Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Cell Death Differ ; 20(2): 259-69, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22976834

RESUMO

mnd2 mice die prematurely as a result of neurodegeneration 30-40 days after birth due to loss of the enzymatic activity of the mitochondrial quality control protease HtrA2/Omi. Here, we show that transgenic expression of human HtrA2/Omi in the central nervous system of mnd2 mice rescues them from neurodegeneration and prevents their premature death. Interestingly, adult transgenic mnd2 mice develop accelerated aging phenotypes, such as premature weight loss, hair loss, reduced fertility, curvature of the spine, heart enlargement, increased autophagy, and death by 12-17 months of age. These mice also have elevated levels of clonally expanded mitochondrial DNA (mtDNA) deletions in their tissues. Our results provide direct genetic evidence linking mitochondrial protein quality control to mtDNA deletions and aging in mammals.


Assuntos
Senilidade Prematura/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Animais , Autofagia , Cardiomiopatias/patologia , Sistema Nervoso Central/metabolismo , DNA Mitocondrial/metabolismo , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Humanos , Camundongos , Camundongos Transgênicos , Fenótipo
2.
Neuroscience ; 214: 68-77, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22531373

RESUMO

Caspases are implicated in neuronal death in neurodegenerative and other central nervous system (CNS) diseases. In a rat model of human immunodeficiency virus type 1 (HIV-1) associated neurocognitive disorders (HAND), we previously characterized HIV-1 envelope gp120-induced neuronal apoptosis by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. In this model, neuronal apoptosis occurred probably via gp120-induced reactive oxygen species (ROS). Antioxidant gene delivery blunted gp120-related apoptosis. Here, we studied the effect of gp120 on different caspases (3, 6, 8, 9) expression. Caspases production increased in the rat caudate-putamen (CP) 6h after gp120 injection into the same structure. The expression of caspases peaked by 24h. Caspases colocalized mainly with neurons. Prior gene delivery of the antioxidant enzymes Cu/Zn superoxide dismutase (SOD1) or glutathione peroxidase (GPx1) into the CP before injecting gp120 there reduced levels of gp120-induced caspases, recapitulating the effect of antioxidant enzymes on gp120-induced apoptosis observed by TUNEL. Thus, HIV-1 gp120 increased caspases expression in the CP. Prior antioxidant enzyme treatment mitigated production of these caspases, probably by reducing ROS levels.


Assuntos
Antioxidantes/administração & dosagem , Inibidores de Caspase/administração & dosagem , Caspases/metabolismo , Técnicas de Transferência de Genes , Glutationa Peroxidase/administração & dosagem , Proteína gp120 do Envelope de HIV/administração & dosagem , Superóxido Dismutase/administração & dosagem , Animais , Caspases/biossíntese , Feminino , Regulação Enzimológica da Expressão Gênica , Glutationa Peroxidase/genética , Proteína gp120 do Envelope de HIV/antagonistas & inibidores , Proteína gp120 do Envelope de HIV/genética , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Glutationa Peroxidase GPX1
3.
Gene Ther ; 19(1): 114-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21614027

RESUMO

Potential genetic treatments for many generalized central nervous system (CNS) diseases require transgene expression throughout the CNS. Using oxidant stress and apoptosis caused by HIV-1 envelope gp120 as a model, we studied pan-CNS neuroprotective gene delivery into the cisterna magna (CM). Recombinant SV40 vectors carrying Cu/Zn superoxide dismutase or glutathione peroxidase were injected into rat CMs following intraperitoneal administration of mannitol. Sustained transgene expression was seen in neurons throughout the CNS. On challenge, 8 weeks later with gp120 injected into the caudate putamen, significant neuroprotection was documented. Thus, intracisternal administration of antioxidant-carrying rSV40 vectors may be useful in treating widespread CNS diseases such as HIV-1-associated neurocognitive disorders characterized by oxidative stress.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vírus 40 dos Símios/metabolismo , Transgenes , Animais , Apoptose , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Feminino , Terapia Genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glutationa Peroxidase/administração & dosagem , Glutationa Peroxidase/genética , Glutationa Peroxidase/farmacologia , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/metabolismo , HIV-1/patogenicidade , Imuno-Histoquímica , Manitol/administração & dosagem , Manitol/farmacologia , Neurônios/citologia , Neurônios/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Vírus 40 dos Símios/genética , Superóxido Dismutase/administração & dosagem , Superóxido Dismutase/genética , Superóxido Dismutase/farmacologia
4.
Neuroscience ; 195: 215-23, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21906658

RESUMO

Using bone marrow (BM)-directed gene transfer and permanent transduction via recombinant SV40-derived vectors, we previously reported that BM-derived cells may be progenitors of CNS cells, such as neurons in normal adult animals. In this study, we asked whether the same was true for the CNS blood vessels, that is, whether marrow-resident precursors can migrate to the vasculature of the CNS. SV40-derived gene delivery vectors, carrying marker epitopes (FLAG or AU1), appended to carrier proteins, were injected directly into the femoral BM of rats or rabbits. Controls received intramarrow SV(BUGT), a control vector. Transgene expression was then examined in the vasculature. Endothelial cells expressing the transgenes were observed in the vessels of the striatum, principally localized in laminin- or CD31-positive structures (markers of brain blood vessels). Results in both animal models and with both transgenes were similar. Thus, under physiologic conditions and in the absence of CNS or vascular injury, BM-derived cells can migrate to, and form an endothelial lining for, brain blood vessels. Intramarrow gene delivery may provide an avenue to deliver genes to the vascular endothelium of the CNS.


Assuntos
Células da Medula Óssea/citologia , Encéfalo/irrigação sanguínea , Movimento Celular/fisiologia , Células Endoteliais/citologia , Terapia Genética/métodos , Animais , Vasos Sanguíneos/citologia , Separação Celular , Feminino , Citometria de Fluxo , Vetores Genéticos , Imuno-Histoquímica , Coelhos , Ratos , Ratos Sprague-Dawley , Vírus 40 dos Símios/genética , Células-Tronco/citologia , Transdução Genética , Transgenes
5.
Gene Ther ; 18(7): 682-91, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21368898

RESUMO

Gene transfer to central nervous system (CNS) has been approached using various vectors. Recombinant SV40-derived vectors (rSV40s) transduce human neurons and microglia effectively in vitro and in rodent brains in vivo, so we tested rSV40s gene transfer to rhesus monkey CNS in vivo, to characterize the distribution, duration and safety of such gene delivery. We used rSV40s carrying HIV-1 RevM10 with a carboxyl-terminal AU1 epitope tag as a marker, and others with the antioxidant enzymes, Cu/Zn superoxide dismutase and glutathione peroxidase. Vectors were injected stereotaxically into the caudate nucleus. Transgene expression was studied at 1 and 6 months by immunostaining serial brain sections. After intraparenchymal administration, numerous transgene-expressing cells were seen, with a longitudinal extent of 20 mm. In neurons and, more rarely, microglial cells, transgene expression remained strong throughout the 6-month study period. Astrocytes and oligodendroglia were not transduced. No evidence of inflammation or tissue damage was observed. SV40-derived vectors may thus be useful for long-term gene expression in the monkey brain and, potentially, in the human brain.


Assuntos
Núcleo Caudado , Técnicas de Transferência de Genes , Vetores Genéticos , Vírus 40 dos Símios/genética , Superóxido Dismutase/genética , Animais , Encéfalo/metabolismo , Glutationa Peroxidase/genética , Macaca mulatta , Vírus 40 dos Símios/imunologia , Transdução Genética , Transgenes
6.
Gene Ther ; 17(2): 227-37, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19890354

RESUMO

In cystic fibrosis (CF), respiratory failure caused by progressive airway obstruction and tissue damage is primarily a result of the aberrant inflammatory responses to lung infections with Pseudomonas aeruginosa. Despite considerable improvement in patient survival, conventional therapies are mainly supportive. Recent progress toward gene therapy for CF has been encouraging; however, several factors such as immune response and transduced cell turnover remain as potential limitations to CF gene therapy. As alternative gene therapy vectors for CF, we examined the feasibility of using recombinant SV40-derived vectors (rSV40s), which may circumvent some of these obstacles. To accommodate the large cystic fibrosis transmembrane conductance regulator (CFTR) cDNA, we removed not only SV40 Tag genes, but also all capsid genes. We, therefore, tested whether 'gutless' rSV40s could be packaged and were able to express a functional human CFTR cDNA. The results from our in vitro analysis determined that rSV40-CFTR was able to successfully result in the expression of CFTR protein, which localized to the plasma membrane and restored channel function to CFTR-deficient cells. Similarly, in vivo experiments delivering rSV40-CFTR to the lungs of Cftr-/- mice resulted in a reduction of the pathology associated with intra-tracheal P. aeruginosa challenge. rSV40-CFTR-treated mice had less weight loss when compared with control-treated mice as well as demonstrably reduced lung inflammation as evidence by histology and reduced inflammatory cytokines in the broncho-alveolar lavage. The reduction in inflammatory cytokine levels led to an evident decrease in neutrophil influx to the airways. These results indicate that further study of the application of rSV40-CFTR to CF gene therapy is warranted.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/terapia , Terapia Genética/métodos , Vetores Genéticos , Vírus 40 dos Símios/genética , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Citocinas/análise , DNA Complementar , Estudos de Viabilidade , Pulmão , Camundongos , Camundongos Knockout , Infecções por Pseudomonas/terapia , Pseudomonas aeruginosa , Transdução Genética
7.
Gene Ther ; 14(22): 1555-63, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17984995

RESUMO

This is the second part of a review summarizing progress and prospects in gene therapy clinical research. Twenty key diseases/strategies are succinctly described and commented on by leaders in the field. This part includes clinical trials for skin diseases, neurological disorders, HIV/AIDS, ornithine transcarbamylase deficiency, alpha(1)-antitrypsin deficiency, haemophilia and cancer.


Assuntos
Terapia Genética/tendências , Ensaios Clínicos como Assunto , Técnicas de Transferência de Genes/efeitos adversos , Técnicas de Transferência de Genes/tendências , Terapia Genética/métodos , Vetores Genéticos , Humanos , Neoplasias/terapia , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/tendências
8.
Gene Ther ; 14(23): 1650-61, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17914406

RESUMO

Human immunodeficiency virus-1 (HIV-1) is the most frequent cause of dementia in adults under 40. We sought to use gene delivery to protect from HIV-1-related neuron loss. Because HIV-1 envelope (Env) gp120 elicits oxidant stress and apoptosis in cultured neurons, we established reproducible parameters of Env-mediated neurotoxicity in vivo, then tested neuroprotection using gene delivery of antioxidant enzymes. We injected 100-500 ng mul(-1)gp120 stereotaxically into rat caudate-putamens (CP) and assayed brains for apoptosis by terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) 6-h to 14-day post-injection. Peak apoptosis occurred 1 day after injection of 250 and 500 ng microl(-1)gp120. TUNEL-positive cells mostly expressed neuronal markers (NeuroTrace), although some expressed CD68 and so were most likely microglial cells. Finally, we compared neuroprotection from gp120-induced apoptosis provided by localized and generalized intra-central nervous system (CNS) gene delivery. Recombinant SV40 vectors carrying Cu/Zn superoxide dismutase (SOD1) or glutathione peroxidase (GPx1) were injected into the CP, where gp120 was administered 4-24 weeks later. Alternatively, we inoculated the vector into the lateral ventricle (LV), with or without prior intraperitoneal (i.p.) administration of mannitol. Intracerebral injection of SV(SOD1) or SV(GPx1) significantly protected neurons from gp120-induced apoptosis throughout the 24-week study. Intraventricular vector administration protected from gp120 neurotoxicity comparably, particularly if preceded by mannitol i.p. Thus, HIV-1 gp120 is neurotoxic in vivo, and intracerebral or intra-ventricular administration of rSV40 vectors carrying antioxidant enzymes is neuroprotective. These findings suggest the potential utility of both localized and widespread gene delivery in treating neuroAIDS and other CNS diseases characterized by excessive oxidative stress.


Assuntos
Complexo AIDS Demência/prevenção & controle , Antioxidantes/uso terapêutico , Terapia Genética/métodos , Proteína gp120 do Envelope de HIV/efeitos adversos , HIV-1 , Neurônios/virologia , Complexo AIDS Demência/virologia , Animais , Apoptose , Encéfalo/metabolismo , Encéfalo/virologia , Feminino , Vetores Genéticos/administração & dosagem , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Injeções , Injeções Intraventriculares , Modelos Animais , Neurônios/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Vírus 40 dos Símios/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Transdução Genética/métodos
9.
Gene Ther ; 14(12): 939-49, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17443215

RESUMO

Gene transfer to the central nervous system (CNS) has been approached using various vectors. Recombinant SV40-derived vectors (rSV40s) transduce neurons and microglia effectively in vitro, so we tested rSV40s gene transfer to the CNS in vivo, and characterized the distribution, duration and cell types transduced. We used rSV40s carrying Human Immunodeficiency Virus Type 1 Net protein (HIV-1 Nef) with a C-terminal FLAG epitope tag as a marker, and another with Cu/Zn superoxide dismutase (SOD1). Rats were given vectors stereotaxically, either intraparenchymally into the caudate-putamen (CP) or into the lateral ventricle (LV). FLAG expression was studied for 3 months by immunostaining serial brain sections. After intraparenchymal administration, numerous transgene-expressing cells were seen, many as far as 4 mm from the injection site. Transgene expression remained strong throughout the 3-month study period. Coimmunostaining for lineage markers showed that neurons and, more rarely, microglial cells were tranduced, except astrocytes and oligodendroglia. After injection into the LV, high levels of transgene expression were detected throughout the frontal cortex by Western analysis. Systemic mannitol-induced hyperosmolarity further augmented LV transgene delivery. SV40-derived vectors may, thus, be useful for long-term gene expression in the brain, whether locally by intraparenchymal administration or diffusely by intraventricular injection, with or without mannitol.


Assuntos
Encéfalo/metabolismo , Produtos do Gene nef/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vírus 40 dos Símios/genética , Transdução Genética/métodos , Animais , Biomarcadores/análise , Encéfalo/virologia , Feminino , Expressão Gênica , Imuno-Histoquímica , Injeções , Manitol/farmacologia , Microglia/enzimologia , Microglia/virologia , Neurônios/enzimologia , Neurônios/virologia , Oligopeptídeos , Concentração Osmolar , Peptídeos/análise , Peptídeos/genética , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/análise , Superóxido Dismutase/genética , Fatores de Tempo , Transgenes , Produtos do Gene nef do Vírus da Imunodeficiência Humana
10.
Cancer Gene Ther ; 14(1): 19-29, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16990845

RESUMO

Pancreatic cancer is one of the most aggressive and devastating human malignancies. There is an urgent need for more effective therapy for patients with advanced disease. In this context, genetic therapy potentially represents a rational new approach to treating pancreatic cancer, which could provide an adjunct to conventional options. Because of the promise of recombinant SV40 vectors, we tested their ability to deliver a transgene, and to target a transcript, so as to inhibit pancreatic tumors growth in vivo. BxPC3 and Capan-1 cells were efficiently transduced using SV40 vectors without selection, as compared to synthetic vectors PEI. SV40 vectors were as efficient as adenoviral vectors, and provided long-term transgene expression. Next, we devised a SV40-derived, targeted gene therapy approach of pancreatic cancer, by combining hTR tumor-specific promoter with sst2 somatostatin receptor tumor-suppressor gene. In vitro cell proliferation was strongly impaired following administration of SV(hTR-sst2). SV40-derived sst2-mediated antiproliferative effect was dependent on the local production of somatostatin. In vivo, intratumoral gene transfer of sst2 using rSV40 vectors resulted in a marked inhibition of Capan-1 tumor progression, and proliferation. These results represent the initial steps toward a novel approach to the gene therapy of pancreatic cancer using SV40 as a vector.


Assuntos
Vírus Defeituosos/fisiologia , Técnicas de Transferência de Genes , Neoplasias Pancreáticas/patologia , Vírus 40 dos Símios/fisiologia , Replicação Viral , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Reação em Cadeia da Polimerase , Transdução Genética
11.
Gene Ther ; 13(23): 1645-56, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16871233

RESUMO

Human immunodeficiency virus-1 (HIV-1) infection in the central nervous system (CNS) may lead to neuronal loss and progressively deteriorating CNS function: HIV-1 gene products, especially gp120, induce free radical-mediated apoptosis. Reactive oxygen species (ROS), are among the potential mediators of these effects. Neurons readily form ROS after gp120 exposure, and so might be protected from ROS-mediated injury by antioxidant enzymes such as Cu/Zn-superoxide dismutase (SOD1) and/or glutathione peroxidase (GPx1). Both enzymes detoxify oxygen free radicals. As they are highly efficient gene delivery vehicles for neurons, recombinant SV40-derived vectors were used for these studies. Cultured mature neurons derived from NT2 cells and primary fetal neurons were transduced with rSV40 vectors carrying human SOD1 and/or GPx1 cDNAs, then exposed to gp120. Apoptosis was measured by terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) assay. Transduction efficiency of both neuron populations was >95%, as assayed by immunostaining. Transgene expression was also ascertained by Western blotting and direct assays of enzyme activity. Gp120 induced apoptosis in a high percentage of unprotected NT2-N. Transduction with SV(SOD1) and SV(GPx1) before gp120 challenge reduced neuronal apoptosis by >90%. Even greater protection was seen in cells treated with both vectors in sequence. Given singly or in combination, they protect neuronal cells from HIV-1-gp120 induced apoptosis. We tested whether rSV40 s can deliver antioxidant enzymes to the CNS in vivo: intracerebral injection of SV(SOD1) or SV(GPx1) into the caudate putamen of rat brain yielded excellent transgene expression in neurons. In vivo transduction using SV(SOD1) also protected neurons from subsequent gp120-induced apoptosis after injection of both into the caudate putamen of rat brain. Thus, SOD1 and GPx1 can be delivered by SV40 vectors in vitro or in vivo. This approach may merit consideration for therapies in HIV-1-induced encephalopathy.


Assuntos
Antioxidantes/administração & dosagem , Terapia Genética/métodos , Proteína gp120 do Envelope de HIV/farmacologia , Infecções por HIV/terapia , HIV-1 , Transdução Genética/métodos , Animais , Apoptose/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/virologia , Células COS , Linhagem Celular , Chlorocebus aethiops , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Glutationa Peroxidase/genética , Infecções por HIV/enzimologia , Humanos , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas , Injeções , Neurônios/enzimologia , Neurônios/patologia , Neurônios/virologia , Ratos , Ratos Sprague-Dawley , Vírus 40 dos Símios/genética , Superóxido Dismutase/genética , Transgenes
12.
Gene Ther ; 11(22): 1627-37, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15295615

RESUMO

The CCR5 chemokine receptor is important for most clinical strains of HIV to establish infection. Individuals with naturally occurring polymorphisms in the CCR5 gene who have reduced or absent CCR5 are apparently otherwise healthy, but are resistant to HIV infection. With the goal of reducing CCR5 and protecting CCR5+ cells from R5-tropic HIV, we used Tag-deleted SV40-derived vectors to deliver several anti-CCR5 transgenes: 2C7, a single-chain Fv (SFv) antibody; VCKA1, a hammerhead ribozyme; and two natural CCR5 ligands, MIP-1alpha and MIP-1beta, modified to direct these chemokines, and hence their receptor to the endoplasmic reticulum. These transgenes were delivered using recombinant, Tag-deleted SV40-derived vectors to human CCR5+ cell lines and primary cells: monocyte-derived macrophages and brain microglia. All transgenes except MIP-1alpha decreased CCR5, as assayed by immunostaining, Northern blotting, and cytofluorimetry (FACS). Individually, all transgenes except MIP-1alpha protected from low challenge doses of HIV. At higher dose HIV challenges, protection provided by all transgenes diminished, the SFv and the ribozyme being most potent. Vectors carrying these two transgenes were used sequentially to deliver combination anti-CCR5 genetic therapy. This approach gave approximately additive reduction in CCR5, as measured by FACS and protected from higher dose HIV challenges. Reducing cell membrane CCR5 using anti-CCR5 transgenes, alone or in combinations, may therefore provide a degree of protection from R5-tropic strains of HIV.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Infecções por HIV/prevenção & controle , HIV-1/metabolismo , Receptores CCR5/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/virologia , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Citometria de Fluxo , Marcação de Genes , Engenharia Genética , Vetores Genéticos/genética , Infecções por HIV/metabolismo , Humanos , Fragmentos de Imunoglobulinas/genética , Proteínas Inflamatórias de Macrófagos/genética , Macrófagos/metabolismo , Macrófagos/virologia , Neuroglia/metabolismo , Neuroglia/virologia , RNA Catalítico/genética , Vírus 40 dos Símios/genética , Transgenes
13.
Gene Ther ; 10(26): 2153-6, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14625571

RESUMO

Constitutive expression of alpha(1)-antitrypsin (alpha(1)AT), a serine protease inhibitor, by a recombinant simian virus-40-based vector blocks both HIV gp160 and p55 processing, and so is a powerful inhibitor of HIV replication. To apply these findings more effectively in devising HIV therapies, we tested HIV LTR conditional promoter, to drive the expression of alpha(1)AT. SV[LTR](AT) was designed so that synthesis of human alpha(1)AT would be trans-activated by HIV infection. Cell lines and primary human lymphocytes were transduced with SV[LTR](AT) without selection and detectable toxicity. Responsiveness of alpha(1)AT expression to HIV Tat or HIV challenge was confirmed by Northern blotting, RT-PCR, cytofluorimetry and immunostaining. SV[LTR](AT)-transduced cells were protected from HIV-1(NL4-3) at a challenge dose of 0.04 MOI (T-cell lines) or 0.2 MOI (peripheral blood lymphocytes). Conditional expression of alpha(1)AT consistently protected T cells from HIV challenge as effectively as did constitutive expression. Combining the efficiency of rSV40 vectors with HIV-responsive expression of a highly effective anti-HIV therapeutic may be an effective approach to gene therapy of HIV replication.


Assuntos
Infecções por HIV/terapia , HIV-1 , Vírus 40 dos Símios/genética , Linfócitos T/virologia , Transdução Genética/métodos , alfa 1-Antitripsina/metabolismo , Southern Blotting , Terapia Genética/métodos , Vetores Genéticos , Infecções por HIV/virologia , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , alfa 1-Antitripsina/administração & dosagem
14.
Gene Ther ; 10(22): 1861-73, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14502215

RESUMO

Chronic infection with hepatitis C virus (HCV) may lead to liver failure and hepatocellular carcinoma. Current treatment for HCV includes high systemic doses of interferonalpha (IFNalpha), which is effective in less than half of patients and may have severe side effects. We designed conditional IFNalpha and IFNgamma expression constructs to be triggered by HCV-induced activation of NFkappaB, and delivered these using highly efficient recombinant Tag-deleted SV40-derived vectors. NFkappaB activates the HIV-1NL4-3 long terminal repeat (HIVLTR) as a promoter, which accounts for the conditional transgene expression. Human hepatocyte lines and primary rat hepatocytes (PRH) were transduced with SV[HIVLTR](IFN) vectors, and transfected with HCV cDNA. Production of human and murine IFNalpha and IFNgamma in cytosol and culture supernatants was measured. HCV activated the HIVLTR to produce and secrete IFNs, and did so largely through the NFkappaB binding sites of the HIVLTR. Levels of IFNs secreted, and the magnitude of induction in response to HCV, were greater in hepatocyte lines than in primary cultured hepatocytes. However, even in the latter, supernatant IFNalpha concentrations achieved by this approach were similar to therapeutic serum concentrations sought in systemic IFNalpha-treated patients. In coculture studies, secreted IFNalpha activated its cognate response elements in untransduced cells, suggesting that its potential inhibitory effects on HCV may not be limited to transduced cells. Although HCV replication in culture is difficult to assess, HCV-induced IFNalpha production demonstrably reduced HCV transcription. Conditional expression of IFNs within the liver may represent an attractive approach to therapy of severe chronic HCV infection that could avoid the side effects of systemic treatment regimens.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Hepacivirus/genética , Neoplasias Hepáticas/terapia , NF-kappa B/imunologia , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/virologia , Linhagem Celular , Expressão Gênica , Vetores Genéticos/administração & dosagem , Hepatócitos/metabolismo , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon gama/genética , Interferon gama/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/virologia , Camundongos , Ratos , Vírus 40 dos Símios/genética , Transdução Genética/métodos , Transfecção/métodos , Transgenes , Células Tumorais Cultivadas
15.
Gene Ther ; 10(6): 467-77, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12621451

RESUMO

The central role of endoconvertases and HIV-1 protease (HIV-1 PR) in the processing of HIV proproteins makes the design of specific inhibitors important in anti-HIV gene therapy. Accordingly, we tested native alpha(1) antitrypsin (alpha(1)AT) delivered by a recombinant simian virus-40-based vector, SV(AT), as an inhibitor of HIV-1 proprotein maturation. Cell lines and primary human lymphocytes were transduced with SV(AT) without selection and detectable toxicity. Expression of alpha(1)AT was confirmed by Northern blotting, immunoprecipitation and immunostaining. SV(AT)-transduced cells showed no evidence of HIV-1-related cytopathic effects when challenged with high doses of HIV-1(NL4-3). As measured by HIV-1 p24 assay, SV(AT)-transduced cells were protected from HIV-1(NL4-3) at challenge dose of 40 000 TCID(50) (MOI = 0.04). In addition, peripheral blood lymphocytes treated with SV(AT) were protected from HIV doses challenge up to 40 000 TCID(50) (MOI = 0.04). By Western blot analyses, the delivered alpha(1)AT inhibited cellular processing of gp160 to gp120 and decreased HIV-1 virion gp120. SV(AT) inhibited processing of p55(Gag) as well. Furthermore, high levels of uncleaved p55(Gag) protein were detected in HIV virus particles recovered from SV(AT)-transduced cells lines and primary lymphocytes. Thus, delivering alpha(1)AT using SV(AT) to human lymphocytes strongly inhibits replication of HIV-1, most likely by inhibiting the activities both of the cellular serine proteases involved in processing gp160 and of the aspartyl protease, HIV-1 PR, which cleaves p55(Gag). alpha(1)AT delivered by SV(AT) may represent a novel and effective strategy for gene therapy to interfere with HIV replication, by blocking a stage in the virus replicative cycle that has until now been inaccessible to gene therapeutic intervention.


Assuntos
Produtos do Gene gag/metabolismo , Terapia Genética/métodos , Infecções por HIV/terapia , Precursores de Proteínas/metabolismo , Inibidores de Serina Proteinase/genética , alfa 1-Antitripsina/genética , Animais , Células COS , Células Cultivadas , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Proteína gp160 do Envelope de HIV/metabolismo , Infecções por HIV/imunologia , Humanos , Linfócitos/imunologia , Inibidores de Serina Proteinase/análise , Inibidores de Serina Proteinase/metabolismo , Vírus 40 dos Símios/genética , Transdução Genética/métodos , Replicação Viral , alfa 1-Antitripsina/análise , alfa 1-Antitripsina/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana , Produtos do Gene pol do Vírus da Imunodeficiência Humana
16.
Gene Ther ; 8(13): 1033-42, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11438838

RESUMO

Vectors based on recombinant SV40 viruses (rSV40) are highly effective in delivering transgene expression driven by constitutive promoters. We tested here whether these vectors could be used with conditional promoters and promoters using RNA polymerase III transcription, with inhibition of HIV-1 by Tat activation response (TAR) decoys as a functional measure of effective transgene delivery and activity. TAR decoys inhibit HIV-1 Tat, a trans-activator of HIV-1 transcription. Tat acts early in the viral replicative cycle and is essential for efficient viral replication. We evaluated rSV40 gene delivery using two different inhibitors of Tat. One was a dual function polyTAR gene encoding 25 sequential TAR elements (TAR(25)), plus an antisense tat, driven either by HIV-1 long terminal repeat (HIV-LTR) as a conditional promoter, or by cytomegalovirus immediate-early promoter (CMV-IEP) as a constitutive promoter. The other inhibitor was a single TAR decoy, driven by the U6 small nuclear RNA promoter (U6-P). These decoys were delivered to unselected cells in two different human T lymphocyte lines and to unstimulated primary human peripheral blood mononuclear cells (pbmc). Gene delivery was confirmed by PCR, and expression by RT-PCR. By in situ hybridization analysis, >95% of cells were transduced. These transgene constructs protected all cell types tested from HIV-1, as measured by syncytia formation and p24 antigen release. Somewhat better inhibition of HIV-1 replication was achieved with HIV-1 long terminal repeat (HIV-1 LTR) as a conditional promoter than with the constitutive CMV-IEP. The U6-P was also very effective, driving a TAR(1) transcript. Cell viability was not detectably affected by TAR decoy expression. Thus, rSV40 vectors effectively deliver HIV-1-inhibitory RNAs using either constitutive or conditional pol II promoters, or using a pol III promoter. The versatility of this gene delivery system may prove to be useful in anti-HIV-1 therapeutics.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , HIV-1/fisiologia , Regiões Promotoras Genéticas/genética , Vírus 40 dos Símios/genética , Sobrevivência Celular/genética , DNA Polimerase III/genética , Expressão Gênica , Genes tat/genética , Humanos , Linfócitos/virologia , Transdução Genética , Transgenes/genética , Replicação Viral/genética
17.
Gene Ther ; 8(5): 408-18, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11313818

RESUMO

CXCR4 is the major co-receptor used by X4 strains of human immunodeficiency virus type I (HIV-1). In HIV-1-infected patients, the appearance of X4 strains (T cell line-tropic) correlates with disease progression. Since its discovery, the CXCR4 co-receptor has been a major target for different agents which block its function, such as stromal-derived factor 1alpha (SDF-1alpha) and the anti-CXCR4 monoclonal antibody, 12G5. In the present studies, the 12G5 hybridoma was used to construct a single-chain variable antibody fragment (SFv). Murine leukemia virus (MLV) and simian virus 40 (SV(40)) were utilized as delivery vehicles for the anti-CXCR4 SFv. Intracellular expression of the anti-CXCR4 SFv led to down-regulation of this critical co-receptor, as demonstrated by immunostaining. This effect significantly and specifically protected transduced cells from challenge with HIV-1, as measured by HIV-1 p24 antigen expression. Inhibition of HIV-1 replication was specific for X4 HIV-1 strains as demonstrated by MAGI assays. HeLa-CD4/betagal-CCR5 cells expressing the anti-CXCR4 SFv showed significant inhibition of infectivity by the X4 HIV-1 strain NL4-3, but not with the R5 HIV-1 strain Bal. Thus, this anti-HIV-1 molecular therapy has the potential to inhibit HIV-1 replication and virion spread. Targeting CXCR4 by intracellular immunization could be of additional benefit to certain HIV-1-infected patients on highly active antiretroviral therapy (HAART).


Assuntos
Regulação para Baixo , Terapia Genética/métodos , Infecções por HIV/terapia , HIV-1 , Receptores CXCR4/metabolismo , Animais , Linhagem Celular , Vetores Genéticos , Infecções por HIV/metabolismo , HIV-1/fisiologia , Humanos , Região Variável de Imunoglobulina/genética , Vírus da Leucemia Murina/genética , Receptores CXCR4/genética , Vírus 40 dos Símios/genética , Linfócitos T/virologia , Replicação Viral
19.
Exp Cell Res ; 263(2): 183-92, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11161717

RESUMO

Surfactant-associated protein A (SP-A) is a component of pulmonary surfactant that binds to a specific receptor (SPAR) on the surface of type II alveolar cells of the lung and regulates gene expression and surfactant secretion. Previously we have shown that activation of SPAR by SP-A binding initiates a signal through pathways that involve tyrosine phosphorylation, include IRS-1, and entail activation of phosphatidylinositol 3-kinase (PI3K). In other cell types, cytokines that activate the PI3K signaling pathway promote cell survival. Therefore we investigated whether there was an effect of SP-A on apoptosis as measured by DNA laddering, FACS analysis, TUNEL assay, and annexin V binding. SP-A protected primary cultures of rat type II alveolar cells against the apoptotic effects of etoposide and UV light and also protected the H441 human Clara lung tumor cell line against staurosporine-induced apoptosis. The protective effects of SP-A were abrogated by inhibition of either tyrosine-specific protein kinase activity or PI3K. SP-A/SPAR interaction thus initiates a signaling pathway that regulates apoptosis in type II cells. These findings may be important in understanding the pathogenesis of acute lung injury and pulmonary tumorigenesis and may suggest new therapeutic options.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Proteolipídeos/metabolismo , Alvéolos Pulmonares/citologia , Surfactantes Pulmonares/metabolismo , Transdução de Sinais , Animais , Separação Celular , Células Cultivadas , Cicloeximida/farmacologia , Dexametasona/farmacologia , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Feminino , Citometria de Fluxo , Glucocorticoides/farmacologia , Glicoproteínas/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/efeitos da radiação , Proteína A Associada a Surfactante Pulmonar , Proteínas Associadas a Surfactantes Pulmonares , Ratos , Estaurosporina/farmacologia , Raios Ultravioleta/efeitos adversos
20.
Gastroenterology ; 119(5): 1348-57, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11054394

RESUMO

BACKGROUND & AIMS: In the quest for a recombinant viral vector for liver-directed gene therapy that would permit both prolonged and efficient transgene expression in quiescent hepatocytes in vivo and repeated administration, we evaluated a recombinant simian virus 40 (rSV40). METHODS: The rSV40 was generated through replacement of the DNA encoding for the T antigens (Tag) by the coding region of human bilirubin-uridine 5'-diphosphate-glucuronosyl-transferase (BUGT) complementary DNA (SV-hBUGT). Helper-free rSV40 units were generated at infectious titers of 5 x 10(9) to 1 x 10(10) infectious units (IU)/mL in a Tag-producing packaging cell line (COS-7 cells). RESULTS: After 1, 3, or 7 daily infusions of 3 x 10(9) IU of SV-hBUGT through an indwelling portal vein catheter in bilirubin-UGT-deficient jaundiced Gunn rats, mean serum bilirubin concentrations decreased by 40%, 60% and 70%, respectively, in 3 weeks and remained at those levels throughout the duration of the study (40 days). Results of liver biopsies from SV-hBUGT-treated Gunn rats, but not from controls, were positive for human BUGT DNA, messenger RNA, and protein. Bilirubin-UGT activity in liver homogenates was 8%-12% of normal, and bilirubin glucuronides were excreted in bile. Immunostaining showed that >50%-60% of hepatocytes stably expressed the transgene. Portal vein infusion of an rSV40 expressing hepatitis B surface antigen (HBsAg) in a naive Gunn rat and a Gunn rat that had received 7 injections of SV-BUGT resulted in approximately equal levels of hepatic expression of HBsAg, indicating that multiple inoculations of SV-BUGT did not elicit neutralizing antibodies. Plasma alanine aminotransferase levels and liver histology remained normal despite repeated injections of rSV40. CONCLUSIONS: rSV40 vectors may represent a significant advance toward gene therapy for metabolic diseases.


Assuntos
Terapia Genética , Icterícia/terapia , Fígado/fisiopatologia , Vírus 40 dos Símios/genética , Animais , Bile/química , Pigmentos Biliares/análise , Bilirrubina/sangue , Bilirrubina/metabolismo , Células COS , Feminino , Expressão Gênica , Vetores Genéticos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Icterícia/fisiopatologia , Masculino , Ratos , Ratos Gunn , Retratamento , Vírus 40 dos Símios/fisiologia , Transgenes/genética , Carga Viral , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...