1
|
Willett BAS, Thompson SB, Chen V, Dareshouri A, Jackson CL, Brunetti T, D'Alessandro A, Klarquist J, Nemkov T, Kedl RM. Mitochondrial protein OPA1 is required for the expansion of effector CD8 T cells. Cell Rep 2025; 44:115610. [PMID: 40261796 DOI: 10.1016/j.celrep.2025.115610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
Short-lived effector cells are characterized metabolically by a highly glycolytic state, driving energy and biomass acquisition, whereas memory-fated T cells have historically been described as meeting these requirements through mitochondrial metabolism. Here, we show that the mitochondrial protein optic atrophy 1 (OPA1) is critical for rapidly dividing CD8 T cells in vivo, the requirement for which is most pronounced in effector CD8 T cells. More specifically, OPA1 supports proper cell cycle initiation and progression and the viability and survival of CD8 T cells during clonal expansion. Use of mice deficient in the mitochondrial membrane fusion proteins Mitofusin 1 and 2 (MFN1/2) in both in vivo proliferation/differentiation assays and ex vivo metabolic analysis indicates that the requirement for OPA1 during cell division supersedes its role in mitochondrial fusion. We conclude that OPA1 is critical for the generation and accumulation of short-lived effector cells that arise during the response to infection.
Collapse
Affiliation(s)
- Benjamin A S Willett
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Scott B Thompson
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vincent Chen
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anza Dareshouri
- Department of Cell and Developmental Biology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Conner L Jackson
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tonya Brunetti
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Travis Nemkov
- Department of Biochemistry & Molecular Genetics, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Xie S, Wei J, Wang X. The intersection of influenza infection and autoimmunity. Front Immunol 2025; 16:1558386. [PMID: 40248710 PMCID: PMC12003283 DOI: 10.3389/fimmu.2025.1558386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
The relationship between viral infection and autoimmune manifestations has been emerging as a significant focus of study, underscoring the intricate interplay between viral infections and the immune system. Influenza infection can result in a spectrum of clinical outcomes, ranging from mild illness to severe disease, including mortality. Annual influenza vaccination remains the most effective strategy for preventing infection and its associated complications. The complications arising from acute influenza infection are attributable not only to the direct effects of the viral infection but also to the dysregulated immune response it elicits. Notably, associations between influenza and various autoimmune diseases, such as Guillain-Barré Syndrome (GBS), Type 1 Diabetes (T1D), and antiphospholipid syndrome, have been reported. While viral infections have long been recognized as potential triggers of autoimmunity, the underlying mechanisms remain to be elucidated. Here, we described the pathophysiology caused by influenza infection and the influenza-associated autoimmune manifestations. Current advances on the understanding of the underlying immune mechanisms that lead to the potential strategies were also summarized.
Collapse
Affiliation(s)
| | | | - Xiaohui Wang
- Guangzhou Institute of Paediatrics, Guangzhou Women and Children’s Medical Center, Guangdong Provincial Research Center for Child Health, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Hildebrand JA, Daniels NR, Dehm EM, Fisher BD, Guter JK, Janse CJ, Lucas ED, Sangala JA, Tankersley TN, Hart GT, Hamilton SE. Severe malaria enforces short-lived effector cell differentiation but does not prevent effective secondary responses by memory CD8 T cells. PLoS Pathog 2025; 21:e1012993. [PMID: 40163479 PMCID: PMC11957282 DOI: 10.1371/journal.ppat.1012993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/18/2025] [Indexed: 04/02/2025] Open
Abstract
Parasitic infections are a major worldwide health burden, yet most studies of CD8 T cell differentiation focus on acute viral and bacterial infections. To understand effector and memory CD8 T cell responses during erythrocytic malaria infection in mice, we utilized transgenic OT-I T cells and compared CD8 T cell responses between infection with OVA-expressing strains of Listeria monocytogenes (Lm) and Plasmodium berghei ANKA (PbA). We find that CD8 T cells expand vigorously during both infections. However, in contrast to Lm infection, PbA infection induces T cells that are heavily biased toward an IL-7Ra-deficient and KLRG1+ short-lived effector cell (SLEC) phenotype at the expense of memory precursor effector cell (MPECs) formation. PbA-induced inflammation, including IFNγ, is partially responsible for this outcome. Following treatment with antimalarial drugs and T cell contraction, PbA-primed memory T cells are rarely found in the blood and peripheral tissues but do maintain a low presence in the spleen and bone marrow. Despite these poor numbers, PbA memory T cells robustly expand upon vaccination or viral infection, control pathogen burden, and form secondary memory pools. Thus, despite PbA enforced SLEC formation and limited memory, effective secondary responses can still proceed.
Collapse
Affiliation(s)
- Jacob A. Hildebrand
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Noah R. Daniels
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Emma M. Dehm
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Benjamin D. Fisher
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joseph K. Guter
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Erin D. Lucas
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jules A. Sangala
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Infectious Disease and Internal Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Trevor N. Tankersley
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Geoffrey T. Hart
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Infectious Disease and Internal Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Sara E. Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
4
|
Liebman LN, Shen Y, Buchwald ZS, Nepiyushchikh Z, Qi Z, García AJ, Dixon JB. Lymphatic vessel network injury reduces local tumor control despite preservation of the tumor-draining lymph node. Sci Rep 2025; 15:3485. [PMID: 39875798 PMCID: PMC11775106 DOI: 10.1038/s41598-025-85670-3] [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: 01/16/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The lymphatic system plays complex, often contradictory, roles in many cancers, including melanoma; these roles include contributions to tumor cell metastasis and immunosuppression in the tumor microenvironment as well as generation of antitumor immunity. Advancing our understanding of lymphatic vessel involvement in regulating tumor growth and immune response may provide new therapeutic targets or treatment plans to enhance the efficacy of existing therapies. We utilized a syngeneic murine melanoma model in which we surgically disrupted the lymphatic vessel network draining from the tumor to the tumor-draining lymph node (TDLN) while leaving the TDLN intact. Although transport of lymphatic-specific molecular weight tracers to the TDLN remains present after surgery, disruption of the tumor-draining lymphatic vessels results in decreased local tumor control, as reflected in an increase in the rate of tumor growth and reduction in effector-like T cell infiltration into the tumor. Our findings suggest that preservation of the functional tumor-draining lymphatic network may be essential in promoting a robust antitumor immune response.
Collapse
Affiliation(s)
- Lauren N Liebman
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yang Shen
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Zachary S Buchwald
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiation Oncology, Emory University, Atlanta, GA, USA
| | - Zhanna Nepiyushchikh
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhiming Qi
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA, 30332, USA
| | - J Brandon Dixon
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA, 30332, USA.
| |
Collapse
|
5
|
Yosri M, Dokhan M, Aboagye E, Al Moussawy M, Abdelsamed HA. Mechanisms governing bystander activation of T cells. Front Immunol 2024; 15:1465889. [PMID: 39669576 PMCID: PMC11635090 DOI: 10.3389/fimmu.2024.1465889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
The immune system is endowed with the capacity to distinguish between self and non-self, so-called immune tolerance or "consciousness of the immune system." This type of awareness is designed to achieve host protection by eliminating cells expressing a wide range of non-self antigens including microbial-derived peptides. Such a successful immune response is associated with the secretion of a whole spectrum of soluble mediators, e.g., cytokines and chemokines, which not only contribute to the clearance of infected host cells but also activate T cells that are not specific to the original cognate antigen. This kind of non-specific T-cell activation is called "bystander activation." Although it is well-established that this phenomenon is cytokine-dependent, there is evidence in the literature showing the involvement of peptide/MHC recognition depending on the type of T-cell subset (naive vs. memory). Here, we will summarize our current understanding of the mechanism(s) of bystander T-cell activation as well as its biological significance in a wide range of diseases including microbial infections, cancer, auto- and alloimmunity, and chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| | - Mohamed Dokhan
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Elizabeth Aboagye
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Mouhamad Al Moussawy
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
- Department of Physiology, Augusta University, Augusta, GA, United States
| |
Collapse
|
6
|
Mian MF, Pa S, Rahman N, Gillgrass A, Kaushic C. NET-EN treatment leads to delayed HSV-2 infection, enhanced mucin and T cell functions in the female genital tract when compared to DMPA in a preclinical mouse model. Front Immunol 2024; 15:1427842. [PMID: 39569191 PMCID: PMC11576457 DOI: 10.3389/fimmu.2024.1427842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Depot-medroxyprogesterone acetate (DMPA) and Norethisterone Enanthate (NET-EN) are progestin-only injectable contraceptives widely used by women in sub-Sharan Africa, where incidence of HIV-1 and HSV-2 infection remains high. Studies indicate that DMPA usage can increase the risk of HSV-2 infection, but limited data indicate no increased risk with use of NET-EN. We therefore investigated the effects of NET-EN and DMPA on susceptibility to vaginal HSV-2 infection in ovariectomized (OVX) mice and effects on immune responses, particularly in the vaginal tract (VT). OVX mice, when treated with NET-EN and infected intravaginally, had delayed genital pathology, decreased viral shedding, and extended survival compared to DMPA- or untreated OVX mice. CD4+ T cells isolated from VT showed no significant change in frequency with either contraceptive. However, DMPA significantly decreased the total number of VT CD4+ and CD8+ T cells and the number of IFN-γ producing CD4 and CD8 T cells and increased the percentage of CD4 and CD8 T cells producing TNF-α compared to untreated mice. In contrast, NET-EN significantly enhanced percentages of CD8+ T cells compared to DMPA treated mice, and frequencies of IFN-γ+ CD4 and CD8 T cells in the VT compared to untreated mice. Comparative analysis of splenic lymphocytes indicated that DMPA treatment resulted in reduction of CD4+ T cell frequency, but enhanced TNF-α+ CD4 T cells compared to untreated mice. NET-EN enhanced the frequency of CD8 T cells, as well as IFN-γ+ and TNF-α+ CD4, and IFN-γ+ CD8 T cells in the spleen compared to untreated mice. Importantly, we found DMPA treatment that significantly reduced mucin production, whereas NET-EN enhanced expression of cell-associated mucin in VT. High levels of mucin in NET-EN mice were associated with lower levels of HSV-2 virus detected in the vaginal tract. This study provides the first evidence that NET-EN treatment can delay HSV-2 infection compared to DMPA.
Collapse
Affiliation(s)
- M Firoz Mian
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sidney Pa
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Nuzhat Rahman
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amy Gillgrass
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
7
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
8
|
Fan J, Xu M, Liu K, Yan W, Wu H, Dong H, Yang Y, Ye W. IL-15-induced CD38 +HLA-DR +CD8 + T cells correlate with liver injury via NKG2D in chronic hepatitis B cirrhosis. Clin Transl Immunology 2024; 13:e70007. [PMID: 39416768 PMCID: PMC11480635 DOI: 10.1002/cti2.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/17/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives CD8+ T cells play a critical role in the immune dysfunction associated with liver cirrhosis. CD38+HLA-DR+CD8+ T cells, or bystander-activated CD8+ T cells, are involved in tissue injury but their specific contribution to liver cirrhosis remains unclear. This study sought to identify the mechanism for CD38+HLA-DR+CD8+ T cell-mediated pathogenesis during liver cirrhosis. Methods The immunophenotype, antigen specificity, cytokine secretion and cytotoxicity-related indicators of CD38+HLA-DR+CD8+ T cells were determined using flow cytometry. The functional properties of these cells were assessed using transcriptome analysis. CD38+HLA-DR+CD8+ T-cell killing was detected using cytotoxicity and antibody-blocking assays. Results The proportion of CD38+HLA-DR+CD8+ T cells was significantly elevated in liver cirrhosis patients and correlated with tissue damage. Transcriptome analysis revealed that these cells had innate-like functional characteristics. This CD8+ T-cell population primarily consisted of effector memory T cells and produced a high level of cytotoxicity-related cytokines, granzyme B and perforin. IL-15 promoted CD38+HLA-DR+CD8+ T-cell activation and proliferation, inducing significant TCR-independent cytotoxicity mediated through NKG2D. Conclusions CD38+HLA-DR+CD8+ T cells correlated with cirrhosis-related liver injury and contributed to liver damage by signalling through NKG2D in a TCR-independent manner.
Collapse
Affiliation(s)
- Jing Fan
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
- Clinical Research Center, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Min Xu
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Ke Liu
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Wanping Yan
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Huanyu Wu
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Hongliang Dong
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yongfeng Yang
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Wei Ye
- Department of Infectious Disease and Liver DiseaseThe Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
9
|
Sacristán C, Youngblood BA, Lu P, Bally APR, Xu JX, McGary K, Hewitt SL, Boss JM, Skok JA, Ahmed R, Dustin ML. Chronic viral infection alters PD-1 locus subnuclear localization in cytotoxic CD8 + T cells. Cell Rep 2024; 43:114547. [PMID: 39083377 PMCID: PMC11522508 DOI: 10.1016/j.celrep.2024.114547] [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: 02/14/2024] [Revised: 06/15/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
During chronic infection, virus-specific CD8+ cytotoxic T lymphocytes (CTLs) progressively lose their ability to mount effective antiviral responses. This "exhaustion" is coupled to persistent upregulation of inhibitory receptor programmed death-1 (PD-1) (Pdcd1)-key in suppressing antiviral CTL responses. Here, we investigate allelic Pdcd1 subnuclear localization and transcription during acute and chronic lymphocytic choriomeningitis virus (LCMV) infection in mice. Pdcd1 alleles dissociate from transcriptionally repressive chromatin domains (lamin B) in virus-specific exhausted CTLs but not in naive or effector CTLs. Relative to naive CTLs, nuclear positioning and Pdcd1-lamina dissociation in exhausted CTLs reflect loss of Pdcd1 promoter methylation and greater PD-1 upregulation, although a direct correlation is not observed in effector cells, 8 days post-infection. Genetic deletion of B lymphocyte-induced maturation protein 1 (Blimp-1) enhances Pdcd1-lamina dissociation in effector CTLs, suggesting that Blimp-1 contributes to maintaining Pdcd1 localization to repressive lamina. Our results identify mechanisms governing Pdcd1 subnuclear localization and the broader role of chromatin dynamics in T cell exhaustion.
Collapse
Affiliation(s)
- Catarina Sacristán
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Ben A Youngblood
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA; Immunology Department, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Peiyuan Lu
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Alexander P R Bally
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Jean Xiaojin Xu
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Katelyn McGary
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Susannah L Hewitt
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Jeremy M Boss
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Jane A Skok
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Rafi Ahmed
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA; The Kennedy Institute of Rheumatology, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Bevilacqua A, Franco F, Lu YT, Rahiman N, Kao KC, Chuang YM, Zhu Y, Held W, Xie X, Gunsalus KC, Xiao Z, Chen SY, Ho PC. PPARβ/δ-orchestrated metabolic reprogramming supports the formation and maintenance of memory CD8 + T cells. Sci Immunol 2024; 9:eadn2717. [PMID: 39178275 DOI: 10.1126/sciimmunol.adn2717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 06/03/2024] [Accepted: 07/31/2024] [Indexed: 08/25/2024]
Abstract
The formation of memory T cells is a fundamental feature of adaptative immunity, allowing the establishment of long-term protection against pathogens. Although emerging evidence suggests that metabolic reprogramming is crucial for memory T cell differentiation and survival, the underlying mechanisms that drive metabolic rewiring in memory T cells remain unclear. Here, we found that up-regulation of the nuclear receptor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) instructs the metabolic reprogramming that occurs during the establishment of central memory CD8+ T cells. PPARβ/δ-regulated changes included suppression of aerobic glycolysis and enhancement of oxidative metabolism and fatty acid oxidation. Mechanistically, exposure to interleukin-15 and expression of T cell factor 1 facilitated activation of the PPARβ/δ pathway, counteracting apoptosis induced by antigen clearance and metabolic stress. Together, our findings indicate that PPARβ/δ is a master metabolic regulator orchestrating a metabolic switch that may be favorable for T cell longevity.
Collapse
Affiliation(s)
- Alessio Bevilacqua
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Fabien Franco
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Ya-Ting Lu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Nabil Rahiman
- Center for Genomics and System Biology (CSGB), New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Kung-Chi Kao
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Yu-Ming Chuang
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Yanan Zhu
- School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, Shaanxi, China
| | - Werner Held
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Xin Xie
- Center for Genomics and System Biology (CSGB), New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China
| | - Kristin C Gunsalus
- Center for Genomics and System Biology (CSGB), New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Zhengtao Xiao
- School of Basic Medical Sciences, Xi'an Jiao Tong University Health Science Center, Xi'an, Shaanxi, China
| | - Shih-Yu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
11
|
Ganusov VV. Appropriate Sampling and Longer Follow-Up Are Required to Rigorously Evaluate Longevity of Humoral Memory After Vaccination. Immunohorizons 2024; 8:397-403. [PMID: 38864816 PMCID: PMC11220738 DOI: 10.4049/immunohorizons.2300057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
One of the goals of vaccination is to induce long-lived immunity against the infection and/or disease. Many studies have followed the generation of humoral immunity to SARS-CoV-2 after vaccination; however, such studies typically varied by the duration of the follow-up and the number of time points at which immune response measurements were done. How these parameters (the number of time points and the overall duration of the follow-up) impact estimates of immunity longevity remain largely unknown. Several studies, including one by Arunachalam et al. (2023. J. Clin. Invest. 133: e167955), evaluated the humoral immune response in individuals receiving either a third or fourth dose of mRNA COVID-19 vaccine; by measuring Ab levels at three time points (prior to vaccination and at 1 and 6 mo), Arunachalam et al. found similar half-life times for serum Abs in the two groups and thus suggested that additional boosting is unnecessary to prolong immunity to SARS-CoV-2. I demonstrate that measuring Ab levels at these three time points and only for 6 mo does not allow one to accurately evaluate the long-term half-life of vaccine-induced Abs. By using the data from a cohort of blood donors followed for several years, I show that after revaccination with vaccinia virus, vaccinia virus-specific Abs decay biphasically, and even the late decay rate exceeds the true slow loss rate of humoral memory observed years prior to the boosting. Mathematical models of Ab response kinetics, parameterized using preliminary data, should be used for power analysis to determine the most appropriate timing and duration of sampling to rigorously determine the duration of humoral immunity after vaccination.
Collapse
Affiliation(s)
- Vitaly V. Ganusov
- Address correspondence and reprint request to: Vitaly V. Ganusov, Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227-5302. E-mail address:
| |
Collapse
|
12
|
Correa-Medero LO, Jankowski SE, Hong HS, Armas ND, Vijendra AI, Reynolds MB, Fogo GM, Awad D, Dils AT, Inoki KA, Williams RG, Ye AM, Svezhova N, Gomez-Rivera F, Collins KL, O'Riordan MX, Sanderson TH, Lyssiotis CA, Carty SA. ER-associated degradation adapter Sel1L is required for CD8 + T cell function and memory formation following acute viral infection. Cell Rep 2024; 43:114156. [PMID: 38687642 PMCID: PMC11194752 DOI: 10.1016/j.celrep.2024.114156] [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: 10/09/2023] [Revised: 03/06/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
The maintenance of antigen-specific CD8+ T cells underlies the efficacy of vaccines and immunotherapies. Pathways contributing to CD8+ T cell loss are not completely understood. Uncovering the pathways underlying the limited persistence of CD8+ T cells would be of significant benefit for developing novel strategies of promoting T cell persistence. Here, we demonstrate that murine CD8+ T cells experience endoplasmic reticulum (ER) stress following activation and that the ER-associated degradation (ERAD) adapter Sel1L is induced in activated CD8+ T cells. Sel1L loss limits CD8+ T cell function and memory formation following acute viral infection. Mechanistically, Sel1L is required for optimal bioenergetics and c-Myc expression. Finally, we demonstrate that human CD8+ T cells experience ER stress upon activation and that ER stress is negatively associated with improved T cell functionality in T cell-redirecting therapies. Together, these results demonstrate that ER stress and ERAD are important regulators of T cell function and persistence.
Collapse
Affiliation(s)
- Luis O Correa-Medero
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Hanna S Hong
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas D Armas
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Mack B Reynolds
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Garrett M Fogo
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dominik Awad
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexander T Dils
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Reid G Williams
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Nadezhda Svezhova
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Kathleen L Collins
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas H Sanderson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shannon A Carty
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Mariniello A, Nasti TH, Chang DY, Hashimoto M, Malik S, McManus DT, Lee J, McGuire DJ, Cardenas MA, Umana P, Nicolini V, Antia R, Saha A, Buchwald Z, Kissick H, Ghorani E, Novello S, Sangiolo D, Scagliotti GV, Ramalingam SS, Ahmed R. Platinum-Based Chemotherapy Attenuates the Effector Response of CD8 T Cells to Concomitant PD-1 Blockade. Clin Cancer Res 2024; 30:1833-1845. [PMID: 37992307 PMCID: PMC11061601 DOI: 10.1158/1078-0432.ccr-23-1316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/27/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023]
Abstract
PURPOSE Combination of chemotherapy with programmed cell death 1 (PD-1) blockade is a front-line treatment for lung cancer. However, it remains unknown whether and how chemotherapy affects the response of exhausted CD8 T cells to PD-1 blockade. EXPERIMENTAL DESIGN We used the well-established mouse model of T-cell exhaustion with chronic lymphocytic choriomeningitis virus (LCMV) infection to assess the effect of chemotherapy (cisplatin+pemetrexed) on T-cell response to PD-1 blockade, in the absence of the impact of chemotherapy on antigen release and presentation observed in tumor models. RESULTS When concomitantly administered with PD-1 blockade, chemotherapy affected the differentiation path of LCMV-specific CD8 T cells from stem-like to transitory effector cells, thereby reducing their expansion and production of IFNγ. After combination treatment, these restrained effector responses resulted in impaired viral control, compared with PD-1 blockade alone. The sequential combination strategy, where PD-1 blockade followed chemotherapy, proved to be superior to the concomitant combination, preserving the proliferative response of exhausted CD8 T cells to PD-1 blockade. Our findings suggest that the stem-like CD8 T cells themselves are relatively unaffected by chemotherapy partly because they are quiescent and maintained by slow self-renewal at the steady state. However, upon the proliferative burst mediated by PD-1 blockade, the accelerated differentiation and self-renewal of stem-like cells may be curbed by concomitant chemotherapy, ultimately resulting in impaired overall CD8 T-cell effector functions. CONCLUSIONS In a translational context, we provide a proof-of-concept to consider optimizing the timing of chemo-immunotherapy strategies for improved CD8 T-cell functions. See related commentary by Vignali and Luke, p. 1705.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Department of Oncology, University of Torino, Turin, Italy
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Tahseen H. Nasti
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel Y. Chang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Masao Hashimoto
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Sakshi Malik
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel T. McManus
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Judong Lee
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Donald J. McGuire
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Maria A. Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Pablo Umana
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Valeria Nicolini
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, Georgia
| | - Ananya Saha
- Department of Biology, Emory University, Atlanta, Georgia
| | - Zachary Buchwald
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Hayden Kissick
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Ehsan Ghorani
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Cancer Immunology and Immunotherapy Unit, Imperial College London, Department of Surgery and Cancer, London, United Kingdom
| | - Silvia Novello
- Department of Oncology, University of Torino, Turin, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Suresh S. Ramalingam
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
14
|
Tiwari R, Singh VK, Rajneesh, Kumar A, Gautam V, Kumar R. MHC tetramer technology: Exploring T cell biology in health and disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:327-345. [PMID: 38762273 DOI: 10.1016/bs.apcsb.2024.02.002] [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: 05/20/2024]
Abstract
Major histocompatibility complex (MHC) tetramers stand as formidable tools within T cell biology, facilitating the exploration and comprehension of immune responses. These artificial molecules, comprising four bound MHC molecules, typically with a specified peptide and a fluorescent label, play a pivotal role in characterizing T cell subsets, monitoring clonal expansion, and unraveling T cell dynamics during responses to infections or immunotherapies. Beyond their applications in T cell biology, MHC tetramers prove valuable in investigating a spectrum of diseases such as infectious diseases, autoimmune disorders, and cancers. Their instrumental role extends to vaccine research and development. Notably, when appropriately configured, tetramers transcend T cell biology research and find utility in exploring natural killer T cells and contributing to specific T cell clonal deletions.
Collapse
Affiliation(s)
- Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
15
|
Sun W, Hughes EP, Kim H, Perovanovic J, Charley KR, Perkins B, Du J, Ibarra A, Syage AR, Hale JS, Williams MA, Tantin D. OCA-B/Pou2af1 is sufficient to promote CD4 + T cell memory and prospectively identifies memory precursors. Proc Natl Acad Sci U S A 2024; 121:e2309153121. [PMID: 38386711 PMCID: PMC10907311 DOI: 10.1073/pnas.2309153121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/12/2024] [Indexed: 02/24/2024] Open
Abstract
The molecular mechanisms leading to the establishment of immunological memory are inadequately understood, limiting the development of effective vaccines and durable antitumor immune therapies. Here, we show that ectopic OCA-B expression is sufficient to improve antiviral memory recall responses, while having minimal effects on primary effector responses. At peak viral response, short-lived effector T cell populations are expanded but show increased Gadd45b and Socs2 expression, while memory precursor effector cells show increased expression of Bcl2, Il7r, and Tcf7 on a per-cell basis. Using an OCA-B mCherry reporter mouse line, we observe high OCA-B expression in CD4+ central memory T cells. We show that early in viral infection, endogenously elevated OCA-B expression prospectively identifies memory precursor cells with increased survival capability and memory recall potential. Cumulatively, the results demonstrate that OCA-B is both necessary and sufficient to promote CD4 T cell memory in vivo and can be used to prospectively identify memory precursor cells.
Collapse
Affiliation(s)
- Wenxiang Sun
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Erik P. Hughes
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Heejoo Kim
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Jelena Perovanovic
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Krystal R. Charley
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Bryant Perkins
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Junhong Du
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Andrea Ibarra
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Amber R. Syage
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - J. Scott Hale
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Matthew A. Williams
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT84112
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT84112
| |
Collapse
|
16
|
Abadie K, Clark EC, Valanparambil RM, Ukogu O, Yang W, Daza RM, Ng KKH, Fathima J, Wang AL, Lee J, Nasti TH, Bhandoola A, Nourmohammad A, Ahmed R, Shendure J, Cao J, Kueh HY. Reversible, tunable epigenetic silencing of TCF1 generates flexibility in the T cell memory decision. Immunity 2024; 57:271-286.e13. [PMID: 38301652 PMCID: PMC10922671 DOI: 10.1016/j.immuni.2023.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/09/2023] [Accepted: 12/07/2023] [Indexed: 02/03/2024]
Abstract
The immune system encodes information about the severity of a pathogenic threat in the quantity and type of memory cells it forms. This encoding emerges from lymphocyte decisions to maintain or lose self-renewal and memory potential during a challenge. By tracking CD8+ T cells at the single-cell and clonal lineage level using time-resolved transcriptomics, quantitative live imaging, and an acute infection model, we find that T cells will maintain or lose memory potential early after antigen recognition. However, following pathogen clearance, T cells may regain memory potential if initially lost. Mechanistically, this flexibility is implemented by a stochastic cis-epigenetic switch that tunably and reversibly silences the memory regulator, TCF1, in response to stimulation. Mathematical modeling shows how this flexibility allows memory T cell numbers to scale robustly with pathogen virulence and immune response magnitudes. We propose that flexibility and stochasticity in cellular decisions ensure optimal immune responses against diverse threats.
Collapse
Affiliation(s)
- Kathleen Abadie
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Elisa C Clark
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Rajesh M Valanparambil
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Obinna Ukogu
- Department of Applied Mathematics, University of Washington, Seattle, WA 98105, USA
| | - Wei Yang
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Kenneth K H Ng
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jumana Fathima
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Allan L Wang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Judong Lee
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tahseen H Nasti
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Avinash Bhandoola
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - Armita Nourmohammad
- Department of Applied Mathematics, University of Washington, Seattle, WA 98105, USA; Department of Physics, University of Washington, Seattle, WA 98105, USA; Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA; Allen Discovery Center for Cell Lineage Tracing, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Junyue Cao
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY 10065, USA.
| | - Hao Yuan Kueh
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
17
|
Balint E, Feng E, Giles EC, Ritchie TM, Qian AS, Vahedi F, Montemarano A, Portillo AL, Monteiro JK, Trigatti BL, Ashkar AA. Bystander activated CD8 + T cells mediate neuropathology during viral infection via antigen-independent cytotoxicity. Nat Commun 2024; 15:896. [PMID: 38316762 PMCID: PMC10844499 DOI: 10.1038/s41467-023-44667-0] [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: 02/09/2022] [Accepted: 12/21/2023] [Indexed: 02/07/2024] Open
Abstract
Although many viral infections are linked to the development of neurological disorders, the mechanism governing virus-induced neuropathology remains poorly understood, particularly when the virus is not directly neuropathic. Using a mouse model of Zika virus (ZIKV) infection, we found that the severity of neurological disease did not correlate with brain ZIKV titers, but rather with infiltration of bystander activated NKG2D+CD8+ T cells. Antibody depletion of CD8 or blockade of NKG2D prevented ZIKV-associated paralysis, suggesting that CD8+ T cells induce neurological disease independent of TCR signaling. Furthermore, spleen and brain CD8+ T cells exhibited antigen-independent cytotoxicity that correlated with NKG2D expression. Finally, viral infection and inflammation in the brain was necessary but not sufficient to induce neurological damage. We demonstrate that CD8+ T cells mediate virus-induced neuropathology via antigen-independent, NKG2D-mediated cytotoxicity, which may serve as a therapeutic target for treatment of virus-induced neurological disease.
Collapse
Affiliation(s)
- Elizabeth Balint
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Emily Feng
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Elizabeth C Giles
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Tyrah M Ritchie
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexander S Qian
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amelia Montemarano
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ana L Portillo
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan K Monteiro
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Bernardo L Trigatti
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
18
|
Fujii SI, Shimizu K. NKT-Licensed In Vivo Dendritic Cell-Based Immunotherapy as Cellular Immunodrugs for Cancer Treatment. Crit Rev Oncog 2024; 29:45-61. [PMID: 38421713 DOI: 10.1615/critrevoncog.2023048735] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
With the advent of new therapies, immunotherapy has gained attention as a critical modality. After the discovery of the natural killer T (NKT) cells ligand, ex vivo cultured dendritic cells (DCs) loaded with NKT ligand (especially α-galactosylceramide (α-GalCer) (DC/Gal) or ex vivo expanded NKT transfer studies were clinically examined in several institutes. To prevent tumoral immune escape, the link between innate and adaptive immunity, in situ selective targeting of DCs has been attempted; however, protocol optimization was required. As a type of DC targeting therapy that combines the benefits of invariant natural killer T (iNKT) cells, we established an all-in-one, off-the-shelf drug, named the artificial adjuvant vector cell (aAVC), which consists of the tumor antigen and the CD1d-iNKT ligand complex. Here, to our knowledge, we first demonstrate the DC/GalCer therapy and NKT transfer therapy. Next, we introduce and discuss the use of aAVC therapy not only for efficient innate and adaptive immunity induction using fully matured DC in situ but also the characterization necessary for locally reprogramming the tumor microenvironment and systemically inducing long-term memory in T cells. We also discuss how the immune network mechanism is controlled by DCs. Next, we performed the first human clinical trial using WT1 antigen-expressing aAVC against relapse and refractory acute myelogenous leukemia. Thus, we highlight the challenges of using aAVCs as prodrugs for actively energizing DCs in vivo, underpinning immunological networks, and developing strategies for providing maximal benefits for patients.
Collapse
Affiliation(s)
- Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), and RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Kanagawa, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), and RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Kanagawa, Japan
| |
Collapse
|
19
|
Wheeler BD, Gagnon JD, Zhu WS, Muñoz-Sandoval P, Wong SK, Simeonov DS, Li Z, DeBarge R, Spitzer MH, Marson A, Ansel KM. The lncRNA Malat1 inhibits miR-15/16 to enhance cytotoxic T cell activation and memory cell formation. eLife 2023; 12:RP87900. [PMID: 38127070 PMCID: PMC10735224 DOI: 10.7554/elife.87900] [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] [Indexed: 12/23/2023] Open
Abstract
Proper activation of cytotoxic T cells via the T cell receptor and the costimulatory receptor CD28 is essential for adaptive immunity against viruses, intracellular bacteria, and cancers. Through biochemical analysis of RNA:protein interactions, we uncovered a non-coding RNA circuit regulating activation and differentiation of cytotoxic T cells composed of the long non-coding RNA Malat1 (Metastasis Associated Lung Adenocarcinoma Transcript 1) and the microRNA family miR-15/16. miR-15/16 is a widely and highly expressed tumor suppressor miRNA family important for cell proliferation and survival. miR-15/16 play important roles in T cell responses to viral infection, including the regulation of antigen-specific T cell expansion and memory. Comparative Argonaute-2 high-throughput sequencing of crosslinking immunoprecipitation (AHC) combined with gene expression profiling in normal and miR-15/16-deficient mouse T cells revealed a large network of hundreds of direct miR-15/16 target mRNAs, many with functional relevance for T cell activation, survival and memory formation. Among these targets, Malat1 contained the largest absolute magnitude miR-15/16-dependent AHC peak. This binding site was among the strongest lncRNA:miRNA interactions detected in the T cell transcriptome. We used CRISPR targeting with homology directed repair to generate mice with a 5-nucleotide mutation in the miR-15/16-binding site in Malat1. This mutation interrupted Malat1:miR-15/16 interaction, and enhanced the repression of other miR-15/16 target genes, including CD28. Interrupting Malat1 interaction with miR-15/16 decreased cytotoxic T cell activation, including the expression of interleukin 2 (IL-2) and a broader CD28-responsive gene program. Accordingly, Malat1 mutation diminished memory cell persistence in mice following LCMV Armstrong and Listeria monocytogenes infection. This study marks a significant advance in the study of long non-coding RNAs in the immune system by ascribing cell-intrinsic, sequence-specific in vivo function to Malat1. These findings have implications for T cell-mediated autoimmune diseases, antiviral and anti-tumor immunity, as well as lung adenocarcinoma and other malignancies where Malat1 is overexpressed.
Collapse
Affiliation(s)
- Benjamin D Wheeler
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Sandler Asthma Basic Research Program, University of California, San FranciscoSan FranciscoUnited States
| | - John D Gagnon
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Sandler Asthma Basic Research Program, University of California, San FranciscoSan FranciscoUnited States
| | - Wandi S Zhu
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Sandler Asthma Basic Research Program, University of California, San FranciscoSan FranciscoUnited States
| | - Priscila Muñoz-Sandoval
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Sandler Asthma Basic Research Program, University of California, San FranciscoSan FranciscoUnited States
| | - Simon K Wong
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
| | - Dimitre S Simeonov
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
| | - Zhongmei Li
- Gladstone-UCSF Institute of Genomic ImmunologySan FranciscoUnited States
| | - Rachel DeBarge
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Gladstone-UCSF Institute of Genomic ImmunologySan FranciscoUnited States
- Department of Otolaryngology-Head and Neck Surgery, University of California San FranciscoSan FranciscoUnited States
| | - Matthew H Spitzer
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Gladstone-UCSF Institute of Genomic ImmunologySan FranciscoUnited States
- Department of Otolaryngology-Head and Neck Surgery, University of California San FranciscoSan FranciscoUnited States
- Parker Institute for Cancer Immunotherapy, San FranciscoSan FranciscoUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Alexander Marson
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Gladstone-UCSF Institute of Genomic ImmunologySan FranciscoUnited States
- Department of Medicine, University of California San FranciscoLexingtonUnited States
| | - K Mark Ansel
- Department of Microbiology & Immunology, University of California San FranciscoSan FranciscoUnited States
- Sandler Asthma Basic Research Program, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
20
|
Khorki ME, Shi T, Cianciolo EE, Burg AR, Chukwuma PC, Picarsic JL, Morrice MK, Woodle ES, Maltzman JS, Ferguson A, Katz JD, Baker BM, Hildeman DA. Prior viral infection primes cross-reactive CD8+ T cells that respond to mouse heart allografts. Front Immunol 2023; 14:1287546. [PMID: 38143762 PMCID: PMC10748599 DOI: 10.3389/fimmu.2023.1287546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/14/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Significant evidence suggests a connection between transplant rejection and the presence of high levels of pre-existing memory T cells. Viral infection can elicit viral-specific memory T cells that cross-react with allo-MHC capable of driving allograft rejection in mice. Despite these advances, and despite their critical role in transplant rejection, a systematic study of allo-reactive memory T cells, their specificities, and the role of cross-reactivity with viral antigens has not been performed. Methods Here, we established a model to identify, isolate, and characterize cross-reactive T cells using Nur77 reporter mice (C57BL/6 background), which transiently express GFP exclusively upon TCR engagement. We infected Nur77 mice with lymphocytic choriomeningitis virus (LCMV-Armstrong) to generate a robust memory compartment, where quiescent LCMV-specific memory CD8+ T cells could be readily tracked with MHC tetramer staining. Then, we transplanted LCMV immune mice with allogeneic hearts and monitored expression of GFP within MHC-tetramer defined viral-specific T cells as an indicator of their ability to cross-react with alloantigens. Results Strikingly, prior LCMV infection significantly increased the kinetics and magnitude of rejection as well as CD8+ T cell recruitment into allogeneic, but not syngeneic, transplanted hearts, relative to non-infected controls. Interestingly, as early as day 1 after allogeneic heart transplant an average of ~8% of MHC-tetramer+ CD8+ T cells expressed GFP, in contrast to syngeneic heart transplants, where the frequency of viral-specific CD8+ T cells that were GFP+ was <1%. These data show that a significant percentage of viral-specific memory CD8+ T cells expressed T cell receptors that also recognized alloantigens in vivo. Notably, the frequency of cross-reactive CD8+ T cells differed depending upon the viral epitope. Further, TCR sequences derived from cross-reactive T cells harbored distinctive motifs that may provide insight into cross-reactivity and allo-specificity. Discussion In sum, we have established a mouse model to track viral-specific, allo-specific, and cross-reactive T cells; revealing that prior infection elicits substantial numbers of viral-specific T cells that cross-react to alloantigen, respond very early after transplant, and may promote rapid rejection.
Collapse
Affiliation(s)
- M. Eyad Khorki
- Division of Nephrology & Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Tiffany Shi
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Eileen E. Cianciolo
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Ashley R. Burg
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - P. Chukwunalu Chukwuma
- Department of Chemistry & Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States
| | - Jennifer L. Picarsic
- Division of Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Mary K. Morrice
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - E. Steve Woodle
- Division of Transplantation, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Jonathan S. Maltzman
- Department of Medicine, Stanford University, Palo Alto, CA, United States
- Geriatric Research and Education Clinical Center, Veterans Affairs (VA) Palo Alto Health Care System, Palo Alto, CA, United States
| | - Autumn Ferguson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jonathan D. Katz
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Brian M. Baker
- Department of Chemistry & Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States
| | - David A. Hildeman
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
21
|
Chen C, Zheng H, Horwitz EM, Ando S, Araki K, Zhao P, Li Z, Ford ML, Ahmed R, Qu CK. Mitochondrial metabolic flexibility is critical for CD8 + T cell antitumor immunity. SCIENCE ADVANCES 2023; 9:eadf9522. [PMID: 38055827 PMCID: PMC10699783 DOI: 10.1126/sciadv.adf9522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/07/2023] [Indexed: 12/08/2023]
Abstract
Mitochondria use different substrates for energy production and intermediatory metabolism according to the availability of nutrients and oxygen levels. The role of mitochondrial metabolic flexibility for CD8+ T cell immune response is poorly understood. Here, we report that the deletion or pharmacological inhibition of protein tyrosine phosphatase, mitochondrial 1 (PTPMT1) significantly decreased CD8+ effector T cell development and clonal expansion. In addition, PTPMT1 deletion impaired stem-like CD8+ T cell maintenance and accelerated CD8+ T cell exhaustion/dysfunction, leading to aggravated tumor growth. Mechanistically, the loss of PTPMT1 critically altered mitochondrial fuel selection-the utilization of pyruvate, a major mitochondrial substrate derived from glucose-was inhibited, whereas fatty acid utilization was enhanced. Persistent mitochondrial substrate shift and metabolic inflexibility induced oxidative stress, DNA damage, and apoptosis in PTPMT1 knockout cells. Collectively, this study reveals an important role of PTPMT1 in facilitating mitochondrial utilization of carbohydrates and that mitochondrial flexibility in energy source selection is critical for CD8+ T cell antitumor immunity.
Collapse
Affiliation(s)
- Chao Chen
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hong Zheng
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Edwin M. Horwitz
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Satomi Ando
- Department of Microbiology and Immunology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Koichi Araki
- Department of Microbiology and Immunology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Peng Zhao
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zhiguo Li
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mandy L. Ford
- Department of Surgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
22
|
Ma J, Chen X, Xue R, Wang F, Dong J, Tao N, Qin Z. Cinnamaldehyde inhibits cytokine storms induced by the ORF3a protein of SARS-CoV-2 via ROS-elimination in activated T cells. Phytother Res 2023; 37:6006-6020. [PMID: 37726983 DOI: 10.1002/ptr.8016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/04/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023]
Abstract
Cytokine storms are the cause of complications in patients with severe COVID-19, and it becomes the target of therapy. Several natural compounds were selected to screen the inhibitory effect on T-cell proliferation by Fluorescence-Activated Cell Sorting (FACS) and cytokine production by enzyme-linked immunosorbent assay (ELISA). Open reading frame 3a (ORF3a) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates the specific T-cell activation model in vivo and in vitro. The coculture system included the macrophage cell line RAW264.7 and splenocytes. Reactive oxygen species (ROS) levels and glycolysis in T cells were evaluated. Cinnamaldehyde effectively inhibits cytokine storms both in vitro and in vivo. It decreased inflammatory cytokine (such as IFN-γ, TNF-α, IL-6, and IL-2) production by murine peripheral blood cells upon direct stimulation with ConA, after immunization with the MHV-A59 virus or ORF3a peptide from SARS-CoV-2. Cinnamaldehyde restored the percentage of T cells, which was originally decreased in the peripheral blood and splenocytes of ORF3a-immunized mice. In a coculture system, cinnamaldehyde reduced the secretion of inflammatory cytokines from macrophages in a T-cell dependent manner. Furthermore, cinnamaldehyde decreased the ROS level in activated T cells, which in turn reduced glycolysis and the activation of T cells. Cinnamaldehyde can be used as a candidate molecule for COVID-19.
Collapse
Affiliation(s)
- Jing Ma
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xu Chen
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fei Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Dong
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Ning Tao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
23
|
Kingstad-Bakke B, Cleven T, Bussan H, Yount BL, Uraki R, Iwatsuki-Horimoto K, Koga M, Yamamoto S, Yotsuyanagi H, Park H, Mishra JS, Kumar S, Baric RS, Halfmann PJ, Kawaoka Y, Suresh M. Airway surveillance and lung viral control by memory T cells induced by COVID-19 mRNA vaccine. JCI Insight 2023; 8:e172510. [PMID: 37796612 PMCID: PMC10721330 DOI: 10.1172/jci.insight.172510] [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: 05/23/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
Although SARS-CoV-2 evolution seeds a continuous stream of antibody-evasive viral variants, COVID-19 mRNA vaccines provide robust protection against severe disease and hospitalization. Here, we asked whether mRNA vaccine-induced memory T cells limit lung SARS-CoV-2 replication and severe disease. We show that mice and humans receiving booster BioNTech mRNA vaccine developed potent CD8 T cell responses and showed similar kinetics of expansion and contraction of granzyme B/perforin-expressing effector CD8 T cells. Both monovalent and bivalent mRNA vaccines elicited strong expansion of a heterogeneous pool of terminal effectors and memory precursor effector CD8 T cells in spleen, inguinal and mediastinal lymph nodes, pulmonary vasculature, and most surprisingly in the airways, suggestive of systemic and regional surveillance. Furthermore, we document that: (a) CD8 T cell memory persists in multiple tissues for > 200 days; (b) following challenge with pathogenic SARS-CoV-2, circulating memory CD8 T cells rapidly extravasate to the lungs and promote expeditious viral clearance, by mechanisms that require CD4 T cell help; and (c) adoptively transferred splenic memory CD8 T cells traffic to the airways and promote lung SARS-CoV-2 clearance. These findings provide insights into the critical role of memory T cells in preventing severe lung disease following breakthrough infections with antibody-evasive SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thomas Cleven
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hailey Bussan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Boyd L. Yount
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryuta Uraki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | | | - Michiko Koga
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shinya Yamamoto
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hongtae Park
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jay S. Mishra
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter J. Halfmann
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
24
|
Dar AA, Kim DD, Gordon SM, Klinzing K, Rosen S, Guha I, Porter N, Ortega Y, Forsyth KS, Roof J, Fazelinia H, Spruce LA, Eisenlohr LC, Behrens EM, Oliver PM. c-Myc uses Cul4b to preserve genome integrity and promote antiviral CD8 + T cell immunity. Nat Commun 2023; 14:7098. [PMID: 37925424 PMCID: PMC10625626 DOI: 10.1038/s41467-023-42765-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
During infection, virus-specific CD8+ T cells undergo rapid bursts of proliferation and differentiate into effector cells that kill virus-infected cells and reduce viral load. This rapid clonal expansion can put T cells at significant risk for replication-induced DNA damage. Here, we find that c-Myc links CD8+ T cell expansion to DNA damage response pathways though the E3 ubiquitin ligase, Cullin 4b (Cul4b). Following activation, c-Myc increases the levels of Cul4b and other members of the Cullin RING Ligase 4 (CRL4) complex. Despite expressing c-Myc at high levels, Cul4b-deficient CD8+ T cells do not expand and clear the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) in vivo. Cul4b-deficient CD8+ T cells accrue DNA damage and succumb to proliferative catastrophe early after antigen encounter. Mechanistically, Cul4b knockout induces an accumulation of p21 and Cyclin E2, resulting in replication stress. Our data show that c-Myc supports cell proliferation by maintaining genome stability via Cul4b, thereby directly coupling these two interdependent pathways. These data clarify how CD8+ T cells use c-Myc and Cul4b to sustain their potential for extraordinary population expansion, longevity and antiviral responses.
Collapse
Affiliation(s)
- Asif A Dar
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Dale D Kim
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott M Gordon
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen Klinzing
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Siera Rosen
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ipsita Guha
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nadia Porter
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yohaniz Ortega
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katherine S Forsyth
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer Roof
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hossein Fazelinia
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lynn A Spruce
- Division of Cell Pathology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurence C Eisenlohr
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward M Behrens
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paula M Oliver
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Lehikoinen J, Valori M, Jääskeläinen AJ, Laakso SM, Arstila TP, Tienari PJ. High Epstein-Barr virus capsid antigen IgG level associates with the carriership of CD8+ T cell somatic mutations in the STAT3 SH2 domain. Clin Immunol 2023; 255:109733. [PMID: 37572949 DOI: 10.1016/j.clim.2023.109733] [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: 07/05/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
High carrier prevalence of STAT3 SH2 domain somatic mutations was recently discovered in CD8+ T cells. We found these low-allele-fraction clones in 26% of donors, without difference between multiple sclerosis (MS) patients and controls. Here we tested whether anti-viral antibodies associate with the carriership of these mutant clones. We compared antibody responses against common viruses in mutation carriers vs. non-carriers. Plasma samples of 152 donors (92 MS patients, 60 controls) were analyzed for antibodies against cytomegalovirus (CMV), Epstein-Barr virus (EBV), human herpesvirus-6A and parvovirus B19. The mutation carrier status associated with EBV VCA IgG level (p = 0.005) and remained significant after logistic regression (p = 0.036). This association was contributed similarly by MS patients and controls. These results suggest that EBV contributes to the generation or growth of these clones. The pathogenic role of the STAT3 mutant clones in MS is presently unclear, but their detailed characterization warrants further study.
Collapse
Affiliation(s)
- Joonas Lehikoinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland.
| | - Miko Valori
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Anne J Jääskeläinen
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital
| | - Sini M Laakso
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| | - T Petteri Arstila
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Bacteriology and Immunology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Brain Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
26
|
Lee S, Lee K, Bae H, Lee K, Lee J, Ma J, Lee YJ, Lee BR, Park WY, Im SJ. Defining a TCF1-expressing progenitor allogeneic CD8 + T cell subset in acute graft-versus-host disease. Nat Commun 2023; 14:5869. [PMID: 37737221 PMCID: PMC10516895 DOI: 10.1038/s41467-023-41357-9] [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: 11/28/2022] [Accepted: 09/01/2023] [Indexed: 09/23/2023] Open
Abstract
Graft-versus-host disease (GvHD) is a severe complication of hematopoietic stem cell transplantation driven by activated allogeneic T cells. Here, we identify a distinct subset of T cell factor-1 (TCF1)+ CD8+ T cells in mouse allogeneic and xenogeneic transplant models of acute GvHD. These TCF1+ cells exhibit distinct characteristics compared to TCF1- cells, including lower expression of inhibitory receptors and higher expression of costimulatory molecules. Notably, the TCF1+ subset displays exclusive proliferative potential and could differentiate into TCF1- effector cells upon antigenic stimulation. Pathway analyses support the role of TCF1+ and TCF1- subsets as resource cells and effector cells, respectively. Furthermore, the TCF1+ CD8+ T cell subset is primarily present in the spleen and exhibits a resident phenotype. These findings provide insight into the differentiation of allogeneic and xenogeneic CD8+ T cells and have implications for the development of immunotherapeutic strategies targeting acute GvHD.
Collapse
Affiliation(s)
- Solhwi Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kunhee Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Hyeonjin Bae
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kyungmin Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Junghwa Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Junhui Ma
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Ye Ji Lee
- GENINUS Inc., Seoul, Republic of Korea
| | | | - Woong-Yang Park
- GENINUS Inc., Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Se Jin Im
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
27
|
Gill AL, Wang PH, Lee J, Hudson WH, Ando S, Araki K, Hu Y, Wieland A, Im S, Gavora A, Medina CB, Freeman GJ, Hashimoto M, Reiner SL, Ahmed R. PD-1 blockade increases the self-renewal of stem-like CD8 T cells to compensate for their accelerated differentiation into effectors. Sci Immunol 2023; 8:eadg0539. [PMID: 37624909 PMCID: PMC10798572 DOI: 10.1126/sciimmunol.adg0539] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
PD-1+TCF-1+ stem-like CD8 T cells act as critical resource cells for maintaining T cell immunity in chronic viral infections and cancer. In addition, they provide the proliferative burst of effector CD8 T cells after programmed death protein 1 (PD-1)-directed immunotherapy. However, it is not known whether checkpoint blockade diminishes the number of these stem-like progenitor cells as effector cell differentiation increases. To investigate this, we used the mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection. Treatment of chronically infected mice with either αPD-1 or αPD-L1 antibody not only increased effector cell differentiation from the virus-specific stem-like CD8 T cells but also increased their proliferation so their numbers were maintained. The increased self-renewal of LCMV-specific stem-like CD8 T cells was mTOR dependent. We used microscopy to understand the division of these progenitor cells and found that after PD-1 blockade, an individual dividing cell could give rise to a differentiated TCF-1- daughter cell alongside a self-renewing TCF-1+ sister cell. This asymmetric division helped to preserve the number of stem-like cells. Moreover, we found that the PD-1+TCF-1+ stem-like CD8 T cells retained their transcriptional program and their in vivo functionality in terms of responding to viral infection and to repeat PD-1 blockade. Together, our results demonstrate that PD-1 blockade does not deplete the stem-like population despite increasing effector differentiation. These findings have implications for PD-1-directed immunotherapy in humans.
Collapse
Affiliation(s)
- Amanda L. Gill
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - Peter H. Wang
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center; New York, NY, 10032, USA
| | - Judong Lee
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - William H. Hudson
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - Satomi Ando
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati, OH, 45229, USA
| | - Koichi Araki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine; Cincinnati, OH, 45229, USA
| | - Yinghong Hu
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - Andreas Wieland
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University; Columbus, OH, 43210, USA
| | - Sejin Im
- Department of Immunology, Sungkyunkwan University School of Medicine; Suwon, Republic of Korea
| | - Autumn Gavora
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - Christopher B. Medina
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Masao Hashimoto
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| | - Steven L. Reiner
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center; New York, NY, 10032, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine; Atlanta, GA, 30329, USA
| |
Collapse
|
28
|
Pircher H, Pinschewer DD, Boehm T. MHC I tetramer staining tends to overestimate the number of functionally relevant self-reactive CD8 T cells in the preimmune repertoire. Eur J Immunol 2023; 53:e2350402. [PMID: 37179469 DOI: 10.1002/eji.202350402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/19/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Previous studies that used peptide-MHC (pMHC) tetramers (tet) to identify self-specific T cells have questioned the effectiveness of thymic-negative selection. Here, we used pMHCI tet to enumerate CD8 T cells specific for the immunodominant gp33 epitope of lymphocytic choriomeningitis virus glycoprotein (GP) in mice transgenically engineered to express high levels of GP as a self-antigen in the thymus. In GP-transgenic mice (GP+ ), monoclonal P14 TCR+ CD8 T cells that express a GP-specific TCR could not be detected by gp33/Db -tet staining, indicative of their complete intrathymic deletion. By contrast, in the same GP+ mice, substantial numbers of polyclonal CD8 T cells identifiable by gp33/Db -tet were present. The gp33-tet staining profiles of polyclonal T cells from GP+ and GP-negative (GP- ) mice were overlapping, but mean fluorescence intensities were ∼15% lower in cells from GP+ mice. Remarkably, the gp33-tet+ T cells in GP+ mice failed to clonally expand after lymphocytic choriomeningitis virus infection, whereas those of GP- mice did so. In Nur77GFP -reporter mice, dose-dependent responses to gp33 peptide-induced TCR stimulation revealed that gp33-tet+ T cells with high ligand sensitivity are lacking in GP+ mice. Hence, pMHCI tet staining identifies self-specific CD8 T cells but tends to overestimate the number of truly self-reactive cells.
Collapse
Affiliation(s)
- Hanspeter Pircher
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Thomas Boehm
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
29
|
Ganusov VV. Appropriate sampling and long follow-up are required to rigorously evaluate longevity of humoral memory after Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.28.23291950. [PMID: 37425815 PMCID: PMC10327268 DOI: 10.1101/2023.06.28.23291950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
One of the goals of vaccination is to induce long-term immunity against the infection and/or disease. However, evaluating the duration of protection following vaccination often requires long-term follow-ups that can conflict with the desire to rapidly publish results. Arunachalam et al. JCI 2023 followed individuals receiving third or fourth dose of mRNA COVID19 vaccines for up to 6 months and in finding that the levels of SARS-CoV2-specific antibodies (Abs) declined with similar rates for the two groups came to the conclusion that additional boosting is unnecessary to prolong immunity to SARS-CoV-2. However, this may be premature conclusion to make. Accordingly, we demonstrate that measuring Ab levels at 3 time points and only for a short (up to 6 month) duration does not allow to accurately and rigorously evaluate the long-term half-life of vaccine-induced Abs. By using the data from a cohort of blood donors followed for several years, we show that after re-vaccination with vaccinia virus (VV), VV-specific Abs decay bi-phasically and even the late decay rate exceeds the true slow loss rate of humoral memory observed years prior to the boosting. We argue that mathematical modeling should be used to better optimize sampling schedules to provide more reliable advice about the duration of humoral immunity after repeated vaccinations.
Collapse
Affiliation(s)
- Vitaly V Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
- Department of Mathematics, University of Tennessee, Knoxville, TN, USA
- Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
30
|
Lee J, Lee K, Bae H, Lee K, Lee S, Ma J, Jo K, Kim I, Jee B, Kang M, Im SJ. IL-15 promotes self-renewal of progenitor exhausted CD8 T cells during persistent antigenic stimulation. Front Immunol 2023; 14:1117092. [PMID: 37409128 PMCID: PMC10319055 DOI: 10.3389/fimmu.2023.1117092] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
In chronic infections and cancer, exhausted CD8 T cells exhibit heterogeneous subpopulations. TCF1+PD-1+ progenitor exhausted CD8 T cells (Tpex) can self-renew and give rise to Tim-3+PD-1+ terminally differentiated CD8 T cells that retain their effector functions. Tpex cells are thus essential to maintaining a pool of antigen-specific CD8 T cells during persistent antigenic stimulation, and only they respond to PD-1-targeted therapy. Despite their potential as a crucial therapeutic target for immune interventions, the mechanisms controlling the maintenance of virus-specific Tpex cells remain to be determined. We observed approximately 10-fold fewer Tpex cells in the spleens of mice chronically infected with lymphocytic choriomeningitis virus (LCMV) one-year post-infection (p.i.) than at three months p.i. Similar to memory CD8 T cells, Tpex cells have been found to undergo self-renewal in the lymphoid organs, prominently the bone marrow, during chronic LCMV infection. Furthermore, ex vivo treatment with IL-15 preferentially induced the proliferation of Tpex cells rather than the terminally differentiated subsets. Interestingly, single-cell RNA sequencing analysis of LCMV-specific exhausted CD8 T cells after ex vivo IL-15 treatment compared with those before treatment revealed increased expression of ribosome-related genes and decreased expression of genes associated with the TCR signaling pathway and apoptosis in both Tpex and Ttex subsets. The exogenous administration of IL-15 to chronically LCMV-infected mice also significantly increased self-renewal of Tpex cells in the spleen and bone marrow. In addition, we assessed the responsiveness of CD8 tumor-infiltrating lymphocytes (TILs) from renal cell carcinoma patients to IL-15. Similar to the data we obtained from chronic viral infection in mice, the expansion of the Tpex subset of PD-1+ CD8 TILs upon ex vivo IL-15 treatment was significantly higher than that of the terminally differentiated subset. These results show that IL-15 could promote self-renewal of Tpex cells, which has important therapeutic implications.
Collapse
Affiliation(s)
- Junghwa Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kyungmin Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Hyeonjin Bae
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kunhee Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Solhwi Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Junhui Ma
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kyungjo Jo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Ijun Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - ByulA Jee
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Se Jin Im
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
31
|
Walvekar P, Kumar P, Choonara YE. Long-acting vaccine delivery systems. Adv Drug Deliv Rev 2023; 198:114897. [PMID: 37225091 DOI: 10.1016/j.addr.2023.114897] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/27/2023] [Accepted: 05/18/2023] [Indexed: 05/26/2023]
Abstract
Bolus vaccines are often administered multiple times due to rapid clearance and reduced transportation to draining lymph nodes resulting in inadequate activation of T and B lymphocytes. In order to achieve adaptive immunity, prolonged exposure of antigens to these immune cells is crucial. Recent research has been focusing on developing long-acting biomaterial-based vaccine delivery systems, which can modulate the release of encapsulated antigens or epitopes to facilitate enhanced antigen presentation in lymph nodes and subsequently achieve robust T and B cell responses. Over the past few years, various polymers and lipids have been extensively explored to develop effective biomaterial-based vaccine strategies. The article reviews relevant polymer and lipid-based strategies used to prepare long-acting vaccine carriers and discusses their results concerning immune responses.
Collapse
Affiliation(s)
- Pavan Walvekar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa.
| |
Collapse
|
32
|
Lee S, Ma J, Im SJ. Expression and function of CD51 on CD8 T cells as an immunomodulatory target. Biochem Biophys Res Commun 2023; 661:56-63. [PMID: 37087799 DOI: 10.1016/j.bbrc.2023.04.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/30/2023] [Accepted: 04/15/2023] [Indexed: 04/25/2023]
Abstract
T cell responses are regulated by co-stimulatory and inhibitory receptors along with T cell receptor- and cytokine-mediated signals. CD51 is a transmembrane glycoprotein of the integrin family that plays a role in cell adhesion, migration, tumorigenesis, and other cellular functions. In this study, we aimed to investigate the expression and function of CD51 on CD8 T cells. Upon in vitro T cell activation, CD51 expression was delayed but subsequently was upregulated in CD8 T cells upon cell division. Furthermore, CD51 was highly expressed in exhausted CD8 T cells in chronic LCMV infection, B16F10 melanoma, and CT26 colon carcinoma, and its expression level increased as cells became more differentiated. Using CRISPR-mediated knockdown, we found that the absence of CD51 led to a lower number of virus-specific CD8 T cells upon chronic lymphocytic choriomeningitis virus (LCMV) infection, although their granzyme B expression and cytokine production were maintained. Blocking CD51 also inhibited the in vitro proliferation of CD8 T cells. These results suggest that CD51 plays an important role in the early expansion of CD8 T cells and may have potential as an immunomodulatory target.
Collapse
Affiliation(s)
- Solhwi Lee
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Junhui Ma
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Se Jin Im
- Department of Immunology, Graduate School of Basic Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
33
|
Osum KC, Jenkins MK. Toward a general model of CD4 + T cell subset specification and memory cell formation. Immunity 2023; 56:475-484. [PMID: 36921574 PMCID: PMC10084496 DOI: 10.1016/j.immuni.2023.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/16/2023] [Indexed: 03/17/2023]
Abstract
In the past few decades, a number of transformative discoveries have been made regarding memory CD8+ T cell biology; meanwhile, the CD4+ T cell field has lagged behind this progress. This perspective focuses on CD4+ helper T (Th) cell subset specification and memory cell formation. Here, we argue that the sheer number of Th effector and memory cell subsets and a focus on their differences have been a barrier to a general model of CD4+ memory T cell formation that applies to all immune responses. We highlight a bifurcation model that relies on an IL-2 signal-dependent switch as an explanation for the balanced production of diverse Th memory cells that participate in cell-mediated or humoral immunity in most contexts.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
34
|
Pyrimidine de novo synthesis inhibition selectively blocks effector but not memory T cell development. Nat Immunol 2023; 24:501-515. [PMID: 36797499 DOI: 10.1038/s41590-023-01436-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 01/13/2023] [Indexed: 02/18/2023]
Abstract
Blocking pyrimidine de novo synthesis by inhibiting dihydroorotate dehydrogenase is used to treat autoimmunity and prevent expansion of rapidly dividing cell populations including activated T cells. Here we show memory T cell precursors are resistant to pyrimidine starvation. Although the treatment effectively blocked effector T cells, the number, function and transcriptional profile of memory T cells and their precursors were unaffected. This effect occurred in a narrow time window in the early T cell expansion phase when developing effector, but not memory precursor, T cells are vulnerable to pyrimidine starvation. This vulnerability stems from a higher proliferative rate of early effector T cells as well as lower pyrimidine synthesis capacity when compared with memory precursors. This differential sensitivity is a drug-targetable checkpoint that efficiently diminishes effector T cells without affecting the memory compartment. This cell fate checkpoint might therefore lead to new methods to safely manipulate effector T cell responses.
Collapse
|
35
|
Longitudinal Analysis of the Phenotype, Transcriptional Profile, and Anatomic Location of Memory CD8 T Cell Subsets after Acute Viral Infection. J Virol 2023; 97:e0155622. [PMID: 36541799 PMCID: PMC9888238 DOI: 10.1128/jvi.01556-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Increased demand for novel, highly effective vaccination strategies necessitates a better understanding of long-lived memory CD8 T cell differentiation. To achieve this understanding, we used the mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. We reexamined classical memory CD8 T cell subsets and performed in-depth, longitudinal analysis of their phenotype, transcriptional programming, and anatomic location within the spleen. All analyses were performed at multiple time points from 8 days to 1 year postinfection. Memory subsets are conventionally defined by their expression of KLRG1 and IL-7Rα, as follows: KLRG1+IL-7Rα- terminal effectors (TEs) and KLRG1-IL-7Rα+ memory precursors (MPs). But we also characterized a third KLRG1+IL-7Rα+ subset which we refer to as KLRG1+ MPs. In these analyses, we defined a comprehensive memory phenotype that is associated with higher levels of CD28 expression. We also demonstrated that MPs, KLRG1+ MPs, and TEs have distinct localization programs within the spleen. We found that MPs became preferentially enriched in the white pulp as early as 1 to 2 weeks postinfection, and their predominance in the white pulp was maintained throughout the course of a year. On the other hand, KLRG1+ MPs and TEs localized to the red pulp just as early, and they consistently localized to the red pulp thereafter. These findings indicate that location may be crucial for memory formation and that white pulp-derived signals may contribute to long-term memory survival. Achieving robust memory responses following vaccination may require more deliberate consideration of which memory phenotypes are induced, as well as where they traffic, as these factors could impact their longevity. IMPORTANCE CD8 T cells play a critical role in viral immunity and it is important to understand how memory cells are formed and what processes lead to their long-term maintenance. Here, we use a mouse model of acute infection to perform an in-depth, longitudinal analysis of memory CD8 T cell differentiation, examining the phenotype and location of memory cells out to 1 year postinfection.
Collapse
|
36
|
Grusdat M, Dostert C, Brenner D. Quantification of lymphocytic choriomeningitis virus specific T cells and LCMV viral titers. Methods Cell Biol 2023; 173:121-131. [PMID: 36653079 DOI: 10.1016/bs.mcb.2022.03.003] [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] [Indexed: 02/04/2023]
Abstract
Lymphocytic choriomeningitis virus (LCMV) is a frequently used animal model to study immune responses against acute and chronic viral infections. LCMV is a non-cytopathic virus, but destruction of infected cells by cytotoxic T lymphocytes (CTLs) can lead to severe damage of tissues. Virus-specific T cell responses have to be balanced. A low virus load leads to a strong T cell response and subsequently to viral control. In contrast, a high viral titer is associated with T cell exhaustion and chronic viral infections. During an intermediate LCMV viral load CD8+ T cells can cause immunopathology, which can have detrimental outcomes. The LCMV infection model offers the opportunity to study virus-specific CD4+ and CD8+ T cell responses during chronic and acute infections by quantifying LCMV-specific T cells by tetramer staining and measuring cytokine production and viral titers in different organs.
Collapse
Affiliation(s)
- Melanie Grusdat
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catherine Dostert
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
37
|
Ando S, Perkins CM, Sajiki Y, Chastain C, Valanparambil RM, Wieland A, Hudson WH, Hashimoto M, Ramalingam SS, Freeman GJ, Ahmed R, Araki K. mTOR regulates T cell exhaustion and PD-1-targeted immunotherapy response during chronic viral infection. J Clin Invest 2023; 133:e160025. [PMID: 36378537 PMCID: PMC9843061 DOI: 10.1172/jci160025] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
T cell exhaustion is a state of T cell dysfunction associated with expression of programmed death 1 (PD-1). Exhausted CD8+ T cells are maintained by self-renewing stem-like T cells that provide differentiated TIM3+ cells, a part of which possesses effector-like properties. PD-1-targeted therapies enhance T cell response by promoting differentiation of stem-like T cells toward TIM3+ cells, but the role of mTOR during T cell exhaustion remains elusive. Here, we showed that mTOR inhibition has distinct outcomes during the beginning of and after the establishment of chronic viral infection. Blocking mTOR during the T cell expansion phase enhanced the T cell response by causing accumulation of stem-like T cells, leading to improved efficacy of PD-1 immunotherapy; whereas, after exhaustion progressed, mTOR inhibition caused immunosuppression, characterized by decreased TIM3+ cells and increased viral load with minimal changes in stem-like T cells. Mechanistically, a cell-intrinsic mTOR signal was vital for differentiation of stem-like T cells into the TIM3+ state in the early and late phases of chronic infection as well as during PD-1 immunotherapy. Thus, PD-1 blockade worked after cessation of mTOR inhibition, but simultaneous treatment failed to induce functional TIM3+ cells, reducing efficacy of PD-1 immunotherapy. Our data demonstrate that mTOR regulates T cell exhaustion and have important implications for combination cancer therapies with PD-1 blockade.
Collapse
Affiliation(s)
- Satomi Ando
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Charles M. Perkins
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yamato Sajiki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Chase Chastain
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Andreas Wieland
- Emory Vaccine Center
- Depatment of Microbiology and Immunology, and
| | | | - Masao Hashimoto
- Emory Vaccine Center
- Depatment of Microbiology and Immunology, and
| | - Suresh S. Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rafi Ahmed
- Emory Vaccine Center
- Depatment of Microbiology and Immunology, and
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Koichi Araki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
38
|
Abstract
The galectin family consists of carbohydrate (glycan) binding proteins that are expressed by a wide variety of cells and bind to galactose-containing glycans. Galectins can be located in the nucleus or the cytoplasm, or can be secreted into the extracellular space. They can modulate innate and adaptive immune cells by binding to glycans on the surface of immune cells or intracellularly via carbohydrate-dependent or carbohydrate-independent interactions. Galectins expressed by immune cells can also participate in host responses to infection by directly binding to microorganisms or by modulating antimicrobial functions such as autophagy. Here we explore the diverse ways in which galectins have been shown to impact immunity and discuss the opportunities and challenges in the field.
Collapse
|
39
|
Grasso EA, Pozzilli V, Tomassini V. Transverse myelitis in children and adults. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:101-117. [PMID: 37620065 DOI: 10.1016/b978-0-323-98817-9.00020-x] [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: 08/26/2023]
Abstract
Transverse myelitis is a noncompressive myelopathy of inflammatory origin. The causes are broad, ranging from infective or toxic to immuno-mediated etiology. They can be manifestations of systemic diseases, such as sarcoidosis and systemic lupus erythematous, or phenotypes of neuroinflammation; in a portion of cases, the etiology remains unknown, leading to the designation idiopathic. The clinical presentation of transverse myelitis depends on the level of spinal cord damage and may include sensorimotor deficits and autonomic dysfunction. The age of onset of the disorder can impact the symptoms and outcomes of affected patients, with differences in manifestation and prognosis between children and adults. Spinal cord magnetic resonance imaging and cerebrospinal fluid examination are the main diagnostic tools that can guide clinicians in the diagnostic process, even though the search for antibodies that target the structural components of the neural tissue (anti-aquaporin4 antibodies and anti-myelin-oligodendrocyte antibodies) helps in the distinction among the immune-mediated phenotypes. Management and outcomes depend on the underlying cause, with different probabilities of relapse according to the phenotypes. Hence, immunosuppression is often recommended for the immune-mediated diseases that may have a higher risk of recurrence. Age at onset has implications for the choice of treatment.
Collapse
Affiliation(s)
- Eleonora Agata Grasso
- Department of Neurosciences, Imaging and Clinical Sciences, Institute of Advanced Biomedical Technologies (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Valeria Pozzilli
- Department of Neurosciences, Imaging and Clinical Sciences, Institute of Advanced Biomedical Technologies (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Valentina Tomassini
- Department of Neurosciences, Imaging and Clinical Sciences, Institute of Advanced Biomedical Technologies (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
40
|
Influenza Virus Infection during Pregnancy as a Trigger of Acute and Chronic Complications. Viruses 2022; 14:v14122729. [PMID: 36560733 PMCID: PMC9786233 DOI: 10.3390/v14122729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus (IAV) infection during pregnancy disrupts maternal and fetal health through biological mechanisms, which are to date poorly characterised. During pregnancy, the viral clearance mechanisms from the lung are sub-optimal and involve hyperactive innate and adaptive immune responses that generate wide-spread inflammation. Pregnancy-related adaptations of the immune and the cardiovascular systems appear to result in delayed recovery post-viral infection, which in turn promotes a prolonged inflammatory phenotype, increasing disease severity, and causing maternal and fetal health problems. This has immediate and long-term consequences for the mother and fetus, with complications including acute cardiopulmonary distress syndrome in the mother that lead to perinatal complications such as intrauterine growth restriction (IUGR), and birth defects; cleft lip, cleft palate, neural tube defects and congenital heart defects. In addition, an increased risk of long-term neurological disorders including schizophrenia in the offspring is reported. In this review we discuss the pathophysiology of IAV infection during pregnancy and its striking similarity to other well-established complications of pregnancy such as preeclampsia. We discuss general features of vascular disease with a focus on vascular inflammation and define the "Vascular Storm" that is triggered by influenza infection during pregnancy, as a pivotal disease mechanism for short and long term cardiovascular complications.
Collapse
|
41
|
Responsiveness to interleukin-15 therapy is shared between tissue-resident and circulating memory CD8 + T cell subsets. Proc Natl Acad Sci U S A 2022; 119:e2209021119. [PMID: 36260745 PMCID: PMC9618124 DOI: 10.1073/pnas.2209021119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interleukin-15 (IL-15) is often considered a central regulator of memory CD8+ T cells, based primarily on studies of recirculating subsets. However, recent work identified IL-15-independent CD8+ T cell memory populations, including tissue-resident memory CD8+ T cells (TRM) in some nonlymphoid tissues (NLTs). Whether this reflects the existence of IL-15-insensitive memory CD8+ T cells is unclear. We report that IL-15 complexes (IL-15c) stimulate rapid proliferation and expansion of both tissue-resident and circulating memory CD8+ T cell subsets across lymphoid and nonlymphoid tissues with varying magnitude by tissue and memory subset, in some sites correlating with differing levels of the IL-2Rβ. This was conserved for memory CD8+ T cells recognizing distinct antigens and elicited by different pathogens. Following IL-15c-induced expansion, divided cells contracted to baseline numbers and only slowly returned to basal proliferation, suggesting a mechanism to transiently amplify memory populations. Through parabiosis, we showed that IL-15c drive local proliferation of TRM, with a degree of recruitment of circulating cells to some NLTs. Hence, irrespective of homeostatic IL-15 dependence, IL-15 sensitivity is a defining feature of memory CD8+ T cell populations, with therapeutic potential for expansion of TRM and other memory subsets in an antigen-agnostic and temporally controlled fashion.
Collapse
|
42
|
Ostovan VR, Sahraian MA, Karazhian N, Rostamihosseinkhani M, Salimi M, Marbooti H. Clinical characteristics, radiological features and prognostic factors of transverse myelitis following COVID-19 vaccination: A systematic review. Mult Scler Relat Disord 2022; 66:104032. [PMID: 35858499 PMCID: PMC9258415 DOI: 10.1016/j.msard.2022.104032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/24/2022] [Accepted: 07/03/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Since introducing COVID-19 vaccines, many neurological complications such as acute transverse myelitis have been reported in the literature. This study aims to identify the clinical characteristics, radiological findings, and prognostic factors in patients with COVID-19 vaccine-associated transverse myelitis (TM). METHODS We systematically reviewed Scopus, Pubmed, Cochrane library, Google Scholar, and preprint databases using appropriate keywords from inception till 8th April 2022. Besides, we manually searched the reference lists of the included studies and relevant previous reviews. RESULTS We included 28 studies identifying 31 post-COVID-19 vaccination myelitis patients (17 female and 14 male). The mean age of the included patients was 52±19 years. ChAdOx1 nCoV-19 vaccine (Oxford-AstraZeneca) was the most common type of vaccine in association with myelitis (12 out of 31), followed by Pfizer (8 out of 31), Moderna (7 out of 31), Sinopharm (3 out of 31), and Janssen vaccine (1 out of 31). The myelitis occurred in 24 and 7 patients after administering the first and second dose of the vaccine, respectively. 21 and 10 patients had good recovery (Modified Rankin Score (MRS) <3 at the follow-up) and poor recovery (MRS≥3 at the follow-up) from myelitis, respectively. Age (OR 1.09, 95%CI 1.01-1.18, pvalue 0.02), and MRS at admission (OR 17.67, 95%CI 1.46-213.76, pvalue 0.024) were two independent risk factors for poor recovery from myelitis. CONCLUSION The patients with higher age and MRS at admission had a worse prognosis and needed timely and more aggressive therapeutic strategies.
Collapse
Affiliation(s)
- Vahid Reza Ostovan
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Sahraian
- MS Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Karazhian
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Marzieh Salimi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hoda Marbooti
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
43
|
Van Roy Z, Kielian T. Exploring epigenetic reprogramming during central nervous system infection. Immunol Rev 2022; 311:112-129. [PMID: 35481573 PMCID: PMC9790395 DOI: 10.1111/imr.13079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022]
Abstract
Epigenetics involves the study of various modes of adaptable transcriptional regulation, contributing to cell identity, characteristics, and function. During central nervous system (CNS) infection, epigenetic mechanisms can exert pronounced control over the maturation and antimicrobial properties of nearly every immune cell type. Epigenetics is a relatively new field, with the first mention of these marks proposed only a half-century ago and a substantial body of immunological epigenetic research emerging only in the last few decades. Here, we review the best-characterized epigenetic marks and their functions as well as illustrate how various immune cell populations responding to CNS infection utilize these marks to organize their activation state and inflammatory processes. We also discuss the metabolic and clinical implications of epigenetic marks and the rapidly expanding set of tools available to researchers that are enabling elucidation of increasingly detailed genetic regulatory pathways. These considerations paint an intricate picture of inflammatory regulation, where epigenetic marks influence genetic, signaling, and environmental elements to orchestrate a tailored immunological response to the threat at hand, cementing epigenetics as an important player in immunity.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Tammy Kielian
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
44
|
Mamootil D, Grewal A. Viral Versus Vaccine-Associated Acute Transverse Myelitis With Neuromyelitis Optica Immunoglobulin G Antibody and Myelin Basic Protein: A Case Report. Cureus 2022; 14:e28922. [PMID: 36225431 PMCID: PMC9541889 DOI: 10.7759/cureus.28922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2022] [Indexed: 12/05/2022] Open
Abstract
Transverse myelitis is a rare spinal cord disorder caused by local inflammation. Usually, this occurs as a complication from infection or autoimmune disease; however, there have been reported idiopathic causes such as vaccinations. A 73-year-old female with a medical history significant for Hashimoto’s thyroiditis presented with new-onset paresthesias in her lower extremities. Her symptom onset was about five weeks after receiving influenza and tetanus, diphtheria, and pertussis (TDaP) vaccines. Magnetic resonance imaging (MRI) of the spine revealed an increased T2 signal of the lower cervical and thoracic spine. Lumbar puncture was also performed, and cerebrospinal fluid (CSF) serology showed elevated myelin basic protein (MBP) at 108.3 ng/mL (reference range: 0-5.5 ng/mL). Serology panel revealed Coxsackie virus type B4 antibody at 1:80 (reference range: <1:10) and Echovirus type 6 antibody at 1:640 (reference range: <1:10). Neuromyelitis optica (NMO) immunoglobulin G (IgG) antibody was 24.6 U/mL (reference range: <2.9 U/mL). She was diagnosed with acute transverse myelitis (ATM) and treated with alternating steroids and plasma exchange (PLEX) therapy for five days each. This case highlights the possible associations of vaccines with transverse myelitis. Although ATM is a rare disorder with serious complications, it has a favorable prognosis in the setting of rapid detection and treatment. Vaccine-related ATM remains controversial, but patients with these adverse reactions need to be cautioned regarding potential recurrence risk.
Collapse
|
45
|
Nath PR, Pal-Nath D, Kaur S, Gangaplara A, Meyer TJ, Cam MC, Roberts DD. Loss of CD47 alters CD8+ T cell activation in vitro and immunodynamics in mice. Oncoimmunology 2022; 11:2111909. [PMID: 36105746 PMCID: PMC9467551 DOI: 10.1080/2162402x.2022.2111909] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
CD47 has established roles in the immune system for regulating macrophage phagocytosis and lymphocyte activation, with growing evidence of its cell-intrinsic regulatory roles in natural killer and CD8+ T cells. CD47 limits antigen-dependent cytotoxic activities of human and murine CD8+ T cells, but its role in T cell activation kinetics remains unclear. Using in vitro and in vivo models, we show here that CD47 differentially regulates CD8+ T cell responses to short- versus long-term activation. Although CD47 was not required for T cell development in mice and early activation in vitro, short-term stimuli elevated pathogen-reactive gene expression and enhanced proliferation and the effector phenotypes of Cd47-deficient relative to Cd47-sufficient CD8+ T cells. In contrast, persistent TCR stimulation limited the effector phenotypes of Cd47 -/- CD8+ T cells and enhanced their apoptosis signature. CD8+ T cell expansion and activation in vivo induced by acute lymphocytic choriomeningitis virus (LCMV) infection did not differ in the absence of CD47. However, the frequency and effector phenotypes of Cd47-/- CD8+ T cells were constrained in chronic LCMV-infected as well as in mice bearing B16 melanoma tumors. Therefore, CD47 regulates CD8+ T cell activation, proliferation, and fitness in a context-dependent manner.
Collapse
Affiliation(s)
- Pulak R. Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical and Translational Immunology Unit, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arunkumar Gangaplara
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute, Bethesda, MD, USA
| | - Margaret C Cam
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute, Bethesda, MD, USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Naoun AA, Raphael I, Forsthuber TG. Immunoregulation via Cell Density and Quorum Sensing-like Mechanisms: An Underexplored Emerging Field with Potential Translational Implications. Cells 2022; 11:cells11152442. [PMID: 35954285 PMCID: PMC9368058 DOI: 10.3390/cells11152442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Quorum sensing (QS) was historically described as a mechanism by which bacteria detect and optimize their population density via gene regulation based on dynamic environmental cues. Recently, it was proposed that QS or similar mechanisms may have broader applications across different species and cell types. Indeed, emerging evidence shows that the mammalian immune system can also elicit coordinated responses on a population level to regulate cell density and function, thus suggesting that QS-like mechanisms may also be a beneficial trait of the immune system. In this review, we explore and discuss potential QS-like mechanisms deployed by the immune system to coordinate cellular-level responses, such as T cell responses mediated via the common gamma chain (γc) receptor cytokines and the aryl hydrocarbon receptors (AhRs). We present evidence regarding a novel role of QS as a multifunctional mechanism coordinating CD4+ and CD8+ T cell behavior during steady state and in response to infection, inflammatory diseases, and cancer. Successful clinical therapies such as adoptive cell transfer for cancer treatment may be re-evaluated to harness the effects of the QS mechanism(s) and enhance treatment responsiveness. Moreover, we discuss how signaling threshold perturbations through QS-like mediators may result in disturbances of the complex crosstalk between immune cell populations, undesired T cell responses, and induction of autoimmune pathology. Finally, we discuss the potential therapeutic role of modulating immune-system-related QS as a promising avenue to treat human diseases.
Collapse
Affiliation(s)
- Adrian A. Naoun
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Itay Raphael
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15217, USA
- Correspondence: (I.R.); (T.G.F.)
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
- Correspondence: (I.R.); (T.G.F.)
| |
Collapse
|
47
|
Jutel M, Torres MJ, Palomares O, Akdis CA, Eiwegger T, Untersmayr E, Barber D, Zemelka-Wiacek M, Kosowska A, Palmer E, Vieths S, Mahler V, Canonica WG, Nadeau K, Shamji MH, Agache I. COVID-19 vaccination in patients receiving allergen immunotherapy (AIT) or biologicals-EAACI recommendations. Allergy 2022; 77:2313-2336. [PMID: 35147230 PMCID: PMC9111382 DOI: 10.1111/all.15252] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 12/16/2022]
Abstract
Immune modulation is a key therapeutic approach for allergic diseases, asthma and autoimmunity. It can be achieved in an antigen-specific manner via allergen immunotherapy (AIT) or in an endotype-driven approach using biologicals that target the major pathways of the type 2 (T2) immune response: immunoglobulin (Ig)E, interleukin (IL)-5 and IL-4/IL-13 or non-type 2 response: anti-cytokine antibodies and B-cell depletion via anti-CD20. Coronavirus disease 2019 (COVID-19) vaccination provides an excellent opportunity to tackle the global pandemics and is currently being applied in an accelerated rhythm worldwide. The vaccine exerts its effects through immune modulation, induces and amplifies the response against the severe acute respiratory syndrome coronavirus (SARS-CoV-2). Thus, as there may be a discernible interference between these treatment modalities, recommendations on how they should be applied in sequence are expected. The European Academy of Allergy and Clinical Immunology (EAACI) assembled an expert panel under its Research and Outreach Committee (ROC). This expert panel evaluated the evidence and have formulated recommendations on the administration of COVID-19 vaccine in patients with allergic diseases and asthma receiving AIT or biologicals. The panel also formulated recommendations for COVID-19 vaccine in association with biologicals targeting the type 1 or type 3 immune response. In formulating recommendations, the panel evaluated the mechanisms of COVID-19 infection, of COVID-19 vaccine, of AIT and of biologicals and considered the data published for other anti-infectious vaccines administered concurrently with AIT or biologicals.
Collapse
Affiliation(s)
- Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wroclaw, Poland
| | - Maria J Torres
- Allergy Unit, Regional University Hospital of Malaga, IBIMA-UMA-ARADyAL-BIONAND, Malaga, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Thomas Eiwegger
- Division of Immunology and Allergy, The Department of 13 Pediatrics, Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Domingo Barber
- Facultad de Medicina, Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | | | - Anna Kosowska
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wroclaw, Poland
| | - Elizabeth Palmer
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London. MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Stefan Vieths
- Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | | | - Walter G Canonica
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Personalized Medicine Asthma, & Allergy Center-IRCCS Humanitas Research Hospital, Milan, Italy
| | - Kari Nadeau
- Division of Pulmonary, Allergy and Critical Care Medicine, Dept of Medicine, Stanford, California, USA
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London. MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | |
Collapse
|
48
|
Mark M, Reich-Zeliger S, Greenstein E, Reshef D, Madi A, Chain B, Friedman N. A hierarchy of selection pressures determines the organization of the T cell receptor repertoire. Front Immunol 2022; 13:939394. [PMID: 35967295 PMCID: PMC9372880 DOI: 10.3389/fimmu.2022.939394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
We systematically examine the receptor repertoire in T cell subsets in young, adult, and LCMV-infected mice. Somatic recombination generates diversity, resulting in the limited overlap between nucleotide sequences of different repertoires even within the same individual. However, statistical features of the repertoire, quantified by the V gene and CDR3 k-mer frequency distributions, are highly conserved. A hierarchy of immunological processes drives the evolution of this structure. Intra-thymic divergence of CD4+ and CD8+ lineages imposes subtle but dominant differences observed across repertoires of all subpopulations in both young and adult mice. Differentiation from naive through memory to effector phenotype imposes an additional gradient of repertoire diversification, which is further influenced by age in a complex and lineage-dependent manner. The distinct repertoire of CD4+ regulatory T cells is more similar to naive cells in young mice and to effectors in adults. Finally, we describe divergent (naive and memory) and convergent (CD8+ effector) evolution of the repertoire following acute infection with LCMV. This study presents a quantitative framework that captures the structure of the repertoire in terms of its fundamental statistical properties and describes how this structure evolves as individual T cells differentiate, migrate and mature in response to antigen exposure.
Collapse
Affiliation(s)
- Michal Mark
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
- *Correspondence: Michal Mark, ; Benny Chain,
| | | | - Erez Greenstein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dan Reshef
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Asaf Madi
- Department of Pathology, Tel-Aviv University, Tel-Aviv, Israel
| | - Benny Chain
- Department of Computer Science, University College London, UCL, London, United Kingdom
- *Correspondence: Michal Mark, ; Benny Chain,
| | - Nir Friedman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
49
|
Nong C, Guan P, Li L, Zhang H, Hu H. Tumor immunotherapy: Mechanisms and clinical applications. MEDCOMM – ONCOLOGY 2022. [DOI: 10.1002/mog2.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Cheng Nong
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Pengbo Guan
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Li
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Huiyuan Zhang
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Hongbo Hu
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- Chongqing International Institution for Immunology Chongqing China
| |
Collapse
|
50
|
Jo T, Noguchi K, Sakai T, Kubota-Koketsu R, Irie S, Matsuo M, Taguchi J, Abe K, Shigematsu K. HTLV-1 Tax-specific memory cytotoxic T lymphocytes in long-term survivors of aggressive-type adult T-cell leukemia/lymphoma. Cancer Med 2022; 11:3238-3250. [PMID: 35315593 PMCID: PMC9468428 DOI: 10.1002/cam4.4689] [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: 07/29/2021] [Revised: 01/24/2022] [Accepted: 02/24/2022] [Indexed: 01/08/2023] Open
Abstract
Purpose Adult T‐cell leukemia/lymphoma (ATLL) is a relatively refractory peripheral T‐cell lymphoma caused by human T‐cell lymphotropic virus type 1 (HTLV‐1). The objective of this study was to investigate the characteristics of long‐term survivors with ATLL. Methods We conducted an observational study of 75 aggressive‐type ATLL patients. Flow cytometry was conducted to analyze HTLV‐1 Tax‐specific cytotoxic T‐lymphocytes (CTLs) and T‐cell receptor Vβ gene repertoire. Results We first evaluated six long‐term survivors among 37 patients who were newly diagnosed with ATLL and then treated with intensive chemotherapy without mogamulizumab, a monoclonal antibody for C‐C chemokine receptor four antigen. Reversal of the CD4‐to‐CD8 ratio (CD4/CD8) in peripheral mononuclear cells was observed in all six patients. Three of these six patients showed reversed CD4/CD8 immediately after herpes virus infection. Four of these six patients who could be examined demonstrated long‐term maintenance of HTLV‐1 Tax‐specific CTLs. We subsequently identified four long‐term survivors among 38 patients who were newly diagnosed with ATLL and then treated with intensive chemotherapy plus mogamulizumab. All four patients showed reversed CD4/CD8, and three of the four patients contracted herpes virus infection during immunochemotherapy. Six of the total 10 patients were subjected to CTL analyses. Tax‐specific CTLs were observed, and the CTLs that were almost entirely composed of memory CTLs in all patients were recorded. HTLV‐1 provirus was also detected in all six patients. Conclusions These data suggest that Tax‐specific memory CTLs probably, together with anticancer agents, eradicate ATLL cells and exhibit long‐term preventive effects from relapse ATLL. Thus, the strong activation of cellular immunity, such as herpes virus infection, seems to be necessary to induce such a potent number of Tax‐specific CTLs.
Collapse
Affiliation(s)
- Tatsuro Jo
- Department of Hematology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Kazuhiro Noguchi
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Takahiro Sakai
- Department of Laboratory, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Ritsuko Kubota-Koketsu
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Sadaharu Irie
- Department of Pharmacy, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Masatoshi Matsuo
- Department of Hematology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Jun Taguchi
- Department of Hematology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Kuniko Abe
- Department of Pathology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Kazuto Shigematsu
- Department of Pathology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| |
Collapse
|