Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.338
Filter
Add more filters

Publication year range
1.
Cell ; 185(2): 266-282.e15, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35026153

ABSTRACT

HIV-1-infected cells that persist despite antiretroviral therapy (ART) are frequently considered "transcriptionally silent," but active viral gene expression may occur in some cells, challenging the concept of viral latency. Applying an assay for profiling the transcriptional activity and the chromosomal locations of individual proviruses, we describe a global genomic and epigenetic map of transcriptionally active and silent proviral species and evaluate their longitudinal evolution in persons receiving suppressive ART. Using genome-wide epigenetic reference data, we show that proviral transcriptional activity is associated with activating epigenetic chromatin features in linear proximity of integration sites and in their inter- and intrachromosomal contact regions. Transcriptionally active proviruses were actively selected against during prolonged ART; however, this pattern was violated by large clones of virally infected cells that may outcompete negative selection forces through elevated intrinsic proliferative activity. Our results suggest that transcriptionally active proviruses are dynamically evolving under selection pressure by host factors.


Subject(s)
HIV-1/genetics , Proviruses/genetics , Transcription, Genetic , Aged , Base Sequence , Biological Evolution , Chromatin/metabolism , Clone Cells , DNA, Viral/genetics , Epigenesis, Genetic/drug effects , Female , Humans , Ionomycin/pharmacology , Male , Middle Aged , Phylogeny , Proviruses/drug effects , RNA, Viral/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects , Virus Integration/genetics , Virus Latency/drug effects , Virus Latency/genetics
2.
Cell ; 166(5): 1257-1268.e12, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565348

ABSTRACT

While an essential role of HIV-1 integrase (IN) for integration of viral cDNA into human chromosome is established, studies with IN mutants and allosteric IN inhibitors (ALLINIs) have suggested that IN can also influence viral particle maturation. However, it has remained enigmatic as to how IN contributes to virion morphogenesis. Here, we demonstrate that IN directly binds the viral RNA genome in virions. These interactions have specificity, as IN exhibits distinct preference for select viral RNA structural elements. We show that IN substitutions that selectively impair its binding to viral RNA result in eccentric, non-infectious virions without affecting nucleocapsid-RNA interactions. Likewise, ALLINIs impair IN binding to viral RNA in virions of wild-type, but not escape mutant, virus. These results reveal an unexpected biological role of IN binding to the viral RNA genome during virion morphogenesis and elucidate the mode of action of ALLINIs.


Subject(s)
Genome, Viral , HIV Integrase/metabolism , HIV-1/growth & development , RNA, Viral/metabolism , Virion/growth & development , HEK293 Cells , HIV Integrase/genetics , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/enzymology , Humans , Morphogenesis , Nucleocapsid/drug effects , Protein Binding , Virion/drug effects , Virion/enzymology , Virus Integration/drug effects
3.
Cell ; 160(3): 420-32, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25635456

ABSTRACT

The barrier to curing HIV-1 is thought to reside primarily in CD4(+) T cells containing silent proviruses. To characterize these latently infected cells, we studied the integration profile of HIV-1 in viremic progressors, individuals receiving antiretroviral therapy, and viremic controllers. Clonally expanded T cells represented the majority of all integrations and increased during therapy. However, none of the 75 expanded T cell clones assayed contained intact virus. In contrast, the cells bearing single integration events decreased in frequency over time on therapy, and the surviving cells were enriched for HIV-1 integration in silent regions of the genome. Finally, there was a strong preference for integration into, or in close proximity to, Alu repeats, which were also enriched in local hotspots for integration. The data indicate that dividing clonally expanded T cells contain defective proviruses and that the replication-competent reservoir is primarily found in CD4(+) T cells that remain relatively quiescent.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , Virus Integration , Virus Latency , Alu Elements , Clone Cells , Defective Viruses/genetics , Defective Viruses/physiology , HIV Infections/drug therapy , HIV-1/genetics , Humans , Immunologic Memory , Proviruses/physiology , Single-Cell Analysis
4.
Cell ; 155(3): 519-29, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243012

ABSTRACT

Despite significant advances in our understanding of HIV, a cure has not been realized for the more than 34 million infected with this virus. HIV is incurable because infected individuals harbor cells where the HIV provirus is integrated into the host's DNA but is not actively replicating and thus is not inhibited by antiviral drugs. Similarly, these latent viruses are not detected by the immune system. In this Review, we discuss HIV-1 latency and the mechanisms that allow this pathogenic retrovirus to hide and persist by exploiting the cellular vehicles of immunological memory.


Subject(s)
HIV Infections/drug therapy , HIV Infections/virology , HIV-1/physiology , Virus Latency , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , HIV Infections/transmission , HIV-1/genetics , Humans , Transcription, Genetic , Virus Integration
5.
N Engl J Med ; 390(22): 2047-2060, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38865660

ABSTRACT

BACKGROUND: The risk of second tumors after chimeric antigen receptor (CAR) T-cell therapy, especially the risk of T-cell neoplasms related to viral vector integration, is an emerging concern. METHODS: We reviewed our clinical experience with adoptive cellular CAR T-cell therapy at our institution since 2016 and ascertained the occurrence of second tumors. In one case of secondary T-cell lymphoma, a broad array of molecular, genetic, and cellular techniques were used to interrogate the tumor, the CAR T cells, and the normal hematopoietic cells in the patient. RESULTS: A total of 724 patients who had received T-cell therapies at our center were included in the study. A lethal T-cell lymphoma was identified in a patient who had received axicabtagene ciloleucel therapy for diffuse large B-cell lymphoma, and both lymphomas were deeply profiled. Each lymphoma had molecularly distinct immunophenotypes and genomic profiles, but both were positive for Epstein-Barr virus and were associated with DNMT3A and TET2 mutant clonal hematopoiesis. No evidence of oncogenic retroviral integration was found with the use of multiple techniques. CONCLUSIONS: Our results highlight the rarity of second tumors and provide a framework for defining clonal relationships and viral vector monitoring. (Funded by the National Cancer Institute and others.).


Subject(s)
Antineoplastic Agents, Immunological , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Lymphoma, T-Cell , Neoplasms, Second Primary , Receptors, Chimeric Antigen , Female , Humans , Middle Aged , Biological Products/adverse effects , Biological Products/therapeutic use , Clonal Hematopoiesis , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/genetics , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/therapy , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/etiology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Virus Integration
6.
Genome Res ; 33(8): 1395-1408, 2023 08.
Article in English | MEDLINE | ID: mdl-37463751

ABSTRACT

A weak palindromic nucleotide motif is the hallmark of retroviral integration site alignments. Given that the majority of target sequences are not palindromic, the current model explains the symmetry by an overlap of the nonpalindromic motif present on one of the half-sites of the sequences. Here, we show that the implementation of multicomponent mixture models allows for different interpretations consistent with the existence of both palindromic and nonpalindromic submotifs in the sets of integration site sequences. We further show that the weak palindromic motifs result from freely combined site-specific submotifs restricted to only a few positions proximal to the site of integration. The submotifs are formed by either palindrome-forming nucleotide preference or nucleotide exclusion. Using the mixture models, we also identify HIV-1-favored palindromic sequences in Alu repeats serving as local hotspots for integration. The application of the novel statistical approach provides deeper insight into the selection of retroviral integration sites and may prove to be a valuable tool in the analysis of any type of DNA motifs.


Subject(s)
Nucleotides , Virus Integration , Virus Integration/genetics , Nucleotide Motifs
7.
PLoS Pathog ; 20(5): e1012261, 2024 May.
Article in English | MEDLINE | ID: mdl-38805555

ABSTRACT

Marek's disease virus (MDV) vaccines were the first vaccines that protected against cancer. The avirulent turkey herpesvirus (HVT) was widely employed and protected billions of chickens from a deadly MDV infection. It is also among the most common vaccine vectors providing protection against a plethora of pathogens. HVT establishes latency in T-cells, allowing the vaccine virus to persist in the host for life. Intriguingly, the HVT genome contains telomeric repeat arrays (TMRs) at both ends; however, their role in the HVT life cycle remains elusive. We have previously shown that similar TMRs in the MDV genome facilitate its integration into host telomeres, which ensures efficient maintenance of the virus genome during latency and tumorigenesis. In this study, we investigated the role of the TMRs in HVT genome integration, latency, and reactivation in vitro and in vivo. Additionally, we examined HVT infection of feather follicles. We generated an HVT mutant lacking both TMRs (vΔTMR) that efficiently replicated in cell culture. We could demonstrate that wild type HVT integrates at the ends of chromosomes containing the telomeres in T-cells, while integration was severely impaired in the absence of the TMRs. To assess the role of TMRs in vivo, we infected one-day-old chickens with HVT or vΔTMR. vΔTMR loads were significantly reduced in the blood and hardly any virus was transported to the feather follicle epithelium where the virus is commonly shed. Strikingly, latency in the spleen and reactivation of the virus were severely impaired in the absence of the TMRs, indicating that the TMRs are crucial for the establishment of latency and reactivation of HVT. Our findings revealed that the TMRs facilitate integration of the HVT genome into host chromosomes, which ensures efficient persistence in the host, reactivation, and transport of the virus to the skin.


Subject(s)
Chickens , Marek Disease , Telomere , Virus Integration , Virus Latency , Animals , Chickens/virology , Telomere/genetics , Telomere/virology , Marek Disease/virology , Marek Disease/immunology , Marek Disease/prevention & control , Genetic Vectors , Herpesvirus 1, Meleagrid/genetics , Herpesvirus 1, Meleagrid/immunology , Marek Disease Vaccines/immunology , Marek Disease Vaccines/genetics , Genome, Viral , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/immunology , Repetitive Sequences, Nucleic Acid , Poultry Diseases/virology , Poultry Diseases/immunology , Poultry Diseases/prevention & control
8.
Blood ; 143(23): 2373-2385, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38452208

ABSTRACT

ABSTRACT: Gene therapy using adeno-associated virus (AAV) vectors is a promising approach for the treatment of monogenic disorders. Long-term multiyear transgene expression has been demonstrated in animal models and clinical studies. Nevertheless, uncertainties remain concerning the nature of AAV vector persistence and whether there is a potential for genotoxicity. Here, we describe the mechanisms of AAV vector persistence in the liver of a severe hemophilia A dog model (male = 4, hemizygous; and female = 4, homozygous), more than a decade after portal vein delivery. The predominant vector form was nonintegrated episomal structures with levels correlating with long-term transgene expression. Random integration was seen in all samples (median frequency, 9.3e-4 sites per cell), with small numbers of nonrandom common integration sites associated with open chromatin. No full-length integrated vectors were found, supporting predominant episomal vector-mediated long-term transgene expression. Despite integration, this was not associated with oncogene upregulation or histopathological evidence of tumorigenesis. These findings support the long-term safety of this therapeutic modality.


Subject(s)
Dependovirus , Factor VIII , Genetic Therapy , Genetic Vectors , Hemophilia A , Liver , Animals , Dogs , Dependovirus/genetics , Hemophilia A/genetics , Hemophilia A/therapy , Genetic Vectors/genetics , Liver/metabolism , Liver/pathology , Male , Genetic Therapy/methods , Female , Factor VIII/genetics , Gene Transfer Techniques , Virus Integration , Transgenes , Disease Models, Animal
9.
EMBO Rep ; 25(2): 725-744, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38177923

ABSTRACT

Viral infection often trigger an ATM serine/threonine kinase (ATM)-dependent DNA damage response in host cells that suppresses viral replication. Viruses evolved different strategies to counteract this antiviral surveillance system. Here, we report that human herpesvirus 6B (HHV-6B) infection causes genomic instability by suppressing ATM signaling in host cells. Expression of immediate-early protein 1 (IE1) phenocopies this phenotype and blocks homology-directed double-strand break repair. Mechanistically, IE1 interacts with NBS1, and inhibits ATM signaling through two distinct domains. HHV-6B seems to efficiently inhibit ATM signaling as further depletion of either NBS1 or ATM do not significantly boost viral replication in infected cells. Interestingly, viral integration of HHV-6B into the host's telomeres is not strictly dependent on NBS1, challenging current models where integration occurs through homology-directed repair. Given that spontaneous IE1 expression has been detected in cells of subjects with inherited chromosomally-integrated form of HHV-6B (iciHHV-6B), a condition associated with several health conditions, our results raise the possibility of a link between genomic instability and the development of iciHHV-6-associated diseases.


Subject(s)
Herpesvirus 6, Human , Immediate-Early Proteins , Roseolovirus Infections , Humans , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/metabolism , Roseolovirus Infections/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Virus Integration , Genomic Instability , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
10.
Nature ; 585(7824): 261-267, 2020 09.
Article in English | MEDLINE | ID: mdl-32848246

ABSTRACT

Sustained, drug-free control of HIV-1 replication is naturally achieved in less than 0.5% of infected individuals (here termed 'elite controllers'), despite the presence of a replication-competent viral reservoir1. Inducing such an ability to spontaneously maintain undetectable plasma viraemia is a major objective of HIV-1 cure research, but the characteristics of proviral reservoirs in elite controllers remain to be determined. Here, using next-generation sequencing of near-full-length single HIV-1 genomes and corresponding chromosomal integration sites, we show that the proviral reservoirs of elite controllers frequently consist of oligoclonal to near-monoclonal clusters of intact proviral sequences. In contrast to individuals treated with long-term antiretroviral therapy, intact proviral sequences from elite controllers were integrated at highly distinct sites in the human genome and were preferentially located in centromeric satellite DNA or in Krüppel-associated box domain-containing zinc finger genes on chromosome 19, both of which are associated with heterochromatin features. Moreover, the integration sites of intact proviral sequences from elite controllers showed an increased distance to transcriptional start sites and accessible chromatin of the host genome and were enriched in repressive chromatin marks. These data suggest that a distinct configuration of the proviral reservoir represents a structural correlate of natural viral control, and that the quality, rather than the quantity, of viral reservoirs can be an important distinguishing feature for a functional cure of HIV-1 infection. Moreover, in one elite controller, we were unable to detect intact proviral sequences despite analysing more than 1.5 billion peripheral blood mononuclear cells, which raises the possibility that a sterilizing cure of HIV-1 infection, which has previously been observed only following allogeneic haematopoietic stem cell transplantation2,3, may be feasible in rare instances.


Subject(s)
Gene Silencing , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Heterochromatin/genetics , Proviruses/genetics , Virus Integration/genetics , Virus Latency/genetics , Adult , Aged , Centromere/genetics , Chromosomes, Human, Pair 19/genetics , DNA, Satellite/genetics , Female , Genome, Viral/genetics , HIV Infections/blood , HIV-1/isolation & purification , Heterochromatin/metabolism , Humans , Male , Middle Aged , Proviruses/isolation & purification , Repressor Proteins/genetics , Transcription Initiation Site
11.
Nature ; 583(7817): 578-584, 2020 07.
Article in English | MEDLINE | ID: mdl-32699395

ABSTRACT

Bats possess extraordinary adaptations, including flight, echolocation, extreme longevity and unique immunity. High-quality genomes are crucial for understanding the molecular basis and evolution of these traits. Here we incorporated long-read sequencing and state-of-the-art scaffolding protocols1 to generate, to our knowledge, the first reference-quality genomes of six bat species (Rhinolophus ferrumequinum, Rousettus aegyptiacus, Phyllostomus discolor, Myotis myotis, Pipistrellus kuhlii and Molossus molossus). We integrated gene projections from our 'Tool to infer Orthologs from Genome Alignments' (TOGA) software with de novo and homology gene predictions as well as short- and long-read transcriptomics to generate highly complete gene annotations. To resolve the phylogenetic position of bats within Laurasiatheria, we applied several phylogenetic methods to comprehensive sets of orthologous protein-coding and noncoding regions of the genome, and identified a basal origin for bats within Scrotifera. Our genome-wide screens revealed positive selection on hearing-related genes in the ancestral branch of bats, which is indicative of laryngeal echolocation being an ancestral trait in this clade. We found selection and loss of immunity-related genes (including pro-inflammatory NF-κB regulators) and expansions of anti-viral APOBEC3 genes, which highlights molecular mechanisms that may contribute to the exceptional immunity of bats. Genomic integrations of diverse viruses provide a genomic record of historical tolerance to viral infection in bats. Finally, we found and experimentally validated bat-specific variation in microRNAs, which may regulate bat-specific gene-expression programs. Our reference-quality bat genomes provide the resources required to uncover and validate the genomic basis of adaptations of bats, and stimulate new avenues of research that are directly relevant to human health and disease1.


Subject(s)
Adaptation, Physiological/genetics , Chiroptera/genetics , Evolution, Molecular , Genome/genetics , Genomics/standards , Adaptation, Physiological/immunology , Animals , Chiroptera/classification , Chiroptera/immunology , DNA Transposable Elements/genetics , Immunity/genetics , Molecular Sequence Annotation/standards , Phylogeny , RNA, Untranslated/genetics , Reference Standards , Reproducibility of Results , Virus Integration/genetics , Viruses/genetics
12.
J Biol Chem ; 300(6): 107374, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762180

ABSTRACT

The pre-integration steps of the HIV-1 viral cycle are some of the most valuable targets of recent therapeutic innovations. HIV-1 integrase (IN) displays multiple functions, thanks to its considerable conformational flexibility. Recently, such flexible proteins have been characterized by their ability to form biomolecular condensates as a result of Liquid-Liquid-Phase-Separation (LLPS), allowing them to evolve in a restricted microenvironment within cells called membrane-less organelles (MLO). The LLPS context constitutes a more physiological approach to study the integration of molecular mechanisms performed by intasomes (complexes containing viral DNA, IN, and its cellular cofactor LEDGF/p75). We investigated here if such complexes can form LLPS in vitro and if IN enzymatic activities were affected by this LLPS environment. We observed that the LLPS formed by IN-LEDGF/p75 functional complexes modulate the in vitro IN activities. While the 3'-processing of viral DNA ends was drastically reduced inside LLPS, viral DNA strand transfer was strongly enhanced. These two catalytic IN activities appear thus tightly regulated by the environment encountered by intasomes.


Subject(s)
HIV Integrase , HIV-1 , Virus Integration , HIV Integrase/metabolism , HIV Integrase/chemistry , HIV Integrase/genetics , HIV-1/metabolism , HIV-1/physiology , Humans , DNA, Viral/metabolism , DNA, Viral/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/chemistry
13.
Genome Res ; 32(1): 55-70, 2022 01.
Article in English | MEDLINE | ID: mdl-34903527

ABSTRACT

Human papillomavirus (HPV) causes 5% of all cancers and frequently integrates into host chromosomes. The HPV oncoproteins E6 and E7 are necessary but insufficient for cancer formation, indicating that additional secondary genetic events are required. Here, we investigate potential oncogenic impacts of virus integration. Analysis of 105 HPV-positive oropharyngeal cancers by whole-genome sequencing detects virus integration in 77%, revealing five statistically significant sites of recurrent integration near genes that regulate epithelial stem cell maintenance (i.e., SOX2, TP63, FGFR, MYC) and immune evasion (i.e., CD274). Genomic copy number hyperamplification is enriched 16-fold near HPV integrants, and the extent of focal host genomic instability increases with their local density. The frequency of genes expressed at extreme outlier levels is increased 86-fold within ±150 kb of integrants. Across 95% of tumors with integration, host gene transcription is disrupted via intragenic integrants, chimeric transcription, outlier expression, gene breaking, and/or de novo expression of noncoding or imprinted genes. We conclude that virus integration can contribute to carcinogenesis in a large majority of HPV-positive oropharyngeal cancers by inducing extensive disruption of host genome structure and gene expression.


Subject(s)
Alphapapillomavirus , Oncogene Proteins, Viral , Oropharyngeal Neoplasms , Alphapapillomavirus/metabolism , Carcinogenesis , Humans , Oncogene Proteins, Viral/genetics , Oropharyngeal Neoplasms/genetics , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Virus Integration/genetics
14.
Genes Cells ; 29(7): 532-548, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38715205

ABSTRACT

Most cervical cancers are caused by human papillomavirus (HPV) infection. In HeLa cells, the HPV18 viral genome is integrated at chromosome 8q24.21 and activates transcription of the proto-oncogene c-Myc. However, the mechanism of how the integrated HPV genome and its transcribed RNAs exhibit transcription activation function has not been fully elucidated. In this study, we found that HPV18 transcripts contain an enhancer RNA-like function to activate proximal genes including CCAT1-5L and c-Myc. We showed that the human genome-integrated HPV18 genes are activated by transcription coregulators including BRD4 and Mediator. The transcribed HPV18 RNAs form a liquid-like condensate at chromosome 8q24.21 locus, which in turn accumulates RNA polymerase II. Moreover, we focused on a relatively uncharacterized transcript from the upstream region of CCAT1, named URC. The URC RNA is transcribed as a chimera RNA with HPV18 and is composed of the 3'-untranslated region of the HPV18 transcript. We experimentally showed that the URC contributes to stabilization of HPV18 RNAs by supplying a polyadenylation site for the HPV18 transcript. Our findings suggest that integrated HPV18 at 8q24.21 locus produces HPV18-URC chimera RNA and promotes tumorigenesis through RNA-based condensate formation.


Subject(s)
Genome, Viral , Human papillomavirus 18 , Proto-Oncogene Mas , Humans , Human papillomavirus 18/genetics , HeLa Cells , RNA, Viral/genetics , RNA, Viral/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Papillomavirus Infections/virology , Papillomavirus Infections/genetics , Virus Integration , Transcription, Genetic , Female , Genome, Human , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Bromodomain Containing Proteins
15.
J Virol ; 98(2): e0165523, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38214547

ABSTRACT

Within-host HIV populations continually diversify during untreated infection, and this diversity persists within infected cell reservoirs during antiretroviral therapy (ART). Achieving a better understanding of on-ART proviral evolutionary dynamics, and a better appreciation of how the overall persisting pool of (largely genetically defective) proviruses differs from the much smaller replication-competent HIV reservoir, is critical to HIV cure efforts. We reconstructed within-host HIV evolutionary histories in blood from seven participants of the Women's Interagency HIV Study who experienced HIV seroconversion, and used these data to characterize the diversity, lineage origins, and ages of proviral env-gp120 sequences sampled longitudinally up to 12 years on ART. We also studied HIV sequences emerging from the reservoir in two participants. We observed that proviral clonality generally increased over time on ART, with clones frequently persisting long term. While on-ART proviral integration dates generally spanned the duration of untreated infection, HIV emerging in plasma was exclusively younger (i.e., dated to the years immediately pre-ART). The genetic and age distributions of distinct proviral sequences remained stable during ART in all but one participant, in whom there was evidence that younger proviruses had been preferentially eliminated after 12 years on ART. Analysis of the gag region in three participants corroborated our env-gp120-based observations, indicating that our observations are not influenced by the HIV region studied. Our results underscore the remarkable genetic stability of the distinct proviral sequences that persist in blood during ART. Our results also suggest that the replication-competent HIV reservoir is a genetically restricted, younger subset of this overall proviral pool.IMPORTANCECharacterizing the genetically diverse HIV sequences that persist in the reservoir despite antiretroviral therapy (ART) is critical to cure efforts. Our observations confirm that proviruses persisting in blood on ART, which are largely genetically defective, broadly reflect the extent of within-host HIV evolution pre-ART. Moreover, on-ART clonal expansion is not appreciably accompanied by the loss of distinct proviral lineages. In fact, on-ART proviral genetic composition remained stable in all but one participant, in whom, after 12 years on ART, proviruses dating to around near ART initiation had been preferentially eliminated. We also identified recombinant proviruses between parental sequence fragments of different ages. Though rare, such sequences suggest that reservoir cells can be superinfected with HIV from another infection era. Overall, our finding that the replication-competent reservoir in blood is a genetically restricted, younger subset of all persisting proviruses suggests that HIV cure strategies will need to eliminate a reservoir that differs in key respects from the overall proviral pool.


Subject(s)
HIV Infections , HIV-1 , Proviruses , Child , Female , Humans , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/genetics , Proviruses/genetics , Viral Load , Virus Integration
16.
J Virol ; 98(2): e0186223, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38294250

ABSTRACT

The primary mode of infection by human T-cell leukemia virus type 1 (HTLV-1) is cell-to-cell transmission during contact between infected cells and target cells. Cell-free HTLV-1 infections are known to be less efficient than infections with other retroviruses, and transmission of free HTLV-1 is considered not to occur in vivo. However, it has been demonstrated that cell-free HTLV-1 virions can infect primary lymphocytes and dendritic cells in vitro, and that virions embedded in biofilms on cell membranes can contribute to transmission. The establishment of an efficient cell-free HTLV-1 infection model would be a useful tool for analyzing the replication process of HTLV-1 and the clonal expansion of infected cells. We first succeeded in obtaining supernatants with high-titer cell-free HTLV-1 using a highly efficient virus-producing cell line. The HTLV-1 virions retained the structural characteristics of retroviruses. Using this cell-free infection model, we confirmed that a variety of cell lines and primary cultured cells can be infected with HTLV-1 and demonstrated that the provirus was randomly integrated into all chromosomes in the target cells. The provirus-integrated cell lines were HTLV-1-productive. Furthermore, we demonstrated for the first time that cell-free HTLV-1 is infectious in vivo using a humanized mouse model. These results indicate that this cell-free infection model recapitulates the HTLV-1 life cycle, including entry, reverse transcription, integration into the host genome, viral replication, and secondary infection. The new cell-free HTLV-1 infection model is promising as a practical resource for studying HTLV-1 infection.IMPORTANCECo-culture of infected and target cells is frequently used for studying HTLV-1 infection. Although this method efficiently infects HTLV-1, the cell mixture is complex, and it is extremely difficult to distinguish donor infected cells from target cells. In contrast, cell-free HTLV-1 infection models allow for more strict experimental conditions. In this study, we established a novel and efficient cell-free HTLV-1 infection model. Using this model, we successfully evaluated the infectivity titers of cell-free HTLV-1 as proviral loads (copies per 100 cells) in various cell lines, primary cultured cells, and a humanized mouse model. Interestingly, the HTLV-1-associated viral biofilms played an important role in enhancing the infectivity of the cell-free infection model. This cell-free HTLV-1 infection model reproduces the replication cycle of HTLV-1 and provides a simple, powerful, and alternative tool for researching HTLV-1 infection.


Subject(s)
Cell-Free System , HTLV-I Infections , Human T-lymphotropic virus 1 , Animals , Humans , Mice , HTLV-I Infections/transmission , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/growth & development , Human T-lymphotropic virus 1/pathogenicity , Human T-lymphotropic virus 1/physiology , Lymphocytes/virology , Proviruses/genetics , Proviruses/metabolism , Virus Replication , Cell-Free System/virology , Cell Line , Cells, Cultured , Virus Internalization , Reverse Transcription , Biofilms , Virus Integration
17.
Blood ; 141(19): 2316-2329, 2023 05 11.
Article in English | MEDLINE | ID: mdl-36790505

ABSTRACT

Adeno-associated virus (AAV) vectors have been successfully exploited in gene therapy applications for the treatment of several genetic disorders. AAV is considered an episomal vector, but it has been shown to integrate within the host cell genome after the generation of double-strand DNA breaks or nicks. Although AAV integration raises some safety concerns, it can also provide therapeutic benefit; the direct intrathymic injection of an AAV harboring a therapeutic transgene results in integration in T-cell progenitors and long-term T-cell immunity. To assess the mechanisms of AAV integration, we retrieved and analyzed hundreds of AAV integration sites from lymph node-derived mature T cells and compared these with liver and brain tissue from treated mice. Notably, we found that although AAV integrations in the liver and brain were distributed across the entire mouse genome, >90% of the integrations in T cells were clustered within the T-cell receptor α, ß, and γ genes. More precisely, the insertion mapped to DNA breaks created by the enzymatic activity of recombination activating genes (RAGs) during variable, diversity, and joining recombination. Our data indicate that RAG activity during T-cell receptor maturation induces a site-specific integration of AAV genomes and opens new therapeutic avenues for achieving long-term AAV-mediated gene transfer in dividing cells.


Subject(s)
Genetic Therapy , Genetic Vectors , Mice , Animals , Genetic Vectors/genetics , Transgenes , Plasmids , Genetic Therapy/methods , Receptors, Antigen, T-Cell/genetics , Dependovirus/genetics , Virus Integration
18.
Trends Immunol ; 43(6): 449-458, 2022 06.
Article in English | MEDLINE | ID: mdl-35490134

ABSTRACT

Several viruses hide in the genome of their host. To complete their replication cycle, they need to integrate in the form of a provirus and express their genes. In vertebrates, integrated viruses can be silenced by chromatin, implying that some specific mechanisms exist to detect non-self genes. The known mechanisms depend on sequence features of retroelements, but the fluctuations of virus expression suggest that other determinants also exist. Here we review the mechanisms allowing chromatin to silence integrated viruses and propose that DNA repair may help flag them as 'non-self' shortly after their genomic insertion.


Subject(s)
Chromatin , Virus Integration , Animals , Chromatin/genetics , Gene Silencing , Humans , Proviruses/genetics , Virus Integration/genetics
19.
Trends Immunol ; 43(8): 617-629, 2022 08.
Article in English | MEDLINE | ID: mdl-35817699

ABSTRACT

Despite antiretroviral therapy (ART), HIV-1 persists as proviruses integrated into the genomic DNA of CD4+ T cells. The mechanisms underlying the persistence and clonal expansion of these cells remain incompletely understood. Cases have been described in which proviral integration can alter host gene expression to drive cellular proliferation. Here, we review observations from other genome-integrating human viruses to propose additional putative modalities by which HIV-1 integration may alter cellular function to favor persistence, such as by altering susceptibility to cytotoxicity in virus-expressing cells. We propose that signals implicating such mechanisms may have been masked thus far by the preponderance of defective and/or nonreactivatable HIV-1 proviruses, but could be revealed by focusing on the integration sites of intact proviruses with expression potential.


Subject(s)
HIV Infections , HIV-1 , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , Humans , Proviruses/genetics , Virus Integration
20.
Nucleic Acids Res ; 51(9): 4237-4251, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36864748

ABSTRACT

Human papillomavirus (HPV) integration is a critical step in cervical cancer development; however, the oncogenic mechanism at the genome-wide transcriptional level is still poorly understood. In this study, we employed integrative analysis on multi-omics data of six HPV-positive and three HPV-negative cell lines. Through HPV integration detection, super-enhancer (SE) identification, SE-associated gene expression and extrachromosomal DNA (ecDNA) investigation, we aimed to explore the genome-wide transcriptional influence of HPV integration. We identified seven high-ranking cellular SEs generated by HPV integration in total (the HPV breakpoint-induced cellular SEs, BP-cSEs), leading to intra-chromosomal and inter-chromosomal regulation of chromosomal genes. The pathway analysis revealed that the dysregulated chromosomal genes were correlated to cancer-related pathways. Importantly, we demonstrated that BP-cSEs existed in the HPV-human hybrid ecDNAs, explaining the above transcriptional alterations. Our results suggest that HPV integration generates cellular SEs that function as ecDNA to regulate unconstrained transcription, expanding the tumorigenic mechanism of HPV integration and providing insights for developing new diagnostic and therapeutic strategies.


Subject(s)
DNA , Enhancer Elements, Genetic , Genome, Human , Human Papillomavirus Viruses , Papillomavirus Infections , Transcription, Genetic , Uterine Cervical Neoplasms , Virus Integration , Female , Humans , Human Papillomavirus Viruses/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Virus Integration/genetics , Enhancer Elements, Genetic/genetics , DNA/genetics , DNA/metabolism , Genome, Human/genetics , Carcinogenesis , Chromosome Breakpoints , Chromosomes, Human/genetics
SELECTION OF CITATIONS
SEARCH DETAIL