1
|
Vemulawada C, Renavikar PS, Crawford MP, Steward-Tharp S, Karandikar NJ. Disruption of IFNγ, GZMB, PRF1, or LYST Results in Reduced Suppressive Function in Human CD8+ T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1722-1732. [PMID: 38607279 PMCID: PMC11105984 DOI: 10.4049/jimmunol.2300388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 03/20/2024] [Indexed: 04/13/2024]
Abstract
An imbalance between proinflammatory and regulatory processes underlies autoimmune disease pathogenesis. We have shown that acute relapses of multiple sclerosis are characterized by a deficit in the immune suppressive ability of CD8+ T cells. These cells play an important immune regulatory role, mediated in part through cytotoxicity (perforin [PRF]/granzyme [GZM]) and IFNγ secretion. In this study, we further investigated the importance of IFNγ-, GZMB-, PRF1-, and LYST-associated pathways in CD8+ T cell-mediated suppression. Using the CRISPR-Cas9 ribonucleoprotein transfection system, we first optimized efficient gene knockout while maintaining high viability in primary bulk human CD8+ T cells. Knockout was confirmed through quantitative real-time PCR assays in all cases, combined with flow cytometry where appropriate, as well as confirmation of insertions and/or deletions at genomic target sites. We observed that the knockout of IFNγ, GZMB, PRF1, or LYST, but not the knockout of IL4 or IL5, resulted in significantly diminished in vitro suppressive ability in these cells. Collectively, these results reveal a pivotal role for these pathways in CD8+ T cell-mediated immune suppression and provide important insights into the biology of human CD8+ T cell-mediated suppression that could be targeted for immunotherapeutic intervention.
Collapse
Affiliation(s)
- Chakrapani Vemulawada
- Department of Pathology, University of Iowa Health Care, 200 Hawkins Dr., Iowa City, IA 52242
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Pranav S. Renavikar
- Department of Pathology, University of Iowa Health Care, 200 Hawkins Dr., Iowa City, IA 52242
| | - Michael P. Crawford
- Department of Pathology, University of Iowa Health Care, 200 Hawkins Dr., Iowa City, IA 52242
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Scott Steward-Tharp
- Department of Pathology, University of Iowa Health Care, 200 Hawkins Dr., Iowa City, IA 52242
| | - Nitin J. Karandikar
- Department of Pathology, University of Iowa Health Care, 200 Hawkins Dr., Iowa City, IA 52242
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| |
Collapse
|
2
|
Kustrimovic N, Gallo D, Piantanida E, Bartalena L, Lai A, Zerbinati N, Tanda ML, Mortara L. Regulatory T Cells in the Pathogenesis of Graves' Disease. Int J Mol Sci 2023; 24:16432. [PMID: 38003622 PMCID: PMC10671795 DOI: 10.3390/ijms242216432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Maintaining a delicate balance between the prompt immune response to pathogens and tolerance towards self-antigens and commensals is crucial for health. T regulatory (Treg) cells are pivotal in preserving self-tolerance, serving as negative regulators of inflammation through the secretion of anti-inflammatory cytokines, interleukin-2 neutralization, and direct suppression of effector T cells. Graves' disease (GD) is a thyroid-specific autoimmune disorder primarily attributed to the breakdown of tolerance to the thyroid-stimulating hormone receptor. Given the limitations of currently available GD treatments, identifying potential pathogenetic factors for pharmacological targeting is of paramount importance. Both functional impairment and frequency reduction of Tregs seem likely in GD pathogenesis. Genome-wide association studies in GD have identified polymorphisms of genes involved in Tregs' functions, such as CD25 (interleukin 2 receptor), and Forkhead box protein P3 (FOXP3). Clinical studies have reported both functional impairment and a reduction in Treg frequency or suppressive actions in GD, although their precise involvement remains a subject of debate. This review begins with an overview of Treg phenotype and functions, subsequently delves into the pathophysiology of GD and into the existing literature concerning the role of Tregs and the balance between Tregs and T helper 17 cells in GD, and finally explores the ongoing studies on target therapies for GD.
Collapse
Affiliation(s)
- Natasa Kustrimovic
- Center for Translational Research on Autoimmune and Allergic Disease—CAAD, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Gallo
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy (M.L.T.)
| | - Eliana Piantanida
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy (M.L.T.)
| | - Luigi Bartalena
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy (M.L.T.)
| | - Adriana Lai
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy (M.L.T.)
| | - Nicola Zerbinati
- Dermatology Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy
| | - Maria Laura Tanda
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy (M.L.T.)
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
3
|
Elizondo CR, Bright JD, Bright RK. Vaccination with a shared oncogenic tumor-self antigen elicits a population of CD8+ T cells with a regulatory phenotype. Hum Vaccin Immunother 2022; 18:2108656. [PMID: 36069634 PMCID: PMC9746449 DOI: 10.1080/21645515.2022.2108656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/29/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer immunotherapy is a powerful tool for inducing antigen-specific antitumor cytotoxic T lymphocytes (CTLs). Next-generation strategies may include vaccination against overexpressed oncogenic tumor-self antigens. Previously, we reported vaccination against the oncogenic tumor-self antigen D52 (D52) was effective in preventing tumor growth. We recently reported that D52-vaccinated IL-10-deficient mice generated a significant memory response against tumor recurrence compared to wild-type mice and that vaccine-induced CD8+ IL-10+ T cells may possess regulatory function. Herein, we extended these studies by testing the hypothesis that D52-vaccine-elicited CD8+ IL-10+ T cells represent a distinct T cell population with a regulatory phenotype. C57Black/6J mice deficient in IL-10 or IFN-γ were vaccinated with the murine orthologue of D52; vaccination of wild-type (wt) mice served as a control for comparison. T cells were isolated from all three groups of vaccinated mice, and RNA was extracted from purified CD8+ T cells for deep sequencing and expression analysis. Chemokine receptor 8 (CCR8) and inducible co-stimulator (ICOS) were overexpressed in CD8+ T cells that produced IL-10 but not IFN-γ. These surface markers are associated with IL-10 producing CD4+ T regulatory cells thus supporting the possibility that CD8+ IL-10+ T cells elicited by D52 vaccination represent a unique regulatory T cell subset. The current phenotypic analyses of D52 vaccine elicited CD8+ T cells strengthen our premise that CD8+ IL-10+ T cells elicited by D52 tumor-self protein vaccination likely contribute to the suppression of memory CTL responses and inhibition of durable tumor immunity.
Collapse
Affiliation(s)
- C. Riccay Elizondo
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jennifer D. Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Robert K. Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Cancer Center, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
4
|
Cai W, Shi L, Zhao J, Xu F, Dufort C, Ye Q, Yang T, Dai X, Lyu J, Jin C, Pu H, Yu F, Hassan S, Sun Z, Zhang W, Hitchens TK, Shi Y, Thomson AW, Leak RK, Hu X, Chen J. Neuroprotection against ischemic stroke requires a specific class of early responder T cells in mice. J Clin Invest 2022; 132:157678. [PMID: 35912857 PMCID: PMC9337834 DOI: 10.1172/jci157678] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/17/2022] [Indexed: 12/20/2022] Open
Abstract
Immunomodulation holds therapeutic promise against brain injuries, but leveraging this approach requires a precise understanding of mechanisms. We report that CD8+CD122+CD49dlo T regulatory-like cells (CD8+ TRLs) are among the earliest lymphocytes to infiltrate mouse brains after ischemic stroke and temper inflammation; they also confer neuroprotection. TRL depletion worsened stroke outcomes, an effect reversed by CD8+ TRL reconstitution. The CXCR3/CXCL10 axis served as the brain-homing mechanism for CD8+ TRLs. Upon brain entry, CD8+ TRLs were reprogrammed to upregulate leukemia inhibitory factor (LIF) receptor, epidermal growth factor–like transforming growth factor (ETGF), and interleukin 10 (IL-10). LIF/LIF receptor interactions induced ETGF and IL-10 production in CD8+ TRLs. While IL-10 induction was important for the antiinflammatory effects of CD8+ TRLs, ETGF provided direct neuroprotection. Poststroke intravenous transfer of CD8+ TRLs reduced infarction, promoting long-term neurological recovery in young males or aged mice of both sexes. Thus, these unique CD8+ TRLs serve as early responders to rally defenses against stroke, offering fresh perspectives for clinical translation.
Collapse
Affiliation(s)
- Wei Cai
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ligen Shi
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jingyan Zhao
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fei Xu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Connor Dufort
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qing Ye
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Xuejiao Dai
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Junxuan Lyu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chenghao Jin
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hongjian Pu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Fang Yu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sulaiman Hassan
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Zeyu Sun
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wenting Zhang
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - T Kevin Hitchens
- Animal Imaging Center and Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Hardardottir L, Bazzano MV, Glau L, Gattinoni L, Köninger A, Tolosa E, Solano ME. The New Old CD8+ T Cells in the Immune Paradox of Pregnancy. Front Immunol 2021; 12:765730. [PMID: 34868016 PMCID: PMC8635142 DOI: 10.3389/fimmu.2021.765730] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
CD8+ T cells are the most frequent T cell population in the immune cell compartment at the feto-maternal interface. Due to their cytotoxic potential, the presence of CD8+ T cells in the immune privileged pregnant uterus has raised considerable interest. Here, we review our current understanding of CD8+ T cell biology in the uterus of pregnant women and discuss this knowledge in relation to a recently published immune cell Atlas of human decidua. We describe how the expansion of CD8+ T cells with an effector memory phenotype often presenting markers of exhaustion is critical for a successful pregnancy, and host defense towards pathogens. Moreover, we review new evidence on the presence of long-lasting immunological memory to former pregnancies and discuss its impact on prospective pregnancy outcomes. The formation of fetal-specific memory CD8+ T cell subests in the uterus, in particular of tissue resident, and stem cell memory cells requires further investigation, but promises interesting results to come. Advancing the knowledge of CD8+ T cell biology in the pregnant uterus will be pivotal for understanding not only tissue-specific immune tolerance but also the etiology of complications during pregnancy, thus enabling preventive or therapeutic interventions in the future.
Collapse
Affiliation(s)
- Lilja Hardardottir
- Laboratory for Translational Perinatology- Focus: Immunology, University Department of Obstetrics and Gynecology, University Hospital Regensburg, Regensburg, Germany
| | - Maria Victoria Bazzano
- Laboratory for Translational Perinatology- Focus: Immunology, University Department of Obstetrics and Gynecology, University Hospital Regensburg, Regensburg, Germany
| | - Laura Glau
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Luca Gattinoni
- Department of Functional Immune Cell Modulation, Regensburg Center for Interventional Immunology, Regensburg, Germany
- University of Regensburg, Regensburg, Germany
| | - Angela Köninger
- Department of Obstetrics and Gynecology of the University of Regensburg at the St. Hedwig Hospital of the Order of St. John, Regensburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria Emilia Solano
- Laboratory for Translational Perinatology- Focus: Immunology, University Department of Obstetrics and Gynecology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
6
|
Du Y, Fang Q, Zheng SG. Regulatory T Cells: Concept, Classification, Phenotype, and Biological Characteristics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:1-31. [PMID: 33523440 DOI: 10.1007/978-981-15-6407-9_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Treg) play an indispensable role in maintaining the body's immune nonresponse to self-antigens and suppressing the body's unwarranted and potentially harmful immune responses. Their absence, reduction, dysfunction, transformation, and instability can lead to numerous autoimmune diseases. There are several distinct subtypes of the Treg cells, although they share certain biological characteristics and have unique phenotypes with different regulatory functions, as well as mechanistic abilities. In this book chapter, we introduce the latest advances in Treg cell subtypes pertaining to classification, phenotype, biological characteristics, and mechanisms. We also highlight the relationship between Treg cells and various diseases, including autoimmune, infectious, as well as tumors and organ transplants.
Collapse
Affiliation(s)
- Yang Du
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, Guangxi, China.,Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Qiannan Fang
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Song-Guo Zheng
- Department of Internal Medicine, Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
7
|
Renavikar PS, Sinha S, Brate AA, Borcherding N, Crawford MP, Steward-Tharp SM, Karandikar NJ. IL-12-Induced Immune Suppressive Deficit During CD8+ T-Cell Differentiation. Front Immunol 2020; 11:568630. [PMID: 33193343 PMCID: PMC7657266 DOI: 10.3389/fimmu.2020.568630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/02/2020] [Indexed: 11/13/2022] Open
Abstract
Autoimmune diseases are characterized by regulatory deficit in both the CD4+ and CD8+ T-cell compartments. We have shown that CD8+ T-cells associated with acute relapse of multiple sclerosis are significantly deficient in their immune suppressive ability. We hypothesized that distinct CD8+ cytotoxic T-cell (Tc) lineages, determined by cytokine milieu during naïve T-cell differentiation, may harbor differential ability to suppress effector CD4+ T-cells. We differentiated purified human naïve CD8+ T-cells in vitro toward Tc0 (media control), Tc1 and Tc17 lineages. Using in vitro flow cytometric suppression assays, we observed that Tc0 and Tc17 cells had similar suppressive ability. In contrast, Tc1 cells showed significant loss of suppressive ability against ex vivo CD4+ T-cells and in vitro-differentiated Th0, Th1 and Th17 cells. Of note, Tc1 cells were also suboptimal in suppressing CD4-induced acute xenogeneic graft versus host disease (xGVHD) in vivo. Tc subtypes derived under various cytokine combinations revealed that IL-12-containing conditions resulted in less suppressive cells exhibiting dysregulated cytotoxic degranulation. RNA sequencing transcriptome analyses indicated differential regulation of inflammatory genes and enrichment in GM-CSF-associated pathways. These studies provide insights into the role of T-cell differentiation in CD8 suppressive biology and may reveal therapeutically targetable pathways to reverse suppressive deficit during immune-mediated disease.
Collapse
Affiliation(s)
- Pranav S Renavikar
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Sushmita Sinha
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Ashley A Brate
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Nicholas Borcherding
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Michael P Crawford
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Scott M Steward-Tharp
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| | - Nitin J Karandikar
- Department of Pathology, University of Iowa Health Care, Iowa City, IA, United States
| |
Collapse
|
8
|
Lees JR. CD8+ T cells: The past and future of immune regulation. Cell Immunol 2020; 357:104212. [PMID: 32979764 DOI: 10.1016/j.cellimm.2020.104212] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/16/2020] [Accepted: 09/01/2020] [Indexed: 02/05/2023]
Abstract
Regulation of the adaptive immune response is critical for health. Regulatory activity can be found in multiple components of the immune system, however, the focus on particular components of the immune regulatory network has left many aspects of this critical immune component understudied. Here we review the evidence for activities of CD8+ T cells in immune homeostasis and regulation of autoimmune reactivity. The heterogeneous nature of identified CD8+ cell types are examined, and common phenotypes associated with functional activities are defined. The varying types of antigen signal crucial for CD8+ T cell regulatory activity are identified and the implications of these activation pathways for control of adaptive responses is considered. Finally, the promising capacity for transgenic antigen receptor directed cytotoxicity as a mechanism for modulation of autoimmunity is detailed.
Collapse
Affiliation(s)
- Jason R Lees
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| |
Collapse
|
9
|
Elizondo CR, Bright JD, Byrne JA, Bright RK. Analysis of the CD8+ IL-10+ T cell response elicited by vaccination with the oncogenic tumor-self protein D52. Hum Vaccin Immunother 2020; 16:1413-1423. [PMID: 31769704 DOI: 10.1080/21645515.2019.1689746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Development of cancer vaccines targeting tumor self-antigens is complex and challenging due to the difficulty of overcoming immune tolerance to self-proteins. Vaccination against tumor self-protein D52 (D52) has been successful, although complete protection appears impaired by immune regulation. Our previous studies suggest that vaccine elicited CD8 + T cells producing interleukin 10 (IL-10) may have a negative impact on tumor protection. Understanding the role CD8+ IL-10 + T cells play in the immune response following vaccination with D52 could result in a more potent vaccine. To address this, we vaccinated IL-10 deficient mice with the murine orthologue of D52; vaccination of wild type (wt) C57BL/6J served as a control for comparison. In separate experiments, D52 vaccinated wt mice were administered IL-10R-specific mAb to neutralize IL-10 function. Interestingly, we observed similar protection against primary tumor challenge in the experimental groups compared to the controls. However, individual IL-10 deficient mice that rejected the primary tumor challenge were re-challenged 140 days post-primary challenge to access vaccine durability and immunologic memory against tumor recurrence. Mice deficient in IL-10 demonstrated a memory response in which 100% of the mice were protected from secondary tumor challenge, while wt mice had diminished recall response (25%) against tumor recurrence. These results with analysis of vaccine-elicited CD8 + T cells for tumor-specific killing and regulatory cell marker expression, add further support to our premise that CD8+ IL-10 + T cells elicited by D52 tumor-self protein vaccine contribute to the suppression of a memory CTL responses and durable tumor immunity.
Collapse
Affiliation(s)
- C Riccay Elizondo
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center , Lubbock, TX, USA
| | - Jennifer D Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center , Lubbock, TX, USA
| | - Jennifer A Byrne
- Faculty of Medicine and Health, The University of Sydney , Westmead, Australia
| | - Robert K Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center , Lubbock, TX, USA.,Cancer Center, Texas Tech University Health Sciences Center , Lubbock, TX, USA
| |
Collapse
|
10
|
The Traditional Chinese Medicine Fufang Shatai Heji (STHJ) Enhances Immune Function in Cyclophosphamide-Treated Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3849847. [PMID: 32063984 PMCID: PMC6998758 DOI: 10.1155/2020/3849847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022]
Abstract
Fufang Shatai Heji (STHJ) is a mixture of traditional Chinese medicines, such as Radix Adenophorae, Radix Pseudostellariae, and Radix Astragali. STHJ is commonly used to treat diseases caused by low immune function, for example, Sjögren's syndrome (SS). The primary objective of this study was to assess the immunopotentiating effect of STHJ using an immunosuppressive mouse model receiving cyclophosphamide (CTX). Following CTX treatment, STHJ was administered by oral gavage for 30 consecutive days. The percentage of specific lymphocyte subpopulations in the spleen was measured by flow cytometry. Levels of inflammatory factors in serum were detected by enzyme-linked immunosorbent assays (ELISAs). The administration of STHJ significantly elevated thymus and spleen indices, increased B cell and natural killer (NK) cell activities, and decreased CD8+ T, CD8+CD122+ T, NKT, and γδT cell activities in the CTX-treated mice. In addition, STHJ upregulated the expression of interleukin- (IL-) 2, IL-6, and tumor necrosis factor-α (TNF-α) and downregulated IL-10 expression in CTX-treated mice. In conclusion, STHJ effectively remitted CTX-induced immunosuppression by modulating the balance of lymphocyte subsets and cytokines. Our results suggest STHJ treatment could be used as an effective therapeutic approach to improve immune function in patients with low immunity.
Collapse
|
11
|
Liu H, Qiu F, Wang Y, Zeng Q, Liu C, Chen Y, Liang CL, Zhang Q, Han L, Dai Z. CD8+CD122+PD-1+ Tregs Synergize With Costimulatory Blockade of CD40/CD154, but Not B7/CD28, to Prolong Murine Allograft Survival. Front Immunol 2019; 10:306. [PMID: 30863408 PMCID: PMC6399415 DOI: 10.3389/fimmu.2019.00306] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/06/2019] [Indexed: 01/22/2023] Open
Abstract
A transplanted organ is always rejected in the absence of any immunosuppressive treatment due to vigorous alloimmunity. However, continuously global immunosuppression with a conventional immunosuppressant may result in severe side effects, including nephrotoxicity, tumors and infections. Tregs have been widely used to inhibit allograft rejection, especially in animal models. However, it's well accepted that administration of Tregs alone is not satisfactory in immune-competent wild-type animals. Therefore, it's imperative to promote Treg therapies under the cover of other approaches, including costimulatory blockade. In the present study, we demonstrated that administration of in vitro-expanded CD8+CD122+PD-1+ Tregs synergized with costimulatory blockade of CD40/CD154, but not B7/CD28, to prolong skin allograft survival in wild-type mice and to reduce cellular infiltration in skin allografts as well. Treg treatment and blockade of CD40/CD154, but not B7/CD28, also exhibited an additive effect on suppression of T cell proliferation in vitro and pro-inflammatory cytokine expression in skin allografts. Importantly, blocking B7/CD28, but not CD40/CD154, costimulation decreased the number of transferred CD8+CD122+PD-1+ Tregs and their expression of IL-10 in recipient mice. Furthermore, it's B7/CD28, but not CD40/CD154, costimulatory blockade that dramatically reduced IL-10 production by CD8+CD122+PD-1+ Tregs in vitro, suggesting that B7/CD28, but not CD40/CD154, costimulation is critical for their production of IL-10. Indeed, infusion of IL-10-deficient CD8+CD122+PD-1+ Tregs failed to synergize with anti-CD154 Ab treatment to further prolong allograft survival. Our data may explain why blocking B7/CD28 costimulatory pathway does not boost IL-10-dependent Treg suppression of alloimmunity. Thus, these findings could be implicated in clinical organ transplantation.
Collapse
Affiliation(s)
- Huazhen Liu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Feifei Qiu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuanzhong Wang
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Qiaohuang Zeng
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Cuihua Liu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuchao Chen
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chun-Ling Liang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qunfang Zhang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ling Han
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhenhua Dai
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
12
|
Shao S, Yu X, Shen L. Autoimmune thyroid diseases and Th17/Treg lymphocytes. Life Sci 2017; 192:160-165. [PMID: 29158050 DOI: 10.1016/j.lfs.2017.11.026] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/10/2017] [Accepted: 11/16/2017] [Indexed: 12/14/2022]
Abstract
Years of researches have demonstrated that the imbalance of Th17 and Tregs contribute to the thyroid autoimmunity and the severity of autoimmune thyroid disease (AITD). The underlying mechanism comprises inherent genetic predisposition, abnormality of Th17 and Treg related biological molecules, and gut microbiota disorder. New therapeutic strategies have been developed to improve the Th17/Treg equilibrium, including regulation of intracellular signaling pathways, neutralization of Th17-related cytokines, as well as manipulation of Th17 and Treg specific transcription factors. Although a few of these agents are applied into AITD, the clinic prospect is promising.
Collapse
Affiliation(s)
- Shiying Shao
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Liya Shen
- Department of Geriatrics, Affiliated Hospital of Jianghan University, Wuhan 430015, PR China.
| |
Collapse
|
13
|
Han R, Yang X, Chen M, Zhang X, Yuan Y, Hu X, Wang M, Liu R, Ma Y, Yang J, Xu S, Shuai Z, Jiang S, Pan F. Changes and clinical significance of CD8+CD122+ T cells in the peripheral blood of patients with ankylosing spondylitis. Clin Rheumatol 2017; 37:639-646. [DOI: 10.1007/s10067-017-3887-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 01/01/2023]
|
14
|
Du W, Dong Q, Lu X, Liu X, Wang Y, Li W, Pan Z, Gong Q, Liang C, Gao G. Iodine-131 therapy alters the immune/inflammatory responses in the thyroids of patients with Graves' disease. Exp Ther Med 2017; 13:1155-1159. [PMID: 28450957 DOI: 10.3892/etm.2017.4047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/01/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to evaluate the serum levels of interleukin-6 (IL-6), CXC chemokine ligand-10 (CXCL-10) and intercellular adhesion molecule-l (ICAM-1) in patients with Graves' disease (GD) following iodine-131 (131I) therapy. A total of 30 patients with GD participated in the present study. Serum cytokine levels were measured with ELISA, and correlation analyses were performed. Serum levels of IL-6, CXCL-10 and ICAM-1 were significantly higher in patients with GD prior to treatment than those in the control subjects (P<0.01). Following 131I therapy, the serum levels of IL-6 and CXCL-10 in patients with GD were markedly increased within the first week, gradually decreased to the pretreatment level in the subsequent six months and decreased further at 18 months post-treatment. However, the serum levels of IL-6 and CXCL-10 in patients with GD at 18 months following 131I therapy remained significantly higher than in control subjects (P<0.01). Conversely, serum ICAM-1 levels in patients with GD were gradually increased in the 12 months following 131I therapy and reached a relatively stable level thereafter. Furthermore, the Pearson's correlation analysis indicated that the serum levels of IL-6, CXCL-10 and ICAM-1 were not associated with free triiodothyronine, the free thyroxine index, and thyroid-stimulating hormone in these patients. 131I therapy was able to alter the immune/inflammatory responses in the thyroids of patients with GD. However, these cytokines (IL-6, CXCL-10, and ICAM-1) are not associated with thyroid function; therefore, they cannot be used as prognostic markers for the 131I therapy of GD.
Collapse
Affiliation(s)
- Wenhua Du
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Qingyu Dong
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Xiaoting Lu
- Department of Clinical Medicine, Taishan Medical University, Taian, Shandong 271000, P.R. China
| | - Xiaomeng Liu
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yueli Wang
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Wenxia Li
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Zhenyu Pan
- Department of Internal Medicine, Shandong Medical College, Linyi, Shandong 276000, P.R. China
| | - Qian Gong
- Department of Internal Medicine, Shandong Medical College, Linyi, Shandong 276000, P.R. China
| | - Cuige Liang
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Guanqi Gao
- Department of Endocrinology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
15
|
Sinha S, Boyden AW, Itani FR, Crawford MP, Karandikar NJ. CD8(+) T-Cells as Immune Regulators of Multiple Sclerosis. Front Immunol 2015; 6:619. [PMID: 26697014 PMCID: PMC4674574 DOI: 10.3389/fimmu.2015.00619] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/26/2015] [Indexed: 11/13/2022] Open
Abstract
The vast majority of studies regarding the immune basis of MS (and its animal model, EAE) have largely focused on CD4(+) T-cells as mediators and regulators of disease. Interestingly, CD8(+) T-cells represent the predominant T-cell population in human MS lesions and are oligoclonally expanded at the site of pathology. However, their role in the autoimmune pathologic process has been both understudied and controversial. Several animal models and MS patient studies support a pathogenic role for CNS-specific CD8(+) T-cells, whereas we and others have demonstrated a regulatory role for these cells in disease. In this review, we describe studies that have investigated the role of CD8(+) T-cells in MS and EAE, presenting evidence for both pathogenic and regulatory functions. In our studies, we have shown that cytotoxic/suppressor CD8(+) T-cells are CNS antigen-specific, MHC class I-restricted, IFNγ- and perforin-dependent, and are able to inhibit disease. The clinical relevance for CD8(+) T-cell suppressive function is best described by a lack of their function during MS relapse, and importantly, restoration of their suppressive function during quiescence. Furthermore, CD8(+) T-cells with immunosuppressive functions can be therapeutically induced in MS patients by glatiramer acetate (GA) treatment. Unlike CNS-specific CD8(+) T-cells, these immunosuppressive GA-induced CD8(+) T-cells appear to be HLA-E restricted. These studies have provided greater fundamental insight into the role of autoreactive as well as therapeutically induced CD8(+) T-cells in disease amelioration. The clinical implications for these findings are immense and we propose that this natural process can be harnessed toward the development of an effective immunotherapeutic strategy.
Collapse
Affiliation(s)
- Sushmita Sinha
- Department of Pathology, University of Iowa , Iowa City, IA , USA
| | | | - Farah R Itani
- Department of Pathology, University of Iowa , Iowa City, IA , USA
| | | | | |
Collapse
|
16
|
Liu J, Chen D, Nie GD, Dai Z. CD8(+)CD122(+) T-Cells: A Newly Emerging Regulator with Central Memory Cell Phenotypes. Front Immunol 2015; 6:494. [PMID: 26539191 PMCID: PMC4610204 DOI: 10.3389/fimmu.2015.00494] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/10/2015] [Indexed: 11/16/2022] Open
Abstract
CD8(+)CD122(+) T-cells have been traditionally described as antigen-specific memory T-cells that respond to previously encountered antigens more quickly and vigorously than their naïve counterparts. However, mounting evidence has demonstrated that murine CD8(+)CD122(+) T-cells exhibit a central memory phenotype (CD44(high)CD62L(high)), regulate T cell homeostasis, and act as regulatory T-cells (Treg) by suppressing both autoimmune and alloimmune responses. Importantly, naturally occurring murine CD8(+)CD122(+) Tregs are more potent in immunosuppression than their CD4(+)CD25(+) counterparts. They appear to be acting in an antigen-non-specific manner. Human CD8(+)CXCR3(+) T-cells are the equivalent of murine CD8(+)CD122(+) Tregs and also exhibit central memory phenotypes. In this mini-review article, we will summarize recent progresses in their phenotypes, homeostatic expansion, antigen-specificity, roles in the suppression of alloimmune and autoimmune responses, and the mechanisms underlying their inhibitory function.
Collapse
Affiliation(s)
- Junfeng Liu
- Section of Immunology, Division of Dermatology, Second Affiliated Hospital, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dacan Chen
- Section of Immunology, Division of Dermatology, Second Affiliated Hospital, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Golay D. Nie
- School of Medicine, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Zhenhua Dai
- Section of Immunology, Division of Dermatology, Second Affiliated Hospital, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
Abstract
The role of CD8+ T cells in the process of autoimmune pathology has been both understudied and controversial. Multiple sclerosis (MS) is an inflammatory, demyelinating disorder of the central nervous system (CNS) with underlying T cell-mediated immunopathology. CD8+ T cells are the predominant T cells in human MS lesions, showing oligoclonal expansion at the site of pathology. It is still unclear whether these cells represent pathogenic immune responses or disease-regulating elements. Through studies in human MS and its animal model, experimental autoimmune encephalomyelitis (EAE), we have discovered two novel CD8+ T cell populations that play an essential immunoregulatory role in disease: (1) MHC class Ia-restricted neuroantigen-specific "autoregulatory" CD8+ T cells and (2) glatiramer acetate (GA/Copaxone(®)) therapy-induced Qa-1/HLA-E-restricted GA-specific CD8+ T cells. These CD8+ Tregs suppress proliferation of pathogenic CD4+ CD25- T cells when stimulated by their cognate antigens. Similarly, CD8+ Tregs significantly suppress EAE when transferred either pre-disease induction or during peak disease. The mechanism of disease inhibition depends, at least in part, on an antigen-specific, contact-dependent process and works through modulation of CD4+ T cell responses as well as antigen-presenting cells through a combination of cytotoxicity and cytokine-mediated modulation. This review provides an overview of our understanding of CD8+ T cells in immune-mediated disease, focusing particularly on our findings regarding regulatory CD8+ T cells both in MS and in EAE. Clinical relevance of these novel CD8-regulatory populations is discussed, providing insights into a potentially intriguing, novel therapeutic strategy for these diseases.
Collapse
|
18
|
Arndt B, Witkowski L, Ellwart J, Seissler J. CD8+ CD122+ PD-1- effector cells promote the development of diabetes in NOD mice. J Leukoc Biol 2014; 97:111-20. [PMID: 25387835 DOI: 10.1189/jlb.3a0613-344rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
It is well established that CD4 and CD8 T cells are required for the initiation of autoimmune diabetes in NOD mice. However, different subsets of CD4 or CD8 cells may play different roles in the initiation of insulitis. In this study, we evaluated the role of the previously described CD8(+) CD122(+) in this process. We found that prediabetic NOD mice have an almost 50% reduction of CD8(+) CD122(+) T cells in their secondary lymphoid organs compared with BL/6 or Balb/c mouse strains. This reduction is explained by the lack of the regulatory CD8(+) CD122(+) PD-1(+) cell population in the NOD mice, as we found that all CD8(+) CD122(+) T cells from prediabetic NOD mice lack PD-1 expression and regulatory function. Depletion of CD8(+) CD122(+) PD-1(-) cells through injection of anti-CD122 mAb in prediabetic female NOD mice reduced the infiltration of mononuclear cells into the Langerhans islets and delayed the onset and decreased the incidence of overt diabetes. In addition, we found that transfer of highly purified and activated CD8(+) CD122(+) PD-1(-) cells, together with diabetogenic splenocytes from NOD donors to NOD SCID recipients, accelerates the diabetes development in these mice. Together, these results demonstrate that CD8(+) CD122(+) PD-1(-) T cells from NOD mice are effector cells that are involved in the pathogenesis of autoimmune diabetes.
Collapse
Affiliation(s)
- Börge Arndt
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| | - Lukas Witkowski
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| | - Joachim Ellwart
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| | - Jochen Seissler
- *Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; Medizinische Klinik und Poliklinik III, Campus Grosshadern, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany; and Helmholtz Zentrum München, Institute of Molecular Immunology (Hämatologikum), Munich, Germany
| |
Collapse
|
19
|
Li S, Xie Q, Zeng Y, Zou C, Liu X, Wu S, Deng H, Xu Y, Li XC, Dai Z. A naturally occurring CD8(+)CD122(+) T-cell subset as a memory-like Treg family. Cell Mol Immunol 2014; 11:326-31. [PMID: 24793406 PMCID: PMC4085522 DOI: 10.1038/cmi.2014.25] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 01/07/2023] Open
Abstract
Despite extensive studies on CD4(+)CD25(+) regulatory T cells (Tregs) during the past decade, the progress on their clinical translation remains stagnant. Mounting evidence suggests that naturally occurring CD8(+)CD122(+) T cells are also Tregs with the capacity to inhibit T-cell responses and suppress autoimmunity as well as alloimmunity. In fact, they are memory-like Tregs that resemble a central memory T cell (TCM) phenotype. The mechanisms underlying their suppression are still not well understood, although they may include IL-10 production. We have recently demonstrated that programmed death-1 (PD-1) expression distinguishes between regulatory and memory CD8(+)CD122(+) T cells and that CD8(+)CD122(+) Tregs undergo faster homeostatic proliferation and are more potent in the suppression of allograft rejection than conventional CD4(+)CD25(+) Tregs. These findings may open a new line of investigation for accelerating effective Treg therapies in the clinic. In this review, we summarize the significant progress in this promising field of CD8(+)CD122(+) Treg research and discuss their phenotypes, suppressive roles in autoimmunity and alloimmunity, functional requirements, mechanisms of action and potential applications in the clinic.
Collapse
Affiliation(s)
- Shanshan Li
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Qingfeng Xie
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Yuqun Zeng
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Chuan Zou
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xusheng Liu
- Department of Nephrology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Shouhai Wu
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Haixia Deng
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Yang Xu
- Section of Immunology, Center for Regenerative and Translational Medicine
| | - Xian C Li
- Immunobiology and Transplantation Research Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Zhenhua Dai
- Section of Immunology, Center for Regenerative and Translational Medicine
| |
Collapse
|
20
|
Robinson MV, Obut TA, Melnikova EV, Trufakin VA. Parameters of cellular and humoral immunity in experimental hyperthyroidism and its correction. Bull Exp Biol Med 2014; 156:473-5. [PMID: 24771430 DOI: 10.1007/s10517-014-2377-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Indexed: 11/28/2022]
Abstract
We demonstrate changes in cellular and humoral immunity in animals with experimental hyperthyroidism induced by chronic administration of potassium iodide (KI) solution. KI increased the weight and cellularity of the thymus and spleen and number of antibody-forming cells to sheep red blood cells and modified the relative content of T cell subpopulations. Phytosorption complex LimfoFit modifying cellular and humoral immunity affected only its individual parameters in hyperthyroid animals.
Collapse
Affiliation(s)
- M V Robinson
- Research Institute of Clinical and Experimental Lymphology, Siberian Division of Russian Academy of Medical Sciences, Novosibirsk, Russia,
| | | | | | | |
Collapse
|
21
|
McLachlan SM, Rapoport B. Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr Rev 2014; 35:59-105. [PMID: 24091783 PMCID: PMC3895862 DOI: 10.1210/er.2013-1055] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/24/2013] [Indexed: 02/06/2023]
Abstract
Thyroid autoimmunity involves loss of tolerance to thyroid proteins in genetically susceptible individuals in association with environmental factors. In central tolerance, intrathymic autoantigen presentation deletes immature T cells with high affinity for autoantigen-derived peptides. Regulatory T cells provide an alternative mechanism to silence autoimmune T cells in the periphery. The TSH receptor (TSHR), thyroid peroxidase (TPO), and thyroglobulin (Tg) have unusual properties ("immunogenicity") that contribute to breaking tolerance, including size, abundance, membrane association, glycosylation, and polymorphisms. Insight into loss of tolerance to thyroid proteins comes from spontaneous and induced animal models: 1) intrathymic expression controls self-tolerance to the TSHR, not TPO or Tg; 2) regulatory T cells are not involved in TSHR self-tolerance and instead control the balance between Graves' disease and thyroiditis; 3) breaking TSHR tolerance involves contributions from major histocompatibility complex molecules (humans and induced mouse models), TSHR polymorphism(s) (humans), and alternative splicing (mice); 4) loss of tolerance to Tg before TPO indicates that greater Tg immunogenicity vs TPO dominates central tolerance expectations; 5) tolerance is induced by thyroid autoantigen administration before autoimmunity is established; 6) interferon-α therapy for hepatitis C infection enhances thyroid autoimmunity in patients with intact immunity; Graves' disease developing after T-cell depletion reflects reconstitution autoimmunity; and 7) most environmental factors (including excess iodine) "reveal," but do not induce, thyroid autoimmunity. Micro-organisms likely exert their effects via bystander stimulation. Finally, no single mechanism explains the loss of tolerance to thyroid proteins. The goal of inducing self-tolerance to prevent autoimmune thyroid disease will require accurate prediction of at-risk individuals together with an antigen-specific, not blanket, therapeutic approach.
Collapse
Affiliation(s)
- Sandra M McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute, and University of California-Los Angeles School of Medicine, Los Angeles, California 90048
| | | |
Collapse
|
22
|
Su J, Xie Q, Xu Y, Li XC, Dai Z. Role of CD8(+) regulatory T cells in organ transplantation. BURNS & TRAUMA 2014; 2:18-23. [PMID: 27574642 PMCID: PMC4994507 DOI: 10.4103/2321-3868.126086] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
CD8+ T cells are regulatory T cells (Tregs) that suppress both alloimmunity and autoimmunity in many animal models. This class of regulatory cells includes the CD8+CD28−, CD8+CD103+, CD8+FoxP3+ and CD8+CD122+ subsets. The mechanisms of action of these regulatory cells are not fully understood; however, the secretion of immunosuppressive cytokines, such as interleukin (IL)-4, IL-10 and transforming growth factor beta (TGF-β) as well as the direct killing of target cells via Fas L/Fas and the perforin/granzyme B pathways have been demonstrated in various models. Further studies are necessary to fully understand the mechanisms underlying the suppressive effects of Tregs and to provide experimental support for potential clinical trials. We recently observed that CD8+CD122+ Tregs more potently suppressed allograft rejection compared to their CD4+CD25+ counterparts, supporting the hypothesis that CD8+ Tregs may represent a new and promising Treg family that can be targeted to prevent allograft rejection in the clinic. In this review, we summarize the progress in the field during the past 7–10 years and discuss CD8+ Treg phenotypes, mechanisms of action, and their potential clinical applications; particularly in composite tissue transplants in burn and trauma patients.
Collapse
Affiliation(s)
- Jiyan Su
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine and Guangzhou University of Chinese Medicine School of Chinese Material Medica, Guangzhou, Guangdong, China
| | - Qingfeng Xie
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine and Guangzhou University of Chinese Medicine School of Chinese Material Medica, Guangzhou, Guangdong, China
| | - Yang Xu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine and Guangzhou University of Chinese Medicine School of Chinese Material Medica, Guangzhou, Guangdong, China
| | - Xian C Li
- Immunobiology and Transplantation Research Center, Houston, Methodist Research Institute, Houston, Texas USA ; Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine and Guangzhou University of Chinese Medicine School of Chinese Material Medica, Guangzhou, Guangdong, China
| | - Zhenhua Dai
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine and Guangzhou University of Chinese Medicine School of Chinese Material Medica, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
The role of T cell immunoglobulin mucin domains 1 and 4 in a herpes simplex virus-induced Behçet's disease mouse model. Mediators Inflamm 2013; 2013:903948. [PMID: 24453431 PMCID: PMC3888750 DOI: 10.1155/2013/903948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/24/2013] [Accepted: 11/18/2013] [Indexed: 01/04/2023] Open
Abstract
The T cell immunoglobulin mucin (TIM) proteins regulate T cell activation and tolerance. TIM-1 plays an important role in the regulation of immune responses and the development of autoimmune diseases. TIM-4 is a natural ligand of TIM-1, and the interaction of TIM-1 and TIM-4 is involved in the regulation of T helper (Th) cell responses and modulation of the Th1/Th2 cytokine balance. Behçet's disease (BD) is a chronic, multisystemic inflammatory disorder with arthritic, intestinal, mucocutaneous, ocular, vascular, and central nervous system involvement. Tim-1 expression was lower in a herpes simplex virus-induced BD mouse model compared to that in asymptomatic BD normal (BDN) mice. Tim-4 expression was higher in BD mice than that in BDN mice. In this study, we investigated the Tim expression in a BD mouse model with BD-like symptoms. Tim-1 and Tim-4 expression was regulated by an expression vector or siRNA injected into the BD mouse model. The Tim-1 vector injected into BD mice resulted in changes in BD-like symptoms and decreased the severity score. Treatment with Tim-4 siRNA also improved BD-like symptoms and decreased the severity score accompanied by upregulation of regulatory T cells. We showed that regulating Tim-1 or Tim-4 affected BD-like symptoms in mice.
Collapse
|
24
|
Iwata H, Bieber K, Tiburzy B, Chrobok N, Kalies K, Shimizu A, Leineweber S, Ishiko A, Vorobyev A, Zillikens D, Köhl J, Westermann J, Seeger K, Manz R, Ludwig RJ. B Cells, Dendritic Cells, and Macrophages Are Required To Induce an Autoreactive CD4 Helper T Cell Response in Experimental Epidermolysis Bullosa Acquisita. THE JOURNAL OF IMMUNOLOGY 2013; 191:2978-88. [DOI: 10.4049/jimmunol.1300310] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Okuno Y, Murakoshi A, Negita M, Akane K, Kojima S, Suzuki H. CD8+ CD122+ regulatory T cells contain clonally expanded cells with identical CDR3 sequences of the T-cell receptor β-chain. Immunology 2013; 139:309-17. [PMID: 23317140 PMCID: PMC3701177 DOI: 10.1111/imm.12067] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 12/26/2012] [Accepted: 01/07/2013] [Indexed: 12/23/2022] Open
Abstract
We identified CD8(+) CD122(+) regulatory T cells (CD8(+) CD122(+) Treg cells) and reported their importance in maintaining immune homeostasis. The absence of CD8(+) CD122(+) Treg cells has been shown to lead to severe systemic autoimmunity in several mouse models, including inflammatory bowel diseases and experimental autoimmune encephalomyelitis. The T-cell receptors (TCRs) expressed on CD8(+) CD122(+) Treg cells recognize the target cells to be regulated. To aid in the identification of the target antigen(s) recognized by TCRs of CD8(+) CD122(+) Treg cells, we compared the TCR diversity of CD8(+) CD122(+) T cells with that of conventional, naive T cells in mice. We analysed the use of TCR-Vβ in the interleukin 10-producing population of CD8(+) CD122(+) T cells marked by high levels of CD49d expression, and found the significantly increased use of Vβ13 in these cells. Immunoscope analysis of the complementarity-determining region 3 (CDR3) of the TCR β-chain revealed remarkable skewing in a pair of Vβ regions, suggesting the existence of clonally expanded cells in CD8(+) CD122(+) T cells. Clonal expansion in Vβ13(+) cells was confirmed by determining the DNA sequences of the CDR3s. The characteristic TCR found in this study is an important building block for further studies to identify the target antigen recognized by CD8(+) CD122(+) Treg cells.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/physiopathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Clone Cells/metabolism
- Complementarity Determining Regions/genetics
- Female
- Humans
- Interleukin-10/metabolism
- Interleukin-2 Receptor beta Subunit/metabolism
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Polymerase Chain Reaction/methods
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Sequence Analysis, DNA
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Yusuke Okuno
- Department of Paediatrics, Nagoya University Graduate School of MedicineNagoya, Japan
- Department of Immunology, Nagoya University Graduate School of MedicineNagoya, Japan
| | - Ayako Murakoshi
- Department of Paediatrics, Nagoya University Graduate School of MedicineNagoya, Japan
- Department of Immunology, Nagoya University Graduate School of MedicineNagoya, Japan
| | - Masashi Negita
- Department of Immunology, Nagoya University Graduate School of MedicineNagoya, Japan
| | - Kazuyuki Akane
- Department of Paediatrics, Nagoya University Graduate School of MedicineNagoya, Japan
| | - Seiji Kojima
- Department of Paediatrics, Nagoya University Graduate School of MedicineNagoya, Japan
| | - Haruhiko Suzuki
- Department of Immunology, Nagoya University Graduate School of MedicineNagoya, Japan
| |
Collapse
|
26
|
Choi J, Lee ES, Choi B, Sohn S. Therapeutic potency of Poly I:C in HSV-induced inflammation through up-regulation of IL-15 receptor alpha. Immunobiology 2013; 218:1119-30. [PMID: 23618691 DOI: 10.1016/j.imbio.2013.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 03/20/2013] [Indexed: 11/19/2022]
Abstract
Interleukin-15 receptor alpha (IL-15Rα) forms stable complex with IL-15 on the cell surface of activated monocytes and mediates the proliferation of memory CD8+ T cells. Recent studies informed that polyinosinic:polycytidylic acid (Poly I:C) is an immunostimulant which boosts the generation of memory T cells through induction of IL-15Rα. The aim of this study is to evaluate the relevance of IL-15Rα in Herpes simplex virus (HSV)-induced Behçet's disease (BD) mouse model and BD patients. The frequencies of IL-15Rα expression in PBMCs of BD patients and BD-like symptomatic mice were analyzed by flow cytometry. In addition, Poly I:C supplementation could reduce inflammation through the up-regulation of memory T cells and IL-15Rα+ cells accompany with down-regulation of pro-inflammatory cytokine, IL-17A in BD mice. In BD patients, the frequencies of IL-15Rα expression in PBMCs were also significantly different between the inactive and active disease states. These results suggest that IL-15Rα is a relevant factor in BD.
Collapse
Affiliation(s)
- Juyoung Choi
- Laboratory of Cell Biology, Institute for Medical Sciences, Ajou University, Suwon, Republic of Korea
| | | | | | | |
Collapse
|
27
|
Rabinowitz K, Mayer L. Working out mechanisms of controlled/physiologic inflammation in the GI tract. Immunol Res 2013; 54:14-24. [PMID: 22466933 DOI: 10.1007/s12026-012-8315-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The mucosal immune system is distinct from its systemic counterpart by virtue of its enormous antigenic exposure (commensal flora, food antigen, pathogens). Despite this, the mucosal immune system maintains a response defined as controlled or physiologic inflammation. This is regulated by many different mechanisms, among which there are physical, cellular and soluble factors. Our laboratory has focused on unique Tregs in the gut controlled by, in one instance, intestinal epithelial cells that serve as non-professional antigen-presenting cells. We believe that intestinal epithelial cells, expressing classical and non-classical MHC molecules, serve to activate Tregs and thus maintain controlled or physiologic inflammation. In this review, we describe regulatory cytokines and T cells that are one part of the emphasis of our laboratory.
Collapse
Affiliation(s)
- Keren Rabinowitz
- Mount Sinai School of Medicine, Immunology Institute, 1425 Madison Avenue, Box 1089, New York, NY 10029, USA
| | | |
Collapse
|
28
|
Morshed SA, Latif R, Davies TF. Delineating the autoimmune mechanisms in Graves' disease. Immunol Res 2013; 54:191-203. [PMID: 22434518 DOI: 10.1007/s12026-012-8312-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The immunologic processes involved in autoimmune thyroid disease (AITD), particularly Graves' disease (GD), are similar to other autoimmune diseases with the emphasis on the antibodies as the most unique aspect. These characteristics include a lymphocytic infiltrate at the target organs, the presence of antigen-reactive T and B cells and antibodies, and the establishment of animal models of GD by antibody transfer or immunization with antigen. Similar to other autoimmune diseases, risk factors for GD include the presence of multiple susceptibility genes, including certain HLA alleles, and the TSHR gene itself. In addition, a variety of known risk factors and precipitators have been characterized including the influence of sex and sex hormones, pregnancy, stress, infection, iodine and other potential environmental factors. The pathogenesis of GD is likely the result of a breakdown in the tolerance mechanisms, both at central and peripheral levels. Different subsets of T and B cells together with their regulatory populations play important roles in the propagation and maintenance of the disease process. Understanding different mechanistic in the complex system biology interplay will help to identify unique factors contributing to the AITD pathogenesis.
Collapse
Affiliation(s)
- Syed A Morshed
- Thyroid Research Unit, Mount Sinai School of Medicine, James J. Peters VA Medical Center, 130 West Kingsbridge Rd, Bronx, New York, NY 10468, USA.
| | | | | |
Collapse
|
29
|
Kucharska AM, Gorska E, Wasik M, Demkow U. Expression of cytotoxic T lymphocyte antigen-4 in T cells from children with Hashimoto's thyroiditis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 756:163-8. [PMID: 22836632 DOI: 10.1007/978-94-007-4549-0_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The cytotoxic T lymphocyte antigen-4 (CTLA-4) (CD152) is a basic negative regulatory molecule of T cell activation and its hypo-function is associated with severe lymphoproliferative syndrome. The aim of the present study was to evaluate the intracellular and surface expression of CTLA-4 on peripheral T cells before and after T cell activation in children with Hashimoto's thyroiditis (HT). Blood samples were obtained from 46 children: 25 with Hashimoto's thyroiditis and 21 controls free of autoimmune disease or thyroid disorders. T cell phenotype was evaluated by flow cytometry with the use of monoclonal antibodies combination: CD4- FITC/ CD28 -PC5/ CD152 -PE and CD8 -FITC/ CD28 -PC5/ CD152 -PE on T cell surface and intracellularly at baseline and after 48 h of T cell culture with the mitogen 48-PHA. We found that the number of T cells with intracellular CD152 expression was comparable in HT patients and controls at baseline and increased after 48-PHA, in CD4 subset only, in both patients and controls. However, the increase was more evident in the HT patients. The number of T cells with the surface expression of CD152 at baseline was significantly lower in the HT patients than in controls (p < 0.0002) in non-stimulated CD4+ and CD8+ T cells. After 48-PHA, surface CD152 expression in CD4+T cells increased in both groups; the increase was greater in controls. In conclusion, impaired function of CTLA-4 in HT patients may depend on the imbalance of intracellular/surface expression of CD152 in T cells.
Collapse
Affiliation(s)
- Anna M Kucharska
- Department of Pediatrics and Endocrinology, Medical University of Warsaw, Warsaw, Poland.
| | | | | | | |
Collapse
|
30
|
Breaking tolerance in transgenic mice expressing the human TSH receptor A-subunit: thyroiditis, epitope spreading and adjuvant as a 'double edged sword'. PLoS One 2012; 7:e43517. [PMID: 22970131 PMCID: PMC3436763 DOI: 10.1371/journal.pone.0043517] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/23/2012] [Indexed: 02/05/2023] Open
Abstract
Transgenic mice with the human thyrotropin-receptor (TSHR) A-subunit targeted to the thyroid are tolerant of the transgene. In transgenics that express low A-subunit levels (Lo-expressors), regulatory T cell (Treg) depletion using anti-CD25 before immunization with adenovirus encoding the A-subunit (A-sub-Ad) breaks tolerance, inducing extensive thyroid lymphocytic infiltration, thyroid damage and antibody spreading to other thyroid proteins. In contrast, no thyroiditis develops in Hi-expressor transgenics or wild-type mice. Our present goal was to determine if thyroiditis could be induced in Hi-expressor transgenics using a more potent immunization protocol: Treg depletion, priming with Complete Freund's Adjuvant (CFA) + A-subunit protein and further Treg depletions before two boosts with A-sub-Ad. As controls, anti-CD25 treated Hi- and Lo-expressors and wild-type mice were primed with CFA+ mouse thyroglobulin (Tg) or CFA alone before A-sub-Ad boosting. Thyroiditis developed after CFA+A-subunit protein or Tg and A-sub-Ad boosting in Lo-expressor transgenics but Hi- expressors (and wild-type mice) were resistant to thyroiditis induction. Importantly, in Lo-expressors, thyroiditis was associated with the development of antibodies to the mouse TSHR downstream of the A-subunit. Unexpectedly, we observed that the effect of bacterial products on the immune system is a “double-edged sword”. On the one hand, priming with CFA (mycobacteria emulsified in oil) plus A-subunit protein broke tolerance to the A-subunit in Hi-expressor transgenics leading to high TSHR antibody levels. On the other hand, prior treatment with CFA in the absence of A-subunit protein inhibited responses to subsequent immunization with A-sub-Ad. Consequently, adjuvant activity arising in vivo after bacterial infections combined with a protein autoantigen can break self-tolerance but in the absence of the autoantigen, adjuvant activity can inhibit the induction of immunity to autoantigens (like the TSHR) displaying strong self-tolerance.
Collapse
|
31
|
Kluger N, Ranki A, Krohn K. APECED: is this a model for failure of T cell and B cell tolerance? Front Immunol 2012; 3:232. [PMID: 22876245 PMCID: PMC3410439 DOI: 10.3389/fimmu.2012.00232] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 07/15/2012] [Indexed: 11/13/2022] Open
Abstract
In APECED, the key abnormality is in the T cell defect that may lead to tissue destruction chiefly in endocrine organs. Besides, APECED is characterized by high-titer antibodies against a wide variety of cytokines that could partly be responsible for the clinical symptoms during APECED, mainly chronic mucocutaneous candidiasis, and linked to antibodies against Th17 cells effector molecules, IL-17 and IL-22. On the other hand, the same antibodies, together with antibodies against type I interferons may prevent the patients from other immunological diseases, such as psoriasis and systemic lupus erythematous. The same effector Th17 cells, present in the lymphocytic infiltrate of target organs of APECED, could be responsible for the tissue destruction. Here again, the antibodies against the corresponding effector molecules, anti-IL-17 and anti-IL-22 could be protective. The occurrence of several effector mechanisms (CD4(+) Th17 cell and CD8(+) CTL and the effector cytokines IL-17 and IL-22), and simultaneous existence of regulatory mechanisms (CD4(+) Treg and antibodies neutralizing the effect of the effector cytokines) may explain the polymorphism of APECED. Almost all the patients develop the characteristic manifestations of the complex, but temporal course and severity of the symptoms vary considerably, even among siblings. The autoantibody profile does not correlate with the clinical picture. One could speculate that a secondary homeostatic balance between the harmful effector mechanisms, and the favorable regulatory mechanisms, finally define both the extent and severity of the clinical condition in the AIRE defective individuals. The proposed hypothesis that in APECED, in addition to strong tissue destructive mechanisms, a controlling regulatory mechanism does exist, allow us to conclude that APECED could be treated, and even cured, with immunological manipulation.
Collapse
Affiliation(s)
- Nicolas Kluger
- Department of Dermatology, Allergology and Venereology, Institute of Clinical Medicine, Skin and Allergy Hospital, Helsinki University Central Hospital, University of Helsinki,Helsinki, Finland
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology, Institute of Clinical Medicine, Skin and Allergy Hospital, Helsinki University Central Hospital, University of Helsinki,Helsinki, Finland
| | - Kai Krohn
- Clinical Research Institute HUCH Ltd,Helsinki, Finland
| |
Collapse
|
32
|
Nagayama Y, Nakahara M, Shimamura M, Horie I, Arima K, Abiru N. Prophylactic and therapeutic efficacies of a selective inhibitor of the immunoproteasome for Hashimoto's thyroiditis, but not for Graves' hyperthyroidism, in mice. Clin Exp Immunol 2012; 168:268-73. [PMID: 22519588 DOI: 10.1111/j.1365-2249.2012.04578.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Major histocompatibility complex (MHC) class I-restricted T cell epitopes are generated mainly by the immunoproteasome in antigen-presenting cells. Therefore, inhibition of activity of this proteolytic complex molecule is thought to be a potential treatment for cell-mediated autoimmune diseases. We therefore studied the efficacy of an immunoproteasome inhibitor, ONX 0914 (formerly PR-957), for the treatment of autoimmune thyroid diseases, including cell-mediated Hashimoto's thyroiditis and autoantibody-mediated Graves' hyperthyroidism using mouse models. Our data show that ONX 0914 was effective prophylactically and therapeutically at suppressing the degree of intrathyroidal lymphocyte infiltration and, to a lesser degree, the titres of anti-thyroglobulin autoantibodies in non-obese diabetic (NOD)-H2(h4) mice, an iodine-induced autoimmune thyroiditis model. It also inhibited differentiation of T cells to T helper type 1 (Th1) and Th17 cells, effector T cell subsets critical for development of thyroiditis in this mouse strain. In contrast, its effect on the Graves' model was negligible. Although ONX 0914 exerts its immune-suppressive effect through not only suppression of immune proteasome but also other mechanism(s), such as inhibition of T cell differentiation, the present results suggest that the immunoproteasome is a novel drug target in treatment of Hashimoto's thyroiditis in particular and cell-mediated autoimmune diseases in general.
Collapse
Affiliation(s)
- Y Nagayama
- Department of Molecular Medicine, Atomic Bomb Disease Institute Division of Immunology, Endocrinology and Metabolism, Department of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | | | | | | | | | | |
Collapse
|
33
|
Kim HJ, Cantor H. Regulation of self-tolerance by Qa-1-restricted CD8(+) regulatory T cells. Semin Immunol 2012; 23:446-52. [PMID: 22136694 DOI: 10.1016/j.smim.2011.06.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/07/2011] [Indexed: 10/14/2022]
Abstract
Mounting an efficient immune response to pathogens while avoiding damage to host tissues is the central task of the immune system. Emerging evidence has highlighted the contribution of the CD8(+) lineage of regulatory T cells to the maintenance of self-tolerance. Specific recognition of the MHC class Ib molecule Qa-1 complexed to peptides expressed by activated CD4(+) T cells by regulatory CD8(+) T cells triggers an inhibitory interaction that prevents autoimmune responses. Conversely, defective Qa-1-restricted CD8(+) regulatory activity can result in development of systemic autoimmune disease. Here, we review recent research into the cellular and molecular basis of these regulatory T cells, their mechanism of suppressive activity and the potential application of these insights into new treatments for autoimmune disease and cancer.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA
| | | |
Collapse
|
34
|
Tsai S, Clemente-Casares X, Santamaria P. CD8(+) Tregs in autoimmunity: learning "self"-control from experience. Cell Mol Life Sci 2011; 68:3781-95. [PMID: 21671120 PMCID: PMC11114820 DOI: 10.1007/s00018-011-0738-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 05/10/2011] [Accepted: 05/17/2011] [Indexed: 10/18/2022]
Abstract
Autoreactive CD8(+) regulatory T cells (Tregs) play important roles as modulators of immune responses against self, and numerical and functional defects in CD8(+) Tregs have been linked to autoimmunity. Several subsets of CD8(+) Tregs have been described. However, the origin of these T cells and how they participate in the natural progression of autoimmunity remain poorly defined. We discuss several lines of evidence suggesting that the autoimmune process itself promotes the development of autoregulatory CD8(+) T cells. We posit that chronic autoantigenic exposure fosters the differentiation of non-pathogenic autoreactive CD8(+) T cells into antigen-experienced, memory-like autoregulatory T cells, to generate a "negative feedback" regulatory loop capable of countering pathogenic autoreactive effectors. This hypothesis predicts that approaches capable of boosting autoregulatory T cell memory will be able to blunt autoimmunity without compromising systemic immunity.
Collapse
Affiliation(s)
- Sue Tsai
- Julia McFarlane Diabetes Research Centre, Faculty of Medicine, The University of Calgary, 3330 Hospital Dr. N.W, Calgary, AB T2N 4N1 Canada
| | - Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre, Faculty of Medicine, The University of Calgary, 3330 Hospital Dr. N.W, Calgary, AB T2N 4N1 Canada
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre, Faculty of Medicine, The University of Calgary, 3330 Hospital Dr. N.W, Calgary, AB T2N 4N1 Canada
- Department of Microbiology and Infectious Diseases, Institute of Inflammation, Infection and Immunity, Faculty of Medicine, The University of Calgary, 3330 Hospital Dr. N.W, Calgary, AB T2N 4N1 Canada
| |
Collapse
|
35
|
Chistiakov DA, Chistiakova EI, Voronova NV, Turakulov RI, Savost'anov KV. A variant of the Il2ra / Cd25 gene predisposing to graves' disease is associated with increased levels of soluble interleukin-2 receptor. Scand J Immunol 2011; 74:496-501. [PMID: 21815908 DOI: 10.1111/j.1365-3083.2011.02608.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alpha-subunit of the IL-2 receptor (IL-2Rα) encoded by the IL2RA/CD25 gene binds IL-2 that plays a pivotal role in the regulation of T cell function. Levels of a soluble form of IL-2Rα (sIL-2Rα) lacking the transmembrane and cytoplasmic domains were shown to be increased in several autoimmune diseases including Graves' disease (GD). Recent studies showed association between the IL2RA/CD25 gene variants and several autoimmune diseases including GD. In this study, we analyzed whether polymorphic markers rs2104286, rs41295061, and rs11594656 located at the IL2RA/CD25 locus confer susceptibility to GD and are related to increased concentrations of sIL-2Rα. A total of 1474 Russian GD patients and 1609 control subjects were genotyped for rs2104286, rs41295061, and rs11594656 using a Taqman assay. Concentrations of sIL-2Rα in sera of affected and non-affected individuals were measured using an ELISA test. A minor allele A of rs41295061 showed significant association with increased risk of GD [odds ratio (OR) = 1.43, P(c) = 0.00102]. The allele A of rs41295061 and allele A of rs11594656 constitute a higher risk haplotype AA (OR = 1.47, P(c) = 0.0477). Compared to carriers of the protective haplogenotype GT/GT, the carriage of two copies of the haplogenotype AA/AA was associated with elevated levels of sIL-2Rα in both GD patients (AA/AA versus GT/GT: 1.35 ± 0.47 ng/ml versus 1.12 ± 0.45 ng/ml, P = 0.0065) and healthy controls (AA/AA versus GT/GT: 0.67 ± 0.28 ng/ml versus 0.51 ± 0.33 ng/ml, P = 0.0098). This is the first report presenting correlation between the carriage of disease-associated variants of IL2RA/CD25 with increased levels of sIL-2Rα in GD.
Collapse
Affiliation(s)
- D A Chistiakov
- Department of Molecular Diagnostics, National Research Center GosNIIgenetika, Moscow, Russia.
| | | | | | | | | |
Collapse
|
36
|
CD4+ T lymphocytes are critical mediators of tumor immunity to simian virus 40 large tumor antigen induced by vaccination with plasmid DNA. J Virol 2011; 85:7216-24. [PMID: 21593176 DOI: 10.1128/jvi.00543-11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A mechanistic analysis of tumor immunity directed toward the viral oncoprotein simian virus 40 (SV40) large tumor antigen (Tag) has previously been described by our laboratory for scenarios of recombinant Tag immunization in BALB/c mice. In the present study, we performed a preliminary characterization of the immune components necessary for systemic tumor immunity induced upon immunization with plasmid DNA encoding SV40 Tag as a transgene (pCMV-Tag). Antibody responses to SV40 Tag were observed via indirect enzyme-linked immunosorbent assay following three intramuscular (i.m.) injections of pCMV-Tag and were typified by a mixed Th1/Th2 response. Complete tumor immunity within a murine model of pulmonary metastasis was achieved upon two i.m. injections of pCMV-Tag, as assessed by examination of tumor foci in mouse lungs, without a detectable antibody response to SV40 Tag. Induction-phase and effector-phase depletions of T cell subsets were performed in vivo via administration of depleting rat monoclonal antibodies, and these experiments demonstrated that CD4(+) T lymphocytes are required in both phases of the adaptive immune response. Conversely, depletion of CD8(+) T lymphocytes did not impair tumor immunity in either immune phase and resulted in the premature production of antibodies to SV40 Tag. Our findings are unique in that a dominant role could be ascribed to CD4(+) T lymphocytes within a model of DNA vaccine-induced tumor immunity to Tag-expressing tumor cells. Additionally, our findings provide insight into the general mechanisms of vaccine-induced tumor immunity directed toward tumors bearing distinct tumor-associated antigens.
Collapse
|
37
|
Zhou F, Zhang GX, Rostami A. 3G11 expression in CD4+ T cell-mediated autoimmunity and immune tolerance. Int Immunopharmacol 2011; 11:593-6. [PMID: 21084064 PMCID: PMC3079069 DOI: 10.1016/j.intimp.2010.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 10/21/2010] [Accepted: 11/01/2010] [Indexed: 11/22/2022]
Abstract
3G11 is a sialylated carbohydrate epitope of the disialoganglioside molecule expressed on mouse CD4(+) T cells. Recent research showed that 3G11 expression is related to the modulation of T cell function, i.e., 3G11(-) T cells exhibit anergic/Treg characteristics and efficiently inhibit autoimmunity in the central nervous system. The relationship between 3G11 expression and immune tolerance is summarized in this literature review.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
38
|
Ueki I, Abiru N, Kobayashi M, Nakahara M, Ichikawa T, Eguchi K, Nagayama Y. B cell-targeted therapy with anti-CD20 monoclonal antibody in a mouse model of Graves' hyperthyroidism. Clin Exp Immunol 2011; 163:309-17. [PMID: 21235532 DOI: 10.1111/j.1365-2249.2010.04301.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Graves' disease is a B cell-mediated and T cell-dependent autoimmune disease of the thyroid which is characterized by overproduction of thyroid hormones and thyroid enlargement by agonistic anti-thyrotrophin receptor (TSHR) autoantibody. In addition to antibody secretion, B cells have recently been recognized to function as antigen-presenting/immune-modulatory cells. The present study was designed to evaluate the efficacy of B cell depletion by anti-mouse (m) CD20 monoclonal antibody (mAb) on Graves' hyperthyroidism in a mouse model involving repeated injection of adenovirus expressing TSHR A-subunit (Ad-TSHR289). We observe that a single injection of 250 µg/mouse anti-mCD20 mAb eliminated B cells efficiently from the periphery and spleen and to a lesser extent from the peritoneum for more than 3 weeks. B cell depletion before immunization suppressed an increase in serum immunoglobulin (Ig)G levels, TSHR-specific splenocyte secretion of interferon (IFN)-γ, anti-TSHR antibody production and development of hyperthyroidism. B cell depletion 2 weeks after the first immunization, a time-point at which T cells were primed but antibody production was not observed, was still effective at inhibiting antibody production and disease development without inhibiting splenocyte secretion of IFN-γ. By contrast, B cell depletion in hyperthyroid mice was therapeutically ineffective. Together, these data demonstrate that B cells are critical not only as antibody-producing cells but also as antigen-presenting/immune-modulatory cells in the early phase of the induction of experimental Graves' hyperthyroidism and, although therapeutically less effective, B cell depletion is highly efficient for preventing disease development.
Collapse
Affiliation(s)
- I Ueki
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Chen CR, Hamidi S, Braley-Mullen H, Nagayama Y, Bresee C, Aliesky HA, Rapoport B, McLachlan SM. Antibodies to thyroid peroxidase arise spontaneously with age in NOD.H-2h4 mice and appear after thyroglobulin antibodies. Endocrinology 2010; 151:4583-93. [PMID: 20573721 PMCID: PMC2940509 DOI: 10.1210/en.2010-0321] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hashimoto's thyroiditis, a common autoimmune disease, is associated with autoantibodies to thyroglobulin (Tg) and thyroid peroxidase (TPO). TPO, unlike abundant and easily purified Tg, is rarely investigated as an autoantigen in animals. We asked whether antibodies (Abs) develop to both TPO and Tg in thyroiditis that is induced (C57BL/6 and DBA/1 mice) or arises spontaneously (NOD.H-2h4 mice). Screening for TPOAbs was performed by flow cytometry using mouse TPO-expressing eukaryotic cells. Sera were also tested for binding to purified mouse Tg and human TPO. The antibody data were compared with the extent of thyroiditis. Immunization with mouse TPO adenovirus broke self-tolerance to this protein in C57BL/6 mice, but thyroiditis was minimal and TgAbs were absent. In DBA/1 mice with extensive granulomatous thyroiditis induced by Tg immunization, TPOAbs were virtually absent despite high levels of TgAbs. In contrast, antibodies to mouse TPO, with minimal cross-reactivity with human TPO, arose spontaneously in older (7-12 months) NOD.H-2h4 mice. Unexpectedly, TgAbs preceded TPOAbs, a time course paralleled in relatives of probands with juvenile Hashimoto's thyroiditis. These findings demonstrate a novel aspect of murine and human thyroid autoimmunity, namely breaking B cell self-tolerance occurs first for Tg and subsequently for TPO.
Collapse
Affiliation(s)
- Chun-Rong Chen
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Research Institute and University of California, Los Angeles School of Medicine, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Filaci G, Fenoglio D, Indiveri F. CD8(+) T regulatory/suppressor cells and their relationships with autoreactivity and autoimmunity. Autoimmunity 2010; 44:51-7. [PMID: 20670118 DOI: 10.3109/08916931003782171] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Regulatory T lymphocytes (Treg) are fundamental for immune homeostasis since they contribute to the induction of peripheral tolerance to autologous antigens and regulate effector immune responses. Treg subsets are present within both the CD4+and the CD8(+) T cell compartments. Considering the CD8(+) Treg, in the last decades several subpopulations, provided with different phenotypes and mechanisms of action, have been characterized. This review is an attempt of integrating in an organic scenario the different CD8(+) Treg subpopulations. Moreover, it summarizes the findings so far achieved on the existence of CD8(+) Treg alterations in autoimmune diseases.
Collapse
Affiliation(s)
- Gilberto Filaci
- Department of Internal Medicine (DIMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.
| | | | | |
Collapse
|
41
|
Horie I, Abiru N, Saitoh O, Ichikawa T, Iwakura Y, Eguchi K, Nagayama Y. Distinct role of T helper Type 17 immune response for Graves' hyperthyroidism in mice with different genetic backgrounds. Autoimmunity 2010; 44:159-65. [PMID: 20670120 DOI: 10.3109/08916931003777247] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
T helper type 17 (Th17) cells, a newly identified effector T-cell subset, have recently been shown to play a role in numerous autoimmune diseases, including iodine-induced autoimmune thyroiditis in non-obese diabetic (NOD)-H2(h4) mice, which had previously been thought Th1-dominant. We here studied the role of Th17 in Graves' hyperthyroidism, another thyroid-specific autoimmune disease, in a mouse model. Two genetically distinct BALB/c and NOD-H2(h4) strains with intact or disrupted IL-17 genes (IL-17(+/+) or IL-17(-/-)) were immunized with adenovirus (Ad) expressing the thyrotropin receptor (TSHR) A-subunit (Ad-TSHR289). Both IL-17(+/+) and IL-17(-/-) mice developed anti-TSHR antibodies and hyperthyroidism at equally high frequencies on the BALB/c genetic background. In contrast, some IL-17(+/+), but none of IL-17(-/-), mice became hyperthyroid on the NOD-H2(h4) genetic background, indicating the crucial role of IL-17 for development of Graves' hyperthyroidism in non-susceptible NOD-H2(h4), but not in susceptible BALB/c mice. In the T-cell recall assay, splenocytes and lymphocytes from the draining lymph nodes from either mouse strains, irrespective of IL-17 gene status, produced IFN-γ and IL-10 but not other cytokines including IL-17 in response to TSHR antigen. Thus, the functional significance of Th17 may not necessarily be predictable from cytokine expression patterns in splenocytes or inflammatory lesions. In conclusion, this is, to our knowledge, the first report showing that the role of Th17 cells for the pathogenesis of a certain autoimmune disease depends on the mouse genetic backgrounds.
Collapse
Affiliation(s)
- Ichiro Horie
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Dinesh RK, Skaggs BJ, Cava AL, Hahn BH, Singh RP. CD8+ Tregs in lupus, autoimmunity, and beyond. Autoimmun Rev 2010; 9:560-8. [PMID: 20385256 PMCID: PMC2879466 DOI: 10.1016/j.autrev.2010.03.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 03/30/2010] [Indexed: 12/12/2022]
Abstract
While CD4(+)CD25(high) regulatory T cells (Tregs) have garnered much attention for their role in the maintenance of immune homeostasis, recent findings have shown that subsets of CD8(+) T cells (CD8(+) Tregs) display immunoregulatory functions as well. Both CD4(+) Tregs and CD8(+) Tregs appear impaired in number and/or function in several autoimmune diseases and in experimental animal models of autoimmunity, suggesting the possibility of immunotherapeutic targeting of these cells for improved management of autoimmune conditions. Our group has developed a strategy to induce CD8(+) Tregs in autoimmune mice through the use of a tolerogenic self-peptide, and new information has been gained on the phenotype, function and role of induced CD8(+) Tregs in autoimmunity. Here we present an overview of the role and mechanisms of action of CD8(+) Tregs in autoimmunity, with a special focus on lupus. We also discuss the potential role of CD8(+) Tregs in other diseases, including chronic infection and cancer.
Collapse
Affiliation(s)
- Ravi K Dinesh
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Brian J Skaggs
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Antonio La Cava
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Bevra H. Hahn
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| | - Ram Pyare Singh
- Division of Rheumatology, Dept of Medicine at the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1670
| |
Collapse
|
43
|
Wang LX, Li Y, Yang G, Pang PY, Haley D, Walker EB, Urba WJ, Hu HM. CD122+CD8+ Treg suppress vaccine-induced antitumor immune responses in lymphodepleted mice. Eur J Immunol 2010; 40:1375-85. [PMID: 20186876 PMCID: PMC3116644 DOI: 10.1002/eji.200839210] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Lymphodeleption prior to adoptive transfer of tumor-specific T cells greatly improves the clinical efficacy of adoptive T-cell therapy for patients with advanced melanoma, and increases the therapeutic efficacy of cancer vaccines in animal models. Lymphodepletion reduces competition between lymphocytes, and thus creates "space" for enhanced expansion and survival of tumor-specific T cells. Within the lymphodepleted host, Ag-specific T cells still need to compete with other lymphocytes that undergo lymphopenia-driven proliferation. Herein, we describe the relative capacity of naïve T cells, Treg, and NK cells to undergo lymphopenia-driven proliferation. We found that the major population that underwent lymphopenia-driven proliferation was the CD122+ memory-like T-cell population (CD122+CD8+ Treg), and these cells competed with Ag-driven proliferation of melanoma-specific T cells. Removal of CD122+CD8+ Treg resulted in a greater expansion of tumor-specific T cells and tumor infiltration of functional effector/memory T cells. Our results demonstrate the lymphopenia-driven proliferation of CD122+CD8+ Treg in reconstituted lymphodepleted mice limited the antitumor efficacy of DC vaccination in conjunction with adoptive transfer of tumor-specific T cells.
Collapse
Affiliation(s)
- Li-Xin Wang
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, P. R. China
- Cancer Research and Biotherapy Center, The Second Affiliated Hospital of Southeast University Nanjing, Jiangsu, P. R. China
| | - Yuhuan Li
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Guojun Yang
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Pui-yi Pang
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Dan Haley
- Laboratory of Immunological Monitoring, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Edwin B. Walker
- Laboratory of Immunological Monitoring, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Walter J. Urba
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
| | - Hong-Ming Hu
- Laboratory of Cancer Immunobiology, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, USA
- Cancer Research and Biotherapy Center, The Second Affiliated Hospital of Southeast University Nanjing, Jiangsu, P. R. China
| |
Collapse
|
44
|
McGlade JP, Strickland DH, Lambert MJM, Gorman S, Thomas JA, Judge MA, Burchell JT, Zosky GR, Hart PH. UV inhibits allergic airways disease in mice by reducing effector CD4 T cells. Clin Exp Allergy 2010; 40:772-85. [PMID: 20214669 DOI: 10.1111/j.1365-2222.2010.03469.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND In human asthma, and experimental allergic airways disease in mice, antigen-presenting cells and CD4(+) effector cells at the airway mucosa orchestrate, and CD4(+)CD25(+) regulatory T cells attenuate, allergen immunity. UV irradiation of skin before sensitization with ovalbumin (OVA) causes significantly reduced asthma-like responses in respiratory tissues. OBJECTIVE To determine whether UV-induced changes in CD11c(+) cells, CD4(+)CD25(+) effector cells or CD4(+)CD25(+) regulatory cells in the trachea and airway draining lymph nodes (ADLNs) were responsible for reduced allergic airways disease. METHODS The phenotype and function of CD11c(+) cells and CD4(+)CD25(+) cells in the trachea and ADLNs of UV- and non-irradiated, OVA-sensitized mice was examined 24 h after a single exposure to aerosolized OVA. RESULTS No changes in the function of CD11c(+) cells from UV-irradiated mice were observed. CD4(+)CD25(+) cells from UV-irradiated, OVA-sensitized mice harvested 24 h after OVA aerosol proliferated less in response to OVA in vitro and were unable to suppress the proliferation of OVA-sensitized responder cells. This result suggested reduced activation of effector T cells in the airway mucosa of UV-irradiated, OVA-sensitized mice. To exclude regulatory cells of any type, there was similar proliferation in vivo to aerosolized OVA by CFSE-loaded, OVA-TCR-specific CD4(+) cells adoptively transferred into UV- and non-irradiated, OVA-sensitized mice. In addition, there was no difference in the expression of regulatory T cell markers (Foxp3, IL-10, TGF-beta mRNA). To examine effector T cells, ADLN cells from UV-irradiated, OVA-sensitized and -challenged mice were cultured with OVA. There was reduced expression of the early activation marker CD69 by CD4(+)CD25(+) cells, and reduced proliferation in the absence of the regulatory cytokine, IL-10. CONCLUSION Reduced allergic airways disease in UV-irradiated mice is due to fewer effector CD4(+)CD25(+) cells in the trachea and ADLNs, and not due to UV-induced regulatory cells.
Collapse
Affiliation(s)
- J P McGlade
- Telethon Institute for Child Health Research and Centre for Child Health Research, The University of Western Australia, West Perth, Western Australia, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The identification of regulatory T cells (Tregs) as regulators of immunological self-tolerance has stimulated tremendous interest in the field. Over the past 12 months, new studies have added greatly to our understanding of the role of Tregs in autoimmune disease, details of which are presented here. RECENT FINDINGS In this review, the mechanism of action of Tregs, their antigen specificity and their frequency and function in different autoimmune diseases is explored. Currently available data on the role of transforming growth factor-beta, the reciprocal relationship between Tregs and Th17 cells, Treg markers, and current therapeutic approaches are evaluated. Other regulatory cells, which have been recently identified to play a significant role in autoimmunity, are described. SUMMARY Increasing insights into understanding the complex mechanisms of action of Tregs have already led to exciting therapeutic advances. This review provides an in-depth analysis of recent advances in the field of Tregs in autoimmunity. It highlights targets for future immunomodulatory therapy that may treat and potentially cure autoimmune disease, and it identifies areas for future research.
Collapse
|
46
|
Misharin AV, Nagayama Y, Aliesky HA, Rapoport B, McLachlan SM. Studies in mice deficient for the autoimmune regulator (Aire) and transgenic for the thyrotropin receptor reveal a role for Aire in tolerance for thyroid autoantigens. Endocrinology 2009; 150:2948-56. [PMID: 19264867 PMCID: PMC2689795 DOI: 10.1210/en.2008-1690] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/20/2009] [Indexed: 01/20/2023]
Abstract
The autoimmune regulator (Aire) mediates central tolerance for many autoantigens, and autoimmunity occurs spontaneously in Aire-deficient humans and mice. Using a mouse model of Graves' disease, we investigated the role of Aire in tolerance to the TSH receptor (TSHR) in Aire-deficient and wild-type mice (hyperthyroid-susceptible BALB/c background). Mice were immunized three times with TSHR A-subunit expressing adenovirus. The lack of Aire did not influence T-cell responses to TSHR protein or TSHR peptides. However, antibody levels were higher in Aire-deficient than wild-type mice after the second (but not the third) immunization. After the third immunization, hyperthyroidism persisted in a higher proportion of Aire-deficient than wild-type mice. Aire-deficient mice were crossed with transgenic strains expressing high or low-intrathyroidal levels of human TSHR A subunits. In the low-expressor transgenics, Aire deficiency had the same effect on the pattern of the TSHR antibody response to immunization as in nontransgenics, although the amplitude of the response was lower in the transgenics. High-expressor A-subunit transgenics were unresponsive to immunization. We examined intrathymic expression of murine TSHR, thyroglobulin, and thyroid peroxidase (TPO), the latter two being the dominant autoantigens in Hashimoto's thyroiditis (particularly TPO). Expression of the TSHR and thyroglobulin were reduced in the absence of Aire. Dramatically, thymic expression of TPO was nearly abolished. In contrast, the human A-subunit transgene, lacking a potential Aire-binding motif, was unaffected. Our findings provide insight into how varying intrathymic autoantigen expression may modulate thyroid autoimmunity and suggest that Aire deficiency may contribute more to developing Hashimoto's thyroiditis than Graves' disease.
Collapse
MESH Headings
- Animals
- Autoantigens/metabolism
- Disease Models, Animal
- Female
- Graves Disease/immunology
- Graves Disease/metabolism
- Graves Disease/pathology
- Hyperthyroidism/immunology
- Hyperthyroidism/metabolism
- Hyperthyroidism/pathology
- Immune Tolerance/immunology
- Immunoglobulins, Thyroid-Stimulating/immunology
- Immunoglobulins, Thyroid-Stimulating/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Receptors, Thyrotropin/genetics
- Receptors, Thyrotropin/immunology
- Receptors, Thyrotropin/metabolism
- T-Lymphocytes, Regulatory/pathology
- Thyroid Gland/immunology
- Thyroid Gland/metabolism
- Thyroid Gland/pathology
- Transcription Factors/genetics
- Transcription Factors/physiology
- AIRE Protein
Collapse
Affiliation(s)
- Alexander V Misharin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and University of California Los Angeles School of Medicine, Los Angeles, California 90048, USA
| | | | | | | | | |
Collapse
|
47
|
Dağdelen S, Kong YCM, Banga JP. Toward better models of hyperthyroid Graves' disease. Endocrinol Metab Clin North Am 2009; 38:343-54, viii. [PMID: 19328415 DOI: 10.1016/j.ecl.2009.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Graves' disease affects only humans. Although it is a treatable illness, medical therapy with antithyroid drugs is imperfect, showing high rates of recurrence. Furthermore, the etiology and treatment of the associated ophthalmopathy still represent problematic issues. Animal models could contribute to the solution of such problems by providing a better understanding of the underlying pathogenesis and could be used for evaluating novel therapeutic strategies. This article discusses the pursuit of a better experimental model for hyperthyroid Graves' disease and outlines how this research has clarified the immunology of the disease.
Collapse
Affiliation(s)
- Selçuk Dağdelen
- Department of Diabetes and Endocrinology, King's College London School of Medicine, Denmark Hill Campus, The Rayne Institute, London, UK.
| | | | | |
Collapse
|
48
|
Misharin A, Hewison M, Chen CR, Lagishetty V, Aliesky HA, Mizutori Y, Rapoport B, McLachlan SM. Vitamin D deficiency modulates Graves' hyperthyroidism induced in BALB/c mice by thyrotropin receptor immunization. Endocrinology 2009; 150:1051-60. [PMID: 18927213 PMCID: PMC2646531 DOI: 10.1210/en.2008-1191] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Accepted: 10/07/2008] [Indexed: 01/08/2023]
Abstract
TSH receptor (TSHR) antibodies and hyperthyroidism are induced by immunizing mice with adenovirus encoding the TSHR or its A-subunit. Depleting regulatory T cells (Treg) exacerbates thyrotoxicosis in susceptible BALB/c mice and induces hyperthyroidism in normally resistant C57BL/6 mice. Vitamin D plays an important role in immunity; high dietary vitamin D intake suppresses (and low intake enhances) adaptive immune responses. Vitamin D-induced immunosuppression may enhance Treg. Therefore, we hypothesized that decreased vitamin D intake would mimic Treg depletion and enhance hyperthyroidism induced by A-subunit adenovirus immunization. BALB/c mice had a reduced ability vs. C57BL/6 mice to generate the active metabolite of vitamin D (1,25-dihydroxyvitamin D3). Vitamin D deficiency induced subtle immune changes in BALB/c (not C57BL/6) mice. Compared with mice fed regular chow, vitamin D-deprived BALB/c mice had fewer splenic B cells and decreased interferon-gamma responses to mitogen and lacked memory T-cell responses to A-subunit protein. However, vitamin D deficiency did not alter TSHR antibody responses measured by ELISA, TSH binding inhibition, or cAMP generation from TSHR-expressing cells. Unexpectedly, compared with vitamin D-sufficient mice, vitamin D-deficient BALB/c mice had lower preimmunization T(4) levels and developed persistent hyperthyroidism. This difference was unrelated to the immunological changes between vitamin D-deficient or -sufficient animals. Previously, we found that different chromosomes or loci confer susceptibility to TSHR antibody induction vs. thyroid function. Our present studies provide evidence that an environmental factor, vitamin D, has only minor effects on induced immunity to the TSHR but directly affects thyroid function in mice.
Collapse
Affiliation(s)
- Alexander Misharin
- Autoimmune Disease Unit, Cedars-Sinai Research Institute and University of California Los Angeles, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Han Y, Guo Q, Zhang M, Chen Z, Cao X. CD69+CD4+CD25−T Cells, a New Subset of Regulatory T Cells, Suppress T Cell Proliferation through Membrane-Bound TGF-β1. THE JOURNAL OF IMMUNOLOGY 2008; 182:111-20. [DOI: 10.4049/jimmunol.182.1.111] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
50
|
Suzuki M, Konya C, Goronzy JJ, Weyand CM. Inhibitory CD8+ T cells in autoimmune disease. Hum Immunol 2008; 69:781-9. [PMID: 18812196 PMCID: PMC2614126 DOI: 10.1016/j.humimm.2008.08.283] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 07/30/2008] [Accepted: 08/12/2008] [Indexed: 11/26/2022]
Abstract
Rheumatologists have long been focused on developing novel immunotherapeutic agents to manage such prototypic autoimmune diseases as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). The ultimate challenge in providing immunosuppressive treatment for patients with RA and SLE has derived from the dilemma that both protective and harmful immune responses result from adaptive immune responses, mediated by highly diverse, antigen-specific T and B cells endowed with powerful effector functions and the ability for long-lasting memory. As regulatory/suppressor T cells can suppress immunity against any antigen, including self-antigens, they emerge as an ideal therapeutic target. Several distinct subtypes of CD8(+) suppressor cells (Ts) have been described that could find application in treating RA or SLE. In a xenograft model of human synovium, CD8(+)CD28(-)CD56(+) T cells effectively suppressed rheumatoid inflammation. Underlying mechanisms involve conditioning of antigen presenting cells (APC). Adoptively transferred CD8(+) T cells characterized by IL-16 secretion have also exhibited disease-inhibitory effects. In mice with polyarthritis, CD8(+) Ts suppressed inflammation by IFNgamma-mediated modulation of the tryptophan metabolism in APC. In SLE animal models, CD8(+) Ts induced by a synthetic peptide exerted suppressive activity mainly via the TGFbeta-Foxp3-PD1 pathway. CD8(+) Ts induced by histone peptides were found to downregulate disease activity by secreting TGFbeta. In essence, disease-specific approaches may be necessary to identify CD8(+) Ts optimally suited to treat immune dysfunctions in different autoimmune syndromes.
Collapse
Affiliation(s)
- Masakatsu Suzuki
- From the Kathleen B. and Mason I. Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322
| | - Christine Konya
- From the Kathleen B. and Mason I. Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322
| | - Jörg J. Goronzy
- From the Kathleen B. and Mason I. Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322
| | - Cornelia M. Weyand
- From the Kathleen B. and Mason I. Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322
| |
Collapse
|