Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.871
Filter
Add more filters

Publication year range
1.
Cell ; 184(4): 1032-1046.e18, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33571428

ABSTRACT

Human immunodeficiency virus (HIV-1) remains a major health threat. Viral capsid uncoating and nuclear import of the viral genome are critical for productive infection. The size of the HIV-1 capsid is generally believed to exceed the diameter of the nuclear pore complex (NPC), indicating that capsid uncoating has to occur prior to nuclear import. Here, we combined correlative light and electron microscopy with subtomogram averaging to capture the structural status of reverse transcription-competent HIV-1 complexes in infected T cells. We demonstrated that the diameter of the NPC in cellulo is sufficient for the import of apparently intact, cone-shaped capsids. Subsequent to nuclear import, we detected disrupted and empty capsid fragments, indicating that uncoating of the replication complex occurs by breaking the capsid open, and not by disassembly into individual subunits. Our data directly visualize a key step in HIV-1 replication and enhance our mechanistic understanding of the viral life cycle.


Subject(s)
Capsid/metabolism , HIV-1/metabolism , Nuclear Pore/metabolism , Active Transport, Cell Nucleus , Capsid/ultrastructure , Cryoelectron Microscopy , HEK293 Cells , HIV Infections/virology , HIV-1/ultrastructure , Humans , Models, Biological , Nuclear Pore/ultrastructure , Nuclear Pore/virology , Reverse Transcription , Virion/metabolism , Virus Internalization , mRNA Cleavage and Polyadenylation Factors/metabolism
2.
Mol Cell ; 82(24): 4647-4663.e8, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36525955

ABSTRACT

To explore genome organization and function in the HIV-infected brain, we applied single-nuclei transcriptomics, cell-type-specific chromosomal conformation mapping, and viral integration site sequencing (IS-seq) to frontal cortex from individuals with encephalitis (HIVE) and without (HIV+). Derepressive changes in 3D genomic compartment structures in HIVE microglia were linked to the transcriptional activation of interferon (IFN) signaling and cell migratory pathways, while transcriptional downregulation and repressive compartmentalization of neuronal health and signaling genes occurred in both HIVE and HIV+ microglia. IS-seq recovered 1,221 brain integration sites showing distinct genomic patterns compared with peripheral lymphocytes, with enrichment for sequences newly mobilized into a permissive chromatin environment after infection. Viral transcription occurred in a subset of highly activated microglia comprising 0.33% of all nuclei in HIVE brain. Our findings point to disrupted microglia-neuronal interactions in HIV and link retroviral integration to remodeling of the microglial 3D genome during infection.


Subject(s)
HIV Infections , Microglia , Humans , Microglia/metabolism , Brain , Macrophage Activation , Macrophages , HIV Infections/genetics
3.
CA Cancer J Clin ; 71(6): 505-526, 2021 11.
Article in English | MEDLINE | ID: mdl-34499351

ABSTRACT

Despite being highly preventable, cervical cancer is the fourth most common cancer and cause of cancer death in women globally. In low-income countries, cervical cancer is often the leading cause of cancer-related morbidity and mortality. Women living with human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome are at a particularly high risk of cervical cancer because of an impaired immune response to human papillomavirus, the obligate cause of virtually all cervical cancers. Globally, approximately 1 in 20 cervical cancers is attributable to HIV; in sub-Saharan Africa, approximately 1 in 5 cervical cancers is due to HIV. Here, the authors provide a critical appraisal of the evidence to date on the impact of HIV disease on cervical cancer risk, describe key methodologic issues, and frame the key outstanding research questions, especially as they apply to ongoing global efforts for prevention and control of cervical cancer. Expanded efforts to integrate HIV care with cervical cancer prevention and control, and vice versa, could assist the global effort to eliminate cervical cancer as a public health problem.


Subject(s)
HIV Infections/epidemiology , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/prevention & control , Early Detection of Cancer , Female , Humans , Immunocompromised Host , Papillomavirus Infections , Papillomavirus Vaccines , Precancerous Conditions/therapy , Primary Prevention , Secondary Prevention
4.
Proc Natl Acad Sci U S A ; 120(22): e2302509120, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37216549

ABSTRACT

Upon its mucosal transmission, HIV type 1 (HIV-1) rapidly targets genital antigen-presenting Langerhans cells (LCs), which subsequently transfer infectious virus to CD4+ T cells. We previously described an inhibitory neuroimmune cross talk, whereby calcitonin gene-related peptide (CGRP), a neuropeptide secreted by peripheral pain-sensing nociceptor neurons innervating all mucosal epithelia and associating with LCs, strongly inhibits HIV-1 transfer. As nociceptors secret CGRP following the activation of their Ca2+ ion channel transient receptor potential vanilloid 1 (TRPV1), and as we reported that LCs secret low levels of CGRP, we investigated whether LCs express functional TRPV1. We found that human LCs expressed mRNA and protein of TRPV1, which was functional and induced Ca2+ influx following activation with TRPV1 agonists, including capsaicin (CP). The treatment of LCs with TRPV1 agonists also increased CGRP secretion, reaching its anti-HIV-1 inhibitory concentrations. Accordingly, CP pretreatment significantly inhibited LCs-mediated HIV-1 transfer to CD4+ T cells, which was abrogated by both TRPV1 and CGRP receptor antagonists. Like CGRP, CP-induced inhibition of HIV-1 transfer was mediated via increased CCL3 secretion and HIV-1 degradation. CP also inhibited direct CD4+ T cells HIV-1 infection, but in CGRP-independent manners. Finally, pretreatment of inner foreskin tissue explants with CP markedly increased CGRP and CCL3 secretion, and upon subsequent polarized exposure to HIV-1, inhibited an increase in LC-T cell conjugate formation and consequently T cell infection. Our results reveal that TRPV1 activation in human LCs and CD4+ T cells inhibits mucosal HIV-1 infection, via CGRP-dependent/independent mechanisms. Formulations containing TRPV1 agonists, already approved for pain relief, could hence be useful against HIV-1.


Subject(s)
Calcitonin Gene-Related Peptide , HIV Infections , Humans , Calcitonin Gene-Related Peptide/pharmacology , T-Lymphocytes/metabolism , Langerhans Cells/metabolism , Mucous Membrane/metabolism , Capsaicin/pharmacology , Pain/metabolism , HIV Infections/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
5.
Clin Microbiol Rev ; 37(1): e0009822, 2024 03 14.
Article in English | MEDLINE | ID: mdl-38299802

ABSTRACT

People with HIV (PWH) are at elevated risk for cardiovascular diseases (CVDs), including myocardial infarction, heart failure, and sudden cardiac death, among other CVD manifestations. Chronic immune dysregulation resulting in persistent inflammation is common among PWH, particularly those with sustained viremia and impaired CD4+ T cell recovery. This inflammatory milieu is a major contributor to CVDs among PWH, in concert with common comorbidities (such as dyslipidemia and smoking) and, to a lesser extent, off-target effects of antiretroviral therapy. In this review, we discuss the clinical and mechanistic evidence surrounding heightened CVD risks among PWH, implications for specific CVD manifestations, and practical guidance for management in the setting of evolving data.


Subject(s)
Cardiovascular Diseases , HIV Infections , Humans , Cardiovascular Diseases/epidemiology , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/epidemiology , Comorbidity
6.
Clin Microbiol Rev ; 37(2): e0009923, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38546225

ABSTRACT

SUMMARYAs Chagas disease remains prevalent in the Americas, it is important that healthcare professionals and researchers are aware of the screening, diagnosis, monitoring, and treatment recommendations for the populations of patients they care for and study. Management of Trypanosoma cruzi infection in immunocompromised hosts is challenging, particularly because, regardless of antitrypanosomal treatment status, immunocompromised patients with Chagas disease are at risk for T. cruzi reactivation, which can be lethal. Evidence-based practices to prevent and manage T. cruzi reactivation vary depending on the type of immunocompromise. Here, we review available data describing Chagas disease epidemiology, testing, and management practices for various populations of immunocompromised individuals, including people with HIV and patients undergoing solid organ and hematopoietic stem cell transplantation.


Subject(s)
Chagas Disease , Immunocompromised Host , Humans , Chagas Disease/diagnosis , Chagas Disease/epidemiology , Chagas Disease/immunology , Chagas Disease/therapy , Trypanosoma cruzi/immunology
7.
Traffic ; 24(3): 114-130, 2023 03.
Article in English | MEDLINE | ID: mdl-35146839

ABSTRACT

The cytoskeleton is an essential component of the cell and it is involved in multiple physiological functions, including intracellular organization and transport. It is composed of three main families of proteinaceous filaments; microtubules, actin filaments and intermediate filaments and their accessory proteins. Motor proteins, which comprise the dynein, kinesin and myosin superfamilies, are a remarkable group of accessory proteins that mainly mediate the intracellular transport of cargoes along with the cytoskeleton. Like other cellular structures and pathways, viruses can exploit the cytoskeleton to promote different steps of their life cycle through associations with motor proteins. The complexity of the cytoskeleton and the differences among viruses, however, has led to a wide diversity of interactions, which in most cases remain poorly understood. Unveiling the details of these interactions is necessary not only for a better comprehension of specific infections, but may also reveal new potential drug targets to fight dreadful diseases such as rabies disease and acquired immunodeficiency syndrome (AIDS). In this review, we describe a few examples of the mechanisms that some human viruses, that is, rabies virus, adenovirus, herpes simplex virus, human immunodeficiency virus, influenza A virus and papillomavirus, have developed to hijack dyneins, kinesins and myosins.


Subject(s)
Cytoskeletal Proteins , Viruses , Humans , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Viruses/metabolism , Microtubules/metabolism , Actin Cytoskeleton/metabolism , Myosins/metabolism , Kinesins/metabolism , Dyneins/metabolism
8.
J Biol Chem ; 300(1): 105526, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043797

ABSTRACT

Despite antiretroviral therapy (ART), chronic forms of HIV-associated neurocognitive disorders (HAND) affect an estimated 50% of individuals living with HIV, greatly impacting their quality of life. The prevailing theory of HAND progression posits that chronic inflammation arising from the activation of latent viral reservoirs leads to progressive damage in the central nervous system (CNS). Recent evidence indicates that blood-brain barrier (BBB) pericytes are capable of active HIV-1 infection; however, their latent infection has not been defined. Given their location and function, BBB pericytes are poised to be a key viral reservoir in the development of HAND. We present the first transcriptional analysis of uninfected, active, and latent human BBB pericytes, revealing distinct transcriptional phenotypes. In addition, we demonstrate that latent infection of BBB pericytes relies on AKT signaling for reservoir survival. These findings provide insight into the state of reservoir maintenance in the CNS during HIV-1 infection and provide novel targets for reservoir clearance.


Subject(s)
Blood-Brain Barrier , Disease Reservoirs , HIV Infections , HIV-1 , Latent Infection , Pericytes , Humans , Blood-Brain Barrier/virology , HIV Infections/drug therapy , HIV Infections/transmission , HIV Infections/virology , Latent Infection/virology , Pericytes/virology , Proto-Oncogene Proteins c-akt/genetics , Quality of Life , Virus Latency , Disease Reservoirs/virology
9.
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
10.
J Virol ; 98(4): e0030824, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38497663

ABSTRACT

Host antiviral proteins inhibit primate lentiviruses and other retroviruses by targeting many features of the viral life cycle. The lentiviral capsid protein and the assembled viral core are known to be inhibited through multiple, directly acting antiviral proteins. Several phenotypes, including those known as Lv1 through Lv5, have been described as cell type-specific blocks to infection against some but not all primate lentiviruses. Here we review important features of known capsid-targeting blocks to infection together with several blocks to infection for which the genes responsible for the inhibition still remain to be identified. We outline the features of these blocks as well as how current methodologies are now well suited to find these antiviral genes and solve these long-standing mysteries in the HIV and retrovirology fields.


Subject(s)
Capsid , Host-Pathogen Interactions , Lentivirus Infections , Lentivirus , Animals , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Lentivirus/metabolism , Lentivirus Infections/metabolism
11.
J Virol ; 98(3): e0156323, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38323811

ABSTRACT

Macrophages are important target cells for diverse viruses and thus represent a valuable system for studying virus biology. Isolation of primary human macrophages is done by culture of dissociated tissues or from differentiated blood monocytes, but these methods are both time consuming and result in low numbers of recovered macrophages. Here, we explore whether macrophages derived from human induced pluripotent stem cells (iPSCs)-which proliferate indefinitely and potentially provide unlimited starting material-could serve as a faithful model system for studying virus biology. Human iPSC-derived monocytes were differentiated into macrophages and then infected with HIV-1, dengue virus, or influenza virus as model human viruses. We show that iPSC-derived macrophages support the replication of these viruses with kinetics and phenotypes similar to human blood monocyte-derived macrophages. These iPSC-derived macrophages were virtually indistinguishable from human blood monocyte-derived macrophages based on surface marker expression (flow cytometry), transcriptomics (RNA sequencing), and chromatin accessibility profiling. iPSC lines were additionally generated from non-human primate (chimpanzee) fibroblasts. When challenged with dengue virus, human and chimpanzee iPSC-derived macrophages show differential susceptibility to infection, thus providing a valuable resource for studying the species-tropism of viruses. We also show that blood- and iPSC-derived macrophages both restrict influenza virus at a late stage of the virus lifecycle. Collectively, our results substantiate iPSC-derived macrophages as an alternative to blood monocyte-derived macrophages for the study of virus biology. IMPORTANCE: Macrophages have complex relationships with viruses: while macrophages aid in the removal of pathogenic viruses from the body, macrophages are also manipulated by some viruses to serve as vessels for viral replication, dissemination, and long-term persistence. Here, we show that iPSC-derived macrophages are an excellent model that can be exploited in virology.


Subject(s)
Dengue Virus , HIV-1 , Induced Pluripotent Stem Cells , Macrophages , Models, Biological , Orthomyxoviridae , Virology , Animals , Humans , Cell Differentiation/genetics , HIV-1/growth & development , HIV-1/physiology , Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Macrophages/metabolism , Macrophages/virology , Orthomyxoviridae/growth & development , Orthomyxoviridae/physiology , Pan troglodytes , Dengue Virus/growth & development , Dengue Virus/physiology , Fibroblasts/cytology , Monocytes/cytology , Virus Replication , Flow Cytometry , Gene Expression Profiling , Chromatin Assembly and Disassembly , Viral Tropism , Virology/methods , Biomarkers/analysis , Biomarkers/metabolism
12.
Mol Ther ; 32(7): 2316-2327, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38734901

ABSTRACT

HIV-1 infection remains a public health problem with no cure. Although antiretroviral therapy (ART) is effective for suppressing HIV-1 replication, it requires lifelong drug administration due to a stable reservoir of latent proviruses and may cause serious side effects and drive the emergence of drug-resistant HIV-1 variants. Gene therapy represents an alternative approach to overcome the limitations of conventional treatments against HIV-1 infection. In this study, we constructed and investigated the antiviral effects of an HIV-1 Tat-dependent conditionally replicating adenovirus, which selectively replicates and expresses the diphtheria toxin A chain (Tat-CRAds-DTA) in HIV-1-infected cells both in vitro and in vivo. We found that Tat-CRAds-DTA could specifically induce cell death and inhibit virus replication in HIV-1-infected cells mediated by adenovirus proliferation and DTA expression. A low titer of progeny Tat-CRAds-DTA was also detected in HIV-1-infected cells. In addition, Tat-CRAds-DTA showed no apparent cytotoxicity to HIV-1-negative cells and demonstrated significant therapeutic efficacy against HIV-1 infection in a humanized mouse model. The findings in this study highlight the potential of Tat-CRAds-DTA as a new gene therapy for the treatment of HIV-1 infection.


Subject(s)
Adenoviridae , Diphtheria Toxin , Genetic Therapy , Genetic Vectors , HIV Infections , HIV-1 , Virus Replication , tat Gene Products, Human Immunodeficiency Virus , Humans , HIV-1/genetics , Diphtheria Toxin/genetics , Animals , Adenoviridae/genetics , HIV Infections/therapy , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , Mice , Genetic Therapy/methods , Genetic Vectors/genetics , Disease Models, Animal , Cell Line , HEK293 Cells , Gene Expression , Peptide Fragments
13.
Semin Immunol ; 51: 101470, 2021 01.
Article in English | MEDLINE | ID: mdl-34272086

ABSTRACT

HIV is a virus that remains a major health concern and results in an infection that has no cure even after over 30 years since its discovery. As such, HIV vaccine discovery continues to be an area of intensive research. In this review, we summarize the most recent HIV vaccine efficacy trials, clinical trials initiated within the last 3 years, and discuss prominent improvements that have been made in prophylactic HIV vaccine designs.


Subject(s)
HIV Infections , HIV-1 , Antibodies, Neutralizing , HIV Antibodies/therapeutic use , HIV Infections/prevention & control , Humans , Vaccinology
14.
Nano Lett ; 24(15): 4423-4432, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38568019

ABSTRACT

The HIV-1 envelope is a heavily glycosylated class 1 trimeric fusion protein responsible for viral entry into CD4+ immune cells. Developing neutralizing antibodies against the specific envelope glycans is an alternative method for antiviral therapies. This work presents the first-ever development and characterization of artificial neutralizing antibodies using molecular imprinting technology to recognize and bind to the envelope protein of HIV-1. The prepared envelope glycan-imprinted nanoparticles (GINPs) can successfully prevent HIV-1 from infecting target cells by shielding the glycans on the envelope protein. In vitro experiments showed that GINPs have strong affinity toward HIV-1 (Kd = 36.7 ± 2.2 nM) and possess high anti-interference and specificity. GINPs demonstrate broad inhibition activity against both tier 1 and tier 2 HIV-1 strains with a pM-level IC50 and exhibit a significant inhibitory effect on long-term viral replication by more than 95%. The strategy provides a promising method for the inhibition and therapy of HIV-1 infection.


Subject(s)
HIV Infections , HIV-1 , Humans , Antibodies, Neutralizing , HIV Antibodies/metabolism , Glycosylation , HIV Infections/drug therapy , Polysaccharides/metabolism
15.
Clin Microbiol Rev ; 36(4): e0001323, 2023 12 20.
Article in English | MEDLINE | ID: mdl-37966222

ABSTRACT

A major impediment for HIV cure is the ability of the virus to integrate its genome in the form of replication-competent proviral DNA into the cellular genome of the host and remain transcriptionally silent and hidden from the host's immune defense mechanisms in latent reservoir cells. These latent reservoirs are highly heterogeneous, long-lived cells that are capable of reactivating to restore the viremic stage in virally suppressed individuals upon treatment interruption, thus necessitating life-long antiretroviral treatment. Latency reversal has become one of the most explored therapeutic approaches for eliminating HIV reservoirs and effecting HIV cure. Various aspects governing the establishment, maintenance, and reversal of HIV latency continue to be an enigma and warrant further research. Quantifying the size of the latent reservoir pool is also a challenge as these cells are very few in number and cannot be easily differentiated from uninfected cells. This article provides a comprehensive review of the in vitro and in vivo models currently available for studying HIV latency as well as the recently developed molecular tools for detection and quantification of latent viral reservoirs.


Subject(s)
HIV Infections , Virus Latency , Humans , HIV Infections/drug therapy , CD4-Positive T-Lymphocytes , Anti-Retroviral Agents/therapeutic use , Models, Theoretical
16.
J Infect Dis ; 230(1): 61-66, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052731

ABSTRACT

BACKGROUND: Abnormal cervical cytology is commonly observed in women with human immunodeficiency virus (WWH). METHODS: A cross-sectional study was conducted with 130 WWH and 147 age-matched healthy controls, who underwent gynecological examinations at Beijing Ditan Hospital. The presence of abnormal cervical cytology in WWH was predicted after performing a logistic regression analysis. RESULTS: Multivariate logistic regression revealed 3 independent factors, among which CD4 cell count ≥350 cells/µL was the protective factor, while human papillomavirus infection and abnormal vaginal pH were the risk factors. CONCLUSIONS: Vaginal microecological disorders can increase the risk of abnormal cervical cytology in WWH.


Subject(s)
HIV Infections , Papillomavirus Infections , Vaginal Diseases , Adult , Female , Humans , Middle Aged , Young Adult , Case-Control Studies , CD4 Lymphocyte Count , Cervix Uteri/pathology , Cervix Uteri/virology , China/epidemiology , Cross-Sectional Studies , HIV Infections/complications , Logistic Models , Papillomavirus Infections/virology , Papillomavirus Infections/diagnosis , Papillomavirus Infections/complications , Risk Factors , Vagina/virology , Vagina/pathology , Vaginal Diseases/virology , Vaginal Diseases/epidemiology
17.
J Infect Dis ; 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39380136

ABSTRACT

Traditional Randomized Controlled Trials often fall short in addressing the specific needs of clinical practice due to their one-size-fits-all treatment approaches. Sequential Multiple Assignment Randomized Trials (SMARTs) offer a dynamic and adaptive approach, allowing for multiple randomizations based on patient responses and evolving conditions. SMARTs enable personalized treatment pathways, such as in the trial for antiretroviral therapy (ART) in South Africa, which adjusts treatment based on patient outcomes. Despite these advantages, the use of SMARTs in infectious diseases remains limited. Greater adoption of SMARTs could promote more personalized treatment approaches, improve flexibility in response to public health needs, and enhance the effectiveness of interventions. However, challenges such as recruitment and increased expertise needed for more complex analyses must be addressed. Additionally, combining SMARTs with other adaptive designs could further improve the relevance and outcomes of clinical research.

18.
J Infect Dis ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38332750

ABSTRACT

In Lusaka, Zambia, we introduced liver fine needle aspiration (FNA) into a research cohort of adults with treatment-naïve chronic hepatitis B virus (HBV) infection, with and without HIV coinfection, as well as with acute HBV infection. Over 117 enrollment and 47 longitudinal FNAs (at 1 year follow-up), we established participant acceptability and safety. We also demonstrated the quality of the material through single cell RNA sequencing of selected enrollment FNAs, which revealed a range of immune cells. This approach can drive new insights into HBV immunology, informing cure strategies, and can improve our understanding of HBV natural history in Africa.

19.
J Infect Dis ; 230(1): 55-60, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052708

ABSTRACT

We assessed cumulative detection and determinants of anal high-grade squamous intraepithelial lesions (HSILs) in men who have sex with men living with human immunodeficiency virus and who underwent 3 visits over 2 years, with cytology and high-resolution anoscopy, within the ANRS-EP57-APACHES study. The cumulative HSIL detection rate was 33% (134 of 410), of which 48% HSILs were detected at baseline. HSIL detection varied considerably by center (from 13% to 51%). The strongest HSIL determinants were baseline human papillomavirus 16 (adjusted odds ratio, 8.2; 95% confidence interval, 3.6-18.9) and p16/Ki67 (4.6 [2.3-9.1]). Repeated annual cytology and high-resolution anoscopy improved HSIL detection but did not fully compensate for between-center heterogeneity.


Subject(s)
Anus Neoplasms , HIV Infections , Homosexuality, Male , Papillomavirus Infections , Squamous Intraepithelial Lesions , Humans , Male , HIV Infections/complications , Squamous Intraepithelial Lesions/virology , Squamous Intraepithelial Lesions/pathology , France/epidemiology , Adult , Anus Neoplasms/virology , Anus Neoplasms/diagnosis , Anus Neoplasms/epidemiology , Anus Neoplasms/pathology , Middle Aged , Papillomavirus Infections/virology , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Follow-Up Studies , Anal Canal/virology , Anal Canal/pathology , Human papillomavirus 16/isolation & purification , Sexual and Gender Minorities
20.
Med Res Rev ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39185567

ABSTRACT

Infections caused by viruses as the smallest infectious agents, pose a major threat to global public health. Viral infections utilize different host mechanisms to facilitate their own propagation and pathogenesis. MicroRNAs (miRNAs), as small noncoding RNA molecules, play important regulatory roles in different diseases, including viral infections. They can promote or inhibit viral infection and have a pro-viral or antiviral role. Also, viral infections can modulate the expression of host miRNAs. Furthermore, viruses from different families evade the host immune response by producing their own miRNAs called viral miRNAs (v-miRNAs). Understanding the replication cycle of viruses and their relation with host miRNAs and v-miRNAs can help to find new treatments against viral infections. In this review, we aim to outline the structure, genome, and replication cycle of various viruses including hepatitis B, hepatitis C, influenza A virus, coronavirus, human immunodeficiency virus, human papillomavirus, herpes simplex virus, Epstein-Barr virus, Dengue virus, Zika virus, and Ebola virus. We also discuss the role of different host miRNAs and v-miRNAs and their role in the pathogenesis of these viral infections.

SELECTION OF CITATIONS
SEARCH DETAIL