Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Commun ; 15(1): 3664, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693123

ABSTRACT

The application of mammalian target of rapamycin inhibition (mTORi) as primary prophylactic therapy to optimize T cell effector function while preserving allograft tolerance remains challenging. Here, we present a comprehensive two-step therapeutic approach in a male patient with metastatic cutaneous squamous cell carcinoma and heart transplantation followed with concomitant longitudinal analysis of systemic immunologic changes. In the first step, calcineurin inhibitor/ mycophenolic acid is replaced by the mTORi everolimus to achieve an improved effector T cell status with increased cytotoxic activity (perforin, granzyme), enhanced proliferation (Ki67) and upregulated activation markers (CD38, CD69). In the second step, talimogene laherparepvec (T-VEC) injection further enhances effector function by switching CD4 and CD8 cells from central memory to effector memory profiles, enhancing Th1 responses, and boosting cytotoxic and proliferative activities. In addition, cytokine release (IL-6, IL-18, sCD25, CCL-2, CCL-4) is enhanced and the frequency of circulating regulatory T cells is increased. Notably, no histologic signs of allograft rejection are observed in consecutive end-myocardial biopsies. These findings provide valuable insights into the dynamics of T cell activation and differentiation and suggest that timely initiation of mTORi-based primary prophylaxis may provide a dual benefit of revitalizing T cell function while maintaining allograft tolerance.


Subject(s)
Carcinoma, Squamous Cell , Graft Rejection , Heart Transplantation , Herpesvirus 1, Human , MTOR Inhibitors , Heart Transplantation/adverse effects , Humans , Male , Graft Rejection/prevention & control , Graft Rejection/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/drug therapy , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Biological Products/pharmacology , Biological Products/therapeutic use , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Middle Aged , Everolimus/pharmacology , Everolimus/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
2.
Vaccines (Basel) ; 12(4)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38675788

ABSTRACT

Vaccines are indispensable tools in the battle against infectious diseases and hold great potential in combating a myriad of other diseases [...].

3.
Front Immunol ; 14: 1213375, 2023.
Article in English | MEDLINE | ID: mdl-37622123

ABSTRACT

Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, IgG , B7-H1 Antigen , Cell Membrane , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use
4.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37380369

ABSTRACT

Cytokine storm induced by anti-human epidermal growth factor receptor-2 (HER2) therapies has not been reported. We report a patient with breast cancer treated with trastuzumab/pertuzumab who developed severe biventricular dysfunction and cardiogenic shock (CS) 6 months after starting double anti-HER2 therapy. The CS was accompanied by severe systemic inflammation, and cardiac MRI (cMRI) showed structural changes typical of myocardial inflammation. The immuno-inflammatory profile showed significantly increased levels of activation of the complement system, proinflammatory cytokines (IL-1ß, IL-6, IL-18, IL-17A, TNF-alpha) with increased activity of classical monocytic, T helper 17 cells (Th17), CD4 T and effector memory CD8 T subsets, whereas NK cell activation was not observed. The data suggest an important role for monocytes as initiators of this FcγR-dependent antibody-dependent cytotoxicity, leading to the overactivation of an adaptive T cell response, in which Th17 cells may act in synergy with T helper 1 cells (Th1) to drive the severe cytokine release syndrome. After discontinuation of trastuzumab/pertuzumab, hypercytokinemia and complement activity normalized along with clinical recovery. Cardiac function returned to baseline within 2 months of initial presentation, together with a resolution of the myocardial inflammation on MRI.


Subject(s)
Breast Neoplasms , Female , Humans , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Cytokines , Shock, Cardiogenic/chemically induced
7.
Viruses ; 14(12)2022 11 28.
Article in English | MEDLINE | ID: mdl-36560669

ABSTRACT

A better understanding of the immunological markers associated with long-lasting immune responses to SARS-CoV-2 infection is of paramount importance. In the present study, we characterized SARS-CoV-2-specific humoral responses in hospitalized (ICU and non-ICU) and non-hospitalized individuals at six months post-onset of symptoms (POS) (N = 95). We showed that the proportion of individuals with detectable anti-SARS-CoV-2 IgG or neutralizing (NAb) responses and the titers of antibodies were significantly reduced in non-hospitalized individuals, compared to ICU- or non-ICU-hospitalized individuals at 6 months POS. Interestingly, SARS-CoV-2-specific memory B cells persist at 6 months POS in both ICU and non-ICU patients and were enriched in cells harboring an activated and/or exhausted phenotype. The frequency/phenotype of SARS-CoV-2-specific memory B cells and the magnitude of IgG or NAb responses at 6 months POS correlated with the serum immune signature detected at patient admission. In particular, the serum levels of CXCL13, IL-1RA, and G-CSF directly correlated with the frequency of Spike-specific B cells and the magnitude of Spike-specific IgG or NAb, while the serum levels of CXCL12 showed an antagonizing effect. Our results indicate that the balance between CXCL12 and CXCL13 is an early marker associated with the magnitude and the quality of the SARS-CoV-2 humoral memory.


Subject(s)
COVID-19 , Chemokine CXCL12 , Chemokine CXCL13 , Cytokines , Immunity, Humoral , Humans , Antibodies, Neutralizing , Antibodies, Viral , Chemokine CXCL12/blood , Chemokine CXCL13/blood , COVID-19/immunology , Cytokines/blood , Immunoglobulin G , SARS-CoV-2
8.
Front Immunol ; 13: 960120, 2022.
Article in English | MEDLINE | ID: mdl-36091040

ABSTRACT

Optimal T follicular helper (Tfh) cells function is important to promote the development of germinal centers and maturation of high affinity antigen-specific B cells. We have found that the expression of CXCR3 defines distinct Tfh subsets: CXCR3+ Th1-like Tfh cells mainly producing single IFN-γ and dual IL-21/IFN-γ and CXCR3- Th2-like Tfh cells mainly producing single IL-4 and dual IL-21/IL-4 cytokines. CXCR3- Th2-like Tfhs are significantly reduced during ongoing HIV replication. While the percentage of Th2-like Tfh cells correlates with that of total and cycling HIV-specific B cells, the percentage of CXCR3+ Th1-like Tfhs correlates with HIV-specific B cells expressing T-bet and CXCR3. Of note, only IL-4 and IL-21 cytokines boosted efficient maturation of HIV-specific B cells while IFN-γ induced expression of T-bet and CXCR3 in B cells. Interestingly, total and HIV-specific CXCR3+ B cells showed lower rate of somatic hypermutation, as compared to CXCR3- B cells. Therefore, the imbalance in Th2/Th1-like Tfhs affects B cell responses in viremic HIV infection.


Subject(s)
HIV Infections , T Follicular Helper Cells , Cytokines/metabolism , Germinal Center/metabolism , HIV Infections/metabolism , Humans , Interleukin-4/metabolism , Viremia
9.
Cell Rep ; 37(2): 109814, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34599871

ABSTRACT

Control of the ongoing SARS-CoV-2 pandemic is endangered by the emergence of viral variants with increased transmission efficiency, resistance to marketed therapeutic antibodies, and reduced sensitivity to vaccine-induced immunity. Here, we screen B cells from COVID-19 donors and identify P5C3, a highly potent and broadly neutralizing monoclonal antibody with picomolar neutralizing activity against all SARS-CoV-2 variants of concern (VOCs) identified to date. Structural characterization of P5C3 Fab in complex with the spike demonstrates a neutralizing activity defined by a large buried surface area, highly overlapping with the receptor-binding domain (RBD) surface necessary for ACE2 interaction. We further demonstrate that P5C3 shows complete prophylactic protection in the SARS-CoV-2-infected hamster challenge model. These results indicate that P5C3 opens exciting perspectives either as a prophylactic agent in immunocompromised individuals with poor response to vaccination or as combination therapy in SARS-CoV-2-infected individuals.


Subject(s)
Broadly Neutralizing Antibodies/therapeutic use , COVID-19 Drug Treatment , SARS-CoV-2/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Broadly Neutralizing Antibodies/immunology , COVID-19/immunology , Cell Line , Cricetinae , Disease Models, Animal , Epitopes/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/metabolism , Neutralization Tests , Protein Binding/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/ultrastructure , Structure-Activity Relationship , Vaccination
10.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33903232

ABSTRACT

The development of follicular helper CD4 T (TFH) cells is a dynamic process resulting in a heterogenous pool of TFH subsets. However, the cellular and molecular determinants of this heterogeneity and the possible mechanistic links between them is not clear. We found that human TFH differentiation is associated with significant changes in phenotypic, chemokine, functional, metabolic and transcriptional profile. Furthermore, this differentiation was associated with distinct positioning to follicular proliferating B cells. Single-cell T cell receptor (TCR) clonotype analysis indicated the transitioning toward PD-1hiCD57hi phenotype. Furthermore, the differentiation of TFH cells was associated with significant reduction in TCR level and drastic changes in immunological synapse formation. TFH synapse lacks a tight cSMAC (central supra molecular activation Cluster) but displays the TCR in peripheral microclusters, which are potentially advantageous in the ability of germinal center (GC) B cells to receive necessary help. Our data reveal significant aspects of human TFH heterogeneity and suggest that the PD-1hiCD57hi TFH cells, in particular, are endowed with distinctive programming and spatial positioning for optimal GC B cell help.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/immunology , Receptors, Antigen, T-Cell/genetics , T Follicular Helper Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD57 Antigens/genetics , Cell Communication/immunology , Cell Differentiation/immunology , Cell Lineage/genetics , Chemokines/genetics , Germinal Center/immunology , Germinal Center/metabolism , Humans , Immunological Synapses/genetics , Immunological Synapses/immunology , Lymphocyte Activation/immunology , Phenotype , Programmed Cell Death 1 Receptor/genetics , Receptors, Antigen, T-Cell/immunology , T Follicular Helper Cells/metabolism , T-Lymphocyte Subsets/immunology
11.
Front Immunol ; 11: 607626, 2020.
Article in English | MEDLINE | ID: mdl-33633728

ABSTRACT

Follicular helper CD4 T (Tfh) cells play an essential role in the formation of germinal centers (GCs), where mature B cells proliferate, differentiate, and provide long-term protective humoral responses. Despite the extensive phenotypic characterization and identification of human Tfh cell subsets, their spatial positioning at tissue level is not well understood. Here, we describe a quantitative multiplexed immunofluorescence approach allowing for the comprehensive in situ characterization of Tfh cells in human tonsils and lymph nodes (LNs) from individuals with angioimmunoblastic T-cell lymphoma (AITL). We have developed eight multiplexed panels comprising a spectrum of Tfh cell markers, like PD-1, CXCR5, and ICOS, along with transcription factors (Bcl6, Tbet, GATA3), to assess their expression, frequencies, spatial distribution and co-localization in a quantitative manner. Combined analysis of relevant markers revealed the presence of several Tfh cell subsets at tissue level based on the differential expression of surface receptors, nuclear factors as well as their distinct localization within the follicular areas. Interestingly, we found a considerable amount of tonsillar Tfh cells expressing high levels of the Th2 regulator GATA3. The co-expression of GATA3, CXCR5, and BCL6, points to an important role of GATA3 for the generation of effector human Tfh cells. Furthermore, our data revealed significantly different Tfh cell profile signatures between health and disease. Therefore, our imaging platform generates meaningful information for the in situ characterization of human Tfh cells and could provide the base for future studies aiming to a comprehensive understanding of Tfh cell tissue heterogeneity.


Subject(s)
Immunoblastic Lymphadenopathy/metabolism , Lymph Nodes/metabolism , Lymphoma, T-Cell/metabolism , Microscopy, Fluorescence , Palatine Tonsil/metabolism , T Follicular Helper Cells/metabolism , Biomarkers/metabolism , Fluorescent Antibody Technique , Humans , Image Processing, Computer-Assisted , Immunoblastic Lymphadenopathy/immunology , Immunoblastic Lymphadenopathy/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Palatine Tonsil/immunology , Palatine Tonsil/pathology , Phenotype , Proteome , Proteomics , T Follicular Helper Cells/immunology
12.
Immunol Rev ; 292(1): 149-163, 2019 11.
Article in English | MEDLINE | ID: mdl-31883174

ABSTRACT

The T-cell response is central in the adaptive immune-mediated elimination of pathogen-infected and/or cancer cells. This activated T-cell response can inflict an overwhelming degree of damage to the targeted cells, which in most instances leads to the control and elimination of foreign invaders. However, in conditions of chronic infection, persistent exposure of T cells to high levels of antigen results in a severe T-cell dysfunctional state called exhaustion. T-cell exhaustion leads to a suboptimal immune-mediated control of multiple viral infections including the human immunodeficiency virus (HIV). In this review, we will discuss the role of T-cell exhaustion in HIV disease progression, the long-term defect of T-cell function even in aviremic patients on antiretroviral therapy (ART), the role of exhaustion-specific markers in maintaining a reservoir of latently infected cells, and exploiting these markers in HIV cure strategies.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Disease Progression , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Lymphocyte Activation/immunology
13.
PLoS Pathog ; 15(7): e1007918, 2019 07.
Article in English | MEDLINE | ID: mdl-31329640

ABSTRACT

T-follicular helper (Tfh) cells, co-expressing PD-1 and TIGIT, serve as a major cell reservoir for HIV-1 and are responsible for active and persistent HIV-1 transcription after prolonged antiretroviral therapy (ART). However, the precise mechanisms regulating HIV-1 transcription in lymph nodes (LNs) remain unclear. In the present study, we investigated the potential role of immune checkpoint (IC)/IC-Ligand (IC-L) interactions on HIV-1 transcription in LN-microenvironment. We show that PD-L1 (PD-1-ligand) and CD155 (TIGIT-ligand) are predominantly co-expressed on LN migratory (CD1chighCCR7+CD127+) dendritic cells (DCs), that locate predominantly in extra-follicular areas in ART treated individuals. We demonstrate that TCR-mediated HIV production is suppressed in vitro in the presence of recombinant PD-L1 or CD155 and, more importantly, when LN migratory DCs are co-cultured with PD-1+/Tfh cells. These results indicate that LN migratory DCs expressing IC-Ls may more efficiently restrict HIV-1 transcription in the extra-follicular areas and explain the persistence of HIV transcription in PD-1+/Tfh cells after prolonged ART within germinal centers.


Subject(s)
HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HIV-1/pathogenicity , Programmed Cell Death 1 Receptor/metabolism , Anti-HIV Agents/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Cell Movement/immunology , Cellular Microenvironment/immunology , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/virology , Germinal Center/immunology , Germinal Center/virology , HIV Infections/drug therapy , HIV-1/immunology , Host Microbial Interactions/immunology , Humans , In Vitro Techniques , Lymph Nodes/immunology , Lymph Nodes/virology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Immunologic/metabolism , Receptors, Virus/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Transcription, Genetic , Virulence
14.
J Exp Med ; 216(7): 1525-1541, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31123083

ABSTRACT

Classical antagonistic antibodies (Abs) targeting PD-1, such as pembrolizumab and nivolumab, act through blockade of the PD-1-PDL-1 interaction. Here, we have identified novel antagonistic anti-PD-1 Abs not blocking the PD-1-PDL-1 interaction. The nonblocking Abs recognize epitopes on PD-1 located on the opposing face of the PDL-1 interaction and overlap with a newly identified evolutionarily conserved patch. These nonblocking Abs act predominantly through the CD28 coreceptor. Importantly, a combination of blocking and nonblocking Abs synergize in the functional recovery of antigen-specific exhausted CD8 T cells. Interestingly, nonblocking anti-PD-1 Abs have equivalent antitumor activity compared with blocker Abs in two mouse tumor models, and combination therapy using both classes of Abs enhanced tumor suppression in the mouse immunogenic tumor model. The identification of the novel nonblocker anti-PD-1 Abs and their synergy with classical blocker Abs may be instrumental in potentiating immunotherapy strategies and antitumor activity.


Subject(s)
Antineoplastic Agents, Immunological/immunology , CD28 Antigens/metabolism , Neoplasms, Experimental/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antineoplastic Agents, Immunological/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Humans , Jurkat Cells , Mice , NF-kappa B/metabolism , Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
15.
J Virol ; 92(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-29976671

ABSTRACT

A recent study conducted in blood has proposed CD32 as the marker identifying the "elusive" HIV reservoir. We have investigated the distribution of CD32+ CD4 T cells in blood and lymph nodes (LNs) of HIV-1-uninfected subjects and viremic untreated and long-term-treated HIV-1-infected individuals and their relationship with PD-1+ CD4 T cells. The frequency of CD32+ CD4 T cells was increased in viremic compared to treated individuals in LNs, and a large proportion (up to 50%) of CD32+ cells coexpressed PD-1 and were enriched within T follicular helper (Tfh) cells. We next investigated the role of LN CD32+ CD4 T cells in the HIV reservoir. Total HIV DNA was enriched in CD32+ and PD-1+ CD4 T cells compared to CD32- and PD-1- cells in both viremic and treated individuals, but there was no difference between CD32+ and PD-1+ cells. There was no enrichment of latently infected cells with inducible HIV-1 in CD32+ versus PD-1+ cells in antiretroviral therapy (ART)-treated individuals. HIV-1 transcription was then analyzed in LN memory CD4 T cell populations sorted on the basis of CD32 and PD-1 expression. CD32+ PD-1+ CD4 T cells were significantly enriched in cell-associated HIV RNA compared to CD32- PD-1- (averages of 5.2-fold in treated individuals and 86.6-fold in viremics), CD32+ PD-1- (2.2-fold in treated individuals and 4.3-fold in viremics), and CD32- PD-1+ (2.2-fold in ART-treated individuals and 4.6-fold in viremics) cell populations. Similar levels of HIV-1 transcription were found in CD32+ PD-1- and CD32- PD-1+ CD4 T cells. Interestingly, the proportion of CD32+ and PD-1+ CD4 T cells negatively correlated with CD4 T cell counts and length of therapy. Therefore, the expression of CD32 identifies, independently of PD-1, a CD4 T cell population with persistent HIV-1 transcription and coexpression of CD32 and PD-1, the CD4 T cell population with the highest levels of HIV-1 transcription in both viremic and treated individuals.IMPORTANCE The existence of long-lived latently infected resting memory CD4 T cells represents a major obstacle to the eradication of HIV infection. Identifying cell markers defining latently infected cells containing replication-competent virus is important in order to determine the mechanisms of HIV persistence and to develop novel therapeutic strategies to cure HIV infection. We provide evidence that PD-1 and CD32 may have a complementary role in better defining CD4 T cell populations infected with HIV-1. Furthermore, CD4 T cells coexpressing CD32 and PD-1 identify a CD4 T cell population with high levels of persistent HIV-1 transcription.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/pathology , HIV-1/growth & development , Lymph Nodes/pathology , T-Lymphocyte Subsets/virology , Transcription, Genetic , Adult , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/chemistry , DNA, Viral/analysis , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Programmed Cell Death 1 Receptor/analysis , RNA, Viral/analysis , Receptors, IgG/analysis , T-Lymphocyte Subsets/chemistry , Young Adult
16.
Front Immunol ; 9: 144, 2018.
Article in English | MEDLINE | ID: mdl-29459864

ABSTRACT

We recently demonstrated that lymph nodes (LNs) PD-1+/T follicular helper (Tfh) cells from antiretroviral therapy (ART)-treated HIV-infected individuals were enriched in cells containing replication competent virus. However, the distribution of cells containing inducible replication competent virus has been only partially elucidated in blood memory CD4 T-cell populations including the Tfh cell counterpart circulating in blood (cTfh). In this context, we have investigated the distribution of (1) total HIV-infected cells and (2) cells containing replication competent and infectious virus within various blood and LN memory CD4 T-cell populations of conventional antiretroviral therapy (cART)-treated HIV-infected individuals. In the present study, we show that blood CXCR3-expressing memory CD4 T cells are enriched in cells containing inducible replication competent virus and contributed the most to the total pool of cells containing replication competent and infectious virus in blood. Interestingly, subsequent proviral sequence analysis did not indicate virus compartmentalization between blood and LN CD4 T-cell populations, suggesting dynamic interchanges between the two compartments. We then investigated whether the composition of blood HIV reservoir may reflect the polarization of LN CD4 T cells at the time of reservoir seeding and showed that LN PD-1+ CD4 T cells of viremic untreated HIV-infected individuals expressed significantly higher levels of CXCR3 as compared to CCR4 and/or CCR6, suggesting that blood CXCR3-expressing CD4 T cells may originate from LN PD-1+ CD4 T cells. Taken together, these results indicate that blood CXCR3-expressing CD4 T cells represent the major blood compartment containing inducible replication competent virus in treated aviremic HIV-infected individuals.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Receptors, CXCR3/immunology , Adult , Anti-Retroviral Agents/therapeutic use , DNA, Viral/blood , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/physiology , Humans , Programmed Cell Death 1 Receptor/immunology , RNA, Viral/analysis , Virus Replication
17.
Curr Opin HIV AIDS ; 12(3): 203-208, 2017 May.
Article in English | MEDLINE | ID: mdl-28422784

ABSTRACT

PURPOSE OF REVIEW: The purpose of the present review is to provide an update on the current development in the field of broadly neutralizing antibodies (bNabs) and their potential use in the prevention and therapeutic settings, and an evaluation of the B-cell abnormalities that may impair antibody responses in HIV infection. RECENT FINDINGS: Major advances have been achieved in the characterization of bNabs directed against different vulnerable regions of HIV Envelope (Env). Recent observations have clearly demonstrated the ability of bNabs to prevent HIV infection in the nonhuman primate model of HIV infection and to suppress viremia in individuals with chronic HIV infection in the absence of antiretroviral therapy. Furthermore, substantial advances have also been obtained in the development of HIV Env proteins and immunization strategies inducing bNabs in small animal models. Several studies have also shed light on the B-cell abnormalities associated with the viremic phase of HIV infection that cause impaired B-cell maturation and antibody responses. Of note, preliminary observations have provided evidence for a correlation between the expansion of a specific population of B cells, for example, germinal center B cells, the expansion of T follicular helper cells (Tfh), and the generation of neutralizing antibodies. SUMMARY: The recent observations on the antiviral effects of bNabs in vivo indicate that bNabs may play a central role in both the prevention and the therapeutic settings. The identification of the role of germinal center B cells and Tfh cells as critical components of the immune response leading to the generation of neutralizing antibodies, will allow the development of specific immunization strategies for the stimulation of germinal center B cells and Tfh cells. A lot of work still remains to be done for the delineation of B-cell and Tfh cell biology from human lymphoid tissues and in the development of HIV Env proteins and immunization strategies leading to the generation of bNabs.


Subject(s)
B-Lymphocytes/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Animals , Antibody Formation , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Humans
18.
Nat Med ; 22(7): 754-61, 2016 07.
Article in English | MEDLINE | ID: mdl-27239760

ABSTRACT

The mechanisms responsible for the persistence of HIV-1 after many years of suppressive antiretroviral therapy (ART) have been only partially elucidated. Most of the studies investigating HIV-1 persistence have been performed with blood, although it is well known that germinal centers (GCs) within lymph nodes (LNs) serve as primary sites for HIV-1 replication. We sought to identify the memory CD4 T cell populations in blood and LNs that are responsible for the production of replication-competent and infectious HIV-1, as well as for active and persistent virus transcription in ART-treated (for 1.5-14.0 years), aviremic (<50 HIV RNA copies/ml) HIV-infected individuals. We demonstrate that LN CD4 T cells that express programmed cell death 1 (PDCD1; also known as PD-1), which are composed of about 65% T follicular helper cells as defined by the expression of the cell surface receptors CXCR5 and PD-1, are the major source of replication-competent HIV-1 and of infectious virus, as compared to any other (CXCR5(-)PD-1(-) and CXCR5(+)PD-1(-)) blood or LN memory CD4 T cell populations. LN PD-1(+) cells accounted for 46% and 96% of the total pools of memory CD4 T cells containing inducible replication-competent or infectious virus, respectively. Notably, higher levels of cell-associated HIV-1 RNA were present in LN PD-1(+) cells after long-term (up to 12 years) ART than in other memory CD4 T cell subpopulations. These results indicate that LN PD-1(+) cells are the major CD4 T cell compartment in the blood and LNs for the production of replication-competent and infectious HIV-1, and for active and persistent virus transcription in long-term-ART-treated aviremic individuals. Thus, these cells may represent a major obstacle to finding a functional cure for HIV-1 infection.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV-1/genetics , Lymph Nodes/virology , T-Lymphocytes, Helper-Inducer/virology , Virus Replication , Adult , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Female , HIV-1/physiology , Humans , Lymph Nodes/cytology , Male , Middle Aged , Programmed Cell Death 1 Receptor/metabolism , RNA, Viral/metabolism , Receptors, CXCR5/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
19.
J Immunol ; 191(2): 540-4, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772031

ABSTRACT

Ag-specific CD8 T cells play a critical role in controlling HIV infection but eventually lose antiviral functions in part because of expression and signaling through the inhibitory programmed death-1 (PD-1) receptor. To better understand the impact of prolonged TCR ligation on regulation of PD-1 expression in HIV-specific CD8 T cells, we investigated the capacity of virus-specific CD8 T cells to modify the PD-1 epigenetic program after reduction in viral load. We observed that the transcriptional regulatory region was unmethylated in the PD-1(hi) HIV-specific CD8 T cells, whereas it remained methylated in donor-matched naive cells at acute and chronic stages of infection. Surprisingly, the PD-1 promoter remained unmethylated in HIV-specific CD8 T cells from subjects with a viral load controlled by antiviral therapy for >2 y or from elite controllers. Together, these data demonstrate that the epigenetic program at the PD-1 locus becomes fixed after prolonged exposure to HIV virus.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA Methylation , HIV Infections/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Antiretroviral Therapy, Highly Active , CD8-Positive T-Lymphocytes/metabolism , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/immunology , Humans , Promoter Regions, Genetic , Transcription, Genetic , Viral Load
20.
Vaccines (Basel) ; 1(4): 481-96, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-26344343

ABSTRACT

Increasing numbers of HIV-infected individuals have access to potent antiretroviral drugs that control viral replication and decrease the risk of transmission. However, there is no cure for HIV and new strategies have to be developed to reach an eradication of the virus or a natural control of viral replication in the absence of drugs (functional cure). Therapeutic vaccines against HIV have been evaluated in many trials over the last 20 years and important knowledge has been gained from these trials. However, the major obstacle to HIV eradication is the persistence of latent proviral reservoirs. Different molecules are currently tested in ART-treated subjects to reactivate these latent reservoirs. Such anti-latency agents should be combined with a vaccination regimen in order to control or eradicate reactivated latently-infected cells. New in vitro assays should also be developed to assess the success of tested therapeutic vaccines by measuring the immune-mediated killing of replication-competent HIV reservoir cells. This review provides an overview of the current strategies to combine HIV vaccines with anti-latency agents that could act as adjuvant on the vaccine-induced immune response as well as new tools to assess the efficacy of these approaches.

SELECTION OF CITATIONS
SEARCH DETAIL
...