Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Virol ; 89(23): 12058-69, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26401032

ABSTRACT

UNLABELLED: Human immunodeficiency virus type 1 (HIV-1) replication requires reverse transcription of its RNA genome into a double-stranded cDNA copy, which is then integrated into the host cell chromosome. The essential steps of reverse transcription and integration are catalyzed by the viral enzymes reverse transcriptase (RT) and integrase (IN), respectively. In vitro, HIV-1 RT can bind with IN, and the C-terminal domain (CTD) of IN is necessary and sufficient for this binding. To better define the RT-IN interaction, we performed nuclear magnetic resonance (NMR) spectroscopy experiments to map a binding surface on the IN CTD in the presence of RT prebound to a duplex DNA construct that mimics the primer-binding site in the HIV-1 genome. To determine the biological significance of the RT-IN interaction during viral replication, we used the NMR chemical shift mapping information as a guide to introduce single amino acid substitutions of nine different residues on the putative RT-binding surface in the IN CTD. We found that six viral clones bearing such IN substitutions (R231E, W243E, G247E, A248E, V250E, and I251E) were noninfectious. Further analyses of the replication-defective IN mutants indicated that the block in replication took place specifically during early reverse transcription. The recombinant INs purified from these mutants, though retaining enzymatic activities, had diminished ability to bind RT in a cosedimentation assay. The results indicate that the RT-IN interaction is functionally relevant during the reverse transcription step of the HIV-1 life cycle. IMPORTANCE: To establish a productive infection, human immunodeficiency virus type 1 (HIV-1) needs to reverse transcribe its RNA genome to create a double-stranded DNA copy and then integrate this viral DNA genome into the chromosome of the host cell. These two essential steps are catalyzed by the HIV-1 enzymes reverse transcriptase (RT) and integrase (IN), respectively. We have shown previously that IN physically interacts with RT, but the importance of this interaction during HIV-1 replication has not been fully characterized. In this study, we have established the biological significance of the HIV-1 RT-IN interaction during the viral life cycle by demonstrating that altering the RT-binding surface on IN disrupts both reverse transcription and viral replication. These findings contribute to our understanding of the RT-IN binding mechanism, as well as indicate that the RT-IN interaction can be exploited as a new antiviral drug target.


Subject(s)
HIV Integrase/metabolism , HIV Reverse Transcriptase/metabolism , HIV-1/physiology , Reverse Transcription/physiology , Virus Replication/physiology , Amino Acid Substitution/genetics , Blotting, Western , DNA Primers/genetics , Escherichia coli , HIV Integrase/genetics , HIV Reverse Transcriptase/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Real-Time Polymerase Chain Reaction
2.
J Virol ; 85(23): 12830-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937660

ABSTRACT

We analyzed xenotropic murine leukemia virus-related virus (XMRV) integration site sequences previously identified from human prostate tissues for single-nucleotide polymorphisms (SNPs) to discriminate between patient and potential cell line sources of the proviruses. The SNPs of two integration sites were identical to those in cell lines but not the patients, whereas the data on the remaining 12 integration sites were inconclusive. Our results provide direct evidence for contamination during analysis of XMRV integration sites.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/virology , Retroviridae Infections/virology , Virus Integration/genetics , Xenotropic murine leukemia virus-related virus/genetics , Animals , Base Sequence , Humans , Male , Mice , Molecular Sequence Data , Proviruses/genetics , Retroviridae Infections/genetics , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured , Xenotropic murine leukemia virus-related virus/isolation & purification , Xenotropic murine leukemia virus-related virus/pathogenicity
3.
Antimicrob Agents Chemother ; 55(11): 5090-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21876045

ABSTRACT

Emergence of drug-resistant mutant viruses during the course of antiretroviral therapy is a major hurdle that limits the success of chemotherapeutic treatment to suppress human immunodeficiency virus type 1 (HIV-1) replication and AIDS progression. Development of new drugs and careful patient management based on resistance genotyping data are important for enhancing therapeutic efficacy. However, identifying changes leading to drug resistance can take years of clinical studies, and conventional in vitro assays are limited in generating reliable drug resistance data. Here we present an efficient in vitro screening assay for selecting drug-resistant variants from a library of randomly mutated HIV-1 strains generated by transposon-directed base-exchange mutagenesis. As a test of principle, we screened a library of mutant HIV-1 strains containing random mutations in the protease gene by using a reporter T-cell line in the presence of the protease inhibitor (PI) nelfinavir (NFV). Analysis of replicating viruses from a single round of infection identified 50 amino acid substitutions at 35 HIV-1 protease residue positions. The selected mutant viruses showed specific resistance to NFV and included most of the known NFV resistance mutations. Therefore, the new assay is efficient for identifying changes leading to drug resistance. The data also provide insights into the molecular mechanisms underlying the development of drug resistance.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/genetics , Cell Line , Drug Resistance, Viral/genetics , Humans , Mutation , Nelfinavir/pharmacology
4.
J Virol ; 84(10): 5181-90, 2010 May.
Article in English | MEDLINE | ID: mdl-20219923

ABSTRACT

After membrane fusion with a target cell, the core of human immunodeficiency virus type 1 (HIV-1) enters into the cytoplasm, where uncoating occurs. The cone-shaped core is composed of the viral capsid protein (CA), which disassembles during uncoating. The underlying factors and mechanisms governing uncoating are poorly understood. Several CA mutations can cause changes in core stability and a block at reverse transcription, demonstrating the requirement for optimal core stability during viral replication. HIV-1 integrase (IN) catalyzes the insertion of the viral cDNA into the host genome, and certain IN mutations are pleiotropic. Similar to some CA mutants, two IN mutants, one with a complete deletion of IN (NL-DeltaIN) and the other with a Cys-to-Ser substitution (NL-C130S), were noninfectious, with a replication block at reverse transcription. Compared to the wild type (WT), the cytoplasmic CA levels of the IN mutants in infected cells were reduced, suggesting accelerated uncoating. The role of IN during uncoating was examined by isolating and characterizing cores from NL-DeltaIN and NL-C130S. Both IN mutants could form functional cores, but the core yield and stability were decreased. Also, virion incorporation of cyclophilin A (CypA), a cellular peptidyl-prolyl isomerase that binds specifically to CA, was decreased in the IN mutants. Cores isolated from WT virus depleted of CypA had an unstable-core phenotype, confirming a role of CypA in promoting optimal core stability. Taken together, our results indicate that IN is required during uncoating for maintaining CypA-CA interaction, which promotes optimal stability of the viral core.


Subject(s)
Cyclophilin A/metabolism , HIV Core Protein p24/metabolism , HIV Integrase/physiology , HIV-1/physiology , Virus Internalization , Virus Replication , Amino Acid Substitution/genetics , Gene Deletion , HIV Integrase/genetics , Humans , Protein Binding
5.
J Virol ; 84(22): 11771-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20844053

ABSTRACT

Retroviral vector-mediated gene therapy has been successfully used to correct genetic diseases. However, a number of studies have shown a subsequent risk of cancer development or aberrant clonal growths due to vector insertion near or within proto-oncogenes. Recent advances in the sequencing technology enable high-throughput clonality analysis via vector integration site (VIS) sequencing, which is particularly useful for studying complex polyclonal hematopoietic progenitor/stem cell (HPSC) repopulation. However, clonal repopulation analysis using the current methods is typically semiquantitative. Here, we present a novel system and standards for accurate clonality analysis using 454 pyrosequencing. We developed a bidirectional VIS PCR method to improve VIS detection by concurrently analyzing both the 5' and the 3' vector-host junctions and optimized the conditions for the quantitative VIS sequencing. The assay was validated by quantifying the relative frequencies of hundreds of repopulating HPSC clones in a nonhuman primate. The reliability and sensitivity of the assay were assessed using clone-specific real-time PCR. The majority of tested clones showed a strong correlation between the two methods. This assay permits high-throughput and sensitive assessment of clonal populations and hence will be useful for a broad range of gene therapy, stem cell, and cancer research applications.


Subject(s)
Hematopoietic Stem Cells/virology , High-Throughput Screening Assays/methods , Lentivirus/physiology , Sequence Analysis, DNA/methods , Virus Integration , Animals , Cells, Cultured , Clone Cells , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/physiology , Hematopoietic Stem Cells/cytology , Lentivirus/genetics , Macaca mulatta
6.
J Virol ; 83(13): 6522-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19369352

ABSTRACT

The ability to traverse an intact nuclear envelope and productively infect nondividing cells is a salient feature of human immunodeficiency virus type 1 (HIV-1) and other lentiviruses, but the viral factors and mechanism of nuclear entry have not been defined. HIV-1 integrase (IN) is implicated to play a role in the nuclear import of the virus, but the cellular pathway for IN trafficking and the role of IN in mediating the nuclear import of viral particles are unknown. Using a semipermeabilized cell assay, we observed that the nuclear import of IN was not the result of passive diffusion but occurred independently of cytosolic factors, metabolic energy, and the classical receptor-mediated, Ran-dependent import pathways. To determine if IN enters the nucleus by interacting with the nucleopore complex (NPC), we found that IN bound directly with the FxFG-rich C-terminal domain of nucleoporin 153 (NUP153C). When added in excess to the import assay, NUP153C inhibited the nuclear import of IN. Known binding partners of NUP153C competed with IN for binding with NUP153 and also inhibited the nuclear import of IN. In cultured cells, overexpression of NUP153C reduced the infectivity of an HIV-derived vector by interfering with the nuclear translocation of the viral cDNA. These results support a functional role for the IN-NUP153 interaction in HIV-1 replication and suggest that HIV-1 subviral particles gain access to the nucleus by interacting directly with the NPC via the binding of particle-associated IN to NUP153C.


Subject(s)
Cell Nucleus/virology , HIV Integrase/metabolism , HIV-1/physiology , Nuclear Pore Complex Proteins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , HIV Infections/virology , HeLa Cells , Humans , Virus Replication
7.
J Virol ; 83(12): 6222-33, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369341

ABSTRACT

Until very recently, quiescent CD4(+) T cells were thought to be resistant to human immunodeficiency virus (HIV) infection. Subsequent studies, attempting to fully elucidate the mechanisms of resistance, showed that quiescent cells could become infected by HIV at low efficiency and form a latently infected population. In this study, we set out to identify the sites of viral integration and to assess the efficiency of the overall integration process in quiescent cells. Based on our results, HIV integration in quiescent CD4(+) T cells occurs in sites similar to those of their prestimulated counterparts. While site selections are similar, the integration process in quiescent cells is plagued by the formation of high levels of incorrectly processed viral ends and abortive two-long-terminal-repeat circles.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV/physiology , Virus Integration , CD4-Positive T-Lymphocytes/cytology , Cell Line , Chromosome Mapping , DNA, Viral , HIV/genetics , HIV Long Terminal Repeat , Humans , Lymphocyte Activation , Sequence Analysis, DNA
8.
Methods ; 47(4): 269-76, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19186211

ABSTRACT

During the life cycle of retroviruses, establishment of a productive infection requires stable joining of a DNA copy of the viral RNA genome into host cell chromosomes. Retroviruses are thus promising vectors for the efficient and stable delivery of genes in therapeutic protocols. Integration of retroviral DNA is catalyzed by the viral enzyme integrase (IN), and one salient feature of retroviral DNA integration is its lack of specificity, as many chromosomal sites can serve as targets for integration. Despite the promise for success in the clinic, one major drawback of the retrovirus-based vector is that any unintended insertion events from the therapy can potentially lead to deleterious effects in patients, as demonstrated by the development of malignancies in both animal and human studies. One approach to directing integration into predetermined DNA sites is fusing IN to a sequence-specific DNA-binding protein, which results in a bias of integration near the recognition site of the fusion partner. Encouraging results have been generated in vitro and in vivo using fusion protein constructs of human immunodeficiency virus type 1 IN and E2C, a designed polydactyl zinc-finger protein that specifically recognizes an 18-base pair DNA sequence. This review focuses on the method for preparing infectious virions containing the IN fusion proteins and on the quantitative PCR assays for determining integration site specificity. Efforts to engineer IN to recognize specific target DNA sequences within the genome may lead to development of effective retroviral vectors that can safely deliver gene-based therapeutics in a clinical setting.


Subject(s)
DNA, Viral/metabolism , HIV Integrase/metabolism , Mutagenesis, Site-Directed/methods , Viral Fusion Proteins/metabolism , Virus Integration/physiology , Zinc Fingers/physiology , Animals , DNA, Viral/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HIV Integrase/genetics , HIV-1/enzymology , HIV-1/genetics , Humans , Viral Fusion Proteins/genetics
9.
J Virol ; 82(20): 9964-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18684813

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) is a new human gammaretrovirus identified in prostate cancer tissue from patients homozygous for a reduced-activity variant of the antiviral enzyme RNase L. Neither a casual relationship between XMRV infection and prostate cancer nor a mechanism of tumorigenesis has been established. To determine the integration site preferences of XMRV and the potential risk of proviral insertional mutagenesis, we carried out a genome-wide analysis of viral integration sites in the prostate cell line DU145 after an acute XMRV infection and compared the integration site pattern of XMRV with those found for murine leukemia virus and two human retroviruses, human immunodeficiency virus type 1 and human T-cell leukemia virus type 1. Among all retroviruses analyzed, XMRV has the strongest preference for transcription start sites, CpG islands, DNase-hypersensitive sites, and gene-dense regions; all are features frequently associated with structurally open transcription regulatory regions of a chromosome. Analyses of XMRV integration sites in tissues from prostate cancer patients found a similar preference for the aforementioned chromosomal features. Additionally, XMRV integration sites in cancer tissues were associated with cancer breakpoints, common fragile sites, microRNA, and cancer-related genes, suggesting a selection process that favors certain chromosomal integration sites. In both acutely infected cells and cancer tissues, no common integration site was detected within or near proto-oncogenes or tumor suppressor genes. These results are consistent with a model in which XMRV may contribute to tumorigenicity via a paracrine mechanism.


Subject(s)
Gammaretrovirus/metabolism , Leukemia Virus, Murine/metabolism , Prostatic Neoplasms/virology , Virus Integration , Animals , Cell Line , Chromosome Mapping , CpG Islands , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gammaretrovirus/genetics , Humans , Leukemia Virus, Murine/genetics , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Data , Prostatic Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Retroviridae Infections/metabolism , Transcription, Genetic
10.
Br J Pharmacol ; 175(8): 1241-1259, 2018 04.
Article in English | MEDLINE | ID: mdl-28910489

ABSTRACT

BACKGROUND AND PURPOSE: Cardiovascular disease associated with antiretroviral therapy (ART) has become a major clinical challenge for HIV-positive patients. However, the role of ART in blood vessel growth is largely unknown. Here, we examined an integral component of ART, nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) and investigated their effects on key microvascular functions, including angiogenesis and lymphangiogenesis. EXPERIMENTAL APPROACH: The angiogenesis/lymphangiogenesis capability of endothelial cells (ECs) was evaluated using migration, proliferation and tube formation assays in vitro, and mouse ear and Matrigel plug assays in vivo. Expressions of signalling molecules and mitochondrial antioxidant catalases were determined using Western blotting. Receptor tyrosine kinase (RTK) internalization and endocytosis were examined using flow cytometry and confocal immunofluorescence microscopy respectively. Mitochondrial DNA copy number and ROS were determined using quantitative real-time PCR and MitoSOX staining respectively. KEY RESULTS: Pharmaceutical doses of NRTIs [azidothymidine (AZT), tenofovir disoproxil fumarate (TDF) and lamivudine (3TC)] inhibited angiogenesis and lymphangiogenesis both in vivo and in vitro by affecting the proliferation and migration of ECs. Correspondingly, NRTIs selectively attenuated the activation and transduction of endothelial RTK signals, VEGFR2 and FGFR1 pathways, in vascular ECs and the VEGFR3 pathway in lymphatic ECs. Both TDF and 3TC restrained RTKs' endocytosis into early endosomes but not internalization, while AZT blocked the protein maturation of RTKs. Excessive ROS levels were detected in NRTI-treated ECs, and the MnSOD mimic MnTMPyP alleviated the angiogenic/lymphangiogenic defects induced by NRTIs. CONCLUSIONS AND IMPLICATIONS: NRTIs negatively regulate angiogenesis and lymphangiogenesis by inducing mitochondrial oxidative stress and subsequently impairing RTK signalling in ECs. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Nucleosides/pharmacology , Nucleotides/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/physiology , Humans , Lymphangiogenesis/drug effects , Neovascularization, Pathologic/drug therapy
11.
Curr HIV Res ; 3(4): 377-92, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16250884

ABSTRACT

Live attenuated HIV vaccines offer a means to introduce exogenous sequences into the viral genome to target the virus elimination in vivo. Foreign genes inserted into the nef region of HIV-1 NL4-3 were found to be rapidly deleted following virus infection and/or replication, in a size dependent manner, in the human fetal Thymus/Liver implants of severe combined immunodeficient mouse (SCID-hu) model. When the murine heat stable antigen (HSA) of 283 bp was substituted into HIV-1 nef region, the viral loads in vivo were comparable to the negative control nef attenuated HIV-1, and the reporter HSA gene was not deleted upon infection. However, the murine Thy1.2 gene (505 bp) substituted into the nef attenuated HIV-1, upon infection and replication, deleted 441 bp in vitro and 437 bp in vivo, of the inserted Thy1.2 gene. When the enhanced green fluorescence protein (eGFP) gene (720 bp) was substituted for nef, virus replication was aborted in vivo in the Thy/Liv implants, as seen by the background levels of viral loads, comparable to mock infected implants, and the eGFP gene was deleted. When the herpes simplex virus thymidine kinase gene, HSV-TK (1.15 kbp), or HSA gene, was substituted into the viral vpr gene, TK but not HSA gene was deleted, upon infection in vitro. Moreover, NL-TKI reporter virus with both intact nef and vpr genes shows deletion of TK gene both in vitro and in vivo. Excision of foreign genes occurred within the exogenous segments but not in the viral own regions. These results suggest that larger "suicide" genes introduced via HIV-1 can be deleted upon infection. However, smaller size nucleotide sequences or genes (approximately 300 bp) inserted in place of viral nef or vpr gene may be used to target the virus or its components, for attack and elimination in vivo, and thus have implications for the development of live attenuated HIV vaccines.


Subject(s)
AIDS Vaccines/genetics , HIV Infections/virology , HIV-1/genetics , Sequence Deletion , AIDS Vaccines/immunology , Animals , Blotting, Western , CD24 Antigen/genetics , Genes, Reporter , Genes, nef/genetics , Genes, vpr/genetics , Green Fluorescent Proteins/genetics , Mice , Mice, SCID , Thy-1 Antigens/genetics , Thymidine Kinase/genetics , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virus Replication
12.
Cell Stem Cell ; 14(4): 473-85, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24702996

ABSTRACT

In mice, clonal tracking of hematopoietic stem cells (HSCs) has revealed variations in repopulation characteristics. However, it is unclear whether similar properties apply in primates. Here, we examined this issue through tracking of thousands of hematopoietic stem and progenitor cells (HSPCs) in rhesus macaques for up to 12 years. Approximately half of the clones analyzed contributed to long-term repopulation (over 3-10 years), arising in sequential groups and likely representing self-renewing HSCs. The remainder contributed primarily for the first year. The long-lived clones could be further subdivided into functional groups contributing primarily to myeloid, lymphoid, or both myeloid and lymphoid lineages. Over time, the 4%-10% of clones with robust dual lineage contribution predominated in repopulation. HSPCs expressing a CCR5 shRNA transgene behaved similarly to controls. Our study therefore documents HSPC behavior in a clinically relevant model over a long time frame and provides a substantial system-level data set that is a reference point for future work.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Tracking , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Myeloid Cells/cytology , Animals , Cells, Cultured , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , Macaca mulatta , Mice , Myeloid Cells/metabolism , RNA, Small Interfering/genetics , Receptors, CCR5/chemistry , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Transgenes/physiology
13.
Immunol Res ; 48(1-3): 27-39, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20717743

ABSTRACT

Xenotropic murine leukemia virus-related virus (XMRV) is a new human retrovirus originally identified in prostate cancer patients with a deficiency in the antiviral enzyme RNase L. XMRV has been detected with varying frequencies in cases of prostate cancer and chronic fatigue syndrome (CFS), as well as in a small proportion of healthy individuals. An etiologic link between XMRV infection and human disease, however, has yet to be established. Here, we summarize existing knowledge regarding the characteristics of XMRV replication, association of XMRV with prostate cancer and CFS, and potential mechanisms of XMRV pathophysiology. We also highlight several areas, such as the establishment of standardized assays and the development of animal models, as future directions to advance our current understanding of XMRV and its relevance to human disease.


Subject(s)
Fatigue Syndrome, Chronic/epidemiology , Prostatic Neoplasms/epidemiology , Retroviridae Infections/epidemiology , Retroviridae Proteins, Oncogenic/metabolism , Xenotropic murine leukemia virus-related virus/physiology , Animals , Disease Models, Animal , Fatigue Syndrome, Chronic/genetics , Genetic Predisposition to Disease , Humans , Male , Mice , Prostatic Neoplasms/genetics , Retroviridae Infections/genetics , Retroviridae Proteins, Oncogenic/genetics , Virus Replication , Xenotropic murine leukemia virus-related virus/pathogenicity
14.
Nucleus ; 1(1): 18-22, 2010.
Article in English | MEDLINE | ID: mdl-21327100

ABSTRACT

The ability to traverse an intact nuclear envelope and productively infect non-dividing cells is a salient feature of human immunodeficiency virus type 1 (HIV-1) and other lentiviruses, but the viral factors and mechanism of nuclear entry have not been defined. We have recently reported a functional role for the nucleoporin NUP153 in the nuclear import of the HIV-1 preintegration complex (PIC). Our findings suggest that HIV-1 sub-viral particles gain access to the nucleus by interacting directly with the nuclear pore complex (NPC) via the binding of PIC-associated integrase (IN) to the C-terminal domain of NUP153. This article discusses how NPC conformation and constitution might influence nuclear import of the PIC, and the subsequent integration of the viral cDNA into actively transcribed genes.


Subject(s)
HIV-1/physiology , Nuclear Pore/metabolism , Virus Replication , Active Transport, Cell Nucleus , HIV Integrase/metabolism , Humans , Models, Molecular , Nuclear Pore Complex Proteins/metabolism , Protein Binding
15.
Immunol Res ; 48(1-3): 14-26, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20721640

ABSTRACT

An early and critical event of the human immunodeficiency virus type 1 (HIV-1) life cycle is uncoating of the viral core. Uncoating involves the disassembly of HIV-1 capsid (CA). The underlying mechanisms governing uncoating are poorly defined, and the role of viral and host factors in uncoating is not well understood. Cyclophilin A and TRIM5α are two cellular factors that interact with CA in exerting their effects on HIV-1 replication. Here, we review the current understanding of uncoating and the new functional role of HIV-1 IN during uncoating.


Subject(s)
Carrier Proteins/metabolism , Cyclophilin A/metabolism , HIV Infections/immunology , HIV Integrase/metabolism , HIV-1/physiology , Virus Integration , Antiviral Restriction Factors , Capsid Proteins/metabolism , HIV Infections/virology , HIV-1/pathogenicity , Host-Pathogen Interactions , Humans , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Viral Core Proteins/metabolism , Virus Replication
16.
PLoS One ; 5(4): e10255, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20421928

ABSTRACT

Xenotropic murine leukemia virus (MLV)-related virus (XMRV) is a new human retrovirus associated with prostate cancer and chronic fatigue syndrome. The causal relationship of XMRV infection to human disease and the mechanism of pathogenicity have not been established. During retrovirus replication, integration of the cDNA copy of the viral RNA genome into the host cell chromosome is an essential step and involves coordinated joining of the two ends of the linear viral DNA into staggered sites on target DNA. Correct integration produces proviruses that are flanked by a short direct repeat, which varies from 4 to 6 bp among the retroviruses but is invariant for each particular retrovirus. Uncoordinated joining of the two viral DNA ends into target DNA can cause insertions, deletions, or other genomic alterations at the integration site. To determine the fidelity of XMRV integration, cells infected with XMRV were clonally expanded and DNA sequences at the viral-host DNA junctions were determined and analyzed. We found that a majority of the provirus ends were correctly processed and flanked by a 4-bp direct repeat of host DNA. A weak consensus sequence was also detected at the XMRV integration sites. We conclude that integration of XMRV DNA involves a coordinated joining of two viral DNA ends that are spaced 4 bp apart on the target DNA and proceeds with high fidelity.


Subject(s)
Leukemia Virus, Murine/genetics , Virus Integration/genetics , Cell Line , DNA, Viral/genetics , Fatigue Syndrome, Chronic , Female , Humans , Male , Prostatic Neoplasms , Retroviridae Infections/virology , Sequence Analysis, DNA , Tumor Virus Infections/virology
17.
J Biol Chem ; 284(12): 7931-9, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19150986

ABSTRACT

Integrase (IN) from human immunodeficiency virus, type 1 (HIV-1) exerts pleiotropic effects in the viral replication cycle. Besides integration, IN mutations can impact nuclear import, viral maturation, and reverse transcription. IN and reverse transcriptase (RT) interact in vitro, and the IN C-terminal domain (CTD) is both necessary and sufficient for binding RT. We used nuclear magnetic resonance spectroscopy to identify a putative RT-binding surface on the IN CTD, and surface plasmon resonance to obtain kinetic parameters and the binding affinity for the IN-RT interaction. An IN K258A substitution that disrupts reverse transcription in infected cells is located at the putative RT-binding surface, and we found that this substitution substantially weakens IN CTD-RT interactions. We also identified two additional IN amino acid substitutions located at the putative RT-binding surface (W243E and V250E) that significantly impair viral replication in tissue culture. These results strengthen the notion that IN-RT interactions are biologically relevant during HIV-1 replication and also provide insights into this interaction at the molecular level.


Subject(s)
HIV Integrase/chemistry , HIV Reverse Transcriptase/chemistry , HIV-1/physiology , Reverse Transcription/physiology , Virus Replication/physiology , Amino Acid Substitution , Binding Sites/physiology , Cell Line , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Humans , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Peptide Mapping , Protein Binding/physiology , Protein Structure, Tertiary/physiology
18.
J Virol ; 81(18): 10037-46, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17626089

ABSTRACT

Reverse transcriptase (RT) and integrase (IN) are two essential enzymes that play a critical role in synthesis and integration of the retroviral cDNA, respectively. For human immunodeficiency virus type 1 (HIV-1), RT and IN physically interact and certain mutations and deletions of IN result in viruses defective in early steps of reverse transcription. However, the mechanism by which IN affects reverse transcription is not understood. We used a cell-free reverse transcription assay with different primers and compositions of deoxynucleoside triphosphates to differentially monitor the effect of IN on the initiation and elongation modes of reverse transcription. During the initiation mode, addition of IN stimulated RT-catalyzed reverse transcription by fourfold. The stimulation was specific to IN and could not be detected when the full-length IN was replaced with truncated IN derivatives. The IN-stimulated initiation was also restricted to the template-primer complex formed using tRNA(3)(Lys) or short RNA oligonucleotides as the primer and not those formed using DNA oligonucleotides as the primer. Addition of IN also produced a threefold stimulation during the elongation mode, which was not primer dependent. The stimulation of both initiation and elongation by IN was retained in the presence of an RT trap. Furthermore, IN had no effect on steps at or before template-primer annealing, including packaging of viral genomic RNA and tRNA(3)(Lys). Taken together, our results showed that IN acts at early steps of reverse transcription by increasing the processivity of RT and suppressing the formation of the pause products.


Subject(s)
HIV Integrase/metabolism , HIV Reverse Transcriptase/metabolism , HIV-1/metabolism , Reverse Transcription/physiology , Virus Replication/physiology , Catalysis , Cell-Free System/chemistry , Cell-Free System/metabolism , Cell-Free System/virology , DNA Primers/chemistry , DNA Primers/metabolism , HIV Integrase/chemistry , HIV Reverse Transcriptase/chemistry , HIV-1/chemistry , Humans , Nucleotides/chemistry , Nucleotides/metabolism , RNA, Transfer, Lys/chemistry , RNA, Transfer, Lys/metabolism , Virus Integration/physiology
19.
J Virol ; 81(7): 3574-82, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17229711

ABSTRACT

Unlike activated T cells, quiescent CD4+ T cells have shown resistance to human immunodeficiency virus (HIV) infection due to a block in the early events of the viral life cycle. To further investigate the nature of this block, we infected quiescent CD4+ T cells with HIV-1(NL4-3) and immediately stimulated them. Compared to activated (prestimulated) cells, these poststimulated cells showed slightly decreased viral entry and delays in the completion of reverse transcription. However, the relative efficiency of integration was similar to that of prestimulated cells. Together, this resulted in decreased expression of tat/rev mRNA and synthesis of viral protein. Furthermore, based on cell cycle staining and BrdU incorporation, poststimulated cells expressing viral protein failed to initiate a second round of their cell cycle, independently of Vpr-mediated arrest. Together, these data demonstrate that the early stages of the HIV life cycle are inefficient in these poststimulated cells and that efficient replication cannot be induced by subsequent activation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV-1/physiology , Lymphocyte Activation/immunology , Virus Replication , CD4-Positive T-Lymphocytes/cytology , Cell Line , Cell Proliferation , Gene Expression Regulation, Viral , Gene Products, gag/metabolism , Gene Products, vpr/metabolism , Humans , Kinetics , RNA, Viral/genetics , Reverse Transcription/genetics , Virus Internalization , vpr Gene Products, Human Immunodeficiency Virus
20.
Proc Natl Acad Sci U S A ; 104(5): 1655-60, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17234809

ABSTRACT

We recently reported identification of a previously undescribed gammaretrovirus genome, xenotropic murine leukemia virus-related virus (XMRV), in prostate cancer tissue from patients homozygous for a reduced activity variant of the antiviral enzyme RNase L. Here we constructed a full-length XMRV genome from prostate tissue RNA and showed that the molecular viral clone is replication-competent. XMRV replication in the prostate cancer cell line DU145 was sensitive to inhibition by IFN-beta. However, LNCaP prostate cancer cells, which are deficient in JAK1 and RNase L, were resistant to the effects of IFN-beta against XMRV. Furthermore, DU145 cells rendered deficient in RNase L with siRNA were partially resistant to IFN inhibition of XMRV. Expression in hamster cells of the xenotropic and polytropic retrovirus receptor 1 allowed these cells to be infected by XMRV. XMRV provirus integration sites were mapped in DNA isolated from human prostate tumor tissue to genes for two transcription factors (NFATc3 and CREB5) and to a gene encoding a suppressor of androgen receptor transactivation (APPBP2/PAT1/ARA67). Our studies demonstrate that XMRV is a virus that has infected humans and is susceptible to inhibition by IFN and its downstream effector, RNase L.


Subject(s)
Interferons/metabolism , Prostatic Neoplasms/genetics , Retroviridae/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Endoribonucleases/metabolism , Humans , Janus Kinase 1/metabolism , Male , Molecular Sequence Data , Open Reading Frames , RNA, Small Interfering/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL