1
|
Caicedo HH, Darrow JJ, Caicedo JC, Pentland A. Prioritizing Early Disease Intervention. Ther Innov Regul Sci 2023; 57:1148-1152. [PMID: 37668879 DOI: 10.1007/s43441-023-00569-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 07/24/2023] [Indexed: 09/06/2023]
Abstract
Scholars and practitioners have described how investing in health care earlier rather than later can be beneficial, from how "biomarkers" offer promise for early disease detection to healthcare system "incentives" that can promote early preventive medicine. Work by health economists has also made clear that the "health capital" of an individual depreciates over time in the absence of investments in health. Yet, our current policy makers and healthcare system continue prioritizing care of late-stage complex symptomatic illness, often when cure is impossible and disease reversal is improbable, thus exacerbating public health burdens. Critically missing are predicates to address this challenge include the following: first, identifying and validating the specific set of presymptomatic biomarkers that will inform the most appropriate intervention timing for those medical conditions amenable to early intervention; second, shifting fundamental health economic incentives to influence the appropriate disease prevention market; and third, formulating and executing a viable economic framework of reimbursement. We examine these predicates and propose actionable policy recommendations that may help align stakeholder interests to improve public health.
Collapse
Affiliation(s)
- H Hugo Caicedo
- Connection Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Corporate Sustainability and Innovation, Harvard University, Cambridge, MA, USA.
| | - Jonathan J Darrow
- Program On Regulation, Therapeutics, and Law, Harvard Medical School, Boston, MA, USA
| | | | - Alex Pentland
- Connection Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
2
|
Van Rampelbergh J, Achenbach P, Leslie RD, Ali MA, Dayan C, Keymeulen B, Owen KR, Kindermans M, Parmentier F, Carlier V, Ahangarani RR, Gebruers E, Bovy N, Vanderelst L, Van Mechelen M, Vandepapelière P, Boitard C. First-in-human, double-blind, randomized phase 1b study of peptide immunotherapy IMCY-0098 in new-onset type 1 diabetes. BMC Med 2023; 21:190. [PMID: 37226224 DOI: 10.1186/s12916-023-02900-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a CD4+ T cell-driven autoimmune disease characterized by the destruction of insulin-producing pancreatic β-cells by CD8+ T cells. Achieving glycemic targets in T1D remains challenging in clinical practice; new treatments aim to halt autoimmunity and prolong β-cell survival. IMCY-0098 is a peptide derived from human proinsulin that contains a thiol-disulfide oxidoreductase motif at the N-terminus and was developed to halt disease progression by promoting the specific elimination of pathogenic T cells. METHODS This first-in-human, 24-week, double-blind phase 1b study evaluated the safety of three dosages of IMCY-0098 in adults diagnosed with T1D < 6 months before study start. Forty-one participants were randomized to receive four bi-weekly injections of placebo or increasing doses of IMCY-0098 (dose groups A/B/C received 50/150/450 μg for priming followed by three further administrations of 25/75/225 μg, respectively). Multiple T1D-related clinical parameters were also assessed to monitor disease progression and inform future development. Long-term follow-up to 48 weeks was also conducted in a subset of patients. RESULTS Treatment with IMCY-0098 was well tolerated with no systemic reactions; a total of 315 adverse events (AEs) were reported in 40 patients (97.6%) and were related to study treatment in 29 patients (68.3%). AEs were generally mild; no AE led to discontinuation of the study or death. No significant decline in C-peptide was noted from baseline to Week 24 for dose A, B, C, or placebo (mean change - 0.108, - 0.041, - 0.040, and - 0.012, respectively), suggesting no disease progression. CONCLUSIONS Promising safety profile and preliminary clinical response data support the design of a phase 2 study of IMCY-0098 in patients with recent-onset T1D. TRIAL REGISTRATION IMCY-T1D-001: ClinicalTrials.gov NCT03272269; EudraCT: 2016-003514-27; and IMCY-T1D-002: ClinicalTrials.gov NCT04190693; EudraCT: 2018-003728-35.
Collapse
Affiliation(s)
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | | | - Mohammad Alhadj Ali
- Diabetes Research Group, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Colin Dayan
- Diabetes Research Group, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Bart Keymeulen
- Member of Belgian Diabetes Registry, Academic Hospital and Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katharine R Owen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | | | | | - Vincent Carlier
- Imcyse S.A., Avenue Pré-Aily 14, Angleur, 4031, Liège, Belgium
| | | | | | - Nicolas Bovy
- Imcyse S.A., Avenue Pré-Aily 14, Angleur, 4031, Liège, Belgium
| | - Luc Vanderelst
- Imcyse S.A., Avenue Pré-Aily 14, Angleur, 4031, Liège, Belgium
| | | | | | - Christian Boitard
- Inserm U1016, Cochin Institute, Paris, France
- Medical Faculty, Université de Paris, Paris, France
| |
Collapse
|
3
|
Roep BO. The need and benefit of immune monitoring to define patient and disease heterogeneity, mechanisms of therapeutic action and efficacy of intervention therapy for precision medicine in type 1 diabetes. Front Immunol 2023; 14:1112858. [PMID: 36733487 PMCID: PMC9887285 DOI: 10.3389/fimmu.2023.1112858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
The current standard of care for type 1 diabetes patients is limited to treatment of the symptoms of the disease, insulin insufficiency and its complications, not its cause. Given the autoimmune nature of type 1 diabetes, immunology is critical to understand the mechanism of disease progression, patient and disease heterogeneity and therapeutic action. Immune monitoring offers the key to all this essential knowledge and is therefore indispensable, despite the challenges and costs associated. In this perspective, I attempt to make this case by providing evidence from the past to create a perspective for future trials and patient selection.
Collapse
|
4
|
Park Y. Interval design to identify the optimal biological dose for immunotherapy. Contemp Clin Trials Commun 2022; 30:101005. [PMID: 36186542 PMCID: PMC9520219 DOI: 10.1016/j.conctc.2022.101005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 07/31/2022] [Accepted: 09/16/2022] [Indexed: 11/04/2022] Open
Abstract
Immunotherapeutics have revolutionized the treatment of metastatic cancers and are expected to play an increasingly prominent role in the treatment of cancer patients. Recent advances in checkpoint inhibition show promising early results in a number of malignancies, and several treatments have been approved for use. However, the immunotherapeutic agents have been shown to have different mechanisms of antitumor activity from cytotoxic agents, and many limitations and challenges encountered in the traditional paradigm were recently pointed out for immunotherapy. I propose a desirability-based method to determine the optimal biological dose of immunotherapeutics by effectively using toxicity, immune response, and tumor response. Moreover, a new dose allocation algorithm of interval designs is proposed to incorporate immune response in addition to toxicity and tumor response. Simulation studies show that the proposed design has desirable operating characteristics compared to existing dose-finding designs. It also inherits the strengths of interval designs for dose-finding trials, yielding good performance with ease of implementation.
Collapse
Affiliation(s)
- Yeonhee Park
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, United States of America
| |
Collapse
|
5
|
Camaya I, Mok TY, Lund M, To J, Braidy N, Robinson MW, Santos J, O'Brien B, Donnelly S. The parasite-derived peptide FhHDM-1 activates the PI3K/Akt pathway to prevent cytokine-induced apoptosis of β-cells. J Mol Med (Berl) 2021; 99:1605-1621. [PMID: 34374810 DOI: 10.1007/s00109-021-02122-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 12/31/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterised by the destruction of the insulin-producing beta (β)-cells within the pancreatic islets. We have previously identified a novel parasite-derived molecule, termed Fasciola hepatica helminth defence molecule 1 (FhHDM-1), that prevents T1D development in non-obese diabetic (NOD) mice. In this study, proteomic analyses of pancreas tissue from NOD mice suggested that FhHDM-1 activated the PI3K/Akt signalling pathway, which is associated with β-cell metabolism, survival and proliferation. Consistent with this finding, FhHDM-1 preserved β-cell mass in NOD mice. Examination of the biodistribution of FhHDM-1 after intraperitoneal administration in NOD mice revealed that the parasite peptide localised to the pancreas, suggesting that it exerted a direct effect on the survival/function of β-cells. This was confirmed in vitro, as the interaction of FhHDM-1 with the NOD-derived β-cell line, NIT-1, resulted in increased levels of phosphorylated Akt, increased NADH and NADPH and reduced activity of the NAD-dependent DNA nick sensor, poly(ADP-ribose) polymerase (PARP-1). As a consequence, β-cell survival was enhanced and apoptosis was prevented in the presence of the pro-inflammatory cytokines that destroy β-cells during T1D pathogenesis. Similarly, FhHDM-1 protected primary human islets from cytokine-induced apoptosis. Importantly, while FhHDM-1 promoted β-cell survival, it did not induce proliferation. Collectively, these data indicate that FhHDM-1 has significant therapeutic applications to promote β-cell survival, which is required for T1D and T2D prevention and islet transplantation. KEY MESSAGES: FhHDM-1 preserves β-cell mass in NOD mice and prevents the development of T1D. FhHDM-1 enhances phosphorylation of Akt in mouse β-cell lines. FhHDM-1 increases levels of NADH/NADPH in mouse β-cell lines in vitro. FhHDM-1 prevents cytokine-induced cell death of mouse β-cell lines and primary human β-cells in vitro via activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Inah Camaya
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Tsz Y Mok
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Maria Lund
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Joyce To
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, Randwick, Australia
| | - Mark W Robinson
- School of Biological Sciences, Queen's University, Belfast, Northern Ireland, UK
| | - Jerran Santos
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Bronwyn O'Brien
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, the University of Technology Sydney, Ultimo, Australia.
| |
Collapse
|
6
|
Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, Verchere CB. The β Cell in Diabetes: Integrating Biomarkers With Functional Measures. Endocr Rev 2021; 42:528-583. [PMID: 34180979 PMCID: PMC9115372 DOI: 10.1210/endrev/bnab021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 02/08/2023]
Abstract
The pathogenesis of hyperglycemia observed in most forms of diabetes is intimately tied to the islet β cell. Impairments in propeptide processing and secretory function, along with the loss of these vital cells, is demonstrable not only in those in whom the diagnosis is established but typically also in individuals who are at increased risk of developing the disease. Biomarkers are used to inform on the state of a biological process, pathological condition, or response to an intervention and are increasingly being used for predicting, diagnosing, and prognosticating disease. They are also proving to be of use in the different forms of diabetes in both research and clinical settings. This review focuses on the β cell, addressing the potential utility of genetic markers, circulating molecules, immune cell phenotyping, and imaging approaches as biomarkers of cellular function and loss of this critical cell. Further, we consider how these biomarkers complement the more long-established, dynamic, and often complex measurements of β-cell secretory function that themselves could be considered biomarkers.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Yi-Chun Chen
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Austin J Taylor
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
7
|
Roep BO, Montero E, van Tienhoven R, Atkinson MA, Schatz DA, Mathieu C. Defining a cure for type 1 diabetes: a call to action. Lancet Diabetes Endocrinol 2021; 9:553-555. [PMID: 34339632 DOI: 10.1016/s2213-8587(21)00181-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Bart O Roep
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA, USA; Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands.
| | - Enrique Montero
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - René van Tienhoven
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Mark A Atkinson
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Desmond A Schatz
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Chantal Mathieu
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Suneja S, Gangopadhyay S, Saini V, Dawar R, Kaur C. Emerging Diabetic Novel Biomarkers of the 21st Century. ANNALS OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES (INDIA) 2021. [DOI: 10.1055/s-0041-1726613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AbstractDiabetes is a growing epidemic with estimated prevalence of infected to reach ~592 million by the year 2035. An effective way to approach is to detect the disease at a very early stage to reduce the complications and improve lifestyle management. Although several traditional biomarkers including glucated hemoglobin, glucated albumin, fructosamine, and 1,5-anhydroglucitol have helped in ease of diagnosis, there is lack of sensitivity and specificity and are inaccurate in certain clinical settings. Thus, search for new and effective biomarkers is a continuous process with an aim of accurate and timely diagnosis. Several novel biomarkers have surged in the present century that are helpful in timely detection of the disease condition. Although it is accepted that a single biomarker will have its inherent limitations, combining several markers will help to identify individuals at high risk of developing prediabetes and eventually its progression to frank diabetes. This review describes the novel biomarkers of the 21st century, both in type 1 and type 2 diabetes mellitus, and their present potential for assessing risk stratification due to insulin resistance that will pave the way for improved clinical outcome.
Collapse
Affiliation(s)
- Shilpa Suneja
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Sukanya Gangopadhyay
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Vandana Saini
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Rajni Dawar
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Charanjeet Kaur
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
9
|
de Wolf ACMT, Herberts CA, Hoefnagel MHN. Dawn of Monitoring Regulatory T Cells in (Pre-)clinical Studies: Their Relevance Is Slowly Recognised. Front Med (Lausanne) 2020; 7:91. [PMID: 32300597 PMCID: PMC7142310 DOI: 10.3389/fmed.2020.00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Tregs) have a prominent role in the control of immune homeostasis. Pharmacological impact on their activity or balance with effector T cells could contribute to (impaired) clinical responses or adverse events. Monitoring treatment-related effects on T cell subsets may therefore be part of (pre-)clinical studies for medicinal products. However, the extent of immune monitoring performed in studies for marketing authorisation and the degree of correspondence with data available in the public domain is not known. We evaluated the presence of T cell immunomonitoring in 46 registration dossiers of monoclonal antibodies indicated for immune-related disorders and published scientific papers. We found that the depth of Treg analysis in registration dossiers was rather small. Nevertheless, data on treatment-related Treg effects are available in public academia-driven studies (post-registration) and suggest that Tregs may act as a biomarker for clinical responses. However, public data are fragmented and obtained with heterogeneity of experimental approaches from a diversity of species and tissues. To reveal the potential added value of T cell (and particular Treg) evaluation in (pre-)clinical studies, more cell-specific data should be acquired, at least for medicinal products with an immunomodulatory mechanism. Therefore, extensive analysis of T cell subset contribution to clinical responses and the relevance of treatment-induced changes in their levels is needed. Preferably, industry and academia should work together to obtain these data in a standardised manner and to enrich our knowledge about T cell activity in disease pathogenesis and therapies. This will ultimately elucidate the necessity of T cell subset monitoring in the therapeutic benefit-risk assessment.
Collapse
|
10
|
Ihantola EL, Ilmonen H, Kailaanmäki A, Rytkönen-Nissinen M, Azam A, Maillère B, Lindestam Arlehamn CS, Sette A, Motwani K, Seay HR, Brusko TM, Knip M, Veijola R, Toppari J, Ilonen J, Kinnunen T. Characterization of Proinsulin T Cell Epitopes Restricted by Type 1 Diabetes-Associated HLA Class II Molecules. THE JOURNAL OF IMMUNOLOGY 2020; 204:2349-2359. [PMID: 32229538 DOI: 10.4049/jimmunol.1901079] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/22/2020] [Indexed: 12/21/2022]
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease in which the insulin-producing β cells within the pancreas are destroyed. Identification of target Ags and epitopes of the β cell-reactive T cells is important both for understanding T1D pathogenesis and for the rational development of Ag-specific immunotherapies for the disease. Several studies suggest that proinsulin is an early and integral target autoantigen in T1D. However, proinsulin epitopes recognized by human CD4+ T cells have not been comprehensively characterized. Using a dye dilution-based T cell cloning method, we generated and characterized 24 unique proinsulin-specific CD4+ T cell clones from the peripheral blood of 17 individuals who carry the high-risk DR3-DQ2 and/or DR4-DQ8 HLA class II haplotypes. Some of the clones recognized previously reported DR4-restricted epitopes within the C-peptide (C25-35) or A-chain (A1-15) of proinsulin. However, we also characterized DR3-restricted epitopes within both the B-chain (B16-27 and B22-C3) and C-peptide (C25-35). Moreover, we identified DQ2-restricted epitopes within the B-chain and several DQ2- or DQ8-restricted epitopes within the C-terminal region of C-peptide that partially overlap with previously reported DQ-restricted epitopes. Two of the DQ2-restricted epitopes, B18-26 and C22-33, were shown to be naturally processed from whole human proinsulin. Finally, we observed a higher frequency of CDR3 sequences matching the TCR sequences of the proinsulin-specific T cell clones in pancreatic lymph node samples compared with spleen samples. In conclusion, we confirmed several previously reported epitopes but also identified novel (to our knowledge) epitopes within proinsulin, which are presented by HLA class II molecules associated with T1D risk.
Collapse
Affiliation(s)
- Emmi-Leena Ihantola
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, FI-70210 Kuopio, Finland
| | - Henna Ilmonen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, FI-70210 Kuopio, Finland
| | - Anssi Kailaanmäki
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, FI-70210 Kuopio, Finland
| | - Marja Rytkönen-Nissinen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, FI-70210 Kuopio, Finland
| | - Aurélien Azam
- Commissariat à l'Energie Atomique et aux Energies Alternatives-Saclay, Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif Sur Yvette, France
| | - Bernard Maillère
- Commissariat à l'Energie Atomique et aux Energies Alternatives-Saclay, Université Paris-Saclay, Service d'Ingénierie Moléculaire des Protéines, 91191 Gif Sur Yvette, France
| | | | - Alessandro Sette
- La Jolla Institute for Immunology, La Jolla, CA 92037.,Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Keshav Motwani
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610
| | - Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610.,Department of Pediatrics, University of Florida, College of Medicine Gainesville, FL 32610
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, FI-33520 Tampere, Finland.,Children's Hospital, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland.,Folkhälsan Research Center, FI-00290 Helsinki, Finland
| | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, FI-90014 Oulu, Finland
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, FI-20521 Turku, Finland.,Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, FI-20520 Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, FI-20520 Turku, Finland.,Clinical Microbiology, Turku University Hospital, FI-20521 Turku, Finland; and
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, FI-70210 Kuopio, Finland; .,Eastern Finland Laboratory Centre (ISLAB), FI-70210 Kuopio, Finland
| |
Collapse
|
11
|
Blahnik G, Uchtenhagen H, Chow IT, Speake C, Greenbaum C, Kwok WW, James EA. Analysis of pancreatic beta cell specific CD4+ T cells reveals a predominance of proinsulin specific cells. Cell Immunol 2018; 335:68-75. [PMID: 30428974 DOI: 10.1016/j.cellimm.2018.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/16/2018] [Accepted: 11/06/2018] [Indexed: 01/01/2023]
Abstract
CD4+ T cell responses are thought to play a role in type 1 diabetes (T1D). However, detection and characterization of T cells that respond to beta cell epitopes in subjects with T1D has been limited by technical obstacles, including the inherently low frequencies in peripheral blood and variable responsiveness of individual subjects to single epitopes. We implemented a multicolor staining approach that allows direct ex vivo characterization of multiple CD4+ T cell specificities in a single sample. Here we demonstrate and apply that multicolor approach to directly measure the frequency and phenotype of beta cell specific CD4+ T cells in T1D patients and HLA matched controls. For this work we utilized five DR0401 restricted peptides from proinsulin, GAD65, IA-2, and IGRP, which were previously reported as disease relevant epitopes. Surprisingly, although responses to each of these peptides can be readily detected after in vitro expansion, our results indicated that only proinsulin specific T cells were consistently detectable at moderate frequencies in subjects with T1D. Characterization of beta cell specific CD4+ T cells revealed only modest differences between subjects with T1D and healthy controls. Subjects with T1D did have higher proportions of CD45RA negative epitope specific T cells than controls. In patients epitope specific T cells were often CXCR3 positive and a substantial proportion were CCR7 negative, suggesting a Th1-like effector phenotype. Finally, we demonstrated that our multicolor staining approach is compatible with class I multimer analysis, facilitating the characterization of self-reactive CD4+ and CD8+ T cells using a single sample.
Collapse
Affiliation(s)
- Gabriele Blahnik
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Hannes Uchtenhagen
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - I-Ting Chow
- Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Cate Speake
- Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Carla Greenbaum
- Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - William W Kwok
- Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Eddie A James
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA.
| |
Collapse
|
12
|
Mathieu C, Lahesmaa R, Bonifacio E, Achenbach P, Tree T. Immunological biomarkers for the development and progression of type 1 diabetes. Diabetologia 2018; 61:2252-2258. [PMID: 30209538 DOI: 10.1007/s00125-018-4726-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
Immune biomarkers of type 1 diabetes are many and diverse. Some of these, such as the autoantibodies, are well established but not discriminative enough to deal with the heterogeneity inherent to type 1 diabetes progression. As an alternative, high hopes are placed on T cell assays, which give insight into the cells that actually target the beta cell or play a crucial role in maintaining tolerance. These assays are approaching a level of robustness that may allow for solid conclusions on both disease progression and therapeutic efficacy of immune interventions. In addition, 'omics' approaches to biomarker discovery are rapidly progressing. The potential emergence of novel biomarkers creates a need for the introduction of bioinformatics and 'big data' analysis systems for the integration of the multitude of biomarker data that will be available, to translate these data into clinical tools. It is worth noting that it is unlikely that the same markers will apply to all individuals. Instead, individualised signatures of biomarkers, combining autoantibodies, T cell profiles and other biomarkers, will need to be used to classify at-risk patients into various categories, thus enabling personalised prediction, prevention and treatment approaches. To achieve this goal, the standardisation of assays for biomarker discovery, the integration of analyses and data from biomarker studies and, most importantly, the careful clinical characterisation of individuals providing samples for these studies are critical. Longitudinal sample-collection initiatives, like INNODIA, should lead to novel biomarker discovery, not only providing a better understanding of type 1 diabetes onset and progression, but also yielding biomarkers of therapeutic efficacy of interventions to prevent or arrest type 1 diabetes.
Collapse
Affiliation(s)
- Chantal Mathieu
- Department of Endocrinology, University Hospital Gasthuisberg, KU Leuven, Herestraat, 49 3000, Leuven, Belgium.
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Ezio Bonifacio
- DFG Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter Achenbach
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Diabetes Research, Munich-Neuherberg, Germany
| | - Timothy Tree
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, Borough Wing Guy's Hospital, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
13
|
Proinsulin C-peptide is an autoantigen in people with type 1 diabetes. Proc Natl Acad Sci U S A 2018; 115:10732-10737. [PMID: 30275329 DOI: 10.1073/pnas.1809208115] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing beta cells, found within the islets of Langerhans in the pancreas, are destroyed by islet-infiltrating T cells. Identifying the antigenic targets of beta-cell reactive T cells is critical to gain insight into the pathogenesis of T1D and develop antigen-specific immunotherapies. Several lines of evidence indicate that insulin is an important target of T cells in T1D. Because many human islet-infiltrating CD4+ T cells recognize C-peptide-derived epitopes, we hypothesized that full-length C-peptide (PI33-63), the peptide excised from proinsulin as it is converted to insulin, is a target of CD4+ T cells in people with T1D. CD4+ T cell responses to full-length C-peptide were detected in the blood of: 14 of 23 (>60%) people with recent-onset T1D, 2 of 15 (>13%) people with long-standing T1D, and 1 of 13 (<8%) HLA-matched people without T1D. C-peptide-specific CD4+ T cell clones, isolated from six people with T1D, recognized epitopes from the entire 31 amino acids of C-peptide. Eighty-six percent (19 of 22) of the C-peptide-specific clones were restricted by HLA-DQ8, HLA-DQ2, HLA-DQ8trans, or HLA-DQ2trans, HLA alleles strongly associated with risk of T1D. We also found that full-length C-peptide was a much more potent agonist of some CD4+ T cell clones than an 18mer peptide encompassing the cognate epitope. Collectively, our findings indicate that proinsulin C-peptide is a key target of autoreactive CD4+ T cells in T1D. Hence, full-length C-peptide is a promising candidate for antigen-specific immunotherapy in T1D.
Collapse
|
14
|
James EA, Abreu JRF, McGinty JW, Odegard JM, Fillié YE, Hocter CN, Culina S, Ladell K, Price DA, Alkanani A, Rihanek M, Fitzgerald-Miller L, Skowera A, Speake C, Gottlieb P, Davidson HW, Wong FS, Roep B, Mallone R. Combinatorial detection of autoreactive CD8 + T cells with HLA-A2 multimers: a multi-centre study by the Immunology of Diabetes Society T Cell Workshop. Diabetologia 2018; 61:658-670. [PMID: 29196783 DOI: 10.1007/s00125-017-4508-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/25/2017] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Validated biomarkers are needed to monitor the effects of immune intervention in individuals with type 1 diabetes. Despite their importance, few options exist for monitoring antigen-specific T cells. Previous reports described a combinatorial approach that enables the simultaneous detection and quantification of multiple islet-specific CD8+ T cell populations. Here, we set out to evaluate the performance of a combinatorial HLA-A2 multimer assay in a multi-centre setting. METHODS The combinatorial HLA-A2 multimer assay was applied in five participating centres using centralised reagents and blinded replicate samples. In preliminary experiments, samples from healthy donors were analysed using recall antigen multimers. In subsequent experiments, samples from healthy donors and individuals with type 1 diabetes were analysed using beta cell antigen and recall antigen multimers. RESULTS The combinatorial assay was successfully implemented in each participating centre, with CVs between replicate samples that indicated good reproducibility for viral epitopes (mean %CV = 33.8). For beta cell epitopes, the assay was very effective in a single-centre setting (mean %CV = 18.4), but showed sixfold greater variability across multi-centre replicates (mean %CV = 119). In general, beta cell antigen-specific CD8+ T cells were detected more commonly in individuals with type 1 diabetes than in healthy donors. Furthermore, CD8+ T cells recognising HLA-A2-restricted insulin and glutamate decarboxylase epitopes were found to occur at higher frequencies in individuals with type 1 diabetes than in healthy donors. CONCLUSIONS/INTERPRETATION Our results suggest that, although combinatorial multimer assays are challenging, they can be implemented in multiple laboratories, providing relevant T cell frequency measurements. Assay reproducibility was notably higher in the single-centre setting, suggesting that biomarker analysis of clinical trial samples would be most successful when assays are performed in a single laboratory. Technical improvements, including further standardisation of cytometry platforms, will likely be necessary to reduce assay variability in the multi-centre setting.
Collapse
Affiliation(s)
- Eddie A James
- Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101, USA.
| | - Joana R F Abreu
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - John W McGinty
- Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101, USA
| | - Jared M Odegard
- Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101, USA
| | - Yvonne E Fillié
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Centre, Leiden, the Netherlands
| | - Claire N Hocter
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Aimon Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marynette Rihanek
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lisa Fitzgerald-Miller
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Cate Speake
- Benaroya Research Institute, 1201 9th Ave, Seattle, WA, 98101, USA
| | - Peter Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Howard W Davidson
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - F Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Bart Roep
- Department of Diabetes Immunology, City of Hope, Duarte, CA, USA
| | | | | |
Collapse
|
15
|
Needell JC, Dinarello CA, Ir D, Robertson CE, Ryan SM, Kroehl ME, Frank DN, Zipris D. Implication of the intestinal microbiome as a potential surrogate marker of immune responsiveness to experimental therapies in autoimmune diabetes. PLoS One 2017; 12:e0173968. [PMID: 28301545 PMCID: PMC5354421 DOI: 10.1371/journal.pone.0173968] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/01/2017] [Indexed: 01/13/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune proinflammatory disease with no effective intervention. A major obstacle in developing new immunotherapies for T1D is the lack of means for monitoring immune responsiveness to experimental therapies. The LEW1.WR1 rat develops autoimmunity following infection with the parvovirus Kilham rat virus (KRV) via mechanisms linked with activation of proinflammatory pathways and alterations in the gut bacterial composition. We used this animal to test the hypothesis that intervention with agents that block innate immunity and diabetes is associated with a shift in the gut microbiota. We observed that infection with KRV results in the induction of proinflammatory gene activation in both the spleen and pancreatic lymph nodes. Furthermore, administering animals the histone deacetylase inhibitor ITF-2357 and IL-1 receptor antagonist (Anakinra) induced differential STAT-1 and the p40 unit of IL-12/IL-23 gene expression. Sequencing of bacterial 16S rRNA genes demonstrated that both ITF-2357 and Anakinra alter microbial diversity. ITF-2357 and Anakinra modulated the abundance of 23 and 8 bacterial taxa in KRV-infected animals, respectively, of which 5 overlapped between the two agents. Lastly, principal component analysis implied that ITF-2357 and Anakinra induce distinct gut microbiomes compared with those from untreated animals or rats provided KRV only. Together, the data suggest that ITF-2357 and Anakinra differentially influence the innate immune system and the intestinal microbiota and highlight the potential use of the gut microbiome as a surrogate means of assessing anti-inflammatory immune effects in type 1 diabetes.
Collapse
Affiliation(s)
- James C. Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Charles A. Dinarello
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- University of Colorado Microbiome Research Consortium (MiRC), Aurora, Colorado, United States of America
| | - Sarah M. Ryan
- Department of Biostatistics and Informatics, Colorado School of Public Health and University of Colorado Denver, Aurora, Colorado, United States of America
| | - Miranda E. Kroehl
- Department of Biostatistics and Informatics, Colorado School of Public Health and University of Colorado Denver, Aurora, Colorado, United States of America
| | - Daniel N. Frank
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- University of Colorado Microbiome Research Consortium (MiRC), Aurora, Colorado, United States of America
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| |
Collapse
|
16
|
McLaughlin RJ, de Haan A, Zaldumbide A, de Koning EJ, de Ru AH, van Veelen PA, van Lummel M, Roep BO. Human islets and dendritic cells generate post-translationally modified islet autoantigens. Clin Exp Immunol 2016; 185:133-40. [PMID: 26861694 DOI: 10.1111/cei.12775] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/15/2016] [Accepted: 02/07/2016] [Indexed: 12/14/2022] Open
Abstract
The initiation of type 1 diabetes (T1D) requires a break in peripheral tolerance. New insights into neoepitope formation indicate that post-translational modification of islet autoantigens, for example via deamidation, may be an important component of disease initiation or exacerbation. Indeed, deamidation of islet autoantigens increases their binding affinity to the T1D highest-risk human leucocyte antigen (HLA) haplotypes HLA-DR3/DQ2 and -DR4/DQ8, increasing the chance that T cells reactive to deamidated autoantigens can be activated upon T cell receptor ligation. Here we investigated human pancreatic islets and inflammatory and tolerogenic human dendritic cells (DC and tolDC) as potential sources of deamidated islet autoantigens and examined whether deamidation is altered in an inflammatory environment. Islets, DC and tolDC contained tissue transglutaminase, the key enzyme responsible for peptide deamidation, and enzyme activity increased following an inflammatory insult. Islets treated with inflammatory cytokines were found to contain deamidated insulin C-peptide. DC, heterozygous for the T1D highest-risk DQ2/8, pulsed with native islet autoantigens could present naturally processed deamidated neoepitopes. HLA-DQ2 or -DQ8 homozygous DC did not present deamidated islet peptides. This study identifies both human islets and DC as sources of deamidated islet autoantigens and implicates inflammatory activation of tissue transglutaminase as a potential mechanism for islet and DC deamidation.
Collapse
Affiliation(s)
- R J McLaughlin
- Department of Immunohematology and Blood Transfusion, Leiden, the Netherlands
| | - A de Haan
- Department of Immunohematology and Blood Transfusion, Leiden, the Netherlands
| | - A Zaldumbide
- Department of Molecular Cell Biology, Leiden, the Netherlands
| | - E J de Koning
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - A H de Ru
- Department of Immunohematology and Blood Transfusion, Leiden, the Netherlands
| | - P A van Veelen
- Department of Immunohematology and Blood Transfusion, Leiden, the Netherlands
| | - M van Lummel
- Department of Immunohematology and Blood Transfusion, Leiden, the Netherlands
| | - B O Roep
- Department of Immunohematology and Blood Transfusion, Leiden, the Netherlands.,Department of Diabetes Immunology, Diabetes and Metabolism Research Institute at the Beckman Research Institute of the City of Hope, Duarte, CA, USA
| |
Collapse
|
17
|
Gibson VB, Nikolic T, Pearce VQ, Demengeot J, Roep BO, Peakman M. Proinsulin multi-peptide immunotherapy induces antigen-specific regulatory T cells and limits autoimmunity in a humanized model. Clin Exp Immunol 2015. [PMID: 26206289 DOI: 10.1111/cei.12687] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Peptide immunotherapy (PIT) is a targeted therapeutic approach, involving administration of disease-associated peptides, with the aim of restoring antigen-specific immunological tolerance without generalized immunosuppression. In type 1 diabetes, proinsulin is a primary antigen targeted by the autoimmune response, and is therefore a strong candidate for exploitation via PIT in this setting. To elucidate the optimal conditions for proinsulin-based PIT and explore mechanisms of action, we developed a preclinical model of proinsulin autoimmunity in a humanized HLA-DRB1*0401 transgenic HLA-DR4 Tg mouse. Once proinsulin-specific tolerance is broken, HLA-DR4 Tg mice develop autoinflammatory responses, including proinsulin-specific T cell proliferation, interferon (IFN)-γ and autoantibody production. These are preventable and quenchable by pre- and post-induction treatment, respectively, using intradermal proinsulin-PIT injections. Intradermal proinsulin-PIT enhances proliferation of regulatory [forkhead box protein 3 (FoxP3(+))CD25(high) ] CD4 T cells, including those capable of proinsulin-specific regulation, suggesting this as its main mode of action. In contrast, peptide delivered intradermally on the surface of vitamin D3-modulated (tolerogenic) dendritic cells, controls autoimmunity in association with proinsulin-specific IL-10 production, but no change in regulatory CD4 T cells. These studies define a humanized, translational model for in vivo optimization of PIT to control autoimmunity in type 1 diabetes and indicate that dominant mechanisms of action differ according to mode of peptide delivery.
Collapse
Affiliation(s)
- V B Gibson
- Department of Immunobiology, Faculty of Life Science and Medicine, King's College London, UK
| | - T Nikolic
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - V Q Pearce
- Department of Immunobiology, Faculty of Life Science and Medicine, King's College London, UK
| | - J Demengeot
- Instituto Gulbenkian De Ciencia, Oeiras, Portugal
| | - B O Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - M Peakman
- Department of Immunobiology, Faculty of Life Science and Medicine, King's College London, UK
| |
Collapse
|
18
|
Abstract
Substantial progress in molecular immunology, coupled with an increasing focus on translational research and an enthusiasm for personalized medicine, has resulted in a rapid expansion in the field of immune biomarkers in recent years. In this Science and Society article, we provide a conceptual overview of the field and discuss the progress that has been made so far, as well as the future potential in the context of the scientific, logistical, financial, legal and ethical framework within which this research is being carried out and translated into clinical use.
Collapse
|
19
|
Haile Y, Nakhaei-Nejad M, Boakye PA, Baker G, Smith PA, Murray AG, Giuliani F, Jahroudi N. Reprogramming of HUVECs into induced pluripotent stem cells (HiPSCs), generation and characterization of HiPSC-derived neurons and astrocytes. PLoS One 2015; 10:e0119617. [PMID: 25789622 PMCID: PMC4366250 DOI: 10.1371/journal.pone.0119617] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/02/2015] [Indexed: 11/30/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic and progressive structural or functional loss of neurons. Limitations related to the animal models of these human diseases have impeded the development of effective drugs. This emphasizes the need to establish disease models using human-derived cells. The discovery of induced pluripotent stem cell (iPSC) technology has provided novel opportunities in disease modeling, drug development, screening, and the potential for “patient-matched” cellular therapies in neurodegenerative diseases. In this study, with the objective of establishing reliable tools to study neurodegenerative diseases, we reprogrammed human umbilical vein endothelial cells (HUVECs) into iPSCs (HiPSCs). Using a novel and direct approach, HiPSCs were differentiated into cells of central nervous system (CNS) lineage, including neuronal, astrocyte and glial cells, with high efficiency. HiPSCs expressed embryonic genes such as nanog, sox2 and Oct-3/4, and formed embryoid bodies that expressed markers of the 3 germ layers. Expression of endothelial-specific genes was not detected in HiPSCs at RNA or protein levels. HiPSC-derived neurons possess similar morphology but significantly longer neurites compared to primary human fetal neurons. These stem cell-derived neurons are susceptible to inflammatory cell-mediated neuronal injury. HiPSC-derived neurons express various amino acids that are important for normal function in the CNS. They have functional receptors for a variety of neurotransmitters such as glutamate and acetylcholine. HiPSC-derived astrocytes respond to ATP and acetylcholine by elevating cytosolic Ca2+ concentrations. In summary, this study presents a novel technique to generate differentiated and functional HiPSC-derived neurons and astrocytes. These cells are appropriate tools for studying the development of the nervous system, the pathophysiology of various neurodegenerative diseases and the development of potential drugs for their treatments.
Collapse
Affiliation(s)
- Yohannes Haile
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | | - Paul A. Boakye
- Department of Pharmacology, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Glen Baker
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Psychiatry (Neurochemical Research Unit), University of Alberta, Edmonton, Canada
| | - Peter A. Smith
- Department of Pharmacology, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Allan G. Murray
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Fabrizio Giuliani
- Department of Medicine, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- * E-mail: (NJ); (FG)
| | - Nadia Jahroudi
- Department of Medicine, University of Alberta, Edmonton, Canada
- * E-mail: (NJ); (FG)
| |
Collapse
|
20
|
Orban T, Beam CA, Xu P, Moore K, Jiang Q, Deng J, Muller S, Gottlieb P, Spain L, Peakman M. Reduction in CD4 central memory T-cell subset in costimulation modulator abatacept-treated patients with recent-onset type 1 diabetes is associated with slower C-peptide decline. Diabetes 2014; 63:3449-57. [PMID: 24834977 PMCID: PMC4171657 DOI: 10.2337/db14-0047] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 05/13/2014] [Indexed: 12/30/2022]
Abstract
We previously reported that continuous 24-month costimulation blockade by abatacept significantly slows the decline of β-cell function after diagnosis of type 1 diabetes. In a mechanistic extension of that study, we evaluated peripheral blood immune cell subsets (CD4, CD8-naive, memory and activated subsets, myeloid and plasmacytoid dendritic cells, monocytes, B lymphocytes, CD4(+)CD25(high) regulatory T cells, and invariant NK T cells) by flow cytometry at baseline and 3, 6, 12, 24, and 30 months after treatment initiation to discover biomarkers of therapeutic effect. Using multivariable analysis and lagging of longitudinally measured variables, we made the novel observation in the placebo group that an increase in central memory (CM) CD4 T cells (CD4(+)CD45R0(+)CD62L(+)) during a preceding visit was significantly associated with C-peptide decline at the subsequent visit. These changes were significantly affected by abatacept treatment, which drove the peripheral contraction of CM CD4 T cells and the expansion of naive (CD45R0(-)CD62L(+)) CD4 T cells in association with a significantly slower rate of C-peptide decline. The findings show that the quantification of CM CD4 T cells can provide a surrogate immune marker for C-peptide decline after the diagnosis of type 1 diabetes and that costimulation blockade may exert its beneficial therapeutic effect via modulation of this subset.
Collapse
Affiliation(s)
| | - Craig A Beam
- Division of Informatics and Biostatistics, Department of Pediatrics, University of South Florida, Tampa, FL
| | - Ping Xu
- Division of Informatics and Biostatistics, Department of Pediatrics, University of South Florida, Tampa, FL
| | - Keith Moore
- David H. Murdock Research Institute, Kannapolis, NC
| | - Qi Jiang
- David H. Murdock Research Institute, Kannapolis, NC
| | - Jun Deng
- David H. Murdock Research Institute, Kannapolis, NC
| | - Sarah Muller
- Division of Informatics and Biostatistics, Department of Pediatrics, University of South Florida, Tampa, FL
| | - Peter Gottlieb
- University of Colorado Barbara Davis Center for Childhood Diabetes, Aurora, CO
| | - Lisa Spain
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Mark Peakman
- Peter Gorer Department of Immunobiology, School of Medicine, King's College London, London, UK
| |
Collapse
|
21
|
Abreu JRF, Roep BO. Targeting proinsulin-reactive CD8+ T cells: a new direction for type 1 diabetes treatment. Expert Rev Clin Immunol 2014; 9:1001-3. [PMID: 24168405 DOI: 10.1586/1744666x.2013.852958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Joana R F Abreu
- Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
22
|
Haile Y, Fu W, Shi B, Westaway D, Baker G, Jhamandas J, Giuliani F. Characterization of the NT2-derived neuronal and astrocytic cell lines as alternative in vitro models for primary human neurons and astrocytes. J Neurosci Res 2014; 92:1187-98. [PMID: 24801011 DOI: 10.1002/jnr.23399] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 03/01/2014] [Accepted: 03/30/2014] [Indexed: 12/22/2022]
Abstract
Primary human fetal neurons and astrocytes (HFNs and HFAs, respectively) provide relevant cell types with which to study in vitro the mechanisms involved in various human neurological diseases, such as multiple sclerosis, Parkinson's disease, and Alzheimer's disease. However, the limited availability of human fetal cells poses a significant problem for the study of these diseases when a human cell model system is required. Thus, generating a readily available alternative cell source with the essential features of human neurons and astrocytes is necessary. The human teratoma-derived NTera2/D1 (NT2) cell line is a promising tool from which both neuronal and glial cells can be generated. Nevertheless, a direct comparison of NT2 neurons and primary HFNs in terms of their morphology physiological and chemical properties is still missing. This study directly compares NT2-derived neurons and primary HFNs using immunocytochemistry, confocal calcium imaging, high-performance liquid chromatography, and high-content analysis techniques. We investigated the morphological similarities and differences, levels of relevant amino acids, and internal calcium fluctuations in response to certain neurotransmitters/stimuli. We also compared NT2-derived astrocytes and HFAs. In most of the parameters tested, both neuronal and astrocytic cell types exhibited similarities to primary human fetal neurons and astrocytes. NT2-derived neurons and astrocytes are reliable in vitro tools and a renewable cell source that can serve as a valid alternative to HFNs/HFAs for mechanistic studies of neurological diseases.
Collapse
Affiliation(s)
- Yohannes Haile
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Type 1 diabetes mellitus (T1DM) is the result of autoimmune destruction of pancreatic β cells in genetically predisposed individuals with impaired immune regulation. The insufficiency in the modulation of immune attacks on the β cells might be partly due to genetic causes; indeed, several of the genetic variants that predispose individuals to T1DM have functional features of impaired immune regulation. Whilst defects in immune regulation in patients with T1DM have been identified, many patients seem to have immune regulatory capacities that are indistinguishable from those of healthy individuals. Insight into the regulation of islet autoimmunity might enable us to restore immune imbalances with therapeutic interventions. In this Review, we discuss the current knowledge on immune regulation and dysfunction in humans that is the basis of tissue-specific immune regulation as an alternative to generalized immune suppression.
Collapse
Affiliation(s)
- Bart O Roep
- Leiden University Medical Center, Department of Immunohaematology & Blood Transfusion, P. O. Box 9600, NL-2300 RC Leiden, Netherlands
| | - Timothy I M Tree
- Department of Immunobiology, King's College London, School of Medicine, London SE1 9RT, UK
| |
Collapse
|
24
|
Sarikonda G, Pettus J, Phatak S, Sachithanantham S, Miller JF, Wesley JD, Cadag E, Chae J, Ganesan L, Mallios R, Edelman S, Peters B, von Herrath M. CD8 T-cell reactivity to islet antigens is unique to type 1 while CD4 T-cell reactivity exists in both type 1 and type 2 diabetes. J Autoimmun 2013; 50:77-82. [PMID: 24387802 DOI: 10.1016/j.jaut.2013.12.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/06/2013] [Accepted: 12/08/2013] [Indexed: 01/19/2023]
Abstract
Previous cross-sectional analyses demonstrated that CD8(+) and CD4(+) T-cell reactivity to islet-specific antigens was more prevalent in T1D subjects than in healthy donors (HD). Here, we examined T1D-associated epitope-specific CD4(+) T-cell cytokine production and autoreactive CD8(+) T-cell frequency on a monthly basis for one year in 10 HD, 33 subjects with T1D, and 15 subjects with T2D. Autoreactive CD4(+) T-cells from both T1D and T2D subjects produced more IFN-γ when stimulated than cells from HD. In contrast, higher frequencies of islet antigen-specific CD8(+) T-cells were detected only in T1D. These observations support the hypothesis that general beta-cell stress drives autoreactive CD4(+) T-cell activity while islet over-expression of MHC class I commonly seen in T1D mediates amplification of CD8(+) T-cells and more rapid beta-cell loss. In conclusion, CD4(+) T-cell autoreactivity appears to be present in both T1D and T2D while autoreactive CD8(+) T-cells are unique to T1D. Thus, autoreactive CD8(+) cells may serve as a more T1D-specific biomarker.
Collapse
Affiliation(s)
| | - Jeremy Pettus
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; University of California San Diego, San Diego, CA, USA
| | - Sonal Phatak
- University of California San Diego, San Diego, CA, USA
| | | | | | | | | | - Ji Chae
- University of California San Diego, San Diego, CA, USA
| | | | - Ronna Mallios
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Steve Edelman
- University of California San Diego, San Diego, CA, USA
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Matthias von Herrath
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Novo Nordisk Type 1 Diabetes R & D Center, Seattle, WA, USA.
| |
Collapse
|
25
|
Pretransplantation GAD-autoantibody status to guide prophylactic antibody induction therapy in simultaneous pancreas and kidney transplantation. Transplantation 2013; 96:745-52. [PMID: 23912172 DOI: 10.1097/tp.0b013e3182a012cc] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Daclizumab and antithymocyte globulin (ATG) have been shown to reduce allograft rejection. We assessed the safety and efficacy of daclizumab or ATG prophylaxis in combination with triple immunotherapy in simultaneous pancreas-kidney transplant (SPKT) recipients. METHODS Thirty-nine type 1 diabetic patients scheduled for primary SPKT were randomized to receive prophylactic therapy with either daclizumab or ATG. A group of 27 patients without prophylactic antibodies was used for retrospective comparison. All patients received cyclosporine and mycophenolate mofetil and gradually tapered prednisone. Autoantibodies and cellular autoreactivity were measured to assess recurrent autoreactive responses. RESULTS Baseline and transplant characteristics were comparable among groups. Both daclizumab and ATG therapy resulted in a significant reduction in acute rejection episodes. The incidence of rejection episodes was significantly higher in pretransplantation GAD autoantibody-positive daclizumab-treated recipients compared with GAD autoantibody-negative or ATG-treated recipients. IA-2 islet autoantibodies showed no association with rejection. There were no significant differences between the groups for in vitro autoreactivity, clinical outcome, or functional parameters. CONCLUSIONS Daclizumab or ATG combined with a maintenance immunosuppressive regime consisting of cyclosporine, mycophenolate mofetil, and prednisolone were well tolerated and equally effective in reducing the incidence of acute rejection episodes in SPKT recipients. Up to 3 years, no adverse sequelae of the immunoprophylaxis or clinical and ex vivo recurrent autoimmunity were observed. We propose that the pretransplantation existence of GAD65 autoantibodies serves as a marker guiding the choice for prophylactic therapy in pancreas transplantation.
Collapse
|
26
|
Sarikonda G, Pettus J, Sachithanantham S, Phatak S, Miller JF, Ganesan L, Chae J, Mallios R, Edelman S, Peters B, von Herrath M. Temporal intra-individual variation of immunological biomarkers in type 1 diabetes patients: implications for future use in cross-sectional assessment. PLoS One 2013; 8:e79383. [PMID: 24223938 PMCID: PMC3817042 DOI: 10.1371/journal.pone.0079383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/21/2013] [Indexed: 12/22/2022] Open
Abstract
Multiple immune parameters such as frequencies of autoreactive CD4+, CD8+ T-cells and CD4+CD25+Foxp3+ T-cells have been explored as biomarkers in human T1D. However, intra-individual temporal variation of these parameters has not been assessed systematically over time. We determined the variation in each of these parameters in a cohort of T1D and healthy donors (HDs), at monthly intervals for one year. Despite low intra- and inter-assay co-efficient of variation (CV), mean CVs for each of the immune parameters were 119.1% for CD4+ T-cell-derived IFN-γ, 50.44% for autoreactive CD8+ T-cells, and 31.24% for CD4+CD25+Foxp3+ T-cells. Further, both HDs and T1D donors had similar CVs. The variation neither correlated with BMI, age, disease duration or insulin usage, nor were there detectable cyclical patterns of variation. However, averaging results from multiple visits for an individual provided a better estimate of the CV between visits. Based on our data we predict that by averaging values from three visits a treatment effect on these parameters with a 50% effect size could be detected with the same power using 1.8–4-fold fewer patients within a trial compared to using values from a single visit. Thus, our present data contribute to a more robust, accurate endpoint design for future clinical trials in T1D and aid in the identification of truly efficacious therapies.
Collapse
Affiliation(s)
- Ghanashyam Sarikonda
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Jeremy Pettus
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- University of California San Diego, San Diego, California, United States of America
| | - Sowbarnika Sachithanantham
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Sonal Phatak
- University of California San Diego, San Diego, California, United States of America
| | - Jacqueline F. Miller
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Lakshmi Ganesan
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- University of California San Diego, San Diego, California, United States of America
| | - Ji Chae
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- University of California San Diego, San Diego, California, United States of America
| | - Ronna Mallios
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Steve Edelman
- University of California San Diego, San Diego, California, United States of America
| | - Bjoern Peters
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Matthias von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
27
|
van Lummel M, Zaldumbide A, Roep BO. Changing faces, unmasking the beta-cell: post-translational modification of antigens in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2013; 20:299-306. [PMID: 23770733 DOI: 10.1097/med.0b013e3283631417] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Description on post-translational modification of islet-autoantigens in type 1 diabetes (T1D). RECENT FINDINGS T1D is an autoimmune disease characterized by progressive destruction of the insulin-producing beta-cells. It is a complex disease process that results from the loss of tolerance to beta-cell autoantigens. This loss of tolerance can be caused by modification of beta-cell autoantigens, generating 'neo-autoantigens', and inducing T-cell responses. Post-translational modifications (PTMs) within the endoplasmic reticulum of stressed beta-cells might impact on the autoantigen T-cell epitope repertoire and on T1D pathogenesis progression. This review summarizes the processes involved in beta-cell stress and PTM of beta-cell autoantigens in T1D. SUMMARY PTMs of beta-cell autoantigens provide a novel hypothesis to understand how autoreactive T-cells can escape immune tolerance and cause destruction of beta-cells ('beta-cell homicide'). Additionally, aberrant proteins produced by stressed beta-cells can cause their own destruction ('beta-cell suicide'). Upon endoplasmic reticulum-stress, proteins are misfolded or modified changing the protein structure. In T1D, this may generate new beta-cell (neo)autoantigens. PTM of islet-autoantigens provides a mechanism by which pathogenic T-cells can escape thymic deletion. This amplifies the immune response when encountering a modified beta-cell neo-autoantigen bound to T1D predisposing human leucocyte antigen molecules in the periphery.
Collapse
Affiliation(s)
- Menno van Lummel
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
28
|
von Herrath M, Peakman M, Roep B. Progress in immune-based therapies for type 1 diabetes. Clin Exp Immunol 2013; 172:186-202. [PMID: 23574316 DOI: 10.1111/cei.12085] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2013] [Indexed: 01/10/2023] Open
Abstract
Immune-based therapies that prevent type 1 diabetes or preserve metabolic function remaining at diagnosis have become a major objective for funding agencies and international trial consortia, and receive backing from notable patient advocate groups. The development of immune-based therapeutic strategies in this arena requires a careful balancing of the risks of the therapy against the potential benefits, because many individuals are diagnosed or identified as being at increased risk of disease in early childhood, a period when manipulation of the developing immune system should be undertaken with caution. In addition, a therapy exists (daily insulin injection) that is life-saving in the acute stages of disease and can be used effectively over a lifetime as maintenance. Conversely, the disease is increasing in incidence; is peaking in ever-younger age groups; carries significant risk of increased morbidity and early mortality; and remains difficult to manage effectively in many settings. With these issues in mind, in this article we review progress towards immune-based strategies for this chronic autoimmune disease.
Collapse
Affiliation(s)
- M von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | | |
Collapse
|
29
|
Ahmed ST, Akirav E, Bradshaw E, Buckner J, McKinney E, Quintana FJ, Waldron-Lynch F, Nepom J. Immunological biomarkers: catalysts for translational advances in autoimmune diabetes. Clin Exp Immunol 2013; 172:178-85. [PMID: 23574315 DOI: 10.1111/cei.12063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2012] [Indexed: 12/20/2022] Open
Abstract
In a recent workshop organized by the JDRF focused on the 'Identification and Utilization of Robust Biomarkers in Type1 Diabetes', leaders in the field of type 1 diabetes (T1D)/autoimmunity and assay technology came together from academia, government and industry to assess the current state of the field, evaluate available resources/technologies and identify gaps that need to be filled for moving the field of T1D research forward. The highlights of this workshop are discussed in this paper, as well as the proposal for a larger, planned consortium effort, incorporating a JDRF Biomarker Core, to foster collaboration and accelerate progress in this critically needed area of T1D research.
Collapse
Affiliation(s)
- S T Ahmed
- JDRF, 26, Broadway, 14th Floor, New York, NY 10004, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Mallone R, Roep BO. Biomarkers for immune intervention trials in type 1 diabetes. Clin Immunol 2013; 149:286-96. [PMID: 23510725 DOI: 10.1016/j.clim.2013.02.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 02/09/2013] [Indexed: 02/07/2023]
Abstract
After many efforts to improve and standardize assays for detecting immune biomarkers in type 1 diabetes (T1D), methods to identify and monitor such correlates of insulitis are coming of age. The ultimate goal is to use these correlates to predict disease progression before onset and regression following therapeutic intervention, which would allow performing smaller and shorter pilot clinical trials with earlier endpoints than those offered by preserved β-cell function or improved glycemic control. Here, too, progress has been made. With the emerging insight that T1D represents a heterogeneous disease, the next challenge is to define patient subpopulations that qualify for personalized medicine or that should be enrolled for immune intervention, to maximize clinical benefit and decrease collateral damage by ineffective or even adverse immune therapeutics. This review discusses the current state of the art, setting the stage for future efforts to monitor disease heterogeneity, progression and therapeutic intervention in T1D.
Collapse
Affiliation(s)
- Roberto Mallone
- Cochin Institute, INSERM U1016, DeAR Lab Avenir, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Assistance Publique Hôpitaux de Paris, Hôtel Dieu, Service de Diabétologie, Paris, France.
| | | |
Collapse
|
31
|
Abstract
Type 1 diabetes (T1D) represents 10 to 15% of all forms of diabetes. Its incidence shows a consistent rise in all countries under survey. Evidence for autoimmunity in human T1D relies on the detection of insulitis, of islet cell antibodies, of activated β-cell-specific T lymphocytes and on the association of T1D with a restricted set of class II major histocompatibility complex (MHC) alleles. However, mechanisms that initiate the failure of immune tolerance to β-cell autoantigens remain elusive in common forms of T1D. T1D commonly develop as a multifactorial disease in which environmental factors concur with a highly multigenic background. The disease is driven by the activation of T-lymphocytes against pancreatic β-cells. Several years elapse between initial triggering of the autoimmune response to β cells, as evidenced by the appearance or islet cell autoantibodies, and the onset of clinical diabetes, defining a prediabetes stage. Active mechanisms hold back autoreactive effector T-cells in prediabetes, in particular a subset of CD4+ T-cells (T(reg)) and other regulatory T-cells, such as invariant NKT cells. There is evidence in experimental models that systemic or local infections can trigger autoimmune reactions to β-cells. However, epidemiological observations that have accumulated over years have failed to identify undisputable environmental factors that trigger T1D. Moreover, multiple environmental factors may intervene in the disease evolution and protective as weel as triggering environmental factors may be involved. Available models also indicate that local signals within the islets are required for full-blown diabetes to develop. Many autoantigens that are expressed by β-cells but also by the other endocrine islet cells and by neurons are recognized by lymphocytes along the development of T1D. The immune image of β-cells is that of native components of the β-cell membrane, as seen by B-lymphocytes, and of fragments of intracellular β-cell proteins in the form of peptides loaded onto class I MHC molecules on the β-cell surface and class I and class II molecules onto professional antigen presenting cells. Given the key role of T lymphocytes in T1D, the cartography of autoantigen-derived peptides that are presented to class I-restricted CD8(+) T-cells and class II-restricted CD4(+) T-cells is of outmost importance and is a necessary step in the development of diagnostic T-cell assays and of immunotherapy of T1D.
Collapse
|
32
|
Abreu JRF, Martina S, Verrijn Stuart AA, Fillié YE, Franken KLMC, Drijfhout JW, Roep BO. CD8 T cell autoreactivity to preproinsulin epitopes with very low human leucocyte antigen class I binding affinity. Clin Exp Immunol 2012; 170:57-65. [PMID: 22943201 DOI: 10.1111/j.1365-2249.2012.04635.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Beta cells presenting islet epitopes are recognized and destroyed by autoreactive CD8 T cells in type 1 diabetes. These islet-specific T cells are believed to react with epitopes binding with high affinity to human leucocyte antigen (HLA) expressed on beta cells. However, this assumption might be flawed in case of islet autoimmunity. We evaluated T cell recognition of the complete array of preproinsulin (PPI) peptides with regard to HLA binding affinity and T cell recognition. In a comprehensive approach, 203 overlapping 9-10mer PPI peptides were tested for HLA-A2 binding and subjected to binding algorithms. Subsequently, a high-throughput assay was employed to detect PPI-specific T cells in patient blood, in which conditional HLA ligands were destabilized by ultraviolet irradiation and HLA molecules refolded with arrays of PPI peptides, followed by quantum-dot labelling and T cell staining. Analysis of patient blood revealed high frequencies of CD8 T cells recognizing very low HLA binding peptides. Of 28 peptides binding to HLA-A2, a majority was predicted not to bind. Unpredicted peptides bound mainly with low affinities. HLA binding affinity and immunogenicity may not correlate in autoimmunity. Algorithms used to predict high-affinity HLA peptide binders discount the majority of low-affinity HLA binding epitopes. Appreciation that peptides binding HLA with very low affinity can act as targets of autoreactive T cells may help to understand loss of tolerance and disease pathogenesis and possibly point to tissue-specific immune intervention targets.
Collapse
Affiliation(s)
- J R F Abreu
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
33
|
Kleijwegt FS, Jansen DTSL, Teeler J, Joosten AM, Laban S, Nikolic T, Roep BO. Tolerogenic dendritic cells impede priming of naïve CD8⁺ T cells and deplete memory CD8⁺ T cells. Eur J Immunol 2012; 43:85-92. [PMID: 23042025 DOI: 10.1002/eji.201242879] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/03/2012] [Accepted: 09/21/2012] [Indexed: 12/22/2022]
Abstract
Type 1 diabetes is a T-cell-mediated autoimmune disease in which autoreactive CD8(+) T cells destroy the insulin-producing pancreatic beta cells. Vitamin D3 and dexamethasone-modulated dendritic cells (Combi-DCs) loaded with islet antigens inducing islet-specific regulatory CD4(+) T cells may offer a tissue-specific intervention therapy. The effect of Combi-DCs on CD8(+) T cells, however, remains unknown. To investigate the interaction of CD8(+) T cells with Combi-DCs presenting epitopes on HLA class I, naive, and memory CD8(+) T cells were co-cultured with DCs and proliferation and function of peptide-specific T cells were analyzed. Antigen-loaded Combi-DCs were unable to prime naïve CD8(+) T cells to proliferate, although a proportion of T cells converted to a memory phenotype. Moreover, expansion of CD8(+) T cells that had been primed by mature monocyte-derived DCs (moDCs) was curtailed by Combi-DCs in co-cultures. Combi-DCs expanded memory T cells once, but CD8(+) T-cell numbers collapsed by subsequent re-stimulation with Combi-DCs. Our data point that (re)activation of CD8(+) T cells by antigen-pulsed Combi-DCs does not promote, but rather deteriorates, CD8(+) T-cell immunity. Yet, Combi-DCs pulsed with CD8(+) T-cell epitopes also act as targets of cytotoxicity, which is undesirable for survival of Combi-DCs infused into patients in therapeutic immune intervention strategies.
Collapse
Affiliation(s)
- Fleur S Kleijwegt
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
34
|
Boitard C, Timsit J. Towards an aetiological treatment of type 1 diabetes? New tools, new strategies. DIABETES & METABOLISM 2012; 38:375-7. [PMID: 23122483 DOI: 10.1016/j.diabet.2012.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 09/14/2012] [Indexed: 11/25/2022]
|
35
|
Mangano K, Fagone P, Di Mauro M, Ascione E, Maiello V, Milicic T, Jotic A, Lalic NM, Saksida T, Stojanovic I, Selmi C, Farina C, Stosic-Grujicic S, Meroni P, Nicoletti F. The immunobiology of apotransferrin in type 1 diabetes. Clin Exp Immunol 2012; 169:244-52. [PMID: 22861364 DOI: 10.1111/j.1365-2249.2012.04619.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The transferrin (Tf) family of iron binding proteins includes important endogenous modulators of the immune function that may modulate autoimmune diseases. To define more clearly the role of apotransferrin (apoTf) in type 1 diabetes we determined the impact of this protein on type 1 diabetes as investigated in islet cells, animal models and patient sera. First, we demonstrated that recombinant apoTf counteracts the cytokine-induced death of murine pancreatic islet cells. Secondly, human apoTf administration favourably influences the course of type 1 diabetes in animal models, resulting in protection against disease development that was associated with reduction of insulitis and reduced levels of proinflammatory cytokines. Finally, we confirmed that patients with newly diagnosed type 1 diabetes manifest significantly lower apoTf serum levels compared to healthy controls and patients with long-lasting disease. In conclusion, our data suggest the apoTf pivotal role in the perpetuation of type 1 diabetes pathology.
Collapse
Affiliation(s)
- K Mangano
- Department of Bio-Medical Sciences, Via Androne 83, 95124, Catania, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Vaccination is the administration of antigenic material to stimulate the immune system to develop adaptive immunity to a disease. As the most successful prophylactic in medical history, there is now an emerging interest as to whether vaccination can be applied in autoimmune and inflammatory conditions. These are diseases of failed immune regulation; vaccination in this context aims to exploit the power of antigenic material to stimulate immune homeostasis in the form of active, adaptive, regulatory immune responses. Type 1 diabetes is an autoimmune disease that could benefit from the therapeutic potential of vaccination. The major conditions necessary to make prophylaxis feasible are in place; the self antigens are known, the failure of existing immune regulation has been demonstrated, early studies of vaccine approaches have proved safe, and the preclinical prodrome of the disease can be easily detected by simple blood tests. Challenges for future implementation include finding the best mode of delivery and the best blend of adjunctive therapies that create the favorable conditions required for a vaccine to be effective.
Collapse
Affiliation(s)
- Mark Peakman
- Department of Immunobiology, King's College London and National Institute for Health Research Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT UK
| |
Collapse
|
37
|
Levy H, Wang X, Kaldunski M, Jia S, Kramer J, Pavletich SJ, Reske M, Gessel T, Yassai M, Quasney MW, Dahmer MK, Gorski J, Hessner MJ. Transcriptional signatures as a disease-specific and predictive inflammatory biomarker for type 1 diabetes. Genes Immun 2012; 13:593-604. [PMID: 22972474 PMCID: PMC4265236 DOI: 10.1038/gene.2012.41] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The complex milieu of inflammatory mediators associated with many diseases is often too dilute to directly measure in the periphery, necessitating development of more sensitive measurements suitable for mechanistic studies, earlier diagnosis, guiding therapeutic decisions and monitoring interventions. We previously demonstrated that plasma samples from recent-onset type 1 diabetes (RO T1D) patients induce a proinflammatory transcriptional signature in freshly drawn peripheral blood mononuclear cells (PBMCs) relative to that of unrelated healthy controls (HC). Here, using cryopreserved PBMC, we analyzed larger RO T1D and HC cohorts, examined T1D progression in pre-onset samples, and compared the RO T1D signature to those associated with three disorders characterized by airway infection and inflammation. The RO T1D signature, consisting of interleukin-1 cytokine family members, chemokines involved in immunocyte chemotaxis, immune receptors and signaling molecules, was detected during early pre-diabetes and found to resolve post-onset. The signatures associated with cystic fibrosis patients chronically infected with Pseudomonas aeruginosa, patients with confirmed bacterial pneumonia, and subjects with H1N1 influenza all reflected immunological activation, yet each were distinct from one another and negatively correlated with that of T1D. This study highlights the remarkable capacity of cells to serve as biosensors capable of sensitively and comprehensively differentiating immunological states.
Collapse
Affiliation(s)
- H Levy
- The Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Verrijn Stuart AA, de Jager W, Klein MR, Teklenburg G, Nuboer R, Hoorweg JJG, de Vroede MAMJ, de Kruijff I, Fick M, Schroor EJ, van der Vlist GJ, Meerding J, Kamphuis S, Prakken BJ. Recognition of heat shock protein 60 epitopes in children with type 1 diabetes. Diabetes Metab Res Rev 2012; 28:527-34. [PMID: 22492505 DOI: 10.1002/dmrr.2306] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Treatment with a specific HSP60 epitope in new onset of type 1 diabetes (T1D) patients has been shown to preserve endogenous insulin production. Previously, recognition of pan HLA-DR-binding HSP60 epitopes in various autoimmune diseases was found; this study investigated recognition of these epitopes in newly diagnosed T1D patients and correlated findings to the occurrence of a partial remission. METHODS Peripheral blood mononuclear cells of 18 children with T1D were prospectively collected at disease onset and a few months after diagnosis. Epitope-specific T-cell proliferation and cytokine production (intracellular and in culture supernatants) were measured. Results were compared with 31 longstanding T1D patients and ten healthy controls. RESULTS Although HSP60 epitope-specific T-cell proliferative responses were detected, overall proliferative responses were low. At onset, epitope-specific intracellular IFN-γ production was higher in T1D patients compared with healthy controls (p < 0.05). At follow-up, both IL-10 and IFN-γ production were higher in those without a partial remission than in those with a partial remission (both p < 0.05). Also, IL-10 and IFN-γ production were higher compared with onset for patients without a PR (both p < 0.01). In supernatants of HSP60 epitope-specific T-cell cultures, no substantial differences in cytokine production were found between T1D patients with and without a partial remission, either at onset or a few months after onset. As patient numbers were small, results should be interpreted with caution. CONCLUSIONS Pan-DR-binding HSP60 peptides induced low peptide-specific proliferative responses and peptide-specific production of some, mainly intracellular, cytokines in T1D patients. Recognition did not differ significantly between patient groups and various time points.
Collapse
Affiliation(s)
- A A Verrijn Stuart
- Department of Paediatric Endocrinology, University Medical Center Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
van der Burg SH, Kalos M, Gouttefangeas C, Janetzki S, Ottensmeier C, Welters MJP, Romero P, Britten CM, Hoos A. Harmonization of immune biomarker assays for clinical studies. Sci Transl Med 2012; 3:108ps44. [PMID: 22072636 DOI: 10.1126/scitranslmed.3002785] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Assays that measure a patient's immune response play an increasingly important role in the development of immunotherapies. The inherent complexity of these assays and independent protocol development between laboratories result in high data variability and poor reproducibility. Quality control through harmonization--based on integration of laboratory-specific protocols with standard operating procedures and assay performance benchmarks--is one way to overcome these limitations. Harmonization guidelines can be widely implemented to address assay performance variables. This process enables objective interpretation and comparison of data across clinical trial sites and also facilitates the identification of relevant immune biomarkers, guiding the development of new therapies.
Collapse
Affiliation(s)
- Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Classical genetic studies established a link between Type 1 diabetes, a common childhood autoimmune disease and genes that encode MHC antigens and several immune-related determinants. The mechanisms by which these genes contribute to the initiation and perpetuation of Type 1 diabetes remain enigmatic. Emerging data indicate a role for epigenetic mechanisms involving hyperacetylation of histones in the differential gene expression and amelioration of autoimmune diabetes in a mouse model. In this article the implications of these and other epigenetic mechanisms including ncRNA-mediated gene regulation in the abrogation of autoimmune diabetes are discussed. Concerted efforts to decipher the epigenetics of Type 1 diabetes may provide novel perspectives on autoimmune diabetogenesis.
Collapse
Affiliation(s)
- Sundararajan Jayaraman
- Deptartment of Medicine, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
41
|
Waldron-Lynch F, Henegariu O, Deng S, Preston-Hurlburt P, Tooley J, Flavell R, Herold KC. Teplizumab induces human gut-tropic regulatory cells in humanized mice and patients. Sci Transl Med 2012; 4:118ra12. [PMID: 22277969 PMCID: PMC4131554 DOI: 10.1126/scitranslmed.3003401] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development and optimization of immune therapies in patients has been hampered by the lack of preclinical models in which their effects on human immune cells can be studied. As a result, observations that have been made in preclinical studies have suggested mechanisms of drug action in murine models that have not been confirmed in clinical studies. Here, we used a humanized mouse reconstituted with human hematopoietic stem cells to study the mechanism of action of teplizumab, an Fc receptor nonbinding humanized monoclonal antibody to CD3 being tested in clinical trials for the treatment of patients with type 1 diabetes mellitus. In this model, human gut-tropic CCR6(+) T cells exited the circulation and secondary lymph organs and migrated to the small intestine. These cells then produced interleukin-10 (IL-10), a regulatory cytokine, in quantities that could be detected in the peripheral circulation. Blocking T cell migration to the small intestine with natalizumab, which prevents cellular adhesion by inhibiting α(4) integrin binding, abolished the treatment effects of teplizumab. Moreover, IL-10 expression by CD4(+)CD25(high)CCR6(+)FoxP3 cells returning to the peripheral circulation was increased in patients with type 1 diabetes treated with teplizumab. These findings demonstrate that humanized mice may be used to identify novel immunologic mechanisms that occur in patients treated with immunomodulators.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- CD3 Complex/immunology
- Cell Movement/drug effects
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Forkhead Transcription Factors/metabolism
- Gastrointestinal Tract/cytology
- Gastrointestinal Tract/drug effects
- Gastrointestinal Tract/immunology
- Humans
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Interleukin-10/metabolism
- Intestine, Small/cytology
- Intestine, Small/drug effects
- Intestine, Small/immunology
- L-Selectin/metabolism
- Mice
- Mucous Membrane/cytology
- Mucous Membrane/drug effects
- Mucous Membrane/immunology
- Natalizumab
- Oligonucleotide Array Sequence Analysis
- Receptors, CCR6/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Frank Waldron-Lynch
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Roep BO, Buckner J, Sawcer S, Toes R, Zipp F. The problems and promises of research into human immunology and autoimmune disease. Nat Med 2012; 18:48-53. [PMID: 22227672 DOI: 10.1038/nm.2626] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Bart O Roep
- Leiden University Medical Center, National Diabetes Expert Center, Department of Immunohaematology and Blood Transfusion, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
43
|
Coppieters KT, Dotta F, Amirian N, Campbell PD, Kay TWH, Atkinson MA, Roep BO, von Herrath MG. Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. ACTA ACUST UNITED AC 2012; 209:51-60. [PMID: 22213807 PMCID: PMC3260877 DOI: 10.1084/jem.20111187] [Citation(s) in RCA: 518] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In situ tetramer staining reveals the presence of islet antigen-reactive CD8+ T cells in pancreatic islets from deceased type 1 diabetes patients. A direct association of islet-autoreactive T cells with β cell destruction in human pancreatic islets from type 1 diabetes (T1D) patients has never been demonstrated, and little is known about disease progression after diagnosis. Frozen pancreas samples were obtained from 45 cadaveric T1D donors with disease durations ranging from 1 wk to >50 yr, 14 nondiabetic controls, 5 nondiabetics with islet autoantibodies, 2 cases of gestational diabetes, and 6 T2D patients. Sections were systematically analyzed for the presence of insulin-sufficient β cells, CD8+ insulitic lesions, and HLA class I hyperexpression. Finally, consecutive sections from HLA-A2–expressing individuals were probed for CD8 T cell reactivity against six defined islet autoantigens associated with T1D by in situ tetramer staining. Both single and multiple CD8 T cell autoreactivities were detected within individual islets in a subset of patients up to 8 yr after clinical diagnosis. Pathological features such as HLA class I hyperexpression and insulitis were specific for T1D and persisted in a small portion of the patients with longstanding disease. Insulitic lesions consistently presented in a multifocal pattern with varying degrees of infiltration and β cell loss across affected organs. Our observations provide the first direct proof for islet autoreactivity within human islets and underscore the heterogeneous and chronic disease course.
Collapse
Affiliation(s)
- Ken T Coppieters
- Type 1 Diabetes Center, the La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Induction of protective genes leads to islet survival and function. J Transplant 2011; 2011:141898. [PMID: 22220267 PMCID: PMC3246756 DOI: 10.1155/2011/141898] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 09/01/2011] [Indexed: 12/16/2022] Open
Abstract
Islet transplantation is the most valid approach to the treatment of type 1 diabetes. However, the function of transplanted islets is often compromised since a large number of β cells undergo apoptosis induced by stress and the immune rejection response elicited by the recipient after transplantation. Conventional treatment for islet transplantation is to administer immunosuppressive drugs to the recipient to suppress the immune rejection response mounted against transplanted islets. Induction of protective genes in the recipient (e.g., heme oxygenase-1 (HO-1), A20/tumor necrosis factor alpha inducible protein3 (tnfaip3), biliverdin reductase (BVR), Bcl2, and others) or administration of one or more of the products of HO-1 to the donor, the islets themselves, and/or the recipient offers an alternative or synergistic approach to improve islet graft survival and function. In this perspective, we summarize studies describing the protective effects of these genes on islet survival and function in rodent allogeneic and xenogeneic transplantation models and the prevention of onset of diabetes, with emphasis on HO-1, A20, and BVR. Such approaches are also appealing to islet autotransplantation in patients with chronic pancreatitis after total pancreatectomy, a procedure that currently only leads to 1/3 of transplanted patients being diabetes-free.
Collapse
|
45
|
Roep BO, Peakman M. Diabetogenic T lymphocytes in human Type 1 diabetes. Curr Opin Immunol 2011; 23:746-53. [PMID: 22051340 DOI: 10.1016/j.coi.2011.10.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 10/12/2011] [Indexed: 01/10/2023]
Abstract
The field of Type 1 diabetes research has been quick to embrace the era of translational medicine in the recent epoch. Building upon some 30 years of intense immunological research, the past decade has been marked by a series of clinical trials designed to evaluate the potential beneficial effects of a range of immune intervention and prevention strategies [1(••),2-5]. At the heart of Type 1 diabetes is an autoimmune process, the consequence of which is immune-mediated destruction of islet β-cells. Although understanding the pathogenesis of islet autoimmunity is critical, there are also good reasons to focus research onto the β-cell destructive process itself. Measuring preservation of function of insulin-producing cells is currently the best means available to evaluate potential beneficial effects of immunotherapy, but there is an urgent need to discover and monitor immunological correlates of this β-cell destructive process. Whilst the best approach to intervention and prevention has yet to emerge, it is logical that future attempts to intelligently design therapeutics for Type 1 diabetes will need to be predicated on a clear understanding of the process of β-cell destruction and the immune components involved. For these reasons, this review will focus on the role of diabetogenic T lymphocytes in this disease-defining event.
Collapse
Affiliation(s)
- Bart O Roep
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands.
| | | |
Collapse
|
46
|
Affiliation(s)
- Bart O Roep
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden NL-2300RC, Netherlands.
| |
Collapse
|
47
|
T cell recognition of autoantigens in human type 1 diabetes: clinical perspectives. Clin Dev Immunol 2011; 2011:513210. [PMID: 21785617 PMCID: PMC3140193 DOI: 10.1155/2011/513210] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 03/18/2011] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease driven by the activation of lymphocytes against pancreatic β-cells. Among β-cell autoantigens, preproinsulin has been ascribed a key role in the T1D process. The successive steps that control the activation of autoreactive lymphocytes have been extensively studied in animal models of T1D, but remains ill defined in man. In man, T lymphocytes, especially CD8+ T cells, are predominant within insulitis. Developing T-cell assays in diabetes autoimmunity is, thus, a major challenge. It is expected to help defining autoantigens and epitopes that drive the disease process, to pinpoint key functional features of epitope-specific T lymphocytes along the natural history of diabetes and to pave the way towards therapeutic strategies to induce immune tolerance to β-cells. New T-cell technologies will allow defining autoreactive T-cell differentiation programs and characterizing autoimmune responses in comparison with physiologically appropriate immune responses. This may prove instrumental in the discovery of immune correlates of efficacy in clinical trials.
Collapse
|
48
|
Waldron-Lynch F, Herold KC. Immunomodulatory therapy to preserve pancreatic β-cell function in type 1 diabetes. Nat Rev Drug Discov 2011; 10:439-52. [DOI: 10.1038/nrd3402] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
49
|
Atkinson MA, Bluestone JA, Eisenbarth GS, Hebrok M, Herold KC, Accili D, Pietropaolo M, Arvan PR, Von Herrath M, Markel DS, Rhodes CJ. How does type 1 diabetes develop?: the notion of homicide or β-cell suicide revisited. Diabetes 2011; 60:1370-9. [PMID: 21525508 PMCID: PMC3292309 DOI: 10.2337/db10-1797] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 02/17/2011] [Indexed: 12/16/2022]
Affiliation(s)
- Mark A Atkinson
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Coppieters KT, Roep BO, von Herrath MG. Beta cells under attack: toward a better understanding of type 1 diabetes immunopathology. Semin Immunopathol 2010; 33:1-7. [PMID: 21170533 DOI: 10.1007/s00281-010-0236-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 12/07/2010] [Indexed: 11/29/2022]
Affiliation(s)
- Ken T Coppieters
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | |
Collapse
|