Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 10(2): e1003969, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586169

ABSTRACT

The large nucleoporin Nup358/RanBP2 forms eight filaments that project from the nuclear pore into the cytoplasm where they function as docking platforms for nucleocytoplasmic transport receptors. RNAi screens have implicated Nup358 in the HIV-1 life cycle. The 164 C-terminal amino acids of this 3,224 amino acid protein are a cyclophilin homology domain (Nup358Cyp), which has potential to bind the HIV-1 capsid and regulate viral progress to integration. Here we examined the virological role of Nup358 in conditional knockout mouse cells and in RNAi-depleted human CD4⁺ T cells. Cre-mediated gene knockout was toxic and diminished HIV-1 infectivity. However, cellular health and HIV-1 susceptibility were coordinately preserved if, prior to gene inactivation, a transposon was used to express all of Nup358 or only the N-terminal 1340 amino acids that contain three FG repeats and a Ran-binding domain. HIV-1, but not N74D capsid-mutant HIV-1, was markedly sensitive to TNPO3 depletion, but they infected 1-1340 segment-complemented Nup358 knockout cells equivalently. Human and mouse CypA both rescued HIV-1 in CypA gene⁻/⁻ Jurkat cells and TRIM-Nup358Cyp fusions derived from each species were equally antiviral; each also inhibited both WT and N74D virus. In the human CD4⁺T cell line SupT1, abrupt Nup358 depletion reduced viral replication but stable Nup358-depleted cells replicated HIV-1 normally. Thus, human CD4⁺ T cells can accommodate to loss of Nup358 and preserve HIV-1 susceptibility. Experiments with cylosporine, viruses with capsids that do not bind cyclophilins, and growth arrest did not uncover viral dependency on the C-terminal domains of Nup358. Our data reinforce the virological importance of TNPO3 and show that Nup358 supports nuclear transport functions important for cellular homeostasis and for HIV-1 nuclear import. However, the results do not suggest direct roles for the Nup358 cyclophilin or SUMO E3 ligase domains in engaging the HIV-1 capsid prior to nuclear translocation.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/metabolism , HIV-1/metabolism , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cyclophilins/metabolism , Humans , Immunoblotting , Mice , Mice, Knockout , Polymerase Chain Reaction , RNA Interference
2.
J Virol ; 86(4): 2312-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22171279

ABSTRACT

A rodent or other small animal model for HIV-1 has not been forthcoming, with the principal obstacles being species-specific restriction mechanisms and deficits in HIV-1 dependency factors. Some Carnivorans may harbor comparatively fewer impediments. For example, in contrast to mice, the domestic cat genome encodes essential nonreceptor HIV-1 dependency factors. All Feliformia species and at least one Caniformia species also lack a major lentiviral restriction mechanism (TRIM5α/TRIMCyp proteins). Here we investigated cells from two species in another carnivore family, the Mustelidae, for permissiveness to the HIV-1 life cycle. Mustela putorius furo (domesticated ferret) primary cells and cell lines did not restrict HIV-1, feline immunodeficiency virus (FIV), equine infectious anemia virus (EIAV), or N-tropic murine leukemia virus (MLV) postentry and supported late HIV-1 life cycle steps comparably to human cells. The ferret TRIM5α gene exon 8, which encodes the B30.2 domain, was found to be pseudogenized. Strikingly, ferret (but not mink) cells engineered to express human HIV-1 entry receptors supported productive spreading replication, amplification, and serial passage of wild-type HIV-1. Nevertheless, produced virions had relatively reduced infectivity and the virus accrued G→A hypermutations, consistent with APOBEC3 protein pressure. Ferret cell-passaged HIV-1 also evolved amino acid changes in the capsid cyclophilin A binding loop. We conclude that the genome of this carnivore can provide essential nonreceptor HIV-1 dependency factors and that ferret APOBEC3 proteins with activity against HIV-1 are likely. Even so, unlike in cat cells, HIV-1 can replicate in ferret cells without vif substitution. The virus evolves in this novel nonprimate cell adaptive landscape. We suggest that further characterization of HIV-1 adaptation in ferret cells and delineation of Mustelidae restriction factor repertoires are warranted, with a view to the potential for an HIV-1 animal model.


Subject(s)
Biological Evolution , Disease Models, Animal , Ferrets , HIV Infections/virology , HIV-1/physiology , Animals , Cell Line , HIV-1/genetics , Humans , Mink , Molecular Sequence Data , Virus Cultivation , Virus Replication
3.
J Virol ; 85(7): 3570-83, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270171

ABSTRACT

Target cell overexpression of the integrase binding domain (IBD) of LEDGF/p75 (LEDGF) inhibits HIV-1 replication. The mechanism and protein structure requirements for this dominant interference are unclear. More generally, how and when HIV-1 uncoating occurs postentry is poorly defined, and it is unknown whether integrase within the evolving viral core becomes accessible to cellular proteins prior to nuclear entry. We used LEDGF dominant interference to address the latter question while characterizing determinants of IBD antiviral activity. Fusions of green fluorescent protein (GFP) with multiple C-terminal segments of LEDGF inhibited HIV-1 replication substantially, but minimal chimeras of either polarity (GFP-IBD or IBD-GFP) were most effective. Combining GFP-IBD expression with LEDGF depletion was profoundly antiviral. CD4(+) T cell lines were rendered virtually uninfectable, with single-cycle HIV-1 infectivity reduced 4 logs and high-input (multiplicity of infection = 5.0) replication completely blocked. We restricted GFP-IBD to specific intracellular locations and found that antiviral activity was preserved when the protein was confined to the cytoplasm or directed to the nuclear envelope. The life cycle block triggered by the cytoplasm-restricted protein manifested after nuclear entry, at the level of integration. We conclude that integrase within the viral core becomes accessible to host cell protein interaction in the cytoplasm. LEDGF dominant interference and depletion impair HIV-1 integration at distinct postentry stages. GFP-IBD may trigger premature or improper integrase oligomerization.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , HIV Integrase/metabolism , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions , Transcription Factors/immunology , Virus Replication/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
4.
J Virol ; 84(22): 11981-93, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20844042

ABSTRACT

HIV-1 and certain other retroviruses initiate plus-strand synthesis in the center of the genome as well as at the standard retroviral 3' polypurine tract. This peculiarity of reverse transcription results in a central DNA "flap" structure that has been of controversial functional significance. We mutated both HIV-1 flap-generating elements, the central polypurine tract (cPPT) and the central termination sequence (CTS). To avoid an ambiguity of previous studies, we did so without affecting integrase coding. DNA flap formation was disrupted but single-cycle infection was unaffected in all target cells tested, regardless of cell cycle status. Spreading HIV-1 infection was also normal in most T cell lines, and flap mutant viruses replicated equivalently to the wild type in nondividing cells, including macrophages. However, spreading infection of flap mutant HIV-1 was impaired in non-vif-permissive cells (HuT78, H9, and primary human peripheral blood mononuclear cells [PBMCs]), suggesting APOBEC3G (A3G) restriction. Single-cycle infections confirmed that vif-intact flap mutant HIV-1 is restricted by producer cell A3G/F. Combining the Δvif and cPPT-CTS mutations increased A3G restriction synergistically. Moreover, RNA interference knockdown of A3G in HuT78 cells released the block to flap mutant HIV-1 replication. Flap mutant HIV-1 also accrued markedly increased A3G-mediated G→A hypermutation compared to that of wild-type HIV-1 (a full log(10) in the 0.36 kb downstream of the mutant cPPT). We suggest that the triple-stranded DNA structure, the flap, is not the consequential outcome. The salient functional feature is central plus-strand initiation, which functions as a second line of defense against single-stranded DNA editing by A3 proteins that survive producer cell degradation by Vif.


Subject(s)
Cytidine Deaminase/metabolism , Cytosine Deaminase/metabolism , HIV Infections/enzymology , HIV-1/genetics , Purines/metabolism , APOBEC-3G Deaminase , Amino Acid Sequence , Base Sequence , Cell Line , Cytidine Deaminase/genetics , Cytosine Deaminase/genetics , HIV Infections/genetics , HIV Infections/virology , HIV-1/physiology , Humans , Molecular Sequence Data , Mutation , RNA, Viral/genetics , RNA, Viral/metabolism , Reverse Transcription , Virus Replication
5.
J Virol ; 84(14): 7378-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20463079

ABSTRACT

Nonprimate animal models of HIV-1 infection are prevented by missing cellular cofactors and by antiviral actions of species-specific host defense factors. These blocks are profound in rodents but may be less abundant in certain Carnivora. Here, we enabled productive, spreading replication and passage of HIV-1 in feline cells. Feline fibroblasts, T-cell lines, and primary peripheral blood mononuclear cells supported early and late HIV-1 life cycle phases in a manner equivalent to that of human cells, except that produced virions had low infectivity. Stable expression of feline immunodeficiency virus (FIV) Vif-green fluorescent protein (GFP) in HIV-1 entry receptor-complemented feline (CrFK) cells enabled robust spreading HIV-1 replication. FIV Vif colocalized with feline APOBEC3 (fA3) proteins, targeted them for degradation, and prevented G-->A hypermutation of the HIV-1 cDNA by fA3CH and fA3H. HIV-1 Vif was inactive against fA3s as expected and even paradoxically augmented restriction in some assays. In an interesting contrast, simian immunodeficiency virus SIVmac Vif had substantial anti-fA3 activities, which were complete against fA3CH and partial against fA3H. Moreover, both primate lentiviral Vifs colocalized with fA3s and could be pulled down from cell lysates by fA3CH. HIV-1 molecular clones that encode FIV Vif or SIVmac Vif (HIV-1(VF) and HIV-1(VS)) were then constructed. These viruses replicated productively in HIV-1 receptor-expressing CrFK cells and could be passaged serially to uninfected cells. Thus, with the exception of entry receptors, the cat genome can supply the dependency factors needed by HIV-1, and a main restriction can be countered by vif chimerism. The results raise the possibility that the domestic cat could yield an animal model of HIV-1 infection.


Subject(s)
Gene Products, vif/metabolism , HIV-1/physiology , Recombinant Fusion Proteins/metabolism , Virus Replication/physiology , Animals , Cats , Cell Line , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Gene Products, vif/genetics , Genes, vif , HIV Infections/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Leukocytes, Mononuclear/virology , Mutation , Recombinant Fusion Proteins/genetics , T-Lymphocytes/virology , Virus Internalization , vif Gene Products, Human Immunodeficiency Virus/metabolism
6.
PLoS Pathog ; 5(7): e1000522, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19609362

ABSTRACT

LEDGF/p75 can tether over-expressed lentiviral integrase proteins to chromatin but how this underlies its integration cofactor role for these retroviruses is unclear. While a single integrase binding domain (IBD) binds integrase, a complex N-terminal domain ensemble (NDE) interacts with unknown chromatin ligands. Whether integration requires chromatin tethering per se, specific NDE-chromatin ligand interactions or other emergent properties of LEDGF/p75 has been elusive. Here we replaced the NDE with strongly divergent chromatin-binding modules. The chimeras rescued integrase tethering and HIV-1 integration in LEDGF/p75-deficient cells. Furthermore, chromatin ligands could reside inside or outside the nucleosome core, and could be protein or DNA. Remarkably, a short Kaposi's sarcoma virus peptide that binds the histone 2A/B dimer converted GFP-IBD from an integration blocker to an integration cofactor that rescues over two logs of infectivity. NDE mutants were corroborative. Chromatin tethering per se is a basic HIV-1 requirement and this rather than engagement of particular chromatin ligands is important for the LEDGF/p75 cofactor mechanism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chromatin/metabolism , HIV Infections/virology , HIV-1/metabolism , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, Viral/metabolism , Antiviral Agents/metabolism , Base Sequence , Cell Line , Cells, Cultured , DNA-Binding Proteins/metabolism , Green Fluorescent Proteins , HIV Integrase/metabolism , HIV-1/physiology , Histones/metabolism , Humans , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , RNA Interference , Recombinant Fusion Proteins/genetics , Transcription Factors/genetics , Virus Replication
7.
Science ; 314(5798): 461-4, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16959972

ABSTRACT

Chromosomal integration enables human immunodeficiency virus (HIV) to establish a permanent reservoir that can be therapeutically suppressed but not eradicated. Participation of cellular proteins in this obligate replication step is poorly understood. We used intensified RNA interference and dominant-negative protein approaches to show that the cellular transcriptional coactivator lens epithelium-derived growth factor (LEDGF)/p75 (p75) is an essential HIV integration cofactor. The mechanism requires both linkages of a molecular tether that p75 forms between integrase and chromatin. Fractionally minute levels of endogenous p75 are sufficient to enable integration, showing that cellular factors that engage HIV after entry may elude identification in less intensive knockdowns. Perturbing the p75-integrase interaction may have therapeutic potential.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD4-Positive T-Lymphocytes/virology , Chromatin/metabolism , HIV Integrase/metabolism , HIV-1/physiology , Transcription Factors/physiology , Virus Integration , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Humans , Intercellular Signaling Peptides and Proteins/metabolism , RNA Interference , Recombinant Fusion Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Virus Replication
8.
J Cell Sci ; 118(Pt 8): 1733-43, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15797927

ABSTRACT

To investigate the basis for the LEDGF/p75 dependence of HIV-1 integrase (IN) nuclear localization and chromatin association, we used cell lines made stably deficient in endogenous LEDGF/p75 by RNAi to analyze determinants of its location in cells and its ability to interact with IN. Deletion of C-terminal LEDGF/p75 residues 340-417 preserved nuclear and chromatin localization but abolished the interaction with IN and the tethering of IN to chromatin. Transfer of this IN-binding domain (IBD) was sufficient to confer HIV-1 IN interaction to GFP. HRP-2, the only other human protein with an identifiable IBD domain, was found to translocate IN to the nucleus of LEDGF/p75(-) cells. However, in contrast to LEDGF/p75, HRP-2 is not chromatin bound and does not tether IN to chromatin. A single classical nuclear localization signal (NLS) in the LEDGF/p75 N-terminal region ((146)RRGRKRKAEKQ(156)) was found by deletion mapping and was shown to be transferable to pyruvate kinase. Four central basic residues in the NLS are critical for its activity. Strikingly, however, stable expression studies with NLS(+/-) and IBD(+/-) mutants revealed that the NLS, although responsible for LEDGF/p75 nuclear import, is dispensable for stable, constitutive nuclear association of LEDGF/p75 and IN. Both wild-type LEDGF/p75 and NLS-mutant LEDGF/p75 remain entirely chromatin associated throughout the cell cycle, and each tethers IN to chromatin. Thus, these experiments reveal stable nuclear sequestration of a transcriptional regulator by chromatin during the nuclear-cytosolic mixing of cell division, which additionally enables stable tethering of IN to chromatin. LEDGF/p75 is a multidomain adaptor protein that interacts with the nuclear import apparatus, lentiviral IN proteins and chromatin by means of an NLS, an IBD and additional chromatin-interacting domains.


Subject(s)
Chromatin/metabolism , HIV Integrase/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nuclear Localization Signals/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence/physiology , Animals , Avian Leukosis Virus/metabolism , Cats , Cell Cycle/genetics , Chlorocebus aethiops , Chromatin/genetics , DNA-Binding Proteins/metabolism , Genes, Regulator/physiology , HIV-1/metabolism , Humans , Mice , Mutation/genetics , NIH 3T3 Cells , Protein Structure, Tertiary/physiology , Protein Transport/physiology , RNA Interference/physiology , Vero Cells , Virus Integration/physiology
9.
J Virol ; 78(17): 9524-37, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15308744

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1), feline immunodeficiency virus (FIV), and Moloney murine leukemia virus (MoMLV) integrases were stably expressed to determine their intracellular trafficking. Each lentiviral integrase localized to cell nuclei in close association with chromatin while the murine oncoretroviral integrase was cytoplasmic. Fusions of pyruvate kinase to the lentiviral integrases did not reveal transferable nuclear localization signals. The intracellular trafficking of each was determined instead by the transcriptional coactivator LEDGF/p75, which was required for nuclear localization. Stable small interfering RNA expression eliminated detectable LEDGF/p75 expression and caused dramatic, stable redistribution of each lentiviral integrase from nucleus to cytoplasm while the distribution of MoMLV integrase was unaffected. In addition, endogenous LEDGF/p75 coimmunoprecipitated specifically with each lentiviral integrase. In vitro integration assays with preintegration complexes (PICs) showed that endogenous LEDGF/p75 is a component of functional HIV-1 and FIV PICs. However, HIV-1 and FIV infection and replication in LEDGF/p75-deficient cells was equivalent to that in control cells, whether cells were dividing or growth arrested. Two-long terminal repeat circle accumulation in nondividing cell nuclei was also equivalent to that of LEDGF/p75 wild-type cells. Virions produced in LEDGF/p75-deficient cells had normal infectivity. We conclude that LEDGF/p75 fully accounts for cellular trafficking of diverse lentiviral, but not oncoretroviral, integrases and is the main lentiviral integrase-to-chromatin tethering factor. While lentiviral PIC nuclear import is unaffected by LEDGF/p75 knockdown, this protein is a component of functional lentiviral PICs. A role in HIV-1 integration site distribution merits investigation.


Subject(s)
Integrases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lentivirus/chemistry , Lentivirus/enzymology , Oncogenic Viruses/enzymology , Retroviridae/enzymology , Virus Integration/physiology , Active Transport, Cell Nucleus , Cell Line , Chromatin/metabolism , Gene Expression , Gene Products, rev/physiology , HIV-1/chemistry , HIV-1/enzymology , HIV-1/physiology , Humans , Immunodeficiency Virus, Feline/chemistry , Immunodeficiency Virus, Feline/enzymology , Immunodeficiency Virus, Feline/physiology , Integrases/genetics , Introns/genetics , Lentivirus/physiology , Nuclear Localization Signals , Protein Binding , Virus Replication/physiology , rev Gene Products, Human Immunodeficiency Virus
10.
J Virol ; 78(6): 2906-20, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990709

ABSTRACT

The circumstances under which unintegrated lentivirus DNA can persist and be a functional template for transcription and protein expression are not clear. We constructed and validated the first class I (nonpleiotropic) integrase (IN) mutants for a non-human lentivirus (feline immunodeficiency virus [FIV]) and analyzed both these and known class I human immunodeficiency virus type 1 IN mutants. The FIV IN mutants (D66V and D66V/D118A) had class I properties: Gag/Pol precursor expression, proteolytic processing, particle formation, and reverse transcriptase (RT) production were normal, while the transduction of dividing fibroblasts was prevented and integration was blocked. When injected into rat retinas, the wild-type (WT) vector produced extensive, persistent transgene expression, compared with only rare positive neuronal cells for the IN mutant vector. In contrast, both WT and mutant vectors produced entirely equivalent, effective transduction levels of primary rat neurons (retinal ganglion cells). By testing the hypothesis that the unexpected retinal neuron transduction was related to cell cycle status, we found that when fibroblasts were growth arrested, transduction and internally promoted transgene expression were not inhibited at all by the class I FIV or HIV-1 IN mutations. Cells were then transduced under aphidicolin arrest and were released from the block 48 h later. Vector expression was stable and durable during repeated passaging in WT vector-transduced cells, while the release of cells transduced with equivalent RT units of class I IN mutant FIV or HIV vector resulted in a steady decline of expression, from 97 to 0% of cells by day 10. Southern blot and PCR analyses showed a lack of integration, irrespective of cell cycle, for the class I mutants and an increase in one- and two-long terminal repeat circular and linear unintegrated DNAs in growth-arrested cells. We conclude that if cell division is prevented, unintegrated FIV and HIV-1 vector DNAs can produce high-level internally promoted transgene expression equivalent to WT vectors. The expression correlates with the unintegrated DNA levels. These observations may facilitate the study of the roles of IN and other preintegration complex components in preintegration phases of infection by (i) providing an alternative way to monitor unintegrated nuclear cDNA forms, (ii) restricting ascertainment to the transcriptionally functional subset of unintegrated DNA, (iii) enabling analysis in individual, nondividing cells, and (iv) uncoupling other potential functions of IN from integration.


Subject(s)
DNA, Viral , Integrases/genetics , Lentivirus/growth & development , Mutation , Virus Integration , Amino Acid Sequence , Animals , Cats , Cell Line , DNA, Viral/genetics , DNA, Viral/metabolism , HIV Integrase/genetics , HeLa Cells , Humans , Immunodeficiency Virus, Feline/enzymology , Immunodeficiency Virus, Feline/genetics , Immunodeficiency Virus, Feline/growth & development , Integrases/metabolism , Lentivirus/genetics , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/virology , Sequence Alignment , Transduction, Genetic
11.
J Gene Med ; 5(12): 1009-17, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14661176

ABSTRACT

BACKGROUND: In neonatal and adult rodent retina, substantial lentiviral vector expression has been detected primarily in retinal pigment epithelium (RPE), except in very young animals (2-5 days post-natal). In non-retinal tissues, studies of lentiviral vectors have utilized various controls. Among the most stringent are class I integrase mutants, which selectively block the integration reaction while leaving all other gag/pol-encoded functions intact. For HIV-1 vectors injected into brain, these have been used to simultaneously control for pseudotransduction and verify that long-term expression requires integration. Such experiments compare particles that differ only in a single amino acid within a single enzyme that forms a very small molar fraction of the virion. Class I integrase mutants have not been described for feline immunodeficiency virus (FIV) integrase, or tested in the eye for any lentiviral vector. METHODS: We compared subretinally and intravitreally injected FIV vectors and followed animals for up to 7 months, a duration that exceeds prior studies. We also compared the wild-type (WT) vector with one incorporating a single class I amino acid mutation in FIV integrase (D66V). A mock vector (packaging construct absent) was an alternative control. All vectors were vesicular stomatitis virus glycoprotein G (VSV-G)-pseudotyped and were injected on day 7 of life. One group of animals received either subretinal or intravitreal injections of WT vector in the right eyes. Control left eyes were injected with mock vector. These animals were sacrificed at 2 or 7 days post-injection. A second group received subretinal injections of either WT vector or equivalent D66V vector (reverse transcriptase-normalized to WT), and were analyzed after 2, 3 and 7 months. All eyes were scored for marker gene (beta-galactosidase) expression by an observer blinded to vector assignments. RESULTS: Subretinal FIV vector injections were much more effective than intravitreal injections. The RPE was the principal retinal layer transduced by the WT vector, and at least 50% of the area of the retina expressed the marker gene at 3 and 7 months. Occasional cells in inner retinal layers also expressed beta-galactosidase at these time points. The sustained retinal expression produced by subretinally injected vector was blocked by the D66V mutation. CONCLUSIONS: These results show that class I integrase mutant FIV vectors are useful control vectors, and that VSV-G-pseudotyped FIV vectors produce extensive retinal expression for at least 215 days, the longest duration yet reported for lentiviral vectors in retina. Transgene expression is mostly restricted to RPE after post-natal day 7 in rats, suggesting that FIV vectors could be used to target RPE for gene therapy.


Subject(s)
Gene Expression Regulation , Genetic Therapy/methods , Immunodeficiency Virus, Feline/genetics , Integrases/genetics , Pigment Epithelium of Eye/virology , Retina/virology , Transgenes , Age Factors , Animals , Genetic Vectors , Immunodeficiency Virus, Feline/pathogenicity , Mutation , Rats , Rats, Sprague-Dawley
12.
Invest Ophthalmol Vis Sci ; 43(12): 3686-90, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12454037

ABSTRACT

PURPOSE: Feline immunodeficiency virus (FIV)-based lentiviral vectors produce effective genetic modification of the trabecular meshwork (TM) of human eyes in organ-perfusion culture, resulting in high-level expression of a beta-galactosidase marker gene (lacZ) without loss of TM cellularity or architecture. However, effects on aqueous outflow physiology have not been determined, and the ability to monitor FIV vector transgene expression in living TM in situ has not been established. In the current study, transgene expression and outflow facility were evaluated in perfused human anterior segments after FIV vector transduction of lacZ or of a marker gene that can be monitored noninvasively, enhanced green fluorescent protein (eGFP). METHODS: Second-generation FIV vectors were made with a protocol for scaled-up production that requires 10 times less input DNA and allows simplified concentration. One vector encodes beta-galactosidase (vector CT26), and the other (bicistronic) encodes eGFP and neomycin phosphotransferase (vector GiNWF). Three pairs of eyes were injected with 1 x 10(8) transducing units (TU) of CT26 in the right eye and with a control (mock lacZ) vector in the left eye. Three others were injected with 1 x 10(8) TU GiNWF in the right eye only, with the left eye serving as an uninjected control. Intraocular pressure was recorded and transduction efficiency was determined. RESULTS: The modified protocol produced high-titer FIV vectors, and coordinate expression of marker genes was observed with the bicistronic vector. In human eyes, the eGFP and lacZ vectors transduced 79% +/- 15% and 82% +/- 4% of TM cells, respectively, without cell loss compared with control eyes. Transduction and marker gene expression caused a transient decrease of outflow facility (30% +/- 22%, P = 0.02), which resolved after 48 to 72 hours. CONCLUSIONS: FIV vectors produce high-level expression of eGFP in the TM of the cultured human eye, with transduction efficiency similar to that obtained with beta-galactosidase vectors. Transduction and expression of these marker genes results in small and transient changes in outflow facility, suggesting suitability of this class of vectors for glaucoma gene therapy.


Subject(s)
Aqueous Humor/metabolism , Genetic Vectors , Immunodeficiency Virus, Feline/genetics , Luminescent Proteins/metabolism , Trabecular Meshwork/metabolism , beta-Galactosidase/metabolism , Aged , Gene Expression , Genetic Markers , Green Fluorescent Proteins , Humans , Immunoenzyme Techniques , Intraocular Pressure , Luminescent Proteins/genetics , Microscopy, Confocal , Organ Culture Techniques , Plasmids , Transfection , beta-Galactosidase/genetics
13.
J Virol ; 76(18): 9152-64, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12186899

ABSTRACT

Caveolin-1 (Cav-1) is a major protein constituent of caveolae, a type of plasma membrane raft. We observed that coexpression of human Cav-1 with human immunodeficiency virus type 1 (HIV-1) blocked virion production from cells that are ordinarily highly permissive. Further investigation showed that this effect is specific, occurs at low ratios of Cav-1 to HIV-1 DNA, depends on expression of Cav-1 protein, and involves severely impaired expression of HIV-1 proteins. Cav-1 also blocked HIV-2 expression. In contrast, Cav-1 did not inhibit protein expression by a paramyxovirus and did not induce apoptosis or affect cellular morphology, cell viability, or cell cycle progression. Although only small amounts of HIV-1 virions were released from Cav-1-transfected cells, these were fully infectious. Deletion mutagenesis showed that the C-terminal 78 residues were as active as the full-length (178-amino-acid) protein in producing the block. In contrast, the 100 most N-terminal amino acids of Cav-1, which include the previously identified oligomerization and scaffolding domains, were shown to be dispensable. Study of single-amino-acid-exchange mutants of Cav-1 established that palmitoylation was not required. Additional deletion mutants then identified the hydrophobic, membrane-associated domain (residues 101 to 135) as the main determinant. Cellular distribution of wild-type and mutant proteins correlated with ability to block HIV-1 expression. Finally, Cav-2 also blocked HIV-1 expression. These data show that coexpression of caveolins can markedly inhibit expression of HIV proviral DNA and establish that the inhibition is mediated by the hydrophobic, membrane-associated domain.


Subject(s)
Caveolins/metabolism , HIV-1/metabolism , Virion/drug effects , Virion/metabolism , Caveolin 1 , Caveolins/chemistry , Caveolins/genetics , Cell Line , Defective Viruses , Fluorescent Antibody Technique , Gene Deletion , HIV-1/genetics , HIV-2/genetics , HIV-2/metabolism , Humans , Proviruses/genetics , Proviruses/metabolism , Respirovirus/metabolism , Transfection , Viral Proteins/drug effects , Viral Proteins/metabolism , Virion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...