Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Biol Chem ; 296: 100363, 2021.
Article in English | MEDLINE | ID: mdl-33539919

ABSTRACT

During the integration step, human immunodeficiency virus type 1 integrase (IN) interacts with viral DNA and the cellular cofactor LEDGF/p75 to effectively integrate the reverse transcript into the host chromatin. Allosteric human immunodeficiency virus type 1 integrase inhibitors (ALLINIs) are a new class of antiviral agents that bind at the dimer interface of the IN catalytic core domain and occupy the binding site of LEDGF/p75. While originally designed to block IN-LEDGF/p75 interactions during viral integration, several of these compounds have been shown to also severely impact viral maturation through an IN multimerization mechanism. In this study, we tested the hypothesis that these dual properties of ALLINIs could be decoupled toward late stage viral replication effects by generating additional contact points between the bound ALLINI and a third subunit of IN. By sequential derivatization at position 7 of a quinoline-based ALLINI scaffold, we show that IN multimerization properties are enhanced by optimizing hydrophobic interactions between the compound and the C-terminal domain of the third IN subunit. These features not only improve the overall antiviral potencies of these compounds but also significantly shift the ALLINIs selectivity toward the viral maturation stage. Thus, we demonstrate that to fully maximize the potency of ALLINIs, the interactions between the inhibitor and all three IN subunits need to be simultaneously optimized.


Subject(s)
HIV Integrase/metabolism , HIV-1/metabolism , Quinolines/pharmacology , Allosteric Regulation/drug effects , Antiviral Agents/pharmacology , HEK293 Cells , HIV Integrase/physiology , HIV Integrase Inhibitors/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Protein Binding/drug effects , Protein Multimerization/drug effects , Quinolines/chemistry , Quinolines/metabolism , Virus Integration/drug effects , Virus Replication/drug effects
2.
Cell Mol Life Sci ; 75(14): 2491-2507, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29417178

ABSTRACT

Integration is central to HIV-1 replication and helps mold the reservoir of cells that persists in AIDS patients. HIV-1 interacts with specific cellular factors to target integration to interior regions of transcriptionally active genes within gene-dense regions of chromatin. The viral capsid interacts with several proteins that are additionally implicated in virus nuclear import, including cleavage and polyadenylation specificity factor 6, to suppress integration into heterochromatin. The viral integrase protein interacts with transcriptional co-activator lens epithelium-derived growth factor p75 to principally position integration within gene bodies. The integrase additionally senses target DNA distortion and nucleotide sequence to help fine-tune the specific phosphodiester bonds that are cleaved at integration sites. Research into virus-host interactions that underlie HIV-1 integration targeting has aided the development of a novel class of integrase inhibitors and may help to improve the safety of viral-based gene therapy vectors.


Subject(s)
Gene Targeting , HIV Integrase/physiology , HIV-1/physiology , Virus Integration/physiology , HIV-1/genetics , Host-Pathogen Interactions/genetics , Humans , Virus Internalization , Virus Latency/physiology
3.
Curr Top Microbiol Immunol ; 389: 93-119, 2015.
Article in English | MEDLINE | ID: mdl-25778682

ABSTRACT

Multimeric HIV-1 integrase (IN) plays an essential, multifunctional role in virus replication and serves as an important therapeutic target. Structural and biochemical studies have revealed the importance of the ordered interplay between IN molecules for its function. In the presence of viral DNA ends, individual IN subunits assemble into a tetramer and form a stable synaptic complex (SSC), which mediates integration of the reverse transcribed HIV-1 genome into chromatin. Cellular chromatin-associated protein LEDGF/p75 engages the IN tetramer in the SSC and directs HIV-1 integration into active genes. A mechanism to deregulate the productive interplay between IN subunits with small molecule inhibitors has recently received considerable attention. Most notably, allosteric IN inhibitors (ALLINIs) have been shown to bind to the IN dimer interface at the LEDGF/p75 binding pocket, stabilize interacting IN subunits, and promote aberrant, higher order IN multimerization. Consequently, these compounds impair formation of the SSC and associated LEDGF/p75-independent IN catalytic activities as well as inhibit LEDGF/p75 binding to the SSC in vitro. However, in infected cells, ALLINIs more potently impaired correct maturation of virus particles than the integration step. ALLINI treatments induced aberrant, higher order IN multimerization in virions and resulted in eccentric, non-infectious virus particles. These studies have suggested that the correctly ordered IN structure is important for virus particle morphogenesis and highlighted IN multimerization as a plausible therapeutic target for developing new inhibitors to enhance treatment options for HIV-1-infected patients.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , HIV Integrase Inhibitors/therapeutic use , HIV Integrase/physiology , Protein Multimerization/drug effects , HIV Integrase/chemistry , Humans , Protein Subunits
4.
Nucleic Acids Res ; 41(5): e61, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23275537

ABSTRACT

Integrating viral vectors are efficient gene transfer tools, but their integration patterns have been associated with genotoxicity and oncogenicity. The recent development of highly specific designer nucleases has enabled target DNA modification and site-specific gene insertion at desired genomic loci. However, a lack of consensus exists regarding a perfect genomic safe harbour (GSH) that would allow transgenes to be stably and reliably expressed without adversely affecting endogenous gene structure and function. Ribosomal DNA (rDNA) has many advantages as a GSH, but efficient means to target integration to this locus are currently lacking. We tested whether lentivirus vector integration can be directed to rDNA by using fusion proteins consisting of the Human Immunodeficiency Virus 1 (HIV-1) integrase (IN) and the homing endonuclease I-PpoI, which has natural cleavage sites in the rDNA. A point mutation (N119A) was introduced into I-PpoI to abolish unwanted DNA cleavage by the endonuclease. The vector-incorporated IN-I-PpoIN119A fusion protein targeted integration into rDNA significantly more than unmodified lentivirus vectors, with an efficiency of 2.7%. Our findings show that IN-fusion proteins can be used to modify the integration pattern of lentivirus vectors, and to package site-specific DNA-recognizing proteins into vectors to obtain safer transgene integration.


Subject(s)
DNA, Ribosomal/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , HIV Integrase/genetics , Mutagenesis, Insertional/methods , Recombinant Fusion Proteins/genetics , Cloning, Molecular , DNA Breaks, Double-Stranded , Deoxyribonucleases, Type II Site-Specific/biosynthesis , Deoxyribonucleases, Type II Site-Specific/physiology , Genetic Vectors , HEK293 Cells , HIV Integrase/biosynthesis , HIV Integrase/physiology , HIV-1/enzymology , HeLa Cells , Humans , Lentivirus/genetics , Physarum polycephalum/enzymology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Transduction, Genetic
5.
Cell Mol Life Sci ; 70(13): 2411-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23417324

ABSTRACT

Higher eukaryotic organisms have a variety of specific and nonspecific defense mechanisms against viral invaders. In animal cells, viral replication may be limited through the decrease in translation. Some viruses, however, have evolved mechanisms that counteract the response of the host. We report that infection by HIV-1 triggers acute decrease in translation. The human protein kinase GCN2 (eIF2AK4) is activated by phosphorylation upon HIV-1 infection in the hours following infection. Thus, infection by HIV-1 constitutes a stress that leads to the activation of GCN2 with a resulting decrease in protein synthesis. We have shown that GCN2 interacts with HIV-1 integrase (IN). Transfection of IN in amino acid-starved cells, where GCN2 is activated, increases the protein synthesis level. These results point to an as yet unknown role of GCN2 as an early mediator in the cellular response to HIV-1 infection, and suggest that the virus is able to overcome the involvement of GCN2 in the cellular response by eliciting methods to maintain protein synthesis.


Subject(s)
HIV-1/pathogenicity , Protein Biosynthesis , Protein Serine-Threonine Kinases/physiology , Gene Silencing , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV Integrase/metabolism , HIV Integrase/physiology , HIV-1/physiology , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological , Virus Replication
6.
J Virol ; 86(12): 6913-23, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22496233

ABSTRACT

An attenuation of the HIV-1 replication capacity (RC) has been observed for immune-mediated escape mutations in Gag restricted by protective HLA alleles. However, the extent to which escape mutations affect other viral proteins during natural infection is not well understood. We generated recombinant viruses encoding plasma HIV-1 RNA integrase sequences from antiretroviral-naïve individuals with early (n = 88) and chronic (n = 304) infections and measured the in vitro RC of each. In contrast to data from previous studies of Gag, we observed little evidence that host HLA allele expression was associated with integrase RC. A modest negative correlation was observed between the number of HLA-B-associated integrase polymorphisms and RC in chronic infection (R = -0.2; P = 0.003); however, this effect was not driven by mutations restricted by protective HLA alleles. Notably, the integrase variants S119R, G163E, and I220L, which represent uncommon polymorphisms associated with HLA-C*05, -A*33, and -B*52, respectively, correlated with lower RC (all q < 0.2). We identified a novel C*05-restricted epitope (HTDNGSNF(114-121)) that likely contributes to the selection of the S119R variant, the polymorphism most significantly associated with lower RC in patient sequences. An NL4-3 mutant encoding the S119R polymorphism displayed a ~35%-reduced function that was rescued by a single compensatory mutation of A91E. Together, these data indicate that substantial HLA-driven attenuation of integrase is not a general phenomenon during HIV-1 adaptation to host immunity. However, uncommon polymorphisms selected by HLA alleles that are not conventionally regarded to be protective may be associated with impaired protein function. Vulnerable epitopes in integrase might therefore be considered for future vaccine strategies.


Subject(s)
HIV Infections/immunology , HIV Integrase/physiology , HIV-1/enzymology , Immune Evasion , Virus Replication , Cohort Studies , HIV Infections/genetics , HIV Infections/virology , HIV Integrase/genetics , HIV Integrase/immunology , HIV-1/genetics , HIV-1/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Molecular Sequence Data
7.
PLoS Pathog ; 7(2): e1001280, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347347

ABSTRACT

Establishment of stable HIV-1 infection requires the efficient integration of the retroviral genome into the host DNA. The molecular mechanism underlying the control of this process by the chromatin structure has not yet been elucidated. We show here that stably associated nucleosomes strongly inhibit in vitro two viral-end integration by decreasing the accessibility of DNA to integrase. Remodeling of the chromatinized template by the SWI/SNF complex, whose INI1 major component interacts with IN, restores and redirects the full-site integration into the stable nucleosome region. These effects are not observed after remodeling by other human remodeling factors such as SNF2H or BRG1 lacking the integrase binding protein INI1. This suggests that the restoration process depends on the direct interaction between IN and the whole SWI/SNF complex, supporting a functional coupling between the remodeling and integration complexes. Furthermore, in silico comparison between more than 40,000 non-redundant cellular integration sites selected from literature and nucleosome occupancy predictions also supports that HIV-1 integration is promoted in the genomic region of weaker intrinsic nucleosome density in the infected cell. Our data indicate that some chromatin structures can be refractory for integration and that coupling between nucleosome remodeling and HIV-1 integration is required to overcome this natural barrier.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , HIV Integrase/physiology , Nucleosomes/metabolism , Nucleosomes/virology , Transcription Factors/physiology , Virus Integration/physiology , Animals , Cell Transformation, Viral/genetics , Cells, Cultured , Chromatin Assembly and Disassembly/physiology , Chromosomal Proteins, Non-Histone/metabolism , Efficiency , Gene Expression Regulation, Viral , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV Integrase/metabolism , HeLa Cells , Humans , Models, Biological , Protein Stability , Spodoptera , Transcription Factors/metabolism , Transcription, Genetic
8.
Biochemistry ; 50(45): 9788-96, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21992419

ABSTRACT

The assembly mechanism for the human immunodeficiency virus type 1 (HIV) synaptic complex (SC) capable of concerted integration is unknown. Molecular and structural studies have established that the HIV SC and prototype foamy virus (PFV) intasome contain a tetramer of integrase (IN) that catalyzes concerted integration. HIV IN purified in the presence of 1 mM EDTA and 10 mM MgSO(4) was predominately a monomer. IN efficiently promoted concerted integration of micromolar concentrations of 3'-OH recessed and blunt-ended U5 long terminal repeat (LTR) oligonucleotide (ODN) substrates (19-42 bp) into circular target DNA. Varying HIV IN to U5 DNA showed that an IN dimer:DNA end molar ratio of 1 was optimal for concerted integration. Integration activities decreased with an increasing length of the ODN, starting from the recessed 18/20 or 19/21 bp set to the 31/33 and 40/42 bp set. Under these conditions, the average fidelity for the HIV 5 bp host site duplication with recessed and blunt-ended substrates was 56%. Modifications of U5 LTR sequences beyond 21 bp from the terminus on longer DNA (1.6 kb) did not alter the ~32 bp DNaseI protective footprint, suggesting viral sequences beyond 21 bp were not essential for IN binding. The results suggest IN binds differentially to an 18/20 bp than to a 40/42 bp ODN substrate for concerted integration. The HIV IN monomer may be a suitable candidate for attempting crystallization of an IN-DNA complex in the absence or presence of strand transfer inhibitors.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/physiology , HIV Long Terminal Repeat/physiology , HIV-1/physiology , Virus Integration/physiology , Base Sequence , HIV Integrase/genetics , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Virus Integration/genetics
9.
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
10.
Uirusu ; 61(1): 73-80, 2011 Jun.
Article in Japanese | MEDLINE | ID: mdl-21972558

ABSTRACT

Reverse transcription of retroviral RNA into double stranded DNA is a characteristic feature of rertoviruses including human immunodeficiency virus type I (HIV-1). There has been accumulating evidence for the involvement of retroviral integrase (IN) in the reverse transcription of viral RNA. Here, we summarized recent our studies demonstrating direct functional roles of IN and its binding partner of host factor, Gemin2 in the reverse transcription. We established new in vitro cell-free assay to mimic natural reverse transcription and found that HIV-1 IN and host factor, Gemin2 synergistically stimulate reverse transcriptase (RT) activity. Analysis of intracellular stability and multimer formation of IN suggest that that high-ordered structures, especially tetramer formation of IN is critical for the function. In addition, Gemin2 might have a role to keep the higher-order structure of IN. Thus, we provide new aspects of reverse transcription of HIV-1 through IN and host factors in addition to RT.


Subject(s)
Genome, Viral/genetics , HIV Integrase/physiology , HIV Reverse Transcriptase/physiology , HIV-1/enzymology , HIV-1/genetics , Reverse Transcription , SMN Complex Proteins/physiology , Cell-Free System , DNA , DNA, Viral , RNA, Viral/genetics , Virus Replication/genetics
11.
Methods ; 47(4): 225-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19389610

ABSTRACT

Significant advances have transpired in the human immunodeficiency virus type 1 (HIV-1) integration field in recent years. Considering its essential nature, integrase has long been a target of interest for antiviral drug development. The most significant advance was the approval of the Merck compound raltegravir, the first licensed integrase inhibitor, in October 2007. Another milestone was the identification and characterization of specific nucleoprotein complexes that mediate integrase 3' processing and DNA strand transfer activities in vitro. Genome-wide distribution analyses have furthermore revealed that different retroviruses differentially target distinctive regions of chromatin during integration. For examples, lentiviruses favor actively transcribed genes whereas gammaretroviruses such as Moloney murine leukemia virus prefer transcriptional start sites. Though the underlying mechanisms are unknown for most retroviruses, the lentiviral preference is in large part guided through the interaction with the integrase binding protein lens epithelium-derived growth factor (LEDGF)/p75. Experimental methods that formed the foundations for each of these advances, as well as other techniques topical to the study of HIV-1 integration, are described in this issue of Methods.


Subject(s)
HIV-1/drug effects , HIV-1/genetics , Virus Integration/drug effects , Animals , Anti-HIV Agents/pharmacology , Gene Targeting/methods , HIV Integrase/physiology , HIV-1/physiology , Humans , Virus Integration/physiology
12.
Ethn Dis ; 20(1 Suppl 1): S1-45-9, 2010.
Article in English | MEDLINE | ID: mdl-20521384

ABSTRACT

An essential step in the life cycle of human immunodeficiency virus type 1 (HIV-1) is integration of the double-stranded retroviral DNA into the genome of the host cell. HIV-1 integrase, the enzyme that inserts the vital DNA into the host chromosome, is an attractive and rational target for anti-AIDS drug design because it is essential for HIV replication and there are no known counterparts in the host cell. Inhibitors of this enzyme have a great potential to complement the therapeutic use of HIV protease and reverse transcriptase inhibitors. Natural products have provided a source of new drug candidates for anti-AIDS therapy. Dicaffeoylquinic acids, isolated from traditional medicinal plants, are a novel class of integrase inhibitors. These compounds are potent inhibitors of HIV-1 replication in cultured cell lines and catalytic activities of integrase in vitro. They are therefore promising compounds for developing new anti-AIDS drugs. To understand how the inhibitors work and therefore design more potent and specific inhibitors, we have used molecular modeling techniques to investigate the binding modes of 3,4-dicaffeoylquinic acid. Our computational modeling study demonstrated that the inhibitor of this compound on HIV integrase is likely to proceed by two different but equivalent mechanisms with one bound to the active site region of the enzyme and another docked into the binding pocket located on the other side of the catalytic site. Our study will be of help to design new pharmaceuticals for the treatment of AIDS.


Subject(s)
HIV Integrase/physiology , HIV-1/physiology , Integrase Inhibitors/pharmacology , Quinic Acid/analogs & derivatives , Virus Replication/drug effects , Acquired Immunodeficiency Syndrome/drug therapy , Catalytic Domain/drug effects , Catalytic Domain/physiology , Computational Biology , Drug Design , Protein Binding , Quinic Acid/antagonists & inhibitors , Structure-Activity Relationship , Virus Integration/drug effects , Virus Replication/physiology
13.
Yao Xue Xue Bao ; 45(2): 215-23, 2010 Feb.
Article in Zh | MEDLINE | ID: mdl-21351431

ABSTRACT

HIV-1 integrase (IN) is an essential enzyme for retroviral replication. There is no analogue for this enzyme in human cells so that inhibition of IN will not bring strong effect on human body. Thus, HIV-1 IN has become a rational target for therapy of AIDS. This review provides a comprehensive report of alpha, gamma-diketo IN inhibitors discovered in recent years. Compilation of such data will prove to be beneficial in developing QSAR, pharmacophore hypothesis generation and validation, virtual screening and synthesis of compounds with higher activity.


Subject(s)
Anti-HIV Agents , HIV Integrase Inhibitors , HIV-1/drug effects , Keto Acids , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV Integrase/chemistry , HIV Integrase/physiology , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , Humans , Keto Acids/chemical synthesis , Keto Acids/chemistry , Keto Acids/pharmacology , Molecular Structure , Quantitative Structure-Activity Relationship
14.
Curr Med Chem ; 26(15): 2574-2600, 2019.
Article in English | MEDLINE | ID: mdl-29623824

ABSTRACT

Currently, used antiretroviral HIV therapy drugs exclusively target critical groups in the enzymes essential for the viral life cycle. Increased mutagenesis of their genes changes these viral enzymes, which once mutated can evade therapeutic targeting, effects which confer drug resistance. To circumvent this, our review addresses a strategy to design and derive HIV-Integrase (HIV-IN) inhibitors which simultaneously target two IN functional domains, rendering it inactive even if the enzyme accumulates many mutations. First we review the enzymatic role of IN to insert the copied viral DNA into a chromosome of the host T lymphocyte, highlighting its main functional and structural features to be subjected to inhibitory action. From a functional and structural perspective we present all classes of HIV-IN inhibitors with their most representative candidates. For each chosen compound we also explain its mechanism of IN inhibition. We use the recently resolved cryo EM IN tetramer intasome DNA complex onto which we dock various reference IN inhibitory chemical scaffolds such as to target adjacent functional IN domains. Pairing compounds with complementary activity, which dock in the vicinity of a IN structural microdomain, we design bifunctional new drugs which may not only be more resilient to IN mutations but also may be more potent inhibitors than their original counterparts. In the end of our review we propose synthesis pathways to link such paired compounds with enhanced synergistic IN inhibitory effects.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/chemistry , DNA/metabolism , Drug Design , HIV Integrase/metabolism , HIV Integrase/physiology , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , HIV-1/enzymology , HeLa Cells , Humans , Molecular Docking Simulation , Protein Binding/drug effects , Protein Domains
15.
Expert Rev Mol Med ; 9(6): 1-19, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17320002

ABSTRACT

Retroviruses, such as human immunodeficiency virus type 1 (HIV-1), are plus-sense RNA viruses that require reverse transcription and then DNA integration to establish a chromosomal provirus as an obligate replication intermediate. The viral enzyme reverse transcriptase synthesises linear double-stranded cDNA, which is the template for the viral enzyme integrase. Integrase catalyses two separate chemical reactions: an initial 3' processing of the nascent cDNA ends, which is followed in the cell nucleus by their covalent attachment to the 5' phosphates of a double-stranded staggered cut in chromosomal DNA. As integrase activity is essential for productive retroviral infection, there is intense interest in developing small-molecule inhibitors of the HIV-1 enzyme to increase the breadth of the antiviral arsenal used to fight HIV/AIDS. Purified integrase protein displays the 3' processing and DNA-strand-transfer activities essential for cDNA integration in integration assays in vitro, but numerous studies indicate that cellular proteins play important roles during integration in infected cells. This review highlights the molecular mechanisms behind HIV-1 integration, focusing on recent insights into functions of human cellular cofactors. The progress towards developing integrase inhibitors for their use in the clinic is also reviewed.


Subject(s)
HIV Infections/therapy , HIV-1/physiology , Virus Integration/physiology , Animals , Coenzymes/physiology , Enzyme Inhibitors/therapeutic use , Gene Targeting , HIV Integrase/chemistry , HIV Integrase/genetics , HIV Integrase/physiology , Humans , Models, Biological , Models, Molecular
16.
Bioorg Med Chem Lett ; 17(23): 6511-5, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17931865

ABSTRACT

A series of potent novel 8-hydroxy-3,4-dihydropyrrolo[1,2-a]pyrazine-1(2H)-one HIV-1 integrase inhibitors was identified. These compounds inhibited the strand transfer process of HIV-1 integrase and viral replication in cells. Compound 12 is active against replication of HIV-1 in cell culture with a CIC(95) of 0.31microM. Further SAR exploration led to the preparation of pseudosymmetrical tricyclic pyrrolopyrazine inhibitors 23 and 24 with further improvement in antiviral activity.


Subject(s)
HIV Integrase Inhibitors/chemistry , HIV Integrase , Pyrazines/chemistry , Cell Line, Tumor , HIV Integrase/physiology , HIV Integrase Inhibitors/pharmacology , Humans , Pyrazines/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/virology
17.
Kokubyo Gakkai Zasshi ; 73(1): 47-52, 2006 Mar.
Article in Japanese | MEDLINE | ID: mdl-16629470

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) is one of the lentiviruses, and unlike other retroviruses, HIV-1 is capable of infecting not only dividing cells but also non-dividing cells. It has been strongly suggested that this property is due to the viral mechanism which facilitates the integration of the viral genome cDNA into the host chromosome more efficiently than other retroviruses. HIV-1 integrase (IN) is a viral protein which catalyzes insertion of the viral genome cDNA into the host cell chromosome. However, it has been suggested that HIV-1 IN also plays putative roles at the steps prior to integration such as uncoating, reverse transcription and nuclear transport of the viral cDNA. In this study, we tried to identify the novel host factor which interacted with HIV-1 IN using two different kinds of yeast two hybrid methods: the conventional yeast two hybrid method and the mating method. First, the full-length cDNA fragment of HIV-1 IN was amplified using polymerase chain reaction (PCR), and the amplified products were ligated into pGBT 9 vector as bait. Plasmid vectors expressing human lymphocytes cDNA library and HeLa cDNA library were used as prey plasmid in the conventional yeast two hybrid method and the mating method, respectively. These plasmids were transformed into the corresponding yeast strain cells, and several positive clones were isolated. As a result, a known gene product was identified as a candidate. Further analysis revealed that this protein was expressed in HeLa, 293 T cells, primary macrophages and activated T lymphocytes, and that suppression of this protein expression affected HIV-1 replication.


Subject(s)
Cyclic AMP Response Element-Binding Protein/isolation & purification , HIV Integrase/physiology , Nerve Tissue Proteins/isolation & purification , RNA-Binding Proteins/isolation & purification , Cyclic AMP Response Element-Binding Protein/physiology , HeLa Cells , Humans , Nerve Tissue Proteins/physiology , RNA-Binding Proteins/physiology , SMN Complex Proteins
18.
Retrovirology ; 2: 62, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16232319

ABSTRACT

BACKGROUND: In addition to mediating the integration process, HIV-1 integrase (IN) has also been implicated in different steps during viral life cycle including reverse transcription and viral DNA nuclear import. Although the karyophilic property of HIV-1 IN has been well demonstrated using a variety of experimental approaches, the definition of domain(s) and/or motif(s) within the protein that mediate viral DNA nuclear import and its mechanism are still disputed and controversial. In this study, we performed mutagenic analyses to investigate the contribution of different regions in the C-terminal domain of HIV-1 IN to protein nuclear localization as well as their effects on virus infection. RESULTS: Our analysis showed that replacing lysine residues in two highly conserved tri-lysine regions, which are located within previously described Region C (235WKGPAKLLWKGEGAVV) and sequence Q (211KELQKQITK) in the C-terminal domain of HIV-1 IN, impaired protein nuclear accumulation, while mutations for RK263,4 had no significant effect. Analysis of their effects on viral infection in a VSV-G pseudotyped RT/IN trans-complemented HIV-1 single cycle replication system revealed that all three C-terminal mutant viruses (KK215,9AA, KK240,4AE and RK263,4AA) exhibited more severe defect of induction of beta-Gal positive cells and luciferase activity than an IN class 1 mutant D64E in HeLa-CD4-CCR5-beta-Gal cells, and in dividing as well as non-dividing C8166 T cells, suggesting that some viral defects are occurring prior to viral integration. Furthermore, by analyzing viral DNA synthesis and the nucleus-associated viral DNA level, the results clearly showed that, although all three C-terminal mutants inhibited viral reverse transcription to different extents, the KK240,4AE mutant exhibited most profound effect on this step, whereas KK215,9AA significantly impaired viral DNA nuclear import. In addition, our analysis could not detect viral DNA integration in each C-terminal mutant infection, even though they displayed various low levels of nucleus-associated viral DNA, suggesting that these C-terminal mutants also impaired viral DNA integration ability. CONCLUSION: All of these results indicate that, in addition to being involved in HIV-1 reverse transcription and integration, the C-terminal tri-lysine regions of IN also contribute to efficient viral DNA nuclear import during the early stage of HIV-1 replication.


Subject(s)
Active Transport, Cell Nucleus , DNA, Viral/metabolism , HIV Integrase/chemistry , HIV-1/genetics , Transcription, Genetic , Amino Acid Sequence , Bacterial Proteins/metabolism , Cell Line , HIV Integrase/physiology , HIV-1/pathogenicity , Humans , Luminescent Proteins/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Mutation , Protein Transport , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/genetics , Virus Integration , Virus Replication
19.
PLoS One ; 10(10): e0141335, 2015.
Article in English | MEDLINE | ID: mdl-26495836

ABSTRACT

The bacterial defense system CRISPR (clustered regularly interspaced short palindromic repeats) has been explored as a powerful tool to edit genomic elements. In this study, we test the potential of CRISPR Csy4 RNA endoribonuclease for targeting HIV-1. We fused human codon-optimized Csy4 endoribonuclease with VPR, a HIV-1 viral preintegration complex protein. An HIV-1 cell model was modified to allow quantitative detection of active virus production. We found that the trans-expressing VPR-Csy4 almost completely blocked viral infection in two target cell lines (SupT1, Ghost). In the MAGI cell assay, where the HIV-1 LTR ß-galactosidase is expressed under the control of the tat gene from an integrated provirus, VPR-Csy4 significantly blocked the activity of the provirus-activated HIV-1 reporter. This proof-of-concept study demonstrates that Csy4 endoribonuclease is a promising tool that could be tailored further to target HIV-1.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , Endoribonucleases/genetics , HIV Infections/prevention & control , HIV-1/physiology , Clustered Regularly Interspaced Short Palindromic Repeats , HEK293 Cells , HIV Integrase/physiology , Humans , Protein Engineering , Virus Assembly , Virus Integration
20.
Front Biosci ; 9: 3187-208, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15353349

ABSTRACT

Integration, catalyzed by the viral integrase (IN) protein, is a crucial step in the life cycle of all retroviruses including human immunodeficiency virus type 1 (HIV-1). Although purified HIV-1 IN protein is sufficient to catalyze the DNA breakage and joining steps of integration in the absence of any other protein factor, a number of studies indicate that cellular proteins participate in the integration process in cells. These host cell proteins have been proposed to act through binding the pre-integrated viral cDNA substrate, by directly interacting with the IN protein, and/or by repairing the single-stranded DNA gaps that occur at viral/chromosomal DNA junctions during integration. In this paper we summarize the identification and potential roles of specific cell factors in HIV-1 integration. We also present experimental results of human cell proteins that coimmunoprecipitated with HIV-1 IN following its expression in HeLa cells and discuss these results in light of the previously-identified integration cofactors.


Subject(s)
HIV Infections/pathology , HIV Integrase/physiology , HIV/metabolism , HeLa Cells/virology , Virus Integration , Animals , Binding Sites , DNA/metabolism , DNA Repair , DNA, Complementary/metabolism , Humans , Protein Binding , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL