1
|
Mauvais FX, van Endert PM. Type 1 Diabetes: A Guide to Autoimmune Mechanisms for Clinicians. Diabetes Obes Metab 2025. [PMID: 40375390 DOI: 10.1111/dom.16460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025]
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic beta cells by autoreactive T lymphocytes, leading to insulin deficiency and lifelong insulin dependence. It develops in genetically predisposed individuals, triggered by environmental or immunological factors. Although the exact causes of T1D remain unknown, the autoimmune pathogenesis of the disease is clearly indicated by the genetic risk conferred by allelic human leukocyte antigens (HLA), the almost obligatory presence of islet cell autoantibodies (AAbs) and immune cell infiltration of pancreatic islets from patients. At the same time, epidemiological data point to a role of environmental factors, notably enteroviral infections, in the disease, although precise causative links between specific pathogens and T1D have been difficult to establish. Studies of human pancreas organs from patients made available through repositories and the advent of high-dimensional high-throughput technologies for genomic and proteomic studies have significantly elucidated our understanding of the disease in recent years and provided mechanistic insights that can be exploited for innovative targeted therapeutic approaches. This short overview will summarise current salient knowledge on immune cell and beta cell dysfunction in T1D pathogenesis. PLAIN LANGUAGE SUMMARY: Type 1 diabetes (T1D) is a chronic disease where the body's own immune system attacks and destroys the insulin-producing beta cells in the pancreas. This leads to a lack of insulin, a hormone essential for regulating blood sugar, which means people with T1D need insulin for life. The disease can develop at any age but is most diagnosed in children and young adults. Despite advances in treatment, T1D still significantly reduces life expectancy, especially in countries with fewer healthcare resources. T1D develops in people with a genetic predisposition, often triggered by environmental factors such as viral infections or changes in the gut microbiome. The disease progresses silently through three stages: Stage 1: Autoantibodies to beta cell components appear, signalling the immune system is reacting against the pancreas, but there are no symptoms; Stage 2: Beta cell function starts to decline, but fasting blood sugar is still normal; Stage 3: Enough beta cells are destroyed that fasting blood sugar rises, and symptoms of diabetes appear. The risk of progressing from stage 1 to full-blown diabetes is about 35-50% within five years, and even higher from stage 2. Over 60 genes are linked to T1D risk, most of which affect how the immune system works. The strongest genetic risk comes from specific versions of histocompatibility genes, which help the immune system distinguish between the body's own cells and invaders. Some types of these genes make it easier for the immune system to mistakenly attack beta cells. However, 90% of people diagnosed with T1D have no family member with T1D, showing that genetics is only part of the story. Environmental factors also play a big role. For example, certain viral infections, especially with viruses infecting the intestine, are associated with a higher risk of developing T1D. The gut microbiome - the community of bacteria living in our intestines - also influences risk, with healthier, more diverse microbiomes appearing to offer some protection. In T1D, immune cells - especially so-called T lymphocytes - mistake beta cells in the pancreas for threats and destroy them. This process is called autoimmunity. The attack is often reflected by the presence of autoantibodies against proteins found in beta cells. Over time, as more beta cells are lost, the body can no longer produce enough insulin, leading to the symptoms of diabetes. Interestingly, not all people with T1D have the same pattern of disease. For example, children diagnosed before age 7 often have more aggressive disease, more autoantibodies, and stronger genetic risk factors than those diagnosed later. Much of our understanding of T1D has come from studying animal models, but new technologies now allow researchers to study human pancreas tissue and blood immune cells in greater detail. Scientists are also exploring how the gut microbiome, diet, and environmental exposures contribute to T1D risk and progression. Treatment currently focuses on replacing insulin, but researchers are working on therapies that target the immune system or aim to protect or replace beta cells. Strategies include immunotherapy, gene therapy, and even modifying the gut microbiome. The goal is to prevent or reverse the disease, not just manage its symptoms. In summary, T1D is a complex autoimmune disease influenced by both genes and the environment. It progresses silently before symptoms appear, and while insulin therapy is life-saving, new research is paving the way for treatments that could one day halt or even prevent the disease.
Collapse
Affiliation(s)
- François-Xavier Mauvais
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Service de Physiologie - Explorations Fonctionnelles Pédiatriques, AP-HP, Hôpital Universitaire Robert Debré, Paris, France
| | - Peter M van Endert
- Université Paris Cité, INSERM, CNRS, Institut Necker Enfants Malades, Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker - Enfants Malades, Paris, France
| |
Collapse
|
2
|
Walker SL, Leete P, Boldison J. Tissue Resident and Infiltrating Immune Cells: Their Influence on the Demise of Beta Cells in Type 1 Diabetes. Biomolecules 2025; 15:441. [PMID: 40149976 PMCID: PMC11939886 DOI: 10.3390/biom15030441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease that results in the selective loss of pancreatic beta cells and an eventual deficit in insulin production to maintain glucose homeostasis. It is now increasingly accepted that this dynamic disease process is multifactorial; involves a variety of immune cells which contribute to an inflamed pancreatic microenvironment; and that the condition is heterogenous, resulting in variable rates of subsequent beta cell damage. In this review, we will explore the current understanding of the cellular interactions between both resident and infiltrating immune cells within the pancreatic environment, highlighting key mechanisms which may promote the beta cell destruction and islet damage associated with T1D.
Collapse
Affiliation(s)
| | | | - Joanne Boldison
- Department of Clinical and Biomedical Sciences, University of Exeter, RILD Building (Level 4), Barrack Road, Exeter EX2 5DW, UK; (S.L.W.); (P.L.)
| |
Collapse
|
3
|
Lacorcia M, Bhattacharjee P, Foster A, Hardy MY, Tye-Din JA, Karas JA, Wentworth JM, Cameron FJ, Mannering SI. BASTA, a simple whole-blood assay for measuring β cell antigen-specific CD4 + T cell responses in type 1 diabetes. Sci Transl Med 2025; 17:eadt2124. [PMID: 40106580 DOI: 10.1126/scitranslmed.adt2124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease where T cells mediate the destruction of the insulin-producing β cells found within the islets of Langerhans in the pancreas. Autoantibodies to β cell antigens are the only tests available to detect β cell autoimmunity. T cell responses to β cell antigens, which are known to cause T1D, can only be measured in research settings because of the complexity of assays and the large blood volumes required. Here, we describe the β cell antigen-specific T cell assay (BASTA). BASTA is a simple whole-blood assay that can detect human CD4+ T cell responses to β cell antigens by measuring antigen-stimulated interleukin-2 (IL-2) production. BASTA is both more sensitive and specific than the CFSE (carboxyfluorescein diacetate succinimidyl ester)-based proliferation assay. We used BASTA to identify the regions of preproinsulin that stimulated T cell responses specifically in blood from people with T1D. BASTA can be done with as little as 2 to 3 milliliters of blood. We found that effector memory CD4+ T cells are the primary producers of IL-2 in response to preproinsulin peptides. We then evaluated responses to individual and pooled preproinsulin peptides in a cross-sectional study of pediatric patients: without T1D, without T1D but with a first-degree relative with T1D, or diagnosed with T1D. In contrast with other preproinsulin peptides, full-length C-peptide (PI33-63) showed high specificity for T1D [area under the curve (AUC) = 0.86)]. We suggest that BASTA will be a useful tool for monitoring changes in β cell-specific CD4+ T cell responses both in research and clinical settings.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Pushpak Bhattacharjee
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Abby Foster
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Melinda Y Hardy
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - John A Karas
- School of Chemistry, University of Melbourne, Parkville, Victoria 3010, Australia
| | - John M Wentworth
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
- Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Fergus J Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Parkville, Victoria 3052, Australia
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
4
|
Forkan CP, Shrestha A, Yu A, Chuang C, Pociot F, Yarani R. Could hypoxic conditioning augment the potential of mesenchymal stromal cell-derived extracellular vesicles as a treatment for type 1 diabetes? Stem Cell Res Ther 2025; 16:37. [PMID: 39901225 PMCID: PMC11792614 DOI: 10.1186/s13287-025-04153-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
Type1 Diabetes (T1D) is an autoimmune disorder characterised by the loss of pancreatic β-cells. This β cell loss occurs primarily through inflammatory pathways culminating in apoptosis. Mesenchymal stromal cells (MSCs) have been heavily studied for therapeutic applications due to their regenerative, anti-apoptotic, immunomodulatory, and anti-inflammatory properties. The therapeutic effects of MSCs are mediated through cell-to-cell contact, differentiation, and the release of paracrine factors, which include the release of extracellular vesicles (EVs). Culturing MSCs in hypoxia, a low oxygen tension state more analogous to their physiological environment, seems to increase the therapeutic efficacy of MSC cell therapy, enhancing their immunomodulatory, anti-inflammatory, and anti-fibrotic properties. This is also the case with MSC-derived EVs, which show altered properties based on the parent cell preconditioning. In this review, we examine the evidence supporting the potential application of hypoxic preconditioning in strengthening MSC-EVs for treating the inflammatory and apoptotic causes of β cell loss in T1D.
Collapse
Affiliation(s)
- Cathal Patrick Forkan
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Aruna Shrestha
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Alfred Yu
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Christine Chuang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark.
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA.
| |
Collapse
|
5
|
Okamura T, Kitagawa N, Kitagawa N, Sakai K, Sumi M, Kobayashi G, Imai D, Matsui T, Hamaguchi M, Fukui M. Single-cell analysis reveals islet autoantigen's immune activation in type 1 diabetes patients. J Clin Biochem Nutr 2025; 76:64-84. [PMID: 39896168 PMCID: PMC11782777 DOI: 10.3164/jcbn.24-86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 02/04/2025] Open
Abstract
In this study, we used single-cell sequencing, which can comprehensively detect the type and number of transcripts per cell, to efficiently stimulate peripheral blood mononuclear cells of type 1 diabetic patients with overlapping peptides of GAD, IA-2, and insulin antigens, and performed gene expression analysis by single-cell variable-diversity-joining sequencing and T-cell receptor repertoire analysis. Twenty male patients with type 1 diabetes mellitus participating in the KAMOGAWA-DM cohort were included. Four of them were randomly selected for BD Rhapsody system after reacting peripheral blood mononuclear cells with overlapping peptides of GAD, IA-2, and insulin antigen. Peripheral blood mononuclear cells were clustered into CD8+ T cells, CD4+ T cells, granulocytes, natural killer cells, dendritic cells, monocytes, and B cells based on Seurat analysis. In the insulin group, gene expression of inflammatory cytokines was elevated in cytotoxic CD8+ T cells and Th1 and Th17 cells, and gene expression related to exhaustion was elevated in regulatory T cells. In T cell receptors of various T cells, the T cell receptor β chain was monoclonally increased in the TRBV28/TRBJ2-7 pairs. This study provides insights into the pathogenesis of type 1 diabetes and provides potential targets for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Noriyuki Kitagawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Diabetology, Kameoka Municipal Hospital, 1-1 Shinonoda, Shino-cho, Kameoka 621-8585, Japan
| | - Nobuko Kitagawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kimiko Sakai
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Madoka Sumi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Genki Kobayashi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Dan Imai
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takaaki Matsui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
6
|
Hanna SJ, Bonami RH, Corrie B, Westley M, Posgai AL, Luning Prak ET, Breden F, Michels AW, Brusko TM. The Type 1 Diabetes T Cell Receptor and B Cell Receptor Repository in the AIRR Data Commons: a practical guide for access, use and contributions through the Type 1 Diabetes AIRR Consortium. Diabetologia 2025; 68:186-202. [PMID: 39467874 PMCID: PMC11663175 DOI: 10.1007/s00125-024-06298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/19/2024] [Indexed: 10/30/2024]
Abstract
Human molecular genetics has brought incredible insights into the variants that confer risk for the development of tissue-specific autoimmune diseases, including type 1 diabetes. The hallmark cell-mediated immune destruction that is characteristic of type 1 diabetes is closely linked with risk conferred by the HLA class II gene locus, in combination with a broad array of additional candidate genes influencing islet-resident beta cells within the pancreas, as well as function, phenotype and trafficking of immune cells to tissues. In addition to the well-studied germline SNP variants, there are critical contributions conferred by T cell receptor (TCR) and B cell receptor (BCR) genes that undergo somatic recombination to yield the Adaptive Immune Receptor Repertoire (AIRR) responsible for autoimmunity in type 1 diabetes. We therefore created the T1D TCR/BCR Repository (The Type 1 Diabetes T Cell Receptor and B Cell Receptor Repository) to study these highly variable and dynamic gene rearrangements. In addition to processed TCR and BCR sequences, the T1D TCR/BCR Repository includes detailed metadata (e.g. participant demographics, disease-associated parameters and tissue type). We introduce the Type 1 Diabetes AIRR Consortium goals and outline methods to use and deposit data to this comprehensive repository. Our ultimate goal is to facilitate research community access to rich, carefully annotated immune AIRR datasets to enable new scientific inquiry and insight into the natural history and pathogenesis of type 1 diabetes.
Collapse
MESH Headings
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/genetics
- Humans
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Autoimmunity
Collapse
Affiliation(s)
- Stephanie J Hanna
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK.
| | - Rachel H Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
| | - Brian Corrie
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- iReceptor Genomic Services, Summerland, BC, Canada
| | | | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix Breden
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- iReceptor Genomic Services, Summerland, BC, Canada
| | - Aaron W Michels
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, USA.
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Bhattacharjee P, Pakusch M, Lacorcia M, Tresoldi E, Rubin AF, Foster A, King L, Chiu CY, Kay TWH, Karas JA, Cameron FJ, Mannering SI. Proinsulin C-peptide is a major source of HLA-DQ8 restricted hybrid insulin peptides recognized by human islet-infiltrating CD4 + T cells. PNAS NEXUS 2024; 3:pgae491. [PMID: 39554513 PMCID: PMC11565411 DOI: 10.1093/pnasnexus/pgae491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/11/2024] [Indexed: 11/19/2024]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that develops when T cells destroy the insulin-producing beta cells that reside in the pancreatic islets. Immune cells, including T cells, infiltrate the islets and gradually destroy the beta cells. Human islet-infiltrating CD4+ T cells recognize peptide epitopes derived from proinsulin, particularly C-peptide. Hybrid insulin peptides (HIPs) are neoepitopes formed by the fusion of two peptides derived from beta cell granule proteins and are known to be the targets of pathogenic CD4+ T cells in the non-obese diabetic (NOD) mouse and human islet-infiltrating CD4+ T cells. Proinsulin is widely recognized as a central antigen in T1D, but its role in forming HIPs is unclear. We developed a method to functionally screen TCRs derived from human islet-infiltrating CD4+ T cells and applied this to the identification of new proinsulin-derived HIPs. We generated a library of 4,488 candidate HIPs formed by fusion of proinsulin fragments and predicted to bind to HLA-DQ8. This library was screened against 109 islet-infiltrating CD4+ T cell receptors (TCRs) isolated from four organ donors who had T1D. We identified 13 unique HIPs recognized by nine different TCRs from two organ donors. HIP-specific T cell avatars responded specifically to a peptide extract from human islets. These new HIPs predominantly stimulated CD4+ T cell proliferation in peripheral blood mononuclear cells from individuals with T1D in contrast to HLA-matched controls. This is the first unbiased functional, islet-infiltrating T cell based, screen to identify proinsulin-derived HIPs. It has revealed many new HIPs and a central role of proinsulin C-peptide in their formation.
Collapse
Affiliation(s)
- Pushpak Bhattacharjee
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Miha Pakusch
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Matthew Lacorcia
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Eleonora Tresoldi
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Alan F Rubin
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Abby Foster
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Laura King
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Chris Y Chiu
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - Thomas W H Kay
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
| | - John A Karas
- School of Chemistry, University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Fergus J Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Parkville, Melbourne, VIC 3052, Australia
- Murdoch Children's Research Institute, Parkville, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC 3065, Australia
- Murdoch Children's Research Institute, Parkville, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, VIC 3065, Australia
| |
Collapse
|
8
|
Groegler J, Callebaut A, James EA, Delong T. The insulin secretory granule is a hotspot for autoantigen formation in type 1 diabetes. Diabetologia 2024; 67:1507-1516. [PMID: 38811417 DOI: 10.1007/s00125-024-06164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/11/2024] [Indexed: 05/31/2024]
Abstract
In type 1 diabetes, the insulin-producing beta cells of the pancreas are destroyed through the activity of autoreactive T cells. In addition to strong and well-documented HLA class II risk haplotypes, type 1 diabetes is associated with noncoding polymorphisms within the insulin gene locus. Furthermore, autoantibody prevalence data and murine studies implicate insulin as a crucial autoantigen for the disease. Studies identify secretory granules, where proinsulin is processed into mature insulin, stored and released in response to glucose stimulation, as a source of antigenic epitopes and neoepitopes. In this review, we integrate established concepts, including the role that susceptible HLA and thymic selection of the T cell repertoire play in setting the stage for autoimmunity, with emerging insights about beta cell and insulin secretory granule biology. In particular, the acidic, peptide-rich environment of secretory granules combined with its array of enzymes generates a distinct proteome that is unique to functional beta cells. These factors converge to generate non-templated peptide sequences that are recognised by autoreactive T cells. Although unanswered questions remain, formation and presentation of these epitopes and the resulting immune responses appear to be key aspects of disease initiation. In addition, these pathways may represent important opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jason Groegler
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aïsha Callebaut
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Thomas Delong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
9
|
Ghasemi Gojani E, Rai S, Norouzkhani F, Shujat S, Wang B, Li D, Kovalchuk O, Kovalchuk I. Targeting β-Cell Plasticity: A Promising Approach for Diabetes Treatment. Curr Issues Mol Biol 2024; 46:7621-7667. [PMID: 39057094 PMCID: PMC11275945 DOI: 10.3390/cimb46070453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The β-cells within the pancreas play a pivotal role in insulin production and secretion, responding to fluctuations in blood glucose levels. However, factors like obesity, dietary habits, and prolonged insulin resistance can compromise β-cell function, contributing to the development of Type 2 Diabetes (T2D). A critical aspect of this dysfunction involves β-cell dedifferentiation and transdifferentiation, wherein these cells lose their specialized characteristics and adopt different identities, notably transitioning towards progenitor or other pancreatic cell types like α-cells. This process significantly contributes to β-cell malfunction and the progression of T2D, often surpassing the impact of outright β-cell loss. Alterations in the expressions of specific genes and transcription factors unique to β-cells, along with epigenetic modifications and environmental factors such as inflammation, oxidative stress, and mitochondrial dysfunction, underpin the occurrence of β-cell dedifferentiation and the onset of T2D. Recent research underscores the potential therapeutic value for targeting β-cell dedifferentiation to manage T2D effectively. In this review, we aim to dissect the intricate mechanisms governing β-cell dedifferentiation and explore the therapeutic avenues stemming from these insights.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.)
| |
Collapse
|
10
|
Linsley PS, Nakayama M, Balmas E, Chen J, Barahmand-Pour-Whitman F, Bansal S, Bottorff T, Serti E, Speake C, Pugliese A, Cerosaletti K. Germline-like TCR-α chains shared between autoreactive T cells in blood and pancreas. Nat Commun 2024; 15:4971. [PMID: 38871688 PMCID: PMC11176301 DOI: 10.1038/s41467-024-48833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
Human type 1 diabetes (T1D) is caused by autoimmune attack on the insulin-producing pancreatic beta cells by islet antigen-reactive T cells. How human islet antigen-reactive (IAR) CD4+ memory T cells from peripheral blood affect T1D progression in the pancreas is poorly understood. Here, we aim to determine if IAR T cells in blood could be detected in pancreas. We identify paired αβ (TRA/TRB) T cell receptors (TCRs) in IAR T cells from the blood of healthy, at-risk, new-onset, and established T1D donors, and measured sequence overlap with TCRs in pancreata from healthy, at risk and T1D organ donors. We report extensive TRA junction sharing between IAR T cells and pancreas-infiltrating T cells (PIT), with perfect-match or single-mismatch TRA junction amino acid sequences comprising ~29% total unique IAR TRA junctions (942/3,264). PIT-matched TRA junctions were largely public and enriched for TRAV41 usage, showing significant nucleotide sequence convergence, increased use of germline-encoded versus non-templated residues in epitope engagement, and a potential for cross-reactivity. Our findings thus link T cells with distinctive germline-like TRA chains in the peripheral blood with T cells in the pancreas.
Collapse
Affiliation(s)
- Peter S Linsley
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elisa Balmas
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Janice Chen
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | | | - Shubham Bansal
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Ty Bottorff
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | | | - Cate Speake
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Alberto Pugliese
- Department of Diabetes Immunology & The Wanek Family Project for Type 1 Diabetes, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | | |
Collapse
|
11
|
Zdinak PM, Trivedi N, Grebinoski S, Torrey J, Martinez EZ, Martinez S, Hicks L, Ranjan R, Makani VKK, Roland MM, Kublo L, Arshad S, Anderson MS, Vignali DAA, Joglekar AV. De novo identification of CD4 + T cell epitopes. Nat Methods 2024; 21:846-856. [PMID: 38658646 PMCID: PMC11093748 DOI: 10.1038/s41592-024-02255-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
CD4+ T cells recognize peptide antigens presented on class II major histocompatibility complex (MHC-II) molecules to carry out their function. The remarkable diversity of T cell receptor sequences and lack of antigen discovery approaches for MHC-II make profiling the specificities of CD4+ T cells challenging. We have expanded our platform of signaling and antigen-presenting bifunctional receptors to encode MHC-II molecules presenting covalently linked peptides (SABR-IIs) for CD4+ T cell antigen discovery. SABR-IIs can present epitopes to CD4+ T cells and induce signaling upon their recognition, allowing a readable output. Furthermore, the SABR-II design is modular in signaling and deployment to T cells and B cells. Here, we demonstrate that SABR-IIs libraries presenting endogenous and non-contiguous epitopes can be used for antigen discovery in the context of type 1 diabetes. SABR-II libraries provide a rapid, flexible, scalable and versatile approach for de novo identification of CD4+ T cell ligands from single-cell RNA sequencing data using experimental and computational approaches.
Collapse
Affiliation(s)
- Paul M Zdinak
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nishtha Trivedi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephanie Grebinoski
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jessica Torrey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eduardo Zarate Martinez
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Microbiology and Immunology Diversity Scholars Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Salome Martinez
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Louise Hicks
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rashi Ranjan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Venkata Krishna Kanth Makani
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mary Melissa Roland
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lyubov Kublo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sanya Arshad
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark S Anderson
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Alok V Joglekar
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Systems Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Bass LE, Bonami RH. Factors Governing B Cell Recognition of Autoantigen and Function in Type 1 Diabetes. Antibodies (Basel) 2024; 13:27. [PMID: 38651407 PMCID: PMC11036271 DOI: 10.3390/antib13020027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Islet autoantibodies predict type 1 diabetes (T1D) but can be transient in murine and human T1D and are not thought to be directly pathogenic. Rather, these autoantibodies signal B cell activity as antigen-presenting cells (APCs) that present islet autoantigen to diabetogenic T cells to promote T1D pathogenesis. Disrupting B cell APC function prevents T1D in mouse models and has shown promise in clinical trials. Autoantigen-specific B cells thus hold potential as sophisticated T1D biomarkers and therapeutic targets. B cell receptor (BCR) somatic hypermutation is a mechanism by which B cells increase affinity for islet autoantigen. High-affinity B and T cell responses are selected in protective immune responses, but immune tolerance mechanisms are known to censor highly autoreactive clones in autoimmunity, including T1D. Thus, different selection rules often apply to autoimmune disease settings (as opposed to protective host immunity), where different autoantigen affinity ceilings are tolerated based on variations in host genetics and environment. This review will explore what is currently known regarding B cell signaling, selection, and interaction with T cells to promote T1D pathogenesis.
Collapse
Affiliation(s)
- Lindsay E. Bass
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Rachel H. Bonami
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
13
|
Honardoost MA, Adinatha A, Schmidt F, Ranjan B, Ghaeidamini M, Arul Rayan N, Gek Liang Lim M, Joanito I, Xiao Xuan Lin Q, Rajagopalan D, Qi Mok S, Hwang YY, Larbi A, Khor CC, Foo R, Boehm BO, Prabhakar S. Systematic immune cell dysregulation and molecular subtypes revealed by single-cell RNA-seq of subjects with type 1 diabetes. Genome Med 2024; 16:45. [PMID: 38539228 PMCID: PMC10976681 DOI: 10.1186/s13073-024-01300-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 01/30/2024] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a prototypic endocrine autoimmune disease resulting from an immune-mediated destruction of pancreatic insulin-secreting β cells. A comprehensive immune cell phenotype evaluation in T1DM has not been performed thus far at the single-cell level. METHODS In this cross-sectional analysis, we generated a single-cell transcriptomic dataset of peripheral blood mononuclear cells (PBMCs) from 46 manifest T1DM (stage 3) cases and 31 matched controls. RESULTS We surprisingly detected profound alterations in circulatory immune cells (1784 dysregulated genes in 13 immune cell types), far exceeding the count in the comparator systemic autoimmune disease SLE. Genes upregulated in T1DM were involved in WNT signaling, interferon signaling and migration of T/NK cells, antigen presentation by B cells, and monocyte activation. A significant fraction of these differentially expressed genes were also altered in T1DM pancreatic islets. We used the single-cell data to construct a T1DM metagene z-score (TMZ score) that distinguished cases and controls and classified patients into molecular subtypes. This score correlated with known prognostic immune markers of T1DM, as well as with drug response in clinical trials. CONCLUSIONS Our study reveals a surprisingly strong systemic dimension at the level of immune cell network in T1DM, defines disease-relevant molecular subtypes, and has the potential to guide non-invasive test development and patient stratification.
Collapse
Affiliation(s)
- Mohammad Amin Honardoost
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
- Cardiovascular Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Andreas Adinatha
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Florian Schmidt
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Bobby Ranjan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Maryam Ghaeidamini
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Nirmala Arul Rayan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Michelle Gek Liang Lim
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Ignasius Joanito
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Quy Xiao Xuan Lin
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Deepa Rajagopalan
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Shi Qi Mok
- Integrated genomics platform, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, 138648, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, 138648, Singapore
| | - Chiea Chuen Khor
- Integrated genomics platform, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore
| | - Roger Foo
- Cardiovascular Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, 138673, Singapore
| | - Bernhard Otto Boehm
- Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Faculty of Life Sciences and Medicine, King's College London, London, WC2R 2LS, UK.
| | - Shyam Prabhakar
- Laboratory of Systems Biology and Data Analytics, Genome Institute of Singapore (GIS), A*STAR (Agency for Science, Technology and Research), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Republic of Singapore.
| |
Collapse
|
14
|
Firdessa Fite R, Bechi Genzano C, Mallone R, Creusot RJ. Epitope-based precision immunotherapy of Type 1 diabetes. Hum Vaccin Immunother 2023; 19:2154098. [PMID: 36656048 PMCID: PMC9980607 DOI: 10.1080/21645515.2022.2154098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Antigen-specific immunotherapies (ASITs) address important clinical needs in treating autoimmune diseases. However, Type 1 diabetes is a heterogeneous disease wherein patient characteristics influence responsiveness to ASITs. Targeting not only disease-relevant T cell populations, but also specific groups of patients using precision medicine is a new goal toward achieving effective treatment. HLA-restricted peptides provide advantages over protein as antigens, however, methods for profiling antigen-specific T cells need to improve in sensitivity, depth, and throughput to facilitate epitope selection. Delivery approaches are highly diverse, illustrating the many ways relevant antigen-presenting cell populations and anatomical locations can be targeted for tolerance induction. The role of persistence of antigen presentation in promoting durable antigen-specific tolerance requires further investigation. Based on the outcome of ASIT trials, the field is moving toward using patient-specific variations to improve efficacy, but challenges still lie on the path to delivering more effective and safer treatment to the T1D patient population.
Collapse
Affiliation(s)
- Rebuma Firdessa Fite
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Camillo Bechi Genzano
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France.,Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Hôpitaux Universitaires de Paris Centre-Université de Paris, Paris, France
| | - Remi J Creusot
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
15
|
Ettinger RA, Buitinga M, Vandamme C, Afonso G, Gomez R, Arribas-Layton D, Bissenova S, Speake C, Reijonen H, Kinnunen T, Overbergh L, Mallone R, Kwok WW, James EA. Technical Validation and Utility of an HLA Class II Tetramer Assay for Type 1 Diabetes: A Multicenter Study. J Clin Endocrinol Metab 2023; 109:183-196. [PMID: 37474341 DOI: 10.1210/clinem/dgad434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/28/2023] [Accepted: 06/17/2023] [Indexed: 07/22/2023]
Abstract
CONTEXT Validated assays to measure autoantigen-specific T-cell frequency and phenotypes are needed for assessing the risk of developing diabetes, monitoring disease progression, evaluating responses to treatment, and personalizing antigen-based therapies. OBJECTIVE Toward this end, we performed a technical validation of a tetramer assay for HLA-DRA-DRB1*04:01, a class II allele that is strongly associated with susceptibility to type 1 diabetes (T1D). METHODS HLA-DRA-DRB1*04:01-restricted T cells specific for immunodominant epitopes from islet cell antigens GAD65, IGRP, preproinsulin, and ZnT8, and a reference influenza epitope, were enumerated and phenotyped in a single staining tube with a tetramer assay. Single and multicenter testing was performed, using a clone-spiked specimen and replicate samples from T1D patients, with a target coefficient of variation (CV) less than 30%. The same assay was applied to an exploratory cross-sectional sample set with 24 T1D patients to evaluate the utility of the assay. RESULTS Influenza-specific T-cell measurements had mean CVs of 6% for the clone-spiked specimen and 11% for T1D samples in single-center testing, and 20% and 31%, respectively, for multicenter testing. Islet-specific T-cell measurements in these same samples had mean CVs of 14% and 23% for single-center and 23% and 41% for multicenter testing. The cross-sectional study identified relationships between T-cell frequencies and phenotype and disease duration, sex, and autoantibodies. A large fraction of the islet-specific T cells exhibited a naive phenotype. CONCLUSION Our results demonstrate that the assay is reproducible and useful to characterize islet-specific T cells and identify correlations between T-cell measures and clinical traits.
Collapse
Affiliation(s)
- Ruth A Ettinger
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Mijke Buitinga
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Céline Vandamme
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Georgia Afonso
- Diabetes and Autoimmunity Research Laboratory, Université Paris Cité, Institut Cochin, CNRS, INSERM, 75014 Paris, France
| | - Rebecca Gomez
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - David Arribas-Layton
- Department of Immunology and Theranostics, City of Hope Medical Center, Beckman Research Institute, Duarte, CA 91010, USA
| | - Samal Bissenova
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Helena Reijonen
- Department of Immunology and Theranostics, City of Hope Medical Center, Beckman Research Institute, Duarte, CA 91010, USA
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
- Eastern Finland Laboratory Centre (ISLAB), 70210 Kuopio, Finland
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Roberto Mallone
- Diabetes and Autoimmunity Research Laboratory, Université Paris Cité, Institut Cochin, CNRS, INSERM, 75014 Paris, France
- Department of Internal Medicine, Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, 75014 Paris, France
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| |
Collapse
|
16
|
Racine JJ, Misherghi A, Dwyer JR, Maser R, Forte E, Bedard O, Sattler S, Pugliese A, Landry L, Elso C, Nakayama M, Mannering S, Rosenthal N, Serreze DV. HLA-DQ8 Supports Development of Insulitis Mediated by Insulin-Reactive Human TCR-Transgenic T Cells in Nonobese Diabetic Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1792-1805. [PMID: 37877672 PMCID: PMC10939972 DOI: 10.4049/jimmunol.2300303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/28/2023] [Indexed: 10/26/2023]
Abstract
In an effort to improve HLA-"humanized" mouse models for type 1 diabetes (T1D) therapy development, we previously generated directly in the NOD strain CRISPR/Cas9-mediated deletions of various combinations of murine MHC genes. These new models improved upon previously available platforms by retaining β2-microglobulin functionality in FcRn and nonclassical MHC class I formation. As proof of concept, we generated H2-Db/H2-Kd double knockout NOD mice expressing human HLA-A*0201 or HLA-B*3906 class I variants that both supported autoreactive diabetogenic CD8+ T cell responses. In this follow-up work, we now describe the creation of 10 new NOD-based mouse models expressing various combinations of HLA genes with and without chimeric transgenic human TCRs reactive to proinsulin/insulin. The new TCR-transgenic models develop differing levels of insulitis mediated by HLA-DQ8-restricted insulin-reactive T cells. Additionally, these transgenic T cells can transfer insulitis to newly developed NSG mice lacking classical murine MHC molecules, but expressing HLA-DQ8. These new models can be used to test potential therapeutics for a possible capacity to reduce islet infiltration or change the phenotype of T cells expressing type 1 diabetes patient-derived β cell autoantigen-specific TCRs.
Collapse
Affiliation(s)
| | - Adel Misherghi
- The Jackson Laboratory, Bar Harbor, ME
- College of the Atlantic, Bar Harbor, ME
| | | | | | | | | | - Susanne Sattler
- Imperial College London, London, United Kingdom
- Medical University Graz, Graz, Austria
| | - Alberto Pugliese
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL
| | - Laurie Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Colleen Elso
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Stuart Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME
- Imperial College London, London, United Kingdom
| | | |
Collapse
|
17
|
Mitchell AM, Baschal EE, McDaniel KA, Fleury T, Choi H, Pyle L, Yu L, Rewers MJ, Nakayama M, Michels AW. Tracking DNA-based antigen-specific T cell receptors during progression to type 1 diabetes. SCIENCE ADVANCES 2023; 9:eadj6975. [PMID: 38064552 PMCID: PMC10708189 DOI: 10.1126/sciadv.adj6975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023]
Abstract
T cells targeting self-proteins are important mediators in autoimmune diseases. T cells express unique cell-surface receptors (TCRs) that recognize peptides presented by major histocompatibility molecules. TCRs have been identified from blood and pancreatic islets of individuals with type 1 diabetes (T1D). Here, we tracked ~1700 known antigen-specific TCR sequences, islet antigen or viral reactive, in bulk TCRβ sequencing from longitudinal blood DNA samples in at-risk cases who progressed to T1D, age/sex/human leukocyte antigen-matched controls, and a new-onset T1D cohort. Shared and frequent antigen-specific TCRβ sequences were identified in all three cohorts, and viral sequences were present across all ages. Islet sequences had different patterns of accumulation based upon antigen specificity in the at-risk cases. Furthermore, 73 islet-antigen TCRβ sequences were present in higher frequencies and numbers in T1D samples relative to controls. The total number of these disease-associated TCRβ sequences inversely correlated with age at clinical diagnosis, indicating the potential to use disease-relevant TCR sequences as biomarkers in autoimmune disorders.
Collapse
Affiliation(s)
- Angela M. Mitchell
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin E. Baschal
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kristen A. McDaniel
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Theodore Fleury
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hyelin Choi
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Laura Pyle
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, CO, USA
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marian J. Rewers
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Maki Nakayama
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aaron W. Michels
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
18
|
Drakul M, Čolić M. Immunomodulatory activity of dipeptidyl peptidase-4 inhibitors in immune-related diseases. Eur J Immunol 2023; 53:e2250302. [PMID: 37732495 DOI: 10.1002/eji.202250302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/22/2023] [Accepted: 09/20/2023] [Indexed: 09/22/2023]
Abstract
Dipeptidyl peptidase-4 (DPP-4), also known as CD26, is a 110-kDa cell surface glycoprotein with enzymatic and signal transducing activity. DPP-4/CD26 is expressed by various cells, including CD4+ and CD8+ T cells, B cells, dendritic cells, macrophages, and NK cells. DPP-4 inhibitors (DPP-4i) were introduced to clinics in 2006 as new oral antihyperglycemic drugs approved for type 2 diabetes mellitus treatment. In addition to glucose-lowering effects, emerging data, from clinical studies and their animal models, suggest that DPP-4i could display anti-inflammatory and immunomodulatory effects as well, but the molecular and immunological mechanisms of these actions are insufficiently investigated. This review focuses on the modulatory activity of DPP-4i in the immune system and the possible application of DPP-4i in other immune-related diseases in patients with or without diabetes.
Collapse
Affiliation(s)
- Marija Drakul
- Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina
| | - Miodrag Čolić
- Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
19
|
James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023; 78:101809. [PMID: 37734713 PMCID: PMC10622886 DOI: 10.1016/j.molmet.2023.101809] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND T1D is an autoimmune disease in which pancreatic islets of Langerhans are infiltrated by immune cells resulting in the specific destruction of insulin-producing islet beta cells. Our understanding of the factors leading to islet infiltration and the interplay of the immune cells with target beta cells is incomplete, especially in human disease. While murine models of T1D have provided crucial information for both beta cell and autoimmune cell function, the translation of successful therapies in the murine model to human disease has been a challenge. SCOPE OF REVIEW Here, we discuss current state of the art and consider knowledge gaps concerning the interface of the islet beta cell with immune infiltrates, with a focus on T cells. We discuss pancreatic and immune cell phenotypes and their impact on cell function in health and disease, which we deem important to investigate further to attain a more comprehensive understanding of human T1D disease etiology. MAJOR CONCLUSIONS The last years have seen accelerated development of approaches that allow comprehensive study of human T1D. Critically, recent studies have contributed to our revised understanding that the pancreatic beta cell assumes an active role, rather than a passive position, during autoimmune disease progression. The T cell-beta cell interface is a critical axis that dictates beta cell fate and shapes autoimmune responses. This includes the state of the beta cell after processing internal and external cues (e.g., stress, inflammation, genetic risk) that that contributes to the breaking of tolerance by hyperexpression of human leukocyte antigen (HLA) class I with presentation of native and neoepitopes and secretion of chemotactic factors to attract immune cells. We anticipate that emerging insights about the molecular and cellular aspects of disease initiation and progression processes will catalyze the development of novel and innovative intervention points to provide additional therapies to individuals affected by T1D.
Collapse
Affiliation(s)
- Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Alok V Joglekar
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amelia K Linnemann
- Center for Diabetes and Metabolic Diseases, and Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sally C Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
20
|
Riaz F, Wei P, Pan F. PPARs at the crossroads of T cell differentiation and type 1 diabetes. Front Immunol 2023; 14:1292238. [PMID: 37928539 PMCID: PMC10623333 DOI: 10.3389/fimmu.2023.1292238] [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: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
T-cell-mediated autoimmune type 1 diabetes (T1D) is characterized by the immune-mediated destruction of pancreatic beta cells (β-cells). The increasing prevalence of T1D poses significant challenges to the healthcare system, particularly in countries with struggling economies. This review paper highlights the multifaceted roles of Peroxisome Proliferator-Activated Receptors (PPARs) in the context of T1D, shedding light on their potential as regulators of immune responses and β-cell biology. Recent research has elucidated the intricate interplay between CD4+ T cell subsets, such as Tregs and Th17, in developing autoimmune diseases like T1D. Th17 cells drive inflammation, while Tregs exert immunosuppressive functions, highlighting the delicate balance crucial for immune homeostasis. Immunotherapy has shown promise in reinstating self-tolerance and restricting the destruction of autoimmune responses, but further investigations are required to refine these therapeutic strategies. Intriguingly, PPARs, initially recognized for their role in lipid metabolism, have emerged as potent modulators of inflammation in autoimmune diseases, particularly in T1D. Although evidence suggests that PPARs affect the β-cell function, their influence on T-cell responses and their potential impact on T1D remains largely unexplored. It was noted that PPARα is involved in restricting the transcription of IL17A and enhancing the expression of Foxp3 by minimizing its proteasomal degradation. Thus, antagonizing PPARs may exert beneficial effects in regulating the differentiation of CD4+ T cells and preventing T1D. Therefore, this review advocates for comprehensive investigations to delineate the precise roles of PPARs in T1D pathogenesis, offering innovative therapeutic avenues that target both the immune system and pancreatic function. This review paper seeks to bridge the knowledge gap between PPARs, immune responses, and T1D, providing insights that may revolutionize the treatment landscape for this autoimmune disorder. Moreover, further studies involving PPAR agonists in non-obese diabetic (NOD) mice hold promise for developing novel T1D therapies.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Ping Wei
- Department of Otolaryngology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
21
|
Hanna SJ, Thayer TC, Robinson EJS, Vinh NN, Williams N, Landry LG, Andrews R, Siah QZ, Leete P, Wyatt R, McAteer MA, Nakayama M, Wong FS, Yang JHM, Tree TIM, Ludvigsson J, Dayan CM, Tatovic D. Single-cell RNAseq identifies clonally expanded antigen-specific T-cells following intradermal injection of gold nanoparticles loaded with diabetes autoantigen in humans. Front Immunol 2023; 14:1276255. [PMID: 37908349 PMCID: PMC10613693 DOI: 10.3389/fimmu.2023.1276255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Gold nanoparticles (GNPs) have been used in the development of novel therapies as a way of delivery of both stimulatory and tolerogenic peptide cargoes. Here we report that intradermal injection of GNPs loaded with the proinsulin peptide C19-A3, in patients with type 1 diabetes, results in recruitment and retention of immune cells in the skin. These include large numbers of clonally expanded T-cells sharing the same paired T-cell receptors (TCRs) with activated phenotypes, half of which, when the TCRs were re-expressed in a cell-based system, were confirmed to be specific for either GNP or proinsulin. All the identified gold-specific clones were CD8+, whilst proinsulin-specific clones were both CD8+ and CD4+. Proinsulin-specific CD8+ clones had a distinctive cytotoxic phenotype with overexpression of granulysin (GNLY) and KIR receptors. Clonally expanded antigen-specific T cells remained in situ for months to years, with a spectrum of tissue resident memory and effector memory phenotypes. As the T-cell response is divided between targeting the gold core and the antigenic cargo, this offers a route to improving resident memory T-cells formation in response to vaccines. In addition, our scRNAseq data indicate that focusing on clonally expanded skin infiltrating T-cells recruited to intradermally injected antigen is a highly efficient method to enrich and identify antigen-specific cells. This approach has the potential to be used to monitor the intradermal delivery of antigens and nanoparticles for immune modulation in humans.
Collapse
Affiliation(s)
- Stephanie J. Hanna
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Terri C. Thayer
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Department of Biological and Chemical Sciences, Roberts Wesleyan University, Rochester, NY, United States
| | - Emma J. S. Robinson
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ngoc-Nga Vinh
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
| | - Nigel Williams
- Division of Psychological Medicine and Clinical Neurosciences, Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
| | - Laurie G. Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Denver, CO, United States
| | - Robert Andrews
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Qi Zhuang Siah
- John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Pia Leete
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, United Kingdom
| | - Rebecca Wyatt
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, United Kingdom
| | | | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Denver, CO, United States
| | - F. Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jennie H. M. Yang
- Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Timothy I. M. Tree
- Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences and Crown Princess Victoria Children´s Hospital, Linköping University, Linköping, Sweden
| | - Colin M. Dayan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Danijela Tatovic
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
22
|
Merzah MH, Diajil AR. Serum and salivary adiponectin levels as predictive markers for diabetes mellitus in children with a family history of diabetes. J Med Life 2023; 16:1561-1565. [PMID: 38313182 PMCID: PMC10835556 DOI: 10.25122/jml-2023-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 02/06/2024] Open
Abstract
Diabetes mellitus (DM) is a chronic, metabolic condition marked by defects in insulin production, action, or both. Environmental and genetic factors can contribute to the onset of diabetes mellitus. Adiponectin, a hormone affecting pancreatic beta cell proliferation, has emerged as a potential indicator of diabetes risk. This cross-sectional study aimed to evaluate serum and salivary adiponectin levels as predictors of diabetes mellitus in children with/without a family history of diabetes mellitus. The study was conducted at Al-Zahra Hospital in Najaf city and included 125 children aged 5 to 16. Data on demographics, including name, age, and gender, were collected, and body mass index (BMI) was assessed. Serum and salivary adiponectin levels were measured and analyzed in relation to family history and BMI. Children with a family history of DM had high serum adiponectin (ADP) levels. Serum adiponectin levels were significantly higher in children with first-degree relatives having a history of diabetes mellitus, except for cases involving mothers and other relatives with diabetes mellitus history (p<0.05). Furthermore, serum adiponectin levels were higher in obese children. Salivary adiponectin levels were significantly elevated in children with a maternal family history of diabetes (p=0.01), while no significant correlation was found with BMI. A significant negative correlation (r=-0.180, p=0.05) between salivary and serum adiponectin concentrations was observed. Compared to children with a normal, healthy weight, children with obesity had decreased salivary adiponectin levels and increased serum adiponectin levels.
Collapse
Affiliation(s)
- Maryam Hamid Merzah
- Department of Oral Diagnosis, Oral Medicine, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Ameena Ryhan Diajil
- Department of Oral Diagnosis, Oral Medicine, College of Dentistry, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
23
|
Siddiqui K, Nawaz SS. Exploration of Immune Targets for Type 1 Diabetes and Latent Autoimmune Disease Immunotherapy. Immunotargets Ther 2023; 12:91-103. [PMID: 37795196 PMCID: PMC10546931 DOI: 10.2147/itt.s417917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/09/2023] [Indexed: 10/06/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that destroys pancreatic beta cells, which produce insulin in the islets of Langerhans. The risk of developing T1D is influenced by environmental factors, genetics, and autoantibodies. Latent autoimmune diabetes in adults (LADA) is a type of T1D that is genetically and phenotypically distinct from classic T1D. This review summarizes the accumulated information on the risk factors for T1D and LADA, and immunotherapy trials that offer insights into potential future combined therapeutic interventions for both T1D and LADA to slow the rate of islet cell loss and preserve beta cell function. Future research should also focus on improving intervention doses, conducting more thorough examinations of intervention responders, and/or combining minimally effective single-target immunotherapies to slow the rate of islet cell loss and preserve beta cell function.
Collapse
Affiliation(s)
- Khalid Siddiqui
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Shaik Sarfaraz Nawaz
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
24
|
Atkinson MA, Mirmira RG. The pathogenic "symphony" in type 1 diabetes: A disorder of the immune system, β cells, and exocrine pancreas. Cell Metab 2023; 35:1500-1518. [PMID: 37478842 PMCID: PMC10529265 DOI: 10.1016/j.cmet.2023.06.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023]
Abstract
Type 1 diabetes (T1D) is widely considered to result from the autoimmune destruction of insulin-producing β cells. This concept has been a central tenet for decades of attempts seeking to decipher the disorder's pathogenesis and prevent/reverse the disease. Recently, this and many other disease-related notions have come under increasing question, particularly given knowledge gained from analyses of human T1D pancreas. Perhaps most crucial are findings suggesting that a collective of cellular constituents-immune, endocrine, and exocrine in origin-mechanistically coalesce to facilitate T1D. This review considers these emerging concepts, from basic science to clinical research, and identifies several key remaining knowledge voids.
Collapse
Affiliation(s)
- Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Raghavendra G Mirmira
- Departments of Medicine and Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
25
|
Kattner N. Immune cell infiltration in the pancreas of type 1, type 2 and type 3c diabetes. Ther Adv Endocrinol Metab 2023; 14:20420188231185958. [PMID: 37529508 PMCID: PMC10387691 DOI: 10.1177/20420188231185958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/16/2023] [Indexed: 08/03/2023] Open
Abstract
The different types of diabetes differ in disease pathogenesis but share the impairment or loss of β-cell function leading to chronic hyperglycaemia. While immune cells are present throughout the whole pancreas in normality, their number and activation is increased in diabetes. Different patterns and composition of inflammation could be observed in type 1, type 2 and type 3c diabetes. Immune cells, pancreatic stellate cells and fibrosis were present in the islet microenvironment and could add to β-cell dysfunction and therefore development and progression of diabetes. First studies investigating the use of anti-inflammatory drugs demonstrate their ability to rescue remaining β-cell function and their potential benefit in diabetes treatment. This article provides an overview of immune cell infiltrates in different types of diabetes, highlights the knowledge of their impact on β-cell function and introduces the potential of immunomodulatory strategies.
Collapse
Affiliation(s)
- Nicole Kattner
- Translational and Clinical Research Institute, Newcastle University, Medical School, Framlington Place, Newcastle upon Tyne, UK
| |
Collapse
|
26
|
Nicholas CA, Smith MJ. Application of single-cell RNA sequencing methods to develop B cell targeted treatments for autoimmunity. Front Immunol 2023; 14:1103690. [PMID: 37520578 PMCID: PMC10382068 DOI: 10.3389/fimmu.2023.1103690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 06/29/2023] [Indexed: 08/01/2023] Open
Abstract
The COVID-19 pandemic coincided with several transformative advances in single-cell analysis. These new methods along with decades of research and trials with antibody therapeutics and RNA based technologies allowed for highly effective vaccines and treatments to be produced at astonishing speeds. While these tools were initially focused on models of infection, they also show promise in an autoimmune setting. Self-reactive B cells play important roles as antigen-presenting cells and cytokine and autoantibody producers for many autoimmune diseases. Yet, current therapies to target autoreactive B cells deplete all B cells irrespective of their pathogenicity. Development of self-reactive B cell targeting therapies that would spare non-pathogenic B cells are needed to treat disease while allowing effective immune responses to other ailments. Single-cell RNA sequencing (scRNA-seq) approaches will aid in identification of the pathogenic self-reactive B cells operative in autoimmunity and help with development of more favorable precision targeted therapies.
Collapse
Affiliation(s)
- Catherine A. Nicholas
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Mia J. Smith
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
27
|
McNitt DH, Joosse BA, Thomas JW, Bonami RH. Productive Germinal Center Responses Depend on the Nature of Stimuli Received by Anti-Insulin B Cells in Type 1 Diabetes-Prone Mice. Immunohorizons 2023; 7:384-397. [PMID: 37261716 PMCID: PMC10448785 DOI: 10.4049/immunohorizons.2300036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023] Open
Abstract
Islet autoantibodies, including those directed at insulin, predict type 1 diabetes (T1D) in mice and humans and signal immune tolerance breach by B lymphocytes. High-affinity insulin autoantibodies and T follicular helper cell involvement implicate germinal centers (GCs) in T1D. The VH125SD BCR transgenic model, in which 1-2% of peripheral B lymphocytes recognize insulin, enables direct study of insulin-binding B cells. Our prior studies showed that anti-insulin B cell receptor transgene site-directed to H chain locus mice fail to generate insulin Ab following T-dependent immunization, but it was unclear whether anti-insulin B cells were blocked for GC initiation, survival, or differentiation into Ab-secreting cells. Here, we show that insulin-binding B cells in T1D-prone anti-insulin B cell receptor transgene site-directed to H chain locus mice can spontaneously adopt a GC phenotype and undergo class switching to the IgG1 isotype, with little if any switching to IgG2b. T-dependent immunizations with insulin SRBC or insulin CFA drove anti-insulin B lymphocytes to adopt a GC phenotype, despite blunted insulin Ab production. Dual immunization against self (insulin) and foreign (4-hydroxy-3-nitrophenylacetyl hapten conjugated to keyhole limpet hemocyanin) Ags showed an anti-insulin (but not anti-4-hydroxy-3-nitrophenylacetyl) Ab block that tracked with increased expression of the apoptosis marker, activated caspase 3, in self-reactive GC B cells. Finally, T-independent immunization with insulin conjugated to Brucella abortus ring test Ag released immune tolerance to allow robust expansion of anti-insulin GC B cells and IgG-switched insulin Ab production. Overall, these data pinpoint GC survival and Ab-secreting cell differentiation as immune tolerance blocks that limit T-dependent, but not T-independent, stimulation of anti-insulin B cell responses.
Collapse
Affiliation(s)
- Dudley H. McNitt
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Bryan A. Joosse
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - James W. Thomas
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of
Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and
Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
28
|
Pieper T, Roth KDR, Glaser V, Riet T, Buitrago-Molina LE, Hagedorn M, Lieber M, Hust M, Noyan F, Jaeckel E, Hardtke-Wolenski M. Generation of Chimeric Antigen Receptors against Tetraspanin 7. Cells 2023; 12:1453. [PMID: 37296574 PMCID: PMC10252682 DOI: 10.3390/cells12111453] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Adoptive transfer of antigen-specific regulatory T cells (Tregs) has shown promising results in the treatment of autoimmune diseases; however, the use of polyspecific Tregs has limited effects. However, obtaining a sufficient number of antigen-specific Tregs from patients with autoimmune disorders remains challenging. Chimeric antigen receptors (CARs) provide an alternative source of T cells for novel immunotherapies that redirect T cells independently of the MHC. In this study, we aimed to generate antibody-like single-chain variable fragments (scFv) and subsequent CARs against tetraspanin 7 (TSPAN7), a membrane protein highly expressed on the surface of pancreatic beta cells, using phage display technology. We established two methods for generating scFvs against TSPAN7 and other target structures. Moreover, we established novel assays to analyze and quantify their binding abilities. The resulting CARs were functional and activated specifically by the target structure, but could not recognize TSPAN7 on the surface of beta cells. Despite this, this study demonstrates that CAR technology is a powerful tool for generating antigen-specific T cells and provides new approaches for generating functional CARs.
Collapse
Affiliation(s)
- Tom Pieper
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Kristian Daniel Ralph Roth
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Medizinische Biotechnologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Viktor Glaser
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Riet
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department I of Internal Medicine, Tumor Genetics, University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50933 Cologne, Germany
| | - Laura Elisa Buitrago-Molina
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Maike Hagedorn
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Maren Lieber
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Medizinische Biotechnologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department of Liver Transplantation, Multi Organ Transplant Program, University Health Network, University of Toronto, Toronto, ON M5T 0S8, Canada
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 47057 Essen, Germany
| |
Collapse
|
29
|
van Tienhoven R, Kracht MJL, van der Slik AR, Thomaidou S, Wolters AHG, Giepmans BNG, Riojas JPR, Nelson MS, Carlotti F, de Koning EJP, Hoeben RC, Zaldumbide A, Roep BO. Presence of immunogenic alternatively spliced insulin gene product in human pancreatic delta cells. Diabetologia 2023; 66:884-896. [PMID: 36884057 PMCID: PMC10036285 DOI: 10.1007/s00125-023-05882-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/23/2022] [Indexed: 03/09/2023]
Abstract
AIMS/HYPOTHESIS Transcriptome analyses revealed insulin-gene-derived transcripts in non-beta endocrine islet cells. We studied alternative splicing of human INS mRNA in pancreatic islets. METHODS Alternative splicing of insulin pre-mRNA was determined by PCR analysis performed on human islet RNA and single-cell RNA-seq analysis. Antisera were generated to detect insulin variants in human pancreatic tissue using immunohistochemistry, electron microscopy and single-cell western blot to confirm the expression of insulin variants. Cytotoxic T lymphocyte (CTL) activation was determined by MIP-1β release. RESULTS We identified an alternatively spliced INS product. This variant encodes the complete insulin signal peptide and B chain and an alternative C-terminus that largely overlaps with a previously identified defective ribosomal product of INS. Immunohistochemical analysis revealed that the translation product of this INS-derived splice transcript was detectable in somatostatin-producing delta cells but not in beta cells; this was confirmed by light and electron microscopy. Expression of this alternatively spliced INS product activated preproinsulin-specific CTLs in vitro. The exclusive presence of this alternatively spliced INS product in delta cells may be explained by its clearance from beta cells by insulin-degrading enzyme capturing its insulin B chain fragment and a lack of insulin-degrading enzyme expression in delta cells. CONCLUSIONS/INTERPRETATION Our data demonstrate that delta cells can express an INS product derived from alternative splicing, containing both the diabetogenic insulin signal peptide and B chain, in their secretory granules. We propose that this alternative INS product may play a role in islet autoimmunity and pathology, as well as endocrine or paracrine function or islet development and endocrine destiny, and transdifferentiation between endocrine cells. INS promoter activity is not confined to beta cells and should be used with care when assigning beta cell identity and selectivity. DATA AVAILABILITY The full EM dataset is available via www.nanotomy.org (for review: http://www.nanotomy.org/OA/Tienhoven2021SUB/6126-368/ ). Single-cell RNA-seq data was made available by Segerstolpe et al [13] and can be found at https://sandberglab.se/pancreas . The RNA and protein sequence of INS-splice was uploaded to GenBank (BankIt2546444 INS-splice OM489474).
Collapse
Affiliation(s)
- René van Tienhoven
- Department of Diabetes and Cancer Discovery Science, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Maria J L Kracht
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arno R van der Slik
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sofia Thomaidou
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anouk H G Wolters
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ben N G Giepmans
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Michael S Nelson
- Light Microscopy Core, City of Hope National Medical Center, Duarte, CA, USA
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob C Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart O Roep
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
30
|
Tippalagama R, Chihab LY, Kearns K, Lewis S, Panda S, Willemsen L, Burel JG, Lindestam Arlehamn CS. Antigen-specificity measurements are the key to understanding T cell responses. Front Immunol 2023; 14:1127470. [PMID: 37122719 PMCID: PMC10140422 DOI: 10.3389/fimmu.2023.1127470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Antigen-specific T cells play a central role in the adaptive immune response and come in a wide range of phenotypes. T cell receptors (TCRs) mediate the antigen-specificities found in T cells. Importantly, high-throughput TCR sequencing provides a fingerprint which allows tracking of specific T cells and their clonal expansion in response to particular antigens. As a result, many studies have leveraged TCR sequencing in an attempt to elucidate the role of antigen-specific T cells in various contexts. Here, we discuss the published approaches to studying antigen-specific T cells and their specific TCR repertoire. Further, we discuss how these methods have been applied to study the TCR repertoire in various diseases in order to characterize the antigen-specific T cells involved in the immune control of disease.
Collapse
|
31
|
Wang F, Liang J, Zhu D, Xiang P, Zhou L, Yang C. Characteristic gene prognostic model of type 1 diabetes mellitus via machine learning strategy. Endocr J 2023; 70:281-294. [PMID: 36477008 DOI: 10.1507/endocrj.ej22-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The present study was designed to detect possible biomarkers associated with Type 1 diabetes mellitus (T1DM) incidence in an effort to develop novel treatments for this condition. Three mRNA expression datasets of peripheral blood mononuclear cells (PBMCs) were obtained from the GEO database. Differentially expressed genes (DEGs) between T1DM patients and healthy controls were identified by Limma package in R, and using the DEGs to conduct GO and DO pathway enrichment. The LASSO-SVM were used to screen the hub genes. We performed immune correlation analysis of hub genes and established a T1DM prognosis model. CIBERSORT algorithm was used to identify the different immune cells in distribution between T1DM and normal samples. The correlation of the hub genes and immune cells was analyzed by Spearman. ROC curves were used to assess the diagnostic value of genes in T1DM. A total of 60 immune related DEGs were obtained from the T1DM and normal samples. Then, DEGs were further screened to obtain 3 hub genes, ANP32A-IT1, ESCO2 and NBPF1. CIBERSORT analysis revealed the percentage of immune cells in each sample, indicating that there was significant difference in monocytes, T cells CD8+, gamma delta T cells, naive CD4+ T cells and activated memory CD4+ T cells between T1DM and normal samples. The area under curve (AUC) of ESCO2, ANP32A-IT1 and NBPF1 were all greater than 0.8, indicating that these three genes have high diagnostic value for T1DM. Together, the findings of these bioinformatics analyses thus identified key hub genes associated with T1DM development.
Collapse
Affiliation(s)
- Fenglin Wang
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
- Hebei North University, Zhangjiakou 075000, China
| | - Jiemei Liang
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
- Hebei North University, Zhangjiakou 075000, China
| | - Di Zhu
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
| | - Pengan Xiang
- Hospital of 94498 Troops, People's Liberation Army, Nanyang 474300, China
| | - Luyao Zhou
- Hebei North University, Zhangjiakou 075000, China
| | - Caizhe Yang
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
| |
Collapse
|
32
|
Garg R, Agarwal A, Katekar R, Dadge S, Yadav S, Gayen JR. Chromogranin A-derived peptides pancreastatin and catestatin: emerging therapeutic target for diabetes. Amino Acids 2023:10.1007/s00726-023-03252-x. [PMID: 36914766 DOI: 10.1007/s00726-023-03252-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/16/2023] [Indexed: 03/16/2023]
Abstract
Chromogranin A (ChgA) is an acidic pro-protein found in neuroendocrine organs, pheochromocytoma chromaffin granules, and tumor cells. Proteolytic processing of ChgA gives rise to an array of biologically active peptides such as pancreastatin (PST), vasostatin, WE14, catestatin (CST), and serpinin, which have diverse roles in regulating cardiovascular functions and metabolism, as well as inflammation. Intricate tissue-specific role of ChgA-derived peptide activity in preclinical rodent models of metabolic syndrome reveals complex effects on carbohydrate and lipid metabolism. Indeed, ChgA-derived peptides, PST and CST, play a pivotal role in metabolic syndrome such as obesity, insulin resistance, and diabetes mellitus. Additionally, supplementation of specific peptide in ChgA-KO mice have an opposing effect on physiological functions, such as PST supplementation reduces insulin sensitivity and enhances inflammatory response. In contrast, CST supplementation enhances insulin sensitivity and reduces inflammatory response. In this review, we focus on the tissue-specific role of PST and CST as therapeutic targets in regulating carbohydrate and lipid metabolism, along with the associated risk factors.
Collapse
Affiliation(s)
- Richa Garg
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Arun Agarwal
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Roshan Katekar
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shailesh Dadge
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shubhi Yadav
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Jiaur R Gayen
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
33
|
Ramos-Martínez E, Ramos-Martínez I, Valencia J, Ramos-Martínez JC, Hernández-Zimbrón L, Rico-Luna A, Pérez-Campos E, Pérez-Campos Mayoral L, Cerbón M. Modulatory role of prolactin in type 1 diabetes. Horm Mol Biol Clin Investig 2023; 44:79-88. [PMID: 35852366 DOI: 10.1515/hmbci-2022-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 06/30/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Patients with type 1 diabetes mellitus have been reported to have elevated prolactin levels and a possible relationship between prolactin levels and the development of the disease has been proposed. However, some studies show that prolactin mediates beneficial functions in beta cells. Therefore, we review information on the roles of prolactin in type 1 diabetes mellitus. CONTENT Here we summarize the functions of prolactin in the immune system and in pancreatic beta cells, in addition, we describe studies related to PRL levels, its regulation and alterations of secretion in patients with type 1 diabetes mellitus. SUMMARY Studies in murine models have shown that prolactin protects beta cells from apoptosis, stimulates their proliferation and promotes pancreatic islet revascularization. In addition, some studies in patients with type 1 diabetes mellitus have shown that elevated prolactin levels correlate with better disease control. OUTLOOK Prolactin treatment appears to be a promising strategy to improve beta-cell vascularization and proliferation in transplantation and immunotherapies.
Collapse
Affiliation(s)
- Edgar Ramos-Martínez
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Ivan Ramos-Martínez
- Departamento de Medicina y Zootecnia de Cerdos, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jorge Valencia
- Endocrine Research Unit, UMAE Hospital de Especialidades, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Juan Carlos Ramos-Martínez
- Cardiology Department, Hospital General Regional Lic Ignacio Garcia Tellez IMSS, Mérida, Yucatán, México
| | - Luis Hernández-Zimbrón
- Escuela Nacional de Estudios Superiores, Licenciatura en Optometría, Unidad León, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Anaiza Rico-Luna
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | | | - Laura Pérez-Campos Mayoral
- Research Centre Medicine UNAM-UABJO. Facultad de Medicina, Universidad Autónoma "Benito Juárez" de Oaxaca, Oaxaca, México
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana. Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
34
|
Guyer P, Arribas-Layton D, Manganaro A, Speake C, Lord S, Eizirik DL, Kent SC, Mallone R, James EA. Recognition of mRNA Splice Variant and Secretory Granule Epitopes by CD4+ T Cells in Type 1 Diabetes. Diabetes 2023; 72:85-96. [PMID: 36201618 PMCID: PMC9797322 DOI: 10.2337/db22-0191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 10/02/2022] [Indexed: 01/19/2023]
Abstract
A recent discovery effort resulted in identification of novel splice variant and secretory granule antigens within the HLA class I peptidome of human islets and documentation of their recognition by CD8+ T cells from peripheral blood and human islets. In the current study, we applied a systematic discovery process to identify novel CD4+ T cell epitopes derived from these candidate antigens. We predicted 145 potential epitopes spanning unique splice junctions and within conventional secretory granule antigens and measured their in vitro binding to DRB1*04:01. We generated HLA class II tetramers for the 35 peptides with detectable binding and used these to assess immunogenicity and isolate T cell clones. Tetramers corresponding to peptides with verified immunogenicity were then used to label T cells specific for these putative epitopes in peripheral blood. T cells that recognize distinct epitopes derived from a cyclin I splice variant, neuroendocrine convertase 2, and urocortin-3 were detected at frequencies that were similar to those of an immunodominant proinsulin epitope. Cells specific for these novel epitopes predominantly exhibited a Th1-like surface phenotype. Among the three epitopes, responses to the cyclin I peptide exhibited a distinct memory profile. Responses to neuroendocrine convertase 2 were detected among pancreatic infiltrating T cells. These results further establish the contribution of unconventional antigens to the loss of tolerance in autoimmune diabetes.
Collapse
Affiliation(s)
- Perrin Guyer
- Center for Translational Immunology, Benaroya Research Institute, Virginia Mason Medical Center, Seattle, WA
| | - David Arribas-Layton
- Center for Translational Immunology, Benaroya Research Institute, Virginia Mason Medical Center, Seattle, WA
| | - Anthony Manganaro
- Division of Diabetes, Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Cate Speake
- Diabetes Clinical Research Program and Center for Interventional Immunology, Benaroya Research Institute, Virginia Mason Medical Center, Seattle, WA
| | - Sandra Lord
- Diabetes Clinical Research Program and Center for Interventional Immunology, Benaroya Research Institute, Virginia Mason Medical Center, Seattle, WA
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Sally C. Kent
- Division of Diabetes, Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Roberto Mallone
- INSERM, CNRS, Institut Cochin, Université de Paris, Paris, France
- Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Hôpitaux Universitaires Paris Centre, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Eddie A. James
- Center for Translational Immunology, Benaroya Research Institute, Virginia Mason Medical Center, Seattle, WA
| |
Collapse
|
35
|
Strollo R, Vinci C, Man YKS, Bruzzaniti S, Piemonte E, Alhamar G, Briganti SI, Malandrucco I, Tramontana F, Fanali C, Garnett J, Buccafusca R, Guyer P, Mamula M, James EA, Pozzilli P, Ludvigsson J, Winyard PG, Galgani M, Nissim A. Autoantibody and T cell responses to oxidative post-translationally modified insulin neoantigenic peptides in type 1 diabetes. Diabetologia 2023; 66:132-146. [PMID: 36207582 PMCID: PMC9729141 DOI: 10.1007/s00125-022-05812-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Antibodies specific to oxidative post-translational modifications (oxPTM) of insulin (oxPTM-INS) are present in most individuals with type 1 diabetes, even before the clinical onset. However, the antigenic determinants of such response are still unknown. In this study, we investigated the antibody response to oxPTM-INS neoepitope peptides (oxPTM-INSPs) and evaluated their ability to stimulate humoral and T cell responses in type 1 diabetes. We also assessed the concordance between antibody and T cell responses to the oxPTM-INS neoantigenic peptides. METHODS oxPTM-INS was generated by exposing insulin to various reactive oxidants. The insulin fragments resulting from oxPTM were fractionated by size-exclusion chromatography further to ELISA and LC-MS/MS analysis to identify the oxidised peptide neoepitopes. Immunogenic peptide candidates were produced and then modified in house or designed to incorporate in silico-oxidised amino acids during synthesis. Autoantibodies to the oxPTM-INSPs were tested by ELISA using sera from 63 participants with new-onset type 1 diabetes and 30 control participants. An additional 18 fresh blood samples from participants with recently diagnosed type 1 diabetes, five with established disease, and from 11 control participants were used to evaluate, in parallel, CD4+ and CD8+ T cell activation by oxPTM-INSPs. RESULTS We observed antibody and T cell responses to three out of six LC-MS/MS-identified insulin peptide candidates: A:12-21 (SLYQLENYCN, native insulin peptide 3 [Nt-INSP-3]), B:11-30 (LVEALYLVCGERGFFYTPKT, Nt-INSP-4) and B:21-30 (ERGFFYTPKT, Nt-INSP-6). For Nt-INSP-4 and Nt-INSP-6, serum antibody binding was stronger in type 1 diabetes compared with healthy control participants (p≤0.02), with oxidised forms of ERGFFYTPKT, oxPTM-INSP-6 conferring the highest antibody binding (83% binders to peptide modified in house by hydroxyl radical [●OH] and >88% to in silico-oxidised peptide; p≤0.001 vs control participants). Nt-INSP-4 induced the strongest T cell stimulation in type 1 diabetes compared with control participants for both CD4+ (p<0.001) and CD8+ (p=0.049). CD4+ response to oxPTM-INSP-6 was also commoner in type 1 diabetes than in control participants (66.7% vs 27.3%; p=0.039). Among individuals with type 1 diabetes, the CD4+ response to oxPTM-INSP-6 was more frequent than to Nt-INSP-6 (66.7% vs 27.8%; p=0.045). Overall, 44.4% of patients showed a concordant autoimmune response to oxPTM-INSP involving simultaneously CD4+ and CD8+ T cells and autoantibodies. CONCLUSIONS/INTERPRETATION Our findings support the concept that oxidative stress, and neoantigenic epitopes of insulin, may be involved in the immunopathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Rocky Strollo
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Vinci
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Y K Stella Man
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sara Bruzzaniti
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Biology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Erica Piemonte
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ghadeer Alhamar
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Silvia Irina Briganti
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Ilaria Malandrucco
- The UOSD of Endocrinology and Metabolic Diseases, Azienda Sanitaria Locale (ASL) Frosinone, Frosinone, Italy
| | - Flavia Tramontana
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Fanali
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - James Garnett
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Roberto Buccafusca
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Perrin Guyer
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Mark Mamula
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Eddie A James
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Paolo Pozzilli
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children's Hospital, Linköping University, Linköping, Sweden
| | - Paul G Winyard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | - Mario Galgani
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
36
|
Foster A, Bhattacharjee P, Tresoldi E, Pakusch M, Cameron FJ, Mannering SI. Glutamine deamidation does not increase the immunogenicity of C-peptide in people with type 1 diabetes. J Transl Autoimmun 2022; 6:100180. [PMID: 36619657 PMCID: PMC9811213 DOI: 10.1016/j.jtauto.2022.100180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Type 1 diabetes (T1D) is a T-cell mediated autoimmune disease in which the insulin-producing beta cells are destroyed. While it is clear that full-length C-peptide, derived from proinsulin, is a major antigen in human T1D it is not clear how and why C-peptide becomes a target of the autoimmune CD4+ T-cell responses in T1D. Neoepitopes formed by the conversion of glutamine (Q) residues to glutamic acid (E) by deamidation are central to the immune pathogenesis of coeliac disease and have been implicated in autoimmune responses in T1D. Here, we asked if the immunogenicity of full-length C-peptide, which comprises four glutamine residues, was enhanced by deamidation, which we mimicked by substituting glutamic acid for glutamine residue. First, we used a panel of 18 well characterized CD4+ T-cell lines specific for epitopes derived from human C-peptide. In all cases, when the substitution fell within the cognate epitope the response was diminished, or in a few cases unchanged. In contrast, when the substitution fell outside the epitope recognized by the TCR responses were unchanged or slightly augmented. Second, we compared CD4+ T-cell proliferation responses, against deamidated and unmodified C-peptide, in the peripheral blood of people with or without T1D using the CFSE-based proliferation assay. While, as reported previously, responses were detected to unmodified C-peptide, no deamidated C-peptide was consistently more stimulatory than native C-peptide. Overall responses were weaker to deamidated C-peptide compared to unmodified C-peptide. Hence, we conclude that deamidated C-peptide does not play a role in beta-cell autoimmunity in people with T1D.
Collapse
Affiliation(s)
- Abby Foster
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, Victoria, 3065, Australia
| | - Pushpak Bhattacharjee
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, Victoria, 3065, Australia
| | - Eleonora Tresoldi
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, Victoria, 3065, Australia
| | - Miha Pakusch
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, Victoria, 3065, Australia
| | - Fergus J. Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Australia,Murdoch Children's Research Institute, Parkville, VIC, Australia,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, Victoria, 3065, Australia,Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria, 3065, Australia,Corresponding author. St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, Victoria, 3065, Melbourne, Australia
| |
Collapse
|
37
|
Mitchell AM, Baschal EE, McDaniel KA, Simmons KM, Pyle L, Waugh K, Steck AK, Yu L, Gottlieb PA, Rewers MJ, Nakayama M, Michels AW. Temporal development of T cell receptor repertoires during childhood in health and disease. JCI Insight 2022; 7:161885. [PMID: 35998036 PMCID: PMC9675557 DOI: 10.1172/jci.insight.161885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/17/2022] [Indexed: 12/05/2022] Open
Abstract
T cell receptor (TCR) sequences are exceptionally diverse and can now be comprehensively measured with next-generation sequencing technologies. However, a thorough investigation of longitudinal TCR repertoires throughout childhood in health and during development of a common childhood disease, type 1 diabetes (T1D), has not been undertaken. Here, we deep sequenced the TCR-β chain repertoires from longitudinal peripheral blood DNA samples at 4 time points beginning early in life (median age of 1.4 years) from children who progressed to T1D (n = 29) and age/sex-matched islet autoantibody-negative controls (n = 25). From 53 million TCR-β sequences, we show that the repertoire is extraordinarily diverse early in life and narrows with age independently of disease. We demonstrate the ability to identify specific TCR sequences, including those known to recognize influenza A and, separately, those specific for insulin and its precursor, preproinsulin. Insulin-reactive TCR-β sequences were more common and frequent in number as the disease progressed in those who developed T1D compared with genetically at risk nondiabetic children, and this was not the case for influenza-reactive sequences. As an independent validation, we sequenced and analyzed TCR-β repertoires from a cohort of new-onset T1D patients (n = 143), identifying the same preproinsulin-reactive TCRs. These results demonstrate an enrichment of preproinsulin-reactive TCR sequences during the progression to T1D, highlighting the importance of using disease-relevant TCR sequences as powerful biomarkers in autoimmune disorders.
Collapse
Affiliation(s)
- Angela M Mitchell
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Erin E Baschal
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Kristen A McDaniel
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Kimber M Simmons
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Laura Pyle
- Department of Biostatistics and Informatics, University of Colorado School of Pubic Health, Aurora, United States of America
| | - Kathleen Waugh
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Andrea K Steck
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Peter A Gottlieb
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Marian J Rewers
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Maki Nakayama
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| | - Aaron W Michels
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, United States of America
| |
Collapse
|
38
|
A gut microbial peptide and molecular mimicry in the pathogenesis of type 1 diabetes. Proc Natl Acad Sci U S A 2022; 119:e2120028119. [PMID: 35878027 PMCID: PMC9351354 DOI: 10.1073/pnas.2120028119] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of pancreatic β-cells. One of the earliest aspects of this process is the development of autoantibodies and T cells directed at an epitope in the B-chain of insulin (insB:9-23). Analysis of microbial protein sequences with homology to the insB:9-23 sequence revealed 17 peptides showing >50% identity to insB:9-23. Of these 17 peptides, the hprt4-18 peptide, found in the normal human gut commensal Parabacteroides distasonis, activated both human T cell clones from T1D patients and T cell hybridomas from nonobese diabetic (NOD) mice specific to insB:9-23. Immunization of NOD mice with P. distasonis insB:9-23 peptide mimic or insB:9-23 peptide verified immune cross-reactivity. Colonization of female NOD mice with P. distasonis accelerated the development of T1D, increasing macrophages, dendritic cells, and destructive CD8+ T cells, while decreasing FoxP3+ regulatory T cells. Western blot analysis identified P. distasonis-reacting antibodies in sera of NOD mice colonized with P. distasonis and human T1D patients. Furthermore, adoptive transfer of splenocytes from P. distasonis-treated mice to NOD/SCID mice enhanced disease phenotype in the recipients. Finally, analysis of human children gut microbiome data from a longitudinal DIABIMMUNE study revealed that seroconversion rates (i.e., the proportion of individuals developing two or more autoantibodies) were consistently higher in children whose microbiome harbored sequences capable of producing the hprt4-18 peptide compared to individuals who did not harbor it. Taken together, these data demonstrate the potential role of a gut microbiota-derived insB:9-23-mimic peptide as a molecular trigger of T1D pathogenesis.
Collapse
|
39
|
Jia X, Zhai TY, Wang B, Zhang JA, Song RH. High-throughput T cell receptor sequencing reveals differential immune repertoires in autoimmune thyroid diseases. Mol Cell Endocrinol 2022; 550:111644. [PMID: 35429598 DOI: 10.1016/j.mce.2022.111644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 01/16/2023]
Abstract
BACKGROUND Autoimmune thyroid diseases (AITDs) are chronic autoimmune diseases specific to thyroid and mainly include Graves' disease (GD) and Hashimoto' thyroiditis (HT). The adaptive immunoreactivity of CD4+ T cells plays a crucial role in the pathogenesis of AITDs, but very little has been known about its changes in disease status. METHODS We collected peripheral CD4+ T cells from 12 GD patients, including 6 newly diagnosed GD (NGD) and 6 refractory GD (RGD) patients, 6 HT patients and 6 healthy controls, and examined the gene expression profiles and colon types of T cells receptor (TCR) β chain complementarity determining region 3 (CDR3) using high-throughput sequencing. RESULTS The TCR repertoire were significantly expanded in AITDs groups, and some TCR genes were expressed more preferentially in AITDs group than in the healthy control group, including TRBV15 (P = 0.001), TRBV4-2 (P = 0.003), TRBV9 (P = 0.007), TRBV3-2 (P = 0.012), TRBV7-8 (P = 0.015), TRBV25-1 (P = 0.019), TRBV12-4 (P = 0.019) and TRBV27 (P = 0.02) in GD patients as well as TRBV29-1 (P = 0.004), TRBV12-4 (P = 0.004), TRBV6-5 (P = 0.011), TRBV7-2 (P = 0.012), TRBV27 (P = 0.012), TRBV9 (P = 0.031) and TRBV4-2 (P = 0.032) in HT patients. Moreover, subgroup analysis showed that the difference in the TCR spectrum between the normal group and NGD was not obvious, but a large number of differential genes appeared in the RGD group. CONCLUSION TCR spectrum has changed in patients with AITDs with expanded repertoire and many upregulated TRBV genes. Moreover, this difference is not apparent in GD patients at the initial stage, but as the disease progresses, the differences in TCR profiles became more pronounced.
Collapse
Affiliation(s)
- Xi Jia
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China
| | - Tian-Yu Zhai
- Department of Endocrinology, Zhongshan Hospital of Fudan University, China
| | - Bing Wang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China
| | - Jin-An Zhang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China.
| | - Rong-Hua Song
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, China.
| |
Collapse
|
40
|
Vecchione A, Madley R, Danzl N, Borsotti C, Marharlooei MK, Li HW, Nauman G, Ding X, Ho SH, Fousteri G, Sykes M. T1D patient-derived hematopoietic stem cells are programmed to generate Tph, Tfh, and autoimmunity-associated B cell subsets in human immune system mice. Clin Immunol 2022; 240:109048. [PMID: 35644520 PMCID: PMC9564152 DOI: 10.1016/j.clim.2022.109048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/03/2022]
Abstract
Interactions between B cells and CD4+ T cells play a central role in the development of Type 1 Diabetes (T1D). Two helper cell subsets, follicular (Tfh) and peripheral (Tph) helper T cells, are increased in patients with T1D but their role in driving B cell autoimmunity is undefined. We used a personalized immune (PI) mouse model to generate human immune systems de novo from hematopoietic stem cells (HSCs) of patients with T1D or from healthy controls (HCs). Both groups developed Tfh and Tph-like cells, and those with T1D-derived immune systems demonstrated increased numbers of Tph-like and Tfh cells compared to HC-derived PI mice. T1D-derived immune systems included increased proportions of unconventional memory CD27-IgD- B cells and reduced proportions of naïve B cells compared to HC PI mice, resembling changes reported for patients with systemic lupus erythematosus. Our findings suggest that T1D HSCs are genetically programmed to produce increased proportions of T cells that promote the development of unconventional, possibly autoreactive memory B cells. PI mice provide an avenue for further understanding of the immune abnormalities that drive autoantibody pathogenesis and T1D.
Collapse
|
41
|
Yang JHM, Ward-Hartstonge KA, Perry DJ, Blanchfield JL, Posgai AL, Wiedeman AE, Diggins K, Rahman A, Tree TIM, Brusko TM, Levings MK, James EA, Kent SC, Speake C, Homann D, Long SA, The Immunology of Diabetes Society T-Cell Cytometry Group. Guidelines for standardizing T-cell cytometry assays to link biomarkers, mechanisms, and disease outcomes in type 1 diabetes. Eur J Immunol 2022; 52:372-388. [PMID: 35025103 PMCID: PMC9006584 DOI: 10.1002/eji.202049067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 11/10/2021] [Accepted: 12/22/2021] [Indexed: 11/11/2022]
Abstract
Cytometric immunophenotyping is a powerful tool to discover and implement T-cell biomarkers of type 1 diabetes (T1D) progression and response to clinical therapy. Although many discovery-based T-cell biomarkers have been described, to date, no such markers have been widely adopted in standard practice. The heterogeneous nature of T1D and lack of standardized assays and experimental design across studies is a major barrier to the broader adoption of T-cell immunophenotyping assays. There is an unmet need to harmonize the design of immunophenotyping assays, including those that measure antigen-agnostic cell populations, such that data collected from different clinical trial sites and T1D cohorts are comparable, yet account for cohort-specific features and different drug mechanisms of action. In these Guidelines, we aim to provide expert advice on how to unify aspects of study design and practice. We provide recommendations for defining cohorts, method implementation, as well as tools for data analysis and reporting by highlighting and building on selected successes. Harmonization of cytometry-based T-cell assays will allow researchers to better integrate findings across trials, ultimately enabling the identification and validation of biomarkers of disease progression and treatment response in T1D.
Collapse
Affiliation(s)
- Jennie H. M. Yang
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King’s College, London, UK
- National Institute of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service Foundation Trust, King’s College London, London, UK
| | - Kirsten A. Ward-Hartstonge
- Department of Surgery, University of British Columbia, Vancouver, California, USA
- BC Children’s Hospital Research Institute, Vancouver, California, USA
| | - Daniel J. Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - J. Lori Blanchfield
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Amanda L. Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Alice E. Wiedeman
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Kirsten Diggins
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Adeeb Rahman
- Human Immune Monitoring Center, Hess Center for Science and Medicine, Icahn School of Medicine, New York, New York, USA
| | - Timothy I. M. Tree
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King’s College, London, UK
- National Institute of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service Foundation Trust, King’s College London, London, UK
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Megan K. Levings
- Department of Surgery, University of British Columbia, Vancouver, California, USA
- BC Children’s Hospital Research Institute, Vancouver, California, USA
- School of Biomedical Engineering, University of British Columbia, California, USA
| | - Eddie A. James
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | - Sally C. Kent
- Diabetes Center of Excellence, University of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Dirk Homann
- Precision Immunology Institute, Icahn School of Medicine, New York, New York, USA
- Diabetes, Obesity, & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - S. Alice Long
- Center for Translational Research, Benaroya Research Institute, Seattle, Washington, USA
| | | |
Collapse
|
42
|
Srivastava N, Hu H, Vomund AN, Peterson OJ, Baker RL, Haskins K, Teyton L, Wan X, Unanue ER. Chromogranin A Deficiency Confers Protection From Autoimmune Diabetes via Multiple Mechanisms. Diabetes 2021; 70:2860-2870. [PMID: 34497137 PMCID: PMC8660984 DOI: 10.2337/db21-0513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022]
Abstract
Recognition of β-cell antigens by autoreactive T cells is a critical step in the initiation of autoimmune type1 diabetes. A complete protection from diabetes development in NOD mice harboring a point mutation in the insulin B-chain 9-23 epitope points to a dominant role of insulin in diabetogenesis. Generation of NOD mice lacking the chromogranin A protein (NOD.ChgA-/-) completely nullified the autoreactivity of the BDC2.5 T cell and conferred protection from diabetes onset. These results raised the issue concerning the dominant antigen that drives the autoimmune process. Here we revisited the NOD.ChgA-/- mice and found that their lack of diabetes development may not be solely explained by the absence of chromogranin A reactivity. NOD.ChgA-/- mice displayed reduced presentation of insulin peptides in the islets and periphery, which corresponded to impaired T-cell priming. Diabetes development in these mice was restored by antibody treatment targeting regulatory T cells or inhibiting transforming growth factor-β and programmed death-1 pathways. Therefore, the global deficiency of chromogranin A impairs recognition of the major diabetogenic antigen insulin, leading to broadly impaired autoimmune responses controlled by multiple regulatory mechanisms.
Collapse
Affiliation(s)
- Neetu Srivastava
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Hao Hu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Anthony N Vomund
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Orion J Peterson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Rocky L Baker
- Department of Immunology and Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO
| | - Kathryn Haskins
- Department of Immunology and Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO
| | - Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA
| | - Xiaoxiao Wan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
43
|
Linsley PS, Barahmand-Pour-Whitman F, Balmas E, DeBerg HA, Flynn KJ, Hu AK, Rosasco MG, Chen J, O'Rourke C, Serti E, Gersuk VH, Motwani K, Seay HR, Brusko TM, Kwok WW, Speake C, Greenbaum CJ, Nepom GT, Cerosaletti K. Autoreactive T cell receptors with shared germline-like α chains in type 1 diabetes. JCI Insight 2021; 6:151349. [PMID: 34806648 PMCID: PMC8663791 DOI: 10.1172/jci.insight.151349] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Human islet antigen reactive CD4+ memory T cells (IAR T cells) play a key role in the pathogenesis of autoimmune type 1 diabetes (T1D). Using single-cell RNA sequencing (scRNA-Seq) to identify T cell receptors (TCRs) in IAR T cells, we have identified a class of TCRs that share TCRα chains between individuals (“public” chains). We isolated IAR T cells from blood of healthy, new-onset T1D and established T1D donors using multiplexed CD154 enrichment and identified paired TCRαβ sequences from 2767 individual cells. More than a quarter of cells shared TCR junctions between 2 or more cells (“expanded”), and 29/47 (~62%) of expanded TCRs tested showed specificity for islet antigen epitopes. Public TCRs sharing TCRα junctions were most prominent in new-onset T1D. Public TCR sequences were more germline like than expanded unique, or “private,” TCRs, and had shorter junction sequences, suggestive of fewer random nucleotide insertions. Public TCRα junctions were often paired with mismatched TCRβ junctions in TCRs; remarkably, a subset of these TCRs exhibited cross-reactivity toward distinct islet antigen peptides. Our findings demonstrate a prevalent population of IAR T cells with diverse specificities determined by TCRs with restricted TCRα junctions and germline-constrained antigen recognition properties. Since these “innate-like” TCRs differ from previously described immunodominant TCRβ chains in autoimmunity, they have implications for fundamental studies of disease mechanisms. Self-reactive restricted TCRα chains and their associated epitopes should be considered in fundamental and translational investigations of TCRs in T1D.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Colin O'Rourke
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | | | | | - Keshav Motwani
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA.,University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Howard R Seay
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA.,University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA.,FlowJo, LLC, Ashland, Oregon, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA.,University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA.,Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Carla J Greenbaum
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | | | | |
Collapse
|
44
|
Nakayama M, Michels AW. Using the T Cell Receptor as a Biomarker in Type 1 Diabetes. Front Immunol 2021; 12:777788. [PMID: 34868047 PMCID: PMC8635517 DOI: 10.3389/fimmu.2021.777788] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022] Open
Abstract
T cell receptors (TCRs) are unique markers that define antigen specificity for a given T cell. With the evolution of sequencing and computational analysis technologies, TCRs are now prime candidates for the development of next-generation non-cell based T cell biomarkers, which provide a surrogate measure to assess the presence of antigen-specific T cells. Type 1 diabetes (T1D), the immune-mediated form of diabetes, is a prototypical organ specific autoimmune disease in which T cells play a pivotal role in targeting pancreatic insulin-producing beta cells. While the disease is now predictable by measuring autoantibodies in the peripheral blood directed to beta cell proteins, there is an urgent need to develop T cell markers that recapitulate T cell activity in the pancreas and can be a measure of disease activity. This review focuses on the potential and challenges of developing TCR biomarkers for T1D. We summarize current knowledge about TCR repertoires and clonotypes specific for T1D and discuss challenges that are unique for autoimmune diabetes. Ultimately, the integration of large TCR datasets produced from individuals with and without T1D along with computational 'big data' analysis will facilitate the development of TCRs as potentially powerful biomarkers in the development of T1D.
Collapse
MESH Headings
- Alleles
- Animals
- Biomarkers
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Disease Susceptibility
- Epitopes/chemistry
- Epitopes/immunology
- Epitopes/metabolism
- Genetic Predisposition to Disease
- Genetic Variation
- Histocompatibility Antigens/genetics
- Histocompatibility Antigens/immunology
- Humans
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Peptides/immunology
- Peptides/metabolism
- Protein Binding
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Aaron W. Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
45
|
Anderson AM, Landry LG, Alkanani AA, Pyle L, Powers AC, Atkinson MA, Mathews CE, Roep BO, Michels AW, Nakayama M. Human islet T cells are highly reactive to preproinsulin in type 1 diabetes. Proc Natl Acad Sci U S A 2021; 118:e2107208118. [PMID: 34611019 PMCID: PMC8521679 DOI: 10.1073/pnas.2107208118] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 01/29/2023] Open
Abstract
Cytotoxic CD8 T lymphocytes play a central role in the tissue destruction of many autoimmune disorders. In type 1 diabetes (T1D), insulin and its precursor preproinsulin are major self-antigens targeted by T cells. We comprehensively examined preproinsulin specificity of CD8 T cells obtained from pancreatic islets of organ donors with and without T1D and identified epitopes throughout the entire preproinsulin protein and defective ribosomal products derived from preproinsulin messenger RNA. The frequency of preproinsulin-reactive T cells was significantly higher in T1D donors than nondiabetic donors and also differed by individual T1D donor, ranging from 3 to over 40%, with higher frequencies in T1D organ donors with HLA-A*02:01. Only T cells reactive to preproinsulin-related peptides isolated from T1D donors demonstrated potent autoreactivity. Reactivity to similar regions of preproinsulin was also observed in peripheral blood of a separate cohort of new-onset T1D patients. These findings have important implications for designing antigen-specific immunotherapies and identifying individuals that may benefit from such interventions.
Collapse
Affiliation(s)
- Amanda M Anderson
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045
| | - Laurie G Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045
| | - Aimon A Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045
| | - Laura Pyle
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, CO 80045
| | - Alvin C Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Medical Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Clayton E Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Bart O Roep
- Department of Diabetes Immunology, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010
- Department of Internal Medicine, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045;
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
46
|
Greaves SA, Ravindran A, Santos RG, Chen L, Falta MT, Wang Y, Mitchell AM, Atif SM, Mack DG, Tinega AN, Maier LA, Dai S, Pinilla C, Grunewald J, Fontenot AP. CD4+ T cells in the lungs of acute sarcoidosis patients recognize an Aspergillus nidulans epitope. J Exp Med 2021; 218:212583. [PMID: 34410304 PMCID: PMC8383815 DOI: 10.1084/jem.20210785] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/18/2021] [Accepted: 07/22/2021] [Indexed: 11/05/2022] Open
Abstract
Löfgren’s syndrome (LS) is an acute form of sarcoidosis characterized by a genetic association with HLA-DRB1*03 (HLA-DR3) and an accumulation of CD4+ T cells of unknown specificity in the bronchoalveolar lavage (BAL). Here, we screened related LS-specific TCRs for antigen specificity and identified a peptide derived from NAD-dependent histone deacetylase hst4 (NDPD) of Aspergillus nidulans that stimulated these CD4+ T cells in an HLA-DR3–restricted manner. Using ELISPOT analysis, a greater number of IFN-γ– and IL-2–secreting T cells in the BAL of DR3+ LS subjects compared with DR3+ control subjects was observed in response to the NDPD peptide. Finally, increased IgG antibody responses to A. nidulans NDPD were detected in the serum of DR3+ LS subjects. Thus, our findings identify a ligand for CD4+ T cells derived from the lungs of LS patients and suggest a role of A. nidulans in the etiology of LS.
Collapse
Affiliation(s)
- Sarah A Greaves
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Avinash Ravindran
- Department of Medicine, Solna, Karolinska University Hospital, Stockholm, Sweden.,Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Radleigh G Santos
- Department of Mathematics, Nova Southeastern University, Ft. Lauderdale, FL
| | - Lan Chen
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Michael T Falta
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Yang Wang
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Angela M Mitchell
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Shaikh M Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Douglas G Mack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Alex N Tinega
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Lisa A Maier
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Medicine, National Jewish Health, Denver, CO
| | - Shaodong Dai
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Clemencia Pinilla
- Center for Translational Science, Florida International University, Port St. Lucie, FL
| | - Johan Grunewald
- Department of Medicine, Solna, Karolinska University Hospital, Stockholm, Sweden
| | - Andrew P Fontenot
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
47
|
Hanna SJ, Tatovic D, Thayer TC, Dayan CM. Insights From Single Cell RNA Sequencing Into the Immunology of Type 1 Diabetes- Cell Phenotypes and Antigen Specificity. Front Immunol 2021; 12:751701. [PMID: 34659258 PMCID: PMC8519581 DOI: 10.3389/fimmu.2021.751701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/14/2021] [Indexed: 01/10/2023] Open
Abstract
In the past few years, huge advances have been made in techniques to analyse cells at an individual level using RNA sequencing, and many of these have precipitated exciting discoveries in the immunology of type 1 diabetes (T1D). This review will cover the first papers to use scRNAseq to characterise human lymphocyte phenotypes in T1D in the peripheral blood, pancreatic lymph nodes and islets. These have revealed specific genes such as IL-32 that are differentially expressed in islet -specific T cells in T1D. scRNAseq has also revealed wider gene expression patterns that are involved in T1D and can predict its development even predating autoantibody production. Single cell sequencing of TCRs has revealed V genes and CDR3 motifs that are commonly used to target islet autoantigens, although truly public TCRs remain elusive. Little is known about BCR repertoires in T1D, but scRNAseq approaches have revealed that insulin binding BCRs commonly use specific J genes, share motifs between donors and frequently demonstrate poly-reactivity. This review will also summarise new developments in scRNAseq technology, the insights they have given into other diseases and how they could be leveraged to advance research in the type 1 diabetes field to identify novel biomarkers and targets for immunotherapy.
Collapse
Affiliation(s)
- Stephanie J. Hanna
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Danijela Tatovic
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Terri C. Thayer
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Department of Biological and Chemical Sciences, School of Natural and Social Sciences, Roberts Wesleyan College, Rochester, NY, United States
| | - Colin M. Dayan
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
48
|
Erdem N, Montero E, Roep BO. Breaking and restoring immune tolerance to pancreatic beta-cells in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2021; 28:397-403. [PMID: 34183540 DOI: 10.1097/med.0000000000000646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) results from the loss of immune tolerance to pancreatic beta-cells leading to their destruction. Immune intervention therapies tested in T1D so far delayed progression but failed to restore tolerance, which partly explains their lack of durable clinical efficacy. RECENT FINDINGS The role of beta-cells and islets themselves in dialogue with their micro- and macro-environment including the immune system and the intestinal microbiome is increasingly evident. Indeed, islets can both maintain and break immune tolerance. Some recent immune therapies in cancer that block immune regulation also break tolerance. Induction of immune tolerance requires activating immune activation too, whereas immune suppression precludes this process. Immunotherapy alone my not suffice without engaging islets to restore tolerance and preserve beta-cell function. SUMMARY New insight into the role of islet tissue and its interaction with its environment in preserving or breaking tolerance has contributed to understand the development of islet autoimmunity and T1D. Knowing which factors in islets and the immune system contribute to maintaining, breaking, and restoring the balance in the immune system is critical to prevent initiation and reverse disease progression, and guides the design of novel tolerogenic strategies for durable therapeutic intervention and remission that target both the immune system and distressed islets.
Collapse
Affiliation(s)
- Neslihan Erdem
- The Arthur Riggs Diabetes & Metabolism Research Institute at the Beckman Research Institute
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Enrique Montero
- The Arthur Riggs Diabetes & Metabolism Research Institute at the Beckman Research Institute
| | - Bart O Roep
- The Arthur Riggs Diabetes & Metabolism Research Institute at the Beckman Research Institute
- Department Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
49
|
Clark M, Kroger CJ, Ke Q, Zhang R, Statum K, Milner JJ, Martin AJ, Wang B, Tisch R. Coreceptor therapy has distinct short- and long-term tolerogenic effects intrinsic to autoreactive effector T cells. JCI Insight 2021; 6:e149130. [PMID: 34314385 PMCID: PMC8492310 DOI: 10.1172/jci.insight.149130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Immunotherapies are needed in the clinic that effectively suppress beta cell autoimmunity and reestablish long-term self-tolerance in type 1 diabetes. We previously demonstrated that nondepleting αCD4 and αCD8α antibodies establish rapid and indefinite remission in recent-onset diabetic NOD mice. Diabetes reversal by coreceptor therapy (CoRT) is induced by suppression of pathogenic effector T cells (Teff) and the selective egress of T cells from the pancreatic lymph nodes and islets that remain free of infiltration long-term. Here, we defined CoRT-induced events regulating early Teff function and pancreatic residency, and long-term tolerance. TCR-driven gene expression controlling autoreactive Teff expansion and proinflammatory activity was suppressed by CoRT, and islet T cell egress was sphingosine-1 phosphate-dependent. In both murine and human T cells, CoRT upregulated the Foxo1 transcriptional axis, which in turn was required for suppression and efficient pancreatic egress of Teff. Interestingly, long-term tolerance induced in late-preclinical NOD mice was marked by reseeding of the pancreas by a reduced CD8+ Teff pool exhibiting an exhausted phenotype. Notably, PD-1 blockade, which rescues exhausted Teff, resulted in diabetes onset in protected animals. These findings demonstrate that CoRT has distinct intrinsic effects on Teff that impact events early in induction and later in maintenance of self-tolerance.
Collapse
Affiliation(s)
- Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Rui Zhang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Karen Statum
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - J Justin Milner
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Aaron J Martin
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Bo Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| | - Roland Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States of America
| |
Collapse
|
50
|
Lin J, Lu Y, Wang B, Jiao P, Ma J. Analysis of immune cell components and immune-related gene expression profiles in peripheral blood of patients with type 1 diabetes mellitus. J Transl Med 2021; 19:319. [PMID: 34311758 PMCID: PMC8314644 DOI: 10.1186/s12967-021-02991-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease caused by severe loss of pancreatic β cells. Immune cells are key mediators of β cell destruction. This study attempted to investigate the role of immune cells and immune-related genes in the occurrence and development of T1DM. METHODS The raw gene expression profile of the samples from 12 T1DM patients and 10 normal controls was obtained from Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified by Limma package in R. The least absolute shrinkage and selection operator (LASSO)-support vector machines (SVM) were used to screen the hub genes. CIBERSORT algorithm was used to identify the different immune cells in distribution between T1DM and normal samples. Correlation of the hub genes and immune cells was analyzed by Spearman, and gene-GO-BP and gene-pathway interaction networks were constructed by Cytoscape plug-in ClueGO. Receiver operating characteristic (ROC) curves were used to assess diagnostic value of genes in T1DM. RESULTS The 50 immune-related DEGs were obtained between the T1DM and normal samples. Then, the 50 immune-related DEGs were further screened to obtain the 5 hub genes. CIBERSORT analysis revealed that the distribution of plasma cells, resting mast cells, resting NK cells and neutrophils had significant difference between T1DM and normal samples. Natural cytotoxicity triggering receptor 3 (NCR3) was significantly related to the activated NK cells, M0 macrophages, monocytes, resting NK cells, and resting memory CD4+ T cells. Moreover, tumor necrosis factor (TNF) was significantly associated with naive B cell and naive CD4+ T cell. NCR3 [Area under curve (AUC) = 0.918] possessed a higher accuracy than TNF (AUC = 0.763) in diagnosis of T1DM. CONCLUSIONS The immune-related genes (NCR3 and TNF) and immune cells (NK cells) may play a vital regulatory role in the occurrence and development of T1DM, which possibly provide new ideas and potential targets for the immunotherapy of diabetes mellitus (DM).
Collapse
Affiliation(s)
- Jian Lin
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin, 130021, P.R. China
| | - Yuanhua Lu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin, 130021, P.R. China
| | - Bizhou Wang
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, Jilin, 130021, P.R. China
| | - Ping Jiao
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin, 130021, P.R. China.
| | - Jie Ma
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin, 130021, P.R. China.
| |
Collapse
|