Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Gene Ther ; 8(16): 1207-13, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509953

ABSTRACT

The development of therapies aimed to promote remyelination is a major issue in chronic inflammatory demyelinating disorders of the central nervous system (CNS) such as multiple sclerosis (MS), where the permanent neurological impairment is due to the axonal loss resulting from recurrent episodes of immune-mediated demyelination. Here, we show that the intrathecal injection of a herpes simplex virus (HSV) type-1 replication-defective multigene vector, engineered with the human fibroblast growth factor (FGF)-II gene (TH:bFGF vector), was able to significantly revert in C57BL/6 mice the clinicopathological signs of chronic experimental autoimmune encephalomyelitis (EAE), the animal model of MS. The treatment with the TH:bFGF vector was initiated within 1 week after the clinical onset of EAE and was effective throughout the whole follow-up period (ie 60 days). The disease-ameliorating effect in FGF-II-treated mice was associated with: (1) CNS production of FGF-II from vector-infected cells which were exclusively located around the CSF space (ependymal, choroidal and leptomeningeal cells); (2) significant decrease (P < 0.01) of the number of myelinotoxic cells (T cells and macrophages) both in the CNS parenchyma and in the leptomeningeal space; and (3) significant increase (P < 0.01) of the number of oligodendrocyte precursors and of myelin-forming oligodendrocytes in areas of demyelination and axonal loss. Our results indicate that CNS gene therapy using HSV-1-derived vector coding for neurotrophic factors (ie FGF-II) is a safe and non-toxic approach that might represent a potential useful 'alternative' tool for the future treatment of immune-mediated demyelinating diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Fibroblast Growth Factor 2/genetics , Genetic Therapy/methods , Animals , Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Genetic Vectors/administration & dosage , Injections, Spinal , Lymphocyte Count , Macrophages/immunology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/therapy , Oligodendroglia/pathology , Simplexvirus/genetics , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
2.
J Immunol ; 167(3): 1821-9, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466408

ABSTRACT

The exclusive detrimental role of proinflammatory cytokines in demyelinating diseases of the CNS, such as multiple sclerosis, is controversial. Here we show that the intrathecal delivery of an HSV-1-derived vector engineered with the mouse IFN-gamma gene leads to persistent (up to 4 wk) CNS production of IFN-gamma and inhibits the course of a chronic-progressive form of experimental autoimmune encephalomyelitis (EAE) induced in C57BL/6 mice by myelin oligodendrocyte glycoprotein (MOG)(35-55). Mice treated with the IFN-gamma-containing vector before EAE onset showed an earlier onset but a milder course of the disease compared with control mice treated with the empty vector. In addition, 83% of IFN-gamma-treated mice completely recovered within 25 days post immunization, whereas control mice did not recover up to 60 days post immunization. Mice treated with the IFN-gamma-containing vector within 1 wk after EAE onset partially recovered from the disease within 25 days after vector injection, whereas control mice worsened. Recovery from EAE in mice treated with IFN-gamma was associated with a significant increase of CNS-infiltrating lymphocytes undergoing apoptosis. During the recovery phase, the mRNA level of TNFR1 was also significantly increased in CNS-infiltrating cells from IFN-gamma-treated mice compared with controls. Our results further challenge the exclusive detrimental role of IFN-gamma in the CNS during EAE/multiple sclerosis, and indicate that CNS-confined inflammation may induce protective immunological countermechanisms leading to a faster clearance of encephalitogenic T cells by apoptosis, thus restoring the immune privilege of the CNS.


Subject(s)
Apoptosis/immunology , Brain/immunology , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interferon-gamma/administration & dosage , Lymphocyte Subsets/pathology , Spinal Cord/immunology , Animals , Antigens, CD/biosynthesis , Apoptosis/genetics , Blood-Brain Barrier/genetics , Blood-Brain Barrier/immunology , Brain/metabolism , Brain/pathology , Cell Movement/genetics , Cerebral Ventricles/immunology , Cerebral Ventricles/virology , Chronic Disease , Cisterna Magna , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Genetic Vectors/administration & dosage , Herpesvirus 1, Human/genetics , Injections , Injections, Spinal , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Lymphocyte Count , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor, Type I , Spinal Cord/metabolism , Spinal Cord/pathology , Subarachnoid Space/immunology , Subarachnoid Space/virology , Virus Replication/genetics
3.
Gene Ther ; 8(1): 13-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11402297

ABSTRACT

Multiple sclerosis (MS) is an immune-mediated inflammatory disease of the central nervous system (CNS) that might benefit from anti-inflammatory therapies. However, systemic delivery of anti-inflammatory drugs in MS patients has so far been disappointing, mostly due to the limited capacity of these molecules to enter the CNS. We injected into the cisterna magna (i.c.) of Biozzi AB/H mice affected by a relapsing-remitting form of experimental autoimmune encephalomyelitis (EAE), the animal model of MS, a non-replicative herpes simplex virus (HSV) type-1-derived vector containing the interleukin (IL)-4 gene (d120:LacZ:IL-4). CNS delivery of the d120:LacZ:IL-4 vector, after EAE onset, induced the in situ production of IL-4 by CNS-resident cells facing the cerebrospinal fluid (CSF) spaces and reduced by 47% (P < 0.02) the disease-related deaths. Compared with mice treated with the control d120:lacZ vector, IL-4-treated mice also showed a shorter duration of the first EAE attack, a longer inter-relapse period, and a reduction in the severity and duration of the first relapse. Protection from EAE progression in IL-4-treated mice was associated with activation of microglia in spinal cord areas where mRNA content of the pro-inflammatory chemokines, macrophage chemoattractant protein-1 (MCP-1) and Rantes, was reduced and that of the anti-inflammatory cytokine IL-4 was increased. Finally, CNS-infiltrating mononuclear cells from IL-4-treated mice produced lower levels of MCP-1 mRNA compared with control mice. Our results, showing that IL-4 gene delivery using HSV-1 vectors induces protection from EAE by in situ modulating the cytokine/chemokine-mediated circuits sustaining effector cell functions, indicate that the intrathecal 'therapeutic' use of nonreplicative HSV-1-derived vectors containing anti-inflammatory molecules might represent an alternative strategy in inflammatory diseases of the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Interleukin-4/genetics , Animals , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Cisterna Magna , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Genetic Vectors , Herpesvirus 1, Human/genetics , Injections, Intraventricular , Interleukin-4/biosynthesis , Mice , Mice, Inbred Strains , Microglia/pathology , Multiple Sclerosis/therapy , RNA, Messenger/genetics , Spinal Cord/metabolism
4.
Hum Gene Ther ; 12(8): 905-20, 2001 May 20.
Article in English | MEDLINE | ID: mdl-11387056

ABSTRACT

Systemic administration of antiinflammatory molecules to patients affected by immune-mediated inflammatory demyelinating diseases of the central nervous system (CNS) has limited therapeutic efficacy due to the presence of the blood-brain barrier (BBB). We found that three of five rhesus monkeys injected intrathecally with a replication-defective herpes simplex virus (HSV) type 1-derived vector engineered with the human interleukin 4 (IL-4) gene were protected from an hyperacute and lethal form of experimental autoimmune encephalomyelitis induced by whole myelin. The intrathecally injected vector consistently diffused within the CNS via the cerebrospinal fluid and infected ependymal cells, which in turn sustained in situ production of IL-4 without overt immunological or toxic side effects. In EAE-protected monkeys, IL-4-gene therapy significantly decreased the number of brain as well as spinal cord inflammatory perivenular infiltrates and the extent of demyelination, necrosis, and axonal loss. The protective effect was associated with in situ downregulation of inflammatory mediators such as tumor necrosis factor alpha (TNF-alpha) and monocyte chemoattractant protein 1 (MCP-1), upregulation of transforming growth factor beta (TGF-beta), and preservation of BBB integrity. Our results indicate that intrathecal delivery of HSV-1-derived vectors containing antiinflammatory cytokine genes may play a major role in the future therapeutic armamentarium of inflammatory CNS-confined demyelinating diseases and, in particular, in the most fulminant forms where conventional therapeutic approaches have, so far, failed to achieve a satisfactory control of the disease evolution.


Subject(s)
Autoimmune Diseases/genetics , Central Nervous System/metabolism , Encephalomyelitis/genetics , Genetic Therapy/methods , Genetic Vectors , Herpesvirus 1, Human/genetics , Interleukin-4/genetics , Animals , Autoimmune Diseases/prevention & control , Blood-Brain Barrier , Brain/pathology , Cell Division , Chemokine CCL2/biosynthesis , Cytokines/biosynthesis , Down-Regulation , Encephalomyelitis/prevention & control , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-4/biosynthesis , Macaca mulatta , Magnetic Resonance Imaging , Male , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/immunology , T-Lymphocytes/cytology , Time Factors , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
5.
Gene Ther ; 7(17): 1483-90, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11001368

ABSTRACT

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


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/adverse effects , Gliosarcoma/therapy , Simplexvirus/enzymology , Thymidine Kinase/genetics , Animals , Antiviral Agents/therapeutic use , Apoptosis , Ganciclovir/therapeutic use , Genetic Engineering , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Male , Rats , Rats, Inbred F344 , Transfection , Tumor Cells, Cultured
6.
Gene Ther ; 7(13): 1087-93, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10918474

ABSTRACT

The peripheral delivery of drugs in patients affected by central nervous system (CNS)-confined diseases is therapeutically ineffective due to the presence of the blood-brain barrier which forms an inaccessible wall to the majority of CNS targeting molecules. When molecules with an anti-inflammatory profile have been systemically administered to patients affected by a chronic inflammatory demyelinating disease of the CNS, such as multiple sclerosis (MS), results have been disappointing. A successful therapeutic approach in MS should therefore consider the delivery of anti-inflammatory molecules directly into the CNS in order to inhibit blood-borne CNS-confined mononuclear cells which act as ultimate effector cells directly destroying oligodendrocytes and/or releasing myelinotoxic substances. Biological and physical vectors engineered with heterologous genes coding for immunomodulatory cytokines with an anti-inflammatory profile might represent the appropriate tool to deliver therapeutic genes into the CNS of patients with MS. So far, cytokine gene therapy has never been attempted in MS, but encouraging results have been obtained in the animal model of MS, experimental autoimmune encephalomyelitis (EAE), using viral vectors or plasmids engineered with cytokine genes and then injected systemically, either in the blood stream or circulating encephalitogenic T cells, or into the CNS. Here, we critically discuss the various attempts made in EAE using gene therapy protocols based on the delivery of immunomodulatory cytokine genes. Special emphasis is put on the use of non-replicative herpes simplex type-1 (HSV)-derived vectors engineered with the gene of the immunomodulatory cytokine interleukin (IL)-4.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Herpesvirus 1, Human , Interleukin-4/genetics , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Th1 Cells/immunology , Th2 Cells/immunology
8.
Virology ; 240(2): 245-53, 1998 Jan 20.
Article in English | MEDLINE | ID: mdl-9454698

ABSTRACT

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


Subject(s)
Cytokines/immunology , Defective Viruses/immunology , Herpes Simplex/immunology , Immunization/methods , Simplexvirus/immunology , Viral Vaccines/immunology , Animals , Cytokines/metabolism , Female , Immunity, Mucosal/immunology , Immunity, Mucosal/physiology , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/metabolism , Viral Vaccines/genetics
9.
Gene Ther ; 5(12): 1593-603, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10023438

ABSTRACT

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


Subject(s)
Gene Deletion , Genes, Immediate-Early , Genetic Therapy/methods , Genetic Vectors , Herpesvirus 1, Human/genetics , Neurons/virology , Viral Proteins , Animals , Cells, Cultured , Chlorocebus aethiops , Gene Expression , Gene Transfer Techniques , Gliosarcoma , Herpesvirus 1, Human/pathogenicity , Immediate-Early Proteins/genetics , Neurons/metabolism , Rats , Tumor Cells, Cultured , Vero Cells , Viral Regulatory and Accessory Proteins
10.
Gene Ther ; 5(11): 1517-30, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9930305

ABSTRACT

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


Subject(s)
Genetic Therapy , Genetic Vectors , Simplexvirus/genetics , Defective Viruses/genetics , Humans
12.
Gene Ther ; 4(10): 1120-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9415320

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is a neurotrophic human pathogen that naturally persists in neurons in a latent state and carries a large number of viral functions which can be replaced by foreign genes to create a vector for gene therapy applications. In this report we describe a two-step method for insertion/deletion mutagenesis of HSV genes and the efficient insertion of transgenes into these locations in the viral genome. The first step is the insertion of a reporter gene (lacZ) cassette flanked by Pacl restriction enzyme sites not otherwise found in the viral genome, using standard marker transfer procedures to interrupt a portion of the target HSV gene. The second step is substitution of the reporter gene with other foreign cDNAs by digestion of the vector DNA with Pacl to remove the lacZ gene and subsequent repair of the vector genome by homologous recombination with a transgene expression plasmid. Potential recombinants identified by a 'clear plaque' phenotype after X-gal staining arose at high frequency (80-100%). Of these recombinants containing the transgene in place of the lacZ gene ranged from 19-65%. Insertion of the transgene expression construct into the viral genome eliminates the Pacl sites, allowing this method to be used repeatedly for the sequential deletion of multiple HSV genes while inserting multiple transgenes. This procedure was repeated in succession to produce a vector carrying two independent expression cassettes at distinct viral loci.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Herpesvirus 1, Human/genetics , DNA, Viral/genetics , Genes, Reporter , Humans , Mutagenesis, Insertional , Time Factors , Transgenes
13.
Gene Ther ; 4(11): 1261-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9425451

ABSTRACT

It was previously shown that a tat mutant (tat22) where cysteine 22 is substituted by glycine behaves as a transdominant negative mutant in Jurkat T cells lytically or latently infected by HIV-1. In this study we demonstrate that tat22 controls HIV-1 replication in primary cells. This effect was observed both after in vitro infection of peripheral blood mononuclear cells (PBMCs) from normal donors and after reactivation of the latent infection in PBMCs from seropositive patients. The antiviral effect of tat22 was limited to conditions of low virus production. The use of tat22 may be promising for a gene therapy approach to AIDS during the asymptomatic phase of the disease allowing control of virus replication in infected cells and inhibition of virus spread to uninfected cells.


Subject(s)
Gene Products, tat/genetics , Genetic Therapy/methods , HIV Infections/immunology , HIV-1/physiology , T-Lymphocytes/virology , Virus Replication , HIV Infections/therapy , Humans , Mutation , tat Gene Products, Human Immunodeficiency Virus
14.
Gene Ther ; 3(3): 235-45, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8646555

ABSTRACT

Tat mutants (tat22, tat37 and tat22/37) were constructed in the transactivation domain, where cysteines at positions 22 or/and 37 were substituted with glycine and serine, respectively. These mutants were expressed either in a BK virus episomal vector or in the retroviral vector LXSN. Constitutive production of tat22 by Jurkat T cells in the context of both vectors blocked HIV-1 replication during lytic infection. Conversely, the tat37 mutant did not show any inhibitory activity and tat22/37 displayed a mild effect on HIV-1 infection only when expressed by the recombinant retrovirus. However, constitutive production of tat22/37 by the BK virus vector in Jurkat T cells chronically infected by HIV-1 was effective in blocking reactivation of viral replication induced by tumor necrosis factor-alpha or human herpesvirus-6. These results suggest that mutants in the transactivation domain of tat may be considered in designing alternative strategies to control HIV-1 replication and reactivation from latency during different phases of infection.


Subject(s)
Cysteine/metabolism , Genes, tat , HIV-1/physiology , Virus Activation/genetics , Virus Latency/genetics , Virus Replication/genetics , Base Sequence , Cell Line , Genes, Dominant , HIV-1/genetics , Humans , Molecular Sequence Data , Mutation , Oligodeoxyribonucleotides , Transcriptional Activation
15.
J Med Virol ; 41(4): 289-95, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8106862

ABSTRACT

A BK virus (BKV) expression vector, specific for human cells, was engineered to express antisense human immunodeficiency virus type 1 (HIV-1) tat cDNA (tat-AS) or a tat mutant in cysteine 22 (tat22). Cysteine residues in the cysteine-rich domain of tat are necessary for tat transactivation of the HIV-1 long terminal repeat (LTR). Both the AS tat and the tat mutant significantly inhibited transactivation by tat when assayed in cells cotransfected with an expression vector where the reporter gene for chloramphenicol acetyl transferase was driven by the HIV-1 LTR. Infection of Jurkat cell clones stably expressing tat22 (Jurkat/tat22) or tat-AS (Jurkat/tat-AS) with HIV-1 did not show differences in virus titer in comparison to HIV-1-infected control cells. However, in two Jurkat/tat22 cell clones, entrance of HIV-1 into latency was accelerated significantly and reactivation of HIV-1 from latency induced by tumor necrosis factor-alpha (TNF-alpha) or tat was blocked. These results suggest that, in a combined and integrated approach to the treatment of acquired immunodeficiency syndrome (AIDS), anti-tat genetic therapy could be successfully applied to maintain virus in latency, thereby extending the duration of the asymptomatic phase preceding full-blown AIDS.


Subject(s)
Genes, tat/genetics , HIV-1/genetics , Virus Activation , Virus Latency , BK Virus , Blotting, Northern , CD4 Antigens/analysis , Cell Line, Transformed , Gene Products, tat/genetics , Genetic Vectors , HIV-1/physiology , Humans , Mutagenesis, Site-Directed , Mutation/genetics , RNA, Antisense/genetics , RNA, Viral/analysis , Transcription, Genetic/genetics , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL