1
|
Qin H, Gao J, Xu W, Song Y, Zhang R, Wang H, Ye Y, Sun J, Jiang J, Liang H, Zhong N, Tian H, Chen X, Peng F, Tu Y. Dendritic cell-based microrobots for enhanced systemic antigen-specific immune tolerance. J Control Release 2025; 381:113566. [PMID: 40010412 DOI: 10.1016/j.jconrel.2025.02.062] [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: 11/25/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Current immunotherapeutic approaches for autoimmune disorders primarily rely on the use of generalized immunosuppressive medications. However, most immune drugs and tolerogenic immunomodulators are insufficient on their own to establish antigen-specific immunological tolerance (ASIT). Therefore, steering antigen-presenting cells (APCs) towards a tolerogenic state with minimal risk of broad immune suppression may be an effective approach. In pursuit of enhanced ASIT, magnetic nanoparticles cloaked with an erythrocyte membrane anchored with the model antigen ovalbumin have been successfully developed, allowing for the in vivo conversion of APCs into tolerogenic microrobots that respond to magnetic activation. Actuated by a rotating magnetic field (RMF), the in situ-formed cell-based microrobots can be guided to inflammatory sites, thereby augmenting systemic and local immune tolerance. These tolerogenic microrobots represent an innovative platform for active immunomodulation and provide precise control over the magnitude and direction of immune responses. This breakthrough offers new insights into the therapeutic management of allergies, autoimmune disorders, and the prevention of anti-drug antibodies in biologic therapies.
Collapse
Affiliation(s)
- Hanfeng Qin
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Junbin Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenxin Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanzhen Song
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ruotian Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hong Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yicheng Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jia Sun
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiamiao Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haiying Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ning Zhong
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hao Tian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaodong Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
2
|
Bessard MA, Moser A, Waeckel-Énée E, Lindo V, Gdoura A, You S, Wong FS, Greer F, van Endert P. Insulin-degrading enzyme regulates insulin-directed cellular autoimmunity in murine type 1 diabetes. Front Immunol 2024; 15:1474453. [PMID: 39600694 PMCID: PMC11588737 DOI: 10.3389/fimmu.2024.1474453] [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: 08/01/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Type 1 diabetes results from the destruction of pancreatic beta cells by autoreactive T cells. As an autoantigen with extremely high expression in beta cells, insulin triggers and sustains the autoimmune CD4+ and CD8+ T cell responses and islet inflammation. We have previously shown that deficiency for insulin-degrading enzyme (IDE), a ubiquitous cytosolic protease with very high affinity for insulin, induces endoplasmic reticulum (ER) stress and proliferation in islet cells and protects non-obese diabetic mice (NOD) from diabetes. Here we wondered whether IDE deficiency affects autoreactive CD8+ T cell responses to insulin and thereby immune pathogenesis in NOD mice. We find that Ide-/- NOD harbor fewer diabetogenic T cells and reduced numbers of CD8+ T cells recognizing the dominant autoantigen insulin and islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP). Using in vitro digestions and cellular antigen presentation assays, we show that generation of the dominant insulin epitope B15-23 involves both the proteasome and IDE. IDE deficiency attenuates MHC-I presentation of the immunodominant insulin epitope by beta cells to cognate CD8+ T cells. Consequently, Ide-/- islets display reduced susceptibility to autoimmune destruction upon grafting, and to killing by insulin-specific CD8+ T cells. Moreover, Ide-/- mice are partly resistant to disease transfer by CD8+ T cells specific for insulin but not for IGRP. Thus, IDE has a dual role in beta cells, regulating ER stress and proliferation while at the same time promoting insulin-directed autoreactive CD8+ T cell responses.
Collapse
Affiliation(s)
- Marie-Andrée Bessard
- Université Paris Cité, Institut National de la Santé et Recherche Médicale (INSERM), Centre National de La Recherche Scientifique (CNRS), Institut Necker Enfants Malades, Paris, France
| | - Anna Moser
- Université Paris Cité, Institut National de la Santé et Recherche Médicale (INSERM), Centre National de La Recherche Scientifique (CNRS), Institut Necker Enfants Malades, Paris, France
| | - Emmanuelle Waeckel-Énée
- Université Paris Cité, Institut National de la Santé et Recherche Médicale (INSERM), Centre National de La Recherche Scientifique (CNRS), Institut Necker Enfants Malades, Paris, France
| | | | - Abdelaziz Gdoura
- Université Paris Cité, Institut National de la Santé et Recherche Médicale (INSERM), Centre National de La Recherche Scientifique (CNRS), Institut Necker Enfants Malades, Paris, France
| | - Sylvaine You
- Université Paris Cité, Institut National de la Santé et Recherche Médicale (INSERM), Centre National de La Recherche Scientifique (CNRS), Institut Cochin, Paris, France
| | - F. Susan Wong
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | - Peter van Endert
- Université Paris Cité, Institut National de la Santé et Recherche Médicale (INSERM), Centre National de La Recherche Scientifique (CNRS), Institut Necker Enfants Malades, Paris, France
- Service Immunologie Biologique, Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Universitaire Necker-Enfants Malades, Paris, France
| |
Collapse
|
3
|
Yang K, Zhang Y, Ding J, Li Z, Zhang H, Zou F. Autoimmune CD8+ T cells in type 1 diabetes: from single-cell RNA sequencing to T-cell receptor redirection. Front Endocrinol (Lausanne) 2024; 15:1377322. [PMID: 38800484 PMCID: PMC11116783 DOI: 10.3389/fendo.2024.1377322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease caused by pancreatic β cell destruction and mediated primarily by autoreactive CD8+ T cells. It has been shown that only a small number of stem cell-like β cell-specific CD8+ T cells are needed to convert normal mice into T1D mice; thus, it is likely that T1D can be cured or significantly improved by modulating or altering self-reactive CD8+ T cells. However, stem cell-type, effector and exhausted CD8+ T cells play intricate and important roles in T1D. The highly diverse T-cell receptors (TCRs) also make precise and stable targeted therapy more difficult. Therefore, this review will investigate the mechanisms of autoimmune CD8+ T cells and TCRs in T1D, as well as the related single-cell RNA sequencing (ScRNA-Seq), CRISPR/Cas9, chimeric antigen receptor T-cell (CAR-T) and T-cell receptor-gene engineered T cells (TCR-T), for a detailed and clear overview. This review highlights that targeting CD8+ T cells and their TCRs may be a potential strategy for predicting or treating T1D.
Collapse
Affiliation(s)
- Kangping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yihan Zhang
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Jiatong Ding
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Zelin Li
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Hejin Zhang
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Fang Zou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Alam A, Dhoundiyal S, Ahmad N, Rao GSNK. Unveiling Diabetes: Categories, Genetics, Diagnostics, Treatments, and Future Horizons. Curr Diabetes Rev 2024; 20:e180823219972. [PMID: 37594107 DOI: 10.2174/1573399820666230818092958] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 08/19/2023]
Abstract
Diabetes mellitus is a global epidemic affecting millions of individuals worldwide. This comprehensive review aims to provide a thorough understanding of the categorization, disease identity, genetic architecture, diagnosis, and treatment of diabetes. The categorization of diabetes is discussed, with a focus on type 1 and type 2 diabetes, as well as the lesser-known types, type 3 and type 4 diabetes. The geographical variation, age, gender, and ethnic differences in the prevalence of type 1 and type 2 diabetes are explored. The impact of disease identity on disease management and the role of autoimmunity in diabetes are examined. The genetic architecture of diabetes, including the interplay between genotype and phenotype, is discussed to enhance our understanding of the underlying mechanisms. The importance of insulin injection sites and the insulin signalling pathway in diabetes management are highlighted. The diagnostic techniques for diabetes are reviewed, along with advancements for improved differentiation between types. Treatment and management approaches, including medications used in diabetes management are presented. Finally, future perspectives are discussed, emphasizing the need for further research and interventions to address the global burden of diabetes. This review serves as a valuable resource for healthcare professionals, researchers, and policymakers, providing insights to develop targeted strategies for the prevention, diagnosis, and management of this complex disease.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Niyaz Ahmad
- Department of Pharmaceutical Analysis, Green Research Lab, Green Industrial Company, Second Industrial Area, Riyadh 14334, Saudi Arabia
| | - G S N Koteswara Rao
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| |
Collapse
|
5
|
Thompson PJ, Pipella J, Rutter GA, Gaisano HY, Santamaria P. Islet autoimmunity in human type 1 diabetes: initiation and progression from the perspective of the beta cell. Diabetologia 2023; 66:1971-1982. [PMID: 37488322 PMCID: PMC10542715 DOI: 10.1007/s00125-023-05970-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 07/26/2023]
Abstract
Type 1 diabetes results from the poorly understood process of islet autoimmunity, which ultimately leads to the loss of functional pancreatic beta cells. Mounting evidence supports the notion that the activation and evolution of islet autoimmunity in genetically susceptible people is contingent upon early life exposures affecting the islets, especially beta cells. Here, we review some of the recent advances and studies that highlight the roles of these changes as well as antigen presentation and stress response pathways in beta cells in the onset and propagation of the autoimmune process in type 1 diabetes. Future progress in this area holds promise for advancing islet- and beta cell-directed therapies that could be implemented in the early stages of the disease and could be combined with immunotherapies.
Collapse
Affiliation(s)
- Peter J Thompson
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.
| | - Jasmine Pipella
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Guy A Rutter
- CRCHUM and Department of Medicine, Université de Montréal, Montréal, QC, Canada.
- Department of Diabetes, Endocrinology and Medicine, Faculty of Medicine, Imperial College, London, UK.
- LKC School of Medicine, Nanyang Technological College, Singapore, Republic of Singapore.
| | - Herbert Y Gaisano
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Pere Santamaria
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
6
|
Lu Y, Ruan Y, Hong P, Rui K, Liu Q, Wang S, Cui D. T-cell senescence: A crucial player in autoimmune diseases. Clin Immunol 2023; 248:109202. [PMID: 36470338 DOI: 10.1016/j.clim.2022.109202] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/24/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Senescent T cells are proliferative disabled lymphocytes that lack antigen-specific responses. The development of T-cell senescence in autoimmune diseases contributes to immunological disorders and disease progression. Senescent T cells lack costimulatory markers with the reduction of T cell receptor repertoire and the uptake of natural killer cell receptors. Senescent T cells exert cytotoxic effects through the expression of perforin, granzymes, tumor necrosis factor, and other molecules without the antigen-presenting process. DNA damage accumulation, telomere damage, and limited DNA repair capacity are important features of senescent T cells. Impaired mitochondrial function and accumulation of reactive oxygen species contribute to T cell senescence. Alleviation of T-cell senescence could provide potential targets for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Yinyun Lu
- Department of Infectious Diseases, Shaoxing People's Hospital, Shaoxing, China
| | - Yongchun Ruan
- Department of Infectious Diseases, Shaoxing People's Hospital, Shaoxing, China
| | - Pan Hong
- Department of Hematology, Shaoxing People's Hospital, Shaoxing, China
| | - Ke Rui
- Department of Transfusion, Shaoxing People's Hospital, Shaoxing, China
| | - Qi Liu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| | - Dawei Cui
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
7
|
Zhang M, Chen Y, Chen R, Wen Y, Huang Q, Liu Y, Zhao C. Research status of the effects of natural oligosaccharides on glucose metabolism. EFOOD 2022. [DOI: 10.1002/efd2.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Minjiao Zhang
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Yaobin Chen
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Ruoxin Chen
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Yuxi Wen
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Department of Analytical and Food Chemistry, Nutrition and Bromatology Group, Faculty of Sciences Universidade de Vigo Ourense Spain
| | - Qihui Huang
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Department of Analytical and Food Chemistry, Nutrition and Bromatology Group, Faculty of Sciences Universidade de Vigo Ourense Spain
| | - Yuanyuan Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
8
|
Erausquin E, Serra P, Parras D, Santamaria P, López-Sagaseta J. Structural plasticity in I-Ag7 links autoreactivity to hybrid insulin peptides in type I diabetes. Front Immunol 2022; 13:924311. [PMID: 35967292 PMCID: PMC9365947 DOI: 10.3389/fimmu.2022.924311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
We recently provided evidence for promiscuous recognition of several different hybrid insulin peptides (HIPs) by the highly diabetogenic, I-Ag7-restricted 4.1-T cell receptor (TCR). To understand the structural determinants of this phenomenon, we solved the structure of an agonistic HIP/I-Ag7 complex, both in isolation as well as bound to the 4.1-TCR. We find that HIP promiscuity of the 4.1-TCR is dictated, on the one hand, by an amino acid sequence pattern that ensures I-Ag7 binding and, on the other hand, by the presence of three acidic residues at positions P5, P7 and P8 that favor an optimal engagement by the 4.1-TCR’s complementary determining regions. Surprisingly, comparison of the TCR-bound and unbound HIP/I-Ag7 structures reveals that 4.1-TCR binding triggers several novel and unique structural motions in both the I-Ag7 molecule and the peptide that are essential for docking. This observation indicates that the type 1 diabetes-associated I-Ag7 molecule is structurally malleable and that this plasticity allows the recognition of multiple peptides by individual TCRs that would otherwise be unable to do so.
Collapse
Affiliation(s)
- Elena Erausquin
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed, Navarra, Spain
- Public University of Navarra (UPNA), Pamplona, Spain
- Navarra University Hospital, Pamplona, Spain
| | - Pau Serra
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Daniel Parras
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pere Santamaria
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Jacinto López-Sagaseta, ; Pere Santamaria,
| | - Jacinto López-Sagaseta
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed, Navarra, Spain
- Public University of Navarra (UPNA), Pamplona, Spain
- Navarra University Hospital, Pamplona, Spain
- *Correspondence: Jacinto López-Sagaseta, ; Pere Santamaria,
| |
Collapse
|
9
|
Yang Y, Santamaria P. Antigen-specific nanomedicines for the treatment of autoimmune disease: target cell types, mechanisms and outcomes. Curr Opin Biotechnol 2022; 74:285-292. [PMID: 35007990 DOI: 10.1016/j.copbio.2021.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/07/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023]
Abstract
Nanoparticle (NP)-based delivery of autoantigenic ligands represents a promising approach to modulate autoimmune responses in vivo. Over the last 15 years, a growing number of compounds have been tested in animal models of various experimental and/or spontaneous autoimmune diseases. Based on the underlying design principles and mechanistic underpinnings, these compounds can be categorized into three broad groups: NPs (or microparticles, MPs) as vehicles for targeted delivery of antigens to tolerogenic antigen-presenting cells (APCs); NPs as scaffolds for targeted delivery of both antigen and immunomodulatory molecules to professional APCs; and NPs as multimerization platforms for direct cognate T-cell targeting via recombinant peptide-major histocompatibility complex molecules (pMHCs). These various compounds operate through different mechanisms of action, eliciting pharmacodynamic effects that range from antigen-specific clonal deletion to induction of comprehensive, yet disease-specific, bystander immunoregulation. Here, we review the outcomes of the various approaches tested to date and discuss their translational significance in the context of mode of action vis-à-vis immunologically complex human autoimmune diseases.
Collapse
Affiliation(s)
- Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1 Canada; Department of Biochemistry and Molecular Biology and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1, Canada.
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1 Canada; Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona 08036, Spain.
| |
Collapse
|
10
|
Reinholdt K, Munk C, Thomsen LT, Dehlendorff C, Carstensen B, Jørgensen ME, Kjaer SK. Increased incidence of genital warts among women and men with type 1 diabetes compared with the general population-results from a nationwide registry-based, cohort study. Acta Diabetol 2022; 59:105-112. [PMID: 34499240 DOI: 10.1007/s00592-021-01786-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/11/2021] [Indexed: 01/23/2023]
Abstract
AIMS To estimate the incidence rates of genital warts (GWs) in women and men with type 1 diabetes compared to persons without diabetes. METHODS In this nationwide registry-based cohort study, we included the entire population aged 15 to 49 years living in Denmark between 1996 and 2016. From national registries, we retrieved individual level information on diabetes status, diagnoses and treatment of GWs, and potential confounding variables. We used Poisson regression to model sex- and age-specific incidence rates of GWs in persons with type 1 diabetes and persons without diabetes. Based on the models, we computed sex-specific incidence rate ratios (IRRs) of GWs in persons with type 1 diabetes compared to persons without diabetes, overall and according to age. RESULTS The analysis included 3,514,824 persons without type 2 diabetes and no GW diagnoses before baseline. The incidence rate of GWs in persons with type 1 diabetes was higher than in those without diabetes, both among women (IRR = 1.59; 95% CI, 1.42-1.78) and men (IRR = 1.36; 95% CI, 1.25-1.48). The pattern of increased incidence rates of GWs in persons with type 1 diabetes was seen at all ages. CONCLUSIONS Persons with type 1 diabetes have higher incidence rates of GWs than persons without diabetes. This supports the importance of HPV vaccination of young girls and boys with type 1 diabetes.
Collapse
Affiliation(s)
- K Reinholdt
- Unit of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - C Munk
- Unit of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - L T Thomsen
- Unit of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - C Dehlendorff
- Statistics and Data Analysis, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - B Carstensen
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - M E Jørgensen
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
- University of Southern Denmark, Odense, Denmark
| | - S K Kjaer
- Unit of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark.
- Department of Gynecology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
11
|
Parras D, Solé P, Delong T, Santamaría P, Serra P. Recognition of Multiple Hybrid Insulin Peptides by a Single Highly Diabetogenic T-Cell Receptor. Front Immunol 2021; 12:737428. [PMID: 34527002 PMCID: PMC8435627 DOI: 10.3389/fimmu.2021.737428] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanisms underlying the major histocompatibility complex class II (MHCII) type 1 diabetes (T1D) association remain incompletely understood. We have previously shown that thymocytes expressing the highly diabetogenic, I-Ag7-restricted 4.1-T-cell receptor (TCR) are MHCII-promiscuous, and that, in MHCII-heterozygous mice, they sequentially undergo positive and negative selection/Treg deviation by recognizing pro- and anti-diabetogenic MHCII molecules on cortical thymic epithelial cells and medullary hematopoietic antigen-presenting cells (APCs), respectively. Here, we use a novel autoantigen discovery approach to define the antigenic specificity of this TCR in the context of I-Ag7. This was done by screening the ability of random epitope-GS linker-I- A β g 7 chain fusion pools to form agonistic peptide-MHCII complexes on the surface of I- A α d chain-transgenic artificial APCs. Pool deconvolution, I-Ag7-binding register-fixing, TCR contact residue mapping, and alanine scanning mutagenesis resulted in the identification of a 4.1-TCR recognition motif XL(G/A)XEXE(D/E)X that was shared by seven agonistic hybrid insulin peptides (HIPs) resulting from the fusion of several different chromogranin A and/or insulin C fragments, including post-translationally modified variants. These data validate a novel, highly sensitive MHCII-restricted epitope discovery approach for orphan TCRs and suggest thymic selection of autoantigen-promiscuous TCRs as a mechanism for the murine T1D-I-Ag7-association.
Collapse
MESH Headings
- Animals
- Autoantigens/genetics
- Autoantigens/immunology
- Autoantigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CHO Cells
- Coculture Techniques
- Cricetulus
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Epitopes
- HEK293 Cells
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Insulin/genetics
- Insulin/immunology
- Insulin/metabolism
- Jurkat Cells
- Mice, Inbred NOD
- Mice, Knockout
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Mice
Collapse
Affiliation(s)
- Daniel Parras
- Institut D’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Patricia Solé
- Institut D’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Thomas Delong
- Skaggs School of Pharmacy and Pharmaceutical Sciences (SSPPS), Department of Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States
| | - Pere Santamaría
- Institut D’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Pau Serra
- Institut D’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
12
|
Low-dose interleukin-2-loaded nanoparticle effect on NK and T-reg cell expression in experimentally induced type 1 diabetes mellitus. GASTROENTEROLOGY REVIEW 2021; 16:67-82. [PMID: 33986891 PMCID: PMC8112267 DOI: 10.5114/pg.2021.104737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
Introduction Type 1 diabetes mellitus is an autoimmune disorder characterized by inflammatory damage to pancreatic β cells resulting in loss of insulin secretion. In autoimmune type 1 diabetes mellitus (T1D) natural killer cells (NK) initiate pancreatic islets cell lyses in autoimmune T1D. Loss of T regulatory cells (Treg) at disease onset facilitates the activation and accumulation of NKs in the pancreatic microenvironment. A proper low-dose interleukin 2 (IL-2) could enhance Tregs and enforce control and regulation of pro-inflammatory NKs. Aim This relation needs to be studied to improve therapeutic strategies aimed at resetting the balance between Tregs and proinflammatory cells. Material and methods We used novel formulations of low-dose IL-2 loaded on chitosan nanoparticles. The study included 116 T1D BALB/c mice experimentally induced by streptozotocin, divided into groups. Their splenocytes were maintained in a short-term culture for assessment of expression of CD4+FOXP3+ Treg and NKp46+NK by both flow cytometry and enzyme-linked immunoassay (ELISA). Morphological, immunohistochemical, and morphometrical analyses were done.In vitro suppressor assay was used to assess the suppressor effect of Treg cells after exogenous IL-2 treatment. Results NK cell expression, NKp46 level, and NK cell functions were modulated more in mice injected with IL-2-loaded chitosan nanoparticles than in other groups. A statistical inverse correlation was found between Treg and NK cell expression in IL-2-loaded chitosan with 0.3 µIU (p = 0.047), and this correlation was related to FOXP3 expression on Treg cells. The modified expression of NK and NKp46 was noticed in mice injected with 0.3 µIU for longer duration (3 weeks) (p < 0.001), but the NK functions did not show any significant changes with prolonged treatment. Conclusions Prolonged administration of low-dose IL-2 results in the vigorous expression of NKp46, indicating a significant role of Tregs in NK stimulation and motivation. Low-dose IL-2 selectively modulates NKp46 NK and FOXP3+ Tregs and increases their expression.
Collapse
|
13
|
Volfson-Sedletsky V, Jones A, Hernandez-Escalante J, Dooms H. Emerging Therapeutic Strategies to Restore Regulatory T Cell Control of Islet Autoimmunity in Type 1 Diabetes. Front Immunol 2021; 12:635767. [PMID: 33815387 PMCID: PMC8015774 DOI: 10.3389/fimmu.2021.635767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Despite many decades of investigation uncovering the autoimmune mechanisms underlying Type 1 Diabetes (T1D), translating these findings into effective therapeutics has proven extremely challenging. T1D is caused by autoreactive T cells that become inappropriately activated and kill the β cells in the pancreas, resulting in insulin insufficiency and hyperglycemia. A large body of evidence supports the idea that the unchecked activation and expansion of autoreactive T cells in T1D is due to defects in immunosuppressive regulatory T cells (Tregs) that are critical for maintaining peripheral tolerance to islet autoantigens. Hence, repairing these Treg deficiencies is a much sought-after strategy to treat the disease. To accomplish this goal in the most precise, effective and safest way possible, restored Treg functions will need to be targeted towards suppressing the autoantigen-specific immune responses only and/or be localized in the pancreas. Here we review the most recent developments in designing Treg therapies that go beyond broad activation or expansion of non-specific polyclonal Treg populations. We focus on two cutting-edge strategies namely ex vivo generation of optimized Tregs for re-introduction in T1D patients vs direct in situ stimulation and restoration of endogenous Treg function.
Collapse
Affiliation(s)
- Victoria Volfson-Sedletsky
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Albert Jones
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Jaileene Hernandez-Escalante
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Hans Dooms
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
14
|
Effect of low dose IL-2 loaded chitosan nanoparticles on natural killer and regulatory T cell expression in experimentally induced autoimmune type 1 diabetes mellitus. Cent Eur J Immunol 2021; 45:382-392. [PMID: 33658887 PMCID: PMC7882410 DOI: 10.5114/ceji.2020.103412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 12/13/2019] [Indexed: 11/20/2022] Open
Abstract
Introduction Natural killer cells (NK) initiate pancreatic islets cell lyses in autoimmune type 1 diabetes mellitus (T1D). Loss of T regulatory cells (Treg) at disease onset facilitates activation and accumulation of NKs in the pancreatic microenvironment. A proper low dose interleukin 2 (IL-2) could enhance Tregs and enforce control and regulation of pro-inflammatory NKs. This relation needs to be studied to improve therapeutic strategies aimed at resetting the balance between Tregs and proinflammatory cells. Material and methods We used novel formulations of low dose IL-2 loaded on chitosan nanoparticles. The study included 116 T1D BALB/c mice experimentally induced by streptozotocin, divided into groups. Their splenocytes were maintained in a short-term culture for assessment of expression of CD4+Foxp3+ Treg and NKp46+NK by both flow cytometry and enzyme linked immunoassay (ELISA). In vitro suppressor-assay was used in order to assess the suppressor effect of Treg cells after exogenous IL-2 treatment. Results NK cell expression, NKp46 level and NK cell functions were modulated in mice injected with IL-2 loaded chitosan nanoparticles than other groups. A statistical inverse correlation was found between Treg and NK cell expression in IL-2 loaded chitosan with (0.3 µIU) (p = 0.047) and this correlation was related to Foxp3 expression on Treg cells. The modified expression of NK and NKp46 was noticed in mice injected with (0.3 µIU) for longer duration (three weeks) (p < 0.001) but the NK functions did not show any significant changes with prolonged treatment. Conclusions Low dose (0.3) µIU IL-2 nanoparticles effectively modulated NK and NKp46 expression. It selectively modulates the suppressive activity of Tregs indicating a significant role of Tregs in NK activation and function by controlling the availability of IL-2 in the microenvironment.
Collapse
|
15
|
Luo YL, Liang LF, Gan YJ, Liu J, Zhang Y, Fan YN, Zhao G, Czarna A, Lu ZD, Du XJ, Shen S, Xu CF, Lian ZX, Wang J. An All-in-One Nanomedicine Consisting of CRISPR-Cas9 and an Autoantigen Peptide for Restoring Specific Immune Tolerance. ACS APPLIED MATERIALS & INTERFACES 2020; 12:48259-48271. [PMID: 33070614 DOI: 10.1021/acsami.0c10885] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanotechnology has shown great promise in treating diverse diseases. However, developing nanomedicines that can cure autoimmune diseases without causing systemic immunosuppression is still quite challenging. Herein, we propose an all-in-one nanomedicine comprising an autoantigen peptide and CRISPR-Cas9 to restore specific immune tolerance by engineering dendritic cells (DCs) into a tolerogenic phenotype, which can expand autoantigen-specific regulatory T (Treg) cells. In brief, we utilized cationic lipid-assisted poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-PLGA) nanoparticles to simultaneously encapsulate an autoimmune diabetes-relevant peptide (2.5mi), a CRISPR-Cas9 plasmid (pCas9), and three guide RNAs (gRNAs) targeting costimulatory molecules (CD80, CD86, and CD40). We demonstrated that the all-in-one nanomedicine was able to effectively codeliver these components into DCs, followed by simultaneous disruption of the three costimulatory molecules and presentation of the 2.5mi peptide on the genome-edited DCs. The resulting tolerogenic DCs triggered the generation and expansion of autoantigen-specific Treg cells by presenting the 2.5mi peptide to CD4+ T cells in the absence of costimulatory signals. Using autoimmune type 1 diabetes (T1D) as a typical disease model, we demonstrated that our nanomedicine prevented autoimmunity to islet components and inhibited T1D development. Our all-in-one nanomedicine achieved codelivery of CRISPR-Cas9 and the peptide to DCs and could be easily applied to other autoimmune diseases by substitution of different autoantigen peptides.
Collapse
Affiliation(s)
- Ying-Li Luo
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Li-Fang Liang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yun-Jiu Gan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, P. R. China
| | - Jing Liu
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yue Zhang
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Ya-Nan Fan
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Gui Zhao
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Anna Czarna
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zi-Dong Lu
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xiao-Jiao Du
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Song Shen
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Cong-Fei Xu
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
| | - Zhe-Xiong Lian
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Jun Wang
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, P. R. China
- Research Institute for Food Nutrition and Human Health, Guangzhou 510641, P. R. China
| |
Collapse
|
16
|
Jamaleddine H, Santamaria P, Khadra A. Quantifying immunoregulation by autoantigen-specific T-regulatory type 1 cells in mice with simultaneous hepatic and extra-hepatic autoimmune disorders. Immunology 2020; 161:209-229. [PMID: 32687611 DOI: 10.1111/imm.13241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/11/2020] [Accepted: 07/12/2020] [Indexed: 12/18/2022] Open
Abstract
Nanoparticles (NPs) displaying autoimmune disease-relevant peptide-major histocompatibility complex class II molecules (pMHCII-NPs) trigger cognate T-regulatory type 1 (Tr1)-cell formation and expansion, capable of reversing organ-specific autoimmune responses. These pMHCII-NPs that display epitopes from mitochondrial protein can blunt the progression of both autoimmune hepatitis (AIH) and experimental autoimmune encephalomyelitis (EAE) in mice carrying either disease. However, with co-morbid mice having both diseases, these pMHCII-NPs selectively treat AIH. In contrast, pMHCII-NPs displaying central nervous system (CNS)-specific epitopes can efficiently treat CNS autoimmunity, both in the absence and presence of AIH, without having any effects on the progression of the latter. Here, we develop a compartmentalized population model of T-cells in co-morbid mice to identify the mechanisms by which Tr1 cells mediate organ-specific immunoregulation. We perform time-series simulations and bifurcation analyses to study how varying physiological parameters, including local cognate antigenic load and rates of Tr1-cell recruitment and retention, affect T-cell allocation and Tr1-mediated immunoregulation. Various regimes of behaviour, including 'competitive autoimmunity' where pMHCII-NP-treatment fails against both diseases, are identified and compared with experimental observations. Our results reveal that a transient delay in Tr1-cell recruitment to the CNS, resulting from inflammation-dependent Tr1-cell allocation, accounts for the liver-centric effects of AIH-specific pMHCII-NPs in co-morbid mice as compared with mice exclusively having EAE. They also suggest that cognate autoantigen expression and local Tr1-cell retention are key determinants of effective regulatory-cell function. These results thus provide new insights into the rules that govern Tr1-cell recruitment and their autoregulatory function.
Collapse
Affiliation(s)
| | - Pere Santamaria
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AL, Canada.,Institut D'Investigacions Biomèdiques August Pi i Sunyer, Carrer del Rosselló, Barcelona, Spain
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, QC, Canada
| |
Collapse
|
17
|
Zhu D, Yan Q, Liu J, Wu X, Jiang Z. Can functional oligosaccharides reduce the risk of diabetes mellitus? FASEB J 2019; 33:11655-11667. [PMID: 31415188 DOI: 10.1096/fj.201802802rrr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diabetes significantly affects the life quality and length of patients with diabetes, and almost half of the 4 million people who die from diabetes are under the age of 60. Because of the increasing number of patients with diabetes and the side effects of antidiabetic drugs, the search for new dietary supplementation from natural resources, especially functional oligosaccharides, has attracted much attention among scientific researchers. Functional oligosaccharides are potential antidiabetic treatments because of their nondigestible, low-calorie, and probiotic features. The antidiabetic activity of multiple functional oligosaccharides such as fructo-oligosaccharides, galacto-oligosaccharides, and xylo-oligosaccharides has been reviewed in this paper. Molecular mechanisms involved in the antidiabetic activity of oligosaccharides have been systematically discussed from multiple perspectives, including the improvement of pancreas function, α-glucosidase inhibition, the relief of insulin and leptin resistance, anti-inflammatory effects, regulation of gut microbiota and hormones, and the intervention of diabetic risk factors. In addition, the antidiabetic effects of functional oligosaccharides through the complex gut-brain-liver axis are summarized. The concepts addressed in this review have important clinical implications, although more works are needed to confirm the antidiabetic mechanisms of functional oligosaccharides, standardize safe dose levels, and clarify their metabolism in the human body.-Zhu, D., Yan, Q., Liu, J., Wu, X., Jiang, Z. Can functional oligosaccharides reduce the risk of diabetes mellitus?
Collapse
Affiliation(s)
- Di Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qiaojuan Yan
- College of Engineering, China Agricultural University, Beijing, China
| | - Jun Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xia Wu
- College of Engineering, China Agricultural University, Beijing, China
| | - Zhengqiang Jiang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
18
|
Zhou X, Zhang S, Yu F, Zhao G, Geng S, Yu W, Wang XY, Wang B. Tolerogenic vaccine composited with islet-derived multipeptides and cyclosporin A induces pTreg and prevents Type 1 diabetes in murine model. Hum Vaccin Immunother 2019; 16:240-250. [PMID: 31070990 PMCID: PMC7062422 DOI: 10.1080/21645515.2019.1616504] [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] [Indexed: 11/17/2022] Open
Abstract
Regulatory T cells (Tregs) play a crucial role in the control of the initiation and progression of type 1 diabetes (T1D). Various immunological interventions including those to ex vivo expansion Tregs transfer, in vivo induction of peripherally derived Treg (pTreg) have been considered as promising approaches for T1D therapy. In this study, we developed a novel tolerogenic vaccine using four autoantigenic peptides of islet-derived with cyclosporine A (CsA) as the pTreg inducer, designated as GAD-IN+CsA. This vaccine immunized into prediabetic NOD mice subcutaneously could induce IL-10 and TGF-β expressing pTregs and lead to suppressing autoreactive T cells responses, resulting in the prevention of T1D in these animals. Furthermore, we demonstrated that CsA with autoantigenic peptides modulates dendritic cells (DCs) to become immature IL-10hiCD40lo DCs. Such modulated DCs could foster naïve CD4+CD25− T cell into Tregs when presenting antigen peptides in vitro. This novel approach offers an alternative strategy to induce pTregs to treat T1D.
Collapse
Affiliation(s)
- Xian Zhou
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shijie Zhang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Fan Yu
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Gan Zhao
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Shuang Geng
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Wencong Yu
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Xuan-Yi Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China.,Children Hospital of Fudan University, Shanghai, China
| |
Collapse
|
19
|
Increased expression of microRNAs, miR-20a and miR-326 in PBMCs of patients with type 1 diabetes. Mol Biol Rep 2018; 45:1973-1980. [PMID: 30194557 DOI: 10.1007/s11033-018-4352-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/31/2018] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder which is characterized by autoimmune attack on β cells of pancreas and lack of insulin. The involvement of microRNAs (miRNAs) in the development of immune system and their differential expression in various autoimmune diseases including T1D have been well established. In this study, the association between expression levels of miR-20a, miR-326 and T1D were evaluated. The expression levels of miR-20a and miR-326 were measured in the PBMCs of 21 T1D patients and 16 healthy controls using qPCR method. In silico analysis was also performed on targetome of miR-20a and miR-326. Both miR-20a (p value: 0.015) and miR-326 (p value: 0.005) were upregulated in the PBMCs of T1D patients compared to healthy controls. Furthermore, different dysregulated miR326-mRNA and miR20a-mRNA interactions were also suggested using integrative computational analysis. The expression level of miR-20a and miR-326 indicates significant association with T1D which suggests the possible regulatory effects of these non-coding RNAs in T1D.
Collapse
|
20
|
|
21
|
Orabona C, Mondanelli G, Pallotta MT, Carvalho A, Albini E, Fallarino F, Vacca C, Volpi C, Belladonna ML, Berioli MG, Ceccarini G, Esposito SM, Scattoni R, Verrotti A, Ferretti A, De Giorgi G, Toni S, Cappa M, Matteoli MC, Bianchi R, Matino D, Iacono A, Puccetti M, Cunha C, Bicciato S, Antognelli C, Talesa VN, Chatenoud L, Fuchs D, Pilotte L, Van den Eynde B, Lemos MC, Romani L, Puccetti P, Grohmann U. Deficiency of immunoregulatory indoleamine 2,3-dioxygenase 1in juvenile diabetes. JCI Insight 2018; 3:96244. [PMID: 29563329 DOI: 10.1172/jci.insight.96244] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 02/13/2018] [Indexed: 12/23/2022] Open
Abstract
A defect in indoleamine 2,3-dioxygenase 1 (IDO1), which is responsible for immunoregulatory tryptophan catabolism, impairs development of immune tolerance to autoantigens in NOD mice, a model for human autoimmune type 1 diabetes (T1D). Whether IDO1 function is also defective in T1D is still unknown. We investigated IDO1 function in sera and peripheral blood mononuclear cells (PBMCs) from children with T1D and matched controls. These children were further included in a discovery study to identify SNPs in IDO1 that might modify the risk of T1D. T1D in children was characterized by a remarkable defect in IDO1 function. A common haplotype, associated with dysfunctional IDO1, increased the risk of developing T1D in the discovery and also confirmation studies. In T1D patients sharing such a common IDO1 haplotype, incubation of PBMCs in vitro with tocilizumab (TCZ) - an IL-6 receptor blocker - would, however, rescue IDO1 activity. In an experimental setting with diabetic NOD mice, TCZ was found to restore normoglycemia via IDO1-dependent mechanisms. Thus, functional SNPs of IDO1 are associated with defective tryptophan catabolism in human T1D, and maneuvers aimed at restoring IDO1 function would be therapeutically effective in at least a subgroup of T1D pediatric patients.
Collapse
Affiliation(s)
- Ciriana Orabona
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria T Pallotta
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Elisa Albini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria L Belladonna
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria G Berioli
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy
| | - Giulia Ceccarini
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Susanna Mr Esposito
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Raffaella Scattoni
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Alberto Verrotti
- Department of Surgical and Biomedical Sciences, University of Perugia, Perugia, Italy.,Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | | | - Giovanni De Giorgi
- Pediatric Clinic of S. Maria della Misericordia Hospital, Perugia, Italy
| | - Sonia Toni
- Juvenile Diabetes Center, Anna Meyer Children's Hospital, Florence, Italy
| | - Marco Cappa
- Unit of Endocrinology and Diabetes, 'Bambino Gesù' Children's Hospital, Rome, Italy
| | - Maria C Matteoli
- Unit of Endocrinology and Diabetes, 'Bambino Gesù' Children's Hospital, Rome, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Davide Matino
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cinzia Antognelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vincenzo N Talesa
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Lucienne Chatenoud
- INSERM U1013, Hôpital Necker-Enfants Malades and Université Paris Descartes, Paris, France
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University, Innsbruck, Austria
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, Walloon Excellence in Life Sciences and Biotechnology and.,De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Benoît Van den Eynde
- Ludwig Institute for Cancer Research, Walloon Excellence in Life Sciences and Biotechnology and.,De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Manuel C Lemos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
22
|
Epps SJ, Boldison J, Stimpson ML, Khera TK, Lait PJP, Copland DA, Dick AD, Nicholson LB. Re-programming immunosurveillance in persistent non-infectious ocular inflammation. Prog Retin Eye Res 2018. [PMID: 29530739 PMCID: PMC6563519 DOI: 10.1016/j.preteyeres.2018.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ocular function depends on a high level of anatomical integrity. This is threatened by inflammation, which alters the local tissue over short and long time-scales. Uveitis due to autoimmune disease, especially when it involves the retina, leads to persistent changes in how the eye interacts with the immune system. The normal pattern of immune surveillance, which for immune privileged tissues is limited, is re-programmed. Many cell types, that are not usually present in the eye, become detectable. There are changes in the tissue homeostasis and integrity. In both human disease and mouse models, in the most extreme cases, immunopathological findings consistent with development of ectopic lymphoid-like structures and disrupted angiogenesis accompany severely impaired eye function. Understanding how the ocular environment is shaped by persistent inflammation is crucial to developing novel approaches to treatment.
Collapse
Affiliation(s)
- Simon J Epps
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK
| | - Joanne Boldison
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Madeleine L Stimpson
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK
| | - Tarnjit K Khera
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK; School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, BS8 1TD, UK
| | - Philippa J P Lait
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK
| | - David A Copland
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK; School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, BS8 1TD, UK; UCL-Institute of Ophthalmology and National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, EC1V 2PD, UK
| | - Lindsay B Nicholson
- Academic Unit of Ophthalmology, Bristol Medical School, Faculty of Health Sciences, University of Bristol, BS8 1TD, UK; School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, BS8 1TD, UK.
| |
Collapse
|
23
|
MHC-mismatched mixed chimerism restores peripheral tolerance of noncross-reactive autoreactive T cells in NOD mice. Proc Natl Acad Sci U S A 2018; 115:E2329-E2337. [PMID: 29463744 PMCID: PMC5877958 DOI: 10.1073/pnas.1720169115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mixed chimerism has shown good potential to cure some autoimmune diseases and prevent tissue rejection. It is known that MHC-mismatched but not -matched mixed chimerism effectively tolerizes autoreactive T cells, even those noncross-reactive T cells that do not directly recognize donor-type antigen presenting cells [i.e., dendritic cells (DCs)]. How this is accomplished remains unknown. These studies have shown that tolerizing peripheral residual host-type noncross-reactive autoreactive T cells requires engraftment of donor-type DCs and involves a host-type DC-mediated increase in donor-type Treg cells, which associates with restoration of tolerogenic features of host-type plasmacytoid DCs and expansion of host-type Treg cells. This study suggests a previously unrecognized tolerance network among donor- and host-type DCs and Treg cells in MHC-mismatched mixed chimeras. Autoimmune type 1 diabetes (T1D) and other autoimmune diseases are associated with particular MHC haplotypes and expansion of autoreactive T cells. Induction of MHC-mismatched but not -matched mixed chimerism by hematopoietic cell transplantation effectively reverses autoimmunity in diabetic nonobese diabetic (NOD) mice, even those with established diabetes. As expected, MHC-mismatched mixed chimerism mediates deletion in the thymus of host-type autoreactive T cells that have T-cell receptor (TCR) recognizing (cross-reacting with) donor-type antigen presenting cells (APCs), which have come to reside in the thymus. However, how MHC-mismatched mixed chimerism tolerizes host autoreactive T cells that recognize only self-MHC–peptide complexes remains unknown. Here, using NOD.Rag1−/−.BDC2.5 or NOD.Rag1−/−.BDC12-4.1 mice that have only noncross-reactive transgenic autoreactive T cells, we show that induction of MHC-mismatched but not -matched mixed chimerism restores immune tolerance of peripheral noncross-reactive autoreactive T cells. MHC-mismatched mixed chimerism results in increased percentages of both donor- and host-type Foxp3+ Treg cells and up-regulated expression of programmed death-ligand 1 (PD-L1) by host-type plasmacytoid dendritic cells (pDCs). Furthermore, adoptive transfer experiments showed that engraftment of donor-type dendritic cells (DCs) and expansion of donor-type Treg cells are required for tolerizing the noncross-reactive autoreactive T cells in the periphery, which are in association with up-regulation of host-type DC expression of PD-L1 and increased percentage of host-type Treg cells. Thus, induction of MHC-mismatched mixed chimerism may establish a peripheral tolerogenic DC and Treg network that actively tolerizes autoreactive T cells, even those with no TCR recognition of the donor APCs.
Collapse
|
24
|
Fatehi F, Kyrychko SN, Ross A, Kyrychko YN, Blyuss KB. Stochastic Effects in Autoimmune Dynamics. Front Physiol 2018; 9:45. [PMID: 29456513 PMCID: PMC5801658 DOI: 10.3389/fphys.2018.00045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/15/2018] [Indexed: 01/05/2023] Open
Abstract
Among various possible causes of autoimmune disease, an important role is played by infections that can result in a breakdown of immune tolerance, primarily through the mechanism of “molecular mimicry”. In this paper we propose and analyse a stochastic model of immune response to a viral infection and subsequent autoimmunity, with account for the populations of T cells with different activation thresholds, regulatory T cells, and cytokines. We show analytically and numerically how stochasticity can result in sustained oscillations around deterministically stable steady states, and we also investigate stochastic dynamics in the regime of bi-stability. These results provide a possible explanation for experimentally observed variations in the progression of autoimmune disease. Computations of the variance of stochastic fluctuations provide practically important insights into how the size of these fluctuations depends on various biological parameters, and this also gives a headway for comparison with experimental data on variation in the observed numbers of T cells and organ cells affected by infection.
Collapse
Affiliation(s)
- Farzad Fatehi
- Department of Mathematics, University of Sussex, Brighton, United Kingdom
| | | | - Aleksandra Ross
- Department of Mathematics, University of Sussex, Brighton, United Kingdom
| | - Yuliya N Kyrychko
- Department of Mathematics, University of Sussex, Brighton, United Kingdom
| | | |
Collapse
|
25
|
|
26
|
Tocheva AS, Mansour S, Holt TGH, Jones S, Chancellor A, Sanderson JP, Eren E, Elliott TJ, Holt RIG, Gadola SD. The Clonal Invariant NKT Cell Repertoire in People with Type 1 Diabetes Is Characterized by a Loss of Clones Expressing High-Affinity TCRs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:1452-1459. [PMID: 28062695 DOI: 10.4049/jimmunol.1600255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 12/02/2016] [Indexed: 02/11/2024]
Abstract
Invariant NKT (iNKT) cells in healthy people express iNKT-TCRs with widely varying affinities for CD1d, suggesting different roles for high- and low-affinity iNKT clones in immune regulation. However, the functional implications of this heterogeneity have not yet been determined. Functionally aberrant iNKT responses have been previously demonstrated in different autoimmune diseases, including human type 1 diabetes, but their relationship to changes in the iNKT clonal repertoire have not been addressed. In this study, we directly compared the clonal iNKT repertoire of people with recent onset type 1 diabetes and age- and gender-matched healthy controls with regard to iNKT-TCR affinity and cytokine production. Our results demonstrate a selective loss of clones expressing high-affinity iNKT-TCRs from the iNKT repertoire of people with type 1 diabetes. Furthermore, this bias in the clonal iNKT repertoire in type 1 diabetes was associated with increased GM-CSF, IL-4, and IL-13 cytokine secretion among Ag-stimulated low-affinity iNKT clones. Thus, qualitative changes of the clonal iNKT repertoire with the potential to affect the regulatory function of this highly conserved T cell population are already established at the early stages in type 1 diabetes. These findings may inform future rationales for the development of iNKT-based therapies aiming to restore immune tolerance in type 1 diabetes.
Collapse
Affiliation(s)
- Anna S Tocheva
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom;
- Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Salah Mansour
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Tristan G H Holt
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom
| | - Samuel Jones
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom
| | - Andrew Chancellor
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom
| | | | - Efrem Eren
- Department of Clinical Immunology, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom
| | - Tim J Elliott
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Richard I G Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom; and
| | - Stephan D Gadola
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton National Health Service Foundation Trust, Southampton SO17 1BJ, United Kingdom;
- Translational medicine, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| |
Collapse
|
27
|
Chan C, Hyslop CM, Shrivastava V, Ochoa A, Reimer RA, Huang C. Oligofructose as an adjunct in treatment of diabetes in NOD mice. Sci Rep 2016; 6:37627. [PMID: 27874076 PMCID: PMC5118692 DOI: 10.1038/srep37627] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/27/2016] [Indexed: 12/15/2022] Open
Abstract
In type 1 diabetes, restoration of normoglycemia can be achieved if the autoimmune attack on beta cells ceases and insulin requirement is met by the residual beta cells. We hypothesize that an adjunctive therapy that reduces insulin demand by increasing insulin sensitivity will improve the efficacy of an immunotherapy in reversing diabetes. We tested the gut microbiota-modulating prebiotic, oligofructose (OFS), as the adjunctive therapy. We treated non-obese diabetic mice with an immunotherapy, monoclonal anti-CD3 antibody (aCD3), with or without concurrent dietary supplement of OFS. After 8 weeks of OFS supplement, the group that received both aCD3 and OFS (aCD3 + OFS) had a higher diabetes remission rate than the group that received aCD3 alone. The aCD3 + OFS group had higher insulin sensitivity accompanied by reduced lymphocytic infiltrate into the pancreatic islets, higher beta-cell proliferation rate, higher pancreatic insulin content, and secreted more insulin in response to glucose. The addition of OFS also caused a change in gut microbiota, with a higher level of Bifidobacterium and lower Clostridium leptum. Hence, our results suggest that OFS can potentially be an effective therapeutic adjunct in the treatment of type 1 diabetes by improving insulin sensitivity and beta-cell function, leading to improved glycemic control.
Collapse
Affiliation(s)
- Clement Chan
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Colin M Hyslop
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vipul Shrivastava
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrea Ochoa
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Raylene A Reimer
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Carol Huang
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
28
|
Stifter K, Schuster C, Schlosser M, Boehm BO, Schirmbeck R. Exploring the induction of preproinsulin-specific Foxp3(+) CD4(+) Treg cells that inhibit CD8(+) T cell-mediated autoimmune diabetes by DNA vaccination. Sci Rep 2016; 6:29419. [PMID: 27406624 PMCID: PMC4942695 DOI: 10.1038/srep29419] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/14/2016] [Indexed: 12/26/2022] Open
Abstract
DNA vaccination is a promising strategy to induce effector T cells but also regulatory Foxp3+ CD25+ CD4+ Treg cells and inhibit autoimmune disorders such as type 1 diabetes. Little is known about the antigen requirements that facilitate priming of Treg cells but not autoreactive effector CD8+ T cells. We have shown that the injection of preproinsulin (ppins)-expressing pCI/ppins vector into PD-1- or PD-L1-deficient mice induced Kb/A12-21-monospecific CD8+ T cells and autoimmune diabetes. A pCI/ppinsΔA12-21 vector (lacking the critical Kb/A12-21 epitope) did not induce autoimmune diabetes but elicited a systemic Foxp3+ CD25+ Treg cell immunity that suppressed diabetes induction by a subsequent injection of the diabetogenic pCI/ppins. TGF-β expression was significantly enhanced in the Foxp3+ CD25+ Treg cell population of vaccinated/ppins-primed mice. Ablation of Treg cells in vaccinated/ppins-primed mice by anti-CD25 antibody treatment abolished the protective effect of the vaccine and enabled diabetes induction by pCI/ppins. Adoptive transfer of Treg cells from vaccinated/ppins-primed mice into PD-L1−/− hosts efficiently suppressed diabetes induction by pCI/ppins. We narrowed down the Treg-stimulating domain to a 15-residue ppins76–90 peptide. Vaccine-induced Treg cells thus play a crucial role in the control of de novo primed autoreactive effector CD8+ T cells in this diabetes model.
Collapse
Affiliation(s)
- Katja Stifter
- Department of Internal Medicine I, Ulm University Medical Center, Ulm, Germany
| | - Cornelia Schuster
- Department of Internal Medicine I, Ulm University Medical Center, Ulm, Germany
| | - Michael Schlosser
- Department of Medical Biochemistry and Molecular Biology, Research Group of Predictive Diagnostics, University Medical Centre Greifswald, Karlsburg, Germany
| | - Bernhard Otto Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore, Singapore.,Imperial College London, London, UK
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
29
|
Hamel Y, Mauvais FX, Pham HP, Kratzer R, Marchi C, Barilleau É, Waeckel-Enée E, Arnoux JB, Hartemann A, Cordier C, Mégret J, Rocha B, de Lonlay P, Beltrand J, Six A, Robert JJ, van Endert P. A unique CD8(+) T lymphocyte signature in pediatric type 1 diabetes. J Autoimmun 2016; 73:54-63. [PMID: 27318739 DOI: 10.1016/j.jaut.2016.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 12/16/2022]
Abstract
Human type 1 diabetes results from a destructive auto-reactive immune response in which CD8(+) T lymphocytes play a critical role. Given the intense ongoing efforts to develop immune intervention to prevent and/or cure the disease, biomarkers suitable for prediction of disease risk and progress, as well as for monitoring of immunotherapy are required. We undertook separate multi-parameter analyses of single naïve and activated/memory CD8(+) T lymphocytes from pediatric and adult patients, with the objective of identifying cellular profiles associated with onset of type 1 diabetes. We observe global perturbations in gene and protein expression and in the abundance of T cell populations characterizing pediatric but not adult patients, relative to age-matched healthy individuals. Pediatric diabetes is associated with a unique population of CD8(+) T lymphocytes co-expressing effector (perforin, granzyme B) and regulatory (transforming growth factor β, interleukin-10 receptor) molecules. This population persists after metabolic normalization and is especially abundant in children with high titers of auto-antibodies to glutamic acid decarboxylase and with elevated HbA1c values. These findings highlight striking differences between pediatric and adult type 1 diabetes, indicate prolonged large-scale perturbations in the CD8(+) T cell compartment in the former, and suggest that CD8(+)CD45RA(-) T cells co-expressing effector and regulatory factors are of interest as biomarkers in pediatric type 1 diabetes.
Collapse
Affiliation(s)
- Yamina Hamel
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - François-Xavier Mauvais
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Hang-Phuong Pham
- Sorbonne Universités, UPMC Université Paris 6, 75015 Paris, France; Institut National de la Sante et de la Recherche Médicale, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Roland Kratzer
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Christophe Marchi
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Émilie Barilleau
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Emmanuelle Waeckel-Enée
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Jean-Baptiste Arnoux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre de référence des Maladies Héréditaires du Métabolisme, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Agnès Hartemann
- Université Pierre & Marie Curie, IHU ICAN, 75013 Paris, France; Service de Diabétologie, Hôpital de la Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, 75013 Paris, France
| | - Corinne Cordier
- Institut National de la Sante et de la Recherche Médicale, US24, 75015 Paris, France; Centre National de la Recherche Scientifique, UMS3633, 75015 Paris, France
| | - Jerome Mégret
- Institut National de la Sante et de la Recherche Médicale, US24, 75015 Paris, France; Centre National de la Recherche Scientifique, UMS3633, 75015 Paris, France
| | - Benedita Rocha
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France
| | - Pascale de Lonlay
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre de référence des Maladies Héréditaires du Métabolisme, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France; Institut Imagine, Institut National de la Sante et de la Recherche Médicale, Unité 1163, 75015 Paris, France
| | - Jacques Beltrand
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Adrien Six
- Sorbonne Universités, UPMC Université Paris 6, 75015 Paris, France; Institut National de la Sante et de la Recherche Médicale, UMRS 959, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Jean-Jacques Robert
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Peter van Endert
- Institut National de la Sante et de la Recherche Médicale, Unité 1151, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, 75015 Paris, France; Centre National de la Recherche Scientifique, UMR8253, 75015 Paris, France.
| |
Collapse
|
30
|
Clemente-Casares X, Blanco J, Ambalavanan P, Yamanouchi J, Singha S, Fandos C, Tsai S, Wang J, Garabatos N, Izquierdo C, Agrawal S, Keough MB, Yong VW, James E, Moore A, Yang Y, Stratmann T, Serra P, Santamaria P. Expanding antigen-specific regulatory networks to treat autoimmunity. Nature 2016; 530:434-40. [PMID: 26886799 DOI: 10.1038/nature16962] [Citation(s) in RCA: 379] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/24/2015] [Indexed: 12/31/2022]
Abstract
Regulatory T cells hold promise as targets for therapeutic intervention in autoimmunity, but approaches capable of expanding antigen-specific regulatory T cells in vivo are currently not available. Here we show that systemic delivery of nanoparticles coated with autoimmune-disease-relevant peptides bound to major histocompatibility complex class II (pMHCII) molecules triggers the generation and expansion of antigen-specific regulatory CD4(+) T cell type 1 (TR1)-like cells in different mouse models, including mice humanized with lymphocytes from patients, leading to resolution of established autoimmune phenomena. Ten pMHCII-based nanomedicines show similar biological effects, regardless of genetic background, prevalence of the cognate T-cell population or MHC restriction. These nanomedicines promote the differentiation of disease-primed autoreactive T cells into TR1-like cells, which in turn suppress autoantigen-loaded antigen-presenting cells and drive the differentiation of cognate B cells into disease-suppressing regulatory B cells, without compromising systemic immunity. pMHCII-based nanomedicines thus represent a new class of drugs, potentially useful for treating a broad spectrum of autoimmune conditions in a disease-specific manner.
Collapse
Affiliation(s)
- Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jesus Blanco
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Poornima Ambalavanan
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jun Yamanouchi
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Santiswarup Singha
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Cesar Fandos
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
| | - Sue Tsai
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jinguo Wang
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Nahir Garabatos
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Cristina Izquierdo
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Smriti Agrawal
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Michael B Keough
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Eddie James
- Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101-2795, USA
| | - Anna Moore
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Thomas Stratmann
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona 08028, Spain
| | - Pau Serra
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC), and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
| |
Collapse
|
31
|
Yu C, Burns JC, Robinson WH, Utz PJ, Ho PP, Steinman L, Frey AB. Identification of Candidate Tolerogenic CD8(+) T Cell Epitopes for Therapy of Type 1 Diabetes in the NOD Mouse Model. J Diabetes Res 2016; 2016:9083103. [PMID: 27069933 PMCID: PMC4812430 DOI: 10.1155/2016/9083103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease in which insulin-producing pancreatic islet β cells are the target of self-reactive B and T cells. T cells reactive with epitopes derived from insulin and/or IGRP are critical for the initiation and maintenance of disease, but T cells reactive with other islet antigens likely have an essential role in disease progression. We sought to identify candidate CD8(+) T cell epitopes that are pathogenic in type 1 diabetes. Proteins that elicit autoantibodies in human type 1 diabetes were analyzed by predictive algorithms for candidate epitopes. Using several different tolerizing regimes using synthetic peptides, two new predicted tolerogenic CD8(+) T cell epitopes were identified in the murine homolog of the major human islet autoantigen zinc transporter ZnT8 (aa 158-166 and 282-290) and one in a non-β cell protein, dopamine β-hydroxylase (aa 233-241). Tolerizing vaccination of NOD mice with a cDNA plasmid expressing full-length proinsulin prevented diabetes, whereas plasmids encoding ZnT8 and DβH did not. However, tolerizing vaccination of NOD mice with the proinsulin plasmid in combination with plasmids expressing ZnT8 and DβH decreased insulitis and enhanced prevention of disease compared to vaccination with the plasmid encoding proinsulin alone.
Collapse
MESH Headings
- Animals
- Autoantibodies/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cation Transport Proteins/genetics
- Cation Transport Proteins/immunology
- Cells, Cultured
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Dopamine beta-Hydroxylase/genetics
- Dopamine beta-Hydroxylase/immunology
- Epitopes, T-Lymphocyte
- Female
- Genetic Therapy/methods
- Humans
- Immune Tolerance
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/pathology
- Lymphocyte Activation
- Mice, Inbred NOD
- Proinsulin/genetics
- Proinsulin/immunology
- Time Factors
- Vaccination
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Zinc Transporter 8
Collapse
Affiliation(s)
- Cailin Yu
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Jeremy C. Burns
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs, Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Paul J. Utz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peggy P. Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan B. Frey
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
- *Alan B. Frey:
| |
Collapse
|
32
|
Affiliation(s)
- Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
33
|
Hyslop CM, Tsai S, Shrivastava V, Santamaria P, Huang C. Prolactin as an Adjunct for Type 1 Diabetes Immunotherapy. Endocrinology 2016; 157:150-65. [PMID: 26512750 DOI: 10.1210/en.2015-1549] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes is caused by autoimmune destruction of β-cells. Although immunotherapy can restore self-tolerance thereby halting continued immune-mediated β-cell loss, residual β-cell mass and function is often insufficient for normoglycemia. Using a growth factor to boost β-cell mass can potentially overcome this barrier and prolactin (PRL) may fill this role. Previous studies have shown that PRL can stimulate β-cell proliferation and up-regulate insulin synthesis and secretion while reducing lymphocytic infiltration of islets, suggesting that it may restore normoglycemia through complementary mechanisms. Here, we test the hypothesis that PRL can improve the efficacy of an immune modulator, the anticluster of differentiation 3 monoclonal antibody (aCD3), in inducing diabetes remission by up-regulating β-cell mass and function. Diabetic nonobese diabetic (NOD) mice were treated with a 5-day course of aCD3 with or without a concurrent 3-week course of PRL. We found that a higher proportion of diabetic mice treated with the aCD3 and PRL combined therapy achieved diabetes reversal than those treated with aCD3 alone. The aCD3 and PRL combined group had a higher β-cell proliferation rate, an increased β-cell fraction, larger islets, higher pancreatic insulin content, and greater glucose-stimulated insulin release. Lineage-tracing analysis found minimal contribution of β-cell neogenesis to the formation of new β-cells. Although we did not detect a significant difference in the number or proliferative capacity of T cells, we observed a higher proportion of insulitis-free islets in the aCD3 and PRL group. These results suggest that combining a growth factor with an immunotherapy may be an effective treatment paradigm for autoimmune diabetes.
Collapse
Affiliation(s)
- Colin M Hyslop
- Department of Biochemistry and Molecular Biology (C.M.H., V.S., C.H.), Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Julia McFarlane Diabetes Research Centre and Department of Microbiology, Immunology and Infectious Diseases (S.T., P.S.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Institut D'Investigacions Biomediques August Pi i Sunyer (P.S.), 08036 Barcelona, Spain; and Department of Pediatrics (C.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Sue Tsai
- Department of Biochemistry and Molecular Biology (C.M.H., V.S., C.H.), Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Julia McFarlane Diabetes Research Centre and Department of Microbiology, Immunology and Infectious Diseases (S.T., P.S.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Institut D'Investigacions Biomediques August Pi i Sunyer (P.S.), 08036 Barcelona, Spain; and Department of Pediatrics (C.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Vipul Shrivastava
- Department of Biochemistry and Molecular Biology (C.M.H., V.S., C.H.), Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Julia McFarlane Diabetes Research Centre and Department of Microbiology, Immunology and Infectious Diseases (S.T., P.S.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Institut D'Investigacions Biomediques August Pi i Sunyer (P.S.), 08036 Barcelona, Spain; and Department of Pediatrics (C.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Pere Santamaria
- Department of Biochemistry and Molecular Biology (C.M.H., V.S., C.H.), Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Julia McFarlane Diabetes Research Centre and Department of Microbiology, Immunology and Infectious Diseases (S.T., P.S.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Institut D'Investigacions Biomediques August Pi i Sunyer (P.S.), 08036 Barcelona, Spain; and Department of Pediatrics (C.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | - Carol Huang
- Department of Biochemistry and Molecular Biology (C.M.H., V.S., C.H.), Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Julia McFarlane Diabetes Research Centre and Department of Microbiology, Immunology and Infectious Diseases (S.T., P.S.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1; Institut D'Investigacions Biomediques August Pi i Sunyer (P.S.), 08036 Barcelona, Spain; and Department of Pediatrics (C.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| |
Collapse
|
34
|
Carrascal J, Carrillo J, Arpa B, Egia-Mendikute L, Rosell-Mases E, Pujol-Autonell I, Planas R, Mora C, Mauricio D, Ampudia RM, Vives-Pi M, Verdaguer J. B-cell anergy induces a Th17 shift in a novel B lymphocyte transgenic NOD mouse model, the 116C-NOD mouse. Eur J Immunol 2015; 46:593-608. [PMID: 26639224 DOI: 10.1002/eji.201445376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 11/09/2015] [Accepted: 12/01/2015] [Indexed: 11/06/2022]
Abstract
Autoreactive B lymphocytes play a key role as APCs in diaebetogenesis. However, it remains unclear whether B-cell tolerance is compromised in NOD mice. Here, we describe a new B lymphocyte transgenic NOD mouse model, the 116C-NOD mouse, where the transgenes derive from an islet-infiltrating B lymphocyte of a (8.3-NODxNOR) F1 mouse. The 116C-NOD mouse produces clonal B lymphocytes with pancreatic islet beta cell specificity. The incidence of T1D in 116C-NOD mice is decreased in both genders when compared with NOD mice. Moreover, several immune selection mechanisms (including clonal deletion and anergy) acting on the development, phenotype, and function of autoreactive B lymphocytes during T1D development have been identified in the 116C-NOD mouse. Surprisingly, a more accurate analysis revealed that, despite their anergic phenotype, 116C B cells express some costimulatory molecules after activation, and induce a T-cell shift toward a Th17 phenotype. Furthermore, this shift on T lymphocytes seems to occur not only when both T and B cells contact, but also when helper T (Th) lineage is established. The 116C-NOD mouse model could be useful to elucidate the mechanisms involved in the generation of Th-cell lineages.
Collapse
Affiliation(s)
- Jorge Carrascal
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Jorge Carrillo
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Berta Arpa
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Leire Egia-Mendikute
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Estela Rosell-Mases
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Irma Pujol-Autonell
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Raquel Planas
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Conchi Mora
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Dídac Mauricio
- Department of Endocrinology, Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Maria Ampudia
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Department, Institut d'Investigacio Germans Trias i Pujol, Badalona, Barcelona, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Verdaguer
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
35
|
Lewis JS, Dolgova NV, Zhang Y, Xia CQ, Wasserfall CH, Atkinson MA, Clare-Salzler MJ, Keselowsky BG. A combination dual-sized microparticle system modulates dendritic cells and prevents type 1 diabetes in prediabetic NOD mice. Clin Immunol 2015; 160:90-102. [PMID: 25842187 DOI: 10.1016/j.clim.2015.03.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/16/2015] [Accepted: 03/24/2015] [Indexed: 12/17/2022]
Abstract
We developed a novel poly(lactic-co-glycolic acid)-based, microparticle (MP) system providing concurrent delivery of multiple encapsulated immuno-suppressive factors and antigen, for in vivo conditioning of dendritic cells (DCs) toward a tolerance promoting pathway. Subcutaneous administration prevents onset of type 1 diabetes (T1D) in NOD mice. Two MP sizes were made: phagocytosable MPs were fabricated encapsulating vitamin D3 or insulin B(9-23) peptide, while unphagocytosable MPs were fabricated encapsulating TGF-β1 or GM-CSF. The combination of Vit D3/TGF-β1 MPs confers an immature and LPS activation-resistant phenotype to DCs, and MP-delivered antigen is efficiently and functionally presented. Notably, two subcutaneous injections into 4week old NOD mice using the combination of MPs encapsulating Vit D3, Ins B, TGF-β1 and GM-CSF protected 40% of mice from T1D development, significant in comparison to the control. This work represents one of the first applications of a biomaterial-based, MP vaccine system to successfully prevent autoimmune diabetes.
Collapse
Affiliation(s)
- Jamal S Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Natalia V Dolgova
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ying Zhang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Chang Qing Xia
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
36
|
Hansen MP, Matheis N, Kahaly GJ. Type 1 diabetes and polyglandular autoimmune syndrome: A review. World J Diabetes 2015; 6:67-79. [PMID: 25685279 PMCID: PMC4317318 DOI: 10.4239/wjd.v6.i1.67] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/11/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder caused by inflammatory destruction of the pancreatic tissue. The etiopathogenesis and characteristics of the pathologic process of pancreatic destruction are well described. In addition, the putative susceptibility genes for T1D as a monoglandular disease and the relation to polyglandular autoimmune syndrome (PAS) have also been well explored. The incidence of T1D has steadily increased in most parts of the world, especially in industrialized nations. T1D is frequently associated with autoimmune endocrine and non-endocrine diseases and patients with T1D are at a higher risk for developing several glandular autoimmune diseases. Familial clustering is observed, which suggests that there is a genetic predisposition. Various hypotheses pertaining to viral- and bacterial-induced pancreatic autoimmunity have been proposed, however a definitive delineation of the autoimmune pathomechanism is still lacking. In patients with PAS, pancreatic and endocrine autoantigens either colocalize on one antigen-presenting cell or are expressed on two/various target cells sharing a common amino acid, which facilitates binding to and activation of T cells. The most prevalent PAS phenotype is the adult type 3 variant or PAS type III, which encompasses T1D and autoimmune thyroid disease. This review discusses the findings of recent studies showing noticeable differences in the genetic background and clinical phenotype of T1D either as an isolated autoimmune endocrinopathy or within the scope of polyglandular autoimmune syndrome.
Collapse
|
37
|
Wang J, Nanjundappa RH, Shameli A, Clemente-Casares X, Yamanouchi J, Elliott JF, Slattery R, Serra P, Santamaria P. The cross-priming capacity and direct presentation potential of an autoantigen are separable and inversely related properties. THE JOURNAL OF IMMUNOLOGY 2014; 193:3296-307. [PMID: 25165150 DOI: 10.4049/jimmunol.1401001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We investigated whether a prevalent epitope of the β-cell-specific autoantigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP206-214) reaches regional Ag-presentation pathways via unprocessed polypeptide chains, as free IGRP206-214 peptide or via preformed IGRP206-214/K(d) complexes. This was accomplished by expressing bacterial artificial chromosome transgenes encoding wild-type (stable) or ubiquitinated (unstable) forms of IGRP in IGRP-deficient NOD mice carrying MHC class I-deficient β-cells, dendritic cells, or B cells. We investigated the ability of the pancreatic lymph nodes of these mice to prime naive IGRP206-214-reactive CD8(+) T cells in vivo, either in response to spontaneous Ag shedding, or to synchronized forms of β-cell necrosis or apoptosis. When IGRP was made unstable by targeting it for proteasomal degradation within β-cells, the cross-priming, autoimmune-initiating potential of this autoantigen (designated autoantigenicity) was impaired. Yet at the same time, the direct presentation, CTL-targeting potential of IGRP (designated pathogenicity) was enhanced. The appearance of IGRP206-214 in regional Ag-presentation pathways was dissociated from transfer of IGRP206-214 or IGRP206-214/K(d) from β cells to dendritic cells. These results indicate that autoantigenicity and pathogenicity are separable and inversely related properties and suggest that pathogenic autoantigens, capable of efficiently priming CTLs while marking target cells for CTL-induced killing, may have a critical balance of these two properties.
Collapse
Affiliation(s)
- Jinguo Wang
- Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Roopa Hebbandi Nanjundappa
- Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Afshin Shameli
- Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jun Yamanouchi
- Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - John F Elliott
- Alberta Diabetes Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; Division of Dermatology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Robyn Slattery
- Department of Immunology, Monash University, Alfred Hospital Medical Research and Education Precinct, Melbourne, Victoria 3004, Australia; and
| | - Pau Serra
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona 08036, Spain
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona 08036, Spain
| |
Collapse
|
38
|
Craig K, d'Agostino C, Poratt D, Walker M. Original hypothesis: Extracorporeal shockwaves as a homeostatic autoimmune restorative treatment (HART) for Type 1 diabetes mellitus. Med Hypotheses 2014; 83:250-3. [PMID: 24947195 DOI: 10.1016/j.mehy.2014.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/07/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
Mononuclear invasion of Langerhans islet and the ensuing insulitis triggers signal-transduction for the autoimmune mediated pancreatic beta-cell (β-cell) apoptosis that severely disrupts insulin production resulting in hyperglycemia associated with Type-1 diabetes (T1DM). Today extensive global research is being conducted to eliminate the need for insulin, and even prevent or find a cure for T1DM. The multifactorial combination of autoimmune dysfunction, Langerhans islet hypoxia, and bio-chemical disruption are seen to be contributory factors for β-cell destruction and the consequential disruption to insulin production. Regeneration of β-cells back to physiological levels may restore homeostatic insulin levels, reversing T1DM. Evidence suggests that there are still functioning pancreatic β-cells even in long standing T1DM providing the potential for their regeneration. Although the exact mechanism of extracorporeal shockwaves (ESW) is yet to be fully elucidated, it is seen to influence a complex spectrum of bio-chemical, cellular and neuronal functions (i.e. suppression of pro-inflammatory immune response, improved tissue hemodynamics, anti-microbial properties, and the induction of progenitor cell expression including proangiogenic factors and nitric oxide syntheses). The rationale for the use of ESW as a therapeutic modality in this instance is attributed to its restorative properties and safety profile demonstrated in urology, cardiology, chronic wounds, osteogenesis, complex pain syndromes, and tendinopathies. ESW may restore autoimmune homeostasis creating a suitable environment for pancreatic β-cell proliferation which in-turn may significantly increase or normalize endogenous insulin secretion reducing or totally eliminating dependency of exogenous insulin. The devastating complications, morbidity and mortality associated with T1DM warrants the exploration of homeostatic autoimmune restorative treatment (HART) modalities that may partially or fully reverse this disease condition. We present our hypothesis discussing ESW as a potential homeostatic autoimmune restorative treatment (HART) option for T1DM.
Collapse
Affiliation(s)
| | - Cristina d'Agostino
- Shock Wave Therapy Research Unit, Rehabilitation Department, Instituto Clinico Humanitas, Milan, Italy. cristina.d'
| | - Daniel Poratt
- Auckland University of Technology, Auckland, New Zealand.
| | | |
Collapse
|
39
|
Holmdahl R, Malmström V, Burkhardt H. Autoimmune priming, tissue attack and chronic inflammation - the three stages of rheumatoid arthritis. Eur J Immunol 2014; 44:1593-9. [PMID: 24737176 DOI: 10.1002/eji.201444486] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 02/27/2014] [Accepted: 04/10/2014] [Indexed: 12/28/2022]
Abstract
Extensive genome-wide association studies have recently shed some light on the causes of chronic autoimmune diseases and have confirmed a central role of the adaptive immune system. Moreover, better diagnostics using disease-associated autoantibodies have been developed, and treatment has improved through the development of biologicals with precise molecular targets. Here, we use rheumatoid arthritis (RA) as a prototype for chronic autoimmune disease to propose that the pathogenesis of autoimmune diseases could be divided into three discrete stages. First, yet unknown environmental challenges seem to activate innate immunity thereby providing an adjuvant signal for the induction of adaptive immune responses that lead to the production of autoantibodies and determine the subsequent disease development. Second, a joint-specific inflammatory reaction occurs. This inflammatory reaction might be clinically diagnosed as the earliest signs of the disease. Third, inflammation is converted to a chronic process leading to tissue destruction and remodeling. In this review, we discuss the stages involved in RA pathogenesis and the experimental approaches, mainly involving animal models that can be used to investigate each disease stage. Although we focus on RA, it is possible that a similar stepwise development of disease also occurs in other chronic autoimmune settings such as multiple sclerosis (MS), type 1 diabetes, and systemic lupus erythematosus.
Collapse
Affiliation(s)
- Rikard Holmdahl
- Department of Medical Biochemistry and Biophysics, Medical Inflammation Research, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
40
|
Li M, Song LJ, Qin XY. Advances in the cellular immunological pathogenesis of type 1 diabetes. J Cell Mol Med 2014; 18:749-58. [PMID: 24629100 PMCID: PMC4119381 DOI: 10.1111/jcmm.12270] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/30/2014] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of insulin-producing pancreatic β cells. In recent years, the incidence of type 1 diabetes continues to increase. It is supposed that genetic, environmental and immune factors participate in the damage of pancreatic β cells. Both the immune regulation and the immune response are involved in the pathogenesis of type 1 diabetes, in which cellular immunity plays a significant role. For the infiltration of CD4(+) and CD8(+) T lymphocyte, B lymphocytes, natural killer cells, dendritic cells and other immune cells take part in the damage of pancreatic β cells, which ultimately lead to type 1 diabetes. This review outlines the cellular immunological mechanism of type 1 diabetes, with a particular emphasis to T lymphocyte and natural killer cells, and provides the effective immune therapy in T1D, which is approached at three stages. However, future studies will be directed at searching for an effective, safe and long-lasting strategy to enhance the regulation of a diabetogenic immune system with limited toxicity and without global immunosuppression.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Lu-Jun Song
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Xin-Yu Qin
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
41
|
A missing PD-L1/PD-1 coinhibition regulates diabetes induction by preproinsulin-specific CD8 T-cells in an epitope-specific manner. PLoS One 2013; 8:e71746. [PMID: 23977133 PMCID: PMC3747217 DOI: 10.1371/journal.pone.0071746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/02/2013] [Indexed: 11/19/2022] Open
Abstract
Coinhibitory PD-1/PD-L1 (B7-H1) interactions provide critical signals for the regulation of autoreactive T-cell responses. We established mouse models, expressing the costimulator molecule B7.1 (CD80) on pancreatic beta cells (RIP-B7.1 tg mice) or are deficient in coinhibitory PD-L1 or PD-1 molecules (PD-L1−/− and PD-1−/− mice), to study induction of preproinsulin (ppins)-specific CD8 T-cell responses and experimental autoimmune diabetes (EAD) by DNA-based immunization. RIP-B7.1 tg mice allowed us to identify two CD8 T-cell specificities: pCI/ppins DNA exclusively induced Kb/A12–21-specific CD8 T-cells and EAD, whereas pCI/ppinsΔA12–21 DNA (encoding ppins without the COOH-terminal A12–21 epitope) elicited Kb/B22–29-specific CD8 T-cells and EAD. Specific expression/processing of mutant ppinsΔA12–21 (but not ppins) in non-beta cells, targeted by intramuscular DNA-injection, thus facilitated induction of Kb/B22–29-specific CD8 T-cells. The A12–21 epitope binds Kb molecules with a very low avidity as compared with B22–29. Interestingly, immunization of coinhibition-deficient PD-L1−/− or PD-1−/− mice with pCI/ppins induced Kb/A12–21-monospecific CD8 T-cells and EAD but injections with pCI/ppinsΔA12–21 did neither recruit Kb/B22–29-specific CD8 T-cells into the pancreatic target tissue nor induce EAD. PpinsΔA12–21/(Kb/B22–29)-mediated EAD was efficiently restored in RIP-B7.1+/PD-L1−/− mice, differing from PD-L1−/− mice only in the tg B7.1 expression in beta cells. Alternatively, an ongoing beta cell destruction and tissue inflammation, initiated by ppins/(Kb/A12–21)-specific CD8 T-cells in pCI/ppins+pCI/ppinsΔA12–21 co-immunized PD-L1−/− mice, facilitated the expansion of ppinsΔA12–21/(Kb/B22–29)-specific CD8 T-cells. CD8 T-cells specific for the high-affinity Kb/B22–29- (but not the low-affinity Kb/A12–21)-epitope thus require stimulatory ´help from beta cells or inflamed islets to expand in PD-L1-deficient mice. The new PD-1/PD-L1 diabetes models may be valuable tools to study under well controlled experimental conditions distinct hierarchies of autoreactive CD8 T-cell responses, which trigger the initial steps of beta cell destruction or emerge during the pathogenic progression of EAD.
Collapse
|
42
|
Naser SA, Thanigachalam S, Dow CT, Collins MT. Exploring the role of Mycobacterium avium subspecies paratuberculosis in the pathogenesis of type 1 diabetes mellitus: a pilot study. Gut Pathog 2013; 5:14. [PMID: 23759115 PMCID: PMC3686596 DOI: 10.1186/1757-4749-5-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/07/2013] [Indexed: 12/11/2022] Open
Abstract
Background Although the etiology of Type 1 Diabetes mellitus (T1DM) has not been determined, genetic polymorphism in key genes, including SLC11A1, and association with Mycobacterium avium subspecies paratuberculosis (MAP) have been reported. We hypothesize that molecular mimicry between MAP Heat shock protein 65 K (Hsp65) and human Glutamic Acid Decarboxylase 65 K (GAD65) may be the trigger leading to autoimmune destruction of beta cells in patients exposed to MAP. Method Peptide sequences of MAP Hsp65 and human GAD65 were investigated for amino acid sequence homology and cross reactivity. A total of 18 blood samples from T1DM and controls were evaluated for the presence of MAP. Results Peptide BLAST analysis revealed a 44% overall identity between MAP Hsp65 and GAD65 with 75% positives in a 16 amino acid region. PyMOL 3D-structural analyses identified the same 16 amino acid region as a potential epitope for antibody binding. Preliminary data suggests a cross reactivity between MAP Hsp65, and a healthy rat pancreatic tissue homogenate against plasma from T1DM patients and rabbit polyclonal anti-MAP IgG. Long-term culture of human blood resulted MAP detection in 3/10 T1DM and 4/8 controls whereas MAP IgG was detected in 5/10 T1DM samples and 3/8 non-diabetic controls. Conclusion The high degree of homology between GAD65 and MAP Hsp65 in an antigenic peptide region supports a possible mycobacterial role in triggering autoimmune destruction of pancreatic cells in T1DM. Reactivity of T1DM patient sera with MAP Hsp65 supports this finding. Culture of MAP from the blood of T1DM patients is intriguing. Overall, the preliminary data are mixed and do not exclude a possible role for MAP in T1DM pathogenesis. A larger study including well-characterized controls is needed to investigate the intriguing question of whether MAP is associated with T1DM or not?
Collapse
Affiliation(s)
- Saleh A Naser
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32816, USA
| | - Saisathya Thanigachalam
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32816, USA
| | - C Thomas Dow
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI, USA
| | - Michael T Collins
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
43
|
Carrero JA, Calderon B, Towfic F, Artyomov MN, Unanue ER. Defining the transcriptional and cellular landscape of type 1 diabetes in the NOD mouse. PLoS One 2013; 8:e59701. [PMID: 23555752 PMCID: PMC3608568 DOI: 10.1371/journal.pone.0059701] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/17/2013] [Indexed: 01/12/2023] Open
Abstract
Our ability to successfully intervene in disease processes is dependent on definitive diagnosis. In the case of autoimmune disease, this is particularly challenging because progression of disease is lengthy and multifactorial. Here we show the first chronological compendium of transcriptional and cellular signatures of diabetes in the non-obese diabetic mouse. Our data relates the immunological environment of the islets of Langerhans with the transcriptional profile at discrete times. Based on these data, we have parsed diabetes into several discrete phases. First, there is a type I interferon signature that precedes T cell activation. Second, there is synchronous infiltration of all immunological cellular subsets and a period of control. Finally, there is the killing phase of the diabetogenic process that is correlated with an NF-kB signature. Our data provides a framework for future examination of autoimmune diabetes and its disease progression markers.
Collapse
Affiliation(s)
- Javier A Carrero
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America.
| | | | | | | | | |
Collapse
|
44
|
Mesenchymal stromal cells as a means of controlling pathological T-cell responses in allogeneic islet transplantation. Curr Opin Organ Transplant 2013; 18:59-64. [DOI: 10.1097/mot.0b013e32835c2adf] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
45
|
Shameli A, Yamanouchi J, Tsai S, Yang Y, Clemente-Casares X, Moore A, Serra P, Santamaria P. IL-2 promotes the function of memory-like autoregulatory CD8+T cells but suppresses their development via FoxP3+Treg cells. Eur J Immunol 2013. [DOI: 10.1002/eji.201242845] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Afshin Shameli
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Jun Yamanouchi
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Sue Tsai
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
- Department of Biochemistry and Molecular Biology; Faculty of Medicine, the University of Calgary; Calgary AB Canada
| | - Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Anna Moore
- Molecular Imaging Laboratory; MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging; Department of Radiology; Massachusetts General Hospital; Charlestown MA USA
| | - Pau Serra
- Institut d'Investigacions Biomédiques August Pi i Sunyer - Hospital Clinic de Barcelona; Centre Esther Koplowitz Barcelona Spain
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
- Institut d'Investigacions Biomédiques August Pi i Sunyer - Hospital Clinic de Barcelona; Centre Esther Koplowitz Barcelona Spain
| |
Collapse
|
46
|
Vaughan K, Peters B, Mallone R, von Herrath M, Roep BO, Sette A. Navigating diabetes-related immune epitope data: resources and tools provided by the Immune Epitope Database (IEDB). Immunome Res 2013; 9. [PMID: 25140192 PMCID: PMC4134942 DOI: 10.4172/1745-7580.1000063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background The Immune Epitope Database (IEDB), originally focused on infectious diseases, was recently expanded to allergy, transplantation and autoimmunity diseases. Here we focus on diabetes, chosen as a prototype autoimmune disease. We utilize a combined tutorial and meta-analysis format, which demonstrates how common questions, related to diabetes epitopes can be answered. Results A total of 409 references are captured in the IEDB describing >2,500 epitopes from diabetes associated antigens. The vast majority of data were derived from GAD, insulin, IA-2/PTPRN, IGRP, ZnT8, HSP, and ICA-1, and the experiments related to T cell epitopes and MHC binding far outnumbers B cell assays. We illustrate how to search by specific antigens, epitopes or host. Other examples include searching for tetramers or epitopes restricted by specific alleles or assays of interest, or searching based on the clinical status of the host. Conclusions The inventory of all published diabetes epitope data facilitates its access for the scientific community. While the global collection of primary data from the literature reflects potential investigational biases present in the literature, the flexible search approach allows users to perform queries tailored to their preferences, including or excluding data as appropriate. Moreover, the analysis highlights knowledge gaps and identifies areas for future investigation.
Collapse
Affiliation(s)
- Kerrie Vaughan
- Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Bjoern Peters
- Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Roberto Mallone
- INSERM, U1016, Cochin Institute, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 82 Avenue Denfert Rochereau, 75674 Paris Cedex 14, France
| | - Matthias von Herrath
- Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Bart O Roep
- Department for Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Alessandro Sette
- Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| |
Collapse
|
47
|
Blyuss K, Nicholson L. The role of tunable activation thresholds in the dynamics of autoimmunity. J Theor Biol 2012; 308:45-55. [DOI: 10.1016/j.jtbi.2012.05.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 11/27/2022]
|
48
|
Abstract
PURPOSE OF REVIEW Regulatory T cells (Treg) maintain immune homeostasis and prevent autoimmune disease. This review summarizes the recent advances in Treg knowledge relevant to type 1 diabetes, focusing on Treg signature, antigen specificity and development and function in the face of inflammation. RECENT FINDINGS Thymus-derived natural regulatory T cells (nTreg) programmed by the transcription factor forkhead box P3 (FOXP3) and peripheral-induced regulatory T cells (iTreg) have largely nonoverlapping T-cell receptor repertoires to self-antigens and jointly contribute to immune homeostasis. Initial reports that CD4CD25 (FOXP3) Treg were impaired in frequency or function in type 1 diabetes have not been confirmed. The Treg-specific demethylated region in the FOXP3 locus in nTreg is, in contrast, methylated in iTreg and conventional T cells (Tconv) and is the only feature that reliably distinguishes activated human nTreg and Tconv. Inflammatory cytokines regulate extrathymic differentiation of nTreg but can also reprogram nTreg into Th17 or Th1 effectors and prevent the differentiation of iTreg. SUMMARY The methylation status of the FOXP3 locus provides a means to re-examine Treg in autoimmune disease. nTreg and iTreg recognize different self-antigens. Shaping of Treg by the cytokine milieu has implications for the application of Treg cell-based immune therapies.
Collapse
Affiliation(s)
- Yuxia Zhang
- The Walter and Eliza Hall Institute of Medical Research, The University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
49
|
Énée É, Kratzer R, Arnoux JB, Barilleau E, Hamel Y, Marchi C, Beltrand J, Michaud B, Chatenoud L, Robert JJ, van Endert P. ZnT8 is a major CD8+ T cell-recognized autoantigen in pediatric type 1 diabetes. Diabetes 2012; 61:1779-84. [PMID: 22586580 PMCID: PMC3379659 DOI: 10.2337/db12-0071] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes results from the destruction of β-cells by an autoimmune T-cell response assisted by antigen-presenting B cells producing autoantibodies. CD8(+) T-cell responses against islet cell antigens, thought to play a central role in diabetes pathogenesis, can be monitored using enzyme-linked immunosorbent spot (ELISpot) assays. However, such assays have been applied to monitoring of adult patients only, leaving aside the large and increasing pediatric patient population. The objective of this study was twofold: 1) to develop a CD8(+) T-cell interferon-γ ELISpot assay for pediatric patients and 2) to determine whether zinc transporter 8 (ZnT8), a recently described target of autoantibodies in a majority of patients, is also recognized by autoreactive CD8(+) T cells. Using DNA immunization of humanized mice, we identified nine HLA-A2-restricted ZnT8 epitopes. Among 36 HLA-A2(+) children with diabetes, 29 responded to ZnT8 epitopes, whereas only 3 of 16 HLA-A2(+) control patients and 0 of 17 HLA-A2(-) control patients responded. Some single ZnT8 epitopes performed as well as the group of epitopes in discriminating between patients and control individuals. Thus, ZnT8 is a major CD8(+) T-cell autoantigen, and ELISpot assays display similar performance in adult and pediatric type 1 diabetes.
Collapse
Affiliation(s)
- Émmanuelle Énée
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Roland Kratzer
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Jean-Baptiste Arnoux
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
- Hôpital Necker, Service d’endocrinologie, Unité fonctionnelle diabétologie, Paris, France
| | - Emilie Barilleau
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Yamina Hamel
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Christophe Marchi
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Jacques Beltrand
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
- Hôpital Necker, Service d’endocrinologie, Unité fonctionnelle diabétologie, Paris, France
| | - Bénédicte Michaud
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Lucienne Chatenoud
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
| | - Jean-Jacques Robert
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
- Hôpital Necker, Service d’endocrinologie, Unité fonctionnelle diabétologie, Paris, France
| | - Peter van Endert
- INSERM, Unité 1013, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine, Paris, France
- Corresponding author: Peter van Endert,
| |
Collapse
|
50
|
Anti-IL-7 receptor-α reverses established type 1 diabetes in nonobese diabetic mice by modulating effector T-cell function. Proc Natl Acad Sci U S A 2012; 109:12674-9. [PMID: 22733769 DOI: 10.1073/pnas.1203795109] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Genetic variation in the IL-7 receptor-α (IL-7R) gene is associated with susceptibility to human type 1 diabetes (T1D). Here we investigate the therapeutic efficacy and mechanism of IL-7Rα antibody in a mouse model of T1D. IL-7Rα antibody induces durable, complete remission in newly onset diabetic mice after only two to three injections. IL-7 increases, whereas IL-7Rα antibody therapy reduces, the IFN-γ-producing CD4(+) (T(H)1) and IFN-γ-producing CD8(+) T cells. Conversely, IL-7 decreases and IL-7Rα antibody enhances the inhibitory receptor Programmed Death 1 (PD-1) expression in the effector T cells. Programmed Death 1 blockade reversed the immune tolerance mediated by the IL-7Rα antibody therapy. Furthermore, IL-7Rα antibody therapy increases the frequency of regulatory T cells without affecting their suppressor activity. The durable efficacy and the multipronged tolerogenic mechanisms of IL-7Rα antibody therapy suggest a unique disease-modifying approach to T1D.
Collapse
|