Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 15.054
1.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article En | MEDLINE | ID: mdl-38928103

The maturation of HIV-1 virions is a crucial process in viral replication. Although T-cells are a primary source of virus production, much of our understanding of virion maturation comes from studies using the HEK293T human embryonic kidney cell line. Notably, there is a lack of comparative analyses between T-cells and HEK293T cells in terms of virion maturation efficiency in existing literature. We previously developed an advanced virion visualization system based on the FRET principle, enabling the effective distinction between immature and mature virions via fluorescence microscopy. In this study, we utilized pseudotyped, single-round infectious viruses tagged with FRET labels (HIV-1 Gag-iFRET∆Env) derived from Jurkat (a human T-lymphocyte cell line) and HEK293T cells to evaluate their virion maturation rates. HEK293T-derived virions demonstrated a maturity rate of 81.79%, consistent with other studies and our previous findings. However, virions originating from Jurkat cells demonstrated a significantly reduced maturation rate of 68.67% (p < 0.0001). Correspondingly, viruses produced from Jurkat cells exhibited significantly reduced infectivity compared to those derived from HEK293T cells, with the relative infectivity measured at 65.3%. This finding is consistent with the observed relative maturation rate of viruses produced by Jurkat cells. These findings suggest that initiation of virion maturation directly correlates with viral infectivity. Our observation highlights the dynamic nature of virus-host interactions and their implications for virion production and infectivity.


Fluorescence Resonance Energy Transfer , HIV-1 , Virion , Humans , HIV-1/physiology , HIV-1/pathogenicity , HEK293 Cells , Virion/metabolism , Jurkat Cells , Fluorescence Resonance Energy Transfer/methods , Virus Replication , Virus Assembly , HIV Infections/virology
2.
Article En | MEDLINE | ID: mdl-38902848

Despite the success of antiretroviral therapy, human immunodeficiency virus (HIV) cannot be cured because of a reservoir of latently infected cells that evades therapy. To understand the mechanisms of HIV latency, we employed an integrated single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin with sequencing (scATAC-seq) approach to simultaneously profile the transcriptomic and epigenomic characteristics of ∼ 125,000 latently infected primary CD4+ T cells after reactivation using three different latency reversing agents. Differentially expressed genes and differentially accessible motifs were used to examine transcriptional pathways and transcription factor (TF) activities across the cell population. We identified cellular transcripts and TFs whose expression/activity was correlated with viral reactivation and demonstrated that a machine learning model trained on these data was 75%-79% accurate at predicting viral reactivation. Finally, we validated the role of two candidate HIV-regulating factors, FOXP1 and GATA3, in viral transcription. These data demonstrate the power of integrated multimodal single-cell analysis to uncover novel relationships between host cell factors and HIV latency.


CD4-Positive T-Lymphocytes , GATA3 Transcription Factor , HIV-1 , Single-Cell Analysis , Virus Activation , Virus Latency , Virus Latency/genetics , Humans , Virus Activation/genetics , Single-Cell Analysis/methods , HIV-1/genetics , HIV-1/physiology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , HIV Infections/virology , HIV Infections/genetics , HIV Infections/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Transcriptome/genetics , Gene Expression Regulation, Viral
3.
PLoS Comput Biol ; 20(6): e1012129, 2024 Jun.
Article En | MEDLINE | ID: mdl-38848426

Understanding the dynamics of acute HIV infection can offer valuable insights into the early stages of viral behavior, potentially helping uncover various aspects of HIV pathogenesis. The standard viral dynamics model explains HIV viral dynamics during acute infection reasonably well. However, the model makes simplifying assumptions, neglecting some aspects of HIV infection. For instance, in the standard model, target cells are infected by a single HIV virion. Yet, cellular multiplicity of infection (MOI) may have considerable effects in pathogenesis and viral evolution. Further, when using the standard model, we take constant infected cell death rates, simplifying the dynamic immune responses. Here, we use four models-1) the standard viral dynamics model, 2) an alternate model incorporating cellular MOI, 3) a model assuming density-dependent death rate of infected cells and 4) a model combining (2) and (3)-to investigate acute infection dynamics in 43 people living with HIV very early after HIV exposure. We find that all models qualitatively describe the data, but none of the tested models is by itself the best to capture different kinds of heterogeneity. Instead, different models describe differing features of the dynamics more accurately. For example, while the standard viral dynamics model may be the most parsimonious across study participants by the corrected Akaike Information Criterion (AICc), we find that viral peaks are better explained by a model allowing for cellular MOI, using a linear regression analysis as analyzed by R2. These results suggest that heterogeneity in within-host viral dynamics cannot be captured by a single model. Depending on the specific aspect of interest, a corresponding model should be employed.


Cell Death , HIV Infections , Models, Biological , HIV Infections/virology , HIV Infections/physiopathology , Humans , Cell Death/physiology , HIV-1/physiology , HIV-1/pathogenicity , Computational Biology , Viral Load , Male , Adult , Acute Disease , Female
4.
PLoS Pathog ; 20(6): e1012281, 2024 Jun.
Article En | MEDLINE | ID: mdl-38848441

Understanding the mechanisms that drive HIV expression and latency is a key goal for achieving an HIV cure. Here we investigate the role of the SETD2 histone methyltransferase, which deposits H3K36 trimethylation (H3K36me3), in HIV infection. We show that prevention of H3K36me3 by a potent and selective inhibitor of SETD2 (EPZ-719) leads to reduced post-integration viral gene expression and accelerated emergence of latently infected cells. CRISPR/Cas9-mediated knockout of SETD2 in primary CD4 T cells confirmed the role of SETD2 in HIV expression. Transcriptomic profiling of EPZ-719-exposed HIV-infected cells identified numerous pathways impacted by EPZ-719. Notably, depletion of H3K36me3 prior to infection did not prevent HIV integration but resulted in a shift of integration sites from highly transcribed genes to quiescent chromatin regions and to polycomb repressed regions. We also observed that SETD2 inhibition did not apparently affect HIV RNA levels, indicating a post-transcriptional mechanism affecting HIV expression. Viral RNA splicing was modestly reduced in the presence of EPZ-719. Intriguingly, EPZ-719 exposure enhanced responsiveness of latent HIV to the HDAC inhibitor vorinostat, suggesting that H3K36me3 can contribute to a repressive chromatin state at the HIV locus. These results identify SETD2 and H3K36me3 as novel regulators of HIV integration, expression and latency.


HIV Infections , HIV-1 , Histone-Lysine N-Methyltransferase , Virus Latency , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Virus Latency/physiology , HIV Infections/virology , HIV Infections/metabolism , HIV Infections/genetics , HIV-1/physiology , HIV-1/genetics , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Viral
5.
Front Immunol ; 15: 1374301, 2024.
Article En | MEDLINE | ID: mdl-38835765

Background: Human immunodeficiency virus (HIV) affects nearly 40 million people globally, with roughly 80% of all people living with HIV receiving antiretroviral therapy. Antiretroviral treatment suppresses viral load in peripheral tissues but does not effectively penetrate the blood-brain barrier. Thus, viral reservoirs persist in the central nervous system and continue to produce low levels of inflammatory factors and early viral proteins, including the transactivator of transcription (Tat). HIV Tat is known to contribute to chronic neuroinflammation and synaptodendritic damage, which is associated with the development of cognitive, motor, and/or mood problems, collectively known as HIV-associated neurocognitive disorders (HAND). Cannabinoid anti-inflammatory effects are well documented, but therapeutic utility of cannabis remains limited due to its psychotropic effects, including alterations within brain regions encoding reward processing and motivation, such as the nucleus accumbens. Alternatively, inhibiting monoacylglycerol lipase (MAGL) has demonstrated therapeutic potential through interactions with the endocannabinoid system. Methods: The present study utilized a reward-related operant behavioral task to quantify motivated behavior in female Tat transgenic mice treated with vehicle or MAGL inhibitor MJN110 (1 mg/kg). Brain tissue was collected to assess dendritic injury and neuroinflammatory profiles, including dendritic microtubule-associated protein (MAP2ab) intensity, microglia density, microglia morphology, astrocyte density, astrocytic interleukin-1ß (IL-1ß) colocalization, and various lipid mediators. Results: No significant behavioral differences were observed; however, MJN110 protected against Tat-induced dendritic injury by significantly upregulating MAP2ab intensity in the nucleus accumbens and in the infralimbic cortex of Tat(+) mice. No or only minor effects were noted for Iba-1+ microglia density and/or microglia morphology. Further, Tat increased GFAP+ astrocyte density in the infralimbic cortex and GFAP+ astrocytic IL-1ß colocalization in the nucleus accumbens, with MJN110 significantly reducing these measures in Tat(+) subjects. Lastly, selected HETE-related inflammatory lipid mediators in the striatum were downregulated by chronic MJN110 treatment. Conclusions: These findings demonstrate anti-inflammatory and neuroprotective properties of MJN110 without cannabimimetic behavioral effects and suggest a promising alternative to cannabis for managing neuroinflammation.


HIV-1 , Mice, Transgenic , Monoacylglycerol Lipases , Neuroinflammatory Diseases , Animals , Mice , HIV-1/physiology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Female , Monoacylglycerol Lipases/antagonists & inhibitors , Monoacylglycerol Lipases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV Infections/drug therapy , Humans , Brain/drug effects , Brain/metabolism , Brain/virology , Brain/pathology , Disease Models, Animal , Microglia/drug effects , Microglia/metabolism , AIDS Dementia Complex/drug therapy
6.
BMC Res Notes ; 17(1): 153, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38835056

OBJECTIVE: Fourth-generation HIV Ag/Ab Combo assay is used for HIV screening of blood for transfusion in developing countries, however, the sensitivity of the assay is questionable during the acute phase of HIV infection. Thus, the study aimed to determine the effect of combining centrifugation with HIV-1 virion lysis on the sensitivity of the fourth-generation HIV Ag/Ab combo assay. RESULTS: When the 50 HIV-1 antibody-negative samples were run on the fourth-generation HIV Ag/Ab combo assay, 8 (16%) were positive following centrifugation, 13 (26%) were positive following lysis while 25 (50%) were positive after combining centrifugation with HIV-1 virion lysis.


Centrifugation , HIV Antibodies , HIV Infections , HIV-1 , Sensitivity and Specificity , Virion , HIV-1/immunology , HIV-1/physiology , Humans , Centrifugation/methods , HIV Infections/diagnosis , HIV Infections/virology , HIV Infections/immunology , HIV Infections/blood , HIV Antibodies/blood , HIV Antibodies/immunology , Virion/isolation & purification , Virion/immunology , HIV Antigens/immunology , HIV Antigens/blood
7.
Retrovirology ; 21(1): 13, 2024 Jun 19.
Article En | MEDLINE | ID: mdl-38898526

Retroviruses exploit host proteins to assemble and release virions from infected cells. Previously, most studies focused on interacting partners of retroviral Gag proteins that localize to the cytoplasm or plasma membrane. Given that several full-length Gag proteins have been found in the nucleus, identifying the Gag-nuclear interactome has high potential for novel findings involving previously unknown host processes. Here we systematically compared nuclear factors identified in published HIV-1 proteomic studies and performed our own mass spectrometry analysis using affinity-tagged HIV-1 and RSV Gag proteins mixed with nuclear extracts. We identified 57 nuclear proteins in common between HIV-1 and RSV Gag, and a set of nuclear proteins present in our analysis and ≥ 1 of the published HIV-1 datasets. Many proteins were associated with nuclear processes which could have functional consequences for viral replication, including transcription initiation/elongation/termination, RNA processing, splicing, and chromatin remodeling. Examples include facilitating chromatin remodeling to expose the integrated provirus, promoting expression of viral genes, repressing the transcription of antagonistic cellular genes, preventing splicing of viral RNA, altering splicing of cellular RNAs, or influencing viral or host RNA folding or RNA nuclear export. Many proteins in our pulldowns common to RSV and HIV-1 Gag are critical for transcription, including PolR2B, the second largest subunit of RNA polymerase II (RNAPII), and LEO1, a PAF1C complex member that regulates transcriptional elongation, supporting the possibility that Gag influences the host transcription profile to aid the virus. Through the interaction of RSV and HIV-1 Gag with splicing-related proteins CBLL1, HNRNPH3, TRA2B, PTBP1 and U2AF1, we speculate that Gag could enhance unspliced viral RNA production for translation and packaging. To validate one putative hit, we demonstrated an interaction of RSV Gag with Mediator complex member Med26, required for RNA polymerase II-mediated transcription. Although 57 host proteins interacted with both Gag proteins, unique host proteins belonging to each interactome dataset were identified. These results provide a strong premise for future functional studies to investigate roles for these nuclear host factors that may have shared functions in the biology of both retroviruses, as well as functions specific to RSV and HIV-1, given their distinctive hosts and molecular pathology.


Gene Products, gag , HIV-1 , Humans , HIV-1/physiology , HIV-1/genetics , Gene Products, gag/metabolism , Gene Products, gag/genetics , Cell Nucleus/metabolism , Cell Nucleus/virology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , Rous sarcoma virus/physiology , Rous sarcoma virus/genetics , Proteomics , Host-Pathogen Interactions , Virus Replication , Host Microbial Interactions , Mass Spectrometry
8.
Nat Commun ; 15(1): 5290, 2024 Jun 21.
Article En | MEDLINE | ID: mdl-38906865

Long-term non-progressors (LTNPs) of HIV-1 infection may provide important insights into mechanisms involved in viral control and pathogenesis. Here, our results suggest that the ribosomal protein lateral stalk subunit P1 (RPLP1) is expressed at higher levels in LTNPs compared to regular progressors (RPs). Functionally, RPLP1 inhibits transcription of clade B HIV-1 strains by occupying the C/EBPß binding sites in the viral long terminal repeat (LTR). This interaction requires the α-helixes 2 and 4 domains of RPLP1 and is evaded by HIV-1 group M subtype C and group N, O and P strains that do not require C/EBPß for transcription. We further demonstrate that HIV-1-induced translocation of RPLP1 from the cytoplasm to the nucleus is essential for antiviral activity. Finally, knock-down of RPLP1 promotes reactivation of latent HIV-1 proviruses. Thus, RPLP1 may play a role in the maintenance of HIV-1 latency and resistance to RPLP1 restriction may contribute to the effective spread of clade C HIV-1 strains.


CCAAT-Enhancer-Binding Protein-beta , HIV Infections , HIV Long Terminal Repeat , HIV-1 , Ribosomal Proteins , HIV-1/genetics , HIV-1/metabolism , HIV-1/physiology , Humans , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , HIV Long Terminal Repeat/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , HIV Infections/virology , HIV Infections/metabolism , HIV Infections/genetics , Transcription, Genetic , Protein Binding , Virus Latency/genetics , Binding Sites , Gene Expression Regulation, Viral , HEK293 Cells , Cell Nucleus/metabolism
9.
Retrovirology ; 21(1): 12, 2024 Jun 17.
Article En | MEDLINE | ID: mdl-38886829

An essential regulatory hub for retroviral replication events, the 5' untranslated region (UTR) encodes an ensemble of cis-acting replication elements that overlap in a logical manner to carry out divergent RNA activities in cells and in virions. The primer binding site (PBS) and primer activation sequence initiate the reverse transcription process in virions, yet overlap with structural elements that regulate expression of the complex viral proteome. PBS-segment also encompasses the attachment site for Integrase to cut and paste the 3' long terminal repeat into the host chromosome to form the provirus and purine residues necessary to execute the precise stoichiometry of genome-length transcripts and spliced viral RNAs. Recent genetic mapping, cofactor affinity experiments, NMR and SAXS have elucidated that the HIV-1 PBS-segment folds into a three-way junction structure. The three-way junction structure is recognized by the host's nuclear RNA helicase A/DHX9 (RHA). RHA tethers host trimethyl guanosine synthase 1 to the Rev/Rev responsive element (RRE)-containing RNAs for m7-guanosine Cap hyper methylation that bolsters virion infectivity significantly. The HIV-1 trimethylated (TMG) Cap licenses specialized translation of virion proteins under conditions that repress translation of the regulatory proteins. Clearly host-adaption and RNA shapeshifting comprise the fundamental basis for PBS-segment orchestrating both reverse transcription of virion RNA and the nuclear modification of m7G-Cap for biphasic translation of the complex viral proteome. These recent observations, which have exposed even greater complexity of retroviral RNA biology than previously established, are the impetus for this article. Basic research to fully comprehend the marriage of PBS-segment structures and host RNA binding proteins that carry out retroviral early and late replication events is likely to expose an immutable virus-specific therapeutic target to attenuate retrovirus proliferation.


5' Untranslated Regions , HIV-1 , RNA, Viral , Virus Replication , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , HIV-1/physiology , HIV-1/genetics , Binding Sites , Gene Expression Regulation, Viral , Reverse Transcription , Retroviridae/physiology , Retroviridae/genetics
10.
Viruses ; 16(6)2024 May 29.
Article En | MEDLINE | ID: mdl-38932164

The HIV-1 nucleocapsid protein (NC) is a multifunctional viral protein necessary for HIV-1 replication. Recent studies have demonstrated that reverse transcription (RT) completes in the intact viral capsid, and the timing of RT and uncoating are correlated. How the small viral core stably contains the ~10 kbp double stranded (ds) DNA product of RT, and the role of NC in this process, are not well understood. We showed previously that NC binds and saturates dsDNA in a non-specific electrostatic binding mode that triggers uniform DNA self-attraction, condensing dsDNA into a tight globule against extending forces up to 10 pN. In this study, we use optical tweezers and atomic force microscopy to characterize the role of NC's basic residues in dsDNA condensation. Basic residue mutations of NC lead to defective interaction with the dsDNA substrate, with the constant force plateau condensation observed with wild-type (WT) NC missing or diminished. These results suggest that NC's high positive charge is essential to its dsDNA condensing activity, and electrostatic interactions involving NC's basic residues are responsible in large part for the conformation, size, and stability of the dsDNA-protein complex inside the viral core. We observe DNA re-solubilization and charge reversal in the presence of excess NC, consistent with the electrostatic nature of NC-induced DNA condensation. Previous studies of HIV-1 replication in the presence of the same cationic residue mutations in NC showed significant defects in both single- and multiple-round viral infectivity. Although NC participates in many stages of viral replication, our results are consistent with the hypothesis that cationic residue mutations inhibit genomic DNA condensation, resulting in increased premature capsid uncoating and contributing to viral replication defects.


DNA, Viral , HIV-1 , Reverse Transcription , HIV-1/genetics , HIV-1/physiology , HIV-1/chemistry , HIV-1/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/chemistry , Humans , Cations/metabolism , Virus Replication , Microscopy, Atomic Force , Virion/metabolism , Virion/genetics , Virion/chemistry , Mutation
11.
Viruses ; 16(6)2024 Jun 03.
Article En | MEDLINE | ID: mdl-38932195

Antiretroviral treatments have notably extended the lives of individuals with HIV and reduced the occurrence of comorbidities, including ocular manifestations. The involvement of endoplasmic reticulum (ER) stress in HIV-1 pathogenesis raises questions about its correlation with cellular senescence or its role in initiating senescent traits. This study investigated how ER stress and dysregulated autophagy impact cellular senescence triggered by HIV-1 Tat in the MIO-M1 cell line (human Müller glial cells). Cells exposed to HIV-1 Tat exhibited increased vimentin expression combined with markers of ER stress (BiP, p-eIF2α), autophagy (LC3, Beclin-1, p62), and the senescence marker p21 compared to control cells. Western blotting and staining techniques like SA-ß-gal were employed to examine these markers. Additionally, treatments with ER stress inhibitor 4-PBA before HIV-1 Tat exposure led to a decreased expression of ER stress, senescence, and autophagy markers. Conversely, pre-treatment with the autophagy inhibitor 3-MA resulted in reduced autophagy and senescence markers but did not alter ER stress markers compared to control cells. The findings suggest a link between ER stress, dysregulated autophagy, and the initiation of a senescence phenotype in MIO-M1 cells induced by HIV-1 Tat exposure.


Autophagy , Cellular Senescence , Endoplasmic Reticulum Stress , HIV-1 , tat Gene Products, Human Immunodeficiency Virus , Humans , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , HIV-1/physiology , Cell Line , Ependymoglial Cells/metabolism , Ependymoglial Cells/virology , HIV Infections/virology
12.
Viruses ; 16(6)2024 Jun 09.
Article En | MEDLINE | ID: mdl-38932227

The HIV envelope glycoprotein (Env) is a trimeric protein that facilitates viral binding and fusion with target cells. As the sole viral protein on the HIV surface, Env is important both for immune responses to HIV and in vaccine designs. Targeting Env in clinical applications is challenging due to its heavy glycosylation, high genetic variability, conformational camouflage, and its low abundance on virions. Thus, there is a critical need to better understand this protein. Flow virometry (FV) is a useful methodology for phenotyping the virion surface in a high-throughput, single virion manner. To demonstrate the utility of FV to characterize Env, we stained HIV virions with a panel of 85 monoclonal antibodies targeting different regions of Env. A broad range of antibodies yielded robust staining of Env, with V3 antibodies showing the highest quantitative staining. A subset of antibodies tested in parallel on viruses produced in CD4+ T cell lines, HEK293T cells, and primary cells showed that the cellular model of virus production can impact Env detection. Finally, in addition to being able to highlight Env heterogeneity on virions, we show FV can sensitively detect differences in Env conformation when soluble CD4 is added to virions before staining.


HIV-1 , Virion , env Gene Products, Human Immunodeficiency Virus , Humans , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , HIV-1/genetics , HIV-1/physiology , HIV-1/immunology , Virion/metabolism , HEK293 Cells , HIV Antibodies/immunology , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/virology
13.
Viruses ; 16(6)2024 Jun 11.
Article En | MEDLINE | ID: mdl-38932230

Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.


HIV Infections , HIV-1 , Immunity, Innate , Virus Replication , HIV-1/genetics , HIV-1/physiology , Humans , HIV Infections/virology , HIV Infections/genetics , HIV Infections/immunology , Gene Expression Regulation, Viral , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Interferon Type I/metabolism , Interferon Type I/genetics , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Interferons/metabolism , Interferons/genetics , Interferons/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
14.
Viruses ; 16(6)2024 Jun 18.
Article En | MEDLINE | ID: mdl-38932267

Viral integration within the host genome plays a pivotal role in carcinogenesis. Various disruptive mechanisms are involved, leading to genomic instability, mutations, and DNA damage. With next-generation sequencing (NGS), we can now precisely identify viral and host genomic breakpoints and chimeric sequences, which are useful for integration site analysis. In this study, we evaluated a commercial hybrid capture NGS panel specifically designed for detecting three key viruses: HPV, HBV, and HIV-1. We also tested workflows for Viral Hybrid Capture (VHC) and Viral Integration Site (VIS) analysis, leveraging customized viral databases in CLC Microbial Genomics. By analyzing sequenced data from virally infected cancer cell lines (including SiHa, HeLa, CaSki, C-33A, DoTc2, 2A3, SCC154 for HPV; 3B2, SNU-182 for HBV; and ACH-2 for HIV-1), we precisely pinpointed viral integration sites. The workflow also highlighted disrupted and neighboring human genes that may play a crucial role in tumor development. Our results included informative virus-host read mappings, genomic breakpoints, and integration circular plots. These visual representations enhance our understanding of the integration process. In conclusion, our seamless end-to-end workflow bridges the gap in understanding viral contributions to cancer development, paving the way for improved diagnostics and treatment strategies.


Carcinogenesis , Genomics , HIV-1 , Hepatitis B virus , High-Throughput Nucleotide Sequencing , Virus Integration , Workflow , Humans , Virus Integration/genetics , Hepatitis B virus/genetics , Hepatitis B virus/physiology , HIV-1/genetics , HIV-1/physiology , High-Throughput Nucleotide Sequencing/methods , Carcinogenesis/genetics , Genomics/methods , Cell Line, Tumor , Papillomaviridae/genetics
15.
Retrovirology ; 21(1): 10, 2024 May 23.
Article En | MEDLINE | ID: mdl-38778414

BACKGROUND: Detection of viruses by host pattern recognition receptors induces the expression of type I interferon (IFN) and IFN-stimulated genes (ISGs), which suppress viral replication. Numerous studies have described HIV-1 as a poor activator of innate immunity in vitro. The exact role that the viral capsid plays in this immune evasion is not fully understood. RESULTS: To better understand the role of the HIV-1 capsid in sensing we tested the effect of making HIV-1 by co-expressing a truncated Gag that encodes the first 107 amino acids of capsid fused with luciferase or GFP, alongside wild type Gag-pol. We found that unlike wild type HIV-1, viral particles produced with a mixture of wild type and truncated Gag fused to luciferase or GFP induced a potent IFN response in THP-1 cells and macrophages. Innate immune activation by Gag-fusion HIV-1 was dependent on reverse transcription and DNA sensor cGAS, suggesting activation of an IFN response by viral DNA. Further investigation revealed incorporation of the Gag-luciferase/GFP fusion proteins into viral particles that correlated with subtle defects in wild type Gag cleavage and a diminished capacity to saturate restriction factor TRIM5α, likely due to aberrant particle formation. We propose that expression of the Gag fusion protein disturbs the correct cleavage and maturation of wild type Gag, yielding viral particles that are unable to effectively shield viral DNA from detection by innate sensors including cGAS. CONCLUSIONS: These data highlight the crucial role of capsid in innate evasion and support growing literature that disruption of Gag cleavage and capsid formation induces a viral DNA- and cGAS-dependent innate immune response. Together these data demonstrate a protective role for capsid and suggest that antiviral activity of capsid-targeting antivirals may benefit from enhanced innate and adaptive immunity in vivo.


HIV-1 , Immunity, Innate , Nucleotidyltransferases , gag Gene Products, Human Immunodeficiency Virus , HIV-1/immunology , HIV-1/genetics , HIV-1/physiology , Humans , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Antiviral Restriction Factors , Macrophages/immunology , Macrophages/virology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , THP-1 Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/immunology , Immune Evasion , Capsid/metabolism , Capsid/immunology , Virus Replication , Virion/metabolism , Virion/genetics , Virion/immunology , Host-Pathogen Interactions/immunology , DNA, Viral/genetics , Cell Line
16.
PLoS Pathog ; 20(5): e1011821, 2024 May.
Article En | MEDLINE | ID: mdl-38781120

The human immunodeficiency virus (HIV) integrates into the host genome forming latent cellular reservoirs that are an obstacle for cure or remission strategies. Viral transcription is the first step in the control of latency and depends upon the hijacking of the host cell RNA polymerase II (Pol II) machinery by the 5' HIV LTR. Consequently, "block and lock" or "shock and kill" strategies for an HIV cure depend upon a full understanding of HIV transcriptional control. The HIV trans-activating protein, Tat, controls HIV latency as part of a positive feed-forward loop that strongly activates HIV transcription. The recognition of the TATA box and adjacent sequences of HIV essential for Tat trans-activation (TASHET) of the core promoter by host cell pre-initiation complexes of HIV (PICH) has been shown to be necessary for Tat trans-activation, yet the protein composition of PICH has remained obscure. Here, DNA-affinity chromatography was employed to identify the mitotic deacetylase complex (MiDAC) as selectively recognizing TASHET. Using biophysical techniques, we show that the MiDAC subunit DNTTIP1 binds directly to TASHET, in part via its CTGC DNA motifs. Using co-immunoprecipitation assays, we show that DNTTIP1 interacts with MiDAC subunits MIDEAS and HDAC1/2. The Tat-interacting protein, NAT10, is also present in HIV-bound MiDAC. Gene silencing revealed a functional role for DNTTIP1, MIDEAS, and NAT10 in HIV expression in cellulo. Furthermore, point mutations in TASHET that prevent DNTTIP1 binding block the reactivation of HIV by latency reversing agents (LRA) that act via the P-TEFb/7SK axis. Our data reveal a key role for MiDAC subunits DNTTIP1, MIDEAS, as well as NAT10, in Tat-activated HIV transcription and latency. DNTTIP1, MIDEAS and NAT10 emerge as cell cycle-regulated host cell transcription factors that can control activated HIV gene expression, and as new drug targets for HIV cure strategies.


Gene Expression Regulation, Viral , HIV Infections , HIV-1 , Promoter Regions, Genetic , Virus Latency , Humans , HIV-1/genetics , HIV-1/physiology , HIV Infections/virology , HIV Infections/metabolism , HIV Infections/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , Viral Transcription
17.
Viruses ; 16(5)2024 04 24.
Article En | MEDLINE | ID: mdl-38793548

Human Immunodeficiency Virus type 1 (HIV-1) latency represents a significant hurdle in finding a cure for HIV-1 infections, despite tireless research efforts. This challenge is partly attributed to the intricate nature of HIV-1 latency, wherein various host and viral factors participate in multiple physiological processes. While substantial progress has been made in discovering therapeutic targets for HIV-1 transcription, targets for the post-transcriptional regulation of HIV-1 infections have received less attention. However, cumulative evidence now suggests the pivotal contribution of post-transcriptional regulation to the viral latency in both in vitro models and infected individuals. In this review, we explore recent insights on post-transcriptional latency in HIV-1 and discuss the potential of its therapeutic targets, illustrating some host factors that restrict HIV-1 at the post-transcriptional level.


HIV Infections , HIV-1 , Virus Latency , Virus Latency/genetics , HIV-1/genetics , HIV-1/physiology , HIV-1/drug effects , Humans , HIV Infections/virology , HIV Infections/drug therapy , Gene Expression Regulation, Viral , RNA Processing, Post-Transcriptional , Host-Pathogen Interactions/genetics
18.
Viruses ; 16(5)2024 04 25.
Article En | MEDLINE | ID: mdl-38793552

The HIV-1 capsid (CA) protein forms the outer shell of the viral core that is released into the cytoplasm upon infection. CA binds various cellular proteins, including CPSF6, that direct HIV-1 integration into speckle-associated domains in host chromatin. Upon HIV-1 infection, CPSF6 forms puncta in the nucleus. Here, we characterised these CPSF6 puncta further in HeLa cells, T-cells and macrophages and confirmed that integration and reverse transcription are not required for puncta formation. Indeed, we found that puncta formed very rapidly after infection, correlating with the time that CA entered the nucleus. In aphidicolin-treated HeLa cells and macrophages, puncta were detected for the length of the experiment, suggesting that puncta are only lost upon cell division. CA still co-localised with CPSF6 puncta at the latest time points, considerably after the peak of reverse transcription and integration. Intriguingly, the number of puncta induced in macrophages did not correlate with the MOI or the total number of nuclear speckles present in each cell, suggesting that CA/CPSF6 is only directed to a few nuclear speckles. Furthermore, we found that CPSF6 already co-localised with nuclear speckles in uninfected T-cells, suggesting that HIV-1 promotes a natural behaviour of CPSF6.


HIV-1 , Macrophages , T-Lymphocytes , mRNA Cleavage and Polyadenylation Factors , HIV-1/physiology , Humans , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , T-Lymphocytes/virology , T-Lymphocytes/metabolism , HeLa Cells , Macrophages/virology , Macrophages/metabolism , Virus Integration , Cell Nucleus/metabolism , Capsid Proteins/metabolism , Capsid Proteins/genetics , HIV Infections/virology , HIV Infections/metabolism , Capsid/metabolism
19.
Viruses ; 16(5)2024 04 27.
Article En | MEDLINE | ID: mdl-38793575

BACKGROUND: EcoHIV is a chimeric HIV that replicates in mice in CD4+ T cells, macrophages, and microglia (but not in neurons), causing lasting neurocognitive impairment resembling neurocognitive disease in people living with HIV. The present study was designed to develop EcoHIV-susceptible primary mouse brain cultures to investigate the indirect effects of HIV infection on neuronal integrity. RESULTS: We used two EcoHIV clones encoding EGFP and mouse bone marrow-derived macrophages (BMM), mixed mouse brain cells, or enriched mouse glial cells from two wild-type mouse strains to test EcoHIV replication efficiency, the identity of productively infected cells, and neuronal apoptosis and integrity. EcoHIV replicated efficiently in BMM. In mixed brain cell cultures, EcoHIV targeted microglia but did not cause neuronal apoptosis. Instead, the productive infection of the microglia activated them and impaired synaptophysin expression, dendritic density, and axonal structure in the neurons. EcoHIV replication in the microglia and neuronal structural changes during infection were prevented by culture with an antiretroviral. CONCLUSIONS: In murine brain cell cultures, EcoHIV replication in the microglia is largely responsible for the aspects of neuronal dysfunction relevant to cognitive disease in infected mice and people living with HIV. These cultures provide a tool for further study of HIV neuropathogenesis and its control.


Brain , Microglia , Neurons , Virus Replication , Animals , Mice , Brain/virology , Brain/pathology , Neurons/virology , Neurons/pathology , Microglia/virology , Cells, Cultured , HIV Infections/virology , Macrophages/virology , Disease Models, Animal , Apoptosis , Humans , HIV-1/physiology , Primary Cell Culture , Mice, Inbred C57BL
20.
Viruses ; 16(5)2024 05 04.
Article En | MEDLINE | ID: mdl-38793610

APOBEC3G (A3G) restricts HIV-1 replication primarily by reducing viral cDNA and inducing G-to-A hypermutations in viral cDNA. HIV-1 encodes virion infectivity factor (Vif) to counteract A3G primarily by excluding A3G viral encapsidation. Even though the Vif-induced exclusion is robust, studies suggest that A3G is still detectable in the virion. The impact of encapsidated A3G in the HIV-1 replication is unclear. Using a highly sensitive next-generation sequencing (NGS)-based G-to-A hypermutation detecting assay, we found that wild-type HIV-1 produced from A3G-expressing T-cells induced higher G-to-A hypermutation frequency in viral cDNA than HIV-1 from non-A3G-expressing T-cells. Interestingly, although the virus produced from A3G-expressing T-cells induced higher hypermutation frequency, there was no significant difference in viral infectivity, revealing a disassociation of cDNA G-to-A hypermutation to viral infectivity. We also measured G-to-A hypermutation in the viral RNA genome. Surprisingly, our data showed that hypermutation frequency in the viral RNA genome was significantly lower than in the integrated DNA, suggesting a mechanism exists to preferentially select intact genomic RNA for viral packing. This study revealed a new insight into the mechanism of HIV-1 counteracting A3G antiviral function and might lay a foundation for new antiviral strategies.


APOBEC-3G Deaminase , DNA, Complementary , HIV-1 , Mutation , Virus Replication , vif Gene Products, Human Immunodeficiency Virus , HIV-1/genetics , HIV-1/physiology , HIV-1/pathogenicity , Humans , APOBEC-3G Deaminase/genetics , APOBEC-3G Deaminase/metabolism , Virus Replication/genetics , DNA, Complementary/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism , DNA, Viral/genetics , HIV Infections/virology , T-Lymphocytes/virology , High-Throughput Nucleotide Sequencing , HEK293 Cells
...