Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 20(7): 742-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23235561

RESUMO

Peripheral neuropathic pain is one of the most common and debilitating complications of diabetes. Several genes have been shown to be effective in reducing neuropathic pain in animal models of diabetes after transfer to the dorsal root ganglion using replication-defective herpes simplex virus (HSV)1-based vectors, yet there has never been a comparative analysis of their efficacy. We compared four different HSV1-based vectors engineered to produce one of two opioid receptor agonists (enkephalin or endomorphin), or one of two isoforms of glutamic acid decarboxylase (GAD65 or GAD67), alone and in combination, in the streptozotocin-induced diabetic rat and mouse models. Our results indicate that a single subcutaneous hindpaw inoculation of vectors expressing GAD65 or GAD67 reduced diabetes-induced mechanical allodynia to a degree that was greater than daily injections of gabapentin in rats. Diabetic mice that developed thermal hyperalgesia also responded to GAD65 or endomorphin gene delivery. The results suggest that either GAD65 or GAD67 vectors are the most effective in the treatment of diabetic pain. The vector combinations, GAD67+endomorphin, GAD67+enkephalin or endomorphin+enkephalin also produced a significant antinociceptive effect but the combination did not appear to be superior to single gene treatment. These findings provide further justification for the clinical development of antinociceptive gene therapies for the treatment of diabetic peripheral neuropathies.


Assuntos
Diabetes Mellitus/terapia , Neuropatias Diabéticas/terapia , Terapia Genética , Simplexvirus/genética , Animais , Complicações do Diabetes , Diabetes Mellitus/genética , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Neuropatias Diabéticas/genética , Modelos Animais de Doenças , Gânglios Espinais/fisiopatologia , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Ratos
2.
Gene Ther ; 14(18): 1344-52, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17611585

RESUMO

Erectile dysfunction (ED) is frequently associated with injury to the cavernous nerve sustained during pelvic surgery. Functional recovery from cavernous nerve injury is generally incomplete and occurs over an extended time frame. We employed a therapeutic gene transfer approach with herpes simplex virus (HSV) vector expressing glial cell line-derived neurotrophic factor (GDNF). Rat cavernous nerve was injured bilaterally using a clamp and dry ice. For HSV-treated groups, 20 microl of purified vector stock was administered directly to and around the damaged nerve. Delivery of an HSV vector expressing both green fluorescent protein (GFP) and lacZ (HSV-LacZ) was used as a control. Intracavernous pressure along with systemic arterial pressure (ICP/AP) was measured 2 and 4 weeks after the nerve injury. Fluorogold (FG) was injected into the penile crus 7 days before killing to assess nerve survival. Approximately 60% of major pelvic ganglion (MPG) cells were GFP positive after viral administration. At 4 weeks after nerve injury, rats treated with HSV-GDNF exhibited significant recovery of ICP/AP compared with control vector or untreated groups. The HSV-GDNF group also yielded more FG-positive MPG cells than the control vector group. HSV vector-mediated delivery of GDNF presents a viable approach for the treatment of ED following cavernous nerve injury.


Assuntos
Disfunção Erétil/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Simplexvirus/genética , Animais , Biomarcadores/análise , Pressão Sanguínea , Disfunção Erétil/metabolismo , Gânglios Espinais/metabolismo , Gânglios Espinais/virologia , Expressão Gênica , Vetores Genéticos/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Injeções , Masculino , Modelos Animais , Óxido Nítrico Sintase Tipo I/análise , Óxido Nítrico Sintase Tipo I/genética , Pênis/lesões , Pênis/inervação , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fatores de Tempo
3.
Gene Ther ; 12(18): 1377-84, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15843809

RESUMO

We examined the utility of herpes simplex virus (HSV) vector-mediated gene transfer of vascular endothelial growth factor (VEGF) in a mouse model of diabetic neuropathy. A replication-incompetent HSV vector with VEGF under the control of the HSV ICP0 promoter (vector T0VEGF) was constructed. T0VEGF expressed and released VEGF from primary dorsal root ganglion (DRG) neurons in vitro, and following subcutaneous inoculation in the foot, expressed VEGF in DRG and nerve in vivo. At 2 weeks after induction of diabetes, subcutaneous inoculation of T0VEGF prevented the reduction in sensory nerve amplitude characteristic of diabetic neuropathy measured 4 weeks later, preserved autonomic function measured by pilocarpine-induced sweating, and prevented the loss of nerve fibers in the skin and reduction of neuropeptide calcitonin gene-related peptide and substance P in DRG neurons of the diabetic mice. HSV-mediated transfer of VEGF to DRG may prove useful in treatment of diabetic neuropathy.


Assuntos
Neuropatias Diabéticas/prevenção & controle , Gânglios Espinais/metabolismo , Terapia Genética/métodos , Simplexvirus/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Diabetes Mellitus Experimental , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/fisiopatologia , Eletrofisiologia , Gânglios Espinais/patologia , Gânglios Espinais/virologia , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica/métodos , Masculino , Camundongos , Medição da Dor , Ratos , Ratos Sprague-Dawley , Pele/inervação , Pele/metabolismo , Transdução Genética/métodos , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Gene Ther ; 11(23): 1675-84, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15306839

RESUMO

Malignant glioma is a fatal human cancer in which surgery, chemo- and radiation therapies are ineffective. Therapeutic gene transfer used in combination with current treatment methods may augment their effectiveness with improved clinical outcome. We have shown that NUREL-C2, a replication-defective multigene HSV-based vector, is effective in treating animal models of glioma. Here, we report safety and biodistribution studies of NUREL-C2 using rhesus macaques as a model host. Increasing total doses (1 x 10(7) to 1 x 10(9) plaque forming units (PFU)) of NUREL-C2 were delivered into the cortex with concomitant delivery of ganciclovir (GCV). The animals were evaluated for changes in behavior, alterations in blood cell counts and chemistry. The results showed that animal behavior was generally unchanged, although the chronic intermediate dose animal became slightly ataxic on day 12 postinjection, a condition resolved by treatment with aspirin. The blood chemistries were unremarkable for all doses. At 4 days following vector injections, magnetic resonance imaging showed inflammatory changes at sites of vector injections concomitant with HSV-TK and TNFalpha expression. The inflammatory response was reduced at 14 days, resolving by 1 month postinjection, a time point when transgene expression also became undetectable. Immunohistochemical staining following animal killing showed the presence of a diffuse low-grade gliosis with infiltrating macrophages localized to the injection site, which also resolved by 1 month postinoculation. Viral antigens were not detected and injected animals did not develop HSV-neutralizing antibodies. Biodistribution studies revealed that vector genomes remained at the site of injection and were not detected in other tissues including contralateral brain. We concluded that intracranial delivery of 1 x 10(9) PFU NUREL-C2, the highest anticipated patient dose, was well tolerated and should be suitable for safety testing in humans.


Assuntos
Encéfalo/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/farmacocinética , Herpesvirus Humano 1/genética , Animais , Anticorpos Antivirais/biossíntese , Encéfalo/patologia , Neoplasias Encefálicas/terapia , Técnicas de Transferência de Genes/efeitos adversos , Terapia Genética/efeitos adversos , Glioma/terapia , Herpesvirus Humano 1/imunologia , Macaca mulatta , Imageamento por Ressonância Magnética , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Distribuição Tecidual , Transgenes , Fator de Necrose Tumoral alfa/metabolismo
5.
Gene Ther ; 7(17): 1483-90, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11001368

RESUMO

Herpes simplex virus (HSV)-mediated delivery of the HSV thymidine kinase (tk) gene to tumor cells in combination with ganciclovir (GCV) administration may provide an effective suicide gene therapy for destruction of malignant glioblastomas. However, because HSV is a highly cytotoxic agent, gene expression from the virus is short-lived which may limit the effectiveness of HSVtk/GCV therapy. Using different replication-defective HSVtk gene vectors, we compared HSV vector backgrounds for their cytotoxic activity on infection of 9L gliosarcoma cells in culture and brain tumors in rats and evaluated the impact of vector toxicity on the effectiveness of tk/GCV-mediated suicide gene therapy. As reported previously for other cell lines, a vector deleted for both copies of the immediate-early (IE) gene ICP4 (SOZ.1) was highly toxic for 9L cells in culture while a vector deleted in addition for the ICP22 and ICP27 IE genes (T.1) reduced or arrested 9L cell proliferation with more limited cell killing. Nevertheless, both vectors supported widespread killing of uninfected cells in the presence of GCV following low multiplicity infections, indicating that vector cytotoxicity did not preempt the production of vector-encoded TK enzyme necessary for the killing of uninfected cells by the HSV-tk/GCV bystander effect. Although an SOZ.1-related vector (SHZ.2) caused tumor cell necrosis in vivo, injection of SHZ.2 at multiple coordinates thoughout the tumor followed by GCV administration failed to prolong markedly the survival of tumor-bearing rats. In contrast, a single injection of T.1 produced a life-extending response to GCV. These results indicate that vector cytotoxicity can limit the efficacy of HSV-tk/GCV treatment in vivo, which may be due to premature termination of tk gene expression with attendant abortion of the bystander effect.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Vetores Genéticos/efeitos adversos , Gliossarcoma/terapia , Simplexvirus/enzimologia , Timidina Quinase/genética , Animais , Antivirais/uso terapêutico , Apoptose , Ganciclovir/uso terapêutico , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Masculino , Ratos , Ratos Endogâmicos F344 , Transfecção , Células Tumorais Cultivadas
6.
Mol Ther ; 1(1): 71-81, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10933914

RESUMO

Tumor cell transduction with the herpes simplex virus (HSV) thymidine kinase (tk) gene and treatment with ganciclovir (GCV) is a widely studied cancer gene therapy. Connexin (Cx)-dependent gap junctions between cells facilitate the intercellular spread of TK-activated GCV, thereby creating a bystander effect that improves tumor cell killing. However, tumor cells often have reduced connexin expression, thus thwarting bystander killing and the effectiveness of TK/GCV gene therapy. To improve the effectiveness of this therapy, we compared an HSV vector (TOCX) expressing Cx43 in addition to TK with an isogenic tk vector (TOZ.1) for their abilities to induce bystander killing of Cx-positive U-87 MG human glioblastoma cells and Cx-negative L929 fibrosarcoma cells in vitro and in vivo. The results showed that low-multiplicity infection of U-87 MG cells with TOCX only minimally increased GCV-mediated cell death compared with infection by TOZ.1, consistent with the endogenous level of Cx in these cells. In contrast, bystander killing of L929 cells was markedly enhanced by vector-mediated expression of Cx. In vivo experiments in which U-87 MG cells were preinfected at low multiplicity and injected into the flanks of nude mice showed complete cures of all animals in the TOCX group following GCV treatment, whereas untreated animals uniformly formed fatal tumors. TOCX injection into U-87 MG intradermal and intracranial tumors resulted in prolonged survival of the host animals in a GCV-dependent manner. Together, these results suggest that the combination of TK and Cx may be beneficial for the treatment of human glioblastoma.


Assuntos
Conexina 43/genética , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Neoplasias Experimentais/terapia , Animais , Antivirais/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/terapia , Terapia Combinada , Feminino , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/terapia , Ganciclovir/farmacologia , Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/terapia , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Timidina Quinase/genética , Transplante Heterólogo , Células Tumorais Cultivadas
7.
Gene Ther ; 7(1): 43-52, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10680015

RESUMO

To develop a cellular vehicle able to reach systemically disseminated areas of angiogenesis, we sought to exploit the natural tropism of circulating endothelial progenitor cells (EPCs). Primate CD34+ EPCs were genetically modified with high efficiency and minimal toxicity using a non-replicative herpes virus vector. These EPCs localized in a skin autograft model of angiogenesis in rhesus monkeys, and sustained the expression of a reporter gene for several weeks while circulating in the blood. In animals infused with autologous CD34+ EPCs transduced with a thymidine kinase-encoding herpes virus, skin autografts and subcutaneous Matrigel pellets impregnated with vascular growth factors underwent necrosis or accelerated regression after administration of ganciclovir. Importantly, the whole intervention was perfectly well tolerated. The accessibility, easy manipulation, lack of immunogenicity of the autologous CD34+ cell vehicles, and tropism for areas of angiogenesis render autologous CD34+ circulating endothelial progenitors as ideal candidates for exploration of their use as cellular vehicles when systemic gene delivery to those areas is required. Gene Therapy (2000) 7, 43-52.


Assuntos
Antígenos CD34/genética , Leucócitos Mononucleares/fisiologia , Transdução Genética/genética , Inibidores da Angiogênese/genética , Animais , Células Cultivadas , Citometria de Fluxo , Técnicas de Transferência de Genes , Humanos , Macaca mulatta , Neovascularização Patológica , Simplexvirus/genética , Pele/irrigação sanguínea , Células-Tronco/fisiologia , beta-Galactosidase/metabolismo
8.
Gene Ther ; 6(5): 904-12, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10505116

RESUMO

We report here the construction and the use of two replication-defective herpes simplex virus vectors, SH FGF-2 and TH FGF-2, which efficiently transfer and express the cDNA for fibroblast-growth-factor-2 (FGF-2) in vitro and in vivo. One mutant was deleted in the immediate-early gene encoding ICP4; the other was deleted in ICP4, ICP22 and ICP27. FGF-2--or the control gene lacZ--were inserted in tk, under control of the human cytomegalovirus immediate-early promoter. Infection of Vero cells with SH FGF-2 induced a dramatic increase in FGF-2 protein levels in the first 2 days after infection, with a rapid return to baseline levels within day 4. In contrast, infection of Vero cells with TH FGF-2 displayed FGF-2 levels progressively increasing up to days 4-5, and slowly returning to baseline. Protein extracts of cells infected with TH FGF-2 induced neuronal differentiation of PC12 cells, indicating that the newly synthesized FGF-2 was biologically active. Robust transient transgene expression was also observed in the rat hippocampus after stereotaxical inoculation of TH FGF-2, but not of TH lacZ or of SH vectors. Enhanced gene expression both in vitro and in vivo by the triple immediate-early gene deletion mutant might be attributed to reduced vector cytotoxicity. The present data suggest that TH FGF-2 is suitable for studies of FGF-2 involvement in neurological disorders.


Assuntos
Epilepsia/terapia , Fator 2 de Crescimento de Fibroblastos/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Simplexvirus/enzimologia , Timidina Quinase/genética , Animais , Química Encefálica , Linhagem Celular , Epilepsia/metabolismo , Expressão Gênica , Masculino , Ratos , Ratos Sprague-Dawley
9.
Gene Ther ; 6(10): 1713-20, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10516720

RESUMO

To evaluate the use of HSV-based vectors for arthritis gene therapy we have constructed a first-generation, ICP4 deficient, replication defective herpes simplex virus (HSV) vector (S/0-) and a second-generation HSV vector derivative (T/0-) deficient for the immediate-early genes ICP4, 22 and 27, each carrying a soluble TNF receptor or IL-1 receptor antagonist transgene cassette. A rabbit synovial-fibroblast line in culture, infected by either vector enabled high-level expression of the transgene product. However, following a single intra-articular injection of the vectors into rabbit knee joints, only the second-generation, HSV T/0- vector expressed detectable levels of soluble TNFR in synovial fluid. Synovial lavage fluid from inoculated joints con- tained up to 12 ng/ml of soluble receptor that persisted at detectable, but reduced levels for at least 7 days. When tested in an experimental model of arthritis generated by intra-articular overexpression of interleukin-1beta using retrovirus transduced synovial cells, the HSV T/0- vector expressing the interleukin-1 receptor antagonist was found to inhibit leukocytosis and synovitis significantly. The improved levels and duration of intra-articular transgene expression achieved via HSV-mediated gene delivery suggest that an HSV vector system could be used for therapeutic applications in patients with rheumatoid arthritis (RA) and other joint-related inflammatory diseases.


Assuntos
Transferência Adotiva , Artrite Experimental/terapia , Marcação de Genes , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Articulação do Joelho , Sialoglicoproteínas/genética , Simplexvirus/genética , Animais , Artrite Experimental/imunologia , Linhagem Celular , Fibroblastos/metabolismo , Expressão Gênica , Membro Posterior , Inflamação/terapia , Injeções Intra-Articulares , Proteína Antagonista do Receptor de Interleucina 1 , Coelhos , Receptores do Fator de Necrose Tumoral/genética , Membrana Sinovial/imunologia
11.
Biochem Biophys Res Commun ; 252(3): 686-90, 1998 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-9837767

RESUMO

Subgroups of the B cell malignancies are known to be associated with Epstein-Barr virus (EBV) infection, especially in immunocompromised patients. These are fatal and refractory to conventional antineoplastic therapy. B cells are usually post-mitotic cells and even mitogen activated or transformed B cells have shown relative resistance against viral mediated gene transfer. To address this issue, we employed a replication-defective herpes simplex virus-1 (HSV-1) to mediate gene transfer into EBV-transformed B cells. The virus expresses the herpes simplex virus thymidine kinase (HSV-TK) and the E. coli lacZ reporter genes and is designated T0Z.1. We used the lymphoblastoid cell line SWEIG as a model for human EBV-related B cell malignancy. This cell line was established by in vitro EBV infection of primary human peripheral blood mononuclear cells. When SWEIG cells were infected with T0Z.1, X-gal staining revealed lacZ expression in more than 20% cells even at multiplicity of infection (MOI) as low as 1 and the expression persisted for at least one week. Ganciclovir (GCV) administration after T0Z.1 infection effectively decreased the number of the infected tumor cells in a dose-responsive manner. Viral toxicity was analyzed by cell proliferation assay (MTS assay) and found to be little even at 10 MOI infection. Three MOI of the virus yielded maximum antineoplastic effect and more than 50% tumor cells were killed by HSV-TK/GCV. These results suggest the potential utility of replication-defective HSV-1 for the treatment of EBV-related B cell malignancies.


Assuntos
Linfócitos B/virologia , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Linfoma de Células B/terapia , Timidina Quinase/genética , Replicação Viral , Antivirais/uso terapêutico , Divisão Celular/efeitos dos fármacos , Transformação Celular Viral , Relação Dose-Resposta a Droga , Ganciclovir/administração & dosagem , Ganciclovir/uso terapêutico , Técnicas de Transferência de Genes , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 4/patogenicidade , Humanos , Linfoma de Células B/virologia , Replicação Viral/genética
12.
Cancer Res ; 58(24): 5731-7, 1998 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9865731

RESUMO

Past studies have documented the promise of herpes simplex virus type 1 (HSV-1) thymidine kinase (TK) suicide gene therapy as a potential antitumor treatment. HSV-TK converts the pro-drug ganciclovir (GCV) into a toxic nucleotide analogue, the incorporation of which into cellular DNA blocks cell proliferation. In this report, we have examined the hypothesis that the effectiveness of HSV-TK suicide gene therapy can be enhanced by coexpression of the antitumor cytokine human tumor necrosis factor-alpha (TNF-alpha) from the same replication-defective HSV-1 vector. In vitro testing demonstrated that TNF-alpha expression from this vector potentiated the killing of both TNF-alpha-sensitive L929 tumor cells and TNF-alpha-resistant U-87 MG cells in the presence of GCV. Furthermore, treatment of established intradermal L929 tumors in vivo with the TNF-alpha/TK vector and GCV resulted in prolonged animal survival compared with treatment with parental HSV-TK vector in the presence or absence of GCV. Treatment of intracerebral U-87 MG tumors showed a clear benefit of TK therapy, but a significant further increase in survival using the TNF-alpha vector could not be demonstrated. We found that potentiation of cell killing in vitro required intracellular TNF-alpha because purified protein added to the culture medium of cells infected with HSV-TK vector failed to have the same effect. Accordingly, potentiation in vivo should depend on efficient infection, but immunohistochemical analysis indicated that virus administration by U-87 MG intratumoral injection was inadequate, resulting in an estimated <1% infection of all tumor cells. Moreover, the majority of infected tumor cells were localized at the tumor margin. Together, these results suggest that TNF-enhanced tk gene therapy should provide a useful treatment for TNF-alpha-sensitive tumors and perhaps also for TNT-alpha-resistant tumors if vector delivery can be improved to increase the percentage of transduced tumor cells.


Assuntos
Terapia Genética , Herpesvirus Humano 1/genética , Fator de Necrose Tumoral alfa/genética , Animais , Chlorocebus aethiops , Resistencia a Medicamentos Antineoplásicos , Estudos de Avaliação como Assunto , Vetores Genéticos , Glioblastoma/terapia , Herpesvirus Humano 1/enzimologia , Humanos , Camundongos , Camundongos Nus , Timidina Quinase/genética , Transdução Genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/uso terapêutico
13.
Gynecol Oncol ; 71(2): 278-87, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9826472

RESUMO

In this report, a replication-defective herpes simplex virus type 1 (HSV-1) vector has been employed to deliver the Escherichia coli LacZ and HSV thymidine kinase (HSVtk) genes to six human ovarian carcinoma cell lines and the efficacy of gene transfer compared to that of adenoviral vectors in vitro. The transduction efficiency of the LacZ-containing virus TOZ.1 was evaluated qualitatively and quantitatively following infection of the different ovarian cancer cell lines. The therapeutic ability of the HSV-T3 vector, which contains the HSVtk gene, was additionally investigated in comparison to the AdCMVHSVTK. Our results show that HSV-1-mediated gene transfer is quantitatively superior to adenoviral vector in five of the six ovarian cancer cell lines at a 100-fold lower dose in vitro. Our preliminary studies suggest that HSV-1 may be a promising alternative vector for ovarian cancer gene therapy.


Assuntos
Terapia Genética , Vetores Genéticos , Herpesvirus Humano 1/genética , Neoplasias Ovarianas/terapia , Timidina Quinase/genética , Divisão Celular , Feminino , Técnicas de Transferência de Genes , Humanos , Óperon Lac , Neoplasias Ovarianas/patologia , Células Tumorais Cultivadas
14.
Neuromuscul Disord ; 8(3-4): 135-48, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9631393

RESUMO

Different viral vectors have been analyzed as gene delivery vehicles to skeletal muscle for potentially therapeutic purposes. In this review, we evaluate the application of retroviral, adenoviral, and herpes simplex viral vectors to deliver genes to skeletal muscle and focus on the dramatic loss of viral transduction detected throughout muscle maturation. Recent results suggested that there are several factors involved in the reduced viral transducibility of mature skeletal muscle: muscle cells become post-mitotic in an early stage, the extracellular matrix develops into a physical barrier, and a loss of myoblast mediation occurs since myoblasts progressively become quiescent. Approaches to improve viral gene delivery to mature skeletal muscle may include the use of particular enzymes to increase the permeability of the extracellular matrix, the pre-treatment of the muscle with a myonecrotic agent to induce myoblast mediation, or the application of the myoblast-mediated ex vivo gene transfer.


Assuntos
Adenoviridae/genética , Envelhecimento/fisiologia , Técnicas de Transferência de Genes , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Retroviridae/genética , Simplexvirus/genética , Animais , Vetores Genéticos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Mutantes
15.
Virology ; 240(2): 245-53, 1998 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-9454698

RESUMO

In this report the ability of enteric immunization with recombinant replication deficient (ICP4-/-) HSV expressing IFN gamma to generate protection and modulate mucosal and systemic immunity was evaluated. ICP4-/-HSV, ICP4-/-HSV expressing IL4, live replicating, and uv HSV were used as controls. Following enteric administration of live HSV, a Th1 cytokine response was induced in the spleen, while both Th1 and notable Th2 cytokine production were detected at mucosal sites. Modulation of mucosal and systemic immune response was achieved when nonreplicating recombinant HSV viruses expressing cytokines were used. Compared to the control replication defective viruses, decreased frequency of Th2 cytokine producing cells in Peyer's patches was observed following enteric administration of nonreplicating HSV expressing IFN gamma. When IFN gamma expressing virus was given enterically, modulation was observed at the systemic level, measured by ELISPOT for cytokine producing cells, ELISA from the in vitro restimulated splenic cell cultures, and by the increase of the IgG2a/IgG1 ratio in the serum. This report provides evidence that replication defective viruses expressing cytokine genes in contrast to uv HSV, are immunogenic when administered enterically and can generate significant immunomodulatory effects at the mucosal and systemic levels.


Assuntos
Citocinas/imunologia , Vírus Defeituosos/imunologia , Herpes Simples/imunologia , Imunização/métodos , Simplexvirus/imunologia , Vacinas Virais/imunologia , Animais , Citocinas/metabolismo , Feminino , Imunidade nas Mucosas/imunologia , Imunidade nas Mucosas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/metabolismo , Vacinas Virais/genética
16.
Gene Ther ; 5(11): 1517-30, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9930305

RESUMO

Some gene therapy applications will require simultaneous expression of multiple gene products to achieve a therapeutic effect. In this study we describe the generation and characterization of replication incompetent herpes simplex virus type 1 (HSV-1) vectors (HX86Z or HX86G) carrying distinct and independently regulated expression cassettes for five transgenes (hIL-2, hGM-CSF, hB7.1, HSV-tk and lacZ or hIFN gamma). The transgenes, representing 12 kb of DNA sequence, were recombined into separate loci of a single mutant virus vector deleted for 11.6 kb of vector sequences representing portions of nine viral genes, ICP4, ICP22, ICP27, ICP47, UL24, UL41, UL44, US10 and US11. Deletion of the immediate--early genes ICP4, ICP22 and ICP27 substantially reduced vector cytotoxicity, prevented early and late viral gene expression and left intact MHC class I antigen expression. Simultaneous expression of multiple transgenes was obtained for up to 7 days in primary human melanoma cells with peak expression at 2-3 days after infection. The transgenes were chosen for their potential to function synergistically in tumor destruction and vaccine gene therapy applications, but the method and vector employed could be applied to other multigene therapy strategies. This study demonstrates the potential for engineering large transgene capacity DNA viruses such as HSV-1 for expression of multiple transgenes.


Assuntos
Terapia Genética , Vetores Genéticos , Simplexvirus/genética , Vírus Defeituosos/genética , Humanos
17.
Gene Ther ; 5(12): 1593-603, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10023438

RESUMO

Herpes simplex virus type 1 (HSV-1) has many attractive features that suggest its utility for gene transfer to neurons. However, viral cytotoxicity and transient transgene expression limit practical applications even in the absence of viral replication. Mutant viruses deleted for the immediate early (IE) gene, ICP4, an essential transcriptional transactivator, are toxic to many cell types in culture in which only the remaining IE genes are expressed. In order to test directly the toxicity of other IE gene products in neurons and develop a mutant background capable of longterm transgene expression, we generated mutants deleted for multiple IE genes in various combinations and tested their relative cytotoxicity in 9L rat gliosarcoma cells, Vero monkey kidney cells, and primary rat cortical and dorsal root neurons in culture. Viral mutants deleted simultaneously for the IE genes encoding ICP4, ICP22 and ICP27 showed substantially reduced cytotoxicity compared with viruses deleted for ICP4 alone or ICP4 in combination with either ICP22, ICP27 or ICP47. Infection of neurons in culture with these triple IE deletion mutants substantially enhanced cell survival and permitted transgene expression for over 21 days. Such mutants may prove useful for efficient gene transfer and extended transgene expression in neurons in vitro and in vivo.


Assuntos
Deleção de Genes , Genes Precoces , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Neurônios/virologia , Proteínas Virais , Animais , Células Cultivadas , Chlorocebus aethiops , Expressão Gênica , Técnicas de Transferência de Genes , Gliossarcoma , Herpesvirus Humano 1/patogenicidade , Proteínas Imediatamente Precoces/genética , Neurônios/metabolismo , Ratos , Células Tumorais Cultivadas , Células Vero , Proteínas Virais Reguladoras e Acessórias
18.
Adv Drug Deliv Rev ; 27(1): 41-57, 1997 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-10837550

RESUMO

Arthritis is presently incurable and poorly treatable, but there are good grounds for expecting gene therapy to improve matters considerably. Although local ex vivo delivery of anti-arthritic genes to the synovial lining of joints has shown considerable promise, intraarticular gene delivery may be desirable. Herpes simplex virus (HSV) may be a viable vector for in vivo transfer of anti-arthritic genes to joints. HSV has the advantages of high infectivity, large carrying capacity and high titer. The large packaging capacity would permit the inclusion of multiple anti-arthritic genes and necessary regulatory elements. Recombinant vectors produced by this laboratory infect synovial cells efficiently, permitting prolonged expression of transgenes in vitro and in vivo without evidence of cytotoxicity. Further improvements to this vector system include taking advantage of an endogenous HSV 'stealthing' gene, ICP47, which interferes with formation of antigen-class I complexes. Inclusion of inducible promoters to appropriately regulate expression of anti-arthritic genes should further improve this system.

19.
Neuromuscul Disord ; 7(5): 299-313, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9267843

RESUMO

The main goal of gene therapy for Duchenne muscular dystrophy (DMD) is to restore dystrophin into as many muscle cells as necessary to be therapeutic. Herpes simplex virus type 1 (HSV-1) represents a promising new viral vector capable of efficient transduction of myofibers in vivo. The viral genome is large and can accommodate multiple or large non-viral genes including the full-length dystrophin. Here we report on the use of a replication defective HSV-1 mutant vector (DZ) deleted for the essential immediate early (IE) gene ICP4 for studies of reporter gene transfer and expression following direct inoculation of mouse skeletal muscle. Our initial experiments showed that HSV-1 can efficiently infect and express a foreign reporter gene in myoblasts and myotubes in vitro. Furthermore, the intramuscular inoculation of HSV-1 resulted in transduction of a significant number of muscle fibers in newborn mice and some muscle fibers in adult animals. We have attempted to exploit these features to develop new HSV mutant vectors for dystrophin gene delivery to DMD muscle, however two impediments to using this virus for muscle gene delivery have to be overcome: namely viral cytotoxicity and the differential transducibility with HSV-1 throughout the development of muscle fibers. To solve the first problem, virus mutants deleted for the immediate early (IE) genes (ICP4, ICP22, ICP27 and UL41) were constructed and the multiple deleted virus was greatly reduced in cytotoxicity relative to our first generation HSV vector strains. Current work is aimed at incorporating full-length dystrophin under muscle specific promoter (muscle creatine kinase MCK) into these new viral vectors. To address the second problem we have analysed by immunohistochemistry the spreading of the HSV-1 in newborn versus adult muscles to determine whether mature basal lamina which surrounds the adult muscle fibers blocks the HSV-1 entry into the mature muscle fibers.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Músculos/fisiologia , Simplexvirus/genética , Animais , Humanos
20.
J Neurovirol ; 3 Suppl 1: S80-8, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9179803

RESUMO

Herpes simplex virus (HSV) represents a candidate gene transfer vector for the treatment of nervous system disease. It has many natural biological features which make it attractive for gene delivery to a variety of tissues. The virus naturally establishes a latency in sensory neurons of the peripheral nervous system, wherein the virus in maintained as an extrachromosomal DNA element in the absence of viral lytic gene expression without altering the metabolism of the host neuron. The virus possesses a neuronal latency-specific promoter system which remains active long-term, while other viral and cellular promoters are repressed. Replication defective virus recombinants have been engineered to delete multiple essential immediate early gene functions rendering these new mutants significantly less cytotoxic to neurons and other cells in culture. Further developments in regulating transgene expression and reducing virus toxicity will continue to aid the design and use of these vectors for therapeutic applications for the nervous system.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Sistema Nervoso , Neurônios/fisiologia , Simplexvirus/fisiologia , Animais , Replicação do DNA , Engenharia Genética/métodos , Genoma Viral , Humanos , Modelos Biológicos , Neurônios/virologia , Simplexvirus/genética , Transfecção , Latência Viral , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...