Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Am J Pathol ; 174(3): 989-98, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19218348

RESUMEN

Polymyositis is a rare and severe inflammatory muscle disorder. Treatments are partially efficacious but have many side effects. New therapeutic approaches must be first tested in a relevant animal model. Regulatory CD4+CD25+ T cells (Tregs) have been rediscovered as a pivotal cell population in the control of autoimmunity, but the connection between polymyositis and Tregs is currently unknown. To develop a reproducible experimental autoimmune myositis model of polymyositis, mice were immunized once a week for 3 weeks with 1 mg of partially purified myosin emulsified in complete Freund's adjuvant. All mice injected with myosin and complete Freund's adjuvant developed myositis. The infiltrates were composed of CD4(+) and CD8(+) cells, as well as macrophages, but did not contain B lymphocytes. In mice that were depleted of Tregs, the myositis was more severe, as determined by quantitative scoring of muscle inflammation (2.36 +/- 0.9 vs. 1.64 +/- 0.8, P = 0.019). In contrast, injection of in vitro expanded polyclonal Tregs at the time of immunization significantly improved the disease (quantitative score of inflammation 0.87 +/- 1.06 vs. 2.4 +/- 0.67, P = 0.047). Transfer of sensitized or CD4(+)-sorted cells from the lymph nodes of experimental autoimmune myositis mice induced myositis in naïve, irradiated, recipient mice. Thus, experimental autoimmune myositis is a reproducible, transferable disease in mice, both aggravated by Treg depletion and improved by polyclonal Treg injection.


Asunto(s)
Enfermedad Autoinmune Experimental del Sistema Nervioso/inmunología , Linfocitos T Reguladores/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Inmunización , Inmunohistoquímica , Inflamación/inmunología , Inflamación/patología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Ratones , Ratones Endogámicos BALB C , Fuerza Muscular , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miosinas/fisiología , Enfermedad Autoinmune Experimental del Sistema Nervioso/patología
2.
Neurosurg Focus ; 20(4): E25, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16709031

RESUMEN

OBJECT: The authors had previously reported on a replication-competent retrovirus (RCR) that has been demonstrated to be stable, capable of effective transduction, and able to prolong survival in an intracranial tumor model in nude mice. The purpose of this study was further investigation of this gene therapy option. METHODS: The transduction efficiency of RCR in RG2, an immunocompetent intracranial tumor model, was tested in Fischer 344 rats. The immune response to the RCR vector was expressed by the quantification of CD4, CD8, and CD11/b in tumors. The pharmaceutical efficacy of the suicide gene CD in converting prodrug 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU) was measured using fluorine-19 nuclear magnetic resonance (19F-NMR) spectroscopy. Animal survival data were plotted on Kaplan-Meier survival curves. Finally, the biodistribution of RCR was determined using quantitative real-time polymerase chain reaction (RT-PCR) for the detection of retroviral env gene. There was no evidence of viral transduction in normal brain cells. Neither severe inflammation nor immunoreaction occurred after intracranial injection of RCR-green fluorescent protein compared with phosphate-buffered saline (PBS). The 19F-NMR spectroscopy studies demonstrated that RCR-CD was able to convert 5-FC to 5-FU effectively in vitro. The infection of RG2 brain tumors with RCR-CD and their subsequent treatment with 5-FC significantly prolonged survival compared with that in animals with RG2 transduced tumors treated with PBS. In contrast to the nude mouse model, evidence of virus dissemination to the systemic organs after intracranial injection was not detected using RT-PCR. CONCLUSIONS: The RCR-mediated suicide gene therapy described in this paper effectively transduced malignant gliomas in an immunocompetent in vivo rodent model, prolonging survival, without evidence of severe intracranial inflammation, and without local transduction of normal brain cells or systemic organs.


Asunto(s)
Neoplasias Encefálicas/terapia , Genes Transgénicos Suicidas/genética , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Glioma/terapia , Viroterapia Oncolítica/métodos , Retroviridae/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , ADN Viral/análisis , ADN Viral/genética , Modelos Animales de Enfermedad , Encefalitis/fisiopatología , Encefalitis/prevención & control , Flucitosina/metabolismo , Terapia Genética/tendencias , Vectores Genéticos/genética , Glioma/genética , Glioma/metabolismo , Humanos , Huésped Inmunocomprometido/genética , Masculino , Viroterapia Oncolítica/tendencias , Ratas , Ratas Endogámicas F344 , Tasa de Supervivencia , Transducción Genética/métodos , Transducción Genética/tendencias , Resultado del Tratamiento , Replicación Viral/genética
3.
Cancer Biol Ther ; 4(9): 987-92, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16082194

RESUMEN

Amphotropic retroviruses with modified envelope displaying single-chain antibody fragment (scFv) directed against the c-Met receptor were recently generated and found to efficiently and selectively deliver genes into hepatocarcinoma cells. A large proportion of human gliomas also frequently overexpresses c-Met. We therefore explored the possibility of infecting glioma cells using such retroviruses bearing an scFv directed against c-Met. In one construct, a urokinase (uPA) cleavage site was inserted between the scFv and the envelope. We assessed the transduction by these chimeric viruses of a panel of seven human glioma cell lines that we characterized for their c-Met and uPA levels. We found that abundance of the c-Met receptor and viral infection were inversely correlated if we used the retrovirus displaying scFv directed against c-Met, suggesting that the chimeric virus binds preferentially to the c-Met receptor, resulting in virus sequestration. Addition of the uPA site between the scFv moiety and the envelope restored the infectivity of the virus, consistent with a "two-step" infection process: (1) virus binding to the c-Met receptor, (2) cleavage of the scFv moiety by uPA, enabling the virus to dissociate from c-Met and entry into the cells via the Pit-2 receptor. Our study has significant implications for the design of targeting strategies for gliomas expressing high levels of c-Met.


Asunto(s)
Marcación de Gen , Glioma/virología , Fragmentos de Inmunoglobulinas/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Retroviridae/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Anticuerpos/genética , Anticuerpos/inmunología , Línea Celular Tumoral , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glioma/genética , Glioma/metabolismo , Humanos
4.
Cancer Gene Ther ; 11(12): 782-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15472716

RESUMEN

In humans, no efficient treatment exists not only against multifocal liver metastases (LM) but also against recurrent microscopic liver metastases within the liver remnant following curative liver resection. Furthermore, in nonmultifocal LM, partial liver resection could be performed, but in more than 50% of the patients, tumor recurrence within liver remnant is observed, partly due to the growth of dormant cancer cells in the setting of postoperative host immune dysfunction. We investigated the therapeutic potential of interleukin-12 (IL-12) immuno-gene therapies in these experimental models under total vascular exclusion (TVE) of the liver. In rats with multiple LM of DHDK12 colon cancer cells, we observed a significant reduction in tumor volume after retroviral-mediated gene transfer of either herpes simplex virus thymidine kinase (HSV1-TK) and ganciclovir (GCV) administration, or IL-12. Combined treatment with HSV1-TK/GCV and IL-12 resulted in improved tumor volume reduction and even survival. In rats with recurrent microscopic DHDK12 LM established after partial liver resection, we observed significantly decreased recurrent tumor volumes and increased survival after retroviral-mediated IL-12 gene transfer. In both settings, immunohistological analysis revealed that IL-12 immuno-gene therapy was accompanied by an infiltration of CD8+ T lymphocytes within the tumors. Altogether, our results suggest that IL-12 adjuvant gene therapy could improve the management of patients with either resectable or unresectable LM.


Asunto(s)
Neoplasias Colorrectales/patología , Terapia Genética , Interleucina-12/uso terapéutico , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Terapia Combinada , Ganciclovir/uso terapéutico , Inmunohistoquímica , Interleucina-12/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Ratas , Simplexvirus/genética , Timidina Quinasa/uso terapéutico , Células Tumorales Cultivadas
5.
Cancer Gene Ther ; 10(1): 30-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12489026

RESUMEN

Poor efficiency of gene transfer into cancer cells constitutes the major bottleneck of current cancer gene therapy. We reasoned that because tumors are masses of rapidly dividing cells, they would be most efficiently transduced with vector systems allowing transgene propagation. We thus designed two replicative retrovirus-derived vector systems: one inherently replicative vector, and one defective vector propagated by a helper retrovirus. In vitro, both systems achieved very efficient transgene propagation. In immunocompetent mice, replicative vectors transduced >85% tumor cells, whereas defective vectors transduced <1% under similar conditions. It is noteworthy that viral propagation could be efficiently blocked by azido-thymidine, in vitro and in vivo. In a model of established brain tumors treated with suicide genes, replicative retroviral vectors (RRVs) were approximately 1000 times more efficient than defective adenoviral vectors. These results demonstrate the advantage and potential of RRVs and strongly support their development for cancer gene therapy.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos , Virus de la Leucemia Murina de Moloney/genética , Neoplasias/terapia , Células 3T3 , Animales , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/genética , Neoplasias/virología , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Replicación Viral
6.
PLoS One ; 5(2): e9299, 2010 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-20174581

RESUMEN

Skeletal muscle is rarely a site of malignant metastasis; the molecular and cellular basis for this rarity is not understood. We report that myogenic cells exert pronounced effects upon co-culture with metastatic melanoma (B16-F10) or carcinoma (LLC1) cells including conversion to the myogenic lineage in vitro and in vivo, as well as inhibition of melanin production in melanoma cells coupled with cytotoxic and cytostatic effects. No effect is seen with non-tumorigenic cells. Tumor suppression assays reveal that the muscle-mediated tumor suppressor effects do not generate resistant clones but function through the down-regulation of the transcription factor MiTF, a master regulator of melanocyte development and a melanoma oncogene. Our findings point to skeletal muscle as a source of therapeutic agents in the treatment of metastatic cancers.


Asunto(s)
Músculo Esquelético/citología , Mioblastos/citología , Neoplasias Experimentales/patología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Citotoxicidad Inmunológica/inmunología , Desmina/genética , Desmina/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Mioblastos/inmunología , Mioblastos/metabolismo , Metástasis de la Neoplasia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Gene Med ; 7(3): 276-87, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15515136

RESUMEN

BACKGROUND: Recently, several cancer gene therapy studies have shown that replication-competent retroviral vectors represent a major improvement over replication-defective ones in terms of transgene propagation efficiency. However, this positive effect is somewhat spoiled by the increased risk of dissemination and oncogenesis that replication-competent retroviral vectors entail. To enhance both their integral safety and their transgene capacity, we developed a semi-replication-competent retroviral vector system. METHODS: The semi-replication-competent retroviral vector system is based on two transcomplementing replication-defective retroviral vectors termed gag-pol vector (GPv) and env vector (Ev). Vector propagation was monitored in vitro and in solid tumors in vivo, using different reporter transgenes for GPv and Ev. Systemic vector dissemination and leukemogenesis was assessed by direct intravenous vector injection and subsequent bone marrow transplantation, in MLV-sensitive mice. RESULTS: In vitro and in vivo the semi-replication-competent retroviral vectors propagate transgenes almost as efficiently as replication-competent ones. The semi-replication-competent retroviral vector system does not lead to detectable dissemination or leukemogenesis as does the replication-competent vector or the parental virus. Additionally, the vector duo allows co-propagation of different transgenes as well as mobilization of a third replication-defective vector. CONCLUSIONS: This study is an initial proof of principle for the use of complementary retroviral vectors to deliver and propagate transgenes in vitro and in solid tumors in vivo, but with reduced pathogenicity compared to its parental virus. In-between replication-defective and replication-competent retroviral vectors, this semi-replicative system offers good grounds for its application in in vitro studies and allows envisioning its further development for cancer gene therapy.


Asunto(s)
Replicación del ADN , Técnicas de Transferencia de Gen , Vectores Genéticos , Retroviridae/genética , Animales , Trasplante de Médula Ósea , Línea Celular , Genes Reporteros , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratas , Retroviridae/metabolismo , Transducción Genética , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA