Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
Add more filters

Publication year range
1.
Cell ; 166(4): 1004-1015, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27453467

ABSTRACT

Targeted HIV cure strategies require definition of the mechanisms that maintain the virus. Here, we tracked HIV replication and the persistence of infected CD4 T cells in individuals with natural virologic control by sequencing viruses, T cell receptor genes, HIV integration sites, and cellular transcriptomes. Our results revealed three mechanisms of HIV persistence operating within distinct anatomic and functional compartments. In lymph node, we detected viruses with genetic and transcriptional attributes of active replication in both T follicular helper (TFH) cells and non-TFH memory cells. In blood, we detected inducible proviruses of archival origin among highly differentiated, clonally expanded cells. Linking the lymph node and blood was a small population of circulating cells harboring inducible proviruses of recent origin. Thus, HIV replication in lymphoid tissue, clonal expansion of infected cells, and recirculation of recently infected cells act together to maintain the virus in HIV controllers despite effective antiviral immunity.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Blood/virology , CD4-Positive T-Lymphocytes/immunology , Chronic Disease , DNA, Viral/genetics , HIV Infections/immunology , HIV-1/genetics , Humans , Leukocytes, Mononuclear , Lymph Nodes/virology , Proviruses/immunology , Sequence Analysis, DNA , Virus Physiological Phenomena , Virus Replication
2.
Cell ; 152(5): 1021-36, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23452851

ABSTRACT

Histone modifications regulate chromatin-dependent processes, yet the mechanisms by which they contribute to specific outcomes remain unclear. H3K4me3 is a prominent histone mark that is associated with active genes and promotes transcription through interactions with effector proteins that include initiation factor TFIID. We demonstrate that H3K4me3-TAF3 interactions direct global TFIID recruitment to active genes, some of which are p53 targets. Further analyses show that (1) H3K4me3 enhances p53-dependent transcription by stimulating preinitiation complex (PIC) formation; (2) H3K4me3, through TAF3 interactions, can act either independently or cooperatively with the TATA box to direct PIC formation and transcription; and (3) H3K4me3-TAF3/TFIID interactions regulate gene-selective functions of p53 in response to genotoxic stress. Our findings indicate a mechanism by which H3K4me3 directs PIC assembly for the rapid induction of specific p53 target genes.


Subject(s)
Histone Code , Histones/metabolism , Transcription Factor TFIID/metabolism , Transcription Initiation, Genetic , Cell Line, Tumor , Humans , Lysine/metabolism , Methylation , TATA Box , TATA-Binding Protein Associated Factors , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , p300-CBP Transcription Factors/metabolism
3.
PLoS Pathog ; 18(8): e1010726, 2022 08.
Article in English | MEDLINE | ID: mdl-36044447

ABSTRACT

Although combination antiretroviral therapy (ART) blocks HIV replication, it is not curative because infected CD4+ T cells that carry intact, infectious proviruses persist. Understanding the behavior of clones of infected T cells is important for understanding the stability of the reservoir; however, the stabilities of clones of infected T cells in persons on long-term ART are not well defined. We determined the relative stabilities of clones of infected and uninfected CD4+ T cells over time intervals of one to four years in three individuals who had been on ART for 9-19 years. The largest clones of uninfected T cells were larger than the largest clones of infected T cells. Clones of infected CD4+ T cells were more stable than clones of uninfected CD4+ T cells of a similar size. Individual clones of CD4+ T cells carrying intact, infectious proviruses can expand, contract, or remain stable over time.


Subject(s)
HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , Clone Cells , DNA, Viral , HIV Infections/drug therapy , HIV-1/genetics , Humans , Proviruses/genetics
4.
Nature ; 559(7714): 405-409, 2018 07.
Article in English | MEDLINE | ID: mdl-29995861

ABSTRACT

Decades of work have aimed to genetically reprogram T cells for therapeutic purposes1,2 using recombinant viral vectors, which do not target transgenes to specific genomic sites3,4. The need for viral vectors has slowed down research and clinical use as their manufacturing and testing is lengthy and expensive. Genome editing brought the promise of specific and efficient insertion of large transgenes into target cells using homology-directed repair5,6. Here we developed a CRISPR-Cas9 genome-targeting system that does not require viral vectors, allowing rapid and efficient insertion of large DNA sequences (greater than one kilobase) at specific sites in the genomes of primary human T cells, while preserving cell viability and function. This permits individual or multiplexed modification of endogenous genes. First, we applied this strategy to correct a pathogenic IL2RA mutation in cells from patients with monogenic autoimmune disease, and demonstrate improved signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR that redirected T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized tumour antigens and mounted productive anti-tumour cell responses in vitro and in vivo. Together, these studies provide preclinical evidence that non-viral genome targeting can enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.


Subject(s)
Cellular Reprogramming/genetics , Gene Editing , Genome, Human/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Autoimmunity/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Male , Mice , Neoplasm Transplantation , Protein Engineering , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
5.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-33958444

ABSTRACT

Prolonged detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA and recurrence of PCR-positive tests have been widely reported in patients after recovery from COVID-19, but some of these patients do not appear to shed infectious virus. We investigated the possibility that SARS-CoV-2 RNAs can be reverse-transcribed and integrated into the DNA of human cells in culture and that transcription of the integrated sequences might account for some of the positive PCR tests seen in patients. In support of this hypothesis, we found that DNA copies of SARS-CoV-2 sequences can be integrated into the genome of infected human cells. We found target site duplications flanking the viral sequences and consensus LINE1 endonuclease recognition sequences at the integration sites, consistent with a LINE1 retrotransposon-mediated, target-primed reverse transcription and retroposition mechanism. We also found, in some patient-derived tissues, evidence suggesting that a large fraction of the viral sequences is transcribed from integrated DNA copies of viral sequences, generating viral-host chimeric transcripts. The integration and transcription of viral sequences may thus contribute to the detection of viral RNA by PCR in patients after infection and clinical recovery. Because we have detected only subgenomic sequences derived mainly from the 3' end of the viral genome integrated into the DNA of the host cell, infectious virus cannot be produced from the integrated subgenomic SARS-CoV-2 sequences.


Subject(s)
COVID-19/genetics , COVID-19/virology , SARS-CoV-2/genetics , Virus Integration/genetics , Animals , COVID-19/metabolism , Chlorocebus aethiops , Genome, Viral , HEK293 Cells , Humans , RNA, Viral/genetics , SARS-CoV-2/metabolism , Vero Cells , Virus Integration/physiology , Virus Replication/genetics , Virus Replication/physiology
6.
PLoS Pathog ; 17(8): e1009812, 2021 08.
Article in English | MEDLINE | ID: mdl-34343212

ABSTRACT

MmuPV1 is a useful model for studying papillomavirus-induced tumorigenesis. We used RNA-seq to look for chimeric RNAs that map to both MmuPV1 and host genomes. In tumor tissues, a higher proportion of total viral reads were virus-host chimeric junction reads (CJRs) (1.9‰ - 7‰) than in tumor-free tissues (0.6‰ - 1.3‰): most CJRs mapped to the viral E2/E4 region. Although most of the MmuPV1 integration sites were mapped to intergenic regions and introns throughout the mouse genome, integrations were seen more than once in several genes: Malat1, Krt1, Krt10, Fabp5, Pard3, and Grip1; these data were confirmed by rapid amplification of cDNA ends (RACE)-Single Molecule Real-Time (SMRT)-seq or targeted DNA-seq. Microhomology sequences were frequently seen at host-virus DNA junctions. MmuPV1 infection and integration affected the expression of host genes. We found that factors for DNA double-stranded break repair and microhomology-mediated end-joining (MMEJ), such as H2ax, Fen1, DNA polymerase Polθ, Cdk1, and Plk1, exhibited a step-wise increase and Mdc1 a decrease in expression in MmuPV1-infected tissues and MmuPV1 tumors relative to normal tissues. Increased expression of mitotic kinases CDK1 and PLK1 appears to be correlated with CtIP phosphorylation in MmuPV1 tumors, suggesting a role for MMEJ-mediated DNA joining in the MmuPV1 integration events that are associated with MmuPV1-induced progression of tumors.


Subject(s)
DNA End-Joining Repair , DNA Repair Enzymes/metabolism , DNA, Viral/genetics , Keratinocytes/metabolism , Papilloma/genetics , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Animals , Animals, Newborn , DNA Breaks, Double-Stranded , DNA Repair Enzymes/genetics , Female , Genome, Viral , Homologous Recombination , Keratinocytes/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Papilloma/virology , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , RNA-Seq
7.
PLoS Pathog ; 17(4): e1009141, 2021 04.
Article in English | MEDLINE | ID: mdl-33826675

ABSTRACT

HIV persists during antiretroviral therapy (ART) as integrated proviruses in cells descended from a small fraction of the CD4+ T cells infected prior to the initiation of ART. To better understand what controls HIV persistence and the distribution of integration sites (IS), we compared about 15,000 and 54,000 IS from individuals pre-ART and on ART, respectively, with approximately 395,000 IS from PBMC infected in vitro. The distribution of IS in vivo is quite similar to the distribution in PBMC, but modified by selection against proviruses in expressed genes, by selection for proviruses integrated into one of 7 specific genes, and by clonal expansion. Clones in which a provirus integrated in an oncogene contributed to cell survival comprised only a small fraction of the clones persisting in on ART. Mechanisms that do not involve the provirus, or its location in the host genome, are more important in determining which clones expand and persist.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/virology , Leukocytes, Mononuclear/virology , Oncogenes/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , DNA, Viral/genetics , Humans , Oncogenes/immunology , Proviruses/genetics , Virus Replication/genetics
8.
Mol Cell ; 59(1): 89-103, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26073541

ABSTRACT

Histone modification and DNA methylation are associated with varying epigenetic "landscapes," but detailed mechanistic and functional links between the two remain unclear. Using the ATRX-DNMT3-DNMT3L (ADD) domain of the DNA methyltransferase Dnmt3a as a paradigm, we apply protein engineering to dissect the molecular interactions underlying the recruitment of this enzyme to specific regions of chromatin in mouse embryonic stem cells (ESCs). By rendering the ADD domain insensitive to histone modification, specifically H3K4 methylation or H3T3 phosphorylation, we demonstrate the consequence of dysregulated Dnmt3a binding and activity. Targeting of a Dnmt3a mutant to H3K4me3 promoters decreases gene expression in a subset of developmental genes and alters ESC differentiation, whereas aberrant binding of another mutant to H3T3ph during mitosis promotes chromosome instability. Our studies support the general view that histone modification "reading" and DNA methylation are closely coupled in mammalian cells, and suggest an avenue for the functional assessment of chromatin-associated proteins.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Embryonic Stem Cells/cytology , Histones/genetics , Protein Engineering , Animals , Cell Differentiation , DNA Helicases/genetics , DNA Methylation , DNA Methyltransferase 3A , Mice , Mice, Inbred C57BL , Mitosis/genetics , Nuclear Proteins/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Structure, Tertiary , X-linked Nuclear Protein
9.
Genes Dev ; 29(21): 2287-97, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26545813

ABSTRACT

The host chromatin-binding factor LEDGF/p75 interacts with HIV-1 integrase and directs integration to active transcription units. To understand how LEDGF/p75 recognizes transcription units, we sequenced 1 million HIV-1 integration sites isolated from cultured HEK293T cells. Analysis of integration sites showed that cancer genes were preferentially targeted, raising concerns about using lentivirus vectors for gene therapy. Additional analysis led to the discovery that introns and alternative splicing contributed significantly to integration site selection. These correlations were independent of transcription levels, size of transcription units, and length of the introns. Multivariate analysis with five parameters previously found to predict integration sites showed that intron density is the strongest predictor of integration density in transcription units. Analysis of previously published HIV-1 integration site data showed that integration density in transcription units in mouse embryonic fibroblasts also correlated strongly with intron number, and this correlation was absent in cells lacking LEDGF. Affinity purification showed that LEDGF/p75 is associated with a number of splicing factors, and RNA sequencing (RNA-seq) analysis of HEK293T cells lacking LEDGF/p75 or the LEDGF/p75 integrase-binding domain (IBD) showed that LEDGF/p75 contributes to splicing patterns in half of the transcription units that have alternative isoforms. Thus, LEDGF/p75 interacts with splicing factors, contributes to exon choice, and directs HIV-1 integration to transcription units that are highly spliced.


Subject(s)
HIV-1/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Virus Integration/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Introns/genetics , Protein Binding , Protein Structure, Tertiary , RNA Splicing
10.
Molecules ; 28(3)2023 Feb 02.
Article in English | MEDLINE | ID: mdl-36771093

ABSTRACT

An efficient one-pot synthetic method has been developed for the preparation of bicyclic carbamoyl pyridones from the known common intermediate methyl 5-((2,4-difluorobenzyl)carbamoyl)-1-(2,2-dimethoxyethyl)-3-methoxy-4-oxo-1,4-dihydropyridine-2-carboxylate (8). The scalable protocol is facile and employs readily available reagents, needing only a single purification as the final step. The utility of the approach was demonstrated by preparing a library of HIV-1 integrase strand transfer inhibitors (INSTIs) that differ by the presence or absence of a double bond in the B-ring of the bicyclic carbamoyl pyridines 6 and 7. Several of the analogs show good antiviral potencies in single-round HIV-1 replication antiviral assays and show no cytotoxicity in cell culture assays. In general, the compounds with a B-ring double bond have higher antiviral potencies than their saturated congeners. Our methodology should be applicable to the synthesis of a range of new metal-chelating analogs.


Subject(s)
HIV Infections , HIV Integrase Inhibitors , HIV Integrase , Humans , Pyridones/chemistry , Raltegravir Potassium/pharmacology , HIV Integrase Inhibitors/chemistry , Drug Resistance, Viral , HIV Integrase/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Heterocyclic Compounds, 3-Ring/pharmacology , HIV Infections/drug therapy
11.
Antimicrob Agents Chemother ; 66(1): e0150021, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34723625

ABSTRACT

Resistance to anti-HIV drugs has been a problem from the beginning of antiviral drug treatments. The recent expansion of combination antiretroviral therapy worldwide has led to an increase in resistance to antiretrovirals; understanding the mechanisms of resistance is increasingly important. In this study, we analyzed reverse transcriptase (RT) variants based on sequences derived from an individual who had low-level rebound viremia while undergoing therapy with abacavir, azidothymidine (AZT) (zidovudine), and (-)-l-2',3'-dideoxy-3'-thiacytidine (3TC) (lamivudine). The RT had mutations at positions 64, 67, 70, 184, and 219 and a threonine insertion after amino acid 69 in RT. The virus remained partially susceptible to the nucleoside RT inhibitor (NRTI) regimen. We show how these mutations affect the ability of NRTIs to inhibit DNA synthesis by RT. The presence of the inserted threonine reduced the susceptibility of the RT mutant to inhibition by tenofovir.


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Amino Acids , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , HIV Reverse Transcriptase/metabolism , HIV-1/genetics , HIV-1/metabolism , Humans , Lamivudine/pharmacology , Mutation/genetics , Reverse Transcriptase Inhibitors/chemistry , Zidovudine/pharmacology
12.
Proc Natl Acad Sci U S A ; 116(51): 25891-25899, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776247

ABSTRACT

Understanding HIV-1 persistence despite antiretroviral therapy (ART) is of paramount importance. Both single-genome sequencing (SGS) and integration site analysis (ISA) provide useful information regarding the structure of persistent HIV DNA populations; however, until recently, there was no way to link integration sites to their cognate proviral sequences. Here, we used multiple-displacement amplification (MDA) of cellular DNA diluted to a proviral endpoint to obtain full-length proviral sequences and their corresponding sites of integration. We applied this method to lymph node and peripheral blood mononuclear cells from 5 ART-treated donors to determine whether groups of identical subgenomic sequences in the 2 compartments are the result of clonal expansion of infected cells or a viral genetic bottleneck. We found that identical proviral sequences can result from both cellular expansion and viral genetic bottlenecks occurring prior to ART initiation and following ART failure. We identified an expanded T cell clone carrying an intact provirus that matched a variant previously detected by viral outgrowth assays and expanded clones with wild-type and drug-resistant defective proviruses. We also found 2 clones from 1 donor that carried identical proviruses except for nonoverlapping deletions, from which we could infer the sequence of the intact parental virus. Thus, MDA-SGS can be used for "viral reconstruction" to better understand intrapatient HIV-1 evolution and to determine the clonality and structure of proviruses within expanded clones, including those with drug-resistant mutations. Importantly, we demonstrate that identical sequences observed by standard SGS are not always sufficient to establish proviral clonality.


Subject(s)
HIV-1/genetics , Virus Integration/genetics , Virus Replication/genetics , Anti-Retroviral Agents/therapeutic use , Base Sequence , Cell Line , DNA, Viral/genetics , Drug Resistance, Viral , HIV Infections/virology , Humans , Leukocytes, Mononuclear/virology , Lymph Nodes/virology , Mutation , Proviruses/genetics , Virus Integration/physiology
13.
PLoS Pathog ; 15(7): e1007869, 2019 07.
Article in English | MEDLINE | ID: mdl-31291371

ABSTRACT

Clonal expansion of HIV infected cells plays an important role in the formation and persistence of the reservoir that allows the virus to persist, in DNA form, despite effective antiretroviral therapy. We used integration site analysis to ask if there is a similar clonal expansion of SIV infected cells in macaques. We show that the distribution of HIV and SIV integration sites in vitro is similar and that both viruses preferentially integrate in many of the same genes. We obtained approximately 8000 integration sites from blood samples taken from SIV-infected macaques prior to the initiation of ART, and from blood, spleen, and lymph node samples taken at necropsy. Seven clones were identified in the pre-ART samples; one persisted for a year on ART. An additional 100 clones were found only in on-ART samples; a number of these clones were found in more than one tissue. The timing and extent of clonal expansion of SIV-infected cells in macaques and HIV-infected cells in humans is quite similar. This suggests that SIV-infected macaques represent a useful model of the clonal expansion of HIV infected cells in humans that can be used to evaluate strategies intended to control or eradicate the viral reservoir.


Subject(s)
HIV Infections/drug therapy , HIV Infections/virology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , Disease Reservoirs/virology , HIV Infections/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , In Vitro Techniques , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/pathogenicity , Viral Load/drug effects , Virus Integration/genetics , Virus Integration/physiology , Virus Replication/drug effects
15.
BMC Genomics ; 21(1): 517, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727364

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

16.
BMC Genomics ; 21(1): 216, 2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32151239

ABSTRACT

BACKGROUND: All retroviruses, including human immunodeficiency virus (HIV), must integrate a DNA copy of their genomes into the genome of the infected host cell to replicate. Although integrated retroviral DNA, known as a provirus, can be found at many sites in the host genome, integration is not random. The adaption of linker-mediated PCR (LM-PCR) protocols for high-throughput integration site mapping, using randomly-sheared genomic DNA and Illumina paired-end sequencing, has dramatically increased the number of mapped integration sites. Analysis of samples from human donors has shown that there is clonal expansion of HIV infected cells and that clonal expansion makes an important contribution to HIV persistence. However, analysis of HIV integration sites in samples taken from patients requires extensive PCR amplification and high-throughput sequencing, which makes the methodology prone to certain specific artifacts. RESULTS: To address the problems with artifacts, we use a comprehensive approach involving experimental procedures linked to a bioinformatics analysis pipeline. Using this combined approach, we are able to reduce the number of PCR/sequencing artifacts that arise and identify the ones that remain. Our streamlined workflow combines random cleavage of the DNA in the samples, end repair, and linker ligation in a single step. We provide guidance on primer and linker design that reduces some of the common artifacts. We also discuss how to identify and remove some of the common artifacts, including the products of PCR mispriming and PCR recombination, that have appeared in some published studies. Our improved bioinformatics pipeline rapidly parses the sequencing data and identifies bona fide integration sites in clonally expanded cells, producing an Excel-formatted report that can be used for additional data processing. CONCLUSIONS: We provide a detailed protocol that reduces the prevalence of artifacts that arise in the analysis of retroviral integration site data generated from in vivo samples and a bioinformatics pipeline that is able to remove the artifacts that remain.


Subject(s)
HIV Infections/genetics , HIV/physiology , Virus Integration , Chromosome Mapping , Computational Biology , DNA, Viral , Genome, Human , Humans , Polymerase Chain Reaction , Proviruses/physiology , Sequence Analysis, DNA
17.
Article in English | MEDLINE | ID: mdl-32601157

ABSTRACT

The currently recommended first-line therapy for HIV-1-infected patients is an integrase (IN) strand transfer inhibitor (INSTI), either dolutegravir (DTG) or bictegravir (BIC), in combination with two nucleoside reverse transcriptase inhibitors (NRTIs). Both DTG and BIC potently inhibit most INSTI-resistant IN mutants selected by the INSTIs raltegravir (RAL) and elvitegravir (EVG). BIC has not been reported to select for resistance in treatment-naive patients, and DTG has selected for a small number of resistant viruses in treatment-naive patients. However, some patients who had viruses with substitutions selected by RAL and EVG responded poorly when switched to DTG-based therapies, and there are mutants that cause a considerable decrease in the potencies of DTG and BIC in in vitro assays. The new INSTI cabotegravir (CAB), which is in late-stage clinical trials, has been shown to select for novel resistant mutants in vitro Thus, it is important to develop new and improved INSTIs that are effective against all the known resistant mutants. This led us to test our best inhibitors, in parallel with DTG, BIC, and CAB, in a single-round infection assay against a panel of the new CAB-resistant mutants. Of the INSTIs we tested, BIC and our compound 4d had the broadest efficacy. Both were superior to DTG, as evidenced by the data obtained with the IN mutant T66I/L74M/E138K/S147G/Q148R/S230N, which was selected by CAB using an EVG-resistant lab strain. These results support the preclinical development of compound 4d and provide information that can be used in the design of additional INSTIs that will be effective against a broad spectrum of resistant mutants.


Subject(s)
HIV Infections , HIV Integrase Inhibitors , HIV Integrase , HIV-1 , Pharmaceutical Preparations , Drug Resistance, Viral/genetics , HIV Infections/drug therapy , HIV Integrase/genetics , HIV Integrase Inhibitors/pharmacology , HIV Integrase Inhibitors/therapeutic use , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Oxazines/pharmacology , Piperazines/pharmacology , Pyridones/pharmacology
18.
J Virol ; 93(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30894467

ABSTRACT

Two mutations, G112D and M230I, were selected in the reverse transcriptase (RT) of human immunodeficiency virus type 1 (HIV-1) by a novel nonnucleoside reverse transcriptase inhibitor (NNRTI). G112D is located near the HIV-1 polymerase active site; M230I is located near the hydrophobic region where NNRTIs bind. Thus, M230I could directly interfere with NNRTI binding but G112D could not. Biochemical and virological assays were performed to analyze the effects of these mutations individually and in combination. M230I alone caused a reduction in susceptibility to NNRTIs, while G112D alone did not. The G112D/M230I double mutant was less susceptible to NNRTIs than was M230I alone. In contrast, both mutations affected the ability of RT to incorporate nucleoside analogs. We suggest that the mutations interact with each other via the bound nucleic acid substrate; the nucleic acid forms part of the polymerase active site, which is near G112D. The positioning of the nucleic acid is influenced by its interactions with the "primer grip" region and could be influenced by the M230I mutation.IMPORTANCE Although antiretroviral therapy (ART) is highly successful, drug-resistant variants can arise that blunt the efficacy of ART. New inhibitors that are broadly effective against known drug-resistant variants are needed, although such compounds might select for novel resistance mutations that affect the sensitivity of the virus to other compounds. Compound 13 selects for resistance mutations that differ from traditional NNRTI resistance mutations. These mutations cause increased sensitivity to NRTIs, such as AZT.


Subject(s)
HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Anti-HIV Agents/pharmacology , Cell Line , Drug Resistance, Viral/genetics , HEK293 Cells , HIV Infections/virology , HIV Reverse Transcriptase/drug effects , HIV-1/genetics , Humans , Mutation/drug effects , Nucleosides/pharmacology , Reverse Transcriptase Inhibitors/pharmacology
19.
J Virol ; 92(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29643235

ABSTRACT

We tested three compounds for their ability to inhibit the RNase H (RH) and polymerase activities of HIV-1 reverse transcriptase (RT). A high-resolution crystal structure (2.2 Å) of one of the compounds showed that it chelates the two magnesium ions at the RH active site; this prevents the RH active site from interacting with, and cleaving, the RNA strand of an RNA-DNA heteroduplex. The compounds were tested using a variety of substrates: all three compounds inhibited the polymerase-independent RH activity of HIV-1 RT. Time-of-addition experiments showed that the compounds were more potent if they were bound to RT before the nucleic acid substrate was added. The compounds significantly inhibited the site-specific cleavage required to generate the polypurine tract (PPT) RNA primer that initiates the second strand of viral DNA synthesis. The compounds also reduced the polymerase activity of RT; this ability was a result of the compounds binding to the RH active site. These compounds appear to be relatively specific; they do not inhibit either Escherichia coli RNase HI or human RNase H2. The compounds inhibit the replication of an HIV-1-based vector in a one-round assay, and their potencies were only modestly decreased by mutations that confer resistance to integrase strand transfer inhibitors (INSTIs), nucleoside analogs, or nonnucleoside RT inhibitors (NNRTIs), suggesting that their ability to block HIV replication is related to their ability to block RH cleavage. These compounds appear to be useful leads that can be used to develop more potent and specific compounds.IMPORTANCE Despite advances in HIV-1 treatment, drug resistance is still a problem. Of the four enzymatic activities found in HIV-1 proteins (protease, RT polymerase, RT RNase H, and integrase), only RNase H has no approved therapeutics directed against it. This new target could be used to design and develop new classes of inhibitors that would suppress the replication of the drug-resistant variants that have been selected by the current therapeutics.


Subject(s)
DNA Replication/drug effects , HIV Infections/drug therapy , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Naphthyridines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H/antagonists & inhibitors , Virus Replication/drug effects , Binding Sites , Catalytic Domain , Crystallography, X-Ray , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , HIV Infections/pathology , HIV Infections/virology , Humans , Naphthyridines/chemistry , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry
20.
PLoS Pathog ; 13(3): e1006283, 2017 03.
Article in English | MEDLINE | ID: mdl-28328934

ABSTRACT

The major obstacle to curing HIV infection is the persistence of cells with intact proviruses that can produce replication-competent virus. This HIV reservoir is believed to exist primarily in CD4+ T-cells and is stable despite years of suppressive antiretroviral therapy. A potential mechanism for HIV persistence is clonal expansion of infected cells, but how often such clones carry replication-competent proviruses has been controversial. Here, we used single-genome sequencing to probe for identical HIV sequence matches among viruses recovered in different viral outgrowth cultures and between the sequences of outgrowth viruses and proviral or intracellular HIV RNA sequences in uncultured blood mononuclear cells from eight donors on suppressive ART with diverse proviral populations. All eight donors had viral outgrowth virus that was fully susceptible to their current ART drug regimen. Six of eight donors studied had identical near full-length HIV RNA sequences recovered from different viral outgrowth cultures, and one of the two remaining donors had identical partial viral sequence matches between outgrowth virus and intracellular HIV RNA. These findings provide evidence that clonal expansion of HIV-infected cells is an important mechanism of reservoir persistence that should be targeted to cure HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/genetics , Proviruses/genetics , Anti-HIV Agents/therapeutic use , Humans , Polymerase Chain Reaction , Viral Load/genetics
SELECTION OF CITATIONS
SEARCH DETAIL