1
|
Gayen S, Mukherjee S, Dasgupta S, Roy S. Emerging druggable targets for immune checkpoint modulation in cancer immunotherapy: the iceberg lies beneath the surface. Apoptosis 2024; 29:1879-1913. [PMID: 39354213 DOI: 10.1007/s10495-024-02022-8] [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] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
The immune system serves as a fundamental defender against the initiation and progression of cancer. Failure of the immune system augments immunosuppressive action that leading to cancer manifestation. This immunosuppressive effect causes from significant alterations in immune checkpoint expression associated with tumoral progression. The tumor microenvironment promotes immune escape mechanisms that further amplifying immunosuppressive actions. Notably, substantial targeting of immune checkpoints has been pragmatic in the advancement of cancer research. This study highlights a comprehensive review of emerging druggable targets aimed at modulating immune checkpoint co-inhibitory as well as co-stimulatory molecules in response to immune system activation. This modulation has prompted to the development of newer therapeutic insights, eventually inducing immunogenic cell death through immunomodulatory actions. The study emphasizes the role of immune checkpoints in immunogenic regulation of cancer pathogenesis and explores potential therapeutic avenues in cancer immunotherapy.Modulation of Immunosuppressive and Immunostimulatory pathways of immune checkpoints in cancer immunotherapy.
Collapse
Affiliation(s)
- Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sandipan Dasgupta
- Department of Pharmaceutical Technology, Maulana Abul Kalam Azad University of Technology, Kolkata, West Bengal, 741249, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
2
|
Dookie RS, Villegas-Mendez A, Cheeseman A, Jones AP, Barroso R, Barrett JR, Draper SJ, Janse CJ, Grogan JL, MacDonald AS, Couper KN. Synergistic blockade of TIGIT and PD-L1 increases type-1 inflammation and improves parasite control during murine blood-stage Plasmodium yoelii non-lethal infection. Infect Immun 2024; 92:e0034524. [PMID: 39324794 PMCID: PMC11556036 DOI: 10.1128/iai.00345-24] [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: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
Pro-inflammatory immune responses are rapidly suppressed during blood-stage malaria but the molecular mechanisms driving this regulation are still incompletely understood. In this study, we show that the co-inhibitory receptors TIGIT and PD-1 are upregulated and co-expressed by antigen-specific CD4+ T cells (ovalbumin-specific OT-II cells) during non-lethal Plasmodium yoelii expressing ovalbumin (PyNL-OVA) blood-stage infection. Synergistic blockade of TIGIT and PD-L1, but not individual blockade of each receptor, during the early stages of infection significantly improved parasite control during the peak stages (days 10-15) of infection. Mechanistically, this protection was correlated with significantly increased plasma levels of IFN-γ, TNF, and IL-2, and an increase in the frequencies of IFN-γ-producing antigen-specific T-bet+ CD4+ T cells (OT-II cells), but not antigen-specific CD8+ T cells (OT-I cells), along with expansion of the splenic red pulp and monocyte-derived macrophage populations. Collectively, our study identifies a novel role for TIGIT in combination with the PD1-PD-L1 axis in regulating specific components of the pro-inflammatory immune response and restricting parasite control during the acute stages of blood-stage PyNL infection.
Collapse
Affiliation(s)
- Rebecca S. Dookie
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ana Villegas-Mendez
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Antonn Cheeseman
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Adam P. Jones
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ruben Barroso
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | | | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Chris J. Janse
- Leiden Malaria Group, Center of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jane L. Grogan
- Department of Cancer Immunology, Genentech, South San Francisco, California, USA
| | - Andrew S. MacDonald
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
3
|
Rishiq A, Liu M, Mandelboim O. Enhancing immunity against Candida albicans infections through TIGIT knockout. mBio 2024; 15:e0116524. [PMID: 39109867 PMCID: PMC11389390 DOI: 10.1128/mbio.01165-24] [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: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 09/12/2024] Open
Abstract
T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT) is an inhibitory receptor expressed by T and natural killer cells. Here, we used TIGIT knockout (KO) mice to demonstrate that mouse TIGIT directly interacts with Candida albicans. Reduced fungal growth and colonization were observed when TIGIT-KO splenocytes were co-cultured with C. albicans compared to the wild type (WT). In a systemic candidiasis model, TIGIT-KO mice exhibited improved survival and reduced body weight loss compared to WT mice. Organ-specific fungal burden assessment revealed significantly lower fungal loads in the kidneys, spleen, and lungs of TIGIT-KO mice. Finally, we show that the agglutinin-like sequence proteins ALS6, ALS7, and ALS9 of C. albicans are ligands for TIGIT and that the absence of these proteins abolishes the TIGIT effect in vivo. Our results identify the significance of TIGIT in modulating host defense against C. albicans and highlight the potential therapeutic implications for C. albicans infections. IMPORTANCE Our results identify the significance of T cell immunoreceptor with immunoglobulin and ITIM domain in modulating host defense against Candida albicans and highlight the potential therapeutic implications for C. albicans infections.
Collapse
Affiliation(s)
- Ahmed Rishiq
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Mingdong Liu
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
4
|
McCulloch TR, Rossi GR, Miranda-Hernandez S, Valencia-Hernandez AM, Alim L, Belle CJ, Krause A, Zacchi LF, Lam PY, Nakamura K, Kupz A, Wells TJ, Souza-Fonseca-Guimaraes F. The immune checkpoint TIGIT is upregulated on T cells during bacterial infection and is a potential target for immunotherapy. Immunol Cell Biol 2024; 102:721-733. [PMID: 38873699 DOI: 10.1111/imcb.12794] [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: 05/20/2024] [Revised: 05/25/2024] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
Antibiotic resistance is a major public health threat, and alternatives to antibiotic therapy are urgently needed. Immunotherapy, particularly the blockade of inhibitory immune checkpoints, is a leading treatment option in cancer and autoimmunity. In this study, we used a murine model of Salmonella Typhimurium infection to investigate whether immune checkpoint blockade could be applied to bacterial infection. We found that the immune checkpoint T-cell immunoglobulin and ITIM domain (TIGIT) was significantly upregulated on lymphocytes during infection, particularly on CD4+ T cells, drastically limiting their proinflammatory function. Blockade of TIGIT in vivo using monoclonal antibodies was able to enhance immunity and improve bacterial clearance. The efficacy of anti-TIGIT was dependent on the capacity of the antibody to bind to Fc (fragment crystallizable) receptors, giving important insights into the mechanism of anti-TIGIT therapy. This research suggests that targeting immune checkpoints, such as TIGIT, has the potential to enhance immune responses toward bacteria and restore antibacterial treatment options in the face of antibiotic resistance.
Collapse
Affiliation(s)
- Timothy R McCulloch
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Gustavo R Rossi
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Socorro Miranda-Hernandez
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | | | - Louisa Alim
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Clemence J Belle
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Andrew Krause
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Lucia F Zacchi
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Pui Yeng Lam
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kyohei Nakamura
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Andreas Kupz
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
5
|
Webb GM, Pessoa CT, McCullen AJ, Hwang JM, Humkey MC, Thormin-Odum R, Kukula KA, Smedley J, Fischer M, Sciurba J, Bochart RM, Shriver-Munsch C, Ndhlovu LC, Sacha JB. Immune restoration by TIGIT blockade is insufficient to control chronic SIV infection. J Virol 2024; 98:e0027324. [PMID: 38775481 PMCID: PMC11237531 DOI: 10.1128/jvi.00273-24] [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: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 06/12/2024] Open
Abstract
TIGIT is a negative immune checkpoint receptor associated with T cell exhaustion in cancer and HIV. TIGIT upregulation in virus-specific CD8+ T cells and NK cells during HIV/SIV infection results in dysfunctional effector capabilities. In vitro studies targeting TIGIT on CD8+ T cells suggest TIGIT blockade as a viable strategy to restore SIV-specific T cell responses. Here, we extend these studies in vivo using TIGIT blockage in nonhuman primates in an effort to reverse T cell and NK cell exhaustion in the setting of SIV infection. We demonstrate that in vivo administration of a humanized anti-TIGIT monoclonal antibody (mAb) is well tolerated in both cynomolgus macaques and rhesus macaques. Despite sustained plasma concentrations of anti-TIGIT mAb, we observed no consistent improvement in NK or T cell cytolytic capacity. TIGIT blockade minimally enhanced T cell proliferation and virus-specific T cell responses in both magnitude and breadth though plasma viral loads in treated animals remained stable indicating that anti-TIGIT mAb treatment alone was insufficient to increase anti-SIV CD8+ T cell function. The enhancement of virus-specific T cell proliferative responses observed in vitro with single or dual blockade of TIGIT and/or PD-1 highlights TIGIT as a potential target to reverse T cell dysfunction. Our studies, however, reveal that targeting the TIGIT pathway alone may be insufficient in the setting of viremia and that combining immune checkpoint blockade with other immunotherapeutics may be a future path forward for improved viral control or elimination of HIV.IMPORTANCEUpregulation of the immune checkpoint receptor TIGIT is associated with HIV-mediated T cell dysfunction and correlates with HIV disease progression. Compelling evidence exists for targeting immune checkpoint receptor pathways that would potentially enhance immunity and refocus effector cell efforts toward viral clearance. In this report, we investigate TIGIT blockade as an immunotherapeutic approach to reverse immune exhaustion during chronic SIV/SHIV infection in a nonhuman primate model of HIV infection. We show that interfering with the TIGIT signaling axis alone is insufficient to improve viral control despite modest improvement in T cell immunity. Our data substantiate the use of targeting multiple immune checkpoint receptors to promote synergy and ultimately eliminate HIV-infected cells.
Collapse
Affiliation(s)
- Gabriela M. Webb
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Cleiton T. Pessoa
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Allyson J. McCullen
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Joseph M. Hwang
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Matthew C. Humkey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Raymond Thormin-Odum
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Kaitlyn A. Kukula
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Miranda Fischer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Joseph Sciurba
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Rachele M. Bochart
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Christine Shriver-Munsch
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Lishomwa C. Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Jonah B. Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
6
|
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
|
7
|
Trivedi P, Jhala G, De George DJ, Chiu C, Selck C, Ge T, Catterall T, Elkerbout L, Boon L, Joller N, Kay TW, Thomas HE, Krishnamurthy B. TIGIT acts as an immune checkpoint upon inhibition of PD1 signaling in autoimmune diabetes. Front Immunol 2024; 15:1370907. [PMID: 38533515 PMCID: PMC10964479 DOI: 10.3389/fimmu.2024.1370907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction Chronic activation of self-reactive T cells with beta cell antigens results in the upregulation of immune checkpoint molecules that keep self-reactive T cells under control and delay beta cell destruction in autoimmune diabetes. Inhibiting PD1/PD-L1 signaling results in autoimmune diabetes in mice and humans with pre-existing autoimmunity against beta cells. However, it is not known if other immune checkpoint molecules, such as TIGIT, can also negatively regulate self-reactive T cells. TIGIT negatively regulates the CD226 costimulatory pathway, T-cell receptor (TCR) signaling, and hence T-cell function. Methods The phenotype and function of TIGIT expressing islet infiltrating T cells was studied in non-obese diabetic (NOD) mice using flow cytometry and single cell RNA sequencing. To determine if TIGIT restrains self-reactive T cells, we used a TIGIT blocking antibody alone or in combination with anti-PDL1 antibody. Results We show that TIGIT is highly expressed on activated islet infiltrating T cells in NOD mice. We identified a subset of stem-like memory CD8+ T cells expressing multiple immune checkpoints including TIGIT, PD1 and the transcription factor EOMES, which is linked to dysfunctional CD8+ T cells. A known ligand for TIGIT, CD155 was expressed on beta cells and islet infiltrating dendritic cells. However, despite TIGIT and its ligand being expressed, islet infiltrating PD1+TIGIT+CD8+ T cells were functional. Inhibiting TIGIT in NOD mice did not result in exacerbated autoimmune diabetes while inhibiting PD1-PDL1 resulted in rapid autoimmune diabetes, indicating that TIGIT does not restrain islet infiltrating T cells in autoimmune diabetes to the same degree as PD1. Partial inhibition of PD1-PDL1 in combination with TIGIT inhibition resulted in rapid diabetes in NOD mice. Discussion These results suggest that TIGIT and PD1 act in synergy as immune checkpoints when PD1 signaling is partially impaired. Beta cell specific stem-like memory T cells retain their functionality despite expressing multiple immune checkpoints and TIGIT is below PD1 in the hierarchy of immune checkpoints in autoimmune diabetes.
Collapse
Affiliation(s)
- Prerak Trivedi
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Gaurang Jhala
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - David J De George
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Chris Chiu
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Claudia Selck
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Tingting Ge
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Tara Catterall
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | - Lorraine Elkerbout
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
| | | | - Nicole Joller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Thomas W Kay
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Helen E Thomas
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Balasubramanian Krishnamurthy
- Immunology and Diabetes Unit, St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| |
Collapse
|
8
|
Joller N, Anderson AC, Kuchroo VK. LAG-3, TIM-3, and TIGIT: Distinct functions in immune regulation. Immunity 2024; 57:206-222. [PMID: 38354701 PMCID: PMC10919259 DOI: 10.1016/j.immuni.2024.01.010] [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: 10/11/2023] [Revised: 12/18/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024]
Abstract
LAG-3, TIM-3, and TIGIT comprise the next generation of immune checkpoint receptors being harnessed in the clinic. Although initially studied for their roles in restraining T cell responses, intense investigation over the last several years has started to pinpoint the unique functions of these molecules in other immune cell types. Understanding the distinct processes that these receptors regulate across immune cells and tissues will inform the clinical development and application of therapies that either antagonize or agonize these receptors, as well as the profile of potential tissue toxicity associated with their targeting. Here, we discuss the distinct functions of LAG-3, TIM-3, and TIGIT, including their contributions to the regulation of immune cells beyond T cells, their roles in disease, and the implications for their targeting in the clinic.
Collapse
Affiliation(s)
- Nicole Joller
- Department of Quantitative Biomedicine, University of Zurich, 8057 Zurich, Switzerland
| | - Ana C Anderson
- Gene Lay Institute of Immunology and Inflammation, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammation, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
9
|
Paine SK, Choudhury P, Alam M, Bhattacharyya C, Pramanik S, Tripathi D, Das C, Patel V, Ghosh S, Chatterjee S, Kanta Mondal L, Basu A. Multi-faceted dysregulated immune response for COVID-19 infection explaining clinical heterogeneity. Cytokine 2024; 174:156434. [PMID: 38141460 DOI: 10.1016/j.cyto.2023.156434] [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: 04/25/2023] [Revised: 10/24/2023] [Accepted: 11/05/2023] [Indexed: 12/25/2023]
Abstract
Clinical heterogeneity and varied prognosis are well noted for SARS-CoV-2 infection. Altered immune response is a major feature for the adverse prognosis however focus on altered immune response has been primarily limited to hyper-inflammatory responses like Cytokine storm. A deeper understanding of viral pathobiology and the interplay of innate and adaptive immune cells against SARS-CoV-2 infection is essential to optimize intervention strategy and future preparedness for SARS-CoV-2 or its related viral diseases. To uncover the immunological signatures driving the progression of SARS-CoV-2 infection, we performed an extensive immunophenotype on blood samples from 79 hospitalized patients with mild/moderate to severe infections as well as from healthy controls and recovered donors to understand the interplay between innate and adaptive responses impacting severity and prognosis. We observed multifarious immune dysregulation, varied across patients of the clinical spectrum. We observed 4 major dysregulations of immune phenotypes 1) depletion of M1φ (impaired antiviral response as APC), 2) immune suppression/exhaustion via activation of repressor like CD4+/CD8+PD1, TIM3, LAG3 3) inappropriate differentiation of lymphocyte (extreme elevated proportion of CD4 naive, memory B and T cells along with reduction of inflammatory activator like TLR2/4/TIGIT) and 4) cytokine storm. Our results show the identification of biomarkers to differentiate the different trajectories for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Suman K Paine
- National Institute of Biomedical Genomics, Kalyani, India.
| | | | - Mahabub Alam
- National Institute of Biomedical Genomics, Kalyani, India
| | | | | | - Devashish Tripathi
- National Institute of Biomedical Genomics, Kalyani, India; Regional Centre for Biotechnology, Delhi, India
| | | | - Vatsal Patel
- National Institute of Biomedical Genomics, Kalyani, India
| | | | | | | | - Analabha Basu
- National Institute of Biomedical Genomics, Kalyani, India.
| |
Collapse
|
10
|
Wang X, Liu Z, Wang D, Zhang Y, Zhang H, Xue F, Wang X, Tang Z, Han X. Immunoswitch Nanomodulators Enable Active Targeting and Selective Proliferation of Regulatory T Cells for Multiple Sclerosis Therapy. ACS NANO 2024; 18:770-782. [PMID: 38113242 DOI: 10.1021/acsnano.3c09225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Interleukin-2 (IL-2) used in multiple sclerosis (MS) therapy modulates the balance between regulatory T (Treg) cells and effector T (Teff) cells. However, the off-target activation of Teff cells by IL-2 limits its clinical application. Therefore, a rapidly prepared immunoswitch nanomodulator termed aT-IL2C NPs was developed, which specifically recognized Treg cells with high TIGIT expression thanks to the presence of an anti-TIGIT and an IL-2/JES6-1 complex (IL2C) being delivered to Treg cells but not to Teff cells with low TIGIT expression. Then, IL2C released IL-2 due to the specific expression of the high-affinity IL-2 receptor on Treg cells, thus enabling the active targeting and selective proliferation of Treg cells. Moreover, the anti-TIGIT of aT-IL2C NPs selectively inhibited the proliferation of Teff cells while leaving the proliferation of Treg cells unaffected. In addition, since the IL-2 receptor on Teff cells had medium-affinity, the IL2C hardly released IL-2 to Teff cells, thus enabling the inhibition of Teff cell proliferation. The treatment of experimental autoimmune encephalomyelitis (EAE) mice with aT-IL2C NPs ameliorated the severity of the EAE and restored white matter integrity. Collectively, this work described a potential promising agent for effective MS therapy.
Collapse
Affiliation(s)
- Xiaoshuang Wang
- No. 1 Department of Neurology, China-Japan Union Hospital of Jilin University, 126 Xiantai Road, Changchun 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
| | - Zhilin Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
| | - Di Wang
- No. 1 Department of Neurology, China-Japan Union Hospital of Jilin University, 126 Xiantai Road, Changchun 130033, China
| | - Yingyu Zhang
- No. 1 Department of Neurology, China-Japan Union Hospital of Jilin University, 126 Xiantai Road, Changchun 130033, China
| | - Honglei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China
| | - Fuxin Xue
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
- Key Laboratory of UV-Emitting Materials and Technology (Northeast Normal University), Ministry of Education, Changchun, Jilin 130024, China
| | - Xianhong Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun 130022, China
| | - Xuemei Han
- No. 1 Department of Neurology, China-Japan Union Hospital of Jilin University, 126 Xiantai Road, Changchun 130033, China
| |
Collapse
|
11
|
Corneillie L, Lemmens I, Weening K, De Meyer A, Van Houtte F, Tavernier J, Meuleman P. Virus-Host Protein Interaction Network of the Hepatitis E Virus ORF2-4 by Mammalian Two-Hybrid Assays. Viruses 2023; 15:2412. [PMID: 38140653 PMCID: PMC10748205 DOI: 10.3390/v15122412] [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: 09/14/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Throughout their life cycle, viruses interact with cellular host factors, thereby influencing propagation, host range, cell tropism and pathogenesis. The hepatitis E virus (HEV) is an underestimated RNA virus in which knowledge of the virus-host interaction network to date is limited. Here, two related high-throughput mammalian two-hybrid approaches (MAPPIT and KISS) were used to screen for HEV-interacting host proteins. Promising hits were examined on protein function, involved pathway(s), and their relation to other viruses. We identified 37 ORF2 hits, 187 for ORF3 and 91 for ORF4. Several hits had functions in the life cycle of distinct viruses. We focused on SHARPIN and RNF5 as candidate hits for ORF3, as they are involved in the RLR-MAVS pathway and interferon (IFN) induction during viral infections. Knocking out (KO) SHARPIN and RNF5 resulted in a different IFN response upon ORF3 transfection, compared to wild-type cells. Moreover, infection was increased in SHARPIN KO cells and decreased in RNF5 KO cells. In conclusion, MAPPIT and KISS are valuable tools to study virus-host interactions, providing insights into the poorly understood HEV life cycle. We further provide evidence for two identified hits as new host factors in the HEV life cycle.
Collapse
Affiliation(s)
- Laura Corneillie
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Irma Lemmens
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Karin Weening
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Freya Van Houtte
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
12
|
de Lima MHF, Machado CC, Nascimento DC, Silva CMS, Toller-Kawahisa JE, Rodrigues TS, Veras FP, Pontelli MC, Castro IA, Zamboni DS, Filho JCA, Cunha TM, Arruda E, da Cunha LD, Oliveira RDR, Cunha FQ, Louzada-Junior P. The TIGIT + T regulatory cells subset associates with nosocomial infection and fatal outcome in COVID-19 patients under mechanical ventilation. Sci Rep 2023; 13:13599. [PMID: 37604833 PMCID: PMC10442317 DOI: 10.1038/s41598-023-39924-7] [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: 11/22/2022] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
The TIGIT+FOXP3+Treg subset (TIGIT+Tregs) exerts robust suppressive activity on cellular immunity and predisposes septic individuals to opportunistic infection. We hypothesized that TIGIT+Tregs could play an important role in intensifying the COVID-19 severity and hampering the defense against nosocomial infections during hospitalization. Herein we aimed to verify the association between the levels of the TIGIT+Tregs with the mechanical ventilation requirement, fatal outcome, and bacteremia during hospitalization. TIGIT+Tregs were immunophenotyped by flow cytometry from the peripheral blood of 72 unvaccinated hospitalized COVID-19 patients at admission from May 29th to August 6th, 2020. The patients were stratified during hospitalization according to their mechanical ventilation requirement and fatal outcome. COVID-19 resulted in a high prevalence of the TIGIT+Tregs at admission, which progressively increased in patients with mechanical ventilation needs and fatal outcomes. The prevalence of TIGIT+Tregs positively correlated with poor pulmonary function and higher plasma levels of LDH, HMGB1, FGL2, and TNF. The non-survivors presented higher plasma levels of IL-33, HMGB1, FGL2, IL-10, IL-6, and 5.54 times more bacteremia than survivors. Conclusions: The expansion of the TIGIT+Tregs in COVID-19 patients was associated with inflammation, lung dysfunction, bacteremia, and fatal outcome.
Collapse
Affiliation(s)
- Mikhael Haruo Fernandes de Lima
- Divisions of Clinical Immunology, Emergency, Infectious Diseases and Intensive Care Unit, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Caio Cavalcante Machado
- Divisions of Clinical Immunology, Emergency, Infectious Diseases and Intensive Care Unit, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Daniele Carvalho Nascimento
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Camila Meirelles S Silva
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Juliana Escher Toller-Kawahisa
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Tamara Silva Rodrigues
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Flavio Protassio Veras
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Marjorie Cornejo Pontelli
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Italo A Castro
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dario Simões Zamboni
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - José-Carlos A Filho
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Eurico Arruda
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Larissa Dias da Cunha
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renê D R Oliveira
- Divisions of Clinical Immunology, Emergency, Infectious Diseases and Intensive Care Unit, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Fernando Q Cunha
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
- Departament of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil.
| | - Paulo Louzada-Junior
- Divisions of Clinical Immunology, Emergency, Infectious Diseases and Intensive Care Unit, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil.
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
13
|
Clement M, Ladell K, Miners KL, Marsden M, Chapman L, Cardus Figueras A, Scott J, Andrews R, Clare S, Kriukova VV, Lupyr KR, Britanova OV, Withers DR, Jones SA, Chudakov DM, Price DA, Humphreys IR. Inhibitory IL-10-producing CD4 + T cells are T-bet-dependent and facilitate cytomegalovirus persistence via coexpression of arginase-1. eLife 2023; 12:e79165. [PMID: 37440306 PMCID: PMC10344424 DOI: 10.7554/elife.79165] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/11/2023] [Indexed: 07/14/2023] Open
Abstract
Inhibitory CD4+ T cells have been linked with suboptimal immune responses against cancer and pathogen chronicity. However, the mechanisms that underpin the development of these regulatory cells, especially in the context of ongoing antigen exposure, have remained obscure. To address this knowledge gap, we undertook a comprehensive functional, phenotypic, and transcriptomic analysis of interleukin (IL)-10-producing CD4+ T cells induced by chronic infection with murine cytomegalovirus (MCMV). We identified these cells as clonally expanded and highly differentiated TH1-like cells that developed in a T-bet-dependent manner and coexpressed arginase-1 (Arg1), which promotes the catalytic breakdown of L-arginine. Mice lacking Arg1-expressing CD4+ T cells exhibited more robust antiviral immunity and were better able to control MCMV. Conditional deletion of T-bet in the CD4+ lineage suppressed the development of these inhibitory cells and also enhanced immune control of MCMV. Collectively, these data elucidated the ontogeny of IL-10-producing CD4+ T cells and revealed a previously unappreciated mechanism of immune regulation, whereby viral persistence was facilitated by the site-specific delivery of Arg1.
Collapse
Affiliation(s)
- Mathew Clement
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Kelly L Miners
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Morgan Marsden
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Lucy Chapman
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Anna Cardus Figueras
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Jake Scott
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Robert Andrews
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Simon Clare
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | - Valeriia V Kriukova
- Center of Life Sciences, Skolkovo Institute of Science and TechnologyMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of SciencesMoscowRussian Federation
- Institute of Clinical Molecular Biology, Christian-Albrecht-University of KielKielGermany
| | - Ksenia R Lupyr
- Center of Life Sciences, Skolkovo Institute of Science and TechnologyMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of SciencesMoscowRussian Federation
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
| | - Olga V Britanova
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of SciencesMoscowRussian Federation
- Institute of Clinical Molecular Biology, Christian-Albrecht-University of KielKielGermany
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
| | - David R Withers
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Simon A Jones
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Dmitriy M Chudakov
- Center of Life Sciences, Skolkovo Institute of Science and TechnologyMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of SciencesMoscowRussian Federation
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Abu Dhabi Stem Cell CenterAl MuntazahUnited Arab Emirates
| | - David A Price
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| | - Ian R Humphreys
- Division of Infection and Immunity, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffUnited Kingdom
| |
Collapse
|
14
|
Zhao J, Li L, Yin H, Feng X, Lu Q. TIGIT: An emerging immune checkpoint target for immunotherapy in autoimmune disease and cancer. Int Immunopharmacol 2023; 120:110358. [PMID: 37262959 DOI: 10.1016/j.intimp.2023.110358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Immune checkpoints (ICs), also referred to as co-inhibitory receptors (IRs), are essential for regulating immune cell function to maintain tolerance and prevent autoimmunity. IRs, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), have been shown to possess immunoregulatory properties that are relevant to various autoimmune diseases and cancers. Tumors are characterized by suppressive microenvironments with elevated levels of IRs on tumor-infiltrating lymphocytes (TILs). Therefore, IR blockade has shown great potential in cancer therapy and has even been approved for clinical use. However, other IRs, including cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT), may also represent promising targets for anti-tumor therapy. The increasing importance of IRs in autoimmune diseases has become apparent. In mouse models, TIGIT pathway blockade or TIGIT deficiency has been linked to T cell overactivation and proliferation, exacerbation of inflammation, and increased susceptibility to autoimmune disorders. On the other hand, TIGIT activation has been shown to alleviate autoimmune disorders in murine models. Given these findings, we examine the effects of TIGIT and its potential as a therapeutic target for both autoimmune diseases and cancers. It is clear that TIGIT represents an emerging and exciting target for immunotherapy in these contexts.
Collapse
Affiliation(s)
- Junpeng Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciencs, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Liming Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciencs, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Huiqi Yin
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciencs, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiwei Feng
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciencs, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciencs, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
15
|
Li H, Zhang J, Su C, Yang Z, Mei X, Zhang Z, Tian X, Wang S. Dynamic changes in TIGIT expression on the T-cell surface and TIGIT-mediated T-cell dysfunction in the brains of mice with chronic Toxoplasma gondii infection. Acta Trop 2023; 241:106871. [PMID: 36863503 DOI: 10.1016/j.actatropica.2023.106871] [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: 01/02/2023] [Revised: 02/09/2023] [Accepted: 02/26/2023] [Indexed: 03/04/2023]
Abstract
The immunosuppressive receptor TIGIT plays a vital role in the regulation of the immune system's response to pathogens. However, the expression pattern of this receptor in mouse brains during infection with Toxoplasma gondii cysts is not known. Here, we provide evidence of immunological changes and TIGIT expression in infected mouse brains through flow cytometry and QPCR. The obtained results show that TIGIT expression on brain T cells rose considerably after infection. T. gondii infection triggered the conversion of TIGIT+ TCM cells to TIGIT+ TEM cells and reduced their cytotoxicity. During the whole period of T. gondii infection, high intensity and persistent expression of IFN-γ and TNF-α in brain and serum of mice. This study shows that chronic T. gondii infection increases TIGIT expression on brain T cells and affects their immune function.
Collapse
Affiliation(s)
- Haoran Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jing Zhang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Changwei Su
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhenke Yang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xuefang Mei
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhenchao Zhang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaowei Tian
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| | - Shuai Wang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
16
|
Bergantini L, d'Alessandro M, Cavallaro D, Pordon E, Cassai L, Gangi S, Meloni F, Montagnani F, Paladini P, Refini RM, Luzzi L, Fossi A, Bargagli E, Bennett D. Immune checkpoint analysis of T-cell responses to pp65 and IE-1 antigens in end-stage lung diseases. Scand J Immunol 2023; 97:e13248. [PMID: 36574966 DOI: 10.1111/sji.13248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/14/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Lung transplant (LTX) patients are at high risk of cytomegalovirus (CMV) infection, which is often associated with high mortality and morbidity. Reactivation of CMV causes cell injury due to the cytopathic effect of viral replication and triggering of T cell immunity. The aim of this study was to compare expression of immune checkpoints (ICs) (PD-1, CTLA-4, LAG-3 and TIGIT) in CD4, CD8 and CD56 and activation markers CD137, CD154 and CD69 of end-stage patients awaiting lung transplant. Eighteen pre-LTX positive for anti-CMV IgG titres and 18 healthy subjects were enrolled. IC and activation markers have been evaluated through flow cytometric analysis in HC and pre-LTX patients. Reactive (QF+) and unreactive (QF-) patients were stratified according to QuantiFERON-CMV assays. ICs' and activation markers' expression were determined before and after in vitro stimulation with pp-65 and IE-1 antigens. Lower expression of PD-1 was observed in CD4 and CD8 cells of pre-LTX patients than controls, whereas CTLA4 appeared upregulated in CD56 and CD8 cells. TIGIT is increased on the surface of CD4, CD8 and NK cells after peptide stimulation in QF-negative patients and PD-1 is only downregulated after stimulation in the QF-positive patients. This study provides new evidence of immune dysregulation in patients with end-stage lung disorders, particularly in relation to immune checkpoint cell biology. The change in QF+ mostly happens on cytotoxic cells NK and CD8, while the changes in QF- were observed in adaptive immune cells, including CD4 and CD8.
Collapse
Affiliation(s)
- Laura Bergantini
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Miriana d'Alessandro
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Dalila Cavallaro
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Elena Pordon
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Lucia Cassai
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Sara Gangi
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Federica Meloni
- University of Pavia, pavia, Italy
- Department of Haematological, Pneumological and Cardiovascular Sciences, Fondazione IRCCS Policlinico San Matteo, pavia, Italy
| | - Francesca Montagnani
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Piero Paladini
- Thoracic Surgery Unit, Cardio-Thoracic and Vascular Department, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Rosa Metella Refini
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Luca Luzzi
- Thoracic Surgery Unit, Cardio-Thoracic and Vascular Department, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Antonella Fossi
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - Elena Bargagli
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| | - David Bennett
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena (Azienda Ospedaliera Universitaria Senese, AOUS), Siena, Italy
| |
Collapse
|
17
|
Hartigan CR, Tong KP, Liu D, Laurie SJ, Ford ML. TIGIT agonism alleviates costimulation blockade-resistant rejection in a regulatory T cell-dependent manner. Am J Transplant 2023; 23:180-189. [PMID: 36695691 PMCID: PMC10062175 DOI: 10.1016/j.ajt.2022.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/16/2022] [Indexed: 01/15/2023]
Abstract
Belatacept-based immunosuppression in kidney transplantation confers fewer off-target toxicities than calcineurin inhibitors but comes at a cost of increased incidence and severity of acute rejection, potentially due to its deleterious effect on both the number and function of Foxp3+ regulatory T cells (Tregs). TIGIT is a CD28 family coinhibitory receptor expressed on several subsets of immune cells including Tregs. We hypothesized that coinhibition through TIGIT signaling could function to ameliorate costimulation blockade-resistant rejection. The results demonstrate that treatment with an agonistic anti-TIGIT antibody, when combined with costimulation blockade by CTLA-4Ig, can prolong allograft survival in a murine skin graft model compared with CTLA-4Ig alone. Further, this prolongation of graft survival is accompanied by an increase in the frequency and number of graft-infiltrating Tregs and a concomitant reduction in the number of CD8+ T cells in the graft. Through the use of Treg-specific TIGIT conditional knockout animals, we demonstrated that the TIGIT-mediated reduction in the graft-infiltrating CD8+ T cell response is dependent on signaling of TIGIT on Foxp3+ Tregs. Our results highlight both the key functional role of TIGIT on Foxp3+ Tregs under conditions in which CTLA-4 is blocked and the therapeutic potential of TIGIT agonism to optimize costimulation blockade-based immunosuppression.
Collapse
Affiliation(s)
- Christina R Hartigan
- Department of Surgery and Emory Transplant Center, Suite 5105, 101 Woodruff Circle, Atlanta, Georgia 30322
| | - Katherine P Tong
- Department of Surgery and Emory Transplant Center, Suite 5105, 101 Woodruff Circle, Atlanta, Georgia 30322
| | - Danya Liu
- Department of Surgery and Emory Transplant Center, Suite 5105, 101 Woodruff Circle, Atlanta, Georgia 30322
| | - Sonia J Laurie
- Department of Surgery and Emory Transplant Center, Suite 5105, 101 Woodruff Circle, Atlanta, Georgia 30322
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Suite 5105, 101 Woodruff Circle, Atlanta, Georgia 30322.
| |
Collapse
|
18
|
Ruedas-Torres I, Sánchez-Carvajal JM, Carrasco L, Pallarés FJ, Larenas-Muñoz F, Rodríguez-Gómez IM, Gómez-Laguna J. PRRSV-1 induced lung lesion is associated with an imbalance between costimulatory and coinhibitory immune checkpoints. Front Microbiol 2023; 13:1007523. [PMID: 36713151 PMCID: PMC9878400 DOI: 10.3389/fmicb.2022.1007523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) induces a dysregulation on the innate and adaptive immune responses. T-cell activation requires a proper interaction and precise balance between costimulatory and coinhibitory molecules, commonly known as immune checkpoints. This study aims to evaluate the expression of immune checkpoints in lung and tracheobronchial lymph node from piglets infected with two PRRSV-1 strains of different virulence during the early stage of infection. Seventy 4-week-old piglets were grouped into three experimental groups: (i) control, (ii) 3249-infected group (low virulent strain), and (iii) Lena-infected group (virulent strain) and were euthanized at 1, 3, 6, 8, and 13 days post-infection (dpi). Lung and tracheobronchial lymph node were collected to evaluate histopathological findings, PRRSV viral load and mRNA expression of costimulatory (CD28, CD226, TNFRSF9, SELL, ICOS, and CD40) and coinhibitory (CTLA4, TIGIT, PD1/PDL1, TIM3, LAG3, and IDO1) molecules through RT-qPCR. Our findings highlight a mild increase of costimulatory molecules together with an earlier and stronger up-regulation of coinhibitory molecules in both organs from PRRSV-1-infected animals, especially in the lung from virulent Lena-infected animals. The simultaneous expression of coinhibitory immune checkpoints could work in synergy to control and limit the inflammation-induced tissue damage. Further studies should be addressed to determine the role of these molecules in later stages of PRRSV infection.
Collapse
|
19
|
Liechti T, Iftikhar Y, Mangino M, Beddall M, Goss CW, O’Halloran JA, Mudd PA, Roederer M. Immune phenotypes that are associated with subsequent COVID-19 severity inferred from post-recovery samples. Nat Commun 2022; 13:7255. [PMID: 36433939 PMCID: PMC9700777 DOI: 10.1038/s41467-022-34638-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
Severe COVID-19 causes profound immune perturbations, but pre-infection immune signatures contributing to severe COVID-19 remain unknown. Genome-wide association studies (GWAS) identified strong associations between severe disease and several chemokine receptors and molecules from the type I interferon pathway. Here, we define immune signatures associated with severe COVID-19 using high-dimensional flow cytometry. We measure the cells of the peripheral immune system from individuals who recovered from mild, moderate, severe or critical COVID-19 and focused only on those immune signatures returning to steady-state. Individuals that suffered from severe COVID-19 show reduced frequencies of T cell, mucosal-associated invariant T cell (MAIT) and dendritic cell (DC) subsets and altered chemokine receptor expression on several subsets, such as reduced levels of CCR1 and CCR2 on monocyte subsets. Furthermore, we find reduced frequencies of type I interferon-producing plasmacytoid DCs and altered IFNAR2 expression on several myeloid cells in individuals recovered from severe COVID-19. Thus, these data identify potential immune mechanisms contributing to severe COVID-19.
Collapse
Affiliation(s)
- Thomas Liechti
- grid.419681.30000 0001 2164 9667ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Maryland, 20892 USA
| | - Yaser Iftikhar
- grid.419681.30000 0001 2164 9667ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Maryland, 20892 USA
| | - Massimo Mangino
- grid.13097.3c0000 0001 2322 6764Department of Twin Research & Genetic Epidemiology, King’s College of London, London, UK ,grid.420545.20000 0004 0489 3985NIHR Biomedical Research Centre at Guy’s and St Thomas’ Foundation Trust, London, SE1 9RT UK
| | - Margaret Beddall
- grid.419681.30000 0001 2164 9667ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Maryland, 20892 USA
| | - Charles W. Goss
- grid.4367.60000 0001 2355 7002Division of Biostatistics, Washington University School of Medicine, St. Louis, MO USA
| | - Jane A. O’Halloran
- grid.4367.60000 0001 2355 7002Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO USA
| | - Philip A. Mudd
- grid.4367.60000 0001 2355 7002Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.4367.60000 0001 2355 7002Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Mario Roederer
- grid.419681.30000 0001 2164 9667ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Maryland, 20892 USA
| |
Collapse
|
20
|
Sodsai P, Ittiwut C, Ruenjaiman V, Ittiwut R, Jantarabenjakul W, Suphapeetiporn K, Shotelersuk V, Hirankarn N. TIGIT Monoallelic Nonsense Variant in Patient with Severe COVID-19 Infection, Thailand. Emerg Infect Dis 2022; 28:2350-2352. [PMID: 36191906 PMCID: PMC9622227 DOI: 10.3201/eid2811.220914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
A heterozygous nonsense variant in the TIGIT gene was identified in a patient in Thailand who had severe COVID-19, resulting in lower TIGIT expression in T cells. The patient’s T cells produced higher levels of cytokines upon stimulation. This mutation causes less-controlled immune responses, which might contribute to COVID-19 severity.
Collapse
|
21
|
Conner M, Hance KW, Yadavilli S, Smothers J, Waight JD. Emergence of the CD226 Axis in Cancer Immunotherapy. Front Immunol 2022; 13:914406. [PMID: 35812451 PMCID: PMC9263721 DOI: 10.3389/fimmu.2022.914406] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/26/2022] [Indexed: 01/31/2023] Open
Abstract
In recent years, a set of immune receptors that interact with members of the nectin/nectin-like (necl) family has garnered significant attention as possible points of manipulation in cancer. Central to this axis, CD226, TIGIT, and CD96 represent ligand (CD155)-competitive co-stimulatory/inhibitory receptors, analogous to the CTLA-4/B7/CD28 tripartite. The identification of PVRIG (CD112R) and CD112 has introduced complexity and enabled additional nodes of therapeutic intervention. By virtue of the clinical progression of TIGIT antagonists and emergence of novel CD96- and PVRIG-based approaches, our overall understanding of the 'CD226 axis' in cancer immunotherapy is starting to take shape. However, several questions remain regarding the unique characteristics of, and mechanistic interplay between, each receptor-ligand pair. This review provides an overview of the CD226 axis in the context of cancer, with a focus on the status of immunotherapeutic strategies (TIGIT, CD96, and PVRIG) and their underlying biology (i.e., cis/trans interactions). We also integrate our emerging knowledge of the immune populations involved, key considerations for Fc gamma (γ) receptor biology in therapeutic activity, and a snapshot of the rapidly evolving clinical landscape.
Collapse
|
22
|
Somasundaram A, Cillo AR, Lampenfeld C, Workman CJ, Kunning S, Oliveri LN, Velez M, Joyce S, Calderon M, Dadey R, Rajasundaram D, Normolle DP, Watkins SC, Herman JG, Kirkwood JM, Lipson EJ, Ferris RL, Bruno TC, Vignali DAA. Systemic immune dysfunction in cancer patients driven by IL6 induction of LAG3 in peripheral CD8+ T cells. Cancer Immunol Res 2022; 10:885-899. [PMID: 35587532 DOI: 10.1158/2326-6066.cir-20-0736] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 06/10/2021] [Accepted: 05/17/2022] [Indexed: 11/16/2022]
Abstract
Many cancer patients do not develop a durable response to the current standard of care immunotherapies, despite substantial advances in targeting immune inhibitory receptors. A potential compounding issue, which may serve as an unappreciated, dominant resistance mechanism, is an inherent systemic immune dysfunction that is often associated with advanced cancer. Minimal response to inhibitory receptor (IR) blockade therapy and increased disease burden have been associated with peripheral CD8+ T-cell dysfunction, characterized by suboptimal T-cell proliferation and chronic expression of IRs (eg. Programmed Death 1 [PD1] and Lymphocyte Activation Gene 3 [LAG3]). Here, we demonstrated that approximately a third of cancer patients analyzed in this study have peripheral CD8+ T cells that expressed robust intracellular LAG3 (LAG3IC), but not surface LAG3 (LAG3SUR) due to A Disintegrin and Metalloproteinase domain-containing protein 10 (ADAM10) cleavage. This associated with poor disease prognosis and decreased CD8+ T-cell function, which could be partially reversed by anti-LAG3. Systemic immune dysfunction was restricted to CD8+ T cells, including, in some cases, a high percentage of peripheral naïve CD8+ T cells, and was driven by the cytokine IL6 via STAT3. These data suggest that additional studies are warrented to determine if the combination of increased LAG3IC in peripheral CD8+ T cells and elevated systemic IL6 can serve as predictive biomarkers and identify which cancer patients may benefit from LAG3 blockade.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maria Velez
- University of Pittsburgh, Pittsburgh, PA, United States
| | - Sonali Joyce
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Michael Calderon
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Rebekah Dadey
- University of Pittsburgh, Pittsburgh, PA, United States
| | | | | | | | | | | | - Evan J Lipson
- Johns Hopkins University School of Medicine, BALTIMORE, MD, United States
| | - Robert L Ferris
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Tullia C Bruno
- University of Colorado Boulder, Pittsburgh, PA, United States
| | | |
Collapse
|
23
|
Pieren DKJ, Smits NAM, Postel RJ, Kandiah V, de Wit J, van Beek J, van Baarle D, Guichelaar T. Co-Expression of TIGIT and Helios Marks Immunosenescent CD8+ T Cells During Aging. Front Immunol 2022; 13:833531. [PMID: 35651622 PMCID: PMC9148977 DOI: 10.3389/fimmu.2022.833531] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aging leads to alterations in the immune system that result in ineffective responsiveness against pathogens. Features of this process, collectively known as immunosenescence, accumulate in CD8+ T cells with age and have been ascribed to differentiation of these cells during the course of life. Here we aimed to identify novel markers in CD8+ T cells associated with immunosenescence. Furthermore, we assessed how these markers relate to the aging-related accumulation of highly differentiated CD27-CD28- cells. We found that co-expression of the transcription factor Helios and the aging-related marker TIGIT identifies CD8+ T cells that fail to proliferate and show impaired induction of activation markers CD69 and CD25 in response to stimulation in vitro. Despite this, in blood of older adults we found TIGIT+Helios+ T cells to become highly activated during an influenza-A virus infection, but these higher frequencies of activated TIGIT+Helios+ T cells associate with longer duration of coughing. Moreover, in healthy individuals, we found that TIGIT+Helios+ CD8+ T cells accumulate with age in the highly differentiated CD27-CD28- population. Interestingly, TIGIT+Helios+ CD8+ T cells also accumulate with age among the less differentiated CD27+CD28- T cells before their transit into the highly differentiated CD27-CD28- stage. This finding suggests that T cells with immunosenescent features become prominent at old age also within the earlier differentiation states of these cells. Our findings show that co-expression of TIGIT and Helios refines the definition of immunosenescent CD8+ T cells and challenge the current dogma of late differentiation stage as proxy for T-cell immunosenescence.
Collapse
Affiliation(s)
- Daan K. J. Pieren
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Noortje A. M. Smits
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Rimke J. Postel
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Vinitha Kandiah
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Debbie van Baarle
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Teun Guichelaar
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
- *Correspondence: Teun Guichelaar,
| |
Collapse
|
24
|
Candida albicans evades NK cell elimination via binding of Agglutinin-Like Sequence proteins to the checkpoint receptor TIGIT. Nat Commun 2022; 13:2463. [PMID: 35513379 PMCID: PMC9072312 DOI: 10.1038/s41467-022-30087-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 04/11/2022] [Indexed: 11/09/2022] Open
Abstract
Candida albicans is the most common fungal pathogen and a prevalent cause of deadly bloodstream infections. Better understanding of the immune response against it, and the ways by which it evades immunity, are crucial for developing new therapeutics against it. Natural Killer (NK) cells are innate lymphocytes best known for their role against viruses and tumors. In recent years it became clear that NK cells also play an important role in anti-fungal immunity. Here we show that while NK cells recognize and eliminate C. albicans, the fungal cells inhibit NK cells by manipulating the immune checkpoint receptor TIGIT (T cell immunoreceptor with Ig and ITIM domains) in both humans and mice. We identify the responsible fungal ligands as members of the Als (Agglutinin-Like Sequences) protein family. Furthermore, we show that blocking this interaction using immunotherapy with a TIGIT-blocking antibody can re-establish anti-Candida immunity and serve as a potential therapeutic tool. Natural killer cells have emerged as critical immune cells in the response to fungal infection. Here the authors identify how Candida albicans evades the natural killer cell response via expression of ligands that directly modify the natural killer cell response and can be therapeutically targeted to restore the anti-Candida immunity.
Collapse
|
25
|
Qiu D, Liu X, Wang W, Jiang X, Wu X, Zheng J, Zhou K, Kong X, Wu X, Jin Z. TIGIT axis: novel immune checkpoints in anti-leukemia immunity. Clin Exp Med 2022; 23:165-174. [PMID: 35419661 DOI: 10.1007/s10238-022-00817-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/25/2022] [Indexed: 12/01/2022]
Abstract
Hematologic malignancy evades immune-mediated recognition through upregulating various checkpoint inhibitory receptors (IRs) on several types of lymphocytes. Immunotherapies targeting IRs have provided ample evidence supporting regulating innate and adaptive immunity and obtaining clinical benefits. Newly described IRs have received considerable attention and are under investigation in cancer immunotherapy. Specifically, T cell immunoglobulin and ITIM domain is a novel inhibitory checkpoint receptor, and its immune checkpoint axis includes additional receptors such as CD96 and CD226, which are very promising targets. However, how the dynamics and functions of these receptor networks remain unknown, this review addresses the recent findings of the relevance of this complex receptor-ligand system and discusses their potential approaches in translating these preclinical findings into novel clinical agents in anti-leukemia immunotherapy.
Collapse
Affiliation(s)
- Dan Qiu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiaxin Liu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Wandi Wang
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xuan Jiang
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiaofang Wu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jiamian Zheng
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Kai Zhou
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xueting Kong
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiuli Wu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Zhenyi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
26
|
Zhang T, Liu H, Jiao L, Zhang Z, He J, Li L, Qiu L, Qian Z, Zhou S, Gong W, Meng B, Ren X, Zhang H, Wang X. Genetic characteristics involving the PD-1/PD-L1/L2 and CD73/A2aR axes and the immunosuppressive microenvironment in DLBCL. J Immunother Cancer 2022; 10:jitc-2021-004114. [PMID: 35365585 PMCID: PMC8977791 DOI: 10.1136/jitc-2021-004114] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
Background Targeting the PD-1/PD-L1/L2 (programmed cell death protein 1/programmed cell death ligand 1/ligand 2) pathway combined with other immunosuppressive signalings, such as CD73/A2aR (A2a adenosine receptor) adenosine signaling, has emerged as a promising strategy for cancer treatment. The genetic characteristics of these immune checkpoints need to be further investigated in diffuse large B-cell lymphoma (DLBCL). Methods We performed whole-exome sequencing/targeted deep sequencing to investigate the genetic characteristics of PD-1/PD-L1/L2 and CD73/A2aR. The immunosuppressive effect of these two pathways on the tumor microenvironment was evaluated via RNA sequencing. Single-cell RNA sequencing was further applied to investigate the dysfunctional CD8+ T cells. In addition, multiplex immunofluorescence staining was used to quantitatively assess the expression of dysfunctional CD8+ T cells in DLBCL. Results SP140 was identified as a novel translocation partner for PD-L1, and a new inversion was detected between PD-L1 and PD-L2, both leading to the upregulation of PD-L1 expression. CD73 genetic mutations did not increase mRNA and protein expression. Patients with genetically altered CD73 tended to have a better overall survival than patients with wild-type CD73. Both PD-1/PD-L1 and CD73/A2aR signaling mediated the immunosuppressive microenvironment in DLBCL. The numbers of CD8+ T cells with PD-1 and A2aR expression were positively correlated with the number of dysfunctional CD8+ T cells (R2=0.974, p=0.013). According to the grades of dysfunctional CD8+ T cells we defined, grade 1 dysfunctional CD8+ T cells, with either PD-1+ or A2aR+, were significantly associated with poorer survival than grade 0 dysfunctional CD8+ T cells, with both PD-1− and A2aR−; and patients with grade 2 dysfunctional CD8+ T cells showed the worst clinical outcomes. Conclusions This study describes the additional genetic basis of PD-L1 overexpression and characterizes certain genetic alterations of CD73/A2aR in DLBCL. The degree of T-cell dysfunction is correlated with clinical outcomes. Strategies that reverse T-cell dysfunction by inhibiting PD-1/PD-L1/L2, particularly in combination with CD73/A2aR, may show potential as effective therapeutic options for DLBCL.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Hengqi Liu
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lei Jiao
- Panovue Biological Technology Co., Ltd, Beijing, China
| | - Zhenzhen Zhang
- Marvel Medical Laboratory, Tianjin Marvelbio Technology Co., Ltd, Tianjin, China
| | - Jin He
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lanfang Li
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lihua Qiu
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zhengzi Qian
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Shiyong Zhou
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Wenchen Gong
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bin Meng
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- Department of Immunology/Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Huilai Zhang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xianhuo Wang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| |
Collapse
|
27
|
Van der Moeren N, Selhorst P, Ha M, Heireman L, Van Gaal PJ, Breems D, Meysman P, Laukens K, Verstrepen W, Van Gasse N, Ogunjimi B, Arien KK, Naesens R. Viral Evolution and Immunology of SARS-CoV-2 in a Persistent Infection after Treatment with Rituximab. Viruses 2022; 14:v14040752. [PMID: 35458482 PMCID: PMC9032773 DOI: 10.3390/v14040752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 12/26/2022] Open
Abstract
Background. Prolonged shedding of SARS-CoV-2 in immunocompromised patients has been described. Furthermore, an accumulation of mutations of the SARS-CoV-2 genome in these patients has been observed. Methods. We describe the viral evolution, immunologic response and clinical course of a patient with a lymphoma in complete remission who had received therapy with rituximab and remained SARS-CoV-2 RT-qPCR positive for 161 days. Results. The patient remained hospitalised for 10 days, after which he fully recovered and remained asymptomatic. A progressive increase in Ct-value, coinciding with a progressive rise in lymphocyte count, was seen from day 137 onward. Culture of a nasopharyngeal swab on day 67 showed growth of SARS-CoV-2. Whole genome sequencing (WGS) demonstrated that the virus belonged to the wildtype SARS-CoV-2 clade 20B/GR, but rapidly accumulated a high number of mutations as well as deletions in the N-terminal domain of its spike protein. Conclusion. SARS-CoV-2 persistence in immunocompromised individuals has important clinical implications, but halting immunosuppressive therapy might result in a favourable clinical course. The long-term shedding of viable virus necessitates customized infection prevention measures in these individuals. The observed accelerated accumulation of mutations of the SARS-CoV-2 genome in these patients might facilitate the origin of new VOCs that might subsequently spread in the general community.
Collapse
Affiliation(s)
- Nathalie Van der Moeren
- Laboratory of Microbiology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium; (L.H.); (W.V.); (N.V.G.); (R.N.)
- Correspondence:
| | - Philippe Selhorst
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium; (P.S.); (K.K.A.)
| | - My Ha
- Centre for Health Economics Research & Modelling Infectious Diseases (CHERMID), Antwerp Centre for Translational Immunology and Virology (ACTIV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Department of Paediatrics, Antwerp University, 2610 Antwerp, Belgium; (M.H.); (P.M.); (K.L.); (B.O.)
| | - Laura Heireman
- Laboratory of Microbiology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium; (L.H.); (W.V.); (N.V.G.); (R.N.)
| | - Pieter-Jan Van Gaal
- Departement of Nephrology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium;
| | - Dimitri Breems
- Department of Haematology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium;
| | - Pieter Meysman
- Centre for Health Economics Research & Modelling Infectious Diseases (CHERMID), Antwerp Centre for Translational Immunology and Virology (ACTIV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Department of Paediatrics, Antwerp University, 2610 Antwerp, Belgium; (M.H.); (P.M.); (K.L.); (B.O.)
| | - Kris Laukens
- Centre for Health Economics Research & Modelling Infectious Diseases (CHERMID), Antwerp Centre for Translational Immunology and Virology (ACTIV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Department of Paediatrics, Antwerp University, 2610 Antwerp, Belgium; (M.H.); (P.M.); (K.L.); (B.O.)
| | - Walter Verstrepen
- Laboratory of Microbiology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium; (L.H.); (W.V.); (N.V.G.); (R.N.)
| | - Natasja Van Gasse
- Laboratory of Microbiology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium; (L.H.); (W.V.); (N.V.G.); (R.N.)
| | - Benson Ogunjimi
- Centre for Health Economics Research & Modelling Infectious Diseases (CHERMID), Antwerp Centre for Translational Immunology and Virology (ACTIV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Department of Paediatrics, Antwerp University, 2610 Antwerp, Belgium; (M.H.); (P.M.); (K.L.); (B.O.)
| | - Kevin K. Arien
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, 2000 Antwerp, Belgium; (P.S.); (K.K.A.)
| | - Reinout Naesens
- Laboratory of Microbiology, Ziekenhuis Netwerk Antwerpen, 2050 Antwerp, Belgium; (L.H.); (W.V.); (N.V.G.); (R.N.)
| |
Collapse
|
28
|
Liechti T, Iftikhar Y, Mangino M, Beddall M, Goss CW, O'Halloran JA, Mudd P, Roederer M. Immune phenotypes that predict COVID-19 severity. RESEARCH SQUARE 2022:rs.3.rs-1378671. [PMID: 35291290 PMCID: PMC8923110 DOI: 10.21203/rs.3.rs-1378671/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Severe COVID-19 causes profound immune perturbations, but pre-infection immune signatures contributing to severe COVID-19 remain unknown. Genome-wide association studies (GWAS) identified strong associations between severe disease and several chemokine receptors and molecules from the type I interferon pathway. Here, we define immune signatures associated with severe COVID-19 using high-dimensional flow cytometry. We measured the peripheral immune system from individuals who recovered from mild, moderate, severe or critical COVID-19 and focused only on those immune signatures returning to steady-state. Individuals that suffered from severe COVID-19 showed reduced frequencies of T cell, MAIT cell and dendritic cell (DCs) subsets and altered chemokine receptor expression on several subsets, such as reduced levels of CCR1 and CCR2 on monocyte subsets. Furthermore, we found reduced frequencies of type I interferon-producing plasmacytoid DCs and altered IFNAR2 expression on several myeloid cells in individuals recovered from severe COVID-19. Thus, these data identify potential immune mechanisms contributing to severe COVID-19.
Collapse
Affiliation(s)
- Thomas Liechti
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, USA, 20892
| | - Yaser Iftikhar
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, USA, 20892
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College of London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London SE1 9RT, UK
| | - Margaret Beddall
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, USA, 20892
| | - Charles W Goss
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jane A O'Halloran
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Mudd
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO, USA, 63110
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, USA, 20892
| |
Collapse
|
29
|
Mi Z, Wang Z, Xue X, Liu T, Wang C, Sun L, Yu G, Zhang Y, Shi P, Sun Y, Yang Y, Ma S, Wang Z, Yu Y, Liu J, Liu H, Zhang F. The immune-suppressive landscape in lepromatous leprosy revealed by single-cell RNA sequencing. Cell Discov 2022; 8:2. [PMID: 35013182 PMCID: PMC8748782 DOI: 10.1038/s41421-021-00353-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022] Open
Abstract
Lepromatous leprosy (L-LEP), caused by the massive proliferation of Mycobacterium leprae primarily in macrophages, is an ideal disease model for investigating the molecular mechanism of intracellular bacteria evading or modulating host immune response. Here, we performed single-cell RNA sequencing of both skin biopsies and peripheral blood mononuclear cells (PBMCs) of L-LEP patients and healthy controls. In L-LEP lesions, we revealed remarkable upregulation of APOE expression that showed a negative correlation with the major histocompatibility complex II gene HLA-DQB2 and MIF, which encodes a pro-inflammatory and anti-microbial cytokine, in the subset of macrophages exhibiting a high expression level of LIPA. The exhaustion of CD8+ T cells featured by the high expression of TIGIT and LAG3 in L-LEP lesions was demonstrated. Moreover, remarkable enhancement of inhibitory immune receptors mediated crosstalk between skin immune cells was observed in L-LEP lesions. For PBMCs, a high expression level of APOE in the HLA-DRhighFBP1high monocyte subset and the expansion of regulatory T cells were found to be associated with L-LEP. These findings revealed the primary suppressive landscape in the L-LEP patients, providing potential targets for the intervention of intracellular bacteria caused persistent infections.
Collapse
Affiliation(s)
- Zihao Mi
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Zhenzhen Wang
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Xiaotong Xue
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Tingting Liu
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Chuan Wang
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Lele Sun
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Gongqi Yu
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Yuan Zhang
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Peidian Shi
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Yonghu Sun
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Yongliang Yang
- grid.460018.b0000 0004 1769 9639Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong China
| | - Shanshan Ma
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Zhe Wang
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Yueqian Yu
- grid.410587.fShandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong China
| | - Jianjun Liu
- grid.418377.e0000 0004 0620 715XHuman Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - Hong Liu
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Furen Zhang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
30
|
Guo Z, Zhao Y, Zhang Z, Li Y. Interleukin-10-Mediated Lymphopenia Caused by Acute Infection with Foot-and-Mouth Disease Virus in Mice. Viruses 2021; 13:v13122358. [PMID: 34960627 PMCID: PMC8708299 DOI: 10.3390/v13122358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/06/2021] [Accepted: 11/20/2021] [Indexed: 12/14/2022] Open
Abstract
Foot-and-mouth disease (FMD) is characterized by a pronounced lymphopenia that is associated with immune suppression. However, the mechanisms leading to lymphopenia remain unclear. In this study, the number of total CD4+, CD8+ T cells, B cells, and NK cells in the peripheral blood were dramatically reduced in C57BL/6 mice infected with foot-and-mouth disease virus (FMDV) serotype O, and it was noted that mice with severe clinical symptoms had expressively lower lymphocyte counts than mice with mild or without clinical symptoms, indicating that lymphopenia was associated with disease severity. A further analysis revealed that lymphocyte apoptosis and trafficking occurred after FMDV infection. In addition, coinhibitory molecules were upregulated in the expression of CD4+ and CD8+ T cells from FMDV-infected mice, including CTLA-4, LAG-3, 2B4, and TIGIT. Interestingly, the elevated IL-10 in the serum was correlated with the appearance of lymphopenia during FMDV infection but not IL-6, IL-2, IL-17, IL-18, IL-1β, TNF-α, IFN-α/β, TGF-β, and CXCL1. Knocking out IL-10 (IL-10-/-) mice or blocking IL-10/IL-10R signaling in vivo was able to prevent lymphopenia via downregulating apoptosis, trafficking, and the coinhibitory expression of lymphocytes in the peripheral blood, which contribute to enhance the survival of mice infected with FMDV. Our findings support that blocking IL-10/IL-10R signaling may represent a novel therapeutic approach for FMD.
Collapse
Affiliation(s)
- Zijing Guo
- State Key Laboratory on Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Z.G.); (Y.Z.)
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Yin Zhao
- State Key Laboratory on Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730030, China; (Z.G.); (Y.Z.)
| | - Zhidong Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
- Correspondence: (Z.Z.); (Y.L.); Tel.: +86-028-85528276 (Y.L.)
| | - Yanmin Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
- Correspondence: (Z.Z.); (Y.L.); Tel.: +86-028-85528276 (Y.L.)
| |
Collapse
|
31
|
Rotte A, Sahasranaman S, Budha N. Targeting TIGIT for Immunotherapy of Cancer: Update on Clinical Development. Biomedicines 2021; 9:1277. [PMID: 34572463 PMCID: PMC8472042 DOI: 10.3390/biomedicines9091277] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint blockers have dramatically improved the chances of survival in patients with metastatic cancer, but only a subset of the patients respond to treatment. Search for novel targets that can improve the responder rates and overcome the limitations of adverse events commonly seen with combination therapies, like PD-1 plus CTLA-4 blockade and PD-1/PD-L1 plus chemotherapy, led to the development of monoclonal antibodies blocking T-cell immunoglobulin and ITIM domain (TIGIT), a inhibitory checkpoint receptor expressed on activated T cells and NK cells. The strategy showed potential in pre-clinical and early clinical studies, and 5 molecules are now in advanced stages of evaluation (phase II and above). This review aims to provide an overview of clinical development of anti-TIGIT antibodies and describes the factors considered and thought process during early clinical development. Critical aspects that can decide the fate of clinical programs, such as origin of the antibody, Ig isotype, FCγR binding, and the dose as well as dosing schedule, are discussed along with the summary of available efficacy and safety data from clinical studies and the challenges in the development of anti-TIGIT antibodies, such as identifying patients who can benefit from therapy and getting payer coverage.
Collapse
Affiliation(s)
- Anand Rotte
- Arcellx, Gaithersburg, MD 20878, USA
- Doloxe, Santa Clara, CA 95050, USA
| | | | | |
Collapse
|
32
|
Li H, Zhang J, Su C, Tian X, Mei X, Zhang Z, Wang M, Li X, Wang S. Dynamic Expressions of TIGIT on Splenic T Cells and TIGIT-Mediated Splenic T Cell Dysfunction of Mice With Chronic Toxoplasma gondii Infection. Front Microbiol 2021; 12:700892. [PMID: 34421855 PMCID: PMC8375303 DOI: 10.3389/fmicb.2021.700892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/12/2021] [Indexed: 12/03/2022] Open
Abstract
As an immunosuppressive receptor, T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) play a critical part in cellular immune regulation mediated by pathogen infection. Whereas, TIGIT expression on splenic T cells in hosts infected with Toxoplasma gondii cysts has not been studied. In this study, we detected TIGIT expression and the changes of immune function in the spleen by flow cytometry and real-time PCR (RT-PCR). We found that TIGIT expression on splenic T cells increased significantly post infection. At the same time, splenic TIGIT+TCM cells were activated and transformed into TIGIT+TEM cells during the infection, and the cytotoxicity of TIGIT+ T cells was reduced in the later stage of infection. This study shows that chronic T. gondii infection can upregulate TIGIT expression on the surface of T cells and affect immune cell function.
Collapse
Affiliation(s)
- Haoran Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jing Zhang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Changwei Su
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaowei Tian
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xuefang Mei
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhenchao Zhang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Mingyong Wang
- Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Xiangrui Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shuai Wang
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
33
|
McCulloch TR, Wells TJ, Souza-Fonseca-Guimaraes F. Towards efficient immunotherapy for bacterial infection. Trends Microbiol 2021; 30:158-169. [PMID: 34253452 DOI: 10.1016/j.tim.2021.05.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
The emergence of multiantibiotic-resistant bacteria, often referred to as superbugs, is leading to infections that are increasingly difficult to treat. Further, bacteria have evolved mechanisms by which they subvert the immune response, meaning that even antibiotic-sensitive bacteria can persist through antibiotic therapy. For these reasons, a broad range of viable therapeutic alternatives or conjunctions to traditional antimicrobial therapy are urgently required to reduce the burden of disease threatened by antibiotic resistance. Immunotherapy has emerged as a leading treatment option in cancer, and researchers are now attempting to apply this to infectious disease. This review summarizes and discusses the recent advances in the field and highlights current and future perspectives of using immunotherapies to treat bacterial infections.
Collapse
Affiliation(s)
- Timothy R McCulloch
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Timothy J Wells
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | | |
Collapse
|
34
|
Pieren DKJ, Smits NAM, Hoeboer J, Kandiah V, Postel RJ, Mariman R, Beek J, Baarle D, Wit J, Guichelaar T. Regulatory KIR + RA + T cells accumulate with age and are highly activated during viral respiratory disease. Aging Cell 2021; 20:e13372. [PMID: 34043881 PMCID: PMC8208794 DOI: 10.1111/acel.13372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/18/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Severe respiratory viral infectious diseases such as influenza and COVID‐19 especially affect the older population. This is partly ascribed to diminished CD8+ T‐cell responses a result of aging. The phenotypical diversity of the CD8+ T‐cell population has made it difficult to identify the impact of aging on CD8+ T‐cell subsets associated with diminished CD8+ T‐cell responses. Here we identify a novel human CD8+ T‐cell subset characterized by expression of Killer‐cell Immunoglobulin‐like Receptors (KIR+) and CD45RA (RA+). These KIR+RA+ T cells accumulated with age in the blood of healthy individuals (20–82 years of age, n = 50), expressed high levels of aging‐related markers of T‐cell regulation, and were functionally capable of suppressing proliferation of other CD8+ T cells. Moreover, KIR+RA+ T cells were a major T‐cell subset becoming activated in older adults suffering from an acute respiratory viral infection (n = 36), including coronavirus and influenza virus infection. In addition, older adults with influenza A infection showed that higher activation status of their KIR+RA+ T cells associated with longer duration of respiratory symptoms. Together, our data indicate that KIR+RA+ T cells are a unique human T‐cell subset with regulatory properties that may explain susceptibility to viral respiratory disease at old age.
Collapse
Affiliation(s)
- Daan K. J. Pieren
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Noortje A. M. Smits
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Jeroen Hoeboer
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Vinitha Kandiah
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Rimke J. Postel
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Rob Mariman
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Josine Beek
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Debbie Baarle
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
- Center for Translational Immunology University Medical Center Utrecht Utrecht University Utrecht The Netherlands
| | - Jelle Wit
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Teun Guichelaar
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| |
Collapse
|
35
|
Künzli M, Reuther P, Pinschewer DD, King CG. Opposing effects of T cell receptor signal strength on CD4 T cells responding to acute versus chronic viral infection. eLife 2021; 10:61869. [PMID: 33684030 PMCID: PMC7943189 DOI: 10.7554/elife.61869] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
A hallmark of adaptive immunity is CD4 T cells’ ability to differentiate into specialized effectors. A long-standing question is whether T cell receptor (TCR) signal strength can dominantly instruct the development of Th1 and T follicular helper (Tfh) cells across distinct infectious contexts. We characterized the differentiation of murine CD4 TCR transgenic T cells responding to altered peptide ligand lymphocytic choriomeningitis viruses (LCMV) derived from acute and chronic parental strains. We found that TCR signal strength exerts opposite and hierarchical effects on the balance of Th1 and Tfh cells responding to acute versus persistent infection. TCR signal strength correlates positively with Th1 generation during acute but negatively during chronic infection. Weakly activated T cells express lower levels of markers associated with chronic T cell stimulation and may resist functional inactivation. We anticipate that the panel of recombinant viruses described herein will be valuable for investigating a wide range of CD4 T cell responses.
Collapse
Affiliation(s)
- Marco Künzli
- Immune Cell Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Peter Reuther
- Division of Experimental Virology, Department of Biomedicine - Haus Petersplatz, University of Basel, Basel, Switzerland
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine - Haus Petersplatz, University of Basel, Basel, Switzerland
| | - Carolyn G King
- Immune Cell Biology Laboratory, Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
36
|
Sun Y, Anyalebechi JC, Sun H, Yumoto T, Xue M, Liu D, Liang Z, Coopersmith CM, Ford ML. Anti-TIGIT differentially affects sepsis survival in immunologically experienced versus previously naive hosts. JCI Insight 2021; 6:141245. [PMID: 33682797 PMCID: PMC8021109 DOI: 10.1172/jci.insight.141245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 01/20/2021] [Indexed: 12/29/2022] Open
Abstract
Mounting evidence suggests that the balance of T cell costimulatory and coinhibitory signals contributes to mortality during sepsis. Here, we identified a critical role of the coinhibitory molecule T cell Ig and ITIM domain (TIGIT) in regulating sepsis mortality. Because TIGIT is significantly upregulated on memory T cells, we developed a "memory mouse" model to study the role of TIGIT during sepsis in a more physiologically relevant context. Mice received sequential pathogen exposure and developed memory T cell frequencies, similar to those observed in adult humans, and were then subjected to sepsis induction via cecal ligation and puncture. Our results show that targeting the TIGIT pathway during sepsis is fundamentally different in previously naive versus memory mice, in that αTIGIT Ab had no effect on survival in previously naive septic mice but sharply worsened survival in memory septic mice. Mechanistically, αTIGIT increased apoptosis of memory T cells, decreased T cell function, and downregulated the costimulatory receptor DNAM on memory CD8+ T cells in memory septic mice, but not in previously naive septic mice. Additionally, αTIGIT diminished Helios expression in Tregs in memory but not previously naive septic mice. These data highlight fundamental differences in the pathophysiological impact of targeting TIGIT in immunologically experienced versus previously naive hosts during sepsis.
Collapse
Affiliation(s)
- Yini Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jerome C. Anyalebechi
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - He Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Hepatobiliary Surgery and Transplantation, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tetsuya Yumoto
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ming Xue
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Danya Liu
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
37
|
Greisen SR, Deleuran B. Checkpoint Molecules in Rheumatology-or the Benefits of Being Exhausted. Curr Rheumatol Rep 2021; 23:22. [PMID: 33651184 DOI: 10.1007/s11926-021-00991-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW This review will focus on the most common co-inhibitory molecules, emphasizing the importance of these in relation to rheumatic disease. RECENT FINDINGS Checkpoint molecules are pivotal in determining the outcome of antigen activation. Checkpoint molecules consist of co-stimulatory and co-inhibitory molecules, where the first activates and the latter inhibits the antigen presentation process. Studies show that increased activity of co-inhibitory molecules is associated with a good prognosis in rheumatic diseases. Opposite, when cancer patients are treated with antibodies blocking the inhibitory pathways, autoimmune diseases, including arthritis, develop as immune-related adverse events (IrAE). This emphasizes the importance of these pathways in autoimmune disease. Co-inhibitory molecules are becoming increasingly interesting as future treatment targets in rheumatic conditions. Treatments with antibodies blocking these pathways result in IrAE, often manifesting as autoimmune rheumatic diseases. Therefore, a need to get acquainted with these molecules is growing so we can cope with future challenges in rheumatic diseases.
Collapse
Affiliation(s)
- Stinne Ravn Greisen
- Department of Biomedicine, Aarhus University, Skou-building, C.F. Møllers Alle 6, DK-8000, Aarhus C, Denmark. .,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Skou-building, C.F. Møllers Alle 6, DK-8000, Aarhus C, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
38
|
Barragué H, Fontaine J, Abravanel F, Mauré E, Péron JM, Alric L, Dubois M, Izopet J, Champagne E. Mobilization of γδ T Cells and IL-10 Production at the Acute Phase of Hepatitis E Virus Infection in Cytomegalovirus Carriers. THE JOURNAL OF IMMUNOLOGY 2021; 206:1027-1038. [PMID: 33483348 DOI: 10.4049/jimmunol.2000187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022]
Abstract
Alterations in the γδ T cell compartment have been reported in immunocompromised individuals infected with hepatitis E virus (HEV)-g3. We now report the analysis of blood γδ T cells from acutely HEV-infected individuals in the absence of immunosuppression. In these patients, non-Vδ2 (ND2) γδ T cells outnumbered otherwise predominant Vδ2 cells selectively in human CMV (HCMV)-seropositive patients and were higher than in HCMVpos controls, mimicking HCMV reactivation, whereas their serum was PCR-negative for HCMV. Stimulation of their lymphocytes with HEV-infected hepatocarcinoma cells led to an HEV-specific response in γδ subsets of HCMVpos individuals. HEV infection was associated with a lowered expression of TIGIT, LAG-3, and CD160 immune checkpoint markers on ND2 effector memory cells in HCMVneg but not in HCMVpos HEV patients. γδ cell lines, predominantly ND2, were generated from patients after coculture with hepatocarcinoma cells permissive to HEV and IL-2/12/18. Upon restimulation with HEV-infected or uninfected cells and selected cytokines, these cell lines produced IFN-γ and IL-10, the latter being induced by IL-12 in IFN-γ-producing cells and upregulated by HEV and IL-18. They were also capable of suppressing the proliferation of CD3/CD28-activated CD4 cells in transwell experiments. Importantly, IL-10 was detected in the plasma of 10 of 10 HCMVpos HEV patients but rarely in controls or HCMVneg HEV patients, implying that γδ cells are probably involved in IL-10 production at the acute phase of infection. Our data indicate that HEV mobilizes a pool of ND2 memory cells in HCMV carriers, promoting the development of an immunoregulatory environment.
Collapse
Affiliation(s)
- Hugo Barragué
- Université Toulouse III Paul-Sabatier, F-31024 Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, INSERM-U1043, CNRS-UMR5282, F-31024 Toulouse, France
| | - Jessica Fontaine
- Université Toulouse III Paul-Sabatier, F-31024 Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, INSERM-U1043, CNRS-UMR5282, F-31024 Toulouse, France
| | - Florence Abravanel
- Centre de Physiopathologie de Toulouse Purpan, INSERM-U1043, CNRS-UMR5282, F-31024 Toulouse, France.,CHU Toulouse, Hôspital Purpan, Laboratoire de Virologie, Centre National de Référence Hépatite E, F-31059 Toulouse, France; and
| | - Emilie Mauré
- Centre de Physiopathologie de Toulouse Purpan, INSERM-U1043, CNRS-UMR5282, F-31024 Toulouse, France
| | - Jean-Marie Péron
- Pôle Hospitalo-Universitaire des Maladies de l'Appareil Digestif, Hôspital Rangueil, F-31059 Toulouse, France
| | - Laurent Alric
- Pôle Hospitalo-Universitaire des Maladies de l'Appareil Digestif, Hôspital Rangueil, F-31059 Toulouse, France
| | - Martine Dubois
- CHU Toulouse, Hôspital Purpan, Laboratoire de Virologie, Centre National de Référence Hépatite E, F-31059 Toulouse, France; and
| | - Jacques Izopet
- Université Toulouse III Paul-Sabatier, F-31024 Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, INSERM-U1043, CNRS-UMR5282, F-31024 Toulouse, France.,CHU Toulouse, Hôspital Purpan, Laboratoire de Virologie, Centre National de Référence Hépatite E, F-31059 Toulouse, France; and
| | - Eric Champagne
- Université Toulouse III Paul-Sabatier, F-31024 Toulouse, France; .,Centre de Physiopathologie de Toulouse Purpan, INSERM-U1043, CNRS-UMR5282, F-31024 Toulouse, France
| |
Collapse
|
39
|
Liang R, Zhu X, Lan T, Ding D, Zheng Z, Chen T, Huang Y, Liu J, Yang X, Shao J, Wei H, Wei B. TIGIT promotes CD8 +T cells exhaustion and predicts poor prognosis of colorectal cancer. Cancer Immunol Immunother 2021; 70:2781-2793. [PMID: 33634371 DOI: 10.1007/s00262-021-02886-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022]
Abstract
TIGIT is a lymphocyte surface receptor, which is mainly expressed on the surface of CD8+T cells. The role of TIGIT in colorectal cancer and its expression pattern in colorectal cancer infiltrating lymphocytes are still controversial. This study aimed at identifying the function of TIGIT in colorectal cancer. Patients with colorectal cancer showed significantly higher TIGIT+CD8+T cell infiltration in tumor tissues, metastases compared with paired PBMC and normal tissues through flow cytometry. TIGIT+CD8+T cells showed an exhausted phenotype and expressed low levels of killer cytokines IFN-γ, IL-2, TNF-α. In addition, more inhibitory receptors such as PD-1, LAG-3, and TIM-3 were expressed on the surface of TIGIT+CD8+T cells. TGF-β1 could promote the expression of TIGIT and inhibit CD8+T cell function in vitro. Moreover, the accumulation of TIGIT+T cells in tumors was associated with advanced disease, predicted early recurrence, and reduced survival rates in colorectal cancer patients. Our results indicate that TIGIT can be a biological marker for the prognosis of colorectal cancer, and TIGIT can be used as a potential target for treatment.
Collapse
Affiliation(s)
- Rongpu Liang
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Xudong Zhu
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Tianyun Lan
- Central Laboratory, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Dongbing Ding
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Zongheng Zheng
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Tufeng Chen
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Yong Huang
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jianpei Liu
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Xiaofeng Yang
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jun Shao
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China.
| | - Bo Wei
- Department of Gastrointestinal Surgery, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China.
| |
Collapse
|
40
|
Panetti C, Kao KC, Joller N. Dampening antiviral immunity can protect the host. FEBS J 2021; 289:634-646. [PMID: 33570771 PMCID: PMC9292735 DOI: 10.1111/febs.15756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
Viral infections are very common, and in most cases, the virus is well controlled and eliminated by the immune system. Nevertheless, in some cases, damage of the host tissue inflicted by the virus itself or by the elicited immune response may result in severe disease courses. Thus, regulatory mechanisms are necessary to control virus‐induced and immune pathology. This ensures immune responses are elicited in a potent but controlled manner. In this review, we will outline how immune regulation may contribute to this process. We focus on regulatory T cells and co‐inhibitory receptors and outline how these two regulatory immune components allow for and may even promote potent but not pathologic immune responses. By enabling a balanced immune response, regulatory mechanisms can thus contribute to pathogen control as well as tissue and host protection.
Collapse
Affiliation(s)
- Camilla Panetti
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Kung-Chi Kao
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, Switzerland
| |
Collapse
|
41
|
Poorebrahim M, Melief J, Pico de Coaña Y, L Wickström S, Cid-Arregui A, Kiessling R. Counteracting CAR T cell dysfunction. Oncogene 2021; 40:421-435. [PMID: 33168929 PMCID: PMC7808935 DOI: 10.1038/s41388-020-01501-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 02/08/2023]
Abstract
In spite of high rates of complete remission following chimeric antigen receptor (CAR) T cell therapy, the efficacy of this approach is limited by generation of dysfunctional CAR T cells in vivo, conceivably induced by immunosuppressive tumor microenvironment (TME) and excessive antigen exposure. Exhaustion and senescence are two critical dysfunctional states that impose a pivotal hurdle for successful CAR T cell therapies. Recently, modified CAR T cells with an "exhaustion-resistant" phenotype have shown superior antitumor functions and prolonged lifespan. In addition, several studies have indicated the feasibility of senescence delay in CAR T cells. Here, we review the latest reports regarding blockade of CAR T cell exhaustion and senescence with a particular focus on the exhaustion-inducing pathways. Subsequently, we describe what potential these latest insights offer for boosting the potency of adoptive cell transfer (ACT) therapies involving CAR T cells. Furthermore, we discuss how induction of costimulation, cytokine exposure, and TME modulation can impact on CAR T cell efficacy and persistence, while potential safety issues associated with reinvigorated CAR T cells will also be addressed.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden. .,Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jeroen Melief
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yago Pico de Coaña
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stina L Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
42
|
TCR Transgenic Mice: A Valuable Tool for Studying Viral Immunopathogenesis Mechanisms. Int J Mol Sci 2020; 21:ijms21249690. [PMID: 33353154 PMCID: PMC7765986 DOI: 10.3390/ijms21249690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 01/07/2023] Open
Abstract
Viral infectious diseases are a significant burden on public health and the global economy, and new viral threats emerge continuously. Since CD4+ and CD8+ T cell responses are essential to eliminating viruses, it is important to understand the underlying mechanisms of anti-viral T cell-mediated immunopathogenesis during viral infections. Remarkable progress in transgenic (Tg) techniques has enabled scientists to more readily understand the mechanisms of viral pathogenesis. T cell receptor (TCR) Tg mice are extremely useful in studying T cell-mediated immune responses because the majority of T cells in these mice express specific TCRs for partner antigens. In this review, we discuss the important studies utilizing TCR Tg mice to unveil underlying mechanisms of T cell-mediated immunopathogenesis during viral infections.
Collapse
|
43
|
Immune Checkpoints in Viral Infections. Viruses 2020; 12:v12091051. [PMID: 32967229 PMCID: PMC7551039 DOI: 10.3390/v12091051] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
As evidence has mounted that virus-infected cells, such as cancer cells, negatively regulate the function of T-cells via immune checkpoints, it has become increasingly clear that viral infections similarly exploit immune checkpoints as an immune system escape mechanism. Although immune checkpoint therapy has been successfully used in cancer treatment, numerous studies have suggested that such therapy may also be highly relevant for treating viral infection, especially chronic viral infections. However, it has not yet been applied in this manner. Here, we reviewed recent findings regarding immune checkpoints in viral infections, including COVID-19, and discussed the role of immune checkpoints in different viral infections, as well as the potential for applying immune checkpoint blockades as antiviral therapy.
Collapse
|
44
|
Sfera A, Osorio C, Jafri N, Diaz EL, Campo Maldonado JE. Intoxication With Endogenous Angiotensin II: A COVID-19 Hypothesis. Front Immunol 2020; 11:1472. [PMID: 32655579 PMCID: PMC7325923 DOI: 10.3389/fimmu.2020.01472] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 has spread rapidly around the globe. However, despite its high pathogenicity and transmissibility, the severity of the associated disease, COVID-19, varies widely. While the prognosis is favorable in most patients, critical illness, manifested by respiratory distress, thromboembolism, shock, and multi-organ failure, has been reported in about 5% of cases. Several studies have associated poor COVID-19 outcomes with the exhaustion of natural killer cells and cytotoxic T cells, lymphopenia, and elevated serum levels of D-dimer. In this article, we propose a common pathophysiological denominator for these negative prognostic markers, endogenous, angiotensin II toxicity. We hypothesize that, like in avian influenza, the outlook of COVID-19 is negatively correlated with the intracellular accumulation of angiotensin II promoted by the viral blockade of its degrading enzyme receptors. In this model, upregulated angiotensin II causes premature vascular senescence, leading to dysfunctional coagulation, and immunity. We further hypothesize that angiotensin II blockers and immune checkpoint inhibitors may be salutary for COVID-19 patients with critical illness by reversing both the clotting and immune defects (Graphical Abstract).
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | - Eddie Lee Diaz
- Patton State Hospital, San Bernardino, CA, United States
| | - Jose E Campo Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| |
Collapse
|