Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
1.
Exp Brain Res ; 239(6): 2001-2014, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33909113

ABSTRACT

Scientists have predominantly assessed anxiety's impact on postural control when anxiety is created by the need to maintain balance (e.g., standing at heights). In the present study, we investigate how postural control and its mechanisms (i.e., vestibular function) are impacted when anxiety is induced by an unrelated task (playing a video game). Additionally, we compare watching and playing a game to dissociate postural adaptations caused by increased engagement rather than anxiety. Participants [N = 25, female = 8, M (SD) age = 23.5 (3.9)] held a controller in four standing conditions of varying surface compliance (firm or foam) and with or without peripheral visual occlusion across four blocks: quiet standing (baseline), watching the game with a visual task (watching), playing the game (low anxiety), and playing under anxiety (high anxiety). We measured sway area, sway frequency, root mean square (RMS) sway, anxiety, and mental effort. Limited sway differences emerged between anxiety blocks (only sway area on firm surface). The watching block elicited more sway than baseline (greater sway area and RMS sway; lower sway frequency), and the low anxiety block elicited more sway than the watching block (greater sway area and RMS sway; higher sway frequency). Mental effort was associated with increased sway area and RMS sway. Our findings indicate that anxiety, when generated through competition, has minimal impact on postural control. Postural control primarily adapts according to mental effort and more cognitively engaging task constraints (i.e., playing versus watching). We speculate increased sway reflects the prioritization of attention to game performance over postural control.


Subject(s)
Video Games , Anxiety , Cognition , Female , Humans , Postural Balance , Standing Position
2.
Cell Death Differ ; 20(2): 259-69, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22976834

ABSTRACT

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.


Subject(s)
Aging, Premature/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Animals , Autophagy , Cardiomyopathies/pathology , Central Nervous System/metabolism , DNA, Mitochondrial/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Humans , Mice , Mice, Transgenic , Phenotype
3.
Neuroscience ; 214: 68-77, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22531373

ABSTRACT

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.


Subject(s)
Antioxidants/administration & dosage , Caspase Inhibitors/administration & dosage , Caspases/metabolism , Gene Transfer Techniques , Glutathione Peroxidase/administration & dosage , HIV Envelope Protein gp120/administration & dosage , Superoxide Dismutase/administration & dosage , Animals , Caspases/biosynthesis , Female , Gene Expression Regulation, Enzymologic , Glutathione Peroxidase/genetics , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp120/genetics , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Glutathione Peroxidase GPX1
4.
Gene Ther ; 19(1): 114-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21614027

ABSTRACT

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.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/administration & dosage , Simian virus 40/metabolism , Transgenes , Animals , Apoptosis , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/virology , Female , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glutathione Peroxidase/administration & dosage , Glutathione Peroxidase/genetics , Glutathione Peroxidase/pharmacology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , HIV-1/pathogenicity , Immunohistochemistry , Mannitol/administration & dosage , Mannitol/pharmacology , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oxidative Stress , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Superoxide Dismutase/administration & dosage , Superoxide Dismutase/genetics , Superoxide Dismutase/pharmacology
5.
Neuroscience ; 195: 215-23, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21906658

ABSTRACT

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.


Subject(s)
Bone Marrow Cells/cytology , Brain/blood supply , Cell Movement/physiology , Endothelial Cells/cytology , Genetic Therapy/methods , Animals , Blood Vessels/cytology , Cell Separation , Female , Flow Cytometry , Genetic Vectors , Immunohistochemistry , Rabbits , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Stem Cells/cytology , Transduction, Genetic , Transgenes
6.
Gene Ther ; 18(7): 682-91, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21368898

ABSTRACT

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.


Subject(s)
Caudate Nucleus , Gene Transfer Techniques , Genetic Vectors , Simian virus 40/genetics , Superoxide Dismutase/genetics , Animals , Brain/metabolism , Glutathione Peroxidase/genetics , Macaca mulatta , Simian virus 40/immunology , Transduction, Genetic , Transgenes
7.
Gene Ther ; 17(2): 227-37, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19890354

ABSTRACT

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.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Genetic Therapy/methods , Genetic Vectors , Simian virus 40/genetics , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytokines/analysis , DNA, Complementary , Feasibility Studies , Lung , Mice , Mice, Knockout , Pseudomonas Infections/therapy , Pseudomonas aeruginosa , Transduction, Genetic
8.
Gene Ther ; 14(22): 1555-63, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17984995

ABSTRACT

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.


Subject(s)
Genetic Therapy/trends , Clinical Trials as Topic , Gene Transfer Techniques/adverse effects , Gene Transfer Techniques/trends , Genetic Therapy/methods , Genetic Vectors , Humans , Neoplasms/therapy , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/trends
9.
Gene Ther ; 14(23): 1650-61, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17914406

ABSTRACT

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.


Subject(s)
AIDS Dementia Complex/prevention & control , Antioxidants/therapeutic use , Genetic Therapy/methods , HIV Envelope Protein gp120/adverse effects , HIV-1 , Neurons/virology , AIDS Dementia Complex/virology , Animals , Apoptosis , Brain/metabolism , Brain/virology , Female , Genetic Vectors/administration & dosage , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Injections , Injections, Intraventricular , Models, Animal , Neurons/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transduction, Genetic/methods
10.
Gene Ther ; 14(12): 939-49, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17443215

ABSTRACT

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.


Subject(s)
Brain/metabolism , Gene Products, nef/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Simian virus 40/genetics , Transduction, Genetic/methods , Animals , Biomarkers/analysis , Brain/virology , Female , Gene Expression , Immunohistochemistry , Injections , Mannitol/pharmacology , Microglia/enzymology , Microglia/virology , Neurons/enzymology , Neurons/virology , Oligopeptides , Osmolar Concentration , Peptides/analysis , Peptides/genetics , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/analysis , Superoxide Dismutase/genetics , Time Factors , Transgenes , nef Gene Products, Human Immunodeficiency Virus
11.
Cancer Gene Ther ; 14(1): 19-29, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16990845

ABSTRACT

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.


Subject(s)
Defective Viruses/physiology , Gene Transfer Techniques , Pancreatic Neoplasms/pathology , Simian virus 40/physiology , Virus Replication , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Polymerase Chain Reaction , Transduction, Genetic
12.
Gene Ther ; 13(23): 1645-56, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16871233

ABSTRACT

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.


Subject(s)
Antioxidants/administration & dosage , Genetic Therapy/methods , HIV Envelope Protein gp120/pharmacology , HIV Infections/therapy , HIV-1 , Transduction, Genetic/methods , Animals , Apoptosis/drug effects , Brain/enzymology , Brain/pathology , Brain/virology , COS Cells , Cell Line , Chlorocebus aethiops , Female , Gene Expression , Genetic Vectors/administration & dosage , Glutathione Peroxidase/genetics , HIV Infections/enzymology , Humans , Immunohistochemistry/methods , In Situ Nick-End Labeling , Injections , Neurons/enzymology , Neurons/pathology , Neurons/virology , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Superoxide Dismutase/genetics , Transgenes
13.
Gene Ther ; 11(22): 1627-37, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15295615

ABSTRACT

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.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , HIV Infections/prevention & control , HIV-1/metabolism , Receptors, CCR5/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Flow Cytometry , Gene Targeting , Genetic Engineering , Genetic Vectors/genetics , HIV Infections/metabolism , Humans , Immunoglobulin Fragments/genetics , Macrophage Inflammatory Proteins/genetics , Macrophages/metabolism , Macrophages/virology , Neuroglia/metabolism , Neuroglia/virology , RNA, Catalytic/genetics , Simian virus 40/genetics , Transgenes
14.
Gene Ther ; 10(26): 2153-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625571

ABSTRACT

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.


Subject(s)
HIV Infections/therapy , HIV-1 , Simian virus 40/genetics , T-Lymphocytes/virology , Transduction, Genetic/methods , alpha 1-Antitrypsin/metabolism , Blotting, Southern , Genetic Therapy/methods , Genetic Vectors , HIV Infections/virology , Humans , Reverse Transcriptase Polymerase Chain Reaction , alpha 1-Antitrypsin/administration & dosage
15.
Gene Ther ; 10(22): 1861-73, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14502215

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/therapy , Genetic Therapy/methods , Hepacivirus/genetics , Liver Neoplasms/therapy , NF-kappa B/immunology , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/virology , Cell Line , Gene Expression , Genetic Vectors/administration & dosage , Hepatocytes/metabolism , Humans , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Liver Neoplasms/immunology , Liver Neoplasms/virology , Mice , Rats , Simian virus 40/genetics , Transduction, Genetic/methods , Transfection/methods , Transgenes , Tumor Cells, Cultured
16.
Gene Ther ; 10(6): 467-77, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12621451

ABSTRACT

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.


Subject(s)
Gene Products, gag/metabolism , Genetic Therapy/methods , HIV Infections/therapy , Protein Precursors/metabolism , Serine Proteinase Inhibitors/genetics , alpha 1-Antitrypsin/genetics , Animals , COS Cells , Cells, Cultured , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HIV Envelope Protein gp160/metabolism , HIV Infections/immunology , Humans , Lymphocytes/immunology , Serine Proteinase Inhibitors/analysis , Serine Proteinase Inhibitors/metabolism , Simian virus 40/genetics , Transduction, Genetic/methods , Virus Replication , alpha 1-Antitrypsin/analysis , alpha 1-Antitrypsin/metabolism , gag Gene Products, Human Immunodeficiency Virus , pol Gene Products, Human Immunodeficiency Virus
17.
Percept Psychophys ; 63(6): 1063-71, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11578050

ABSTRACT

In two priming experiments, we manipulated the perceptual quality of the target or the distractor on the prime trial; the stimuli were repeated or novel. Negative priming was found to be contingent on stimulus repetition, because it was obtained with repeated items but not with novel items. Prime trial perceptual degradation modulated negative priming for repeated items but had no effect on priming in ignored repetition conditions using novel stimuli. These patterns were obtained even when the effect of perceptual degradation was (1) greater than the effect of stimulus repetition and (2) greater for novel words than for repeated words. Although stimulus repetition increases perceptual fluency, the activation of perceptual representations by itself is not sufficient to produce negative priming. Instead, we suggest that negative priming is a manifestation of an activation-sensitive inhibitory mechanism that functions to reduce response competition.


Subject(s)
Attention , Paired-Associate Learning , Pattern Recognition, Visual , Reaction Time , Adolescent , Adult , Color Perception , Female , Humans , Inhibition, Psychological , Male , Psychophysics
18.
J Autism Dev Disord ; 31(3): 257-63, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11518480

ABSTRACT

Earlier investigations have found mixed evidence of working memory impairment in autism. The present study examined working memory in a high-functioning autistic sample, relative to both a clinical control group diagnosed with Tourette Syndrome and a typically developing control group. No group differences were found across three tasks and five dependent measures of working memory. Performance was significantly correlated with both age and IQ. It is concluded that working memory is not one of the executive functions that is seriously impaired in autism. We also suggest that the format of administration of working memory tasks may be important in determining whether or not performance falls in the impaired range.


Subject(s)
Autistic Disorder , Memory/physiology , Adolescent , Child , Female , Humans , Intelligence , Intelligence Tests , Male , Neuropsychological Tests , Space Perception/physiology , Tourette Syndrome
19.
Gene Ther ; 8(13): 1033-42, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438838

ABSTRACT

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.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , HIV-1/physiology , Promoter Regions, Genetic/genetics , Simian virus 40/genetics , Cell Survival/genetics , DNA Polymerase III/genetics , Gene Expression , Genes, tat/genetics , Humans , Lymphocytes/virology , Transduction, Genetic , Transgenes/genetics , Virus Replication/genetics
20.
Gene Ther ; 8(5): 408-18, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11313818

ABSTRACT

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).


Subject(s)
Down-Regulation , Genetic Therapy/methods , HIV Infections/therapy , HIV-1 , Receptors, CXCR4/metabolism , Animals , Cell Line , Genetic Vectors , HIV Infections/metabolism , HIV-1/physiology , Humans , Immunoglobulin Variable Region/genetics , Leukemia Virus, Murine/genetics , Receptors, CXCR4/genetics , Simian virus 40/genetics , T-Lymphocytes/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...