Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cell ; 184(15): 3899-3914.e16, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34237254

ABSTRACT

The impact of the microbiome on HIV disease is widely acknowledged although the mechanisms downstream of fluctuations in microbial composition remain speculative. We detected rapid, dynamic changes in translocated microbial constituents during two years after cART initiation. An unbiased systems biology approach revealed two distinct pathways driven by changes in the abundance ratio of Serratia to other bacterial genera. Increased CD4 T cell numbers over the first year were associated with high Serratia abundance, pro-inflammatory innate cytokines, and metabolites that drive Th17 gene expression signatures and restoration of mucosal integrity. Subsequently, decreased Serratia abundance and downregulation of innate cytokines allowed re-establishment of systemic T cell homeostasis promoting restoration of Th1 and Th2 gene expression signatures. Analyses of three other geographically distinct cohorts of treated HIV infection established a more generalized principle that changes in diversity and composition of translocated microbial species influence systemic inflammation and consequently CD4 T cell recovery.


Subject(s)
Gastrointestinal Microbiome , HIV Infections/immunology , HIV Infections/microbiology , Antiretroviral Therapy, Highly Active , Biodiversity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokines/blood , Cohort Studies , Glycolysis , HIV Infections/blood , HIV Infections/drug therapy , Humans , Inflammation/genetics , Inflammation/pathology , Mitochondria/metabolism , Monocytes/metabolism , Nucleic Acids/blood , Principal Component Analysis , Serratia/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Transcription, Genetic , Uganda , Viral Load/immunology
2.
Nat Immunol ; 20(8): 1059-1070, 2019 08.
Article in English | MEDLINE | ID: mdl-31308541

ABSTRACT

Dysfunction of virus-specific CD4+ T cells in chronic human infections is poorly understood. We performed genome-wide transcriptional analyses and functional assays of CD4+ T cells specific for human immunodeficiency virus (HIV) from HIV-infected people before and after initiation of antiretroviral therapy (ART). A follicular helper T cell (TFH cell)-like profile characterized HIV-specific CD4+ T cells in viremic infection. HIV-specific CD4+ T cells from people spontaneously controlling the virus (elite controllers) robustly expressed genes associated with the TH1, TH17 and TH22 subsets of helper T cells. Viral suppression by ART resulted in a distinct transcriptional landscape, with a reduction in the expression of genes associated with TFH cells, but persistently low expression of genes associated with TH1, TH17 and TH22 cells compared to the elite controller profile. Thus, altered differentiation is central to the impairment of HIV-specific CD4+ T cells and involves both gain of function and loss of function.


Subject(s)
Anti-HIV Agents/therapeutic use , Gene Expression/drug effects , HIV Infections/drug therapy , HIV Infections/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Gene Expression Profiling , HIV Infections/virology , Humans , Receptors, CXCR5/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Viral Load/drug effects , Virus Replication/drug effects
3.
J Immunol ; 201(3): 971-981, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29934472

ABSTRACT

Immune exhaustion is an important feature of chronic infections, such as HIV, and a barrier to effective immunity against cancer. This dysfunction is in part controlled by inhibitory immune checkpoints. Blockade of the PD-1 or IL-10 pathways can reinvigorate HIV-specific CD4 T cell function in vitro, as measured by cytokine secretion and proliferative responses upon Ag stimulation. However, whether this restoration of HIV-specific CD4 T cells can improve help to other cell subsets impaired in HIV infection remains to be determined. In this study, we examine a cohort of chronically infected subjects prior to initiation of antiretroviral therapy (ART) and individuals with suppressed viral load on ART. We show that IFN-γ induction in NK cells upon PBMC stimulation by HIV Ag varies inversely with viremia and depends on HIV-specific CD4 T cell help. We demonstrate in both untreated and ART-suppressed individuals that dual PD-1 and IL-10 blockade enhances cytokine secretion of NK cells via restored HIV-specific CD4 T cell function, that soluble factors contribute to these immunotherapeutic effects, and that they depend on IL-2 and IL-12 signaling. Importantly, we show that inhibition of the PD-1 and IL-10 pathways also increases NK degranulation and killing of target cells. This study demonstrates a previously underappreciated relationship between CD4 T cell impairment and NK cell exhaustion in HIV infection, provides a proof of principle that reversal of adaptive immunity exhaustion can improve the innate immune response, and suggests that immune checkpoint modulation that improves CD4/NK cell cooperation can be used as adjuvant therapy in HIV infection.


Subject(s)
Anti-Retroviral Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , HIV Infections/drug therapy , Killer Cells, Natural/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cohort Studies , HIV Infections/immunology , HIV-1/drug effects , HIV-1/immunology , Humans , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-2/immunology , K562 Cells , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Programmed Cell Death 1 Receptor/immunology , Viral Load/drug effects , Viral Load/immunology
4.
AIDS Res Ther ; 14(1): 40, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28893281

ABSTRACT

Despite the tremendous success of anti-retroviral therapy (ART) no current treatment can eradicate latent HIV reservoirs from HIV-infected individuals or generate, effective HIV-specific immunity. Technological limitations have hampered the identification and characterization of both HIV-infected cells and HIV-specific responses in clinical samples directly ex vivo. RNA-flow cytometric fluorescence in situ hybridisation (RNA Flow-FISH) is a powerful technique, which enables detection of mRNAs in conjunction with proteins at a single-cell level. Here, we describe how we are using this technology to address some of the major questions remaining in the HIV field in the era of ART. We discuss how CD4 T cell responses to HIV antigens, both following vaccination and HIV infection, can be characterized by measurement of cytokine mRNAs. We describe how our development of a dual HIV mRNA/protein assay (HIVRNA/Gag assay) enables high sensitivity detection of very rare HIV-infected cells and aids investigations into the translation-competent latent reservoir in the context of HIV cure.


Subject(s)
Flow Cytometry/methods , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/immunology , In Situ Hybridization, Fluorescence/methods , RNA/analysis , AIDS Vaccines , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , CD4-Positive T-Lymphocytes/virology , Cytokines/genetics , Cytokines/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Vaccination , Virus Latency
5.
PLoS Pathog ; 10(5): e1004136, 2014 May.
Article in English | MEDLINE | ID: mdl-24830456

ABSTRACT

Earlier studies had suggested that epigenetic mechanisms play an important role in the control of human cytomegalovirus (HCMV) infection. Here we show that productive HCMV infection is indeed under the control of histone H3K27 trimethylation. The histone H3K27 methyltransferase EZH2, and its regulators JARID2 and NDY1/KDM2B repress GFI1, a transcriptional repressor of the major immediate-early promoter (MIEP) of HCMV. Knocking down EZH2, NDY1/KDM2B or JARID2 relieves the repression and results in the upregulation of GFI1. During infection, the incoming HCMV rapidly downregulates the GFI1 mRNA and protein in both wild-type cells and in cells in which EZH2, NDY1/KDM2B or JARID2 were knocked down. However, since the pre-infection levels of GFI1 in the latter cells are significantly higher, the virus fails to downregulate it to levels permissive for MIEP activation and viral infection. Following the EZH2-NDY1/KDM2B-JARID2-independent downregulation of GFI1 in the early stages of infection, the virus also initiates an EZH2-NDY1/ΚDM2Β-JARID2-dependent program that represses GFI1 throughout the infection cycle. The EZH2 knockdown also delays histone H3K27 trimethylation in the immediate early region of HCMV, which is accompanied by a drop in H3K4 trimethylation that may contribute to the shEZH2-mediated repression of the major immediate early HCMV promoter. These data show that HCMV uses multiple mechanisms to allow the activation of the HCMV MIEP and to prevent cellular mechanisms from blocking the HCMV replication program.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus/physiology , DNA-Binding Proteins/genetics , F-Box Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Polycomb Repressive Complex 2/physiology , Promoter Regions, Genetic , Transcription Factors/genetics , Antigens, Viral/genetics , Cells, Cultured , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein , HEK293 Cells , HeLa Cells , Humans , Immediate-Early Proteins/genetics , Signal Transduction/physiology , Transcription Factors/metabolism , Viral Proteins/genetics , Viral Proteins/physiology , Virus Replication/genetics
6.
EBioMedicine ; 84: 104254, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36150362

ABSTRACT

BACKGROUND: Immune checkpoint blockade (ICB) partially reverses the dysfunctional state of antigen-specific T cell in chronic infections. However, its impact on the diverse subsets of CD4+ T cells in humans is largely unknown. METHODS: We examined immune checkpoint (IC) expression and function in HIV-specific CD4+ T cells of viremic individuals (≥5000 vRNA cp/ml, n = 17) prior to ART and persons with spontaneous (n = 11) or therapy-induced (n = 16) viral suppression (<40 cp/ml). We investigated IC patterns associated with exhaustion-related transcription factors and chemokine receptors using activation-induced marker assays. We determined effector functions representative of TFH, TH1, and TH17/TH22 using RNA flow cytometric fluorescence in situ hybridization (FISH). We compared increase in cytokine expression upon ICB across functions and patient status. FINDINGS: Expression of dysfunction-related molecules, such as transcription factors and ICs PD-1, TIGIT, and CD200, followed a hierarchy associated with infection status and effector profile. In vitro responsiveness to PD-L1 blockade varied with defined functions rather than IC levels: frequencies of cells with TH1- and TH17/TH22-, but not TFH-related functions, increased. Cells co-expressing TH1 and TFH functions showed response to ICB, suggesting that the cell's state rather than function dictates responsiveness to PD-L1 blockade. Response to PD-L1 blockade was strongest in viremic participants and reduced after ART initiation. INTERPRETATION: Our data highlight a polarization-specific regulation of IC expression and differing sensitivities of antigen-specific T helper subsets to PD-1-mediated inhibition. This heterogeneity may direct and constrain ICB efficacy in restoring CD4+ T cell function in HIV infection and other diseases. FUNDING: NIH, CIHR, CFI, FRQS.


Subject(s)
B7-H1 Antigen , HIV Infections , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors , In Situ Hybridization, Fluorescence , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , RNA/therapeutic use , Receptors, Chemokine/metabolism , Receptors, Chemokine/therapeutic use , Receptors, Immunologic/metabolism , Transcription Factors/metabolism
7.
EBioMedicine ; 54: 102727, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32268275

ABSTRACT

BACKGROUND: Untreated HIV infection leads to alterations in HIV-specific CD4+ T cells including increased expression of co-inhibitory receptors (IRs) and skewing toward a T follicular helper cell (Tfh) signature. However, which changes are maintained after suppression of viral replication with antiretroviral therapy (ART) is poorly known. METHODS: We analyzed blood CD4+ T cells specific to HIV and comparative viral antigens in ART-treated people using a cytokine-independent activation-induced marker assay alone or in combination with functional readouts. FINDINGS: In intra-individual comparisons, HIV-specific CD4+ T cells were characterized by a larger fraction of circulating Tfh (cTfh) cells than CMV- and HBV-specific cells and preferentially expressed multiple IRs and showed elevated production of the Tfh cytokines CXCL13 and IL-21. In addition, HIV-specific cTfh exhibited a predominant Th1-like phenotype and function when compared to cTfh of other specificities, contrasting with a reduction in Th1-functions in HIV-specific non-cTfh. Using longitudinal samples, we demonstrate that this distinct HIV-specific cTfh profile was induced during chronic untreated HIV infection, persisted on ART and correlated with the translation-competent HIV reservoir but not with the total HIV DNA reservoir. INTERPRETATION: Expansion and altered features of HIV-specific cTfh cells are maintained during ART and may be driven by persistent HIV antigen expression. FUNDING: This work was supported by the National Institutes of Health (NIH), the Canadian Institutes of Health Research (CIHR) and the FRQS AIDS and Infectious Diseases Network.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/blood , T Follicular Helper Cells/immunology , Th1 Cells/immunology , Cells, Cultured , Chemokine CXCL13/metabolism , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Interleukins/metabolism
8.
J Clin Invest ; 129(6): 2463-2479, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30912767

ABSTRACT

Rationale Tumor infiltrating lymphocytes are widely associated with positive outcomes, yet carry key indicators of a systemic failed immune response against unresolved cancer. Cancer immunotherapies can reverse their tolerance phenotypes, while preserving tumor-reactivity and neoantigen-specificity shared with circulating immune cells. Objectives We performed comprehensive transcriptomic analyses to identify gene signatures common to circulating and tumor infiltrating lymphocytes in the context of clear cell renal cell carcinoma. Modulated genes also associated with disease outcome were validated in other cancer types. Findings Using bioinformatics, we identified practical diagnostic markers and actionable targets of the failed immune response. On circulating lymphocytes, three genes, LEF1, FASLG, and MMP9, could efficiently stratify patients from healthy control donors. From their associations with resistance to cancer immunotherapies and microbial infections, we uncovered not only pan-cancer, but pan-pathology failed immune response profiles. A prominent lymphocytic matrix metallopeptidase cell migration pathway, is central to a panoply of diseases and tumor immunogenicity, correlates with multi-cancer recurrence, and identifies a feasible, non-invasive approach to pan-pathology diagnoses. Conclusions The non-invasive differently expressed genes we have identified warrant future investigation towards the development of their potential in precision diagnostics and precision pan-disease immunotherapeutics.


Subject(s)
Carcinoma, Renal Cell , Gene Expression Regulation, Neoplastic/immunology , Immunotherapy , Kidney Neoplasms , Lymphocytes, Tumor-Infiltrating , Neoplasm Proteins , Tumor Microenvironment/immunology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Female , Gene Expression Profiling , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology
9.
Toxicol Lett ; 176(1): 77-84, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-18054450

ABSTRACT

Staphylococcal enterotoxin A (SEA) is a potent stimulator of CD4+ and CD8+ T cells, the immunotoxic action of which remains unclear. We investigated the in vitro effects of SEA on freshly isolated human peripheral blood lymphocytes depleted of CD8+ T cells. Proliferation and flow cytometry analysis indicated that SEA generated an activation-induced cell death (AICD) phenomenon that was characterized by an increased expression of the chemokine receptor CCR5 on the CD4+/CD45RO+ T cell subset. Incubation of cells with glycoprotein secretion inhibitor monensin A completely blocked cell proliferation, affecting mainly the CD4+/CD45RO+ T cell subset. The IL-2 mRNA levels were increased just hours after SEA stimulation, accompanied by an increase in the expression of CD25, indicating a possible involvement of IL-2 in the AICD process. We observed a 15-fold mRNA reduction of the transcription factor Yin Yang 1 (YY1) at the proliferation peak, and an increase of the receptors CCR5, CD95 and DR5 on the CD45RO+/CD4+ T cell subset. These findings suggest that SEA triggers a TCR-mediated AICD mechanism in CD4+ T cells, the intracellular signalling of which is probably modulated, at least, by YY1.


Subject(s)
Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , Enterotoxins/toxicity , Lymphocyte Activation/drug effects , Signal Transduction/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Humans , Inflammation Mediators/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Leukocyte Common Antigens/analysis , Monensin/pharmacology , RNA, Messenger/metabolism , Receptors, CCR5/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Time Factors , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , fas Receptor/metabolism
10.
PLoS One ; 12(10): e0186998, 2017.
Article in English | MEDLINE | ID: mdl-29065175

ABSTRACT

The identification and study of antigen-specific CD4 T cells, both in peripheral blood and in tissues, is key for a broad range of immunological research, including vaccine responses and infectious diseases. Detection of these cells is hampered by both their rarity and their heterogeneity, in particular with regards to cytokine secretion profiles. These factors prevent the identification of the total pool of antigen-specific CD4 T cells by classical methods. We have developed assays for the highly sensitive detection of such cells by measuring the upregulation of surface activation induced markers (AIM). Here, we compare two such assays based on concurrent expression of CD69 plus CD40L (CD154) or expression of OX40 plus CD25, and we develop additional AIM assays based on OX40 plus PD-L1 or 4-1BB. We compare the relative sensitivity of these assays for detection of vaccine and natural infection-induced CD4 T cell responses and show that these assays identify distinct, but overlapping populations of antigen-specific CD4 T cells, a subpopulation of which can also be detected on the basis of cytokine synthesis. Bystander activation had minimal effect on AIM markers. However, some T regulatory cells upregulate CD25 upon antigen stimulation. We therefore validated AIM assays designed to exclude most T regulatory cells, for both human and non-human primate (NHP, Macaca mulatta) studies. Overall, through head-to-head comparisons and methodological improvements, we show that AIM assays represent a sensitive and valuable method for the detection of antigen-specific CD4 T cells.


Subject(s)
Antigens/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Animals , Antigens, CD/immunology , Bystander Effect , Cohort Studies , Humans , Macaca mulatta
11.
Curr Opin HIV AIDS ; 9(5): 446-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25023623

ABSTRACT

PURPOSE OF REVIEW: To describe recent advances in the understanding of virus-specific CD4 T cell dysfunction in chronic viral infections, with an emphasis on HIV disease. We highlight features that are distinctive for CD4 T cells, as opposed to their CD8 T cell counterparts. RECENT FINDINGS: CD4 T cell activation and differentiation are tightly controlled. Regulation of these processes depends on the context of initial encounter of the naïve CD4 T cell with the cognate antigen and on ongoing external cues to the antigen-experienced CD4 T cell, in particular the inflammatory environment, which is prominent in HIV infection. Virus-specific CD4 T cell dysfunction results from a combination of an exhaustion program and skewing in T helper lineage differentiation which impact function. The CD4 and CD8 T cell exhaustion programs present similarities and distinct features. The sets of inhibitory coreceptors expression differ, although programmed-death 1 (PD-1) and T cell immunoglobulin mucin-3 (Tim-3) are upregulated on both HIV-specific CD4 and CD8 T cells, cytotoxic T-lymphocyte antigen 4 (CTLA-4) is largely specific to CD4 T cells, whereas 2B4 and CD160 are biased toward CD8 T cells. SUMMARY: Understanding the molecular basis of HIV-specific CD4 T cell exhaustion and identifying key differences with CD8 T cell impairment will be critical to design effective therapeutic and preventive immunotherapies against HIV.


Subject(s)
CD4-Positive T-Lymphocytes , Chronic Disease , HIV Infections , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , Humans , Lymphocyte Activation
SELECTION OF CITATIONS
SEARCH DETAIL