP) target reporter gene. The silencing of EGFP gene expression was quantified over time by fluorescence imaging in the living animal. Our study indicates that electric field can be used as an efficient method for siRNA delivery and associated gene silencing into cells of solid tumors in vivo.


Asunto(s)
Electroporación/métodos , Terapia Genética/métodos , Neoplasias/terapia , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Animales , Femenino , Silenciador del Gen , Marcación de Gen , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Gut ; 56(1): 107-14, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16891358

RESUMEN

BACKGROUND: Peritoneal carcinomatosis from pancreatic cancer has a poor prognosis with a median survival of 3.1 months. This is mainly due to lack of effective treatment. Interleukin 12 (IL12) is a proinflammatory cytokine that has a potent antitumoral effect by stimulating innate and adoptive immunity. AIM: To examine the antitumoral effect and toxicity of intraperitoneal delivery of IL12 using an ex vivo gene therapy approach in a murine model of pancreatic peritoneal carcinomatosis. METHODS: Peritoneal carcinomatosis was generated by direct intraperitoneal inoculation of the pancreatic cancer cell line Capan-1 in athymic mice. Syngenic fibroblasts were genetically modified in vitro to secrete IL12 using a polycistronic TFG murine IL12 retroviral vector coding for both p35 and p40 murine IL12 subunits. Ex vivo gene therapy involved injection of the genetically modified fibroblasts intraperitoneally twice a week for 4 weeks. RESULTS: Treatment of pre-established peritoneal carcinomatosis with fibroblasts genetically modified to express IL12 induced a marked inhibition of tumour growth as measured by comparison of the weights of the intraperitoneal tumour nodules in the treated and control animals (3.52 (SD 0.47) v 0.93 (SD 0.21) g, p<0.05) and improved survival. This effect was associated with infiltration of the peritoneal tumour nodules with macrophages. Peritoneal lavage confirmed enhancement of the innate peritoneal inflammatory activity, with an increased number of activated macrophages and natural killer cells. Moreover, macrophages harvested from animals with peritoneal carcinomatosis and treated with IL12-expressing fibroblasts expressed an activated proinflammatory antitumoral M1 phenotype that included strongly enhanced reactive oxygen species and nitric oxide production. There was no treatment-related toxicity. CONCLUSION: Multiple injections of genetically modified fibroblasts to express IL12 is an effective and well-tolerated treatment for experimental murine pancreatic peritoneal carcinomatosis via activated innate immunity and in particular activated M1 macrophages.


Asunto(s)
Antineoplásicos/inmunología , Fibroblastos/inmunología , Terapia Genética/métodos , Interleucina-12/inmunología , Neoplasias Peritoneales/terapia , Animales , División Celular/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo/métodos , Inmunidad Innata/inmunología , Inmunohistoquímica/métodos , Inyecciones Intraperitoneales , Interleucina-12/administración & dosificación , Interleucina-12/genética , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Óxido Nítrico/biosíntesis , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/inmunología , Especies Reactivas de Oxígeno/metabolismo
7.
Gene Ther ; 13(19): 1422-9, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16724094

RESUMEN

Posterior capsule opacification is the main complication of cataract surgery. Using adenovirus-mediated gene transfer, we recently reported that it was feasible to prevent PCO by overexpressing pro-apoptotic molecules such as pro-caspase 3 or Bax in the residual lens epithelial cells post-cataract surgery. However, this approach is feasible only if gene transfer can be restricted to the residual cells responsible for PCO. Initially, we tested an adenovirus (human serotype 5, HAd5), a lentivirus (HIV) and an oncoretrovirus (MLV) vector for the their in vivo transduction efficiency of rabbit lens cells. We found that HAd5 vectors were the most efficient (>90% of the cells could be transduced). Six potential lens-specific promoters were then cloned into HAd5 vectors and assayed for their ability to target expression to a specific population of cells, using in vitro, ex vivo and in vivo rabbit tissues and human lens capsular bags. We found that the LEP503, MIP and Filensin promoters induced strong lens-specific expression of a reporter gene, in human lens cells. Following this ex vivo assay, we showed in a rabbit PCO model that gene transfer could be spatially restricted to the capsular bag by confining the vector with Matrigel. Our combined approach using a lens-specific promoter and a biocompatible gel should render feasible a novel therapeutic strategy for PCO that targets the remaining lens cells.


Asunto(s)
Catarata/prevención & control , Cristalinas/genética , Terapia Genética/métodos , Cápsula del Cristalino/metabolismo , Regiones Promotoras Genéticas , Transducción Genética/métodos , Adenoviridae/genética , Anciano , Anciano de 80 o más Años , Animales , Cámara Anterior , Acuaporinas/genética , Catarata/metabolismo , Catarata/patología , Colágeno , Proteínas de Unión al ADN/genética , Combinación de Medicamentos , Proteínas del Ojo/genética , Expresión Génica , Marcación de Gen , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Humanos , Inmunohistoquímica/métodos , Inyecciones , Proteínas de Filamentos Intermediarios/genética , Laminina , Cápsula del Cristalino/patología , Glicoproteínas de Membrana/genética , Microscopía de Contraste de Fase , Modelos Animales , Proteoglicanos , Conejos , Recurrencia , beta-Galactosidasa/genética
8.
Gene Ther ; 13(5): 440-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16251995

RESUMEN

Posterior capsule opacification (PCO) is a common complication of cataract surgery. Using adenovirus(Ad)-mediated gene transfer, we overexpressed the proapoptotic molecules p53, procaspase 3, Bax, and TRAIL to induce therapeutic programmed cell death of residual lens cells to prevent PCO. Overexpressed TRAIL did not induce apoptosis in cultured rabbit lens cells or in human lens cells. Overexpressed p53 induced apoptosis of lens cells in vitro and ex vivo, but was unable to prevent PCO in vivo. Overexpressed procaspase 3 was associated with engagement of many components of the apoptotic pathway, including cleavage of intracellular caspase targets such as PARP and inter-nucleosome DNA fragmentation. Even when only slightly overexpressed, Bax caused apoptosis of transduced rabbit and human lens cells by engaging the mitochondrial pathway, including catalytic activation of the caspases. A single in vivo injection of Ad vectors expressing either Bax or procaspase 3 into the capsular bag at the end of phacoemulsification prevented PCO in rabbits. These experiments show that Ad-mediated Bax or procaspase 3 overexpression is capable of inducing therapeutic programmed cell death in vitro and in vivo in residual lens cells and preventing PCO in a rabbit model of PCO. Manipulation of proapoptotic molecule expression could be a novel gene therapy approach for prevention of PCO.


Asunto(s)
Terapia Genética/métodos , Cápsula del Cristalino/patología , Facoemulsificación , Complicaciones Posoperatorias/terapia , Adenoviridae/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Caspasa 3 , Caspasas/genética , Línea Celular Transformada , Proliferación Celular , Células Epiteliales/patología , Epitelio Corneal/patología , Regulación de la Expresión Génica , Genes p53 , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Glicoproteínas de Membrana/genética , Complicaciones Posoperatorias/patología , Conejos , Ligando Inductor de Apoptosis Relacionado con TNF , Transducción Genética/métodos , Factor de Necrosis Tumoral alfa/genética , Proteína X Asociada a bcl-2/genética
9.
Proc Natl Acad Sci U S A ; 97(21): 11626-31, 2000 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-11027361

RESUMEN

Protein isoprenylation is a lipid posttranslational modification required for the function of many proteins that share a carboxyl-terminal CAAX motif. The X residue determines which isoprenoid will be added to the cysteine. When X is a methionine or serine, the farnesyl-transferase transfers a farnesyl, and when X is a leucine or isoleucine, the geranygeranyl-transferase I, a geranylgeranyl group. But despite its CKVL motif, RhoB was reported to be both geranylgeranylated and farnesylated. Thus, the determinants of RhoB prenylation appear more complex than initially thought. To determine the role of RhoB CAAX motif, we designed RhoB mutants with modified CAAX sequence expressed in baculovirus-infected insect cells. We demonstrated that RhoB was prenylated as a function of the three terminal amino acids, i.e., RhoB bearing the CAIM motif of lamin B or CLLL motif of Rap1A was farnesylated or geranylgeranylated, respectively. Next, we produced a specific polyclonal antibody against farnesyl cysteine methyl ester allowing prenylation analysis avoiding the metabolic labeling restrictions. We confirmed that the unique modification of the RhoB CAAX box was sufficient to direct the RhoB distinct prenylation in mammalian cells and, inversely, that a RhoA-CKVL chimera could be alternatively prenylated. Moreover, the immunoprecipitation of endogenous RhoB from cells with the anti-farnesyl cysteine antibody suggested that wild-type RhoB is farnesylated in vivo. Taken together, our results demonstrated that the three last carboxyl amino acids are the main determinants for RhoB prenylation and described an anti-farnesyl cysteine antibody as a useful tool for understanding the cellular control of protein farnesylation.


Asunto(s)
Aminoácidos/metabolismo , Cisteína/inmunología , Proteína de Unión al GTP rhoB/metabolismo , Transferasas Alquil y Aril/metabolismo , Secuencias de Aminoácidos , Animales , Secuencia de Bases , Células COS , Cisteína/metabolismo , Cartilla de ADN , Farnesiltransferasa , Mutagénesis , Reacción en Cadena de la Polimerasa , Prenilación de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Spodoptera , Proteína de Unión al GTP rhoB/química , Proteína de Unión al GTP rhoB/genética
10.
J Biol Chem ; 275(40): 31001-8, 2000 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-10896672

RESUMEN

The importance of post-translational geranylgeranylation of the GTPase RhoA for its ability to induce cellular proliferation and malignant transformation is not well understood. In this manuscript we demonstrate that geranylgeranylation is required for the proper cellular localization of V14RhoA and for its ability to induce actin stress fiber and focal adhesion formation. Furthermore, V14RhoA geranylgeranylation was also required for suppressing p21(WAF) transcription, promoting cell cycle progression and cellular proliferation. The ability of V14RhoA to induce focus formation and enhance plating efficiency and oncogenic Ras anchorage-dependent growth was also dependent on its geranylgeranylation. The only biological activity of V14RhoA that was not dependent on its prenylation was its ability to induce serum response element transcriptional activity. Furthermore, we demonstrate that a farnesylated form of V14RhoA was also able to bind RhoGDI-1, was able to induce cytoskeleton organization, proliferation, and transformation, and was just as potent as geranylgeranylated V14RhoA at suppressing p21(WAF) transcriptional activity. These results demonstrate that RhoA geranylgeranylation is required for its biological activity and that the nature of the lipid modification is not critical.


Asunto(s)
Citoesqueleto/metabolismo , Elementos de Respuesta/genética , Transcripción Genética , Proteína de Unión al GTP rhoA/metabolismo , Células 3T3 , Actinas/metabolismo , Animales , Células COS , Ciclo Celular , División Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Citoesqueleto/fisiología , Proteínas de Unión al ADN , Detergentes/farmacología , Adhesiones Focales/metabolismo , GTP Fosfohidrolasas/metabolismo , Genes ras/genética , Glutatión Transferasa/metabolismo , Metabolismo de los Lípidos , Ratones , Microscopía Fluorescente , Proteínas Nucleares , Octoxinol , Plásmidos/metabolismo , Polietilenglicoles/farmacología , Regiones Promotoras Genéticas , Prenilación de Proteína , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Respuesta Sérica , Fibras de Estrés/metabolismo , Factores de Tiempo , Transfección , Vinculina/metabolismo
11.
Crit Rev Oncol Hematol ; 35(1): 33-48, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10863150

RESUMEN

Aggressive non-Hodgkin's lymphona include diffuse large B-cell lymphoma, anaplastic large cell lymphona, and different peripheral T-cell lymphomas. An international prognostic index has been developed including age, serum LDH, performance status, and extranodal involvement. For localized aggressive lymphoma, the preferred treatment is 3-4 CHOP and radiation therapy, with a cure rate of 70-80%. For disseminated aggressive lymphoma, current regimens have a cure rate of less than 40%. Innovative strategies, including dose escalation, autologus stem cell support, new drugs, and immunotherapy are being explored to improve these results.


Asunto(s)
Linfoma no Hodgkin/terapia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Combinada , Trasplante de Células Madre Hematopoyéticas , Humanos , Linfoma no Hodgkin/patología , Linfoma no Hodgkin/fisiopatología , Terapia Recuperativa
12.
Cancer Gene Ther ; 7(12): 1543-56, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11228533

RESUMEN

The expression of costimulatory molecules such as CD70 or CD80 by gene-modified tumor cells has been shown to enhance the antitumor immune response based mainly on T lymphocytes. However, most human tumors show defects of major histocompatibility complex (MHC) expression, preventing them from being recognized by MHC-restricted T cells. To investigate if coexpression of CD70 and CD80 costimulatory molecules induces comparable antitumor responses in low and high MHC-expressing tumor cells, we used two low immunogenic murine tumor models, the B16.F10 melanoma and the TS/A mammary adenocarcinoma cell lines expressing, respectively, low and high levels of MHC class I molecules. Transfection of both CD70 and CD80 genes resulted in an increased capacity of gene-modified tumor cells to costimulate in vitro the proliferation and cytokine production of optimally activated lymphoid cells. Coexpression of CD70 and CD80 by the two tumor cell lines, TS/A and B16.F10, resulted in both cases in partial regression of subcutaneous tumors. Immunochemical analysis and studies in nude mice showed that, even in the B16.F10 model, T cells had a significant role in the antitumor response induced by combining CD70 and CD80. However, rejection of the CD70/CD80-transfected tumor cells appeared more effective in the MHC class I high TS/A model, leading to a protection against parental tumor cells. B16.F10 and TS/A transfectants were then tested with fibroblasts genetically modified to secrete interleukin-12 (IL-12) as a therapeutic vaccine in mice bearing parental tumors. In the two models tested, the injections of irradiated IL-12 and CD70/CD80 gene-modified cells generated an antitumor response to established tumors leading to the slowing down of the tumor growth rate. Although the mechanisms remain to be defined, these findings suggest that the combination of several immuno-modulatory molecules could provide additional strategies for cancer immuno-gene therapy, even for MHC expression-deficient tumors.


Asunto(s)
Antígenos CD , Antígeno B7-1/biosíntesis , Genes MHC Clase I/genética , Terapia Genética/métodos , Neoplasias Mamarias Experimentales/terapia , Melanoma Experimental/terapia , Proteínas de la Membrana/biosíntesis , Animales , Ligando CD27 , Cartilla de ADN , Femenino , Citometría de Flujo , Expresión Génica , Vectores Genéticos , Humanos , Inmunofenotipificación , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Retroviridae/genética , Transfección
13.
Neuroendocrinology ; 70(5): 332-42, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10567859

RESUMEN

In this study, we compared the effects of different chloride (Cl(-)) substitutes - methane sulfonate (CH(3)SO(-)(3)), bromide (Br(-)), nitrate (NO(-)(3)), thiocyanate (SCN(-)) and perchlorate (ClO(-)(4)) - on the secretory activity and calcium current activation of rat lactotropes in primary culture. We observed that CH(3)SO(-)(3) decreased basal prolactin (PRL) secretion. Br(-) had no effect, whereas the more lyotropic anions, such as NO(-3), SCN(-) and C1O(-4), increased basal PRL secretion. The latter three substitutes induced a significant shift in the voltage dependence of T-type calcium channel activation towards hyperpolarized values. However, this shift alone cannot explain the increase in secretion. Anion permeability studies also demonstrated that the organic anion CH(3)SO(-3) was less permeant than Cl(-), whereas monovalent inorganic anions were more permeant, with the following anion permeability sequence: SCN(-) > ClO(-4) > NO(-3) > Br(-). In conclusion, deprivation of Cl(-) ions has converse consequences on basal and induced secretion; permeating anions result in a transient increase in intracellular Ca(2+) ions. This process involves voltage-dependent Ca(2+) channels. We propose that an alteration in intracellular anion concentrations may influence the activation of internal effectors such as G proteins or channel proteins and, therefore, interfere with exocytosis. These effects are correlated with an external action of lyotropic anions, particularly NO(-3), ClO(-4) and SCN(-), on the gating properties of T-type calcium channels, probably through changes in cell surface charges. The results demonstrate the modulatory effect of anions on the secretory activity of rat lactotropes and underline the specific role played by chloride in stimulus-secretion coupling.


Asunto(s)
Calcio/farmacocinética , Mesilatos/farmacología , Hipófisis/metabolismo , Prolactina/metabolismo , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Animales , Bromuros/farmacología , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/fisiología , Células Cultivadas , Cloruros/farmacología , Femenino , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Nitratos/farmacología , Técnicas de Placa-Clamp , Percloratos/farmacología , Hipófisis/citología , Hipófisis/efectos de los fármacos , Ratas , Ratas Wistar , Compuestos de Sodio/farmacología , Tiocianatos/farmacología
14.
Curr Eye Res ; 19(6): 472-82, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10550788

RESUMEN

PURPOSE: The most common complication of cataract surgery is the development of posterior capsule opacification (PCO). Hyperplasia of the lens epithelium is one of the main cellular events following phacoemulsification and was found to be an important feature contributing to opacification of the posterior capsule. We investigated the feasibility of killing the residual lens epithelial cells by retroviral-mediated transfer of the herpes simplex virus-thymidine kinase (HSV-tk) gene, a well-studied suicide gene, into rabbit lens epithelial cells followed by ganciclovir (GCV) treatment. METHODS: The capacity of retroviral vectors to transfer genes into rabbit lens epithelial cells was determined either in vitro (culture of rabbit lens epithelial cells) or in vivo (experimental model of PCO in rabbits) using cDNA encoding the beta-galactosidase (LacZ) reporter gene. To evaluate the efficiency of suicide gene therapy (infection with retroviral vectors encoding the HSV-tk gene followed by GCV treatment) we determined the sensitivity of HSV-tk infected lens epithelial cells to different concentrations of GCV in vitro. Then, in an experimental model of PCO, rabbits were treated with HSV-tk retroviral vectors at the end of the surgery and they received repeated intracameral and intravitreal injections of GCV at the concentration determined by the in vitro experiments. RESULTS: Infection efficiency using LacZ retroviral vectors was about 29% in vitro and 10% in vivo. After infection of the HSV-tk cDNA in vitro, the cell killing effect of GCV was evaluated. A significant enhancement (four- to five-fold) of the cell sensitivity to GCV was shown in FLY-DFGtk as compared with mock infected (P < 0.01) cells even without selection of the HSV-tk positive cells. The GCV concentration leading to 50% reduction in cell number (IC50) was 50 microg/ml. In vivo infection with a HSV-tk vector led to the tk gene transfer into lens epithelial cells. Despite this local HSV-tk gene expression, we could not prevent capsule opacification. CONCLUSIONS: Lens epithelial cells were successfully infected both in vitro and in vivo by beta-galactosidase and HSV-tk genes via retroviral vectors. In vitro infected lens epithelial cells displayed a strong sensitivity to GCV treatment. In vivo, we could not prevent capsule opacification in the rabbit model, very likely due to the limited level of the HSV-tk gene expression. However, our results suggest that virus-mediated suicide gene therapy might be a feasible treatment strategy to prevent capsule opacification with a more powerful vector.


Asunto(s)
Catarata/prevención & control , Técnicas de Transferencia de Gen , Cápsula del Cristalino/metabolismo , Timidina Quinasa/genética , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Catarata/genética , Recuento de Células/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/metabolismo , Femenino , Galactósidos/metabolismo , Ganciclovir/farmacología , Ganciclovir/uso terapéutico , Regulación Enzimológica de la Expresión Génica , Terapia Genética , Herpesvirus Humano 1/enzimología , Herpesvirus Humano 1/genética , Indoles/metabolismo , Cápsula del Cristalino/citología , Cápsula del Cristalino/patología , Conejos , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Retroviridae/genética , Coloración y Etiquetado , Transfección , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
15.
Hum Gene Ther ; 10(14): 2365-72, 1999 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-10515456

RESUMEN

The most common complication of cataract surgery is the development of posterior capsule opacification (PCO). Hyperplasia of the lens epithelium is one of the main cellular events following phacoemulsification, and has been found to be an important feature contributing to opacification of the posterior capsule. Adenoviral vector-mediated transfer is a suitable method for transducing the herpes simplex virus thymidine kinase gene (HSV-tk) into proliferating cells, allowing for the selective killing of these cells by ganciclovir (GCV) treatment. To determine the potential of gene transduction for lens epithelial cells, we studied the transduction of rabbit lens epithelial cells with adenoviral vectors containing either the Escherichia coli beta-galactosidase (lacZ) gene or the HSV-tk gene in vitro and in vivo in an experimental model of PCO. The efficiency of lacZ gene transfer in rabbit lens epithelial cells was at least 95% both in vitro and in vivo. In vivo transduction with HSV-tk adenoviral vector followed by GCV treatment significantly inhibited the development of PCO (p<0.001). These results suggest that adenoviral vector-mediated transfer of HSV-tk into the proliferating lens epithelial cells is feasible and may provide a novel therapeutic strategy for PCO.


Asunto(s)
Catarata/prevención & control , Terapia Genética , Cápsula del Cristalino/patología , Facoemulsificación/efectos adversos , Adenoviridae/genética , Animales , Antivirales/farmacología , Catarata/etiología , Catarata/patología , Células Epiteliales/metabolismo , Estudios de Factibilidad , Ganciclovir/farmacología , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Hiperplasia , Técnicas In Vitro , Microscopía de Contraste de Fase , Conejos , Simplexvirus/genética , Timidina Quinasa/genética , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
16.
Cancer Gene Ther ; 5(3): 163-75, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9622100

RESUMEN

CD70 (CD27 ligand (CD27L)), CD153 (CD30L), and CD154 (CD40L) are members of the tumor necrosis factor family of costimulatory molecules and expressed on the surface of T cells that are important for both T- and B-cell help. We examined the capacity for expression of these tumor necrosis factor family members on tumor cells to induce an antitumor response either in the presence or absence of interleukin 12. Retroviral vectors were constructed that expressed high levels of membrane bound CD70, CD153, or CD154 following infection and selection of the murine tumor lines MCA 207 or TS/A. The genetically modified tumor cells expressing these molecules were able to stimulate splenic cell proliferation, demonstrating that the expressed costimulatory molecules were biologically active. When tested for tumor establishment, the expression of either CD70 or CD154 was able to slow tumor growth. Similarly, CD70 or CD154 were able to induce antitumor immunity at a higher frequency when tested in vaccination and therapy models. CD70 was able to induce antitumor immunity at a level similar to CD80 when tested either in the presence or absence of interleukin 12. Moreover, coexpression of CD70 and CD80 was able to synergize the induction of a higher frequency of antitumor immunity in a vaccination model. Taken together, our results suggest that CD154 and in particular CD70 are able to contribute to the induction of the immune response to tumor in murine models and thus may be of use for human clinical trials.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos CD , Neoplasias Mamarias Experimentales/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/inmunología , Células 3T3 , Adenocarcinoma/terapia , Animales , Secuencia de Bases , Ligando CD27 , Ligando de CD40 , Vacunas contra el Cáncer/uso terapéutico , Cartilla de ADN , Frecuencia de los Genes , Vectores Genéticos , Interleucina-12/genética , Interleucina-12/uso terapéutico , Neoplasias Mamarias Experimentales/terapia , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Retroviridae/genética , Bazo/inmunología , Células Tumorales Cultivadas
18.
Cancer Res ; 57(7): 1364-70, 1997 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9102225

RESUMEN

The basic fibroblast growth factor-(bFGF) mediated signal transduction pathway has been implicated in cellular resistance to ionizing radiation. bFGF is synthesized from the same mRNA in four isoforms resulting from alternative initiations of translation at three CUG start codons (24, 21.5, and 21 kDa) and one AUG start codon (18 kDa). We analyzed the implication of high- and low-molecular forms of bFGF in radioresistance acquisition. For this, we transfected HeLa cells with retroviral vector containing either the CUG-initiated 24-kDa molecular form (HeLa 3A cells), the AUG-initiated 18-kDa molecular bFGF form (HeLa 5A cells), or the vector alone (HeLa PINA cells). A significantly increased radioresistance was obtained only in HeLa 3A cells (Dq = 810 +/- 24 cGy) compared with wild-type cells (Dq = 253 +/- 49 cGy) or HeLa PINA cells (Dq = 256 +/- 29 cGy; P < 0.001). This radioprotective effect was independent of an inhibition of radiation-induced apoptosis but related to an increased G2 duration after irradiation and to an hyperphosphorylation of p34cdc2 kinase. Knowledge of the high-molecular bFGF form-induced radioresistance pathway could offer novel targets for decreasing the radioresistance phenotype of tumors expressing high amounts of bFGF, such as glioblastoma.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fase G2/efectos de la radiación , Tolerancia a Radiación/fisiología , Protectores contra Radiación/farmacología , Tirfostinos , Apoptosis/efectos de la radiación , Western Blotting , Catecoles/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Fase G2/fisiología , Células HeLa , Humanos , Nitrilos/farmacología , Fosforilación/efectos de la radiación , Proteínas Tirosina Quinasas/antagonistas & inhibidores
19.
Ann N Y Acad Sci ; 795: 284-93, 1996 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-8958940

RESUMEN

The recent identification of tumor-associated antigens (TAA) and TAA-derived peptides presented by MHC molecules to T cells has provided the tools to design and test clinical vaccines for treating human malignancies, such as melanoma. While the most effective adjuvant for use in vaccine formulation remains unclear, autologous dendritic cells (DC) appear to be good candidate adjuvants. We have previously shown that syngeneic bone marrow-derived DC when pulsed ex vivo with relevant TAA-derived peptides can effectively vaccinate mice against a subsequent challenge with tumor or can effectively treat animals bearing established tumors. In this report, we have engineered murine interleukin-12 (mIL-12), a potent stimulator of cell-mediated immunity, into murine DC using retroviral-mediated or plasmid-based transfection procedures. Transfectants produced up to 25 ng rIL-12/10(6) cells/48 hours. These engineered cells are capable of promoting enhanced anti-tumor, antigen-specific CTL responses compared to nontransduced DC.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Interleucina-12/administración & dosificación , Sarcoma Experimental/terapia , Adyuvantes Inmunológicos , Animales , Antígenos de Neoplasias/inmunología , Citotoxicidad Inmunológica , Células Dendríticas , Femenino , Fibrosarcoma/terapia , Terapia Genética , Inmunidad Celular , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección
20.
Cell Growth Differ ; 6(11): 1415-23, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8562480

RESUMEN

3-Hydroxy-3-methylglutaryl CoA reductase (HMG-CoA reductase) plays a rate-limiting role in isoprenoid biosynthesis and is associated with cell proliferation and transformation. Although an elevated level of HMG-CoA reductase activity is consistently detected in cancer cell lines and tumors, the question remains whether HMG-CoA reductase activity may have a causative role in cell transformation. We have stably transfected the A549 human adenocarcinoma cells with both bicistronic and retroviral expression vectors, including the whole cDNA of human HMG-CoA reductase. Stably transfected cells showed strong morphological changes and disorganization in the filamentous actin architecture, became contact inhibited, and had a lower doubling time. Moreover, they exhibited anchorage-independent growth reduction and lost their capability to induce tumors in nude mice. Surprisingly, no quantitative modification of enzyme activity was observed following transfection, although expression of HMG-CoA reductase cDNA was shown by Northern blot analysis. When endogenous and transfected reductase activity was bypassed by the addition of mevalonate and compactin, a competitive inhibitor, the filamentous actin distribution in HMG-CoA reductase-transfected cells became very similar to that of control cells, demonstrating the role of exogenous HMG-CoA reductase activity in this process. All of our data together strongly suggest that phenotype reversion is dependent on exogenous HMG-CoA reductase expression and that enzymatic activity is implied in this mechanism. HMG-CoA reductase cDNA expression, by expression of a particular form of reductase, might be a negative regulator of cell growth and thus reverse the phenotype of tumor cells.


Asunto(s)
Acilcoenzima A/genética , Adenocarcinoma/enzimología , Acilcoenzima A/metabolismo , Agar , Animales , División Celular/fisiología , Línea Celular Transformada , Clonación Molecular , Citoesqueleto/enzimología , ADN Complementario/genética , Humanos , Cinética , Neoplasias Pulmonares , Ratones , Ratones Desnudos , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fenotipo , ARN Mensajero/análisis , Transformación Genética , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...