Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nat Methods ; 7(11): 905-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20953176

ABSTRACT

We administered recombinant SV40-derived viral vectors (rSV40s) intravenously to mice with or without prior intraperitoneal injection of mannitol to deliver transgenes to the central nervous system (CNS). We detected transgene-expressing cells (mainly neurons) most prominently in the cortex and spinal cord; prior intraperitoneal mannitol injection increased CNS gene delivery tenfold. Intravenous injection of rSV40s, particularly with mannitol pretreatment, resulted in extensive expression of multiple transgenes throughout the CNS.


Subject(s)
Cerebral Cortex/metabolism , Gene Transfer Techniques , Simian virus 40/genetics , Spinal Cord/metabolism , Transgenes , Animals , COS Cells , Chlorocebus aethiops , Genetic Therapy , Injections, Intravenous , Male , Mice , Mice, Inbred BALB C
2.
Neurobiol Dis ; 45(2): 657-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22036626

ABSTRACT

HIV-associated neurocognitive disorder (HAND) is an increasingly common, progressive disease characterized by neuronal loss and progressively deteriorating CNS function. HIV-1 gene products, particularly gp120 and Tat elicit reactive oxygen species (ROS) that lead to oxidant injury and cause neuron apoptosis. Understanding of, and developing therapies for, HAND requires accessible models of the disease. We have devised experimental approaches to studying the acute and chronic effects of Tat on the CNS. We studied acute exposure by injecting recombinant Tat protein into the caudate-putamen (CP). Ongoing Tat expression, which more closely mimics HIV-1 infection of the brain, was studied by delivering Tat-expression over time using an SV40-derived gene delivery vector, SV(Tat). Both acute and chronic Tat exposure induced lipid peroxidation and neuronal apoptosis. Finally, prior administration of recombinant SV40 vectors carrying antioxidant enzymes, copper/zinc superoxide dismutase (SOD1) or glutathione peroxidase (GPx1), protected from Tat-induced apoptosis and oxidative injury. Thus, injection of recombinant HIV-1 Tat and the expression vector, SV(Tat), into the rat CP cause respectively acute or ongoing apoptosis and oxidative stress in neurons and may represent useful animal models for studying the pathogenesis and, potentially, treatment of HIV-1 Tat-related damage.


Subject(s)
AIDS Dementia Complex/therapy , Antioxidants/administration & dosage , Disease Models, Animal , Genetic Therapy/methods , HIV Infections/therapy , HIV-1 , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , Apoptosis/physiology , Female , Genetic Vectors , HIV Infections/complications , Immunohistochemistry , In Situ Nick-End Labeling , Lipid Peroxidation/physiology , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Simian virus 40/genetics , tat Gene Products, Human Immunodeficiency Virus/administration & dosage
3.
FASEB J ; 25(2): 737-53, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20940264

ABSTRACT

Chemokines may play a role in leukocyte migration across the blood-brain barrier (BBB) during neuroinflammation and other neuropathological processes, such as epilepsy. We investigated the role of the chemokine receptor CCR5 in seizures. We used a rat model based on intraperitoneal kainic acid (KA) administration. Four months before KA injection, adult rats were given femoral intramarrow inoculations of SV (RNAiR5-RevM10.AU1), which carries an interfering RNA (RNAi) against CCR5, plus a marker epitope (AU1), or its monofunctional RNAi-carrying homologue, SV(RNAiR5). This treatment lowered expression of CCR5 in circulating cells. In control rats, seizures induced elevated expression of CCR5 ligands MIP-1α and RANTES in the microvasculature, increased BBB leakage and CCR5(+) cells, as well as neuronal loss, inflammation, and gliosis in the hippocampi. Animals given either the bifunctional or the monofunctional vector were largely protected from KA-induced seizures, neuroinflammation, BBB damage, and neuron loss. Brain CCR5 mRNA was reduced. Rats receiving RNAiR5-bearing vectors showed far greater repair responses: increased neuronal proliferation, and decreased production of MIP-1α and RANTES. Controls received unrelated SV(BUGT) vectors. Decrease in CCR5 in circulating cells strongly protected from excitotoxin-induced seizures, BBB leakage, CNS injury, and inflammation, and facilitated neurogenic repair.


Subject(s)
Chemokine CCL3/metabolism , Chemokine CCL5/metabolism , Kainic Acid/toxicity , Receptors, CCR5/metabolism , Seizures/chemically induced , Animals , Blood-Brain Barrier/drug effects , Brain/cytology , Brain/drug effects , Chemokine CCL3/genetics , Chemokine CCL5/genetics , Female , Gene Expression Regulation/drug effects , Gliosis , Hippocampus/cytology , Hippocampus/drug effects , Ligands , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Neurotoxins/toxicity , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, CCR5/genetics , Regeneration , Seizures/prevention & control
4.
ScientificWorldJournal ; 2012: 482575, 2012.
Article in English | MEDLINE | ID: mdl-22448134

ABSTRACT

The blood-brain barrier (BBB) is compromised in many systemic and CNS diseases, including HIV-1 infection of the brain. We studied BBB disruption caused by HIV-1 envelope glycoprotein 120 (gp120) as a model. Exposure to gp120, whether acute [by direct intra-caudate-putamen (CP) injection] or chronic [using SV(gp120), an experimental model of ongoing production of gp120] disrupted the BBB, and led to leakage of vascular contents. Gp120 was directly toxic to brain endothelial cells. Abnormalities of the BBB reflect the activity of matrix metalloproteinases (MMPs). These target laminin and attack the tight junctions between endothelial cells and BBB basal laminae. MMP-2 and MMP-9 were upregulated following gp120-injection. Gp120 reduced laminin and tight junction proteins. Reactive oxygen species (ROS) activate MMPs. Injecting gp120 induced lipid peroxidation. Gene transfer of antioxidant enzymes protected against gp120-induced BBB abnormalities. NMDA upregulates the proform of MMP-9. Using the NMDA receptor (NMDAR-1) inhibitor, memantine, we observed partial protection from gp120-induced BBB injury. Thus, (1) HIV-envelope gp120 disrupts the BBB; (2) this occurs via lesions in brain microvessels, MMP activation and degradation of vascular basement membrane and vascular tight junctions; (3) NMDAR-1 activation plays a role in this BBB injury; and (4) antioxidant gene delivery as well as NMDAR-1 antagonists may protect the BBB.


Subject(s)
Blood-Brain Barrier , HIV Envelope Protein gp120/physiology , Animals , Basement Membrane/pathology , Brain/blood supply , Cells, Cultured , Endothelium, Vascular/pathology , Glutathione Peroxidase/genetics , HIV-1 , Humans , Matrix Metalloproteinases/biosynthesis , Microvessels/pathology , Oxidative Stress/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Tight Junctions/pathology , Glutathione Peroxidase GPX1
5.
Eur J Neurosci ; 34(12): 2015-23, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22092673

ABSTRACT

Matrix metalloproteinases (MMPs) are implicated in diverse processes, such as neuroinflammation, leakiness of the blood-brain barrier (BBB) and direct cellular damage in neurodegenerative and other CNS diseases. Tissue destruction by MMPs is regulated by their endogenous tissue inhibitors (TIMPs). TIMPs prevent excessive MMP-related degradation of extracellular matrix components. In a rat model of human immunodeficiency virus (HIV)-related encephalopathy, we described MMP-2 and MMP-9 upregulation by HIV-1 envelope gp120, probably via gp120-induced reactive oxygen species. Antioxidant gene delivery blunted gp120-induced MMP production. We also studied the effect of gp120 on TIMP-1 and TIMP-2 production. TIMP-1 and TIMP-2 levels increased 6 h after gp120 injection into rat caudate-putamen (CP). TIMP-1 and TIMP-2 colocalized mainly with neurons (92 and 95%, respectively). By 24 h, expression of these protease inhibitors diverged, as TIMP-1 levels remained high but TIMP-2 subsided. Gene delivery of the antioxidant enzymes Cu/Zn superoxide dismutase or glutathione peroxidase into the CP before injecting gp120 there reduced levels of gp120-induced TIMP-1 and TIMP-2, recapitulating the effect of antioxidant enzymes on gp120-induced MMP-2 and MMP-9. A significant correlation was observed between MMP/TIMP upregulation and BBB leakiness. Thus, HIV-1 gp120 upregulated TIMP-1 and TIMP-2 in the CP. Prior antioxidant enzyme treatment mitigated production of these TIMPs, probably by reducing MMP expression.


Subject(s)
AIDS Dementia Complex/physiopathology , Caudate Nucleus/drug effects , HIV Envelope Protein gp120/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Putamen/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , AIDS Dementia Complex/pathology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiology , Caudate Nucleus/cytology , Caudate Nucleus/physiology , Disease Models, Animal , Female , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , HIV Envelope Protein gp120/genetics , Humans , Putamen/cytology , Putamen/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Up-Regulation
6.
Neurobiol Dis ; 38(2): 313-25, 2010 May.
Article in English | MEDLINE | ID: mdl-20219678

ABSTRACT

HIV-1 effects on the blood-brain barrier (BBB) structure and function are still poorly understood in animal models based on direct administration of recombinant HIV proteins. We therefore injected HIV-1 envelope glycoprotein, gp120, into rat caudate-putamens (CPs) and examined vascular integrity and function. Gp120 coimmunostained with endothelial cell marker, CD31. It induced apoptosis of endothelial cells in vitro and in vivo. BBB function was assessed by administering Evans Blue (EB) intravenously before injecting gp120. EB leaked near the site of gp120 administration. Within 1h after intra-CP gp120 injection, structures positive for endothelial markers ICAM-1 and RECA-1 were greatly decreased. Vascular density assessed by laminin immunostaining remained decreased 1 month after gp120 injection. RECA-1-positive cells expressed hydroxynonenal, a marker of lipid peroxidation and rSV40-mediated gene delivery of antioxidant enzymes protected the BBB from gp120-related injury. Extravasated IgG accumulated following intra-CP SV(gp120) injection, an experimental model of continuing gp120 exposure. Thus: acute and chronic exposure to gp120 disrupts the BBB; gp120-mediated BBB abnormalities are related to lesions of brain microvessels; and gp120 is directly toxic to brain endothelial cells.


Subject(s)
Blood-Brain Barrier/metabolism , Caudate Nucleus/metabolism , Glutathione Peroxidase/genetics , HIV Envelope Protein gp120/administration & dosage , Superoxide Dismutase/genetics , Animals , Antioxidants , Apoptosis/physiology , Blood-Brain Barrier/pathology , Caudate Nucleus/pathology , Cells, Cultured , Female , Fluorescent Antibody Technique , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Glutathione Peroxidase/administration & dosage , Humans , In Situ Nick-End Labeling , Neurons/metabolism , Neurons/pathology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Superoxide Dismutase/administration & dosage
7.
Cerebellum ; 9(4): 587-97, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20700772

ABSTRACT

There are several diseases for which gene transfer therapy to the cerebellum might be practicable. In these studies, we used recombinant Tag-deleted SV40-derived vectors (rSV40s) to study gene delivery targeting the cerebellum. These vectors transduce neurons and microglia very effectively in vitro and in vivo, and so we tested them to evaluate gene transfer to the cerebellum in vivo. Using a rSV40 vector carrying human immunodeficiency virus (HIV)-Nef with a C-terminal FLAG epitope, we characterized the distribution, duration, and cell types transduced. Rats received test and control vectors by stereotaxic injection into the cerebellum. Transgene expression was assessed 1, 2, and 4 weeks later by immunostaining of serial brain sections. FLAG epitope-expressing cells were seen, at all times after vector administration, principally detected in the Purkinje cells of the cerebellum, identified as immunopositive for calbindin. Occasional microglial cells were tranduced; transgene expression was not detected in astrocytes or oligodendrocytes. No inflammatory or other reaction was detected at any time. Thus, SV40-derived vectors can deliver effective, safe, and durable transgene expression to the cerebellum.


Subject(s)
Cerebellum/metabolism , Transduction, Genetic/methods , Animals , Cell Count/methods , Cerebellum/cytology , Female , Gene Expression/physiology , Gene Products, nef/genetics , Gene Products, nef/metabolism , Genetic Vectors/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Statistics, Nonparametric
8.
J Neuropathol Exp Neurol ; 68(5): 456-73, 2009 May.
Article in English | MEDLINE | ID: mdl-19525894

ABSTRACT

Human immunodeficiency virus 1 (HIV-1) encephalopathy is thought to result in part from the toxicity of HIV-1 envelope glycoprotein gp120 for neurons. Experimental systems for studying the effects of gp120 and other HIV proteins on the brain have been limited to the acute effects of recombinant proteins in vitro or in vivo in simian immunodeficiency virus-infected monkeys. We describe an experimental rodent model of ongoing gp120-induced neurotoxicity in which HIV-1 envelope is expressed in the brain using an SV40-derived gene delivery vector, SV(gp120). When it is inoculated stereotaxically into the rat caudate putamen, SV(gp120) caused a partly hemorrhagic lesion in which neuron and other cell apoptosis continues for at least 12 weeks. Human immunodeficiency virus gp120 is expressed throughout this time, and some apoptotic cells are gp120 positive. Malondialdehyde and 4-hydroxynonenal assays indicated that there was lipid peroxidation in these lesions. Prior administration of recombinant SV40 vectors carrying antioxidant enzymes, copper/ zinc superoxide dismutase or glutathione peroxidase, was protective against SV(gp120)-induced oxidative injury and apoptosis. Thus, in vivo inoculation of SV(gp120) into the rat caudate putamen causes ongoing oxidative stress and apoptosis in neurons and may therefore represent a useful animal model for studying the pathogenesis and treatment of HIV-1 envelope-related brain damage.


Subject(s)
AIDS Dementia Complex/etiology , Disease Models, Animal , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp120/metabolism , HIV-1/pathogenicity , AIDS Dementia Complex/genetics , Adenoviruses, Simian , Aldehydes/metabolism , Animals , Calcium-Binding Proteins , Cell Death , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Viral/physiology , Genetic Vectors/administration & dosage , Glial Fibrillary Acidic Protein/metabolism , Glutathione Peroxidase/metabolism , Humans , In Situ Nick-End Labeling , Indoles , Microfilament Proteins , Neurons/pathology , Neurons/virology , Phosphopyruvate Hydratase/metabolism , Putamen/virology , Rats , Rats, Nude , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors , Transduction, Genetic/methods
9.
Neurobiol Dis ; 34(3): 462-76, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19327399

ABSTRACT

Toxicity of HIV-1 envelope glycoprotein (gp120) for substantia nigra (SN) neurons may contribute to the Parkinsonian manifestations often seen in HIV-1-associated dementia (HAD). We studied the neurotoxicity of gp120 for dopaminergic neurons and potential neuroprotection by antioxidant gene delivery. Rats were injected stereotaxically into their caudate-putamen (CP); CP and (substantia nigra) SN neuron loss was quantified. The area of neuron loss extended several millimeters from the injection site, approximately 35% of the CP area. SN neurons, outside of this area of direct neurotoxicity, were also severely affected. Dopaminergic SN neurons (expressing tyrosine hydroxylase, TH, in the SN and dopamine transporter, DAT, in the CP) were mostly affected: intra-CP gp120 caused approximately 50% DAT+ SN neuron loss. Prior intra-CP gene delivery of Cu/Zn superoxide dismutase (SOD1) or glutathione peroxidase (GPx1) protected SN neurons from intra-CP gp120. Thus, SN dopaminergic neurons are highly sensitive to HIV-1 gp120-induced neurotoxicity, and antioxidant gene delivery, even at a distance, is protective.


Subject(s)
Dopamine/metabolism , Glutathione Peroxidase/metabolism , HIV Envelope Protein gp120/toxicity , Neurons/physiology , Superoxide Dismutase/metabolism , Animals , Caudate Nucleus/pathology , Caudate Nucleus/physiopathology , Cell Death , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Genetic Vectors , Glutathione Peroxidase/genetics , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Tyrosine 3-Monooxygenase/metabolism
11.
Virus Res ; 118(1-2): 87-97, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16414141

ABSTRACT

Antiretroviral chemotherapy penetrates the CNS poorly. CNS HIV, thus sheltered, may injure the brain and complicate control of systemic HIV infection. Microglial cells play a major role in HIV persistence in the CNS but are rarely targeted for gene delivery. Because recombinant SV40 vectors (rSV40s) transduce other phagocytic cells efficiently, we tested rSV40 delivery of anti-HIV genetic therapy to microglial cells. Microglia prepared as enriched cultures from human fetal brain, were transduced with marker vectors, SV(RFP) and SV(Nef/FLAG), respectively, carrying DsRed and HIV-1 Nef bearing a FLAG epitope. By immunostaining and FACS, 95% of unselected cells expressed the transgenes, without detectable toxicity. Microglia were transduced with SV(AT), carrying human alpha1-antitrypsin (alpha1AT), which blocks Env and Gag processing. SV(AT)-treated microglia strongly resisted challenge with HIV-1BaL, even when microglia were transduced with SV(AT) following HIV challenge. Thus, rSV40s effectively transduce microglia and protect them from HIV.


Subject(s)
Genetic Therapy/methods , HIV-1/physiology , Microglia/virology , Virus Replication , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , HIV Core Protein p24/analysis , HIV-1/genetics , Humans , Simian virus 40/genetics , Transduction, Genetic , alpha 1-Antitrypsin/genetics
12.
Mol Biotechnol ; 34(2): 257-70, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17172671

ABSTRACT

Among the goals of gene therapy is long-term expression of delivered transgenes. Recombinant Tagdeleted SV40 vectors (rSV40s) are especially well suited for this purpose. rSV40s deliver transgene expression that endures for extended periods of time in tissue culture and in vivo, in both dividing and nondividing cells. These vectors are particularly effective in transducing some cell types that have been almost unapproachable using other gene delivery systems, such as quiescent hematopoietic progenitor cells and their differentiated derivatives. Other cellular targets include neurons, brain microglia, hepatocytes, dendritic cells, vascular endothelium, and others. Because rSV40s do not elicit neutralizing antibodies they are useful for in vivo gene delivery in settings where more than one administration may be desirable. The key characteristics of these vectors include their high production titers and therefore suitability for large cell pools, effectiveness in delivering intracellular proteins, and untranslated RNAs, maintenance of transgene expression at constant levels for extended times, suitability for constitutive or conditional promoters and for combinatorial gene delivery and ability to integrate into genomes of both dividing and nondividing cells.


Subject(s)
Gene Expression , Genetic Therapy/methods , Genetic Vectors/genetics , Simian virus 40/genetics , Simian virus 40/immunology , Animals , Cell Division , Gene Transfer Techniques , Genetic Therapy/adverse effects , Humans , Mutagenesis, Insertional , Virus Integration
13.
World J Stem Cells ; 8(4): 136-57, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27114746

ABSTRACT

Neurogenesis takes place in the adult mammalian brain in three areas: Subgranular zone of the dentate gyrus (DG); subventricular zone of the lateral ventricle; olfactory bulb. Different molecular markers can be used to characterize the cells involved in adult neurogenesis. It has been recently suggested that a population of bone marrow (BM) progenitor cells may migrate to the brain and differentiate into neuronal lineage. To explore this hypothesis, we injected recombinant SV40-derived vectors into the BM and followed the potential migration of the transduced cells. Long-term BM-directed gene transfer using recombinant SV40-derived vectors leads to expression of the genes delivered to the BM firstly in circulating cells, then after several months in mature neurons and microglial cells, and thus without central nervous system (CNS) lesion. Most of transgene-expressing cells expressed NeuN, a marker of mature neurons. Thus, BM-derived cells may function as progenitors of CNS cells in adult animals. The mechanism by which the cells from the BM come to be neurons remains to be determined. Although the observed gradual increase in transgene-expressing neurons over 16 mo suggests that the pathway involved differentiation of BM-resident cells into neurons, cell fusion as the principal route cannot be totally ruled out. Additional studies using similar viral vectors showed that BM-derived progenitor cells migrating in the CNS express markers of neuronal precursors or immature neurons. Transgene-positive cells were found in the subgranular zone of the DG of the hippocampus 16 mo after intramarrow injection of the vector. In addition to cells expressing markers of mature neurons, transgene-positive cells were also positive for nestin and doublecortin, molecules expressed by developing neuronal cells. These cells were actively proliferating, as shown by short term BrdU incorporation studies. Inducing seizures by using kainic acid increased the number of BM progenitor cells transduced by SV40 vectors migrating to the hippocampus, and these cells were seen at earlier time points in the DG. We show that the cell membrane chemokine receptor, CCR5, and its ligands, enhance CNS inflammation and seizure activity in a model of neuronal excitotoxicity. SV40-based gene delivery of RNAi targeting CCR5 to the BM results in downregulating CCR5 in circulating cells, suggesting that CCR5 plays an important role in regulating traffic of BM-derived cells into the CNS, both in the basal state and in response to injury. Furthermore, reduction in CCR5 expression in circulating cells provides profound neuroprotection from excitotoxic neuronal injury, reduces neuroinflammation, and increases neuronal regeneration following this type of insult. These results suggest that BM-derived, transgene-expressing, cells can migrate to the brain and that they become neurons, at least in part, by differentiating into neuron precursors and subsequently developing into mature neurons.

14.
Biochim Biophys Acta ; 1638(3): 197-207, 2003 Jul 30.
Article in English | MEDLINE | ID: mdl-12878320

ABSTRACT

Proprotein processing is essential for HIV infectivity. Cellular trans-Golgi network (TGN) serine proteases (e.g., furin) are required to cleave HIV envelope gp160 to gp120. In addition, HIV protease (PR), an aspartyl protease, cleaves p55(Gag) to p24, etc., in budding virions. alpha1-Antitrypsin (alpha(1)AT) is cleaved by serine proteases, causing a conformational change in alpha(1)AT that sequesters and so inactivates the protease. alpha(1)AT blocks both gp160 and p55 processing, and so is a powerful inhibitor of HIV replication. We hypothesized that alpha(1)AT inhibited gp160 and p55 processing via different mechanisms, and that in both cases, alpha(1)AT bound and was itself cleaved by the proteases whose activities were blocked. alpha(1)AT delivered by SV(AT), a recombinant, Tag-deleted SV40-derived vector, localized to the TGN, co-precipitated with furin, and depleted furin from the TGN. After SV(AT) transduction and HIV challenge, alpha(1)AT was detected in resulting nascent immature HIV-1 virions. alpha(1)AT also blocked incorporation of the enzymatically active dimeric form of PR into HIV virions. Western analysis using recombinant proteins showed that alpha(1)AT directly bound HIV PR, and was cleaved by it. The simultaneous inhibition of two different steps in HIV morphogenesis both increases alpha(1)AT antilentiviral activity and decreases the possibility that HIV mutations will allow escape from inhibition.


Subject(s)
HIV Protease Inhibitors/metabolism , HIV Protease/metabolism , HIV-1/physiology , Subtilisins/antagonists & inhibitors , T-Lymphocytes/virology , alpha 1-Antitrypsin/metabolism , Animals , COS Cells , Furin , Golgi Apparatus/enzymology , HIV Envelope Protein gp160/metabolism , HIV Protease/chemistry , HIV-1/metabolism , HeLa Cells , Humans , Protein Conformation , Subtilisins/analysis , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , Transduction, Genetic , alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/genetics
15.
Curr Gene Ther ; 5(2): 151-65, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15853724

ABSTRACT

The natural function of viruses is to deliver their genetic material to cells. Among the most effective of viruses in doing that is Simian Virus-40 (SV40). The properties that make SV40 a successful virus make it an attractive candidate for use as a gene delivery vehicle: high titer replication, infectivity for almost all nucleated cell types whether the cells are dividing or resting, potential for integration into cellular DNA, a peculiar pathway for entering cells that bypasses the cells' antigen processing apparatus, very high stability, and the apparent ability to activate expression of its own capsid genes in trans. Exploiting these and other characteristics of wild type (wt) SV40, increasing numbers of laboratories are studying recombinant (r) SV40-derived vectors. Among the uses to which these vectors have been applied are: delivering therapy to inhibit HIV, hepatitis C virus (HCV) and other viruses; correction of inherited hepatic and other protein deficiencies; immunizing against lentiviral and other antigens; treatment of inherited and acquired diseases of the central nervous system; protecting the lung and other organs from free radical-induced injury; and many others. The effectiveness of these vectors is a reflection of the adaptive evolution that produced their parent virus, wt SV40. This article explores how and why these vectors work, their strengths and their limitations, and provides a functional model for their exploitation for experimental and clinical applications.


Subject(s)
Genetic Therapy , Genetic Vectors , Simian virus 40/genetics , Animals , Humans , Transduction, Genetic , Transgenes , Virus Replication
16.
Methods Enzymol ; 392: 227-41, 2005.
Article in English | MEDLINE | ID: mdl-15644185

ABSTRACT

The rapid increase in the study of small interfering RNA (siRNA) as a means to decrease expression of targeted genes has led to concerns about possible unexpected consequences of constitutive siRNA expression. We therefore devised a conditional siRNA expression system in which siRNA targeting hepatitis C virus (HCV) would be produced in response to HCV. We found that HCV acts via NFkappaB to stimulate the HIV long terminal repeat (LTR) as a promoter. We exploited this observation by designing conditional siRNA transcription constructs to be triggered by HCV-induced activation of NFkappaB. These were delivered by using highly efficient recombinant Tag-deleted SV40-derived vectors. Conditional activation of HIV-LTR and consequent siRNA synthesis in cells expressing HCV were observed. HCV-specific RNAi decreased HCV RNA greatly within 4 days, using transient transfection of the whole HCV genome as a model of acute HCV entry into transduced cells. We then tested the effectiveness of rSV40-delivered anti-HCV siRNA in cells stably transfected with the whole HCV genome to simulate hepatocytes chronically infected with HCV. There is considerable need for regulated production of siRNAs activated by a particular set of conditions (HCV in this case) but quiescent otherwise. Approaches described here may serve as a paradigm for such conditional siRNA expression.


Subject(s)
RNA Interference , Animals , Base Sequence , Blotting, Northern , Cell Line , DNA Primers , DNA, Complementary , Hepacivirus/genetics , Plasmids , RNA, Viral/analysis , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
J Interferon Cytokine Res ; 23(12): 709-21, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14769147

ABSTRACT

Chronic infection with hepatitis B virus (HBV) has potentially devastating consequences and is very difficult to treat. Therapy with recombinant interferons (IFN), especially IFN-alpha, may be effective. The blood IFN-alpha levels that are needed to maintain therapeutic IFN-alpha levels in the liver, however, often cause severe side effects. Gene delivery to the liver may provide a solution. Using a long-term expression construct could provide the desired levels of IFN locally without the need to maintain potentially problematic blood levels. Recombinant, Tag-deleted SV40-derived vectors transduce hepatocytes efficiently and provide permanent transgene expression. We designed an expression construct that was effective against HBV and whose activity was limited to HBV-infected cells. To do this, we exploited the ability of HBV X protein to activate NF-kappaB and, via NF-kappaB, to activate promoter activity of HIV long terminal repeat (LTR) in hepatocytes. Using HIVLTR as a conditional promoter upstream of human and murine IFN-alpha and IFN-gamma cDNAs, rSV40 vectors were used to test the responsiveness of IFN to HBV and the ability of these IFNs to inhibit HBV transcripts and protein production and to activate IFN signaling in neighboring untransduced cells. We found that in hepatocyte cell lines and in primary hepatocytes, HBV activated the promoter activity of the HIVLTR via NF-kappaB. When whole HBV genome was delivered to cells by transfection to simulate HBV infection, IFN expression was activated, IFNs were produced and secreted, and they protected cells from HBV. Levels of IFN proteins that were secreted in this context were comparable to targeted blood levels needed to control chronic hepatitis viral infection. Further, IFNs that were elicited and secreted in this manner were able to activate IFN-induced signaling pathways in neighboring, untransduced cells and so were likely to provide protection even to cells that the rSV40 vector did not transduce. Gene delivery using such rSV40 vectors expressing IFNs conditionally in response to HBV may be an attractive therapeutic option for the treatment of chronic hepatitis B.


Subject(s)
Genetic Therapy/methods , Hepatitis B virus/physiology , Hepatitis B/genetics , Hepatitis B/therapy , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation , HIV Long Terminal Repeat/genetics , Haplorhini , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon-alpha/genetics , Interferon-gamma/genetics , Liver/metabolism , Liver/pathology , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Signal Transduction , Simian virus 40/genetics
18.
J Interferon Cytokine Res ; 23(9): 477-88, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14565857

ABSTRACT

In human immunodeficiency virus type 1 (HIV-1)-infected individuals, virus-induced production of interferon alpha (IFN-alpha) is impaired. In order to obtain regulated expression of IFN-alpha that responds to HIV-1 infection, a recombinant SV40 vector was designed that carries the human IFN-alpha2 cDNA under the control of the HIV-1 long terminal repeat (LTR) (SV[HIVLTR]IFN). Thus, the IFN-alpha2 gene would be trans-activated on infection with HIV-1. This vector was tested to determine if central nervous system (CNS) cell types that may be potential HIV-1 targets could be transduced and protected from HIV. SV[HIVLTR]IFN transduced NT2 cells, a human neuronal precursor cell line, mature neurons derived from NT2 precursor cells, and human primary monocyte-derived macrophages. IFN-alpha2 expression was retained in mature neurons after SV[HIVLTR]IFN-transduced NT2 precursor cells were induced to differentiate using retinoic acid. IFN-alpha expression was detected only after exposing transduced cells to HIV. Furthermore, SV[HIVLTR]IFN-delivered IFN-alpha2 expression significantly inhibited replication of multiple strains of HIV in both NT2 and NT2-derived mature neurons. SV[HIVLTR]IFN transduction also inhibited HIV-1(BaL) replication in human primary monocyte-derived macrophages. Therefore, we have demonstrated the effectiveness of IFN-alpha2, delivered by an SV40 vector driven by HIV-1 LTR as a promoter, to protect several CNS-based, potentially HIV-susceptible cell types. These findings may have implications for therapy of HIV-1 infection in the CNS.


Subject(s)
Genetic Therapy , Genetic Vectors , HIV Infections/therapy , HIV-1/drug effects , Interferon Type I/genetics , Simian virus 40 , Central Nervous System Diseases/therapy , Humans , Interferon Type I/metabolism , Recombinant Proteins
19.
Oligonucleotides ; 13(5): 281-94, 2003.
Article in English | MEDLINE | ID: mdl-15000819

ABSTRACT

Transducing macrophages and other phagocytic cells has been problematic because these cells are largely nondividing and can phagocytose and degrade viral gene delivery vectors. Because of their carriage of the CCR5 chemokine receptor that functions as a coreceptor for most clinical strains of HIV, these cells are also key targets in early HIV infection and dissemination. We describe here a strategy to transduce these phagocytes, reduce cell membrane CCR5, and protect from infection with R5-tropic HIV. Recombinant Tag-deleted SV40 vectors were used to transduce unselected CCR5-bearing cell lines and primary cells with >98% efficiency. rSV40s were designed to express two different anti-CCR5 small interfering RNAs (siRNAs), driven by the adenoviral VA1 polymerase III (pol III) promoter, which localizes the transcripts in the cytoplasm. Transduction with both siRNAs substantially reduced CCR5 mRNA, which in turn decreased detectable cell membrane CCR5. Both CCR5+ cell lines and primary cells were used: SupT1/CCR5 cells, monocyte-derived macrophages (MDM), and primary human brain microglia. In addition, one siRNA, siRNA R5 #5, was designed to recognize conserved sequences in both murine and human CCR5 mRNA and effectively reduced CCR5 transcript in cells of both species. These siRNAs largely protected CCR5+ cell lines and primary human macrophages and brain microglia from challenge with R5-tropic HIV. Therefore, strategies to target CCR5 using rSV40-delivered, VA promoter-driven siRNAs may be useful therapeutic options for treating HIV infection.


Subject(s)
HIV-1/physiology , Macrophages/immunology , Macrophages/virology , Microglia/immunology , RNA, Small Interfering/genetics , Receptors, CCR5/genetics , Simian virus 40/genetics , Animals , Brain/cytology , COS Cells , Cell Differentiation , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Fetus , Gene Expression Regulation/immunology , Genetic Vectors , Humans , Macrophages/cytology , Mice , Microglia/virology , Monocytes/cytology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Transfection
20.
Curr Opin Mol Ther ; 4(4): 313-23, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12222869

ABSTRACT

The limited success of gene therapy as an approach to treating human disease largely reflects the limitations of the gene delivery vectors that have been used. Poor titers, low transduction efficiency, waning transgene expression and immunogenicity have remained obstacles in the field. As a consequence, much research in normal, immunocompetent animals has not demonstrated therapeutic levels of gene delivery, and results from most human clinical trials have been predictably discouraging. Recombinant gene transfer vectors derived from SV40 virus (rSV40) are potentially of great interest for those working in gene therapy, since these vectors are not subject to many of the problems that have limited gene delivery using other vector systems. rSV40 is made at a very high titer and infects - and so transduces - almost all nucleated cell types very efficiently, regardless of lineage or whether they are resting or dividing; they integrate and are not susceptible to transgene silencing; and they elicit no detectable immune response on the part of normal animals and so can be used to deliver multiple transgenes over time and in sequence. The recent development of 'gutless' rSV40 vectors has expanded the range of potential therapeutic transgenes that can be delivered with this system and added flexibility to the expression configurations that can be accommodated. All of these functional characteristics of SV40-derived vectors have their bases in the biology of SV40 and similar viruses, and have important implications for the potential utility of rSV40 vectors in gene therapeutics. Like all viral gene delivery systems, these vectors have their idiosyncrasies and limitations. They also allow gene delivery that bypasses many of the difficulties that have plagued the field from its inception.


Subject(s)
Genetic Therapy , Genetic Vectors , Simian virus 40 , Animals , Humans , Insecta/genetics , Mice , Transduction, Genetic , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL