1
|
Zong Y, Deng K, Chong WP. Regulation of Treg cells by cytokine signaling and co-stimulatory molecules. Front Immunol 2024; 15:1387975. [PMID: 38807592 PMCID: PMC11131382 DOI: 10.3389/fimmu.2024.1387975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
CD4+CD25+Foxp3+ regulatory T cells (Tregs), a vital component of the immune system, are responsible for maintaining immune homeostasis and preventing excessive immune responses. This review explores the signaling pathways of the cytokines that regulate Treg cells, including transforming growth factor beta (TGF-β), interleukin (IL)-2, IL-10, and IL-35, which foster the differentiation and enhance the immunosuppressive capabilities of Tregs. It also examines how, conversely, signals mediated by IL-6 and tumor necrosis factor -alpha (TNF-α) can undermine Treg suppressive functions or even drive their reprogramming into effector T cells. The B7 family comprises indispensable co-stimulators for T cell activation. Among its members, this review focuses on the capacity of CTLA-4 and PD-1 to regulate the differentiation, function, and survival of Tregs. As Tregs play an essential role in maintaining immune homeostasis, their dysfunction contributes to the pathogenesis of autoimmune diseases. This review delves into the potential of employing Treg-based immunotherapy for the treatment of autoimmune diseases, transplant rejection, and cancer. By shedding light on these topics, this article aims to enhance our understanding of the regulation of Tregs by cytokines and their therapeutic potential for various pathological conditions.
Collapse
Affiliation(s)
- Yuan Zong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, China
| | - Kaihang Deng
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Wai Po Chong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen, China
| |
Collapse
|
2
|
Falcon DM, Byrne KA, Sales MA, Erf GF. Spontaneous immunological activities in the target tissue of vitiligo-prone Smyth and vitiligo-susceptible Brown lines of chicken. Front Immunol 2024; 15:1386727. [PMID: 38720888 PMCID: PMC11076693 DOI: 10.3389/fimmu.2024.1386727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Vitiligo is an acquired de-pigmentation disorder characterized by the post-natal loss of epidermal melanocytes (pigment-producing cells) resulting in the appearance of white patches in the skin. The Smyth chicken is the only model for vitiligo that shares all the characteristics of the human condition including: spontaneous post-natal loss of epidermal melanocytes, interactions between genetic, environmental and immunological factors, and associations with other autoimmune diseases. In addition, an avian model for vitiligo has the added benefit of an easily accessible target tissue (a growing feather) that allows for the repeated sampling of an individual and thus the continuous monitoring of local immune responses over time. Methods Using a combination of flow cytometry and gene expression analyses, we sought to gain a comprehensive understanding of the initiating events leading to expression of vitiligo in growing feathers by monitoring the infiltration of leukocytes and concurrent immunological activities in the target tissue beginning prior to visual onset and continuing throughout disease development. Results Here, we document a sequence of immunologically significant events, including characteristic rises in infiltrating B and αβ T cells as well as evidence of active leukocyte recruitment and cell-mediated immune activities (CCL19, IFNG, GZMA) leading up to visual vitiligo onset. Examination of growing feathers from vitiligo-susceptible Brown line chickens revealed anti-inflammatory immune activities which may be responsible for preventing vitiligo (IL10, CTLA4, FOXP3). Furthermore, we detected positive correlations between infiltrating T cells and changes in their T cell receptor diversity supporting a T cell-specific immune response. Conclusion Collectively, these results further support the notion of cell-mediated immune destruction of epidermal melanocytes in the pulp of growing feathers and open new avenues of study in the vitiligo-prone Smyth and vitiligo-susceptible Brown line chickens.
Collapse
Affiliation(s)
| | | | | | - Gisela F. Erf
- Division of Agriculture, Department of Poultry Science, University of Arkansas System, Fayetteville, AR, United States
| |
Collapse
|
3
|
Kim GR, Choi JM. Current Understanding of Cytotoxic T Lymphocyte Antigen-4 (CTLA-4) Signaling in T-Cell Biology and Disease Therapy. Mol Cells 2022; 45:513-521. [PMID: 35950451 PMCID: PMC9385567 DOI: 10.14348/molcells.2022.2056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/21/2022] Open
Abstract
Cytotoxic T lymphocyte antigen-4 (CTLA-4) is an immune checkpoint molecule that is mainly expressed on activated T cells and regulatory T (Treg) cells that inhibits T-cell activation and regulates immune homeostasis. Due to the crucial functions of CTLA-4 in T-cell biology, CTLA-4-targeted immunotherapies have been developed for autoimmune disease as well as cancers. CTLA-4 is known to compete with CD28 to interact with B7, but some studies have revealed that its downstream signaling is independent of its ligand interaction. As a signaling domain of CTLA-4, the tyrosine motif plays a role in inhibiting T-cell activation. Recently, the lysine motif has been shown to be required for the function of Treg cells, emphasizing the importance of CTLA-4 signaling. In this review, we summarize the current understanding of CTLA-4 biology and molecular signaling events and discuss strategies to target CTLA-4 signaling for immune modulation and disease therapy.
Collapse
Affiliation(s)
- Gil-Ran Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, Korea
- Institute for Rheumatology Research, Hanyang University, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
4
|
Kim G, Kim W, Lim S, Lee H, Koo J, Nam K, Kim S, Park S, Choi J. In Vivo Induction of Regulatory T Cells Via CTLA-4 Signaling Peptide to Control Autoimmune Encephalomyelitis and Prevent Disease Relapse. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004973. [PMID: 34306974 PMCID: PMC8292875 DOI: 10.1002/advs.202004973] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/01/2021] [Indexed: 05/22/2023]
Abstract
Regulatory T cells play a key role in immune tolerance to self-antigens, thereby preventing autoimmune diseases. However, no drugs targeting Treg cells have been approved for clinical trials yet. Here, a chimeric peptide is generated by conjugation of the cytoplasmic domain of CTLA-4 (ctCTLA-4) with dNP2 for intracellular delivery, dNP2-ctCTLA-4, and evaluated Foxp3 expression during Th0, Th1, Treg, and Th17 differentiation dependent on TGF-β. The lysine motif of ctCTLA-4, not tyrosine motif, is required for Foxp3 expression for Treg induction and amelioration of experimental autoimmune encephalomyelitis (EAE). Transcriptome analysis reveals that dNP2-ctCTLA-4-treated T cells express Treg transcriptomic patterns with properties of suppressive functions. In addition, the molecular interaction between the lysine motif of ctCTLA-4 and PKC-η is critical for Foxp3 expression. Although both CTLA-4-Ig and dNP2-ctCTLA-4 treatment in vivo ameliorated EAE progression, only dNP2-ctCTLA-4 requires Treg cells for inhibition of disease progression and prevention of relapse. Furthermore, the CTLA-4 signaling peptide is able to induce human Tregs in vitro and in vivo as well as from peripheral blood mononuclear cells (PBMCs) of multiple sclerosis patients. These results collectively suggest that the chimeric CTLA-4 signaling peptide can be used for successful induction of regulatory T cells in vivo to control autoimmune diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Gil‐Ran Kim
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesHanyang UniversitySeoul04763Republic of Korea
| | - Won‐Ju Kim
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesHanyang UniversitySeoul04763Republic of Korea
| | - Sangho Lim
- Hubrecht Institute for Developmental Biology and Stem Cell Research‐KNAW, University Medical Centre UtrechtUtrecht3584 CTNetherland
| | - Hong‐Gyun Lee
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesHanyang UniversitySeoul04763Republic of Korea
| | - Ja‐Hyun Koo
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesHanyang UniversitySeoul04763Republic of Korea
| | - Kyung‐Ho Nam
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesHanyang UniversitySeoul04763Republic of Korea
| | - Sung‐Min Kim
- Department of NeurologyCollege of MedicineSeoul National UniversitySeoul National University HospitalSeoul03080Republic of Korea
| | - Sung‐Dong Park
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesHanyang UniversitySeoul04763Republic of Korea
| | - Je‐Min Choi
- Department of Life ScienceCollege of Natural SciencesHanyang UniversityResearch institute for Natural SciencesResearch Institute for Convergence of Basic SciencesHanyang UniversitySeoul04763Republic of Korea
| |
Collapse
|
5
|
Liu Y, Zhang Y, Zhang M, Meng J, Ma Q, Hao Z, Zheng M, Zhang L, Chen X, Liang C. Activated autophagy restored the impaired frequency and function of regulatory T cells in chronic prostatitis. Prostate 2021; 81:29-40. [PMID: 33085775 DOI: 10.1002/pros.24073] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/04/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Chronic prostatitis or chronic pelvic pain syndrome (CP/CPPS) is a disease with an unclear pathogenesis. Recent studies have reported that regulatory T (Treg) cells might be involved in the development of CP/CPPS. In this study we aimed to examine the functional role of Treg cells and explore the possible regulatory mechanism of Treg cells in CP/CPPS. METHODS An experimental autoimmune prostatitis (EAP) mouse model was constructed; the numbers and functions of Treg cells in the EAP and control groups were tested. Then, cell differentiation experiments were conducted to evaluate the regulatory effect of autophagy on Treg cell differentiation. Furthermore, autologous CD4+ CD25- cells and CD4+ CD25+ cells from the two groups were magnetically sorted and cocultured to observe differences in cellular inhibitory functions. Finally, in an in vivo experiment, rapamycin was intraperitoneally injected into EAP mice for 4 weeks to observe the therapeutic effects. RESULTS We found that the number and function of Treg cells in the EAP group were diminished compared to those in the control group. Meanwhile, the tolerance of pain in EAP mice had also decreased. Moreover, after using the autophagy activator rapamycin, the expression of the inflammatory cytokines interleukin-1β was decreased and the pain symptoms were alleviated. A mechanistic study found that autophagy activation promoted the differentiation of Treg and increased the suppressive functions of Treg cells, along with the elevated expression of GATA-3 and cytotoxic T lymphocyte antigen 4 (CTLA-4). Furthermore, in vivo administration of the autophagy activator rapamycin had similar effects on recovering the frequency and function of Treg cells as well as the expression of GATA-3 and CTLA-4. CONCLUSION The impaired frequency and function of Treg cells may contribute to the progression of CP/CPPS, and autophagy is a protective mechanism that promotes the differentiation of Treg cells and restores the suppressive functions of Treg cells. Autophagy may be a novel therapeutic option for patients with CP/CPPS.
Collapse
Affiliation(s)
- Yi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Yong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Jialin Meng
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Qingqing Ma
- Department of Urology, Medical Research Center, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Meijuan Zheng
- Department of Urology, Medical Research Center, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Xianguo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Urology, Institute of Urology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
6
|
Schnell A, Bod L, Madi A, Kuchroo VK. The yin and yang of co-inhibitory receptors: toward anti-tumor immunity without autoimmunity. Cell Res 2020; 30:285-299. [PMID: 31974523 PMCID: PMC7118128 DOI: 10.1038/s41422-020-0277-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022] Open
Abstract
Co-inhibitory receptors are important regulators of T-cell function that define the balance between tolerance and autoimmunity. The immune regulatory function of co-inhibitory receptors, including CTLA-4, PD-1, TIM-3, TIGIT, and LAG-3, was first discovered in the setting of autoimmune disease models, in which their blockade or deficiency resulted in induction or exacerbation of the disease. Later on, co-inhibitory receptors on lymphocytes have also been found to influence outcomes in tumor and chronic viral infection settings. These receptors suppress T-cell function in the tumor microenvironment (TME), thereby making the T cells dysfunctional. Based on this observation, blockade of co-inhibitory receptors (also known as checkpoint molecules) has emerged as a successful treatment option for a number of human cancers. However, severe autoimmune-like side effects limit the use of therapeutics that block individual or combinations of co-inhibitory receptors for cancer treatment. In this review we provide an overview of the role of co-inhibitory receptors in autoimmunity and anti-tumor immunity. We then discuss current approaches and future directions to leverage our knowledge of co-inhibitory receptors to target them in tumor immunity without inducing autoimmunity.
Collapse
Affiliation(s)
- Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lloyd Bod
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
7
|
Martinov T, Fife BT. Type 1 diabetes pathogenesis and the role of inhibitory receptors in islet tolerance. Ann N Y Acad Sci 2020; 1461:73-103. [PMID: 31025378 PMCID: PMC6994200 DOI: 10.1111/nyas.14106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) affects over a million Americans, and disease incidence is on the rise. Despite decades of research, there is still no cure for this disease. Exciting beta cell replacement strategies are being developed, but in order for such approaches to work, targeted immunotherapies must be designed. To selectively halt the autoimmune response, researchers must first understand how this response is regulated and which tolerance checkpoints fail during T1D development. Herein, we discuss the current understanding of T1D pathogenesis in humans, genetic and environmental risk factors, presumed roles of CD4+ and CD8+ T cells as well as B cells, and implicated autoantigens. We also highlight studies in non-obese diabetic mice that have demonstrated the requirement for CD4+ and CD8+ T cells and B cells in driving T1D pathology. We present an overview of central and peripheral tolerance mechanisms and comment on existing controversies in the field regarding central tolerance. Finally, we discuss T cell- and B cell-intrinsic tolerance mechanisms, with an emphasis on the roles of inhibitory receptors in maintaining islet tolerance in humans and in diabetes-prone mice, and strategies employed to date to harness inhibitory receptor signaling to prevent or reverse T1D.
Collapse
Affiliation(s)
- Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
8
|
Dougan M, Pietropaolo M. Time to dissect the autoimmune etiology of cancer antibody immunotherapy. J Clin Invest 2020; 130:51-61. [PMID: 31895048 PMCID: PMC6934191 DOI: 10.1172/jci131194] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy has transformed the treatment landscape for a wide range of human cancers. Immune checkpoint inhibitors (ICIs), monoclonal antibodies that block the immune-regulatory "checkpoint" receptors CTLA-4, PD-1, or its ligand PD-L1, can produce durable responses in some patients. However, coupled with their success, these treatments commonly evoke a wide range of immune-related adverse events (irAEs) that can affect any organ system and can be treatment-limiting and life-threatening, such as diabetic ketoacidosis, which appears to be more frequent than initially described. The majority of irAEs from checkpoint blockade involve either barrier tissues (e.g., gastrointestinal mucosa or skin) or endocrine organs, although any organ system can be affected. Often, irAEs resemble spontaneous autoimmune diseases, such as inflammatory bowel disease, autoimmune thyroid disease, type 1 diabetes mellitus (T1D), and autoimmune pancreatitis. Yet whether similar molecular or pathologic mechanisms underlie these apparent autoimmune adverse events and classical autoimmune diseases is presently unknown. Interestingly, evidence links HLA alleles associated with high risk for autoimmune disease with ICI-induced T1D and colitis. Understanding the genetic risks and immunologic mechanisms driving ICI-mediated inflammatory toxicities may not only identify therapeutic targets useful for managing irAEs, but may also provide new insights into the pathoetiology and treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Massimo Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
9
|
Okazaki T, Okazaki IM. Stimulatory and Inhibitory Co-signals in Autoimmunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:213-232. [PMID: 31758536 DOI: 10.1007/978-981-32-9717-3_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Co-receptors cooperatively regulate the function of immune cells to optimize anti-infectious immunity while limiting autoimmunity by providing stimulatory and inhibitory co-signals. Among various co-receptors, those in the CD28/CTLA-4 family play fundamental roles in the regulation of lymphocytes by modulating the strength, quality, and/or duration of the antigen receptor signal. The development of the lethal lymphoproliferative disorder and various tissue-specific autoimmune diseases in mice deficient for CTLA-4 and PD-1, respectively, clearly demonstrates their pivotal roles in the development and the maintenance of immune tolerance. The recent success of immunotherapies targeting CTLA-4 and PD-1 in the treatment of various cancers highlights their critical roles in the regulation of cancer immunity in human. In addition, the development of multifarious autoimmune diseases as immune-related adverse events of anti-CTLA-4 and anti-PD-1/PD-L1 therapies and the successful clinical application of the CD28 blocking therapy using CTLA-4-Ig to the treatment of arthritis assure their crucial roles in the regulation of autoimmunity in human. Accumulating evidences in mice and humans indicate that genetic and environmental factors strikingly modify effects of the targeted inhibition and potentiation of co-signals. In this review, we summarize our current understanding of the roles of CD28, CTLA-4, and PD-1 in autoimmunity. Deeper understandings of the context-dependent and context-independent functions of co-signals are essential for the appropriate usage and the future development of innovative immunomodulatory therapies for a diverse array of diseases.
Collapse
Affiliation(s)
- Taku Okazaki
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan.
| | - Il-Mi Okazaki
- Division of Immune Regulation, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
10
|
Al-Koofee DA, Jasim AA, Al-Issawi HA. Identification of +49A/G polymorphism of cytotoxic T lymphocyte-associated protein-4 gene in type 1 diabetes patients in Kerbala province/Iraq. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Lingel H, Brunner-Weinzierl MC. CTLA-4 (CD152): A versatile receptor for immune-based therapy. Semin Immunol 2019; 42:101298. [DOI: 10.1016/j.smim.2019.101298] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/05/2019] [Indexed: 12/31/2022]
|
12
|
Amaya-Uribe L, Rojas M, Azizi G, Anaya JM, Gershwin ME. Primary immunodeficiency and autoimmunity: A comprehensive review. J Autoimmun 2019; 99:52-72. [PMID: 30795880 DOI: 10.1016/j.jaut.2019.01.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023]
Abstract
The primary immunodeficiency diseases (PIDs) include many genetic disorders that affect different components of the innate and adaptive responses. The number of distinct genetic PIDs has increased exponentially with improved methods of detection and advanced laboratory methodology. Patients with PIDs have an increased susceptibility to infectious diseases and non-infectious complications including allergies, malignancies and autoimmune diseases (ADs), the latter being the first manifestation of PIDs in several cases. There are two types of PIDS. Monogenic immunodeficiencies due to mutations in genes involved in immunological tolerance that increase the predisposition to develop autoimmunity including polyautoimmunity, and polygenic immunodeficiencies characterized by a heterogeneous clinical presentation that can be explained by a complex pathophysiology and which may have a multifactorial etiology. The high prevalence of ADs in PIDs demonstrates the intricate relationships between the mechanisms of these two conditions. Defects in central and peripheral tolerance, including mutations in AIRE and T regulatory cells respectively, are thought to be crucial in the development of ADs in these patients. In fact, pathology that leads to PID often also impacts the Treg/Th17 balance that may ease the appearance of a proinflammatory environment, increasing the odds for the development of autoimmunity. Furthermore, the influence of chronic and recurrent infections through molecular mimicry, bystander activation and super antigens activation are supposed to be pivotal for the development of autoimmunity. These multiple mechanisms are associated with diverse clinical subphenotypes that hinders an accurate diagnosis in clinical settings, and in some cases, may delay the selection of suitable pharmacological therapies. Herein, a comprehensively appraisal of the common mechanisms among these conditions, together with clinical pearls for treatment and diagnosis is presented.
Collapse
Affiliation(s)
- Laura Amaya-Uribe
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Doctoral Program in Biomedical Sciences, Universidad Del Rosario, Bogota, Colombia
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, CA, USA.
| |
Collapse
|
13
|
Seitz C, Liu S, Klocke K, Joly AL, Czarnewski PV, Tibbitt CA, Parigi SM, Westerberg LS, Coquet JM, Villablanca EJ, Wing K, Andersson J. Multi-faceted inhibition of dendritic cell function by CD4 +Foxp3 + regulatory T cells. J Autoimmun 2019; 98:86-94. [PMID: 30616979 DOI: 10.1016/j.jaut.2018.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/22/2022]
Abstract
CTLA-4 is required for CD4+Foxp3+ regulatory T (Treg) cell function, but its mode of action remains incompletely defined. Herein we generated Ctla-4ex2fl/flFoxp3-Cre mice with Treg cells exclusively expressing a naturally occurring, ligand-independent isoform of CTLA-4 (liCTLA-4) that cannot interact with the costimulatory molecules CD80 and CD86. The mice did not exhibit any signs of effector T cell activation early in life, however, at 6 months of age they exhibited excessive T cell activation and inflammation in lungs. In contrast, mice with Treg cells completely lacking CTLA-4 developed lymphoproliferative disease characterized by multi-organ inflammation early in life. In vitro, Treg cells exclusively expressing liCTLA-4 inhibited CD80 and CD86 expression on dendritic cells (DC). Conversely, Treg cells required the extra-cellular part of CTLA-4 to up-regulate expression of the co-inhibitory molecule PD-L2 on DCs. Transcriptomic analysis of suppressed DCs revealed that Treg cells induced a specific immunosuppressive program in DCs.
Collapse
Affiliation(s)
- Christina Seitz
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Sang Liu
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Katrin Klocke
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anne-Laure Joly
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Christopher A Tibbitt
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sara M Parigi
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan M Coquet
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Kajsa Wing
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - John Andersson
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
14
|
Vonberg AD, Acevedo-Calado M, Cox AR, Pietropaolo SL, Gianani R, Lundy SK, Pietropaolo M. CD19+IgM+ cells demonstrate enhanced therapeutic efficacy in type 1 diabetes mellitus. JCI Insight 2018; 3:99860. [PMID: 30518692 DOI: 10.1172/jci.insight.99860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
We describe a protective effect on autoimmune diabetes and reduced destructive insulitis in NOD.scid recipients following splenocyte injections from diabetic NOD donors and sorted CD19+ cells compared with NOD.scid recipients receiving splenocytes alone. This protective effect was age specific (only CD19+ cells from young NOD donors exerted this effect; P < 0.001). We found that the CD19+IgM+ cell is the primary subpopulation of B cells that delayed transfer of diabetes mediated by diabetogenic T cells from NOD mice (P = 0.002). Removal of IgM+ cells from the CD19+ pool did not result in protection. Notably, protection conferred by CD19+IgM+ cotransfers were not dependent on the presence of Tregs, as their depletion did not affect their ability to delay onset of diabetes. Blockade of IL-10 with neutralizing antibodies at the time of CD19+ cell cotransfers also abrogated the therapeutic effect, suggesting that IL-10 secretion was an important component of protection. These results were strengthened by ex vivo incubation of CD19+ cells with IL-5, resulting in enhanced proliferation and IL-10 production and equivalently delayed diabetes progression (P = 0.0005). The potential to expand CD19+IgM+ cells, especially in response to IL-5 stimulation or by pharmacologic agents, may be a new therapeutic option for type 1 diabetes.
Collapse
Affiliation(s)
- Andrew D Vonberg
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Maria Acevedo-Calado
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Aaron R Cox
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Susan L Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Roberto Gianani
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Massimo Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| |
Collapse
|
15
|
Lim S, Kirkiles-Smith NC, Pober JS, Bothwell ALM, Choi JM. Regulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection. Biomaterials 2018; 183:128-138. [PMID: 30165256 DOI: 10.1016/j.biomaterials.2018.08.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Nancy C Kirkiles-Smith
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
16
|
Zhong JX, Chen J, Rao X, Duan L. Dichotomous roles of co-stimulatory molecules in diabetes mellitus. Oncotarget 2018; 9:2902-2911. [PMID: 29416823 PMCID: PMC5788691 DOI: 10.18632/oncotarget.23102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 11/15/2017] [Indexed: 11/25/2022] Open
Abstract
Numerous studies have established the importance of immune dysfunction in the development of diabetes mellitus, including typ1 and typ2 diabetes, and it is worth noting that T cell activation acts a key role in the pathogenesis of loss of β cell mass, adipose inflammation and insulin resistance. Regarding as an important checkpoint in the process of T cell activation, co-stimulatory molecules interaction between antigen present cells and T cells have been identified the critical role in the development of diabetes mellitus. Thus, blockage of co-stimulatory dyads interaction between antigen present cells and T cells was supposed to a potential of therapeutic strategies. However, studies also showed that inhibition or deletion of some co-stimulatory molecules do not always reduce the development of diabetes, and even exacerbate the disease activity. Here, in this context, we highlight the dichotomous role of co-stimulatory molecules interaction in the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Ji-Xin Zhong
- Department of Endocrinology, Central Hospital of Wuhan, Wuhan, Hubei, China 430061
| | - Jie Chen
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA 44106
- Basic Medical Department of Medical College, Xiamen University, Xiamen, China 361102
| | - Xiaoquan Rao
- Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA 44106
| | - Lihua Duan
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China 361003
| |
Collapse
|
17
|
Abstract
The immune system is guided by a series of checks and balances, a major component of which is a large array of co-stimulatory and co-inhibitory pathways that modulate the host response. Although co-stimulation is essential for boosting and shaping the initial response following signaling through the antigen receptor, inhibitory pathways are also critical for modulating the immune response. Excessive co-stimulation and/or insufficient co-inhibition can lead to a breakdown of self-tolerance and thus to autoimmunity. In this review, we will focus on the role of co-stimulatory and co-inhibitory pathways in two systemic (systemic lupus erythematosus and rheumatoid arthritis) and two organ-specific (multiple sclerosis and type 1 diabetes) emblematic autoimmune diseases. We will also discuss how mechanistic analysis of these pathways has led to the identification of potential therapeutic targets and initiation of clinical trials for autoimmune diseases, as well as outline some of the challenges that lie ahead.
Collapse
Affiliation(s)
- Qianxia Zhang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA.
| |
Collapse
|
18
|
Qiao YC, Pan YH, Ling W, Tian F, Chen YL, Zhang XX, Zhao HL. The Yin and Yang of regulatory T cell and therapy progress in autoimmune disease. Autoimmun Rev 2017; 16:1058-1070. [PMID: 28778708 DOI: 10.1016/j.autrev.2017.08.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
Autoimmune diseases (ADs) are primarily mediated by the failure of immunological self-tolerance. Regulatory T cells (Tregs) play a critical role in the maintenance of induced tolerance to peripheral self-antigens, suppressing immoderate immune responses deleterious to the host and preventing the AD development. Tregs and suppressive cytokines are homeostatic with effective cells plus pro-inflammatory cytokines in healthy hosts which is defined as "Yang", and ADs are usually induced in case of disturbed homeostasis, which is defined as "Yin". Indeed, the Yin-Yang balance could explain the pathogenic mechanism of ADs. Tregs not only suppress CD4+ and CD8+ T cells but also can suppress other immune cells such as B cell, natural killer cell, DC and other antigen-presenting cell through cell-cell contact or secreting suppressive cytokines. In Tregs, Foxp3 as an intracellular protein displays a more specific marker than currently used other cell-surface markers (such as CD25, CD40L, CTLA-4, ICOS and GITR) in defining the naturally occurring CD4+ Tregs. Though the precise mechanism for the opposite effects of Tregs has not been fully elucidated, the importance of Tregs in ADs has been proved to be associated with kinds of immunocytes. At present, the surface marker, frequency and function of Tregs existed conflicts and hence the Tregs therapy in ADs faces challenges. Though some success has been achieved with Tregs therapy in few ADs both in murine models and humans, more effort should paid to meet the future challenges. This review summarizes the progress and discusses the phenotypic, numeric and functional abnormalities of Tregs and is the first time to systematically review the progress of Tregs therapy in kinds of ADs.
Collapse
Affiliation(s)
- Yong-Chao Qiao
- Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yan-Hong Pan
- Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China
| | - Wei Ling
- Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Fang Tian
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yin-Ling Chen
- Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Xiao-Xi Zhang
- Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China
| | - Hai-Lu Zhao
- Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
19
|
Kim I, Wu G, Chai NN, Klein AS, Jordan SC. Immunological characterization of de novo and recall alloantibody suppression by CTLA4Ig in a mouse model of allosensitization. Transpl Immunol 2016; 38:84-92. [PMID: 27507323 DOI: 10.1016/j.trim.2016.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/15/2016] [Accepted: 08/05/2016] [Indexed: 12/14/2022]
Abstract
It is well known that CTLA4Ig inhibits allogenic T-cell activation in transplantation. The immunological features and mechanisms associated with alloantibody suppression by CTLA4Ig, however, are poorly understood. Here, we used a mouse model of allosensitization to evaluate the efficacy of CTLA4Ig (abatacept) in suppression of donor-specific antibody (DSA) during de novo and recall alloantibody responses. We found that abatacept inhibited de novo DSA IgM and IgG responses to HLA-A2 expressing skin grafts. Abatacept administered during primary T cell priming also reduced recall IgG responses induced by re-immunization. Suppression of de novo DSA responses by abatacept is associated with a reduction in splenic expression of the germinal center activation marker GL7 and a reduction of CD4(+)PD1(+)CXCR5(+) follicular T helper (Tfh) subset in splenic lymphocytes detected by flow cytometry. The efficacy of abatacept on recall DSA suppression is moderate. In vitro experiments demonstrated that abatacept inhibited DSA IgG secretion by CD138(+) plasma cells isolated from allograft recipients. Additional experiments using an IgG1 secreting mouse hybridoma cell line showed that abatacept binds to CD80 expressed on these cells with subsequent inhibition of cell proliferation and reduction in IgG ELISpot formation. In conclusion, CTLA4Ig is a potent suppressor of de novo DSA responses and also affects recall responses. The data suggests modification of recall DSA responses is due to a direct suppressive effect on plasma cells.
Collapse
Affiliation(s)
- Irene Kim
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gordon Wu
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Ning-Ning Chai
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew S Klein
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stanley C Jordan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
20
|
Shishido A, Caicedo A, Rodriguez-Diaz R, Pileggi A, Berggren PO, Abdulreda MH. Clinical intraocular islet transplantation is not a number issue. CELLR4-- REPAIR, REPLACEMENT, REGENERATION, & REPROGRAMMING 2016; 4:e2120. [PMID: 29497631 PMCID: PMC5828508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
It is now well established that beta cell replacement through pancreatic islet transplantation results in significant improvement in the quality-of-life of type 1 diabetes (T1D) patients. This is achieved through improved control and prevention of severe drops in blood sugar levels. Islet transplant therapy is on the verge of becoming standard-of-care in the USA. Yet, as with other established transplantation therapies, there remain hurdles to overcome to bring islet transplantation to full fruition as a long-lasting therapy of T1D. One of these hurdles is establishing reliable new sites, other than the liver, where durable efficacy and survival of transplanted islets can be achieved. In this article, we discuss the anterior chamber of the eye as a new site for clinical islet transplantation in the treatment of T1D. We specifically focus on the common conceptions, and preconceptions, on the requirements of islet mass, and whether or not the anterior chamber can accommodate sufficient islets to achieve meaningful efficacy and significant impact on hyperglycemia in clinical application.
Collapse
Affiliation(s)
- A Shishido
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Caicedo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - R Rodriguez-Diaz
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Pileggi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Departments of Microbiology and Immunology and Biomedical Engineering, University of Miami, Miami, FL, USA
| | - P-O Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - M H Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
21
|
Abstract
Co-stimulatory and co-inhibitory molecules direct the "second signal," which largely determines the outcome of the "first signal" generated by the interaction of T cell receptor (TCR) with cognate MHC-peptide complex. The co-stimulatory and co-inhibitory signals are key mechanistic contributors to the regulation of adaptive immunity, especially the T cell-mediated immune response. Regulatory T cells (Tregs) are a special population of T cells, which unlike other T cells function as "attenuators" to suppress T cell immunity. Dysregulation of either the "second signal" or Tregs leads to an unbalanced immune system, which can result in a range of immune-related disorders, including autoimmune diseases, chronic infections, and tumors. In contrast, precise manipulation of these two systems offers tremendous clinical opportunities to treat these same diseases. Co-stimulatory and co-inhibitory molecules modulate immunity at molecular level, whereas Tregs delicately control the immune response at cellular level. Accumulating evidence has demonstrated that these two regulatory strategies converge and synergize with each other. This review discusses recent progress on the roles of co-stimulatory and co-inhibitory signals in the context of Tregs.
Collapse
Affiliation(s)
- Weifeng Liu
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA.,b Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Steven C Almo
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Xingxing Zang
- b Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA.,c Department of Medicine , Albert Einstein College of Medicine , Bronx , NY , USA.,d Department of Urology , Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
22
|
Jakubczik F, Jones K, Nichols J, Mansfield W, Cooke A, Holmes N. A SNP in the Immunoregulatory Molecule CTLA-4 Controls mRNA Splicing In Vivo but Does Not Alter Diabetes Susceptibility in the NOD Mouse. Diabetes 2016; 65:120-8. [PMID: 26450994 PMCID: PMC4693968 DOI: 10.2337/db15-1175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/01/2015] [Indexed: 11/13/2022]
Abstract
CTLA-4 is a critical "checkpoint" regulator in autoimmunity. Variation in CTLA-4 isoform expression has been linked to type 1 diabetes development in human and NOD mouse studies. In the NOD mouse, a causative link between increased expression of the minor isoform ligand-independent CTLA-4 and a reduction in diabetes has become widely accepted. Altered splicing of CTLA-4 has been attributed to a single nucleotide polymorphism (SNP) in Ctla4 exon2 (e2_77A/G). To investigate this link, we have used NOD embryonic stem (ES) cells to generate a novel NOD transgenic line with the 77A/G SNP. This strain phenocopies the increase in splicing toward the liCTLA4 isoform seen in B10 Idd5.1 mice. Crucially, the SNP does not alter the spontaneous incidence of diabetes, the incidence of cyclophosphamide-induced diabetes, or the activation of diabetogenic T-cell receptor transgenic CD4(+) T cells after adoptive transfer. Our results show that one or more of the many other linked genetic variants between the B10 and NOD genome are required for the diabetes protection conferred by Idd5.1. With the NOD mouse model closely mimicking the human disease, our data demonstrate that knock-in transgenic mice on the NOD background can test causative mutations relevant in human diabetes.
Collapse
Affiliation(s)
- Fabian Jakubczik
- Department of Pathology, University of Cambridge, Cambridge, U.K
| | - Ken Jones
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge, U.K. Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, U.K
| | - Jennifer Nichols
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge, U.K. Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, U.K
| | - William Mansfield
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge, U.K
| | - Anne Cooke
- Department of Pathology, University of Cambridge, Cambridge, U.K
| | - Nick Holmes
- Department of Pathology, University of Cambridge, Cambridge, U.K.
| |
Collapse
|
23
|
Michels A, Zhang L, Khadra A, Kushner JA, Redondo MJ, Pietropaolo M. Prediction and prevention of type 1 diabetes: update on success of prediction and struggles at prevention. Pediatr Diabetes 2015; 16. [PMID: 26202050 PMCID: PMC4592445 DOI: 10.1111/pedi.12299] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is the archetypal example of a T cell-mediated autoimmune disease characterized by selective destruction of pancreatic β cells. The pathogenic equation for T1DM presents a complex interrelation of genetic and environmental factors, most of which have yet to be identified. On the basis of observed familial aggregation of T1DM, it is certain that there is a decided heritable genetic susceptibility for developing T1DM. The well-known association of T1DM with certain human histocompatibility leukocyte antigen (HLA) alleles of the major histocompatibility complex (MHC) was a major step toward understanding the role of inheritance in T1DM. Type 1 diabetes is a polygenic disease with a small number of genes having large effects (e.g., HLA) and a large number of genes having small effects. Risk of T1DM progression is conferred by specific HLA DR/DQ alleles [e.g., DRB1*03-DQB1*0201 (DR3/DQ2) or DRB1*04-DQB1*0302 (DR4/DQ8)]. In addition, the HLA allele DQB1*0602 is associated with dominant protection from T1DM in multiple populations. A concordance rate lower than 100% between monozygotic twins indicates a potential involvement of environmental factors on disease development. The detection of at least two islet autoantibodies in the blood is virtually pre-diagnostic for T1DM. The majority of children who carry these biomarkers, regardless of whether they have an a priori family history of the disease, will develop insulin-requiring diabetes. Facilitating pre-diagnosis is the timing of seroconversion which is most pronounced in the first 2 yr of life. Unfortunately the significant progress in improving prediction of T1DM has not yet been paralleled by safe and efficacious intervention strategies aimed at preventing the disease. Herein we summarize the chequered history of prediction and prevention of T1DM, describing successes and failures alike, and thereafter examine future trends in the exciting, partially explored field of T1DM prevention.
Collapse
Affiliation(s)
- Aaron Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado
| | - Li Zhang
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, QC Canada
| | - Jake A. Kushner
- Division of Diabetes Pediatric Endocrinology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Maria J. Redondo
- Division of Diabetes Pediatric Endocrinology, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas
| | - Massimo Pietropaolo
- Division of Diabetes, Endocrinology and Metabolism, McNair Medical Institute, Baylor College of Medicine, Houston, Texas,To Whom Correspondence May be Addressed: Massimo Pietropaolo, M.D., Division of Diabetes, Endocrinology and Metabolism, Alkek Building for Biomedical Research, R 609, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030
| |
Collapse
|
24
|
Salant DJ. Podocyte Expression of B7-1/CD80: Is it a Reliable Biomarker for the Treatment of Proteinuric Kidney Diseases with Abatacept? J Am Soc Nephrol 2015; 27:963-5. [PMID: 26400567 DOI: 10.1681/asn.2015080947] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- David J Salant
- Renal Section and Department of Medicine, Boston University Medical Center, Boston, Massachusetts
| |
Collapse
|
25
|
Lim S, Kim WJ, Kim YH, Lee S, Koo JH, Lee JA, Yoon H, Kim DH, Park HJ, Kim HM, Lee HG, Yun Kim J, Lee JU, Hun Shin J, Kyun Kim L, Doh J, Kim H, Lee SK, Bothwell ALM, Suh M, Choi JM. dNP2 is a blood-brain barrier-permeable peptide enabling ctCTLA-4 protein delivery to ameliorate experimental autoimmune encephalomyelitis. Nat Commun 2015; 6:8244. [PMID: 26372309 PMCID: PMC4579786 DOI: 10.1038/ncomms9244] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 07/31/2015] [Indexed: 01/06/2023] Open
Abstract
Central nervous system (CNS)-infiltrating effector T cells play critical roles in the development and progression of multiple sclerosis (MS). However, current drugs for MS are very limited due to the difficulty of delivering drugs into the CNS. Here we identify a cell-permeable peptide, dNP2, which efficiently delivers proteins into mouse and human T cells, as well as various tissues. Moreover, it enters the brain tissue and resident cells through blood vessels by penetrating the tightly organized blood-brain barrier. The dNP2-conjugated cytoplasmic domain of cytotoxic T-lymphocyte antigen 4 (dNP2-ctCTLA-4) negatively regulates activated T cells and shows inhibitory effects on experimental autoimmune encephalomyelitis in both preventive and therapeutic mouse models, resulting in the reduction of demyelination and CNS-infiltrating T helper 1 and T helper 17 cells. Thus, this study demonstrates that dNP2 is a blood-brain barrier-permeable peptide and dNP2-ctCTLA-4 could be an effective agent for treating CNS inflammatory diseases such as MS.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Won-Ju Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Yeon-Ho Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Sohee Lee
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 440-746, Republic of Korea.,Samsung Advanced Institute for Health Sciences &Technology (SAIHST), Seoul 135-710, Republic of Korea
| | - Ja-Hyun Koo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Jung-Ah Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Heeseok Yoon
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Do-Hyun Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Hong-Jai Park
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Hye-Mi Kim
- Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Hong-Gyun Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Ji Yun Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Jae-Ung Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea
| | - Jae Hun Shin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Lark Kyun Kim
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Junsang Doh
- Department of Mechanical Engineering, School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Hongtae Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 440-746, Republic of Korea.,Department of Biological Science, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Sang-Kyou Lee
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Minah Suh
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 440-746, Republic of Korea.,Samsung Advanced Institute for Health Sciences &Technology (SAIHST), Seoul 135-710, Republic of Korea.,Department of Biological Science, Sungkyunkwan University, Suwon 440-746, Republic of Korea.,Department of Biomedical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.,Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 440-746, Republic of Korea
| |
Collapse
|
26
|
Ovcinnikovs V, Walker LSK. Regulatory T Cells in Autoimmune Diabetes: Mechanisms of Action and Translational Potential. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 136:245-77. [PMID: 26615100 DOI: 10.1016/bs.pmbts.2015.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Since the discovery of specialized T cells with regulatory function, harnessing the power of these cells to ameliorate autoimmunity has been a major goal. Here we collate the evidence that regulatory T cells (Treg) can inhibit Type 1 diabetes in animal models and humans. We discuss the anatomical sites and molecular mechanisms of Treg suppressive function in the Type 1 diabetes setting, citing evidence that Treg can function in both the pancreatic lymph nodes and within the pancreatic lesion. Involvement of the CTLA-4 pathway, as well as TGF-β and IL-2 deprivation will be considered. Finally, we summarize current efforts to manipulate Treg therapeutically in individuals with Type 1 diabetes. The translation of this research area from bench to bedside is still in its infancy, but the remarkable therapeutic potential of successfully manipulating Treg populations is clear to see.
Collapse
Affiliation(s)
- Vitalijs Ovcinnikovs
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, London, United Kingdom.
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, London, United Kingdom
| |
Collapse
|
27
|
Hou TZ, Qureshi OS, Wang CJ, Baker J, Young SP, Walker LSK, Sansom DM. A transendocytosis model of CTLA-4 function predicts its suppressive behavior on regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:2148-59. [PMID: 25632005 DOI: 10.4049/jimmunol.1401876] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Manipulation of the CD28/CTLA-4 pathway is at the heart of a number of immunomodulatory approaches used in both autoimmunity and cancer. Although it is clear that CTLA-4 is a critical regulator of T cell responses, the immunological contexts in which CTLA-4 controls immune responses are not well defined. In this study, we show that whereas CD80/CD86-dependent activation of resting human T cells caused extensive T cell proliferation and robust CTLA-4 expression, in this context CTLA-4 blocking Abs had no impact on the response. In contrast, in settings where CTLA-4(+) cells were present as "regulators," inhibition of resting T cell responses was dependent on CTLA-4 expression and specifically related to the number of APC. At low numbers of APC or low levels of ligand, CTLA-4-dependent suppression was highly effective whereas at higher APC numbers or high levels of ligand, inhibition was lost. Accordingly, the degree of suppression correlated with the level of CD86 expression remaining on the APC. These data reveal clear rules for the inhibitory function of CTLA-4 on regulatory T cells, which are predicted by its ability to remove ligands from APC.
Collapse
Affiliation(s)
- Tie Zheng Hou
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| | - Omar S Qureshi
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Chun Jing Wang
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| | - Jennifer Baker
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stephen P Young
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Lucy S K Walker
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| | - David M Sansom
- Division of Infection and Immunity, Department of Immunology, Institute of Immunity and Transplantation, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom; and
| |
Collapse
|
28
|
Evidence for genes controlling resistance to Heligmosomoides bakeri on mouse chromosome 1. Parasitology 2014; 142:566-75. [PMID: 25377239 DOI: 10.1017/s0031182014001644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Resistance to infections with Heligmosomoides bakeri is associated with a significant quantitative trait locus (QTL-Hbnr1) on mouse chromosome 1 (MMU1). We exploited recombinant mice, with a segment of MMU1 from susceptible C57Bl/10 mice introgressed onto MMU1 in intermediate responder NOD mice (strains 1094 and 6109). BALB/c (intermediate responder) and C57Bl/6 mice (poor responder) were included as control strains and strain 1098 (B10 alleles on MMU3) as NOD controls. BALB/c mice resisted infection rapidly and C57Bl/6 accumulated heavy worm burdens. Fecal egg counts dropped by weeks 10-11 in strain 1098, but strains 1094 and 6109 continued to produce eggs, harbouring more worms when autopsied (day 77). PubMed search identified 3 genes (Ctla4, Cd28, Icos) as associated with 'Heligmosomoides' in the B10 insert. Single nucleotide polymorphism (SNP) differences in Ctla4 could be responsible for regulatory changes in gene function, and a SNP within a splice site in Cd28 could have an impact on function, but no polymorphisms with predicted effects on function were found in Icos. Therefore, one or more genes encoded in the B10 insert into NOD mice contribute to the response phenotype, narrowing down the search for genes underlying the H. bakeri resistance QTL, and suggest Cd28 and Ctla4 as candidate genes.
Collapse
|
29
|
Devarajan P, Miska J, Lui JB, Swieboda D, Chen Z. Opposing effects of CTLA4 insufficiency on regulatory versus conventional T cells in autoimmunity converge on effector memory in target tissue. THE JOURNAL OF IMMUNOLOGY 2014; 193:4368-80. [PMID: 25246499 DOI: 10.4049/jimmunol.1400876] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Quantitative variations in CTLA4 expression, because of genetic polymorphisms, are associated with various human autoimmune conditions, including type 1 diabetes (T1D). Extensive studies have demonstrated that CTLA4 is not only essential for the suppressive role of regulatory T cells (T(reg)) but also required for intrinsic control of conventional T (T(conv)) cells. We report that a modest insufficiency of CTLA4 in mice, which mimics the effect of some human CTLA4 genetic polymorphisms, accompanied by a T1D-permissive MHC locus, was sufficient to induce juvenile-onset diabetes on an otherwise T1D-resistant genetic background. Reduction in CTLA4 levels had an unanticipated effect in promoting Treg function both in vivo and in vitro. It led to an increase in T(reg) memory in both lymphoid and nonlymphoid target tissue. Conversely, modulating CTLA4 by either RNA interference or Ab blockade promoted conventional effector memory T cell formation in the T(conv) compartment. The CD4(+) conventional effector memory T cells, including those within target tissue, produced IL-17 or IFN-γ. Blocking IL-7 signaling reduced the Th17 autoimmune compartment but did not suppress the T1D induced by CTLA4 insufficiency. Enhanced effector memory formation in both T(conv) and T(reg) lineages may underpin the apparently dichotomized impact of CTLA4 insufficiency on autoimmune pathogenesis. Therefore, although the presence of CTLA4 plays a critical role in controlling homeostasis of T cells, its quantitative variation may impose diverse or even opposing effects on distinct lineages of T cells, an optimal sum of which is necessary for preservation of T cell immunity while suppressing tissue damage.
Collapse
Affiliation(s)
- Priyadharshini Devarajan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136; and
| | - Jason Miska
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136; and
| | - Jen Bon Lui
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136; and
| | - Dominika Swieboda
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136; and
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136; and Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
30
|
Gardner D, Jeffery LE, Sansom DM. Understanding the CD28/CTLA-4 (CD152) pathway and its implications for costimulatory blockade. Am J Transplant 2014; 14:1985-91. [PMID: 25098238 DOI: 10.1111/ajt.12834] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 01/25/2023]
Abstract
T cell activation is a key event in the adaptive immune system and vital in the generation of protective cellular and humoral immunity. Activation is required to generate CD4 effector T cell responses and provide help for B cell and cytotoxic T cell responses. While defective T responses to foreign antigen result in infectious pathology, over-reactive T cell responses against self-antigens result in autoimmunity and, in a transplantation setting, tissue rejection. Understanding how T cell activation is normally regulated is critical to therapeutic intervention and the CD28/CTLA-4 (CD152) pathway represents the initial activation checkpoint in molecular terms. In particular, while the CTLA-4 pathway is well established as an essential regulator of self-reactivity, its mechanism of action is still uncertain. Such mechanistic issues are important given its central position in T cell activation and the increasing number of therapeutic modalities aimed at manipulating the CD28/CTLA-4 pathway. Here, we provide an updated view of CTLA-4 biology, reviewing the established features of the system and highlighting its interplay with CD28. We then discuss how recent progress in our understanding of this pathway affects our interpretations following intervention.
Collapse
Affiliation(s)
- D Gardner
- University of Birmingham, MRC Centre for Immune Regulation, Birmingham, UK
| | | | | |
Collapse
|
31
|
Zhong J, Rao X, Braunstein Z, Taylor A, Narula V, Hazey J, Mikami D, Needleman B, Rutsky J, Sun Q, Deiuliis JA, Satoskar AR, Rajagopalan S. T-cell costimulation protects obesity-induced adipose inflammation and insulin resistance. Diabetes 2014; 63:1289-302. [PMID: 24222350 PMCID: PMC4179314 DOI: 10.2337/db13-1094] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A key pathophysiologic role for activated T-cells in mediating adipose inflammation and insulin resistance (IR) has been recently postulated. However, mechanisms underlying their activation are poorly understood. In this study, we demonstrated a previously unrecognized homeostatic role for the costimulatory B7 molecules (CD80 and CD86) in preventing adipose inflammation. Instead of promoting inflammation, which was found in many other disease conditions, B7 costimulation reduced adipose inflammation by maintaining regulatory T-cell (Treg) numbers in adipose tissue. In both humans and mice, expression of CD80 and CD86 was negatively correlated with the degree of IR and adipose tissue macrophage infiltration. Decreased B7 expression in obesity appeared to directly impair Treg proliferation and function that lead to excessive proinflammatory macrophages and the development of IR. CD80/CD86 double knockout (B7 KO) mice had enhanced adipose macrophage inflammation and IR under both high-fat and normal diet conditions, accompanied by reduced Treg development and proliferation. Adoptive transfer of Tregs reversed IR and adipose inflammation in B7 KO mice. Our results suggest an essential role for B7 in maintaining Tregs and adipose homeostasis and may have important implications for therapies that target costimulation in type 2 diabetes.
Collapse
Affiliation(s)
- Jixin Zhong
- Division of Cardiology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Xiaoquan Rao
- Division of Cardiology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Zachary Braunstein
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
| | - Anne Taylor
- Department of Surgery, The Ohio State University, Columbus, OH
| | - Vimal Narula
- Department of Surgery, The Ohio State University, Columbus, OH
| | - Jeffrey Hazey
- Department of Surgery, The Ohio State University, Columbus, OH
| | - Dean Mikami
- Department of Surgery, The Ohio State University, Columbus, OH
| | | | - Jessica Rutsky
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
| | - Qinghua Sun
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
| | - Jeffrey A. Deiuliis
- Division of Cardiology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Abhay R. Satoskar
- Division of Experimental Pathology, The Ohio State University, Columbus, OH
| | - Sanjay Rajagopalan
- Division of Cardiology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
- Corresponding author: Sanjay Rajagopalan,
| |
Collapse
|
32
|
Mourich DV, Oda SK, Schnell FJ, Crumley SL, Hauck LL, Moentenich CA, Marshall NB, Hinrichs DJ, Iversen PL. Alternative splice forms of CTLA-4 induced by antisense mediated splice-switching influences autoimmune diabetes susceptibility in NOD mice. Nucleic Acid Ther 2014; 24:114-26. [PMID: 24494586 DOI: 10.1089/nat.2013.0449] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Activated and regulatory T cells express the negative co-stimulatory molecule cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) that binds B7 on antigen-presenting cells to mediate cellular responses. Single nucleotide polymorphisms in the CTLA-4 gene have been found to affect alternative splicing and are linked to autoimmune disease susceptibility or resistance. Increased expression of a soluble splice form (sCTLA-4), lacking the transmembrane domain encoded by exon 3, has been shown to accelerate autoimmune pathology. In contrast, an exon 2-deficient form lacking the B7 ligand binding domain (liCTLA-4), expressed by diabetes resistant mouse strains has been shown to be protective when expressed as a transgene in diabetes susceptible non-obese diabetic (NOD) mice. We sought to employ an antisense-targeted splice-switching approach to independently produce these CTLA-4 splice forms in NOD mouse T cells and observe their relative impact on spontaneous autoimmune diabetes susceptibility. In vitro antisense targeting of the splice acceptor site for exon 2 produced liCTLA-4 while targeting exon 3 produced the sCTLA-4 form in NOD T cells. The liCTLA-4 expressing T cells exhibited reduced activation, proliferation and increased adhesion to intercellular adhesion molecule-1 (ICAM-1) similar to treatment with agonist α-CTLA-4. Mice treated to produce liCTLA-4 at the time of elevated blood glucose levels exhibited a significant reduction in the incidence of insulitis and diabetes, whereas a marked increase in the incidence of both was observed in animals treated to produce sCTLA-4. These findings provide further support that alternative splice forms of CTLA-4 affects diabetes susceptibility in NOD mice and demonstrates the therapeutic utility of antisense mediated splice-switching for modulating immune responses.
Collapse
|
33
|
Abstract
T cell activation is a key event in the adaptive immune response and vital to the generation of both cellular and humoral immunity. Activation is required not only for effective CD4 T cell responses but also to provide help for B cells and the generation of cytotoxic T cell responses. Unsurprisingly, impaired T cell activation results in infectious pathology, whereas dysregulated activation can result in autoimmunity. The decision to activate is therefore tightly regulated and the CD28/CTLA-4 pathway represents this apical decision point at the molecular level. In particular, CTLA-4 (CD152) is an essential checkpoint control for autoimmunity; however, the molecular mechanism(s) by which CTLA-4 achieves its regulatory function are not well understood, especially how it functionally intersects with the CD28 pathway. In this chapter, we review the established molecular and cellular concepts relating to CD28 and CTLA-4 biology, and attempt to integrate these by discussing the transendocytosis of ligands as a new model of CTLA-4 function.
Collapse
Affiliation(s)
- Blagoje Soskic
- School of Immunity and Infection, University of Birmingham, Birmingham, United Kingdom
| | | | - Tiezheng Hou
- UCL Institute of Immunity and Transplantation, Royal Free Campus, London, United Kingdom
| | - David M Sansom
- UCL Institute of Immunity and Transplantation, Royal Free Campus, London, United Kingdom.
| |
Collapse
|
34
|
Abstract
An increasing body of evidence suggests that immune-mediated processes affect female reproductive success at multiple levels. Crosstalk between endocrine and immune systems regulates a large number of biological processes that affect target tissues, and this crosstalk involves gene expression, cytokine and/or lymphokine release and hormone action. In addition, endocrine-immune interactions have a major role in the implantation process of the fetal (paternally derived) semi-allograft, which requires a reprogramming process of the maternal immune system from rejection to temporary tolerance for the length of gestation. Usually, the female immune system is supportive of all of these processes and, therefore, facilitates reproductive success. Abnormalities of the female immune system, including autoimmunity, potentially interfere at multiple levels. The relevance of the immune system to female infertility is increasingly recognized by investigators, but clinically is often not adequately considered and is, therefore, underestimated. This Review summarizes the effect of individual autoimmune endocrine diseases on female fertility, and points towards selected developments expected in the near future.
Collapse
Affiliation(s)
- Aritro Sen
- The Center for Human Reproduction (CHR), 21 East 69th Street, New York, NY 10021, USA
| | - Vitaly A Kushnir
- The Center for Human Reproduction (CHR), 21 East 69th Street, New York, NY 10021, USA
| | - David H Barad
- The Center for Human Reproduction (CHR), 21 East 69th Street, New York, NY 10021, USA
| | - Norbert Gleicher
- The Center for Human Reproduction (CHR), 21 East 69th Street, New York, NY 10021, USA
| |
Collapse
|
35
|
Advances in our understanding of the pathophysiology of Type 1 diabetes: lessons from the NOD mouse. Clin Sci (Lond) 2013; 126:1-18. [PMID: 24020444 DOI: 10.1042/cs20120627] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
T1D (Type 1 diabetes) is an autoimmune disease caused by the immune-mediated destruction of pancreatic β-cells. Studies in T1D patients have been limited by the availability of pancreatic samples, a protracted pre-diabetic phase and limitations in markers that reflect β-cell mass and function. The NOD (non-obese diabetic) mouse is currently the best available animal model of T1D, since it develops disease spontaneously and shares many genetic and immunopathogenic features with human T1D. Consequently, the NOD mouse has been extensively studied and has made a tremendous contribution to our understanding of human T1D. The present review summarizes the key lessons from NOD mouse studies concerning the genetic susceptibility, aetiology and immunopathogenic mechanisms that contribute to autoimmune destruction of β-cells. Finally, we summarize the potential and limitations of immunotherapeutic strategies, successful in NOD mice, now being trialled in T1D patients and individuals at risk of developing T1D.
Collapse
|
36
|
Walker LSK. Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun 2013; 45:49-57. [PMID: 23849743 PMCID: PMC3989116 DOI: 10.1016/j.jaut.2013.06.006] [Citation(s) in RCA: 294] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 01/12/2023]
Abstract
Both the CTLA-4 pathway and regulatory T cells (Treg) are essential for the control of immune homeostasis. Their therapeutic relevance is highlighted by the increasing use of anti-CTLA-4 antibody in tumor therapy and the development of Treg cell transfer strategies for use in autoimmunity and transplantation settings. The CTLA-4 pathway first came to the attention of the immunological community in 1995 with the discovery that mice deficient in Ctla-4 suffered a fatal lymphoproliferative syndrome. Eight years later, mice lacking the critical Treg transcription factor Foxp3 were shown to exhibit a remarkably similar phenotype. Much of the debate since has centered on the question of whether Treg suppressive function requires CTLA-4. The finding that it does in some settings but not in others has provoked controversy and inevitable polarization of opinion. In this article, I suggest that CTLA-4 and Treg represent complementary and largely overlapping mechanisms of immune tolerance. I argue that Treg commonly use CTLA-4 to effect suppression, however CTLA-4 can also function in the non-Treg compartment while Treg can invoke CTLA-4-independent mechanisms of suppression. The notion that Foxp3 and CTLA-4 direct independent programs of immune regulation, which in practice overlap to a significant extent, will hopefully help move us towards a better appreciation of the underlying biology and therapeutic significance of these pathways.
Collapse
Affiliation(s)
- Lucy S K Walker
- Institute of Immunity & Transplantation, University College London Medical School, Royal Free Campus, Rowland Hill Street, London NW3 2PF, UK.
| |
Collapse
|
37
|
Bour-Jordan H, Thompson HL, Giampaolo JR, Davini D, Rosenthal W, Bluestone JA. Distinct genetic control of autoimmune neuropathy and diabetes in the non-obese diabetic background. J Autoimmun 2013; 45:58-67. [PMID: 23850635 PMCID: PMC4156399 DOI: 10.1016/j.jaut.2013.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 02/01/2023]
Abstract
The non-obese diabetic (NOD) mouse is susceptible to the development of autoimmune diabetes but also multiple other autoimmune diseases. Over twenty susceptibility loci linked to diabetes have been identified in NOD mice and progress has been made in the definition of candidate genes at many of these loci (termed Idd for insulin-dependent diabetes). The susceptibility to multiple autoimmune diseases in the NOD background is a unique opportunity to examine susceptibility genes that confer a general propensity for autoimmunity versus susceptibility genes that control individual autoimmune diseases. We previously showed that NOD mice deficient for the costimulatory molecule B7-2 (NOD-B7-2KO mice) were protected from diabetes but spontaneously developed an autoimmune peripheral neuropathy. Here, we took advantage of multiple NOD mouse strains congenic for Idd loci to test the role of these Idd loci the development of neuropathy and determine if B6 alleles at Idd loci that are protective for diabetes will also be for neuropathy. Thus, we generated NOD-B7-2KO strains congenic at Idd loci and examined the development of neuritis and clinical neuropathy. We found that the NOD-H-2(g7) MHC region is necessary for development of neuropathy in NOD-B7-2KO mice. In contrast, other Idd loci that significantly protect from diabetes did not affect neuropathy when considered individually. However, we found potent genetic interactions of some Idd loci that provided almost complete protection from neuritis and clinical neuropathy. In addition, defective immunoregulation by Tregs could supersede protection by some, but not other, Idd loci in a tissue-specific manner in a model where neuropathy and diabetes occurred concomitantly. Thus, our study helps identify Idd loci that control tissue-specific disease or confer general susceptibility to autoimmunity, and brings insight to the Treg-dependence of autoimmune processes influenced by given Idd region in the NOD background.
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- University of California in San Francisco, 513 Parnassus Avenue, Box 0400, San Francisco, CA 94143-0400, USA
| | | | | | | | | | | |
Collapse
|