1
|
Opsteen S, Fram T, Files JK, Levitan EB, Goepfert P, Erdmann N. Impact of Chronic HIV Infection on Acute Immune Responses to SARS-CoV-2. J Acquir Immune Defic Syndr 2024; 96:92-100. [PMID: 38408318 PMCID: PMC11009054 DOI: 10.1097/qai.0000000000003399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024]
Abstract
ABSTRACT There is mounting evidence that HIV infection is a risk factor for severe presentations of COVID-19. We hypothesized that the persistent immune activation associated with chronic HIV infection contributes to worsened outcomes during acute COVID-19. The goals of this study were to provide an in-depth analysis of immune response to acute COVID-19 and investigate relationships between immune responses and clinical outcomes in an unvaccinated, sex- and race-matched cohort of people with HIV (PWH, n = 20) and people without HIV (PWOH, n = 41). We performed flow cytometric analyses on peripheral blood mononuclear cells from PWH and PWOH experiencing acute COVID-19 (≤21-day postsymptom onset). PWH were younger (median 52 vs 65 years) and had milder COVID-19 (40% vs 88% hospitalized) compared with PWOH. Flow cytometry panels included surface markers for immune cell populations, activation and exhaustion surface markers (with and without SARS-CoV-2-specific antigen stimulation), and intracellular cytokine staining. We observed that PWH had increased expression of activation (eg, CD137 and OX40) and exhaustion (eg, PD1 and TIGIT) markers as compared to PWOH during acute COVID-19. When analyzing the impact of COVID-19 severity, we found that hospitalized PWH had lower nonclassical (CD16 + ) monocyte frequencies, decreased expression of TIM3 on CD4 + T cells, and increased expression of PDL1 and CD69 on CD8 + T cells. Our findings demonstrate that PWH have increased immune activation and exhaustion as compared to a cohort of predominately older, hospitalized PWOH and raises questions on how chronic immune activation affects acute disease and the development of postacute sequelae.
Collapse
Affiliation(s)
- Skye Opsteen
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Tim Fram
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Jacob K. Files
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Emily B. Levitan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Nathaniel Erdmann
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| |
Collapse
|
2
|
Manandhar P, Szymczak-Workman AL, Kane LP. Tim-3 Is Not Required for Establishment of CD8+ T Cell Memory to Lymphocytic Choriomeningitis Virus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:466-474. [PMID: 38108417 PMCID: PMC10906969 DOI: 10.4049/jimmunol.2300401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Tim-3 is a transmembrane protein that is best known for being highly expressed on terminally exhausted CD8+ T cells associated with chronic infection and tumors, although its expression is not limited to those settings. Tim-3 is also expressed by CD8+ T cells during acute infection and by multiple other immune cell types, including CD4+ Th1 and regulatory T cells, dendritic cells, and mast cells. In this study, we investigated the role of Tim-3 signaling on CD8+ T cell memory using a Tim-3 conditional knockout mouse model and mice lacking the signaling portion of the Tim-3 cytoplasmic domain. Together, our results indicate that Tim-3 has at most a modest effect on the formation and function of CD8+ memory T cells.
Collapse
Affiliation(s)
- Priyanka Manandhar
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| | | | - Lawrence P. Kane
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
3
|
Muñoz-Muela E, Trujillo-Rodríguez M, Serna-Gallego A, Saborido-Alconchel A, Ruiz-Mateos E, López-Cortés LF, Gutiérrez-Valencia A. HIV-1-specific T-cell responses and exhaustion profiles in people with HIV after switching to dual therapy vs. maintaining triple therapy based on integrase inhibitors. Biomed Pharmacother 2023; 168:115750. [PMID: 37871555 DOI: 10.1016/j.biopha.2023.115750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Dual therapy (DT) has shown comparable results to triple therapy (TT) in efficacy and other immunological aspects. However, there are still some concerns about DT, including several immunological features. Therefore, we evaluated whether HIV-1-specific memory T-cell responses and exhaustion phenotypes are adversely influenced after simplification to DT. METHODS HIV-1-specific CD4+ and CD8+ T-cell responses were assessed by intracellular cytokine and degranulation marker staining, and polyfunctionality indexes after stimulation with a Gag peptide pool. Exhaustion phenotypes were evaluated by PD-1, TIM-3, and LAG-3 expression in CD4+ and CD8+ T cells. RESULTS Forty participants in the TRIDUAL trial (ClinicalTrials.gov: NCT03447873) who were randomized to continue integrase inhibitor-based TT (n = 20) or to switch to DT (dolutegravir or darunavir/cobicistat plus lamivudine) (n = 20). After 96 weeks, the magnitude of CD4+ and CD8+ T-cell responses was similar in both treatment arms (p = 0.221 and p = 0.602, respectively). The CD4+ polyfunctionality index decreased in the TT arm (p = 0.013) and remained stable in the DT arm, while the polyfunctionality of CD8+ T cells was unchanged in both arms. There was a significant decrease in the expression of PD-1, TIM-3, and the co-expression of PD-1+TIM-3+LAG-3+, and PD-1 +TIM-3 + in both CD4+ and CD8+ T cells. However, the decrease in the expression of exhaustion markers did not improve HIV-1-specific T-cell responses. CONCLUSIONS Our results suggest that simplification to DT does not negatively influence the HIV-1-specific T-cell response or the exhaustion phenotype after 96 weeks of follow-up.
Collapse
Affiliation(s)
- Esperanza Muñoz-Muela
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - María Trujillo-Rodríguez
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Ana Serna-Gallego
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Abraham Saborido-Alconchel
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Ezequiel Ruiz-Mateos
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Luis F López-Cortés
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain.
| | - Alicia Gutiérrez-Valencia
- Enfermedades Infecciosas, Microbiología y Parasitología, Instituto de Biomedicina de Sevilla/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| |
Collapse
|
4
|
Xie L, Fang J, Yu J, Zhang W, He Z, Ye L, Wang H. The role of CD4 + T cells in tumor and chronic viral immune responses. MedComm (Beijing) 2023; 4:e390. [PMID: 37829505 PMCID: PMC10565399 DOI: 10.1002/mco2.390] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Immunotherapies are mainly aimed to promote a CD8+ T cell response rather than a CD4+ T cell response as cytotoxic T lymphocytes (CTLs) can directly kill target cells. Recently, CD4+ T cells have received more attention due to their diverse roles in tumors and chronic viral infections. In antitumor and antichronic viral responses, CD4+ T cells relay help signals through dendritic cells to indirectly regulate CD8+ T cell response, interact with B cells or macrophages to indirectly modulate humoral immunity or macrophage polarization, and inhibit tumor blood vessel formation. Additionally, CD4+ T cells can also exhibit direct cytotoxicity toward target cells. However, regulatory T cells exhibit immunosuppression and CD4+ T cells become exhausted, which promote tumor progression and chronic viral persistence. Finally, we also outline immunotherapies based on CD4+ T cells, including adoptive cell transfer, vaccines, and immune checkpoint blockade. Overall, this review summarizes diverse roles of CD4+ T cells in the antitumor or protumor and chronic viral responses, and also highlights the immunotherapies based on CD4+ T cells, giving a better understanding of their roles in tumors and chronic viral infections.
Collapse
Affiliation(s)
- Luoyingzi Xie
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Jingyi Fang
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Juncheng Yu
- Department of Thoracic SurgeryXinqiao Hospital Third Military Medical University (Army Medical University)ChongqingChina
| | - Weinan Zhang
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Zhiqiang He
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Lilin Ye
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
| |
Collapse
|
5
|
Jimenez-Leon MR, Gasca-Capote C, Tarancon-Diez L, Dominguez-Molina B, Lopez-Verdugo M, Ritraj R, Gallego I, Alvarez-Rios AI, Vitalle J, Bachiller S, Camacho-Sojo MI, Perez-Gomez A, Espinosa N, Roca-Oporto C, Rafii-El-Idrissi Benhnia M, Gutierrez-Valencia A, Lopez-Cortes LF, Ruiz-Mateos E. Toll-like receptor agonists enhance HIV-specific T cell response mediated by plasmacytoid dendritic cells in diverse HIV-1 disease progression phenotypes. EBioMedicine 2023; 91:104549. [PMID: 37018973 PMCID: PMC10106920 DOI: 10.1016/j.ebiom.2023.104549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Plasmacytoid dendritic cells (pDCs) sense viral and bacterial products through Toll-like receptor (TLR)-7 and -9 and translate this sensing into Interferon-α (IFN-α) production and T-cell activation. The understanding of the mechanisms involved in pDCs stimulation may contribute to HIV-cure immunotherapeutic strategies. The objective of the present study was to characterize the immunomodulatory effects of TLR agonist stimulations in several HIV-1 disease progression phenotypes and in non HIV-1 infected donors. METHODS pDCs, CD4 and CD8 T-cells were isolated from 450 ml of whole blood from non HIV-1 infected donors, immune responders (IR), immune non responders (INR), viremic (VIR) and elite controller (EC) participants. pDCs were stimulated overnight with AT-2, CpG-A, CpG-C and GS-9620 or no stimuli. After that, pDCs were co-cultured with autologous CD4 or CD8 T-cells and with/without HIV-1 (Gag peptide pool) or SEB (Staphylococcal Enterotoxin B). Cytokine array, gene expression and deep immunophenotyping were assayed. FINDINGS pDCs showed an increase of activation markers levels, interferon related genes, HIV-1 restriction factors and cytokines levels after TLR stimulation in the different HIV-disease progression phenotypes. This pDC activation was prominent with CpG-C and GS-9620 and induced an increase of HIV-specific T-cell response even in VIR and INR comparable with EC. This HIV-1 specific T-cell response was associated with the upregulation of HIV-1 restriction factors and IFN-α production by pDC. INTERPRETATION These results shed light on the mechanisms associated with TLR-specific pDCs stimulation associated with the induction of a T-cell mediated antiviral response which is essential for HIV-1 eradication strategies. FUNDING This work was supported by Gilead fellowship program, the Instituto de Salud Carlos III (Fondo Europeo de Desarrollo Regional, FEDER, "a way to make Europe") and the Red Temática de Investigación Cooperativa en SIDA and by the Spanish National Research Council (CSIC).
Collapse
|
6
|
Arnolds KL, Yamada E, Neff CP, Schneider JM, Palmer BE, Lozupone CA. Disruption of Genes Encoding Putative Zwitterionic Capsular Polysaccharides of Diverse Intestinal Bacteroides Reduces the Induction of Host Anti-Inflammatory Factors. MICROBIAL ECOLOGY 2023; 85:1620-1629. [PMID: 35596750 PMCID: PMC10167101 DOI: 10.1007/s00248-022-02037-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/08/2022] [Indexed: 05/10/2023]
Abstract
Bacterial zwitterionic capsular polysaccharides (ZPS), such as polysaccharide A (PSA) of the intestinal commensal Bacteroides fragilis, have been shown to modulate T cells, including inducing anti-inflammatory IL-10-secreting T regulatory cells (Tregs). We previously used a genomic screen to identify diverse host-associated bacteria with the predicted genetic capacity to produce ZPSs related to PSA of B. fragilis and hypothesized that genetic disruption (KO) of a key functional gene within these operons would reduce the anti-inflammatory activity of these bacteria. We found that ZPS-KO bacteria in two common gut commensals, Bacteroides uniformis and Bacteroides cellulosilyticus, had a reduced ability to induce Tregs and IL-10 in stimulations of human peripheral blood mononuclear cells (PBMCs). Additionally, we found that macrophage stimulated with either wildtype B. fragilis or B. uniformis produced significantly more IL-10 than KOs, indicating a potentially novel function of ZPS of shifting the cytokine response in macrophages to a more anti-inflammatory state. These findings support the hypothesis that these related ZPS may represent a shared strategy to modulate host immune responses.
Collapse
Affiliation(s)
- Kathleen L Arnolds
- Department of Immunology and Microbiology, University of Colorado Anschutz, Aurora, CO, USA
| | - Eiko Yamada
- Department of Medicine, University of Colorado Anschutz, Aurora, CO, USA
| | - C Preston Neff
- Department of Medicine, University of Colorado Anschutz, Aurora, CO, USA
| | | | - Brent E Palmer
- Department of Medicine, University of Colorado Anschutz, Aurora, CO, USA
| | | |
Collapse
|
7
|
Brunet-Ratnasingham E, Morou A, Dubé M, Niessl J, Baxter AE, Tastet O, Brassard N, Ortega-Delgado G, Charlebois R, Freeman GJ, Tremblay C, Routy JP, Kaufmann DE. Immune checkpoint expression on HIV-specific CD4+ T cells and response to their blockade are dependent on lineage and function. EBioMedicine 2022; 84:104254. [PMID: 36150362 PMCID: PMC9508408 DOI: 10.1016/j.ebiom.2022.104254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 11/29/2022] Open
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
Collapse
Affiliation(s)
- Elsa Brunet-Ratnasingham
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Université de Montréal, Montreal, Quebec, Canada
| | - Antigoni Morou
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Université de Montréal, Montreal, Quebec, Canada
| | - Mathieu Dubé
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Julia Niessl
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Université de Montréal, Montreal, Quebec, Canada
| | - Amy E Baxter
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Université de Montréal, Montreal, Quebec, Canada
| | - Olivier Tastet
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Nathalie Brassard
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Gloria Ortega-Delgado
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Roxanne Charlebois
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Cécile Tremblay
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illnesses Service and Division of Hematology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Daniel E Kaufmann
- Research Centre of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
8
|
Tessema B, Boldt A, König B, Maier M, Sack U. Flow-Cytometry Intracellular Detection and Quantification of HIV1 p24 Antigen and Immunocheckpoint Molecules in T Cells among HIV/AIDS Patients. HIV AIDS (Auckl) 2022; 14:365-379. [PMID: 35958525 PMCID: PMC9359413 DOI: 10.2147/hiv.s374369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Belay Tessema
- Department of Medical Microbiology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Institute of Medical Microbiology and Virology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Correspondence: Belay Tessema, Department of Medical Microbiology, College of Medicine and Health Sciences, University of Gondar, 196, Gondar, Ethiopia, Tel +251-91-930-6918, Email
| | - Andreas Boldt
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Brigitte König
- Institute of Medical Microbiology and Virology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Melanie Maier
- Department of Virology, Institute of Medical Microbiology and Virology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Ulrich Sack
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
9
|
Expression Profile and Biological Role of Immune Checkpoints in Disease Progression of HIV/SIV Infection. Viruses 2022; 14:v14030581. [PMID: 35336991 PMCID: PMC8955100 DOI: 10.3390/v14030581] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
During HIV/SIV infection, the upregulation of immune checkpoint (IC) markers, programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), T cell immunoglobulin and ITIM domain (TIGIT), lymphocyte-activation gene-3 (LAG-3), T cell immunoglobulin and mucin domain-3 (Tim-3), CD160, 2B4 (CD244), and V-domain Ig suppressor of T cell activation (VISTA), can lead to chronic T cell exhaustion. These ICs play predominant roles in regulating the progression of HIV/SIV infection by mediating T cell responses as well as enriching latent viral reservoirs. It has been demonstrated that enhanced expression of ICs on CD4+ and CD8+ T cells could inhibit cell proliferation and cytokine production. Overexpression of ICs on CD4+ T cells could also format and prolong HIV/SIV persistence. IC blockers have shown promising clinical results in HIV therapy, implying that targeting ICs may optimize antiretroviral therapy in the context of HIV suppression. Here, we systematically review the expression profile, biological regulation, and therapeutic efficacy of targeted immune checkpoints in HIV/SIV infection.
Collapse
|
10
|
Lu C, Chen H, Wang C, Yang F, Li J, Liu H, Chen G. An Emerging Role of TIM3 Expression on T Cells in Chronic Kidney Inflammation. Front Immunol 2022; 12:798683. [PMID: 35154075 PMCID: PMC8825483 DOI: 10.3389/fimmu.2021.798683] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/16/2021] [Indexed: 12/29/2022] Open
Abstract
T cell immunoglobulin domain and mucin domain 3 (TIM3) was initially identified as an inhibitory molecule on IFNγ-producing T cells. Further research discovered the broad expression of TIM3 on different immune cells binding to multiple ligands. Apart from its suppressive effects on the Th1 cells, recent compelling experiments highlighted the indispensable role of TIM3 in the myeloid cell-mediated inflammatory response, supporting that TIM3 exerts pleiotropic effects on both adaptive and innate immune cells in a context-dependent manner. A large number of studies have been conducted on TIM3 biology in the disease settings of infection, cancer, and autoimmunity. However, there is a lack of clinical evidence to closely evaluate the role of T cell-expressing TIM3 in the pathogenesis of chronic kidney disease (CKD). Here, we reported an intriguing case of Mycobacterium tuberculosis (Mtb) infection that was characterized by persistent overexpression of TIM3 on circulating T cells and ongoing kidney tubulointerstitial inflammation for a period of 12 months. In this case, multiple histopathological biopsies revealed a massive accumulation of recruited T cells and macrophages in the enlarged kidney and liver. After standard anti-Mtb treatment, repeated renal biopsy identified a dramatic remission of the infiltrated immune cells in the tubulointerstitial compartment. This is the first clinical report to reveal a time-course expression of TIM3 on the T cells, which is pathologically associated with the progression of severe kidney inflammation in a non-autoimmunity setting. Based on this case, we summarize the recent findings on TIM3 biology and propose a novel model of CKD progression due to the aberrant crosstalk among immune cells.
Collapse
Affiliation(s)
- Can Lu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huihui Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Chang Wang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Fei Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Jun Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| |
Collapse
|
11
|
Chiu CY, Chang JJ, Dantanarayana AI, Soloman A, Evans VA, Pascoe R, Gubser C, Trautman L, Fromentin R, Chomont N, McMahon JH, Cameron PU, Rasmussen TA, Lewin SR. Combination Immune Checkpoint Blockade Enhances IL-2 and CD107a Production from HIV-Specific T Cells Ex Vivo in People Living with HIV on Antiretroviral Therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:54-62. [PMID: 34853078 PMCID: PMC8702486 DOI: 10.4049/jimmunol.2100367] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/13/2021] [Indexed: 01/03/2023]
Abstract
In people with HIV (PWH) on antiretroviral therapy (ART), immune dysfunction persists, including elevated expression of immune checkpoint (IC) proteins on total and HIV-specific T cells. Reversing immune exhaustion is one strategy to enhance the elimination of HIV-infected cells that persist in PWH on ART. We aimed to evaluate whether blocking CTL-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), T cell Ig domain and mucin domain 3 (TIM-3), T cell Ig and ITIM domain (TIGIT) and lymphocyte activation gene-3 (LAG-3) alone or in combination would enhance HIV-specific CD4+ and CD8+ T cell function ex vivo. Intracellular cytokine staining was performed using human PBMCs from PWH on ART (n = 11) and expression of CD107a, IFN-γ, TNF-α, and IL-2 was quantified with HIV peptides and Abs to IC. We found the following: 1) IC blockade enhanced the induction of CD107a and IL-2 but not IFN-γ and TNF-α in response to Gag and Nef peptides; 2) the induction of CD107a and IL-2 was greatest with multiple combinations of two Abs; and 3) Abs to LAG-3, CTLA-4, and TIGIT in combinations showed synergistic induction of IL-2 in HIV-specific CD8+ and CD107a and IL-2 production in HIV-specific CD4+ and CD8+ T cells. These results demonstrate that the combination of Abs to LAG-3, CTLA-4, or TIGIT can increase the frequency of cells expressing CD107a and IL-2 that associated with cytotoxicity and survival of HIV-specific CD4+ and CD8+ T cells in PWH on ART. These combinations should be further explored for an HIV cure.
Collapse
Affiliation(s)
- Chris Y. Chiu
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Judy J. Chang
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Ashanti I. Dantanarayana
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Ajantha Soloman
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Vanessa A. Evans
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Rachel Pascoe
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Céline Gubser
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Lydie Trautman
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Rémi Fromentin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec H2X 3E4, Canada
| | - Nicolas Chomont
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec H2X 3E4, Canada;,Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - James H. McMahon
- Department of Infectious Diseases, Monash University and the Alfred Hospital, Melbourne, Victoria 3010, Australia
| | - Paul U. Cameron
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia;,Department of Infectious Diseases, Monash University and the Alfred Hospital, Melbourne, Victoria 3010, Australia
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia;,Department of Infectious Diseases, Monash University and the Alfred Hospital, Melbourne, Victoria 3010, Australia;,Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000
| |
Collapse
|
12
|
Premeaux TA, Moser CB, McKhann A, Hoenigl M, Laws EI, Aquino DL, Lederman MM, Landay AL, Gianella S, Ndhlovu LC. Plasma galectin-9 as a predictor of adverse non-AIDS events in persons with chronic HIV during suppressive antiretroviral therapy. AIDS 2021; 35:2489-2495. [PMID: 34366381 PMCID: PMC8631144 DOI: 10.1097/qad.0000000000003048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND People with HIV (PWH) on antiretroviral therapy (ART) still experience an increased risk of morbidity and mortality, presumably driven by chronic inflammation, yet predictors of discrete or combinatorial outcomes remain unclear. Galectin-9 (Gal-9), a driver of both inflammatory and immunosuppressive responses, has been associated with HIV disease progression and multimorbidity. OBJECTIVE To determine whether plasma Gal-9 levels are associated with the occurrence of specific non-AIDS events (NAEs) in PWH initiating ART. DESIGN We performed a nested case-control study of PWH enrolled from 2001 to 2009 and evaluated pre-ART (66 cases, 97 controls), a year post-ART (112 cases, 211 controls), and immediately preceding an event (89 cases, 162 controls). Events included myocardial infarction/stroke, malignancy, serious bacterial infection, or death. METHODS Plasma Gal-9 levels were assessed by ELISA. Conditional logistic regression assessed associations with NAEs and Spearman's correlations compared Gal-9 with other previously assessed biomarkers. RESULTS NAEs occurred at a median of 2.8 years (1.7-4.6) after ART initiation. Higher Gal-9 levels were associated with increased risk of NAEs at year 1 and preevent [odds ratio (OR) per 1 interquartile range = 1.4-1.6; all P < 0.05], specifically myocardial infarction/stroke at year 1 (OR = 1.9; P = 0.029). Gal-9 also correlated with multiple inflammatory and immune activation predictors of NAEs (all timepoints). CONCLUSION Elevated Gal-9 levels are predictive of deleterious NAEs, particularly cardiovascular complications. Whether the Gal-9 pathway, potentially binding to its putative ligands, is active in the pathogenesis of these outcomes warrants further investigation to determine if targeting Gal-9 may slow or reverse the risk of NAEs.
Collapse
Affiliation(s)
- Thomas A. Premeaux
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Carlee B. Moser
- Center for Biostatistics in AIDS Research in the Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Ashley McKhann
- Center for Biostatistics in AIDS Research in the Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Martin Hoenigl
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA
| | - Elizabeth I. Laws
- Department of Cell and Molecular Biology, University of Hawaii, Honolulu, HI
| | - Draven L. Aquino
- Department of Cell and Molecular Biology, University of Hawaii, Honolulu, HI
| | - Michael M. Lederman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
13
|
Wang D, Jiang Y, Song Y, Zeng Y, Li C, Wang X, Liu Y, Xiao J, Kong Y, Zhao H. Altered T-Cell Subsets are Associated with Dysregulated Cytokine Secretion of CD4 + T Cells During HIV Infection. J Inflamm Res 2021; 14:5149-5163. [PMID: 34675594 PMCID: PMC8504938 DOI: 10.2147/jir.s333902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/27/2021] [Indexed: 11/23/2022] Open
Abstract
Background CD4+ T cells play a critical role in the regulation of immunopathogenesis in HIV infection. Previous studies have shown contradictory results of the CD4+ T-cell responses in people living with HIV (PLHIV). Methods A cross-sectional study was performed on 40 healthy controls, 134 ART-naïve PLHIV, and 34 individuals who experienced 3-year ART with low baseline CD4 count from 4 August 2016 to 23 January 2019. We determined the frequencies of CD4+ T-cell subsets and described the cytokine secretion pattern of total and subsets of CD4+ T cells in these individuals. Results We found that CD4+ T cells in PLHIV displayed enhanced secretion of pro-inflammation cytokines and polyfunctionality due to HIV disease progression (r = -0.282, P = 0.0035 for IFN-γ; r = -0.412, P = 0.0002 for TNF-α; r = -0.243, P < 0.0001 for GM-CSF; r = -0.252, P = 0.0093 for IFN-γ+ TNF-α+ cells). However, the altered T-cell subsets, as presented by the loss of naïve cells and expansion of memory/effector population in PLHIV, were associated with discordant results in total and subsets of CD4+ T cells. As major cytokine-producing T subsets, effector/memory CD4 subsets showed impaired cytokine production (P < 0.05). We further demonstrated that 3-year ART treatment could improve CD4 counts by increasing the pool of naïve T cells but could not restore cytokine secretion in CD4+ T-cell subsets (P < 0.05). Conclusion These data identified the impaired capacity of cytokine secretion in CD4+ T-cell subsets due to HIV disease progression, and the altered T-cell subsets were associated with pseudo-elevation of cytokine production in total CD4+ T cells. This study collectively suggested the importance of therapies that can preserve and/or enhance the function of CD4+ T cells in strategies of HIV remission.
Collapse
Affiliation(s)
- Di Wang
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yu Jiang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yangzi Song
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yongqin Zeng
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Cuilin Li
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xinyue Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ying Liu
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jiang Xiao
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yaxian Kong
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hongxin Zhao
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
14
|
Jacob RA, Edgar CR, Prévost J, Trothen SM, Lurie A, Mumby MJ, Galbraith A, Kirchhoff F, Haeryfar SMM, Finzi A, Dikeakos JD. The HIV-1 accessory protein Nef increases surface expression of the checkpoint receptor Tim-3 in infected CD4 + T cells. J Biol Chem 2021; 297:101042. [PMID: 34358561 PMCID: PMC8390549 DOI: 10.1016/j.jbc.2021.101042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Prolonged immune activation drives the upregulation of multiple checkpoint receptors on the surface of virus-specific T cells, inducing their exhaustion. Reversing HIV-1-induced T cell exhaustion is imperative for efficient virus clearance; however, viral mediators of checkpoint receptor upregulation remain largely unknown. The enrichment of checkpoint receptors on T cells upon HIV-1 infection severely constrains the generation of an efficient immune response. Herein, we examined the role of HIV-1 Nef in mediating the upregulation of checkpoint receptors on peripheral blood mononuclear cells. We demonstrate that the HIV-1 accessory protein Nef upregulates cell surface levels of the checkpoint receptor T-cell immunoglobulin mucin domain-3 (Tim-3) and that this is dependent on Nef's dileucine motif LL164/165. Furthermore, we used a bimolecular fluorescence complementation assay to demonstrate that Nef and Tim-3 form a complex within cells that is abrogated upon mutation of the Nef dileucine motif. We also provide evidence that Nef moderately promotes Tim-3 shedding from the cell surface in a dileucine motif–dependent manner. Treating HIV-1-infected CD4+ T cells with a matrix metalloprotease inhibitor enhanced cell surface Tim-3 levels and reduced Tim-3 shedding. Finally, Tim-3-expressing CD4+ T cells displayed a higher propensity to release the proinflammatory cytokine interferon-gamma. Collectively, our findings uncover a novel mechanism by which HIV-1 directly increases the levels of a checkpoint receptor on the surface of infected CD4+ T cells.
Collapse
Affiliation(s)
- Rajesh Abraham Jacob
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Cassandra R Edgar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
| | - Steven M Trothen
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Antony Lurie
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Mitchell J Mumby
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Alexa Galbraith
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, UIm, Germany
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
15
|
Kataoka S, Manandhar P, Lee J, Workman CJ, Banerjee H, Szymczak-Workman AL, Kvorjak M, Lohmueller J, Kane LP. The costimulatory activity of Tim-3 requires Akt and MAPK signaling and its recruitment to the immune synapse. Sci Signal 2021; 14:eaba0717. [PMID: 34131021 PMCID: PMC9741863 DOI: 10.1126/scisignal.aba0717] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Expression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic activation, including during chronic infection and in solid tumors. Thus, Tim-3 is generally thought of as an inhibitory protein. We and others previously reported that under some circumstances, Tim-3 exerts paradoxical costimulatory activity in T cells (and other cells), including enhancement of the phosphorylation of ribosomal S6 protein. Here, we examined the upstream signaling pathways that control Tim-3-mediated increases in phosphorylated S6 in T cells. We also defined the localization of Tim-3 relative to the T cell immune synapse and its effects on downstream signaling. Recruitment of Tim-3 to the immune synapse was mediated exclusively by the transmembrane domain, replacement of which impaired the ability of Tim-3 to costimulate T cell receptor (TCR)-dependent S6 phosphorylation. Furthermore, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in a chimeric antigen receptor still enabled T cell activation. Together, our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.
Collapse
Affiliation(s)
- Shunsuke Kataoka
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Asahi Kasei Pharma Corporation, Shizuoka, Japan
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Priyanka Manandhar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Judong Lee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hridesh Banerjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | - Michael Kvorjak
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jason Lohmueller
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
16
|
Pérez-Antón E, Egui A, Thomas MC, Carrilero B, Simón M, López-Ruz MÁ, Segovia M, López MC. A proportion of CD4+ T cells from patients with chronic Chagas disease undergo a dysfunctional process, which is partially reversed by benznidazole treatment. PLoS Negl Trop Dis 2021; 15:e0009059. [PMID: 33539379 PMCID: PMC7888659 DOI: 10.1371/journal.pntd.0009059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 02/17/2021] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Background Signs of senescence and the late stages of differentiation associated with the more severe forms of Chagas disease have been described in the Trypanosoma cruzi antigen-specific CD4+ T-cell population. However, the mechanisms involved in these functions are not fully known. To date, little is known about the possible impact of benznidazole treatment on the T. cruzi-specific functional response of CD4+ T cells. Methodology/Principal findings The functional capacity of CD4+ T cells was analyzed by cytometric assays in chronic Chagas disease patients, with indeterminate form (IND) and cardiac alterations (CCC) (25 and 15, respectively) before and after benznidazole treatment. An increase in the multifunctional capacity (expression of IFN-γ, IL-2, TNF-α, perforin and/or granzyme B) of the antigen-specific CD4+ T cells was observed in indeterminate versus cardiac patients, which was associated with the reduced coexpression of inhibitory receptors (2B4, CD160, CTLA-4, PD-1 and/or TIM-3). The functional profile of these cells shows statistically significant differences between IND and CCC (p<0.001), with a higher proportion of CD4+ T cells coexpressing 2 and 3 molecules in IND (54.4% versus 23.1% and 4.1% versus 2.4%, respectively). A significant decrease in the frequencies of CD4+ T cells that coexpress 2, 3 and 4 inhibitory receptors was observed in IND after 24–48 months of treatment (p<0.05, p<0.01 and p<0.05, respectively), which was associated with an increase in antigen-specific multifunctional activity. The IND group showed, at 9–12 months after treatment, an increase in the CD4+ T cell subset coproducing three molecules, which were mainly granzyme B+, perforin+ and IFN-γ+ (1.4% versus 4.5%). Conclusions/Significance A CD4+ T cell dysfunctional process was detected in chronic Chagas disease patients, being more exacerbated in those patients with cardiac symptoms. After short-term benznidazole treatment (9–12 months), indeterminate patients showed a significant increase in the frequency of multifunctional antigen-specific CD4+ T cells. Trypanosoma cruzi infection triggers several immune mechanisms in the host that do not result in a total clearance of the parasite, the persistence of which leads to the chronicity of Chagas disease. The mechanisms by which some chronic patients remain asymptomatic or become symptomatic are not entirely clear. The aim of the present manuscript is to study the CD4+ T cell population and its functional capacity in patients with different forms of chronic disease. The obtained results indicate that cells from indeterminate patients have an enhanced multifunctional profile, which is associated with the reduced expression of inhibitory molecules. CD4+ T cells from chronic patients with cardiac alterations show lower functional activity against specific antigens of the parasite and increased coexpression of inhibitory molecules. After benznidazole treatment, antigen-specific CD4+ T cells, especially those from indeterminate patients, are more likely to show a multifunctional profile and a decline in the coexpression of inhibitory receptors. These results allow us to make progress in clarifying the mechanisms that may influence disease progression and to realize the importance of antiparasitic treatment for the enhancement of the activity of the immune system.
Collapse
Affiliation(s)
- Elena Pérez-Antón
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
| | - Adriana Egui
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
| | - M. Carmen Thomas
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
| | - Bartolomé Carrilero
- Unidad Regional de Medicina Tropical, Hospital Virgen de la Arrixaca; El Palmar, Murcia, Spain
| | - Marina Simón
- Unidad Regional de Medicina Tropical, Hospital Virgen de la Arrixaca; El Palmar, Murcia, Spain
| | | | - Manuel Segovia
- Unidad Regional de Medicina Tropical, Hospital Virgen de la Arrixaca; El Palmar, Murcia, Spain
| | - Manuel Carlos López
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas; Granada, Spain
- * E-mail:
| |
Collapse
|
17
|
Nandi D, Pathak S, Verma T, Singh M, Chattopadhyay A, Thakur S, Raghavan A, Gokhroo A, Vijayamahantesh. T cell costimulation, checkpoint inhibitors and anti-tumor therapy. J Biosci 2021. [PMID: 32345776 DOI: 10.1007/s12038-020-0020-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The hallmarks of the adaptive immune response are specificity and memory. The cellular response is mediated by T cells which express cell surface T cell receptors (TCRs) that recognize peptide antigens in complex with major histocompatibility complex (MHC) molecules on antigen presenting cells (APCs). However, binding of cognate TCRs with MHC-peptide complexes alone (signal 1) does not trigger optimal T cell activation. In addition to signal 1, the binding of positive and negative costimulatory receptors to their ligands modulates T cell activation. This complex signaling network prevents aberrant activation of T cells. CD28 is the main positive costimulatory receptor on naı¨ve T cells; upon activation, CTLA4 is induced but reduces T cell activation. Further studies led to the identification of additional negative costimulatory receptors known as checkpoints, e.g. PD1. This review chronicles the basic studies in T cell costimulation that led to the discovery of checkpoint inhibitors, i.e. antibodies to negative costimulatory receptors (e.g. CTLA4 and PD1) which reduce tumor growth. This discovery has been recognized with the award of the 2018 Nobel prize in Physiology/Medicine. This review highlights the structural and functional roles of costimulatory receptors, the mechanisms by which checkpoint inhibitors work, the challenges encountered and future prospects.
Collapse
Affiliation(s)
- Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru 560 012, India
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Collora JA, Liu R, Albrecht K, Ho YC. The single-cell landscape of immunological responses of CD4+ T cells in HIV versus severe acute respiratory syndrome coronavirus 2. Curr Opin HIV AIDS 2021; 16:36-47. [PMID: 33165008 PMCID: PMC8162470 DOI: 10.1097/coh.0000000000000655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW CD4 T cell loss is the hallmark of uncontrolled HIV-1 infection. Strikingly, CD4 T cell depletion is a strong indicator for disease severity in the recently emerged coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We reviewed recent single-cell immune profiling studies in HIV-1 infection and COVID-19 to provide critical insight in virus-induced immunopathogenesis. RECENT FINDINGS Cytokine dysregulation in HIV-1 leads to chronic inflammation, while severe SARS-CoV-2 infection induces cytokine release syndrome and increased mortality. HIV-1-specific CD4 T cells are dysfunctional, while SARS-CoV-2-specific CD4 T cells exhibit robust Th1 function and correlate with protective antibody responses. In HIV-1 infection, follicular helper T cells (TFH) are susceptible to HIV-1 infection and persist in immune-sanctuary sites in lymphoid tissues as an HIV-1 reservoir. In severe SARS-CoV-2 infection, TFH are absent in lymphoid tissues and are associated with diminished protective immunity. Advancement in HIV-1 DNA, RNA, and protein-based single-cell capture methods can overcome the rarity and heterogeneity of HIV-1-infected cells and identify mechanisms of HIV-1 persistence and clonal expansion dynamics. SUMMARY Single-cell immune profiling identifies a high-resolution picture of immune dysregulation in HIV-1 and SARS-CoV-2 infection and informs outcome prediction and therapeutic interventions.
Collapse
Affiliation(s)
- Jack A Collora
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | |
Collapse
|
19
|
Chen H, Moussa M, Catalfamo M. The Role of Immunomodulatory Receptors in the Pathogenesis of HIV Infection: A Therapeutic Opportunity for HIV Cure? Front Immunol 2020; 11:1223. [PMID: 32714317 PMCID: PMC7343933 DOI: 10.3389/fimmu.2020.01223] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Immune activation is the hallmark of HIV infection and plays a role in the pathogenesis of the disease. In the context of suppressed HIV RNA replication by combination antiretroviral therapy (cART), there remains immune activation which is associated to the HIV reservoirs. Persistent virus contributes to a sustained inflammatory environment promoting accumulation of "activated/exhausted" T cells with diminished effector function. These T cells show increased expression of immunomodulatory receptors including Programmed cell death protein (PD1), Cytotoxic T Lymphocyte Associated Protein 4 (CTLA4), Lymphocyte activation gene 3 (LAG3), T cell immunoglobulin and ITIM domain (TIGIT), T cell immunoglobulin and mucin domain containing 3 (TIM3) among others. More importantly, recent reports had demonstrated that, HIV infected T cells express checkpoint receptors, contributing to their survival and promoting maintenance of the viral reservoir. Therapeutic strategies are focused on viral reservoir elimination and/or those to achieve sustained cART-free virologic remission. In this review, we will discuss the immunological basis and the latest advances of the use of checkpoint inhibitors to treat HIV infection.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
- CMRS/Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Maha Moussa
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
| |
Collapse
|
20
|
Fenwick C, Joo V, Jacquier P, Noto A, Banga R, Perreau M, Pantaleo G. T-cell exhaustion in HIV infection. Immunol Rev 2020; 292:149-163. [PMID: 31883174 PMCID: PMC7003858 DOI: 10.1111/imr.12823] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022]
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.
Collapse
Affiliation(s)
- Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Patricia Jacquier
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Riddhima Banga
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
21
|
Tim-3: A co-receptor with diverse roles in T cell exhaustion and tolerance. Semin Immunol 2020; 42:101302. [PMID: 31604535 DOI: 10.1016/j.smim.2019.101302] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022]
Abstract
T cell inhibitory co-receptors play a crucial role in maintaining the balance between physiologic immune responses and maladaptive ones. T cell immunoglobulin and mucin domain-containing-3 (Tim-3) is a unique inhibitory co-receptor in that its expression is chiefly restricted to interferon (IFN)γ-producing CD4+ and CD8+ T cells. Early reports firmly established its importance in maintaining peripheral tolerance in transplantation and autoimmunity. However, it has become increasingly clear that Tim-3 expression on T cells, together with other check-point molecules, in chronic infections and cancers can hinder productive immune responses. In this review, we outline what is currently known about the regulation of Tim-3 expression, its ligands and signaling. We discuss both its salutary and deleterious function in immune disorders, as well as the T cell-extrinsic and -intrinsic factors that regulate its function.
Collapse
|
22
|
Zhao L, Yu G, Han Q, Cui C, Zhang B. TIM-3: An emerging target in the liver diseases. Scand J Immunol 2020; 91:e12825. [PMID: 31486085 DOI: 10.1111/sji.12825] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/27/2019] [Accepted: 08/31/2019] [Indexed: 12/17/2022]
Abstract
T cell immunoglobulin domain and mucin domain-containing molecule 3 (TIM-3) is found expression in the surface of terminally differentiated T cells and belongs to the TIM family of type Ⅰ transmembrane proteins. It binds to the ligand Galectin-9 and mediates T cell apoptosis. As the research progresses, TIM-3 is also expressed in Th17, NK, monocyte, which binds to ligand and induce immune peripheral tolerance in both mice and man. Numerous researches have demonstrated that TIM-3 influences liver diseases, including liver-associated chronic viral infection, liver fibrosis, liver cancer et al and suggest new approaches to intervention. Currently, targeted therapy of TIM-3 is a new treatment in the field of immunization. Although many studies have proven that TIM-3 has an inhibitory effect in vivo, the specific mechanism is not clear. Herein, we summarize the important role of TIM-3 in the regulation of liver disease and prospects for future clinical research. TIM-3 will provide new targets for improving clinical liver disease.
Collapse
Affiliation(s)
- Lizhen Zhao
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| | - Guoyi Yu
- Editorial Office of Journal of Qingdao University (Medical Science), Qingdao, China
| | - Qi Han
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| | - Congxian Cui
- Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Bei Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Abstract
A disease of more than 39.6 million people worldwide, HIV-1 infection has no curative therapy. To date, one man has achieved a sterile cure, with millions more hoping to avoid the potential pitfalls of lifelong antiretroviral therapy and other HIV-related disorders, including neurocognitive decline. Recent developments in immunotherapies and gene therapies provide renewed hope in advancing efforts toward a sterilizing or functional cure. On the horizon is research concentrated in multiple separate but potentially complementary domains: vaccine research, viral transcript editing, T-cell effector response targeting including checkpoint inhibitors, and gene editing. Here, we review the concept of targeting the HIV-1 tissue reservoirs, with an emphasis on the central nervous system, and describe relevant new work in functional cure research and strategies for HIV-1 eradication.
Collapse
|
24
|
Liu R, Simonetti FR, Ho YC. The forces driving clonal expansion of the HIV-1 latent reservoir. Virol J 2020; 17:4. [PMID: 31910871 PMCID: PMC6947923 DOI: 10.1186/s12985-019-1276-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
Despite antiretroviral therapy (ART) which halts HIV-1 replication and reduces plasma viral load to clinically undetectable levels, viral rebound inevitably occurs once ART is interrupted. HIV-1-infected cells can undergo clonal expansion, and these clonally expanded cells increase over time. Over 50% of latent reservoirs are maintained through clonal expansion. The clonally expanding HIV-1-infected cells, both in the blood and in the lymphoid tissues, contribute to viral rebound. The major drivers of clonal expansion of HIV-1-infected cells include antigen-driven proliferation, homeostatic proliferation and HIV-1 integration site-dependent proliferation. Here, we reviewed how viral, immunologic and genomic factors contribute to clonal expansion of HIV-1-infected cells, and how clonal expansion shapes the HIV-1 latent reservoir. Antigen-specific CD4+ T cells specific for different pathogens have different clonal expansion dynamics, depending on antigen exposure, cytokine profiles and exhaustion phenotypes. Homeostatic proliferation replenishes the HIV-1 latent reservoir without inducing viral expression and immune clearance. Integration site-dependent proliferation, a mechanism also deployed by other retroviruses, leads to slow but steady increase of HIV-1-infected cells harboring HIV-1 proviruses integrated in the same orientation at specific sites of certain cancer-related genes. Targeting clonally expanding HIV-1 latent reservoir without disrupting CD4+ T cell function is a top priority for HIV-1 eradication.
Collapse
Affiliation(s)
- Runxia Liu
- Department of Microbial Pathogenesis, Yale University, New Haven, CT, 06519, USA
| | | | - Ya-Chi Ho
- Department of Microbial Pathogenesis, Yale University, New Haven, CT, 06519, USA.
| |
Collapse
|
25
|
Treating cancer with immunotherapy in HIV-positive patients: A challenging reality. Crit Rev Oncol Hematol 2019; 145:102836. [PMID: 31918216 DOI: 10.1016/j.critrevonc.2019.102836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/24/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023] Open
Abstract
Immunotherapy has widely changed the management of different malignancies. However, efficacy and safety of immune checkpoint inhibitors (ICIs) are not well established in people living with HIV (PLWH). Population of HIV-positive patients has deeply changed after the introduction of modern antiretroviral therapy (ART) and available data of immunotherapy in this subgroup are inadequate considering that cancer has become a leading cause of death and morbidity in this population. Moreover, there are many similarities between cancer and infectious antigen stimulation so that ICIs are even under evaluation as specific HIV treatment. Most of literature on this topic is based on small case series that suggest that immunotherapy for PLWH seems to be as effective as in HIV-negative population with a good safety profile. In this article we review literature on HIV and immunotherapy and we collect many case series available in different malignancies, with a brief focus on lung cancer.
Collapse
|
26
|
Puertas MC, Gómez-Mora E, Santos JR, Moltó J, Urrea V, Morón-López S, Hernández-Rodríguez A, Marfil S, Martínez-Bonet M, Matas L, Muñoz-Fernández MA, Clotet B, Blanco J, Martinez-Picado J. Impact of intensification with raltegravir on HIV-1-infected individuals receiving monotherapy with boosted PIs. J Antimicrob Chemother 2019; 73:1940-1948. [PMID: 29635527 PMCID: PMC6005067 DOI: 10.1093/jac/dky106] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/05/2018] [Indexed: 01/09/2023] Open
Abstract
Background Monotherapy with ritonavir-boosted PIs (PI/r) has been used to simplify treatment of HIV-1-infected patients. In previous studies raltegravir intensification evidenced ongoing viral replication and reduced T cell activation, preferentially in subjects receiving PI-based triple ART. However, data about low-level viral replication and its consequences in patients receiving PI/r monotherapy are scarce. Methods We evaluated the impact of 24 weeks of intensification with raltegravir on markers of viral persistence, cellular immune activation and inflammation biomarkers in 33 patients receiving maintenance PI/r monotherapy with darunavir or lopinavir boosted with ritonavir. ClinicalTrials.gov identifier: NCT01480713. Results The addition of raltegravir to PI/r monotherapy resulted in a transient increase in 2-LTR (long-terminal repeat) circles in a significant proportion of participants, along with decreases in CD8+ T cell activation levels and a temporary increase in the expression of the exhaustion marker CTLA-4 in peripheral T lymphocytes. Intensification with raltegravir also reduced the number of samples with intermediate levels of residual viraemia (10–60 HIV-1 RNA copies/mL) compared with samples taken during PI/r monotherapy. However, there were no changes in cell-associated HIV-1 DNA in peripheral CD4+ T cells or soluble inflammatory biomarkers (CD14, IP-10, IL-6, C-reactive protein and D-dimer). Conclusions Intensification of PI/r monotherapy with raltegravir revealed persistent low-level viral replication and reduced residual viraemia in some patients during long-term PI/r monotherapy. The concomitant change in T cell phenotype suggests an association between active viral production and T cell activation. These results contribute to understanding the lower efficacy rates of PI/r monotherapies compared with triple therapies in clinical trials.
Collapse
Affiliation(s)
- Maria C Puertas
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Elisabet Gómez-Mora
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - José R Santos
- 'Lluita Contra la Sida' Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - José Moltó
- 'Lluita Contra la Sida' Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Víctor Urrea
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Sara Morón-López
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | | | - Silvia Marfil
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immuno Molecular Biology, Section of Immunology, Hospital General Universitario Gregorio Marañon, IiSGM, Madrid, Spain.,Spanish HIV HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Lurdes Matas
- Servicio de Microbiologia, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mª Angeles Muñoz-Fernández
- Laboratory of Immuno Molecular Biology, Section of Immunology, Hospital General Universitario Gregorio Marañon, IiSGM, Madrid, Spain.,Spanish HIV HGM BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Bonaventura Clotet
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,'Lluita Contra la Sida' Foundation, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Universitat de Vic - Universitat Central de Catalunya (UVic-UCC), Vic, Spain
| | - Julià Blanco
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,Universitat de Vic - Universitat Central de Catalunya (UVic-UCC), Vic, Spain
| | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain.,Universitat de Vic - Universitat Central de Catalunya (UVic-UCC), Vic, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
27
|
Barham MS, Abrahams DA, Khayumbi J, Ongalo J, Tonui J, Campbell A, de Kock M, Ouma SG, Odhiambo FH, Hanekom WA, Gandhi NR, Day CL. HIV Infection Is Associated With Downregulation of BTLA Expression on Mycobacterium tuberculosis-Specific CD4 T Cells in Active Tuberculosis Disease. Front Immunol 2019; 10:1983. [PMID: 31497018 PMCID: PMC6712065 DOI: 10.3389/fimmu.2019.01983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Nearly a quarter of the global population is infected with Mycobacterium tuberculosis (Mtb), with 10 million people developing active tuberculosis (TB) annually. Co-infection with human immunodeficiency virus (HIV) has long been recognized as a significant risk factor for progression to TB disease, yet the mechanisms whereby HIV impairs T cell-mediated control of Mtb infection remain poorly defined. We hypothesized that HIV infection may promote upregulation of inhibitory receptors on Mtb-specific CD4 T cells, a mechanism that has been associated with antigen-specific T cell dysfunction in chronic infections. Using cohorts of HIV-infected and HIV-uninfected individuals with latent Mtb infection (LTBI) and with active TB disease, we stimulated peripheral blood mononuclear cells (PBMC) for 6 hours with Mtb peptide pools and evaluated co-expression profiles of the inhibitory receptors BTLA, CTLA-4, and PD-1 on IFN-γ+/TNF-α+ Mtb-specific CD4 T cells. Mtb-specific CD4 T cells in all participant groups expressed predominately either one or no inhibitory receptors, unlike cytomegalovirus- and HIV-specific CD4 T cells circulating in the same individuals, which were predominately CTLA-4+PD-1+. There were no significant differences in inhibitory receptor expression profiles of Mtb-specific CD4 T cells between HIV-uninfected and HIV-infected individuals with LTBI. Surprisingly, BTLA expression, both alone and in combination with CTLA-4 and PD-1, was markedly downregulated on Mtb-specific CD4 T cells in HIV-infected individuals with active TB. Together, these data provide novel evidence that the majority of Mtb-specific CD4 T cells do not co-express multiple inhibitory receptors, regardless of HIV infection status; moreover, they highlight a previously unrecognized role of BTLA expression on Mtb-specific CD4 T cells that could be further explored as a potential biomarker of Mtb infection status, particularly in people living with HIV, the population at greatest risk for development of active TB disease.
Collapse
Affiliation(s)
- Morgan S Barham
- Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Deborah A Abrahams
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jeremiah Khayumbi
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joshua Ongalo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joan Tonui
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Angela Campbell
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Marwou de Kock
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Samuel Gurrion Ouma
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative, School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Neel R Gandhi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Cheryl L Day
- Emory Vaccine Center, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
28
|
Oniszczuk J, Sendeyo K, Chhuon C, Savas B, Cogné E, Vachin P, Henique C, Guerrera IC, Astarita G, Frontera V, Pawlak A, Audard V, Sahali D, Ollero M. CMIP is a negative regulator of T cell signaling. Cell Mol Immunol 2019; 17:1026-1041. [PMID: 31395948 PMCID: PMC7609264 DOI: 10.1038/s41423-019-0266-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 07/10/2019] [Indexed: 11/24/2022] Open
Abstract
Upon their interaction with cognate antigen, T cells integrate different extracellular and intracellular signals involving basal and induced protein–protein interactions, as well as the binding of proteins to lipids, which can lead to either cell activation or inhibition. Here, we show that the selective T cell expression of CMIP, a new adapter protein, by targeted transgenesis drives T cells toward a naïve phenotype. We found that CMIP inhibits activation of the Src kinases Fyn and Lck after CD3/CD28 costimulation and the subsequent localization of Fyn and Lck to LRs. Video microscopy analysis showed that CMIP blocks the recruitment of LAT and the lipid raft marker cholera toxin B at the site of TCR engagement. Proteomic analysis identified several protein clusters differentially modulated by CMIP and, notably, Cofilin-1, which is inactivated in CMIP-expressing T cells. Moreover, transgenic T cells exhibited the downregulation of GM3 synthase, a key enzyme involved in the biosynthesis of gangliosides. These results suggest that CMIP negatively impacts proximal signaling and cytoskeletal rearrangement and defines a new mechanism for the negative regulation of T cells that could be a therapeutic target.
Collapse
Affiliation(s)
- Julie Oniszczuk
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Kelhia Sendeyo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Cerina Chhuon
- Proteomic Platform Necker, PPN-3P5, Structure Fédérative de Recherche SFR Necker US24, 75015, Paris, France
| | - Berkan Savas
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Etienne Cogné
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Pauline Vachin
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Carole Henique
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Ida Chiara Guerrera
- Proteomic Platform Necker, PPN-3P5, Structure Fédérative de Recherche SFR Necker US24, 75015, Paris, France
| | - Giuseppe Astarita
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| | - Vincent Frontera
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Andre Pawlak
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| | - Vincent Audard
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France.,AP-HP, Groupe Henri-Mondor Albert-Chenevier, Service de Néphrologie, F-94010, Créteil, France.,Institut Francilien De Recherche En Néphrologie Et Transplantation, F-94010, Créteil, France
| | - Dil Sahali
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France. .,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France. .,AP-HP, Groupe Henri-Mondor Albert-Chenevier, Service de Néphrologie, F-94010, Créteil, France. .,Institut Francilien De Recherche En Néphrologie Et Transplantation, F-94010, Créteil, France.
| | - Mario Ollero
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 955, Equipe 21, F-94010, Créteil, France.,Faculté de Médecine, Université Paris Est, UMRS 955, Equipe 21, F-94010, Créteil, France
| |
Collapse
|
29
|
Hope JL, Stairiker CJ, Bae EA, Otero DC, Bradley LM. Striking a Balance-Cellular and Molecular Drivers of Memory T Cell Development and Responses to Chronic Stimulation. Front Immunol 2019; 10:1595. [PMID: 31379821 PMCID: PMC6650570 DOI: 10.3389/fimmu.2019.01595] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/26/2019] [Indexed: 01/11/2023] Open
Abstract
Effective adaptive immune responses are characterized by stages of development and maturation of T and B cell populations that respond to disturbances in the host homeostasis in cases of both infections and cancer. For the T cell compartment, this begins with recognition of specific peptides by naïve, antigen-inexperienced T cells that results in their activation, proliferation, and differentiation, which generates an effector population that clears the antigen. Loss of stimulation eventually returns the host to a homeostatic state, with a heterogeneous memory T cell population that persists in the absence of antigen and is primed for rapid responses to a repeat antigen exposure. However, in chronic infections and cancers, continued antigen persistence impedes a successful adaptive immune response and the formation of a stereotypical memory population of T cells is compromised. With repeated antigen stimulation, responding T cells proceed down an altered path of differentiation that allows for antigen persistence, but much less is known regarding the heterogeneity of these cells and the extent to which they can become “memory-like,” with a capacity for self-renewal and recall responses that are characteristic of bona fide memory cells. This review focuses on the differentiation of CD4+ and CD8+ T cells in the context of chronic antigen stimulation, highlighting the central observations in both human and mouse studies regarding the differentiation of memory or “memory-like” T cells. The importance of both the cellular and molecular drivers of memory T cell development are emphasized to better understand the consequences of persisting antigen on T cell fates. Integrating what is known and is common across model systems and patients can instruct future studies aimed at further understanding T cell differentiation and development, with the goal of developing novel methods to direct T cells toward the generation of effective memory populations.
Collapse
Affiliation(s)
- Jennifer L Hope
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Christopher J Stairiker
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Eun-Ah Bae
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Dennis C Otero
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Linda M Bradley
- Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The purpose of the present review is to describe the major barriers to HIV eradication and assess the most promising cure strategies under investigation. RECENT FINDINGS There are significant challenges to achieve HIV eradication. These include the establishment of persistent latently infected cells, systemic chronic immune activation, and immune dysfunction. Since the announcement of the first HIV cure involving the Berlin patient, several attempts to reproduce these results have failed. Thus, it is widely accepted that long-term HIV remission would be a more feasible approach. Optimization of ART, immune-based therapies, therapeutic vaccinations, and gene editing, amongst others, are strategies aimed at controlling HIV in the absence of ART. These new strategies alone or in combination are being developed in preclinical studies and clinical trials and will provide further insight into whether long-term HIV remission is possible. SUMMARY The present review discusses several mechanisms that mediate the persistence of the HIV reservoir, clinical cases that provide hope in finding a functional cure of HIV, and promising interventional strategies being tested in preclinical studies and clinical trials that attempt to reduce the HIV reservoirs and/or boost the immune responses to control HIV in the absence of ART.
Collapse
|
31
|
Abstract
CD8+ T cells are important for the protective immunity against intracellular pathogens and tumor. In the case of chronic infection or cancer, CD8+ T cells are exposed to persistent antigen and/or inflammatory signals. This excessive amount of signals often leads CD8+ T cells to gradual deterioration of T cell function, a state called "exhaustion." Exhausted T cells are characterized by progressive loss of effector functions (cytokine production and killing function), expression of multiple inhibitory receptors (such as PD-1 and LAG3), dysregulated metabolism, poor memory recall response, and homeostatic proliferation. These altered functions are closely related with altered transcriptional program and epigenetic landscape that clearly distinguish exhausted T cells from normal effector and memory T cells. T cell exhaustion is often associated with inefficient control of persisting infections and cancers, but re-invigoration of exhausted T cells with inhibitory receptor blockade can promote improved immunity and disease outcome. Accumulating evidences support the therapeutic potential of targeting exhausted T cells. However, exhausted T cells comprise heterogenous cell population with distinct responsiveness to intervention. Understanding molecular mechanism of T cell exhaustion is essential to establish rational immunotherapeutic interventions.
Collapse
|
32
|
Garcia-Bates TM, Palma ML, Shen C, Gambotto A, Macatangay BJC, Ferris RL, Rinaldo CR, Mailliard RB. Contrasting Roles of the PD-1 Signaling Pathway in Dendritic Cell-Mediated Induction and Regulation of HIV-1-Specific Effector T Cell Functions. J Virol 2019; 93:e02035-18. [PMID: 30541848 PMCID: PMC6384070 DOI: 10.1128/jvi.02035-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022] Open
Abstract
Eliciting highly functional CD8+ cytotoxic T lymphocyte (CTL) responses against a broad range of epitopes will likely be required for immunotherapeutic control of HIV-1 infection. However, the combination of CTL exhaustion and the ability of HIV-1 to rapidly establish CTL escape variants presents major hurdles toward this goal. Our previous work highlighted the use of monocyte-derived, mature, high-interleukin-12 (IL-12)-producing type 1 polarized dendritic cells (MDC1) to selectively induce more potent effector CTLs derived from naive, rather than memory, CD8+ T cell precursors isolated from HIV-1-positive participants in the Multicenter AIDS Cohort Study. In this study, we report that these highly stimulatory antigen-presenting cells also express enhanced levels of the coinhibitory molecule programmed cell death ligand 1 (PD-L1), the ligand for PD-1, which is further upregulated upon subsequent stimulation with the CD4+ T helper cell-derived factor CD40L. Interestingly, blocking the PD-1 signaling pathway during MDC1 induction of HIV-1-specific CTL responses inhibited the priming, activation, and differentiation of naive CD8+ T cells into effector T cells expressing high levels of T-box transcription factor (T-bethi) and eomesodermin (Eomes+). In contrast, PD-1 blockade enhanced the overall magnitude of memory HIV-specific CTL responses and reversed the exhausted memory phenotype from a T-betlow/Eomes+ to a T-bethi/Eomes+ phenotype. These results indicate that the PD-L1/PD-1 signaling pathway has a previously unappreciated dual role in the induction and regulation of HIV-1-specific CTL immunity, which is greatly determined by the context and differentiation stage of the responsive CD8+ T cells.IMPORTANCE Targeting the PD-1/PD-L1 immune checkpoint axis with signaling inhibitors has proven to be a powerful immunotherapeutic strategy to enhance the functional quality and survival of existing antigen-specific effector T cells. However, our study demonstrates that the context and timing of PD-1 signaling in T cells greatly impact the outcome of the effector response. In particular, we show that PD-1 activation plays a positive role during the DC-mediated initiation stage of the primary T cell response, while it serves as an inhibitory mechanism during the effector phase of the response. Therefore, caution should be taken in the design of therapies that include targeting of the PD-1/PD-L1 signaling pathway in order to avoid potential negative impacts on the induction of de novo T cell responses.
Collapse
Affiliation(s)
- Tatiana M Garcia-Bates
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Mariana L Palma
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Chengli Shen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bernard J C Macatangay
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles R Rinaldo
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robbie B Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
33
|
Jones D, Como CN, Jing L, Blackmon A, Neff CP, Krueger O, Bubak AN, Palmer BE, Koelle DM, Nagel MA. Varicella zoster virus productively infects human peripheral blood mononuclear cells to modulate expression of immunoinhibitory proteins and blocking PD-L1 enhances virus-specific CD8+ T cell effector function. PLoS Pathog 2019; 15:e1007650. [PMID: 30870532 PMCID: PMC6435197 DOI: 10.1371/journal.ppat.1007650] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/26/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022] Open
Abstract
Varicella zoster virus (VZV) is a lymphotropic alpha-herpesvirinae subfamily member that produces varicella on primary infection and causes zoster, vascular disease and vision loss upon reactivation from latency. VZV-infected peripheral blood mononuclear cells (PBMCs) disseminate virus to distal organs to produce clinical disease. To assess immune evasion strategies elicited by VZV that may contribute to dissemination of infection, human PBMCs and VZV-specific CD8+ T cells (V-CD8+) were mock- or VZV-infected and analyzed for immunoinhibitory protein PD-1, PD-L1, PD-L2, CTLA-4, LAG-3 and TIM-3 expression using flow cytometry. All VZV-infected PBMCs (monocytes, NK, NKT, B cells, CD4+ and CD8+ T cells) and V-CD8+ showed significant elevations in PD-L1 expression compared to uninfected cells. VZV induced PD-L2 expression in B cells and V-CD8+. Only VZV-infected CD8+ T cells, NKT cells and V-CD8+ upregulated PD-1 expression, the immunoinhibitory receptor for PD-L1/PD-L2. VZV induced CTLA-4 expression only in V-CD8+ and no significant changes in LAG-3 or TIM-3 expression were observed in V-CD8+ or PBMC T cells. To test whether PD-L1, PD-L2 or CTLA-4 regulates V-CD8+ effector function, autologous PBMCs were VZV-infected and co-cultured with V-CD8+ cells in the presence of blocking antibodies against PD-L1, PD-L2 or CTLA-4; ELISAs revealed significant elevations in IFNγ only upon blocking of PD-L1. Together, these results identified additional immune cells that are permissive to VZV infection (monocytes, B cells and NKT cells); along with a novel mechanism for inhibiting CD8+ T cell effector function through induction of PD-L1 expression.
Collapse
Affiliation(s)
- Dallas Jones
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Christina N. Como
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Anna Blackmon
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles Preston Neff
- Department of Medicine, Division of Allergy and Clinical Immunology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Owen Krueger
- Department of Medicine, Division of Allergy and Clinical Immunology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Andrew N. Bubak
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Brent E. Palmer
- Department of Medicine, Division of Allergy and Clinical Immunology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - David M. Koelle
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Benaroya Research Institute, Seattle, Washington, United States of America
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Maria A. Nagel
- Department of Neurology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
34
|
Yasuma-Mitobe K, Matsuoka M. The Roles of Coinhibitory Receptors in Pathogenesis of Human Retroviral Infections. Front Immunol 2018; 9:2755. [PMID: 30538707 PMCID: PMC6277675 DOI: 10.3389/fimmu.2018.02755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/08/2018] [Indexed: 12/14/2022] Open
Abstract
Costimulatory and coinhibitory receptors play a key role in regulating immune responses to infection and cancer. Coinhibitory receptors include programmed cell death 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and T cell immunoglobulin and ITIM domain (TIGIT), which suppress immune responses. Coinhibitory receptors are highly expressed on exhausted virus-specific T cells, indicating that viruses evade host immune responses through enhanced expression of these molecules. Human retroviruses, human immunodeficiency virus (HIV) and human T-cell leukemia virus type 1 (HTLV-1), infect T cells, macrophages and dendritic cells. Therefore, one needs to consider the effects of coinhibitory receptors on both uninfected effector T cells and infected target cells. Coinhibitory receptors are implicated not only in the suppression of immune responses to viruses by inhibition of effector T cells, but also in the persistence of infected cells in vivo. Here we review recent studies on coinhibitory receptors and their roles in retroviral infections such as HIV and HTLV-1.
Collapse
Affiliation(s)
- Keiko Yasuma-Mitobe
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masao Matsuoka
- Department of Hematology, Rheumatology and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Laboratory of Virus Control, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
35
|
Kumar NA, van der Sluis RM, Mota T, Pascoe R, Evans VA, Lewin SR, Cameron PU. Myeloid Dendritic Cells Induce HIV Latency in Proliferating CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:1468-1477. [PMID: 30030324 DOI: 10.4049/jimmunol.1701233] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 06/27/2018] [Indexed: 02/04/2023]
Abstract
HIV latency occurs predominantly in long-lived resting CD4+ T cells; however, latent infection also occurs in T cell subsets, including proliferating CD4+ T cells. We compared the establishment and maintenance of latent infection in nonproliferating and proliferating human CD4+ T cells cocultured with syngeneic myeloid dendritic cells (mDC). Resting CD4+ T cells were labeled with the proliferation dye eFluor 670 and cultured alone or with mDC, plasmacytoid dendritic cells, or monocytes in the presence of staphylococcal enterotoxin B (SEB). Cells were cultured for 24 h and infected with CCR5-tropic enhanced GFP (EGFP) reporter HIV. Five days postinfection, nonproductively infected EGFP- CD4+ T cells that were either nonproliferating (eFluor 670hi) or proliferating (eFluor 670lo) were sorted and cultured for an additional 7 d (day 12) with IL-7 and antiretrovirals. At day 5 postinfection, sorted, nonproductively infected T cells were stimulated with anti-CD3/CD28, and induced expression of EGFP was measured to determine the frequency of latent infection. Integrated HIV in these cells was confirmed using quantitative PCR. By these criteria, latent infection was detected at day 5 and 12 in proliferating T cells cocultured with mDC and monocytes but not plasmacytoid dendritic cells, where CD4+ T cells at day 12 were poor. At day 5 postinfection, nonproliferating T cells expressing SEB-specific TCR Vβ-17 were enriched in latent infection compared with non-SEB-specific TCR Vβ-8.1. Together, these data show that both nonproliferating and proliferating CD4+ T cells can harbor latent infection during SEB-stimulated T cell proliferation and that the establishment of HIV latency in nonproliferating T cells is linked to expression of specific TCR that respond to SEB.
Collapse
Affiliation(s)
- Nitasha A Kumar
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Renee M van der Sluis
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Talia Mota
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Rachel Pascoe
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Vanessa A Evans
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia.,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia; and.,Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Paul U Cameron
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia; .,Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia; and.,Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| |
Collapse
|
36
|
Sperk M, Domselaar RV, Neogi U. Immune Checkpoints as the Immune System Regulators and Potential Biomarkers in HIV-1 Infection. Int J Mol Sci 2018; 19:ijms19072000. [PMID: 29987244 PMCID: PMC6073446 DOI: 10.3390/ijms19072000] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 11/21/2022] Open
Abstract
Immune checkpoints are several co-stimulatory and inhibitory pathways that regulate T cell immune responses. Most of the discoveries about immune checkpoints were made in cancer research where inhibitory immune checkpoints cause immune exhaustion and down-regulate anti-tumor responses. In addition to cancer, immune checkpoints are exploited in chronic infectious diseases. In human immunodeficiency virus (HIV) infection, the immune checkpoint molecule called programmed cell death protein 1 (PD-1) has been determined as being a major regulatory factor for T cell exhaustion. Recent studies with antibodies blocking either PD-1 ligand 1 (PD-L1) or PD-1 show not only promising results in the enhancement of HIV-specific immune responses but even in reducing the latent HIV reservoir. Apart from the therapeutic target for a functional cure of HIV-1, immune checkpoint molecules might be used as biomarkers for monitoring disease progression and therapeutic response. In this review, we will summarize and discuss the inhibitory immune checkpoint molecules PD-1, cytotoxic T-lymphocyte-associated protein 4 (CTLA4), lymphocyte-activation gene 3 (LAG3), and T cell immunoglobulin and mucin-domain-containing-3 (TIM3) as well as the co-stimulatory molecules CD40L and CD70, including their role in immunity, with a particular focus on HIV infection, and being potential targets for a functional HIV cure.
Collapse
Affiliation(s)
- Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
| | - Robert van Domselaar
- Department of Medicine Huddinge, Unit of Infectious Diseases, Karolinska Institutet, Karolinska University Hospital, 141 86 Stockholm, Sweden.
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 141 86 Stockholm, Sweden.
| |
Collapse
|
37
|
CRISPR/Cas9-mediated deletion of miR-146a enhances antiviral response in HIV-1 infected cells. Genes Immun 2018; 20:327-337. [PMID: 29961753 DOI: 10.1038/s41435-018-0036-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023]
Abstract
The human immunodeficiency virus type 1 (HIV-1) causes persistent infection in human and induces miR-146a expression in infected cells. miR-146a represses the innate immune response by inhibiting the expression of TRAF6 and IRAK1 genes, thus negatively controls the NF-κB-related cytokines and interferon stimulated genes. Here we reported that lentiviral CRISPR/Cas9 system was highly efficient in introducing mutations in the precursor miR-146a genomic sequences, resulting in a loss of miR-146a expression and function. miR-146a ablation led to increasing cytokines production in LPS-stimulated A549 cells. Moreover, miR-146a knockout in HIV-1 infected MT2 cells markedly increased the expression of cytokines and HIV-1 restriction factors and reversed T cell exhaustion markers expression, thus influencing HIV-1 replication. Our study indicates that lentiviral CRISPR/Cas9-mediated gene editing is an effective approach to abrogate miR-146a expression, which consequently inhibits HIV-1 replication as well as proviral reactivation by enhancing the expression of cytokines and HIV-1 restriction factors.
Collapse
|
38
|
Liu F, Zeng G, Zhou S, He X, Sun N, Zhu X, Hu A. Blocking Tim-3 or/and PD-1 reverses dysfunction of tumor-infiltrating lymphocytes in HBV-related hepatocellular carcinoma. Bull Cancer 2018; 105:493-501. [PMID: 29576222 DOI: 10.1016/j.bulcan.2018.01.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/15/2018] [Accepted: 01/29/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND The immunosuppression of tumor-infiltrating lymphocytes (TILs) is associated with rapid progression of hepatitis B virus-related hepatocellular carcinoma (HBV-HCC). T cell Ig- and mucin-domain-containing molecule-3 (Tim-3) and programmed cell death 1 (PD-1) are important inhibitory molecules expressed on the surface of T cells, but their roles in the function of TILs in HBV-HCC are poorly understood. We aimed to study the roles of these two markers in HBV-HCC. METHODS Ninety patients with pathologically confirmed HBV-associated HCC were enrolled in our study. Blood samples, paired fresh tumor tissues and adjacent tissues were collected, and isolating peripheral blood mononuclear cells, TILs and adjacent-infiltrating lymphocytes were isolated from these samples. The patients were followed-up to allow survival analysis. RESULTS Tim-3 or/and PD-1 was up-regulated expressed on CD4+ and CD8+ TILs in HBV-HCC patients and a higher proportion of TILs expressed PD-1 alone. Tim-3+ and PD-1+ TILs greatly decreased secretion of IFN-? and TNF-a. Expression of Tim-3 and PD-1 on TILs negatively correlated with disease-free survival of HCC patients. Direct blockade of Tim-3 and PD-1 in vitro significantly enhanced TILs proliferation and secretion of IFN-? and TNF-a. CONCLUSION Expression of Tim-3 and/or PD-1 on TILs impairs their function and correlates negatively with disease-free survival in HBV-HCC. Direct blockade of Tim-3 and PD-1 restores anti-tumor effects of TILs, which suggests a potential target for novel immunotherapy in HBV-HCC.
Collapse
Affiliation(s)
- Furong Liu
- Sun Yat-Sen university, The First Affiliated Hospital, department of surgical intensive care unit, 510080 Guangzhou, China
| | - Gucheng Zeng
- Sun Yat-Sen university, department of microbiology, Zhongshan School of Medicine, Guangzhou, China
| | - Shaotang Zhou
- Military Hospital of China, department of hepatobiliary surgery, 302, Beijing, China
| | - Xiaoshun He
- Sun Yat-Sen university, The First Affiliated Hospital, department of surgical intensive care unit, 510080 Guangzhou, China
| | - Nianfeng Sun
- Shandong university, Qilu Hospital, department of general surgery, Jinan, China
| | - Xiaofeng Zhu
- Sun Yat-Sen university, The First Affiliated Hospital, department of surgical intensive care unit, 510080 Guangzhou, China
| | - Anbin Hu
- Sun Yat-Sen university, The First Affiliated Hospital, department of surgical intensive care unit, 510080 Guangzhou, China.
| |
Collapse
|
39
|
Tim-3 co-stimulation promotes short-lived effector T cells, restricts memory precursors, and is dispensable for T cell exhaustion. Proc Natl Acad Sci U S A 2018; 115:2455-2460. [PMID: 29463725 DOI: 10.1073/pnas.1712107115] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tim-3 is highly expressed on a subset of T cells during T cell exhaustion in settings of chronic viral infection and tumors. Using lymphocytic choriomeningitis virus (LCMV) Clone 13, a model for chronic infection, we found that Tim-3 was neither necessary nor sufficient for the development of T cell exhaustion. Nonetheless, expression of Tim-3 was sufficient to drive resistance to PD-L1 blockade therapy during chronic infection. Strikingly, expression of Tim-3 promoted the development of short-lived effector T cells, at the expense of memory precursor development, after acute LCMV infection. These effects were accompanied by increased Akt/mTOR signaling in T cells expressing endogenous or ectopic Tim-3. Conversely, Akt/mTOR signaling was reduced in effector T cells from Tim-3-deficient mice. Thus, Tim-3 is essential for optimal effector T cell responses, and may also contribute to exhaustion by restricting the development of long-lived memory T cells. Taken together, our results suggest that Tim-3 is actually more similar to costimulatory receptors that are up-regulated after T cell activation than to a dominant inhibitory protein like PD-1. These findings have significant implications for the development of anti-Tim-3 antibodies as therapeutic agents.
Collapse
|
40
|
Gorman JV, Colgan JD. Acute stimulation generates Tim-3-expressing T helper type 1 CD4 T cells that persist in vivo and show enhanced effector function. Immunology 2018; 154:418-433. [PMID: 29315553 DOI: 10.1111/imm.12890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022] Open
Abstract
T-cell immunoglobulin and mucin domain 3 (Tim-3) is a surface receptor expressed by T helper type 1 (Th1) effector CD4 T cells, which are critical for defence against intracellular pathogens and have been implicated in autoimmune disease. Previous studies showed that Tim-3 expression makes Th1 cells more susceptible to apoptosis and also marks functionally impaired T cells that arise due to chronic stimulation. However, other studies suggested that Tim-3-expressing Th1 cells do not always have these properties. To further define the relationship between Tim-3 and Th1 cell function, we analysed the characteristics of Th1 cells that expressed Tim-3 in response to brief stimulation in vitro or an acute viral infection in vivo. As expected, cultured CD4 T cells began expressing Tim-3 during Th1 differentiation and secondary stimulation generated Tim-3- and Tim-3+ fractions that were separated and further analysed. When injected into naive mice, Tim-3+ cells down-regulated Tim-3 and survived equally well compared with Tim-3- cells. Further, Tim-3- and Tim-3+ Th1 cells had similar functional responses when transferred into naive mice that were subsequently infected with lymphocytic choriomeningitis virus (LCMV). Cultured Th1 cells that expressed Tim-3 following T-cell receptor stimulation had a greater capacity to express signature Th1 cytokines than their Tim-3- counterparts and showed differential expression of genes that regulate CD4 T-cell function. Consistent with these findings, Tim-3+ Th1 cells generated in response to LCMV infection displayed augmented effector function relative to Tim-3- cells. These results suggest that Tim-3 expression by Th1 cells responding to acute stimulation can mark cells that are functionally competent and have an augmented ability to produce cytokines.
Collapse
Affiliation(s)
- Jacob V Gorman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John D Colgan
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
41
|
Marra A, Scognamiglio G, Peluso I, Botti G, Fusciello C, Filippelli A, Ascierto PA, Pepe S, Sabbatino F. Immune Checkpoint Inhibitors in Melanoma and HIV Infection. Open AIDS J 2017; 11:91-100. [PMID: 29290886 PMCID: PMC5730951 DOI: 10.2174/1874613601711010091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022] Open
Abstract
Introduction: Immunotherapy with immune checkpoint inhibitors increases the overall survival of patients with metastatic melanoma regardless of their oncogene addicted mutations. However, no data is available from clinical trials of effective therapies in subgroups of melanoma patients that carry chronic infective diseases such as HIV. Evidences suggest a key role of the immune checkpoint molecules as a mechanism of immune escape not only from melanoma but also from HIV host immune response. Conclusion: In this article, firstly, we will describe the role of the immune checkpoint molecules in HIV chronic infection. Secondly, we will summarize the most relevant clinical evidences utilizing immune checkpoint inhibitors for the treatment of melanoma patients. Lastly, we will discuss the potential implications as well as the potential applications of immune checkpoint molecule-based immunotherapy in patients with melanoma and HIV infection.
Collapse
Affiliation(s)
- Antonio Marra
- Department of Medical Oncology, San Gerardo Hospital, via G. B. Pergolesi, 20052 Monza, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Ilaria Peluso
- Hematology Unit, Department of Clinical and Surgical Medicine, University of Naples Federico II, via S. Pansini, 80131 Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Celeste Fusciello
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Amelia Filippelli
- Pharmacology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Stefano Pepe
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| |
Collapse
|
42
|
McGary CS, Deleage C, Harper J, Micci L, Ribeiro SP, Paganini S, Kuri-Cervantes L, Benne C, Ryan ES, Balderas R, Jean S, Easley K, Marconi V, Silvestri G, Estes JD, Sekaly RP, Paiardini M. CTLA-4 +PD-1 - Memory CD4 + T Cells Critically Contribute to Viral Persistence in Antiretroviral Therapy-Suppressed, SIV-Infected Rhesus Macaques. Immunity 2017; 47:776-788.e5. [PMID: 29045906 PMCID: PMC5679306 DOI: 10.1016/j.immuni.2017.09.018] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 06/13/2017] [Accepted: 09/27/2017] [Indexed: 01/09/2023]
Abstract
Antiretroviral therapy (ART) suppresses viral replication in HIV-infected individuals but does not eliminate the reservoir of latently infected cells. Recent work identified PD-1+ follicular helper T (Tfh) cells as an important cellular compartment for viral persistence. Here, using ART-treated, SIV-infected rhesus macaques, we show that CTLA-4+PD-1- memory CD4+ T cells, which share phenotypic markers with regulatory T cells, were enriched in SIV DNA in blood, lymph nodes (LN), spleen, and gut, and contained replication-competent and infectious virus. In contrast to PD-1+ Tfh cells, SIV-enriched CTLA-4+PD-1- CD4+ T cells were found outside the B cell follicle of the LN, predicted the size of the persistent viral reservoir during ART, and significantly increased their contribution to the SIV reservoir with prolonged ART-mediated viral suppression. We have shown that CTLA-4+PD-1- memory CD4+ T cells are a previously unrecognized component of the SIV and HIV reservoir that should be therapeutically targeted for a functional HIV-1 cure.
Collapse
Affiliation(s)
- Colleen S McGary
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Justin Harper
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Luca Micci
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Susan P Ribeiro
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sara Paganini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Clarisse Benne
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Emily S Ryan
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | - Sherrie Jean
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Atlanta, GA 30329, USA
| | - Vincent Marconi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Guido Silvestri
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Rafick-Pierre Sekaly
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA.
| |
Collapse
|
43
|
Hsu DC, Ananworanich J. Immune Interventions to Eliminate the HIV Reservoir. Curr Top Microbiol Immunol 2017; 417:181-210. [PMID: 29071472 DOI: 10.1007/82_2017_70] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inducing HIV remission is a monumental challenge. A potential strategy is the "kick and kill" approach where latently infected cells are first activated to express viral proteins and then eliminated through cytopathic effects of HIV or immune-mediated killing. However, pre-existing immune responses to HIV cannot eradicate HIV infection due to the presence of escape variants, inadequate magnitude, and breadth of responses as well as immune exhaustion. The two major approaches to boost immune-mediated elimination of infected cells include enhancing cytotoxic T lymphocyte mediated killing and harnessing antibodies to eliminate HIV. Specific strategies include increasing the magnitude and breadth of T cell responses through therapeutic vaccinations, reversing the effects of T cell exhaustion using immune checkpoint inhibition, employing bispecific T cell targeting immunomodulatory proteins or dual-affinity re-targeting molecules to direct cytotoxic T lymphocytes to virus-expressing cells and broadly neutralizing antibody infusions. Methods to steer immune responses to tissue sites where latently infected cells are located need to be further explored. Ultimately, strategies to induce HIV remission must be tolerable, safe, and scalable in order to make a global impact.
Collapse
Affiliation(s)
- Denise C Hsu
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Jintanat Ananworanich
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA. .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA. .,US Military HIV Research Program (MHRP), 6720-A Rockledge Drive, Suite 400, Bethesda, MD, 20817, USA.
| |
Collapse
|
44
|
CD8+ T cells of chronic HCV-infected patients express multiple negative immune checkpoints following stimulation with HCV peptides. Cell Immunol 2016; 313:1-9. [PMID: 28104239 DOI: 10.1016/j.cellimm.2016.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/08/2016] [Accepted: 12/12/2016] [Indexed: 01/25/2023]
Abstract
Hepatitis C virus (HCV)-specific CD4+ and CD8+ T cells are key to successful viral clearance in HCV disease. Accumulation of exhausted HCV-specific T cells during chronic infection results in considerable loss of protective functional immune responses. The role of T-cell exhaustion in chronic HCV disease remains poorly understood. Here, we studied the frequency of HCV peptide-stimulated T cells expressing negative immune checkpoints (PD-1, CTLA-4, TRAIL, TIM-3 and BTLA) by flow cytometry, and measured the levels of Th1/Th2/Th17 cytokines secreted by T cells by a commercial Multi-Analyte ELISArray™ following in vitro stimulation of T cells using HCV peptides and phytohemagglutinin (PHA). HCV peptide-stimulated CD4+ and CD8+ T cells of chronic HCV (CHC) patients showed significant increase of CTLA-4. Furthermore, HCV peptide-stimulated CD4+ T cells of CHC patients also displayed relatively higher levels of PD-1 and TRAIL, whereas TIM-3 was up-regulated on HCV peptide-stimulated CD8+ T cells. Whereas the levels of IL-10 and TGF-β1 were significantly increased, the levels of pro-inflammatory cytokines IL-2, TNF-α, IL-17A and IL-6 were markedly decreased in the T cell cultures of CHC patients. Chronic HCV infection results in functional exhaustion of CD4+ and CD8+ T cells likely contributing to viral persistence.
Collapse
|
45
|
Thompson M, Heath SL, Sweeton B, Williams K, Cunningham P, Keele BF, Sen S, Palmer BE, Chomont N, Xu Y, Basu R, Hellerstein MS, Kwa S, Robinson HL. DNA/MVA Vaccination of HIV-1 Infected Participants with Viral Suppression on Antiretroviral Therapy, followed by Treatment Interruption: Elicitation of Immune Responses without Control of Re-Emergent Virus. PLoS One 2016; 11:e0163164. [PMID: 27711228 PMCID: PMC5053438 DOI: 10.1371/journal.pone.0163164] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/14/2016] [Indexed: 12/01/2022] Open
Abstract
GV-TH-01, a Phase 1 open-label trial of a DNA prime—Modified Vaccinia Ankara (MVA) boost vaccine (GOVX-B11), was undertaken in HIV infected participants on antiretroviral treatment (ART) to evaluate safety and vaccine-elicited T cell responses, and explore the ability of elicited CD8+ T cells to control viral rebound during analytical treatment interruption (TI). Nine men who began antiretroviral therapy (ART) within 18 months of seroconversion and had sustained plasma HIV-1 RNA <50 copies/mL for at least 6 months were enrolled. Median age was 38 years, median pre-ART HIV-1 RNA was 140,000 copies/ml and mean baseline CD4 count was 755/μl. Two DNA, followed by 2 MVA, inoculations were given 8 weeks apart. Eight subjects completed all vaccinations and TI. Clinical and laboratory adverse events were generally mild, with no serious or grade 4 events. Only reactogenicity events were considered related to study drug. No treatment emergent viral resistance was seen. The vaccinations did not reduce viral reservoirs and virus re-emerged in all participants during TI, with a median time to re-emergence of 4 weeks. Eight of 9 participants had CD8+ T cells that could be stimulated by vaccine-matched Gag peptides prior to vaccination. Vaccinations boosted these responses as well as eliciting previously undetected CD8+ responses. Elicited T cells did not display signs of exhaustion. During TI, temporal patterns of viral re-emergence and Gag-specific CD8+ T cell expansion suggested that vaccine-specific CD8+ T cells had been stimulated by re-emergent virus in only 2 of 8 participants. In these 2, transient decreases in viremia were associated with Gag selection in known CD8+ T cell epitopes. We hypothesize that escape mutations, already archived in the viral reservoir, plus a poor ability of CD8+ T cells to traffic to and control virus at sites of re-emergence, limited the therapeutic efficacy of the DNA/MVA vaccine. TRIAL REGISTRATION clinicaltrials.gov NCT01378156.
Collapse
Affiliation(s)
- Melanie Thompson
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Sonya L. Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Bentley Sweeton
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Kathy Williams
- AIDS Research Consortium of Atlanta, Atlanta, Georgia, United States of America
| | - Pamela Cunningham
- Alabama Vaccine Research Clinic, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sharon Sen
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Brent E. Palmer
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Nicolas Chomont
- Centre de recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Canada
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Rahul Basu
- GeoVax, Inc., Atlanta, Georgia, United States of America
| | | | - Suefen Kwa
- GeoVax, Inc., Atlanta, Georgia, United States of America
| | | |
Collapse
|
46
|
Neff CP, Rhodes ME, Arnolds KL, Collins CB, Donnelly J, Nusbacher N, Jedlicka P, Schneider JM, McCarter MD, Shaffer M, Mazmanian SK, Palmer BE, Lozupone CA. Diverse Intestinal Bacteria Contain Putative Zwitterionic Capsular Polysaccharides with Anti-inflammatory Properties. Cell Host Microbe 2016; 20:535-547. [PMID: 27693306 DOI: 10.1016/j.chom.2016.09.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 07/19/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
Zwitterionic capsular polysaccharides (ZPSs) are bacterial products that modulate T cells, including inducing anti-inflammatory IL-10-secreting T regulatory cells (Tregs). However, only a few diverse bacteria are known to modulate the host immune system via ZPS. We present a genomic screen for bacteria encoding ZPS molecules. We identify diverse host-associated bacteria, including commensals and pathogens with known anti-inflammatory properties, with the capacity to produce ZPSs. Human mononuclear cells stimulated with lysates from putative ZPS-producing bacteria induce significantly greater IL-10 production and higher proportions of Tregs than lysates from non-ZPS-encoding relatives or a commensal strain of Bacteroides cellulosilyticus in which a putative ZPS biosynthetic operon was genetically disrupted. Similarly, wild-type B. cellulosilyticus DSM 14838, but not a close relative lacking a putative ZPS, attenuated experimental colitis in mice. Collectively, this screen identifies bacterial strains that may use ZPSs to interact with the host as well as those with potential probiotic properties.
Collapse
Affiliation(s)
- C Preston Neff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Matthew E Rhodes
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kathleen L Arnolds
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Colm B Collins
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jody Donnelly
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nichole Nusbacher
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jennifer M Schneider
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Martin D McCarter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael Shaffer
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarkis K Mazmanian
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Brent E Palmer
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Catherine A Lozupone
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
47
|
Schade H, Sen S, Neff CP, Freed BM, Gao D, Gutman JA, Palmer BE. Programmed Death 1 Expression on CD4 + T Cells Predicts Mortality after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2016; 22:2172-2179. [PMID: 27519280 DOI: 10.1016/j.bbmt.2016.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Excessive or persistent programmed death 1 (PD-1) expression on virus- or tumor-specific T cells during chronic viral infection or malignancy has been associated with impaired immune control. To assess the role of the PD-1 pathway in allogeneic stem cell transplantation (SCT), we examined PD-1 expression and maturation phenotype on T cells from 42 patients early (day 55 to 85) after cord blood (CB), matched unrelated donor, and matched related donor transplantation. Expression of PD-1 on CD4+ T cells was significantly elevated in all transplantation types, with the highest level observed in CB subjects. Elevated PD-1 expression on CD4+ T cells early after transplantation was observed in nonsurvivors (median, 40.2%; range, 15.1 to 86.1) compared with survivors (median, 23.6%; range, 8.4 to 55.2; P = .001), indicating its association with increased risk for mortality, especially with CB transplantations, where PD-1 was increased in nonsurvivors (median, 64.6%; range, 36.5 to 86.1) compared with survivors (median, 34.1%; range, 15.9 to 55.2; P = .01). Furthermore, T cell subset analysis revealed that PD-1 expression was further elevated on CD4+ T central memory in nonsurvivors (median, 49.8%; range, 15.1 to 83.4) compared with survivors (median, 24.8%; range, 8.9 to 71.3; P = .002) and on T effector memory cells in nonsurvivors (median, 69.1%; range, 24.7 to 92.6) compared with survivors (median, 43.7%; range, 13.9 to 96.5; P = .0003). Our findings suggest that elevation of PD-1 expression on CD4+ T cells is associated with mortality in CB and possibly all SCT recipients.
Collapse
Affiliation(s)
- Henning Schade
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Hematology and Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sharon Sen
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - C Preston Neff
- Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brian M Freed
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dexiang Gao
- Department of Pediatrics and Department of Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jonathan A Gutman
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Hematology and Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brent E Palmer
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Division of Allergy and Clinical Immunology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
48
|
Fromentin R, Bakeman W, Lawani MB, Khoury G, Hartogensis W, DaFonseca S, Killian M, Epling L, Hoh R, Sinclair E, Hecht FM, Bacchetti P, Deeks SG, Lewin SR, Sékaly RP, Chomont N. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART. PLoS Pathog 2016; 12:e1005761. [PMID: 27415008 PMCID: PMC4944956 DOI: 10.1371/journal.ppat.1005761] [Citation(s) in RCA: 326] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022] Open
Abstract
HIV persists in a small pool of latently infected cells despite antiretroviral therapy (ART). Identifying cellular markers expressed at the surface of these cells may lead to novel therapeutic strategies to reduce the size of the HIV reservoir. We hypothesized that CD4+ T cells expressing immune checkpoint molecules would be enriched in HIV-infected cells in individuals receiving suppressive ART. Expression levels of 7 immune checkpoint molecules (PD-1, CTLA-4, LAG-3, TIGIT, TIM-3, CD160 and 2B4) as well as 4 markers of HIV persistence (integrated and total HIV DNA, 2-LTR circles and cell-associated unspliced HIV RNA) were measured in PBMCs from 48 virally suppressed individuals. Using negative binomial regression models, we identified PD-1, TIGIT and LAG-3 as immune checkpoint molecules positively associated with the frequency of CD4+ T cells harboring integrated HIV DNA. The frequency of CD4+ T cells co-expressing PD-1, TIGIT and LAG-3 independently predicted the frequency of cells harboring integrated HIV DNA. Quantification of HIV genomes in highly purified cell subsets from blood further revealed that expressions of PD-1, TIGIT and LAG-3 were associated with HIV-infected cells in distinct memory CD4+ T cell subsets. CD4+ T cells co-expressing the three markers were highly enriched for integrated viral genomes (median of 8.2 fold compared to total CD4+ T cells). Importantly, most cells carrying inducible HIV genomes expressed at least one of these markers (median contribution of cells expressing LAG-3, PD-1 or TIGIT to the inducible reservoir = 76%). Our data provide evidence that CD4+ T cells expressing PD-1, TIGIT and LAG-3 alone or in combination are enriched for persistent HIV during ART and suggest that immune checkpoint blockers directed against these receptors may represent valuable tools to target latently infected cells in virally suppressed individuals.
Collapse
Affiliation(s)
- Rémi Fromentin
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Wendy Bakeman
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
| | - Mariam B. Lawani
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
| | - Gabriela Khoury
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Wendy Hartogensis
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Sandrina DaFonseca
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
| | - Marisela Killian
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Lorrie Epling
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Rebecca Hoh
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Elizabeth Sinclair
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Frederick M. Hecht
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Peter Bacchetti
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, United States of America
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Sharon R. Lewin
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | | | - Nicolas Chomont
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Vaccine and Gene Therapy Institute Florida, Port St Lucie, Florida, United States of America
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal, Faculty of Medicine, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
49
|
Sabins NC, Harman BC, Barone LR, Shen S, Santulli-Marotto S. Differential Expression of Immune Checkpoint Modulators on In Vitro Primed CD4(+) and CD8(+) T Cells. Front Immunol 2016; 7:221. [PMID: 27379090 PMCID: PMC4909735 DOI: 10.3389/fimmu.2016.00221] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/23/2016] [Indexed: 12/13/2022] Open
Abstract
PD-1, TIM-3, and LAG-3 are molecules shown to have immune modulatory properties, and although initially classified as indicators of T cell hyporesponsiveness, it has become clear that they are also associated with the normal course of T cell activation. Functional studies have focused mainly on CD8+ T cells during chronic inflammation due to interest in co-opting the cellular immune response to eliminate viral or cancerous threats; however, there remains a relative lack of data regarding the expression of these molecules on CD4+ T cells. Here, we report that expression of the immune checkpoint (IC) molecules PD-1, LAG-3, and TIM-3 are differentially expressed on CD4+ and CD8+ T cells in the allogeneic response resulting from a mixed lymphocyte reaction. In these studies, PD-1 expression is higher on CD4+ T cells compared to CD8+ T cells. In contrast, TIM-3 is expressed at higher levels on CD8+ T cells compared to CD4+ T cells with an apparent reciprocity in that PD-1+ CD4+ T cells are frequently TIM-3lo/−, while TIM-3-expressing CD8+ T cells are largely PD-1lo/−. In addition, there is a decrease in the frequency of TIM-3+ CD4+ cells producing IFN-γ and IL-5 compared to TIM-3+ CD8+ cells. Lastly, the memory T cell phenotype within each IC-expressing subset differs between CD4+ and CD8+ T cells. These findings highlight key differences in IC expression patterns between CD4+ and CD8+ T cells and may allow for more effective therapeutic targeting of these molecules in the future.
Collapse
Affiliation(s)
- Nina C Sabins
- Biologics Research, Janssen R&D , Spring House, PA , USA
| | | | - Linda R Barone
- Biologics Research, Janssen R&D , Spring House, PA , USA
| | - Shixue Shen
- Biologics Research, Janssen R&D , Spring House, PA , USA
| | | |
Collapse
|
50
|
Liu Y, Gao LF, Liang XH, Ma CH. Role of Tim-3 in hepatitis B virus infection: An overview. World J Gastroenterol 2016; 22:2294-2303. [PMID: 26900291 PMCID: PMC4735003 DOI: 10.3748/wjg.v22.i7.2294] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/08/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection has received increasing public attention. HBV is the prototypical member of hepadnaviruses, which naturally infect only humans and great apes and induce the acute and persistent chronic infection of hepatocytes. A large body of evidence has demonstrated that dysfunction of the host anti-viral immune response is responsible for persistent HBV replication, unresolved inflammation and disease progression. Many regulatory factors are involved in immune dysfunction. Among these, T cell immunoglobulin domain and mucin domain-3 (Tim-3), one of the immune checkpoint proteins, has attracted increasing attention due to its critical role in regulating both adaptive and innate immune cells. In chronic HBV infection, Tim-3 expression is elevated in many types of immune cells, such as T helper cells, cytotoxic T lymphocytes, dendritic cells, macrophages and natural killer cells. Tim-3 over-expression is often accompanied by impaired function of the above-mentioned immunocytes, and Tim-3 inhibition can at least partially rescue impaired immune function and thus promote viral clearance. A better understanding of the regulatory role of Tim-3 in host immunity during HBV infection will shed new light on the mechanisms of HBV-related liver disease and suggest new therapeutic methods for intervention.
Collapse
|