Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
JCI Insight ; 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39288262

ABSTRACT

Elite controllers (EC), a unique group of people with HIV (PWH), exhibit remarkable control of viral replication in the absence of antiretroviral therapy. In this study, we comprehensively characterized the NK cell repertoire in EC after long-term viral control. Phenotypic profiling of NK cells revealed profound differences compared with other PWH, but marked similarities to uninfected individuals, with a distinctive prevalence of NKG2C+CD57+memory-like NK cells. Functional analyses indicated that EC had limited production of functional molecules upon NK stimulation and consequently reduced natural cytotoxicity against non-HIV target cells. Importantly, EC showed an exceptional ability to kill primary HIV-infected cells by the antibody-dependent cell cytotoxicity (ADCC) adaptive mechanism, which was achieved by a specific memory-like NK population expressing CD16, NKG2A, NKG2C, CD57 and CXCR3. In-depth single-cell RNA sequencing unveiled a unique transcriptional signature in these NK cells linked to increased cell metabolism, migration, chemotaxis, effector functions, cytokine secretion, and antiviral response. Our findings underscore a pivotal role of NK cells in the immune control of HIV and identify specific NK cells as emerging targets for immunotherapies.

2.
JCI Insight ; 9(15)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900571

ABSTRACT

Men who have sex with men (MSM) with HIV are at high risk for squamous intraepithelial lesion (SIL) and anal cancer. Identifying local immunological mechanisms involved in the development of anal dysplasia could aid treatment and diagnostics. Here, we studied 111 anal biopsies obtained from 101 MSM with HIV, who participated in an anal screening program. We first assessed multiple immune subsets by flow cytometry, in addition to histological examination, in a discovery cohort. Selected molecules were further evaluated by immunohistochemistry in a validation cohort. Pathological samples were characterized by the presence of resident memory T cells with low expression of CD103 and by changes in natural killer cell subsets, affecting residency and activation. Furthermore, potentially immunosuppressive subsets, including CD15+CD16+ mature neutrophils, gradually increased as the anal lesion progressed. Immunohistochemistry verified the association between the presence of CD15 in the epithelium and SIL diagnosis for the correlation with high-grade SIL. A complex immunological environment with imbalanced proportions of resident effectors and immune-suppressive subsets characterized pathological samples. Neutrophil infiltration, determined by CD15 staining, may represent a valuable pathological marker associated with the grade of dysplasia.


Subject(s)
Anus Neoplasms , HIV Infections , Lewis X Antigen , Humans , Male , HIV Infections/immunology , HIV Infections/complications , HIV Infections/pathology , Anus Neoplasms/pathology , Anus Neoplasms/immunology , Adult , Middle Aged , Lewis X Antigen/metabolism , Homosexuality, Male , Squamous Intraepithelial Lesions/pathology , Anal Canal/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Antigens, CD/metabolism , Neutrophils/immunology , Neutrophils/pathology , Neutrophils/metabolism , Biopsy , Immunohistochemistry , Integrin alpha Chains/metabolism
3.
Nat Commun ; 15(1): 2100, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38453949

ABSTRACT

Increased recruitment of transitional and non-classical monocytes in the lung during SARS-CoV-2 infection is associated with COVID-19 severity. However, whether specific innate sensors mediate the activation or differentiation of monocytes in response to different SARS-CoV-2 proteins remain poorly characterized. Here, we show that SARS-CoV-2 Spike 1 but not nucleoprotein induce differentiation of monocytes into transitional or non-classical subsets from both peripheral blood and COVID-19 bronchoalveolar lavage samples in a NFκB-dependent manner, but this process does not require inflammasome activation. However, NLRP3 and NLRC4 differentially regulated CD86 expression in monocytes in response to Spike 1 and Nucleoprotein, respectively. Moreover, monocytes exposed to Spike 1 induce significantly higher proportions of Th1 and Th17 CD4 + T cells. In contrast, monocytes exposed to Nucleoprotein reduce the degranulation of CD8 + T cells from severe COVID-19 patients. Our study provides insights in the differential impact of innate sensors in regulating monocytes in response to different SARS-CoV-2 proteins, which might be useful to better understand COVID-19 immunopathology and identify therapeutic targets.


Subject(s)
COVID-19 , Inflammasomes , Humans , Calcium-Binding Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , COVID-19/pathology , Inflammasomes/metabolism , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleoproteins/metabolism , SARS-CoV-2/metabolism
4.
Curr Opin HIV AIDS ; 19(2): 69-78, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38169333

ABSTRACT

PURPOSE OF REVIEW: The complex nature and distribution of the HIV reservoir in tissue of people with HIV remains one of the major obstacles to achieve the elimination of HIV persistence. Challenges include the tissue-specific states of latency and viral persistence, which translates into high levels of reservoir heterogeneity. Moreover, the best strategies to reach and eliminate these reservoirs may differ based on the intrinsic characteristics of the cellular and anatomical reservoir to reach. RECENT FINDINGS: While major focus has been undertaken for lymphoid tissues and follicular T helper cells, evidence of viral persistence in HIV and non-HIV antigen-specific CD4 + T cells and macrophages resident in multiple tissues providing long-term protection presents new challenges in the quest for an HIV cure. Considering the microenvironments where these cellular reservoirs persist opens new venues for the delivery of drugs and immunotherapies to target these niches. New tools, such as single-cell RNA sequencing, CRISPR screenings, mRNA technology or tissue organoids are quickly developing and providing detailed information about the complex nature of the tissue reservoirs. SUMMARY: Targeting persistence in tissue reservoirs represents a complex but essential step towards achieving HIV cure. Combinatorial strategies, particularly during the early phases of infection to impact initial reservoirs, capable of reaching and reactivating multiple long-lived reservoirs in the body may lead the path.


Subject(s)
HIV Infections , Humans , HIV Infections/drug therapy , Virus Latency , CD4-Positive T-Lymphocytes
6.
Cell Rep Med ; 4(10): 101202, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37741278

ABSTRACT

Human immunodeficiency virus (HIV) infection induces immunological dysfunction, which limits the elimination of HIV-infected cells during treated infection. Identifying and targeting dysfunctional immune cells might help accelerate the purging of the persistent viral reservoir. Here, we show that chronic HIV infection increases natural killer (NK) cell populations expressing the negative immune regulator KLRG1, both in peripheral blood and lymph nodes. Antiretroviral treatment (ART) does not reestablish these functionally impaired NK populations, and the expression of KLRG1 correlates with active HIV transcription. Targeting KLRG1 with specific antibodies significantly restores the capacity of NK cells to kill HIV-infected cells, reactivates latent HIV present in CD4+ T cells co-expressing KLRG1, and reduces the intact HIV genomes in samples from ART-treated individuals. Our data support the potential use of immunotherapy against the KLRG1 receptor to impact the viral reservoir during HIV persistence.


Subject(s)
HIV Infections , HIV-1 , Receptors, Immunologic , Humans , HIV Infections/drug therapy , HIV Infections/genetics , Killer Cells, Natural , Lectins, C-Type/genetics , Receptors, Immunologic/genetics , Virus Latency
7.
Commun Biol ; 6(1): 487, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37165099

ABSTRACT

Latency is a major barrier towards virus elimination in HIV-1-infected individuals. Yet, the mechanisms that contribute to the maintenance of HIV-1 latency are incompletely understood. Here we describe the Schlafen 12 protein (SLFN12) as an HIV-1 restriction factor that establishes a post-transcriptional block in HIV-1-infected cells and thereby inhibits HIV-1 replication and virus reactivation from latently infected cells. The inhibitory activity is dependent on the HIV-1 codon usage and on the SLFN12 RNase active sites. Within HIV-1-infected individuals, SLFN12 expression in PBMCs correlated with HIV-1 plasma viral loads and proviral loads suggesting a link with the general activation of the immune system. Using an RNA FISH-Flow HIV-1 reactivation assay, we demonstrate that SLFN12 expression is enriched in infected cells positive for HIV-1 transcripts but negative for HIV-1 proteins. Thus, codon-usage dependent translation inhibition of HIV-1 proteins participates in HIV-1 latency and can restrict the amount of virus release after latency reversal.


Subject(s)
CD4-Positive T-Lymphocytes , HIV-1 , Codon Usage , HIV-1/physiology , RNA, Viral/genetics , Virus Latency/genetics
8.
Front Microbiol ; 14: 1185504, 2023.
Article in English | MEDLINE | ID: mdl-37206325

ABSTRACT

The emergent human coronavirus SARS-CoV-2 and its resistance to current drugs makes the need for new potent treatments for COVID-19 patients strongly necessary. Dextran sulfate (DS) polysaccharides have long demonstrated antiviral activity against different enveloped viruses in vitro. However, their poor bioavailability has led to their abandonment as antiviral candidates. Here, we report for the first time the broad-spectrum antiviral activity of a DS-based extrapolymeric substance produced by the lactic acid bacterium Leuconostoc mesenteroides B512F. Time of addition assays with SARS-CoV-2 pseudoviruses in in vitro models confirm the inhibitory activity of DSs in the early stages of viral infection (viral entry). In addition, this exopolysaccharide substance also reports broad-spectrum antiviral activity against several enveloped viruses such as SARS-CoV-2, HCoV229E, HSV-1, in in vitro models and in human lung tissue. The toxicity and antiviral capacity of DS from L. mesenteroides was tested in vivo in mouse models which are susceptible to SARS-CoV-2 infection. The described DS, administered by inhalation, a new route of administration for these types of polymers, shows strong inhibition of SARS-CoV-2 infection in vivo, significantly reducing animal mortality and morbidity at non-toxic doses. Therefore, we suggest that it may be considered as a potential candidate for antiviral therapy against SARS-CoV-2.

9.
Nat Commun ; 14(1): 1887, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37019909

ABSTRACT

Resident memory T cells (TRM) present at the respiratory tract may be essential to enhance early SARS-CoV-2 viral clearance, thus limiting viral infection and disease. While long-term antigen-specific TRM are detectable beyond 11 months in the lung of convalescent COVID-19 patients, it is unknown if mRNA vaccination encoding for the SARS-CoV-2 S-protein can induce this frontline protection. Here we show that the frequency of CD4+ T cells secreting IFNγ in response to S-peptides is variable but overall similar in the lung of mRNA-vaccinated patients compared to convalescent-infected patients. However, in vaccinated patients, lung responses present less frequently a TRM phenotype compared to convalescent infected individuals and polyfunctional CD107a+ IFNγ+ TRM are virtually absent in vaccinated patients. These data indicate that mRNA vaccination induces specific T cell responses to SARS-CoV-2 in the lung parenchyma, although to a limited extend. It remains to be determined whether these vaccine-induced responses contribute to overall COVID-19 control.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Memory T Cells , Immunologic Memory , Lung , Vaccination , Antibodies, Viral
10.
J Enzyme Inhib Med Chem ; 37(1): 2158-2168, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35943189

ABSTRACT

Zinc pyrithione (1a), together with its analogues 1b-h and ruthenium pyrithione complex 2a, were synthesised and evaluated for the stability in biologically relevant media and anti-SARS-CoV-2 activity. Zinc pyrithione revealed potent in vitro inhibition of cathepsin L (IC50=1.88 ± 0.49 µM) and PLPro (IC50=0.50 ± 0.07 µM), enzymes involved in SARS-CoV-2 entry and replication, respectively, as well as antiviral entry and replication properties in an ex vivo system derived from primary human lung tissue. Zinc complexes 1b-h expressed comparable in vitro inhibition. On the contrary, ruthenium complex 2a and the ligand pyrithione a itself expressed poor inhibition in mentioned assays, indicating the importance of the selection of metal core and structure of metal complex for antiviral activity. Safe, effective, and preferably oral at-home therapeutics for COVID-19 are needed and as such zinc pyrithione, which is also commercially available, could be considered as a potential therapeutic agent against SARS-CoV-2.


Subject(s)
COVID-19 Drug Treatment , Ruthenium , Antiviral Agents/pharmacology , Cathepsin L , Humans , Organometallic Compounds , Pyridines , SARS-CoV-2
11.
Elife ; 112022 05 26.
Article in English | MEDLINE | ID: mdl-35616530

ABSTRACT

Human immunodeficiency virus (HIV) establishes a persistent infection in heterogeneous cell reservoirs, which can be maintained by different mechanisms including cellular proliferation, and represent the main obstacle to curing the infection. The expression of the Fcγ receptor CD32 has been identified as a marker of the active cell reservoirs in people on antiretroviral therapy (ART), but if its expression has any role in conferring advantage for viral persistence is unknown. Here, we report that HIV-infected cells expressing CD32 have reduced susceptibility to natural killer (NK) antibody-dependent cell cytotoxicity (ADCC) by a mechanism compatible with the suboptimal binding of HIV-specific antibodies. Infected CD32 cells have increased proliferative capacity in the presence of immune complexes, and are more resistant to strategies directed to potentiate NK function. Remarkably, reactivation of the latent reservoir from antiretroviral-treated people living with HIV increases the pool of infected CD32 cells, which are largely resistant to the ADCC immune mechanism. Thus, we report the existence of reservoir cells that evade part of the NK immune response through the expression of CD32.


Subject(s)
HIV Infections , HIV-1 , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes , HIV Antibodies , HIV-1/physiology , Humans , Immunity
12.
PLoS Pathog ; 18(1): e1010171, 2022 01.
Article in English | MEDLINE | ID: mdl-35025963

ABSTRACT

The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Lung/virology , SARS-CoV-2/physiology , Virus Internalization , Adult , Animals , Antiviral Agents/pharmacology , COVID-19/immunology , COVID-19/pathology , Cells, Cultured , Chlorocebus aethiops , Drug Evaluation, Preclinical , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Inflammation/pathology , Inflammation/therapy , Inflammation/virology , Lung/pathology , SARS-CoV-2/drug effects , Vero Cells , Virus Internalization/drug effects
13.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34948390

ABSTRACT

Since the start of the COVID-19 outbreak, pharmaceutical companies and research groups have focused on the development of vaccines and antiviral drugs against SARS-CoV-2. Here, we apply a drug repurposing strategy to identify drug candidates that are able to block the entrance of the virus into human cells. By combining virtual screening with in vitro pseudovirus assays and antiviral assays in Human Lung Tissue (HLT) cells, we identify entrectinib as a potential antiviral drug.


Subject(s)
Benzamides/pharmacology , COVID-19 Drug Treatment , Indazoles/pharmacology , SARS-CoV-2/drug effects , Animals , Antiviral Agents/pharmacology , Benzamides/metabolism , COVID-19/metabolism , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical , Drug Repositioning/methods , Humans , Indazoles/metabolism , Lung/pathology , Lung/virology , Molecular Docking Simulation , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Vero Cells , Virus Attachment/drug effects
14.
Nano Today ; 362021 Feb.
Article in English | MEDLINE | ID: mdl-34394703

ABSTRACT

HIV represents a persistent infection which negatively alters the immune system. New tools to reinvigorate different immune cell populations to impact HIV are needed. Herein, a novel nanotool for the specific enhancement of the natural killer (NK) immune response towards HIV-infected T-cells has been developed. Bispecific Au nanoparticles (BiAb-AuNPs), dually conjugated with IgG anti-HIVgp120 and IgG anti-human CD16 antibodies, were generated by a new controlled, linker-free and cooperative conjugation method promoting the ordered distribution and segregation of antibodies in domains. The cooperatively-adsorbed antibodies fully retained the capabilities to recognize their cognate antigen and were able to significantly enhance cell-to-cell contact between HIV-expressing cells and NK cells. As a consequence, the BiAb-AuNPs triggered a potent cytotoxic response against HIV-infected cells in blood and human tonsil explants. Remarkably, the BiAb-AuNPs were able to significantly reduce latent HIV infection after viral reactivation in a primary cell model of HIV latency. This novel molecularly-targeted strategy using a bispecific nanotool to enhance the immune system represents a new approximation with potential applications beyond HIV.

15.
Nat Commun ; 12(1): 3010, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021148

ABSTRACT

Resident memory T cells (TRM) positioned within the respiratory tract are probably required to limit SARS-CoV-2 spread and COVID-19. Importantly, TRM are mostly non-recirculating, which reduces the window of opportunity to examine these cells in the blood as they move to the lung parenchyma. Here, we identify circulating virus-specific T cell responses during acute infection with functional, migratory and apoptotic patterns modulated by viral proteins and associated with clinical outcome. Disease severity is associated predominantly with IFNγ and IL-4 responses, increased responses against S peptides and apoptosis, whereas non-hospitalized patients have increased IL-12p70 levels, degranulation in response to N peptides and SARS-CoV-2-specific CCR7+ T cells secreting IL-10. In convalescent patients, lung-TRM are frequently detected even 10 months after initial infection, in which contemporaneous blood does not reflect tissue-resident profiles. Our study highlights a balanced anti-inflammatory antiviral response associated with a better outcome and persisting TRM cells as important for future protection against SARS-CoV-2 infection.


Subject(s)
COVID-19/immunology , Immunologic Memory/immunology , Lung/immunology , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COVID-19/virology , Cell Movement/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Lung/virology , SARS-CoV-2/physiology , T-Lymphocytes/metabolism
16.
Nat Commun ; 10(1): 4739, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31628331

ABSTRACT

HIV viral reservoirs are established very early during infection. Resident memory T cells (TRM) are present in tissues such as the lower female genital tract, but the contribution of this subset of cells to the pathogenesis and persistence of HIV remains unclear. Here, we show that cervical CD4+TRM display a unique repertoire of clusters of differentiation, with enrichment of several molecules associated with HIV infection susceptibility, longevity and self-renewing capacities. These protein profiles are enriched in a fraction of CD4+TRM expressing CD32. Cervical explant models show that CD4+TRM preferentially support HIV infection and harbor more viral DNA and protein than non-TRM. Importantly, cervical tissue from ART-suppressed HIV+ women contain high levels of viral DNA and RNA, being the TRM fraction the principal contributor. These results recognize the lower female genital tract as an HIV sanctuary and identify CD4+TRM as primary targets of HIV infection and viral persistence. Thus, strategies towards an HIV cure will need to consider TRM phenotypes, which are widely distributed in tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Immunologic Memory/immunology , Adult , Aged , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cervix Uteri/drug effects , Cervix Uteri/virology , Disease Reservoirs/virology , Female , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , Humans , Middle Aged , Mucous Membrane/drug effects , Mucous Membrane/virology , Viral Load/drug effects , Viral Load/genetics , Viral Load/immunology
17.
Nat Commun ; 10(1): 3705, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31420544

ABSTRACT

The identification of exclusive markers to target HIV-reservoir cells will represent a significant advance in the search for therapies to cure HIV. Here, we identify the B lymphocyte antigen CD20 as a marker for HIV-infected cells in vitro and in vivo. The CD20 molecule is dimly expressed in a subpopulation of CD4-positive (CD4+) T lymphocytes from blood, with high levels of cell activation and heterogeneous memory phenotypes. In lymph node samples from infected patients, CD20 is present in productively HIV-infected cells, and ex vivo viral infection selectively upregulates the expression of CD20 during early infection. In samples from patients on antiretroviral therapy (ART) this subpopulation is significantly enriched in HIV transcripts, and the anti-CD20 monoclonal antibody Rituximab induces cell killing, which reduces the pool of HIV-expressing cells when combined with latency reversal agents. We provide a tool for targeting this active HIV-reservoir after viral reactivation in patients while on ART.


Subject(s)
Antigens, CD20/metabolism , CD4-Positive T-Lymphocytes/drug effects , HIV Infections/metabolism , Immunologic Factors/pharmacology , Rituximab/pharmacology , Virus Activation , Virus Latency , Anti-HIV Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Flow Cytometry , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1 , Humans , Immunologic Memory , Leukocytes, Mononuclear , Lymph Nodes/cytology , Lymphocyte Activation/immunology , RNA, Viral , Rituximab/therapeutic use
18.
PLoS Pathog ; 15(8): e1007991, 2019 08.
Article in English | MEDLINE | ID: mdl-31425551

ABSTRACT

Latency reversal agents (LRAs) have proven to induce HIV-1 transcription in vivo but are ineffective at decreasing the size of the latent reservoir in antiretroviral treated patients. The capacity of the LRAs to perturb the viral reservoir present in distinct subpopulations of cells is currently unknown. Here, using a new RNA FISH/flow ex vivo viral reactivation assay, we performed a comprehensive assessment of the viral reactivation capacity of different families of LRAs, and their combinations, in different CD4+ T cell subsets. We observed that a median of 16.28% of the whole HIV-reservoir induced HIV-1 transcripts after viral reactivation, but only 10.10% of these HIV-1 RNA+ cells produced the viral protein p24. Moreover, none of the LRAs were powerful enough to reactivate HIV-1 transcription in all CD4+ T cell subpopulations. For instance, the combination of Romidepsin and Ingenol was identified as the best combination of drugs at increasing the proportion of HIV-1 RNA+ cells, in most, but not all, CD4+ T cell subsets. Importantly, memory stem cells were identified as highly resistant to HIV-1 reactivation, and only the combination of Panobinostat and Bryostatin-1 significantly increased the number of cells transcribing HIV within this subset. Overall, our results validate the use of the RNA FISH/flow technique to assess the potency of LRAs among different CD4+ T cell subsets, manifest the intrinsic differences between cells that encompass the latent HIV reservoir, and highlight the difficulty to significantly impact the latent infection with the currently available drugs. Thus, our results have important implications for the rational design of therapies aimed at reversing HIV latency from diverse cellular reservoirs.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Virus Activation/immunology , Virus Latency/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Depsipeptides/pharmacology , Diterpenes/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Humans , Viral Load , Virus Activation/drug effects , Virus Latency/drug effects
19.
Front Immunol ; 10: 825, 2019.
Article in English | MEDLINE | ID: mdl-31114569

ABSTRACT

Antigen presenting cells from the cervical mucosa are thought to amplify incoming HIV-1 and spread infection systemically without being productively infected. Yet, the molecular mechanism at the cervical mucosa underlying this viral transmission pathway remains unknown. Here we identified a subset of HLA-DR+ CD14+ CD11c+ cervical DCs at the lamina propria of the ectocervix and the endocervix that expressed the type-I interferon inducible lectin Siglec-1 (CD169), which promoted viral uptake. In the cervical biopsy of a viremic HIV-1+ patient, Siglec-1+ cells harbored HIV-1-containing compartments, demonstrating that in vivo, these cells trap viruses. Ex vivo, a type-I interferon antiviral environment enhanced viral capture and trans-infection via Siglec-1. Nonetheless, HIV-1 transfer via cervical DCs was effectively prevented with antibodies against Siglec-1. Our findings contribute to decipher how cervical DCs may boost HIV-1 replication and promote systemic viral spread from the cervical mucosa, and highlight the importance of including inhibitors against Siglec-1 in microbicidal strategies.


Subject(s)
Cervix Uteri/immunology , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/physiology , Sialic Acid Binding Ig-like Lectin 1/immunology , Virus Replication/immunology , Adult , Aged , Aged, 80 and over , Biological Transport, Active/immunology , Cervix Uteri/pathology , Cervix Uteri/virology , Dendritic Cells/pathology , Dendritic Cells/virology , Female , HEK293 Cells , HIV Infections/pathology , Humans , Interferon Type I/immunology , Middle Aged , Mucous Membrane/immunology , Mucous Membrane/pathology , Mucous Membrane/virology
20.
J Immunol Methods ; 460: 26-35, 2018 09.
Article in English | MEDLINE | ID: mdl-29894750

ABSTRACT

In the field of sexually transmitted infections (STI), the cervicovaginal explant (CVEx) model, not only provides the opportunity to study the different immunological arms present in these tissues under steady state conditions, but also their response against ex vivo infection with relevant pathogens. The methodology associated to the establishment of the HIV infection model in the cervicovaginal tissue was described in detail by Grivel et al. earlier (Grivel and Margolis, 2009). With this model as a foundation, we illustrate different approaches to obtain a large number of immunological readouts from a single piece of tissue, thus maximizing the immunological output obtained. Additionally, we discuss several ideas to study some of the immunological subsets present in this mucosal tissue by enriching them with the addition of distinct chemokines or specifically inducing their activation. Importantly, most of the methodology and concepts proposed here can be applied to study the immune subsets resident in other tissues. In the field of mucosal immunology, the possibility of studying resident immune subsets from tissue explants offers a great opportunity to understand the real players against invading pathogens and localized pathologies. Furthermore, this model allows for addressing the therapeutic benefit of modulating the activity of certain molecules and immune subsets against invading pathogens, which may infer their contribution to pathogen control and direct novel therapeutic interventions.


Subject(s)
Cervix Uteri , HIV-1/immunology , Models, Immunological , Tissue Culture Techniques , Vagina , Cervix Uteri/immunology , Cervix Uteri/physiology , Cervix Uteri/virology , Female , HIV Infections/pathology , HIV Infections/virology , Humans , Mucous Membrane/immunology , Mucous Membrane/virology , Vagina/immunology , Vagina/pathology , Vagina/virology
SELECTION OF CITATIONS
SEARCH DETAIL