1
|
Dendritic Cells and Their Immunotherapeutic Potential for Treating Type 1 Diabetes. Int J Mol Sci 2022; 23:ijms23094885. [PMID: 35563276 PMCID: PMC9099521 DOI: 10.3390/ijms23094885] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic beta cells through a process that is primarily mediated by T cells. Emerging evidence suggests that dendritic cells (DCs) play a crucial role in initiating and developing this debilitating disease. DCs are professional antigen-presenting cells with the ability to integrate signals arising from tissue infection or injury that present processed antigens from these sites to naïve T cells in secondary lymphoid organs, thereby triggering naïve T cells to differentiate and modulate adaptive immune responses. Recent advancements in our knowledge of the various subsets of DCs and their cellular structures and methods of orchestration over time have resulted in a better understanding of how the T cell response is shaped. DCs employ various arsenal to maintain their tolerance, including the induction of effector T cell deletion or unresponsiveness and the generation and expansion of regulatory T cell populations. Therapies that suppress the immunogenic effects of dendritic cells by blocking T cell costimulatory pathways and proinflammatory cytokine production are currently being sought. Moreover, new strategies are being developed that can regulate DC differentiation and development and harness the tolerogenic capacity of these cells. Here, in this report, we focus on recent advances in the field of DC immunology and evaluate the prospects of DC-based therapeutic strategies to treat T1D.
Collapse
|
2
|
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
|
3
|
Rodriguez-Calvo T, Johnson JD, Overbergh L, Dunne JL. Neoepitopes in Type 1 Diabetes: Etiological Insights, Biomarkers and Therapeutic Targets. Front Immunol 2021; 12:667989. [PMID: 33953728 PMCID: PMC8089389 DOI: 10.3389/fimmu.2021.667989] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms underlying type 1 diabetes (T1D) pathogenesis remain largely unknown. While autoantibodies to pancreatic beta-cell antigens are often the first biological response and thereby a useful biomarker for identifying individuals in early stages of T1D, their role in T1D pathogenesis is not well understood. Recognition of these antigenic targets by autoreactive T-cells plays a pathological role in T1D development. Recently, several beta-cell neoantigens have been described, indicating that both neoantigens and known T1D antigens escape central or peripheral tolerance. Several questions regarding the mechanisms by which tolerance is broken in T1D remain unanswered. Further delineating the timing and nature of antigenic responses could allow their use as biomarkers to improve staging, as targets for therapeutic intervention, and lead to a better understanding of the mechanisms leading to loss of tolerance. Multiple factors that contribute to cellular stress may result in the generation of beta-cell derived neoepitopes and contribute to autoimmunity. Understanding the cellular mechanisms that induce beta-cells to produce neoantigens has direct implications on development of therapies to intercept T1D disease progression. In this perspective, we will discuss evidence for the role of neoantigens in the pathogenesis of T1D, including antigenic responses and cellular mechanisms. We will additionally discuss the pathways leading to neoepitope formation and the cross talk between the immune system and the beta-cells in this regard. Ultimately, delineating the timing of neoepitope generation in T1D pathogenesis will determine their role as biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Teresa Rodriguez-Calvo
- Institute of Diabetes Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Munich, Germany
| | - James D. Johnson
- Diabetes Research Group, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Lut Overbergh
- Laboratory Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Jessica L. Dunne
- Janssen Research and Development, LLC, Raritan, NJ, United States
| |
Collapse
|
4
|
von Herrath M, Bain SC, Bode B, Clausen JO, Coppieters K, Gaysina L, Gumprecht J, Hansen TK, Mathieu C, Morales C, Mosenzon O, Segel S, Tsoukas G, Pieber TR. Anti-interleukin-21 antibody and liraglutide for the preservation of β-cell function in adults with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Diabetes Endocrinol 2021; 9:212-224. [PMID: 33662334 DOI: 10.1016/s2213-8587(21)00019-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Type 1 diabetes is characterised by progressive loss of functional β-cell mass, necessitating insulin treatment. We aimed to investigate the hypothesis that combining anti-interleukin (IL)-21 antibody (for low-grade and transient immunomodulation) with liraglutide (to improve β-cell function) could enable β-cell survival with a reduced risk of complications compared with traditional immunomodulation. METHODS This randomised, parallel-group, placebo-controlled, double-dummy, double-blind, phase 2 trial was done at 94 sites (university hospitals and medical centres) in 17 countries. Eligible participants were adults aged 18-45 years with recently diagnosed type 1 diabetes and residual β-cell function. Individuals with unstable type 1 diabetes (defined by an episode of severe diabetic ketoacidosis within 2 weeks of enrolment) or active or latent chronic infections were excluded. Participants were randomly assigned (1:1:1:1), with stratification by baseline stimulated peak C-peptide concentration (mixed-meal tolerance test [MMTT]), to the combination of anti-IL-21 and liraglutide, anti-IL-21 alone, liraglutide alone, or placebo, all as an adjunct to insulin. Investigators, participants, and funder personnel were masked throughout the treatment period. The primary outcome was the change in MMTT-stimulated C-peptide concentration at week 54 (end of treatment) relative to baseline, measured via the area under the concentration-time curve (AUC) over a 4 h period for the full analysis set (intention-to-treat population consisting of all participants who were randomly assigned). After treatment cessation, participants were followed up for an additional 26-week off-treatment observation period. This trial is registered with ClinicalTrials.gov, NCT02443155. FINDINGS Between Nov 10, 2015, and Feb 27, 2019, 553 adults were assessed for eligibility, of whom 308 were randomly assigned to receive either anti-IL-21 plus liraglutide, anti-IL-21, liraglutide, or placebo (77 assigned to each group). Compared with placebo (ratio to baseline 0·61, 39% decrease), the decrease in MMTT-stimulated C-peptide concentration from baseline to week 54 was significantly smaller with combination treatment (0·90, 10% decrease; estimated treatment ratio 1·48, 95% CI 1·16-1·89; p=0·0017), but not with anti-IL-21 alone (1·23, 0·97-1·57; p=0·093) or liraglutide alone (1·12, 0·87-1·42; p=0·38). Despite greater insulin use in the placebo group, the decrease in HbA1c (a key secondary outcome) at week 54 was greater with all active treatments (-0·50 percentage points) than with placebo (-0·10 percentage points), although the differences versus placebo were not significant. The effects diminished upon treatment cessation. Changes in immune cell subsets across groups were transient and mild (<10% change over time). The most frequently reported adverse events included gastrointestinal disorders, in keeping with the known side-effect profile of liraglutide. The rate of hypoglycaemic events did not differ significantly between active treatment groups and placebo, with an exception of a lower rate in the liraglutide group than in the placebo group during the treatment period. No events of diabetic ketoacidosis were observed. One participant died while on liraglutide (considered unlikely to be related to trial treatment) in connection with three reported adverse events (hypoglycaemic coma, pneumonia, and brain oedema). INTERPRETATION The combination of anti-IL-21 and liraglutide could preserve β-cell function in recently diagnosed type 1 diabetes. The efficacy of this combination appears to be similar to that seen in trials of other disease-modifying interventions in type 1 diabetes, but with a seemingly better safety profile. Efficacy and safety should be further evaluated in a phase 3 trial programme. FUNDING Novo Nordisk.
Collapse
Affiliation(s)
| | | | - Bruce Bode
- Atlanta Diabetes Associates, Atlanta, GA, USA; Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | - Chantal Mathieu
- Clinical and Experimental Endocrinology, UZ Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Cristobal Morales
- Endocrinology and Nutrition Department, Virgen Macarena Hospital, Seville, Spain
| | - Ofri Mosenzon
- Diabetes Unit, Department of Endocrinology and Metabolism, Hadassah Medical Centre, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - George Tsoukas
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Thomas R Pieber
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | | |
Collapse
|
5
|
Volfson-Sedletsky V, Jones A, Hernandez-Escalante J, Dooms H. Emerging Therapeutic Strategies to Restore Regulatory T Cell Control of Islet Autoimmunity in Type 1 Diabetes. Front Immunol 2021; 12:635767. [PMID: 33815387 PMCID: PMC8015774 DOI: 10.3389/fimmu.2021.635767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Despite many decades of investigation uncovering the autoimmune mechanisms underlying Type 1 Diabetes (T1D), translating these findings into effective therapeutics has proven extremely challenging. T1D is caused by autoreactive T cells that become inappropriately activated and kill the β cells in the pancreas, resulting in insulin insufficiency and hyperglycemia. A large body of evidence supports the idea that the unchecked activation and expansion of autoreactive T cells in T1D is due to defects in immunosuppressive regulatory T cells (Tregs) that are critical for maintaining peripheral tolerance to islet autoantigens. Hence, repairing these Treg deficiencies is a much sought-after strategy to treat the disease. To accomplish this goal in the most precise, effective and safest way possible, restored Treg functions will need to be targeted towards suppressing the autoantigen-specific immune responses only and/or be localized in the pancreas. Here we review the most recent developments in designing Treg therapies that go beyond broad activation or expansion of non-specific polyclonal Treg populations. We focus on two cutting-edge strategies namely ex vivo generation of optimized Tregs for re-introduction in T1D patients vs direct in situ stimulation and restoration of endogenous Treg function.
Collapse
Affiliation(s)
- Victoria Volfson-Sedletsky
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Albert Jones
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Jaileene Hernandez-Escalante
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Hans Dooms
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
6
|
Bassin EJ, Piganelli JD, Little SR. Auto-antigen and Immunomodulatory Agent-Based Approaches for Antigen-Specific Tolerance in NOD Mice. Curr Diab Rep 2021; 21:9. [PMID: 33547977 DOI: 10.1007/s11892-021-01376-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) can be managed by insulin replacement, but it is still associated with an increased risk of microvascular/cardiovascular complications. There is considerable interest in antigen-specific approaches for treating T1D due to their potential for a favorable risk-benefit ratio relative to non-specific immune-based treatments. Here we review recent antigen-specific tolerance approaches using auto-antigen and/or immunomodulatory agents in NOD mice and provide insight into seemingly contradictory findings. RECENT FINDINGS Although delivery of auto-antigen alone can prevent T1D in NOD mice, this approach may be prone to inconsistent results and has not demonstrated an ability to reverse established T1D. Conversely, several approaches that promote presentation of auto-antigen in a tolerogenic context through cell/tissue targeting, delivery system properties, or the delivery of immunomodulatory agents have had success in reversing recent-onset T1D in NOD mice. While initial auto-antigen based approaches were unable to substantially influence T1D progression clinically, recent antigen-specific approaches have promising potential.
Collapse
Affiliation(s)
- Ethan J Bassin
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jon D Piganelli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh, 4401 Penn Avenue, 6125 Rangos Research Center, Pittsburgh, PA, 15224, USA.
| | - Steven R Little
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Chemical Engineering, University of Pittsburgh, 3700 O'Hara Street, 940 Benedum Hall, Pittsburgh, PA, 15261, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmaceutical Science, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Mao RF, Chen YY, Zhang J, Chang X, Wang YF. Type 1 diabetes mellitus and its oral tolerance therapy. World J Diabetes 2020; 11:400-415. [PMID: 33133388 PMCID: PMC7582116 DOI: 10.4239/wjd.v11.i10.400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/27/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
As a T cell-mediated autoimmune disease, type 1 diabetes mellitus (T1DM) is marked by insulin defect resulting from the destruction of pancreatic β-cells. The understanding of various aspects of T1DM, such as its epidemiology, pathobiology, pathogenesis, clinical manifestations, and complications, has been greatly promoted by valuable research performed during the past decades. However, these findings have not been translated into an effective treatment. The ideal treatment should safely repair the destroyed immune balance in a long-lasting manner, preventing or stopping the destruction of β-cells. As a type of immune hypo-responsiveness to the orally administrated antigen, oral tolerance may be induced by enhancement of regulatory T cells (Tregs) or by anergy/deletion of T cells, depending on the dosage of orally administrated antigen. Acting as an antigen-specific immunotherapy, oral tolerance therapy for T1DM has been mainly performed using animal models and some clinical trials have been completed or are still ongoing. Based on the review of the proposed mechanism of the development of T1DM and oral tolerance, we give a current overview of oral tolerance therapy for T1DM conducted in both animal models and clinical trials.
Collapse
Affiliation(s)
- Rui-Feng Mao
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Science, Huaiyin Normal University, Huai'an 223300, Jiangsu Province, China
| | - Ying-Ying Chen
- School of Life Sciences, Huaiyin Normal University, Huai'an 223300, Jiangsu Province, China
| | - Ji Zhang
- School of Life Sciences, Huaiyin Normal University, Huai'an 223300, Jiangsu Province, China
| | - Xin Chang
- Department of Ultrasound Medicine, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, Jiangsu Province, China
| | - Ye-Fu Wang
- College of Life Sciences, Wuhan University, Wuhan 430072, Hubei Province, China
| |
Collapse
|
8
|
Wang Q, Ren L, Wan Y, Prud'homme GJ. GABAergic regulation of pancreatic islet cells: Physiology and antidiabetic effects. J Cell Physiol 2019; 234:14432-14444. [PMID: 30693506 DOI: 10.1002/jcp.28214] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/15/2019] [Indexed: 01/24/2023]
Abstract
Diabetes occurs when pancreatic β-cell death exceeds β-cell growth, which leads to loss of β-cell mass. An effective therapy must have two actions: promotion of β-cell replication and suppression of β-cell death. Previous studies have established an important role for γ-aminobutyric acid (GABA) in islet-cell hormone homeostasis, as well as the maintenance of the β-cell mass. GABA exerts paracrine actions on α cells in suppressing glucagon secretion, and it has autocrine actions on β cells that increase insulin secretion. Multiple studies have shown that GABA increases the mitotic rate of β cells. In mice, following β-cell depletion with streptozotocin, GABA therapy can restore the β-cell mass. Enhanced β-cell replication appears to depend on growth and survival pathways involving Akt activation. Some studies have also suggested that it induces transdifferentiation of α cells into β cells, but this has been disputed and requires further investigation. In addition to proliferative effects, GABA protects β cells against injury and markedly reduces their apoptosis under a variety of conditions. The antiapoptotic effects depend at least in part on the enhancement of sirtuin-1 and Klotho activity, which both inhibit activation of the NF-κB inflammatory pathway. Importantly, in xenotransplanted human islets, GABA therapy stimulates β-cell replication and insulin secretion. Thus, the intraislet GABAergic system is a target for the amelioration of diabetes therapy, including β-cell survival and regeneration. GABA (or GABAergic drugs) can be combined with other antidiabetic drugs for greater effect.
Collapse
Affiliation(s)
- Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Liwei Ren
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Gerald J Prud'homme
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Vazquez-Mateo C, Collins J, Goldberg SJ, Lawson M, Hernandez-Escalante J, Dooms H. Combining anti-IL-7Rα antibodies with autoantigen-specific immunotherapy enhances non-specific cytokine production but fails to prevent Type 1 Diabetes. PLoS One 2019; 14:e0214379. [PMID: 30908554 PMCID: PMC6433345 DOI: 10.1371/journal.pone.0214379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/12/2019] [Indexed: 01/09/2023] Open
Abstract
Autoantigen-specific methods to prevent and treat Type 1 Diabetes (T1D) carry high hopes to permanently cure this disease, but have largely failed in clinical trials. One suggested approach to increase the efficacy of islet antigen-specific vaccination is to combine it with a modulator of the T cell response, with the goal of reducing effector differentiation and promoting regulatory T cells (Tregs). Here we asked if addition of antibodies that block the IL-7/IL-7Rα pathway altered the T cell response to islet antigen vaccination and prevented T1D in non-obese diabetic (NOD) mice. Anti-IL-7Rα monoclonal antibodies (mAbs) reduced the numbers of islet antigen-specific T cells generated after vaccination with islet peptides and alum. However, addition of anti-IL-7Rα antibodies to peptide/alum vaccination unexpectedly increased non-specific IFN-γ, IL-2 and IL-10 cytokine production and did not result in improved prevention of T1D onset. In a second approach, we used a conjugate vaccine to deliver islet autoantigens, using Keyhole Limpet Hemocyanin (KLH) as a carrier. Islet antigen-KLH vaccination led to a significant expansion of antigen-specific Tregs and delayed diabetes onset in NOD mice. These outcomes were not further improved by addition of anti-IL-7Rα antibodies. To the contrary, blocking IL-7Rα during vaccination led to non-specific cytokine production and reduced the efficacy of a KLH-conjugated vaccine to prevent T1D. Our study thus revealed that adding anti-IL-7Rα antibodies during autoantigen immunization did not improve the efficacy of such vaccinations to prevent T1D, despite altering some aspects of the T cell response in a potentially advantageous way. Further refinement of this approach will be required to separate the beneficial from the adverse effects of anti-IL-7Rα antibodies to treat autoimmune disease.
Collapse
Affiliation(s)
- Cristina Vazquez-Mateo
- Arthritis Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Justin Collins
- Arthritis Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sarah J. Goldberg
- Arthritis Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Maxx Lawson
- Arthritis Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jaileene Hernandez-Escalante
- Arthritis Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Hans Dooms
- Arthritis Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
10
|
Xin GLL, Khee YP, Ying TY, Chellian J, Gupta G, Kunnath AP, Nammi S, Collet T, Hansbro PM, Dua K, Chellappan DK. Current Status on Immunological Therapies for Type 1 Diabetes Mellitus. Curr Diab Rep 2019; 19:22. [PMID: 30905013 DOI: 10.1007/s11892-019-1144-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) occurs when there is destruction of beta cells within the islets of Langerhans in the pancreas due to autoimmunity. It is considered a complex disease, and different complications can surface and worsen the condition if T1D is not managed well. Since it is an incurable disease, numerous treatments and therapies have been postulated in order to control T1D by balancing hyperglycemia control while minimizing hypoglycemic episodes. The purpose of this review is to primarily look into the current state of the available immunological therapies and their advantages for the treatment of T1D. RECENT FINDINGS Over the years, immunological therapy has become the center of attraction to treat T1D. Immunomodulatory approaches on non-antigens involving agents such as cyclosporine A, mycophenolate mofetil, anti-CD20, cytotoxic T cells, anti-TNF, anti-CD3, and anti-thymocyte globulin as well as immunomodulative approaches on antigens such as insulin, glutamic acid decarboxylase, and heat shock protein 60 have been studied. Aside from these two approaches, studies and trials have also been conducted on regulatory T cells, dendritic cells, interleukin 2, interleukin 4, M2 macrophages, and rapamycin/interleukin 2 combination therapy to test their effects on patients with T1D. Many of these agents have successfully suppressed T1D in non-obese diabetic (NOD) mice and in human trials. However, some have shown negative results. To date, the insights into the management of the immune system have been increasing rapidly to search for potential therapies and treatments for T1D. Nevertheless, some of the challenges are still inevitable. A lot of work and effort need to be put into the investigation on T1D through immunological therapy, particularly to reduce complications to improve and enhance clinical outcomes.
Collapse
Affiliation(s)
- Griselda Lim Loo Xin
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Yap Pui Khee
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Tan Yoke Ying
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, 302017, India
| | - Anil Philip Kunnath
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Sydney, NSW, 2751, Australia
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, 2751, Australia
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, 4059, Australia
| | - Philip Michael Hansbro
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, Newcastle, NSW, 2308, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, Newcastle, NSW, 2308, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
11
|
Nakayama M, Michels AW. Determining Antigen Specificity of Human Islet Infiltrating T Cells in Type 1 Diabetes. Front Immunol 2019; 10:365. [PMID: 30906293 PMCID: PMC6418007 DOI: 10.3389/fimmu.2019.00365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/13/2019] [Indexed: 01/07/2023] Open
Abstract
Type 1 diabetes, the immune mediated form of diabetes, represents a prototypical organ specific autoimmune disease in that insulin producing pancreatic islets are specifically targeted by T cells. The disease is now predictable in humans with the measurement of type 1 diabetes associated autoantibodies (islet autoantibodies) in the peripheral blood which are directed against insulin and beta cell proteins. With an increasing incidence of disease, especially in young children, large well-controlled clinical prevention trials using antigen specific immunotherapy have been completed but with limited clinical benefit. To improve outcomes, it is critical to understand the antigen and T cell receptor repertoires of those cells that infiltrate the target organ, pancreatic islets, in human type 1 diabetes. With international networks to identify organ donors with type 1 diabetes, improved immunosequencing platforms, and the ability to reconstitute T cell receptors of interest into immortalized cell lines allows antigen discovery efforts for rare tissue specific T cells. Here we review the disease pathogenesis of type 1 diabetes with a focus on human islet infiltrating T cell antigen discovery efforts, which provides necessary knowledge to define biomarkers of disease activity and improve antigen specific immunotherapy approaches for disease prevention.
Collapse
Affiliation(s)
- Maki Nakayama
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
12
|
Rosenthal KS, Carambula R, Zimmerman DH. Why Don't We Have a Vaccine Against Autoimmune Diseases? - A Review. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2019; 10:574. [PMID: 31328022 PMCID: PMC6640150 DOI: 10.4172/2155-9899.1000574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
This review examines some of the reasons why we don't have a vaccine against autoimmune diseases and highlights the progress that has been made. Many autoimmune diseases, such as rheumatoid arthritis (RA), multiple sclerosis (MS) and type 1 diabetes (T1D), are driven by autoimmune T cell responses. Unlike vaccines for most infectious diseases, which elicit antibody responses, are intended for immuno-naive individuals and considered preventative, a vaccine for an autoimmune disease must be therapeutic and resolve or control the on-going autoimmune response and condition in the diseased host. Despite these differences, many of the same considerations for infectious disease vaccines must also be addressed to develop a therapeutic vaccine for autoimmune diseases. The disease initiator/triggers, antigens and autoantigens, nature of the immunopathogenic and protective/therapeutic immune response will be compared for infectious and autoimmune diseases as will approaches for developing vaccines including formulations, animal models and indicators of success. The rationale for a therapeutic vaccine for RA will be discussed in greater detail with a relatively limited discussion of T1D, MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Ken S Rosenthal
- Roseman University College of Medicine, 10530 Discovery Dr, Las Vegas, USA
- Northeast Ohio Medical University, Rootstown, OH, USA
| | | | | |
Collapse
|
13
|
A multi-epitope DNA vaccine enables a broad engagement of diabetogenic T cells for tolerance in Type 1 diabetes. J Autoimmun 2018; 98:13-23. [PMID: 30454875 DOI: 10.1016/j.jaut.2018.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is caused by diabetogenic T cells that evaded tolerance mechanisms and react against multiple β-cell antigens. Antigen-specific therapy to reinstate tolerance (typically using a single β-cell antigen) has so far proved unsuccessful in T1D patients. Plasmid DNA (pDNA)-mediated expression of proinsulin has demonstrated transient protection in clinical trials, but long-lasting tolerance is yet to be achieved. We aimed to address whether pDNA delivery of multiple epitopes/mimotopes from several β-cell antigens efficiently presented to CD4+ and CD8+ T cells could also induce tolerance. This approach significantly delayed T1D development, while co-delivery of pDNA vectors expressing four full antigens protected more mice. Delivery of multiple epitopes resulted in a broad engagement of specific T cells, eliciting a response distinct from endogenous epitopes draining from islets. T-cell phenotypes also varied with antigen specificity. Unexpectedly, the repertoire of T cells reactive to the same epitope was highly polyclonal. Despite induction of some CD25+ Foxp3+ regulatory T cells, protection from disease did not persist after treatment discontinuation. These data demonstrate that epitope-based tolerogenic DNA vaccines constitute effective precision medicine tools to target a broad range of specific CD4+ and CD8+ diabetogenic T-cell populations for prevention or treatment of T1D.
Collapse
|
14
|
Cook DP, Gysemans C, Mathieu C. Lactococcus lactis As a Versatile Vehicle for Tolerogenic Immunotherapy. Front Immunol 2018; 8:1961. [PMID: 29387056 PMCID: PMC5776164 DOI: 10.3389/fimmu.2017.01961] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022] Open
Abstract
Genetically modified Lactococcus lactis bacteria have been engineered as a tool to deliver bioactive proteins to mucosal tissues as a means to exert both local and systemic effects. They have an excellent safety profile, the result of years of human consumption in the food industry, as well as a lack of toxicity and immunogenicity. Also, containment strategies have been developed to promote further application as clinical protein-based therapeutics. Here, we review technological advancements made to enhanced the potential of L. lactis as live biofactories and discuss some examples of tolerogenic immunotherapies mediated by mucosal drug delivery via L. lactis. Additionally, we highlight their use to induce mucosal tolerance by targeted autoantigen delivery to the intestine as an approach to reverse autoimmune type 1 diabetes.
Collapse
Affiliation(s)
- Dana P Cook
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Knip M. Metabolically inactive insulin: friend or foe in the prevention of autoimmune diabetes? Diabetologia 2017; 60:1382-1384. [PMID: 28580535 DOI: 10.1007/s00125-017-4319-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/09/2017] [Indexed: 11/29/2022]
Abstract
About 20 years ago an American study suggested that daily subcutaneous injections of a metabolically inactive insulin analogue with a single amino acid substitution (aspartic acid instead of phenylalanine) at position 25 of the B chain was as effective as intact insulin in preventing autoimmune diabetes in NOD mice. In this issue of Diabetologia Grönholm et al (DOI: 10.1007/s00125-017-4276-5 ) report that parenteral administration of the same insulin analogue has no preventive effect whatsoever on the development of diabetes in NOD mice; in fact, high doses of the metabolically inactive insulin accelerated disease development. The authors were also unable to show any tolerogenic effect of an insulin peptide mimetope given via a subcutaneous osmotic pump. These data do not support the use of metabolically inactive insulin for the prevention of autoimmune diabetes and question whether insulin alone, intact or inactivated has any role in preventing progression to symptomatic diabetes. Future and ongoing intervention trials in humans with preclinical type 1 diabetes should indicate whether the administration of oral insulin has any protective, neutral or even predisposing effects on the development of symptomatic diabetes.
Collapse
Affiliation(s)
- Mikael Knip
- Children's Hospital, University of Helsinki, and Helsinki University Hospital, P.O. Box 22, (Stenbäckinkatu 11), 00014, Helsinki, Finland.
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland.
- Folkhälsan Research Center, Helsinki, Finland.
| |
Collapse
|
16
|
Vazquez-Mateo C, Collins J, Fleury M, Dooms H. Broad induction of immunoregulatory mechanisms after a short course of anti-IL-7Rα antibodies in NOD mice. BMC Immunol 2017; 18:18. [PMID: 28356069 PMCID: PMC5372316 DOI: 10.1186/s12865-017-0201-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/22/2017] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Type 1 diabetes is an autoimmune disease caused by T cell-mediated destruction of the insulin-producing β-cells in the pancreas. Therefore, approaches that effectively halt the pathogenic T cell response are predicted to have preventive or therapeutic benefit for type 1 diabetes patients. We previously demonstrated that long-term blocking of IL-7 signaling, which is critical for the survival and function of T cells, prevented and reversed type 1 diabetes in non-obese diabetic mice. However, such persistent inhibition of T cell responses raises concerns about causing immunodeficiency. Here, we asked whether a reduced duration of the treatment with anti-IL-7Rα antibodies retained efficacy in preventing diabetes. Moreover, we sought to identify immunoregulatory mechanisms induced by anti-IL-7Rα administration. RESULTS Anti-IL-7Rα antibodies were administered to prediabetic NOD mice for 3 weeks and blood samples were taken at the end of treatment and 2 weeks later to analyze changes in T cell phenotypes in response to IL-7Rα blockade. We found that the co-inhibitory receptors LAG-3, Tim-3 and PD-1 were increased on peripheral blood CD4+ and CD8+ T cells from anti-IL-7Rα-treated mice. Expression of these receptors contributed to reduced T cell cytokine production in response to TCR stimulation. In addition, the frequency of Tregs within the circulating CD4+ T cells was increased at the end of anti-IL-7Rα antibody treatment and these Tregs showed a more activated phenotype. In vitro restimulation assays revealed that effector T cells from anti-IL-7Rα-treated mice were more sensitive to co-inhibitory receptor induction after TCR stimulation. Importantly, these changes were accompanied by delayed type 1 diabetes disease kinetics. CONCLUSIONS Together, our data show that short-term blockade of IL-7Rα induces detectable changes in co-inhibitory receptor expression and Treg frequencies in peripheral blood of NOD mice. These changes appear to have long-lasting effects by delaying or preventing type 1 diabetes incidence. Hence, our study provides further support for using anti-IL-7Rα antibodies to modulate autoreactive T cell responses.
Collapse
Affiliation(s)
- Cristina Vazquez-Mateo
- Department of Medicine, Arthritis Center/Rheumatology Section, Boston University School of Medicine, 72 East Concord Street, E519, Boston, MA, 02118, USA
| | - Justin Collins
- Department of Medicine, Arthritis Center/Rheumatology Section, Boston University School of Medicine, 72 East Concord Street, E519, Boston, MA, 02118, USA
| | - Michelle Fleury
- Department of Medicine, Arthritis Center/Rheumatology Section, Boston University School of Medicine, 72 East Concord Street, E519, Boston, MA, 02118, USA
| | - Hans Dooms
- Department of Medicine, Arthritis Center/Rheumatology Section, Boston University School of Medicine, 72 East Concord Street, E519, Boston, MA, 02118, USA.
| |
Collapse
|
17
|
Verbeke CS, Gordo S, Schubert DA, Lewin SA, Desai RM, Dobbins J, Wucherpfennig KW, Mooney DJ. Multicomponent Injectable Hydrogels for Antigen-Specific Tolerogenic Immune Modulation. Adv Healthc Mater 2017; 6:10.1002/adhm.201600773. [PMID: 28116870 PMCID: PMC5518671 DOI: 10.1002/adhm.201600773] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/14/2016] [Indexed: 12/27/2022]
Abstract
Biomaterial scaffolds that enrich and modulate immune cells in situ can form the basis for potent immunotherapies to elicit immunity or reëstablish tolerance. Here, the authors explore the potential of an injectable, porous hydrogel to induce a regulatory T cell (Treg) response by delivering a peptide antigen to dendritic cells in a noninflammatory context. Two methods are described for delivering the BDC peptide from pore-forming alginate gels in the nonobese diabetic mouse model of type 1 diabetes: encapsulation in poly(lactide-co-glycolide) (PLG) microparticles, or direct conjugation to the alginate polymer. While particle-based delivery leads to antigen-specific T cells responses in vivo, PLG particles alter the phenotype of the cells infiltrating the gels. Following gel-based peptide delivery, transient expansion of endogenous antigen-specific T cells is observed in the draining lymph nodes. Antigen-specific T cells accumulate in the gels, and, strikingly, ≈60% of the antigen-specific CD4+ T cells in the gels are Tregs. Antigen-specific T cells are also enriched in the pancreatic islets, and administration of peptide-loaded gels does not accelerate diabetes. This work demonstrates that a noninflammatory biomaterial system can generate antigen-specific Tregs in vivo, which may enable the development of new therapies for the treatment of transplant rejection or autoimmune diseases.
Collapse
Affiliation(s)
- Catia S Verbeke
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Susana Gordo
- Dana Farber Cancer Institute, Boston, MA, 02215, USA
| | | | - Sarah A Lewin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Rajiv M Desai
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | | | | | - David J Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
18
|
Graves CL, Li J, LaPato M, Shapiro MR, Glover SC, Wallet MA, Wallet SM. Intestinal Epithelial Cell Regulation of Adaptive Immune Dysfunction in Human Type 1 Diabetes. Front Immunol 2017; 7:679. [PMID: 28119693 PMCID: PMC5222791 DOI: 10.3389/fimmu.2016.00679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/21/2016] [Indexed: 01/29/2023] Open
Abstract
Environmental factors contribute to the initiation, progression, and maintenance of type 1 diabetes (T1D), although a single environmental trigger for disease has not been identified. Studies have documented the contribution of immunity within the gastrointestinal tract (GI) to the expression of autoimmunity at distal sites. Intestinal epithelial cells (IECs) regulate local and systemic immunologic homeostasis through physical and biochemical interactions with innate and adaptive immune populations. We hypothesize that a loss in the tolerance-inducing nature of the GI tract occurs within T1D and is due to altered IECs' innate immune function. As a first step in addressing this hypothesis, we contrasted the global immune microenvironment within the GI tract of individuals with T1D as well as evaluated the IEC-specific effects on adaptive immune cell phenotypes. The soluble and cellular immune microenvironment within the duodenum, the soluble mediator profile of primary IECs derived from the same duodenal tissues, and the effect of the primary IECs' soluble mediator profile on T-cell expansion and polarization were evaluated. Higher levels of IL-17C and beta-defensin 2 (BD-2) mRNA in the T1D-duodenum were observed. Higher frequencies of type 1 innate lymphoid cells (ILC1) and CD8+CXCR3+ T-cells (Tc1) were also observed in T1D-duodenal tissues, concomitant with lower frequencies of type 3 ILC (ILC3) and CD8+CCR6+ T-cells (Tc17). Higher levels of proinflammatory mediators (IL-17C and BD-2) in the absence of similar changes in mediators associated with homeostasis (interleukin 10 and thymic stromal lymphopoietin) were also observed in T1D-derived primary IEC cultures. T1D-derived IEC culture supernatants induced more robust CD8+ T-cell proliferation along with enhanced polarization of Tc1 populations, at the expense of Tc17 polarization, as well as the expansion of CXCR3+CCR6+/- Tregs, indicative of a Th1-like and less regulatory phenotype. These data demonstrate a proinflammatory microenvironment of the T1D-duodenum, whereby IECs have the potential to contribute to the expansion and polarization of innate and adaptive immune cells. Although these data do not discern whether these observations are not simply a consequence of T1D, the data indicate that the T1D-GI tract has the capacity to foster a permissive environment under which autoreactive T-cells could be expanded and polarized.
Collapse
Affiliation(s)
- Christina L. Graves
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Jian Li
- Department of Gastroenterology, Hepatology, and Nutrition, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Melissa LaPato
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Melanie R. Shapiro
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| | - Sarah C. Glover
- Department of Gastroenterology, Hepatology, and Nutrition, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Mark A. Wallet
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida Health Science Center, Gainesville, FL, USA
| | - Shannon M. Wallet
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, USA
| |
Collapse
|
19
|
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease that leads to destruction of pancreatic β cells, lifelong dependence on insulin, and increased morbidity and mortality from diabetes-related complications. Preservation of residual β cells at diagnosis is a major goal because higher levels of endogenous insulin secretion are associated with better short- and long-term outcomes. For the past 3 decades, a variety of immune interventions have been evaluated in the setting of new-onset T1D, including nonspecific immunosuppression, pathway-specific immune modulation, antigen-specific therapies, and cellular therapies. To date, no single intervention has produced durable remission off therapy in most treated patients, but the field has gained valuable insights into disease mechanisms and potential immunologic correlates of success. In particular, T-cell–directed therapies, including therapies that lead to partial depletion or modulation of effector T cells and preservation or augmentation of regulatory T cells, have shown the most success and will likely form the backbone of future approaches. The next phase will see evaluation of rational combinations, comprising one or more of the following: an effector T-depleting or -modulating drug, a cytokine-based tolerogenic (regulatory T-cells–promoting) agent, and an antigen-specific component. The long term goal is to reestablish immunologic tolerance to β cells, thereby preserving residual β cells early after diagnosis or enabling restoration of β-cell mass from autologous stem cells or induced neogenesis in patients with established T1D.
Collapse
|
20
|
Dastagir SR, Postigo-Fernandez J, Xu C, Stoeckle JH, Firdessa-Fite R, Creusot RJ. Efficient Presentation of Multiple Endogenous Epitopes to Both CD4 + and CD8 + Diabetogenic T Cells for Tolerance. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 4:27-38. [PMID: 28344989 PMCID: PMC5363322 DOI: 10.1016/j.omtm.2016.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/13/2016] [Indexed: 10/28/2022]
Abstract
Antigen-specific immunotherapy of type 1 diabetes, typically via delivery of a single native β cell antigen, has had little clinical benefit to date. With increasing evidence that diabetogenic T cells react against multiple β cell antigens, including previously unappreciated neo-antigens that can be emulated by mimotopes, a shift from protein- to epitope-based therapy is warranted. To this end, we aimed to achieve efficient co-presentation of multiple major epitopes targeting both CD4+ and CD8+ diabetogenic T cells. We have compared native epitopes versus mimotopes as well as various targeting signals in an effort to optimize recognition by both types of T cells in vitro. Optimal engagement of all T cells was achieved with segregation of CD8 and CD4 epitopes, the latter containing mimotopes and driven by endosome-targeting signals, after delivery into either dendritic or stromal cells. The CD4+ T cell responses elicited by the endogenously delivered epitopes were comparable with high concentrations of soluble peptide and included functional regulatory T cells. This work has important implications for the improvement of antigen-specific therapies using an epitope-based approach to restore tolerance in type 1 diabetes and in a variety of other diseases requiring concomitant targeting of CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Shamael R Dastagir
- Columbia Center for Translational Immunology and Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Jorge Postigo-Fernandez
- Columbia Center for Translational Immunology and Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Chunliang Xu
- Columbia Center for Translational Immunology and Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - James H Stoeckle
- Columbia Center for Translational Immunology and Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Rebuma Firdessa-Fite
- Columbia Center for Translational Immunology and Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Rémi J Creusot
- Columbia Center for Translational Immunology and Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
21
|
Prezioso G, Comegna L, Di Giulio C, Franchini S, Chiarelli F, Blasetti A. C1858T Polymorphism of Protein Tyrosine Phosphatase Non-receptor Type 22 (PTPN22): an eligible target for prevention of type 1 diabetes? Expert Rev Clin Immunol 2016; 13:189-196. [PMID: 27892782 DOI: 10.1080/1744666x.2017.1266257] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION In type 1 diabetes (T1D), several genetic factors are associated to β-cell autoimmunity onset and clinical progression. HLA-genes play a major role in susceptibility and initiation of β-cell autoimmunity, whereas non-HLA genes may influence the destruction rate. Areas covered: Our review focuses on the possible role of the PTPN22 C1858 T variant as a prognostic factor, given its influence on disease variability. Moreover, we present the potential role of C1858 T as a target for tertiary prevention trials and new therapeutic strategies, such as the LYP inhibitors. We used PubMed for literature research; key words were 'PTPN22', 'C1858 T polymorphism', 'lymphoid-specific tyrosine phosphatase' and 'type 1 diabetes'. We selected publications between 2000 and 2016. Expert commentary: Current data suggest that PTPN22 can be a promising target for therapeutic interventions and identification of at-risk subjects in autoimmune diseases such as T1D.
Collapse
Affiliation(s)
- Giovanni Prezioso
- a Department of Pediatrics , 'G. D'Annunzio' University , Chieti , Italy
| | - Laura Comegna
- a Department of Pediatrics , 'G. D'Annunzio' University , Chieti , Italy
| | - Concetta Di Giulio
- a Department of Pediatrics , 'G. D'Annunzio' University , Chieti , Italy
| | - Simone Franchini
- a Department of Pediatrics , 'G. D'Annunzio' University , Chieti , Italy
| | | | - Annalisa Blasetti
- a Department of Pediatrics , 'G. D'Annunzio' University , Chieti , Italy
| |
Collapse
|
22
|
Rahimi M, Vinciguerra M, Daghighi M, Özcan B, Akbarkhanzadeh V, Sheedfar F, Amini M, Mazza T, Pazienza V, Motazacker MM, Mahmoudi M, De Rooij FWM, Sijbrands E, Peppelenbosch MP, Rezaee F. Age-related obesity and type 2 diabetes dysregulate neuronal associated genes and proteins in humans. Oncotarget 2016; 6:29818-32. [PMID: 26337083 PMCID: PMC4745765 DOI: 10.18632/oncotarget.4904] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/07/2015] [Indexed: 12/29/2022] Open
Abstract
Despite numerous developed drugs based on glucose metabolism interventions for treatment of age-related diseases such as diabetes neuropathies (DNs), DNs are still increasing in patients with type 1 or type 2 diabetes (T1D, T2D). We aimed to identify novel candidates in adipose tissue (AT) and pancreas with T2D for targeting to develop new drugs for DNs therapy. AT-T2D displayed 15 (e.g. SYT4 up-regulated and VGF down-regulated) and pancreas-T2D showed 10 (e.g. BAG3 up-regulated, VAV3 and APOA1 down-regulated) highly differentially expressed genes with neuronal functions as compared to control tissues. ELISA was blindly performed to measure proteins of 5 most differentially expressed genes in 41 human subjects. SYT4 protein was upregulated, VAV3 and APOA1 were down-regulated, and BAG3 remained unchanged in 1- Obese and 2- Obese-T2D without insulin, VGF protein was higher in these two groups as well as in group 3- Obese-T2D receiving insulin than 4-lean subjects. Interaction networks analysis of these 5 genes showed several metabolic pathways (e.g. lipid metabolism and insulin signaling). Pancreas is a novel site for APOA1 synthesis. VGF is synthesized in AT and could be considered as good diagnostic, and even prognostic, marker for age-induced diseases obesity and T2D. This study provides new targets for rational drugs development for the therapy of age-related DNs.
Collapse
Affiliation(s)
- Mehran Rahimi
- Faculty of Medical Science, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manlio Vinciguerra
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, UK.,Gastroenterology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mojtaba Daghighi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Behiye Özcan
- Department of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Fareeba Sheedfar
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marzyeh Amini
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Valerio Pazienza
- Gastroenterology Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mahdi M Motazacker
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Morteza Mahmoudi
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States.,Department of Nanotechnology and Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Felix W M De Rooij
- Department of Cardiovascular Genetics, Metabolism, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eric Sijbrands
- Department of Cardiovascular Genetics, Metabolism, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, University of Rotterdam, Rotterdam, The Netherlands
| | - Farhad Rezaee
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, University of Rotterdam, Rotterdam, The Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Northrup L, Christopher MA, Sullivan BP, Berkland C. Combining antigen and immunomodulators: Emerging trends in antigen-specific immunotherapy for autoimmunity. Adv Drug Deliv Rev 2016; 98:86-98. [PMID: 26546466 DOI: 10.1016/j.addr.2015.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/05/2023]
Abstract
A majority of current therapies for autoimmune diseases are general immunosuppressants, which can compromise patient response to opportunistic infection and lead to adverse events. Using antigen-specific immunotherapy (ASIT) to selectively disarm autoimmune diseases, without suppressing the global immune response, would be a transformative therapy for patients. ASIT has been used historically in allergy hyposensitization therapy to induce tolerance to an allergen. Similar strategies to induce immune tolerance toward autoantigens responsible for autoimmune disease have been attempted but have yielded limited clinical success. Recent studies of ASIT for autoimmunity have explored combination therapy, combining the disease-causing autoantigen with an immunomodulatory compound. ASIT combination therapy may direct the immune response in an antigen-specific manner, potentially reversing the root cause of autoimmunity while limiting side effects. This review analyzes recent advances in ASIT applied to autoimmune diseases, emphasizing current combination therapies and future strategies.
Collapse
Affiliation(s)
- Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
24
|
Blasetti A, Di Giulio C, Tumini S, Provenzano M, Rapino D, Comegna L, Prezioso G, Chiuri R, Franchini S, Chiarelli F, Stuppia L. Role of the C1858T polymorphism of protein tyrosine phosphatase non-receptor type 22 (PTPN22) in children and adolescents with type 1 diabetes. THE PHARMACOGENOMICS JOURNAL 2016; 17:186-191. [DOI: 10.1038/tpj.2016.6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 11/24/2015] [Accepted: 01/20/2016] [Indexed: 01/18/2023]
|
25
|
Creusot RJ, Battaglia M, Roncarolo MG, Fathman CG. Concise Review: Cell-Based Therapies and Other Non-Traditional Approaches for Type 1 Diabetes. Stem Cells 2016; 34:809-19. [PMID: 26840009 PMCID: PMC5021120 DOI: 10.1002/stem.2290] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/07/2015] [Indexed: 01/01/2023]
Abstract
The evolution of Type 1 diabetes (T1D) therapy has been marked by consecutive shifts, from insulin replacement to immunosuppressive drugs and targeted biologics (following the understanding that T1D is an autoimmune disease), and to more disease‐specific or patient‐oriented approaches such as antigen‐specific and cell‐based therapies, with a goal to provide efficacy, safety, and long‐term protection. At the same time, another important paradigm shift from treatment of new onset T1D patients to prevention in high‐risk individuals has taken place, based on the hypothesis that therapeutic approaches deemed sufficiently safe may show better efficacy if applied early enough to maintain endogenous β cell function, a concept supported by many preclinical studies. This new strategy has been made possible by capitalizing on a variety of biomarkers that can more reliably estimate the risk and rate of progression of the disease. More advanced (“omic”‐based) biomarkers that also shed light on the underlying contributors of disease for each individual will be helpful to guide the choice of the most appropriate therapies, or combinations thereof. In this review, we present current efforts to stratify patients according to biomarkers and current alternatives to conventional drug‐based therapies for T1D, with a special emphasis on cell‐based therapies, their status in the clinic and potential for treatment and/or prevention. Stem Cells2016;34:809–819
Collapse
Affiliation(s)
- Remi J Creusot
- Department of Medicine, Columbia Center for Translational Immunology and Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, USA
| | - Manuela Battaglia
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria-Grazia Roncarolo
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine Stanford, CA, USA
| | - C Garrison Fathman
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine Stanford, CA, USA
| |
Collapse
|
26
|
Yu C, Burns JC, Robinson WH, Utz PJ, Ho PP, Steinman L, Frey AB. Identification of Candidate Tolerogenic CD8(+) T Cell Epitopes for Therapy of Type 1 Diabetes in the NOD Mouse Model. J Diabetes Res 2016; 2016:9083103. [PMID: 27069933 PMCID: PMC4812430 DOI: 10.1155/2016/9083103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease in which insulin-producing pancreatic islet β cells are the target of self-reactive B and T cells. T cells reactive with epitopes derived from insulin and/or IGRP are critical for the initiation and maintenance of disease, but T cells reactive with other islet antigens likely have an essential role in disease progression. We sought to identify candidate CD8(+) T cell epitopes that are pathogenic in type 1 diabetes. Proteins that elicit autoantibodies in human type 1 diabetes were analyzed by predictive algorithms for candidate epitopes. Using several different tolerizing regimes using synthetic peptides, two new predicted tolerogenic CD8(+) T cell epitopes were identified in the murine homolog of the major human islet autoantigen zinc transporter ZnT8 (aa 158-166 and 282-290) and one in a non-β cell protein, dopamine β-hydroxylase (aa 233-241). Tolerizing vaccination of NOD mice with a cDNA plasmid expressing full-length proinsulin prevented diabetes, whereas plasmids encoding ZnT8 and DβH did not. However, tolerizing vaccination of NOD mice with the proinsulin plasmid in combination with plasmids expressing ZnT8 and DβH decreased insulitis and enhanced prevention of disease compared to vaccination with the plasmid encoding proinsulin alone.
Collapse
MESH Headings
- Animals
- Autoantibodies/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cation Transport Proteins/genetics
- Cation Transport Proteins/immunology
- Cells, Cultured
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Dopamine beta-Hydroxylase/genetics
- Dopamine beta-Hydroxylase/immunology
- Epitopes, T-Lymphocyte
- Female
- Genetic Therapy/methods
- Humans
- Immune Tolerance
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/pathology
- Lymphocyte Activation
- Mice, Inbred NOD
- Proinsulin/genetics
- Proinsulin/immunology
- Time Factors
- Vaccination
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Zinc Transporter 8
Collapse
Affiliation(s)
- Cailin Yu
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Jeremy C. Burns
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Geriatric Research Education and Clinical Center, Veterans Affairs, Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Paul J. Utz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peggy P. Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan B. Frey
- Department of Cell Biology, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA
- *Alan B. Frey:
| |
Collapse
|
27
|
Affiliation(s)
- Dirk Homann
- Diabetes, Obesity and Metabolism Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
28
|
Fousteri G, Jofra T, Di Fonte R, Battaglia M. Combination of an Antigen-Specific Therapy and an Immunomodulatory Treatment to Simultaneous Block Recurrent Autoimmunity and Alloreactivity in Non-Obese Diabetic Mice. PLoS One 2015; 10:e0127631. [PMID: 26080071 PMCID: PMC4469694 DOI: 10.1371/journal.pone.0127631] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/16/2015] [Indexed: 12/12/2022] Open
Abstract
Restoration of endogenous insulin production by islet transplantation is considered a curative option for patients with type 1 diabetes. However, recurrent autoimmunity and alloreactivity cause graft rejection hindering successful transplantation. Here we tested whether transplant tolerance to allogeneic islets could be achieved in non-obese diabetic (NOD) mice by simultaneously tackling autoimmunity via antigen-specific immunization, and alloreactivity via granulocyte colony stimulating factor (G-CSF) and rapamycin (RAPA) treatment. Immunization with insB9-23 peptide alone or in combination with two islet peptides (IGRP206-214 and GAD524-543) in incomplete Freund’s adjuvant (IFA) were tested for promoting syngeneic pancreatic islet engraftment in spontaneously diabetic NOD mice. Treatment with G-CSF/RAPA alone or in combination with insB9-23/IFA was examined for promoting allogeneic islet engraftment in the same mouse model. InsB9-23/IFA immunization significantly prolonged syngeneic pancreatic islet survival in NOD mice by a mechanism that necessitated the presence of CD4+CD25+ T regulatory (Treg) cells, while combination of three islet epitopes was less efficacious in controlling recurrent autoimmunity. G-CSF/RAPA treatment was unable to reverse T1D or control recurrent autoimmunity but significantly prolonged islet allograft survival in NOD mice. Blockade of interleukin-10 (IL-10) during G-CSF/RAPA treatment resulted in allograft rejection suggesting that IL-10-producing cells were fundamental to achieve transplant tolerance. G-CSF/RAPA treatment combined with insB9-23/IFA did not further increase the survival of allogeneic islets. Thus, insB9-23/IFA immunization controls recurrent autoimmunity and G-CSF/RAPA treatment limits alloreactivity, however their combination does not further promote allogeneic pancreatic islet engraftment in NOD mice.
Collapse
Affiliation(s)
- Georgia Fousteri
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
- * E-mail: (GF); (MB)
| | - Tatiana Jofra
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Roberta Di Fonte
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Manuela Battaglia
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
- * E-mail: (GF); (MB)
| |
Collapse
|
29
|
Sarikonda G, Sachithanantham S, Miller JF, Pagni PP, Coppieters KT, von Herrath M. The Hsp60 peptide p277 enhances anti-CD3 mediated diabetes remission in non-obese diabetic mice. J Autoimmun 2015; 59:61-6. [PMID: 25772283 DOI: 10.1016/j.jaut.2015.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/07/2023]
Abstract
Type 1 diabetes (T1D) is characterized by the immune-mediated destruction of pancreatic beta cells leading to inadequate glycemic control. Trials with immunomodulatory monotherapies have shown that the disease course can in principle be altered. The observed preservation of endogenous insulin secretion however is typically transient and chronic treatment is often associated with significant side effects. Here we combined anti-CD3 with the Hsp60 peptide p277, two drugs that have been evaluated in Phase 3 trials, to test for enhanced efficacy. Female NOD mice with recent onset diabetes were given 5 μg anti-CD3 i.v., on three consecutive days in combination with 100 μg of p277 peptide in IFA s.c., once weekly for four weeks. Anti-CD3 alone restored normoglycemia in 44% of the mice while combination therapy with anti-CD3 and p277 induced stable remission in 83% of mice. The observed increase in protection occurred only in part through TLR2 signaling and was characterized by increased Treg numbers and decreased insulitis. These results have important implications for the design of combination therapies for the treatment of T1D.
Collapse
Affiliation(s)
- Ghanashyam Sarikonda
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | - Jacqueline F Miller
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Philippe P Pagni
- Type 1 Diabetes R&D Center, Novo Nordisk, Inc., Seattle, WA, USA
| | - Ken T Coppieters
- Type 1 Diabetes R&D Center, Novo Nordisk, Inc., Seattle, WA, USA
| | - Matthias von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Type 1 Diabetes R&D Center, Novo Nordisk, Inc., Seattle, WA, USA.
| |
Collapse
|
30
|
Skowera A, Ladell K, McLaren JE, Dolton G, Matthews KK, Gostick E, Kronenberg-Versteeg D, Eichmann M, Knight RR, Heck S, Powrie J, Bingley PJ, Dayan CM, Miles JJ, Sewell AK, Price DA, Peakman M. β-cell-specific CD8 T cell phenotype in type 1 diabetes reflects chronic autoantigen exposure. Diabetes 2015; 64:916-925. [PMID: 25249579 PMCID: PMC4557541 DOI: 10.2337/db14-0332] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Autoreactive CD8 T cells play a central role in the destruction of pancreatic islet β-cells that leads to type 1 diabetes, yet the key features of this immune-mediated process remain poorly defined. In this study, we combined high-definition polychromatic flow cytometry with ultrasensitive peptide-human leukocyte antigen class I tetramer staining to quantify and characterize β-cell-specific CD8 T cell populations in patients with recent-onset type 1 diabetes and healthy control subjects. Remarkably, we found that β-cell-specific CD8 T cell frequencies in peripheral blood were similar between subject groups. In contrast to healthy control subjects, however, patients with newly diagnosed type 1 diabetes displayed hallmarks of antigen-driven expansion uniquely within the β-cell-specific CD8 T cell compartment. Molecular analysis of selected β-cell-specific CD8 T cell populations further revealed highly skewed oligoclonal T cell receptor repertoires comprising exclusively private clonotypes. Collectively, these data identify novel and distinctive features of disease-relevant CD8 T cells that inform the immunopathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Ania Skowera
- Department of Immunobiology, King’s College London School of Medicine, London, UK
| | - Kristin Ladell
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - James E. McLaren
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Garry Dolton
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Katherine K. Matthews
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Emma Gostick
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | | - Martin Eichmann
- Department of Immunobiology, King’s College London School of Medicine, London, UK
| | - Robin R. Knight
- Department of Immunobiology, King’s College London School of Medicine, London, UK
| | - Susanne Heck
- National Institute for Health Research Biomedical Research Centre at Guy’s & St Thomas’ National Health Service Foundation Trust and King’s College London, London, UK
| | - Jake Powrie
- Department of Diabetes and Endocrinology, Guy’s & St Thomas’ National Health Service Foundation Trust, London, UK
| | | | - Colin M. Dayan
- Institute of Molecular & Experimental Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - John J. Miles
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Andrew K. Sewell
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - David A. Price
- Institute of Infection & Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Mark Peakman
- Department of Immunobiology, King’s College London School of Medicine, London, UK
| |
Collapse
|
31
|
Johannesson B, Sui L, Freytes DO, Creusot RJ, Egli D. Toward beta cell replacement for diabetes. EMBO J 2015; 34:841-55. [PMID: 25733347 DOI: 10.15252/embj.201490685] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/22/2015] [Indexed: 12/31/2022] Open
Abstract
The discovery of insulin more than 90 years ago introduced a life-saving treatment for patients with type 1 diabetes, and since then, significant progress has been made in clinical care for all forms of diabetes. However, no method of insulin delivery matches the ability of the human pancreas to reliably and automatically maintain glucose levels within a tight range. Transplantation of human islets or of an intact pancreas can in principle cure diabetes, but this approach is generally reserved for cases with simultaneous transplantation of a kidney, where immunosuppression is already a requirement. Recent advances in cell reprogramming and beta cell differentiation now allow the generation of personalized stem cells, providing an unlimited source of beta cells for research and for developing autologous cell therapies. In this review, we will discuss the utility of stem cell-derived beta cells to investigate the mechanisms of beta cell failure in diabetes, and the challenges to develop beta cell replacement therapies. These challenges include appropriate quality controls of the cells being used, the ability to generate beta cell grafts of stable cellular composition, and in the case of type 1 diabetes, protecting implanted cells from autoimmune destruction without compromising other aspects of the immune system or the functionality of the graft. Such novel treatments will need to match or exceed the relative safety and efficacy of available care for diabetes.
Collapse
Affiliation(s)
| | - Lina Sui
- Naomi Berrie Diabetes Center & Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Donald O Freytes
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Remi J Creusot
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA
| | - Dieter Egli
- The New York Stem Cell Foundation Research Institute, New York, NY, USA Naomi Berrie Diabetes Center & Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
32
|
Sachdeva N, Paul M, Badal D, Kumar R, Jacob N, Dayal D, Bhansali A, Arora SK, Bhadada SK. Preproinsulin specific CD8+ T cells in subjects with latent autoimmune diabetes show lower frequency and different pathophysiological characteristics than those with type 1 diabetes. Clin Immunol 2015; 157:78-90. [DOI: 10.1016/j.clim.2015.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/02/2014] [Accepted: 01/10/2015] [Indexed: 01/08/2023]
|
33
|
Smilek DE, Ehlers MR, Nepom GT. Restoring the balance: immunotherapeutic combinations for autoimmune disease. Dis Model Mech 2014; 7:503-13. [PMID: 24795433 PMCID: PMC4007402 DOI: 10.1242/dmm.015099] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autoimmunity occurs when T cells, B cells or both are inappropriately activated, resulting in damage to one or more organ systems. Normally, high-affinity self-reactive T and B cells are eliminated in the thymus and bone marrow through a process known as central immune tolerance. However, low-affinity self-reactive T and B cells escape central tolerance and enter the blood and tissues, where they are kept in check by complex and non-redundant peripheral tolerance mechanisms. Dysfunction or imbalance of the immune system can lead to autoimmunity, and thus elucidation of normal tolerance mechanisms has led to identification of therapeutic targets for treating autoimmune disease. In the past 15 years, a number of disease-modifying monoclonal antibodies and genetically engineered biologic agents targeting the immune system have been approved, notably for the treatment of rheumatoid arthritis, inflammatory bowel disease and psoriasis. Although these agents represent a major advance, effective therapy for other autoimmune conditions, such as type 1 diabetes, remain elusive and will likely require intervention aimed at multiple components of the immune system. To this end, approaches that manipulate cells ex vivo and harness their complex behaviors are being tested in preclinical and clinical settings. In addition, approved biologic agents are being examined in combination with one another and with cell-based therapies. Substantial development and regulatory hurdles must be overcome in order to successfully combine immunotherapeutic biologic agents. Nevertheless, such combinations might ultimately be necessary to control autoimmune disease manifestations and restore the tolerant state.
Collapse
Affiliation(s)
- Dawn E Smilek
- The Immune Tolerance Network, 185 Berry Street #3515, San Francisco, CA 94107, USA
| | | | | |
Collapse
|
34
|
Drescher KM, von Herrath M, Tracy S. Enteroviruses, hygiene and type 1 diabetes: toward a preventive vaccine. Rev Med Virol 2014; 25:19-32. [PMID: 25430610 DOI: 10.1002/rmv.1815] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/26/2014] [Accepted: 10/02/2014] [Indexed: 12/24/2022]
Abstract
Enteroviruses and humans have long co-existed. Although recognized in ancient times, poliomyelitis and type 1 diabetes (T1D) were exceptionally rare and not epidemic, due in large part to poor sanitation and personal hygiene which resulted in repeated exposure to fecal-oral transmitted viruses and other infectious agents and viruses and the generation of a broad protective immunity. As a function of a growing acceptance of the benefits of hygienic practices and microbiologically clean(er) water supplies, the likelihood of exposure to diverse infectious agents and viruses declined. The effort to vaccinate against poliomyelitis demonstrated that enteroviral diseases are preventable by vaccination and led to understanding how to successfully attenuate enteroviruses. Type 1 diabetes onset has been convincingly linked to infection by numerous enteroviruses including the group B coxsackieviruses (CVB), while studies of CVB infections in NOD mice have demonstrated not only a clear link between disease onset but an ability to reduce the incidence of T1D as well: CVB infections can suppress naturally occurring autoimmune T1D. We propose here that if we can harness and develop the capacity to use attenuated enteroviral strains to induce regulatory T cell populations in the host through vaccination, then a vaccine could be considered that should function to protect against both autoimmune as well as virus-triggered T1D. Such a vaccine would not only specifically protect from certain enterovirus types but more importantly, also reset the organism's regulatory rheostat making the further development of pathogenic autoimmunity less likely.
Collapse
Affiliation(s)
- Kristen M Drescher
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, USA
| | | | | |
Collapse
|
35
|
Coppieters KT, von Herrath MG. Metabolic syndrome - Removing roadblocks to therapy: Antigenic immunotherapies. Mol Metab 2014; 3:275-83. [PMID: 24749057 PMCID: PMC3986497 DOI: 10.1016/j.molmet.2013.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 01/08/2023] Open
Abstract
Up to 25 per cent of the world׳s adult population may have the metabolic syndrome, a condition closely associated with central obesity. The metabolic syndrome is a major risk factor for cardiovascular disease and type 2 diabetes and therefore represents an important worldwide health problem. In addition to metabolic abnormalities such as raised fasting plasma glucose, high cholesterol and high blood pressure, there is consensus that obese subjects develop a state of low-grade chronic immune activation. This sustained pro-inflammatory response in fat tissue is thought to worsen insulin resistance and dyslipidemia. Likewise, the immune system contributes to the detrimental cascade of events leading to plaque formation in atherosclerosis. It has long been assumed that the innate arm of the immune system was the only key player, but emerging evidence suggests that there is in fact a sizeable adaptive immune component to obesity and cardiovascular disease. From a therapeutic perspective, it could be envisioned that immune modulation drugs such as cytokine inhibitors, co-stimulation blockers or anti-T cell agents could offer benefit. It is questionable, however, whether chronic treatment with for instance biologicals will have a favorable risk/benefit profile in a silent condition such as the metabolic syndrome. An attractive alternative could be the development of antigen-specific T cell therapies, not unlike those currently in various phases of development for type 1 diabetes. In this article, we will give an overview of antigen-specific treatment modalities in type 1 diabetes, followed by a review of the evidence for T cell involvement in obesity and atherosclerosis.
Collapse
Affiliation(s)
| | - Matthias G. von Herrath
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA
- Type 1 Diabetes Center, The La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA, USA
- Corresponding author at: Type 1 Diabetes Center, The La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA, USA. Tel.: +1 858 752 6817; fax: +1 858 752 6993.
| |
Collapse
|
36
|
Morel PA. Dendritic cell subsets in type 1 diabetes: friend or foe? Front Immunol 2013; 4:415. [PMID: 24367363 PMCID: PMC3853773 DOI: 10.3389/fimmu.2013.00415] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/13/2013] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a T cell mediated autoimmune disease characterized by immune mediated destruction of the insulin-producing β cells in the islets of Langerhans. Dendritic cells (DC) have been implicated in the pathogenesis of T1D and are also used as immunotherapeutic agents. Plasmacytoid (p)DC have been shown to have both protective and pathogenic effects and a newly described merocytic DC population has been shown to break tolerance in the mouse model of T1D, the non-obese diabetic (NOD) mouse. We have used DC populations to prevent the onset of T1D in NOD mice and clinical trials of DC therapy in T1D diabetes have been initiated. In this review we will critically examine the recent published literature on the role of DC subsets in the induction and regulation of the autoimmune response in T1D.
Collapse
Affiliation(s)
- Penelope A Morel
- Department of Immunology, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|