1
|
Liao CC, Hsieh CC, Shia WC, Chou MY, Huang CC, Lin JH, Lee SH, Sung HH. Refined protocol for newly onset identification in non-obese diabetic mice: an animal-friendly, cost-effective, and efficient alternative. Lab Anim Res 2024; 40:16. [PMID: 38649958 PMCID: PMC11034171 DOI: 10.1186/s42826-024-00202-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Therapeutic interventions for diabetes are most effective when administered in the newly onset phase, yet determining the exact onset moment can be elusive in practice. Spontaneous autoimmune diabetes among NOD mice appears randomly between 12 and 32 weeks of age with an incidence range from 60 to 90%. Furthermore, the disease often progresses rapidly to severe diabetes within days, resulting in a very short window of newly onset phase, that poses significant challenge in early diagnosis. Conventionally, extensive blood glucose (BG) testing is typically required on large cohorts throughout several months to conduct prospective survey. We incorporated ultrasensitive urine glucose (UG) testing into an ordinary BG survey process, initially aiming to elucidate the lag period required for excessive glucose leaking from blood to urine during diabetes progression in the mouse model. RESULTS The observations unexpectedly revealed that small amounts of glucose detected in the urine often coincide with, sometimes even a couple days prior than elevated BG is diagnosed. Accordingly, we conducted the UG-based survey protocol in another cohort that was validated to accurately identified every individual near onset, who could then be confirmed by following few BG tests to fulfill the consecutive BG + criteria. This approach required fewer than 95 BG tests, compared to over 700 tests with traditional BG survey, to diagnose all the 37-38 diabetic mice out of total 60. The average BG level at diagnosis was slightly below 350 mg/dl, lower than the approximately 400 mg/dl observed with conventional BG monitoring. CONCLUSIONS We demonstrated a near perfect correlation between BG + and ultrasensitive UG + results in prospective survey with no lag period detected under twice weekly of testing frequency. This led to the refined protocol based on surveying with noninvasive UG testing, allowing for the early identification of newly onset diabetic mice with only a few BG tests required per mouse. This protocol significantly reduces the need for extensive blood sampling, lancet usage, labor, and animal distress, aligning with the 3Rs principle. It presents a convenient, accurate, and animal-friendly alternative for early diabetes diagnosis, facilitating research on diagnosis, pathogenesis, prevention, and treatment.
Collapse
Affiliation(s)
- Chia-Chi Liao
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chia-Chun Hsieh
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Chung Shia
- Molecular Medicine Laboratory, Department of Research, Changhua Christian Hospital, Changhua, Taiwan
| | - Min-Yuan Chou
- Biomedical Technology and Device Research Lab, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chuan-Chuan Huang
- Biomedical Technology and Device Research Lab, Industrial Technology Research Institute, Hsinchu, Taiwan
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jhih-Hong Lin
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, Taiwan
| | - Shu-Hsien Lee
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, Taiwan
| | - Hsiang-Hsuan Sung
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan.
| |
Collapse
|
2
|
Jing Z, Li Y, Ma Y, Zhang X, Liang X, Zhang X. Leverage biomaterials to modulate immunity for type 1 diabetes. Front Immunol 2022; 13:997287. [PMID: 36405706 PMCID: PMC9667795 DOI: 10.3389/fimmu.2022.997287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/20/2022] [Indexed: 09/08/2024] Open
Abstract
The pathogeny of type 1 diabetes (T1D) is mainly provoked by the β-cell loss due to the autoimmune attack. Critically, autoreactive T cells firsthand attack β-cell in islet, that results in the deficiency of insulin in bloodstream and ultimately leads to hyperglycemia. Hence, modulating immunity to conserve residual β-cell is a desirable way to treat new-onset T1D. However, systemic immunosuppression makes patients at risk of organ damage, infection, even cancers. Biomaterials can be leveraged to achieve targeted immunomodulation, which can reduce the toxic side effects of immunosuppressants. In this review, we discuss the recent advances in harness of biomaterials to immunomodulate immunity for T1D. We investigate nanotechnology in targeting delivery of immunosuppressant, biological macromolecule for β-cell specific autoreactive T cell regulation. We also explore the biomaterials for developing vaccines and facilitate immunosuppressive cells to restore immune tolerance in pancreas.
Collapse
Affiliation(s)
- Zhangyan Jing
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuan Li
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yumeng Ma
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaozhou Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Key Laboratory of Stem Cell and Regenerative Tissue Engineering, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Xin Liang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Key Laboratory of Stem Cell and Regenerative Tissue Engineering, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Xudong Zhang
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Kim S, Shukla RK, Yu H, Baek A, Cressman SG, Golconda S, Lee GE, Choi H, Reneau JC, Wang Z, Huang CA, Liyanage NPM, Kim S. CD3e-immunotoxin spares CD62L lo Tregs and reshapes organ-specific T-cell composition by preferentially depleting CD3e hi T cells. Front Immunol 2022; 13:1011190. [PMID: 36389741 PMCID: PMC9643874 DOI: 10.3389/fimmu.2022.1011190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/04/2022] [Indexed: 02/03/2023] Open
Abstract
CD3-epsilon(CD3e) immunotoxins (IT), a promising precision reagent for various clinical conditions requiring effective depletion of T cells, often shows limited treatment efficacy for largely unknown reasons. Tissue-resident T cells that persist in peripheral tissues have been shown to play pivotal roles in local and systemic immunity, as well as transplant rejection, autoimmunity and cancers. The impact of CD3e-IT treatment on these local cells, however, remains poorly understood. Here, using a new murine testing model, we demonstrate a substantial enrichment of tissue-resident Foxp3+ Tregs following CD3e-IT treatment. Differential surface expression of CD3e among T-cell subsets appears to be a main driver of Treg enrichment in CD3e-IT treatment. The surviving Tregs in CD3e-IT-treated mice were mostly the CD3edimCD62Llo effector phenotype, but the levels of this phenotype markedly varied among different lymphoid and nonlymphoid organs. We also found notable variations in surface CD3e levels among tissue-resident T cells of different organs, and these variations drive CD3e-IT to uniquely reshape T-cell compositions in local organs. The functions of organs and anatomic locations (lymph nodes) also affected the efficacy of CD3e-IT. The multi-organ pharmacodynamics of CD3e-IT and potential treatment resistance mechanisms identified in this study may generate new opportunities to further improve this promising treatment.
Collapse
Affiliation(s)
- Shihyoung Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Rajni Kant Shukla
- Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH, United States
| | - Hannah Yu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Alice Baek
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Sophie G. Cressman
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Sarah Golconda
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Ga-Eun Lee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Hyewon Choi
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - John C. Reneau
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Zhirui Wang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Christene A. Huang
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Namal P. M. Liyanage
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States,Department of Microbial Immunity and Infection, The Ohio State University, Columbus, OH, United States,Infectious Disease Institute, The Ohio State University, Columbus, OH, United States,*Correspondence: Namal P. M. Liyanage, ; Sanggu Kim,
| | - Sanggu Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States,Infectious Disease Institute, The Ohio State University, Columbus, OH, United States,*Correspondence: Namal P. M. Liyanage, ; Sanggu Kim,
| |
Collapse
|
4
|
Anderson RL, DiMeglio LA, Mander AP, Dayan CM, Linsley PS, Herold KC, Marinac M, Ahmed ST. Innovative Designs and Logistical Considerations for Expedited Clinical Development of Combination Disease-Modifying Treatments for Type 1 Diabetes. Diabetes Care 2022; 45:2189-2201. [PMID: 36150059 PMCID: PMC9911317 DOI: 10.2337/dc22-0308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
It has been 100 years since the life-saving discovery of insulin, yet daily management of type 1 diabetes (T1D) remains challenging. Even with closed-loop systems, the prevailing need for persons with T1D to attempt to match the kinetics of insulin activity with the kinetics of carbohydrate metabolism, alongside dynamic life factors affecting insulin requirements, results in the need for frequent interventions to adjust insulin dosages or consume carbohydrates to correct mismatches. Moreover, peripheral insulin dosing leaves the liver underinsulinized and hyperglucagonemic and peripheral tissues overinsulinized relative to their normal physiologic roles in glucose homeostasis. Disease-modifying therapies (DMT) to preserve and/or restore functional β-cell mass with controlled or corrected autoimmunity would simplify exogenous insulin need, thereby reducing disease mortality, morbidity, and management burdens. However, identifying effective DMTs for T1D has proven complex. There is some consensus that combination DMTs are needed for more meaningful clinical benefit. Other complexities are addressable with more innovative trial designs and logistics. While no DMT has yet been approved for marketing, existing regulatory guidance provides opportunities to further "de-risk" development. The T1D development ecosystem can accelerate progress by using more innovative ways for testing DMTs for T1D. This perspective outlines suggestions for accelerating evaluation of candidate T1D DMTs, including combination therapies, by use of innovative trial designs, enhanced logistical coordination of efforts, and regulatory guidance for expedited development, combination therapies, and adaptive designs.
Collapse
Affiliation(s)
| | - Linda A. DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Adrian P. Mander
- Centre for Trials Research, Cardiff University School of Medicine, Cardiff, U.K
| | - Colin M. Dayan
- Centre for Endocrine and Diabetes Science, Cardiff University School of Medicine, Cardiff, U.K
| | - Peter S. Linsley
- Systems Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Kevan C. Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT
| | | | - Simi T. Ahmed
- New York Stem Cell Foundation Research Institute, New York, NY
| |
Collapse
|
5
|
Karacay C, Prietl B, Harer C, Ehall B, Haudum CW, Bounab K, Franz J, Eisenberg T, Madeo F, Kolb D, Hingerl K, Hausl M, Magnes C, Mautner SI, Kotzbeck P, Pieber TR. The effect of spermidine on autoimmunity and beta cell function in NOD mice. Sci Rep 2022; 12:4502. [PMID: 35296698 PMCID: PMC8927410 DOI: 10.1038/s41598-022-08168-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 02/28/2022] [Indexed: 12/21/2022] Open
Abstract
Spermidine is a natural polyamine which was shown to prolong lifespan of organisms and to improve cardiac and cognitive function. Spermidine was also reported to reduce inflammation and modulate T-cells. Autophagy is one of the mechanisms that spermidine exerts its effect. Autophagy is vital for β-cell homeostasis and autophagy deficiency was reported to lead to exacerbated diabetes in mice. The effect of spermidine in type 1 diabetes pathogenesis remains to be elucidated. Therefore, we examined the effect of spermidine treatment in non-obese diabetic (NOD) mice, a mouse model for type 1 diabetes. NOD mice were given untreated or spermidine-treated water ad libitum from 4 weeks of age until diabetes onset or 35 weeks of age. We found that treatment with 10 mM spermidine led to higher diabetes incidence in NOD mice despite unchanged pancreatic insulitis. Spermidine modulated tissue polyamine levels and elevated signs of autophagy in pancreas. Spermidine led to increased proportion of pro-inflammatory T-cells in pancreatic lymph nodes (pLN) in diabetic mice. Spermidine elevated the proportion of regulatory T-cells in early onset mice, whereas it reduced the proportion of regulatory T-cells in late onset mice. In summary spermidine treatment led to higher diabetes incidence and elevated proportion of T-cells in pLN.
Collapse
Affiliation(s)
- Ceren Karacay
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,BioTechMed Graz, Graz, Austria
| | - Barbara Prietl
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,CBmed GmbH- Center for Biomarker Research in Medicine, Graz, Austria
| | - Clemens Harer
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,BioTechMed Graz, Graz, Austria
| | - Barbara Ehall
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,BioTechMed Graz, Graz, Austria
| | - Christoph W Haudum
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,CBmed GmbH- Center for Biomarker Research in Medicine, Graz, Austria
| | - Kaddour Bounab
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Joakim Franz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Tobias Eisenberg
- BioTechMed Graz, Graz, Austria.,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.,Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Frank Madeo
- BioTechMed Graz, Graz, Austria.,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.,Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria.,Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Kerstin Hingerl
- Core Facility Ultrastructure Analysis, Center for Medical Research (ZMF), Medical University of Graz, Graz, Austria.,Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Markus Hausl
- Joanneum Research Forschungsgesellschaft mbH HEALTH - Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Christoph Magnes
- Joanneum Research Forschungsgesellschaft mbH HEALTH - Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Selma I Mautner
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Joanneum Research Forschungsgesellschaft mbH HEALTH - Institute for Biomedicine and Health Sciences, Graz, Austria
| | - Petra Kotzbeck
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,BioTechMed Graz, Graz, Austria.,Division of Plastic, Aesthetic and Reconstructive Surgery, Medical University of Graz, Graz, Austria.,Joanneum Research Forschungsgesellschaft mbH COREMED - Cooperative Centre for Regenerative Medicine, Graz, Austria
| | - Thomas R Pieber
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria. .,BioTechMed Graz, Graz, Austria. .,CBmed GmbH- Center for Biomarker Research in Medicine, Graz, Austria. .,Joanneum Research Forschungsgesellschaft mbH HEALTH - Institute for Biomedicine and Health Sciences, Graz, Austria.
| |
Collapse
|
6
|
Circulating miRNAs can serve as potential diagnostic biomarkers in chronic myelogenous leukemia patients. Leuk Res Rep 2021; 16:100257. [PMID: 34401317 PMCID: PMC8355908 DOI: 10.1016/j.lrr.2021.100257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 11/21/2022] Open
Abstract
Introduction Chronic Myelogenous Leukemia (CML) is a myeloproliferative disorder described as a malignant blood disorder by accounts for 15–20% of all adult leukemia. MicroRNAs (miRNAs) play an important role in post-transcriptional regulation of gene expressions. Expression level of tumor suppressor-miRNAs, described as miRNAs that target the oncogens, can contribute to diagnosis and prognosis of some malignant disorders including CML. We theorized that according to the excessive proliferation and alteration in miRNA expressions, there could be a change in the expression of miRNAs in plasma carried by exosomes. Methods We consequently decided to detect the differences between normal and aberrant miRNA expression in human plasma sample to find out the possibility of diagnosis by these alterations. The expression of candidate miRNAs were compared using RNA extracted from the plasma of 50 patients, as well as 30 healthy individuals. We analysed the plasma miR-16-1, miR-20, miR-106, miR-126, miR-155, miR-222, and miR-451 expression levels in CML patients by individual real-time quantitative RT-PCR. Results All selected miRNAs were found to be upregulated in newly diagnosed CML patients compared to the control, while upregulation of only three (miR-20, 106 and 222) were significant (17.4, 19 and 74.95 fold change, respectively; p<0.0001). In conclusion microRNAs have a potential use in treatment of CML, as they can target the genes involved in cell cycle, MAPK, growth inhibition, TGF beta, and p53 signaling pathways. Therefore, these miRNA signatures provide the basis for their utilization as biomarkers in CML.
Collapse
|
7
|
Datta SK. Harnessing Tolerogenic Histone Peptide Epitopes From Nucleosomes for Selective Down-Regulation of Pathogenic Autoimmune Response in Lupus (Past, Present, and Future). Front Immunol 2021; 12:629807. [PMID: 33936042 PMCID: PMC8080879 DOI: 10.3389/fimmu.2021.629807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Autoantigen-directed tolerance can be induced by certain nucleosomal histone peptide epitope/s in nanomolar dosage leading to sustained remission of disease in mice with spontaneous SLE. By contrast, lupus is accelerated by administration of intact (whole) histones, or whole nucleosomes in microparticles from apoptotic cells, or by post-translationally acetylated histone-peptides. Low-dose therapy with the histone-peptide epitopes simultaneously induces TGFβ and inhibits IL-6 production by DC in vivo, especially pDC, which then induce CD4+CD25+ Treg and CD8+ Treg cells that suppress pathogenic autoimmune response. Both types of induced Treg cells are FoxP3+ and act by producing TGFβ at close cell-to-cell range. No anaphylactic adverse reactions, or generalized immunosuppression have been detected in mice injected with the peptides, because the epitopes are derived from evolutionarily conserved histones in the chromatin; and the peptides are expressed in the thymus during ontogeny, and their native sequences have not been altered. The peptide-induced Treg cells can block severe lupus on adoptive transfer reducing inflammatory cell reaction and infiltration in the kidney. In Humans, similar potent Treg cells are generated by the histone peptide epitopes in vitro in lupus patients’ PBMC, inhibiting anti-dsDNA autoantibody and interferon production. Furthermore, the same types of Treg cells are generated in lupus patients who are in very long-term remission (2-8 years) after undergoing autologous hematopoietic stem cell transplantation. These Treg cells are not found in lupus patients treated conventionally into clinical remission (SLEDAI of 0); and consequently they still harbor pathogenic autoimmune cells, causing subclinical damage. Although antigen-specific therapy with pinpoint accuracy is suitable for straight-forward organ-specific autoimmune diseases, Systemic Lupus is much more complex. The histone peptide epitopes have unique tolerogenic properties for inhibiting Innate immune cells (DC), T cells and B cell populations that are both antigen-specifically and cross-reactively involved in the pathogenic autoimmune response in lupus. The histone peptide tolerance is a natural and non-toxic therapy suitable for treating early lupus, and also maintaining lupus patients after toxic drug therapy. The experimental steps, challenges and possible solutions for successful therapy with these peptide epitopes are discussed in this highly focused review on Systemic Lupus.
Collapse
Affiliation(s)
- Syamal K Datta
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
8
|
Graßhoff H, Comdühr S, Monne LR, Müller A, Lamprecht P, Riemekasten G, Humrich JY. Low-Dose IL-2 Therapy in Autoimmune and Rheumatic Diseases. Front Immunol 2021; 12:648408. [PMID: 33868284 PMCID: PMC8047324 DOI: 10.3389/fimmu.2021.648408] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Treg) are crucial for the maintenance of peripheral tolerance and for the control of ongoing inflammation and autoimmunity. The cytokine interleukin-2 (IL-2) is essentially required for the growth and survival of Treg in the peripheral lymphatic tissues and thus plays a vital role in the biology of Treg. Most autoimmune and rheumatic diseases exhibit disturbances in Treg biology either at a numerical or functional level resulting in an imbalance between protective and pathogenic immune cells. In addition, in some autoimmune diseases, a relative deficiency of IL-2 develops during disease pathogenesis leading to a disturbance of Treg homeostasis, which further amplifies the vicious cycle of tolerance breach and chronic inflammation. Low-dose IL-2 therapy aims either to compensate for this IL-2 deficiency to restore a physiological state or to strengthen the Treg population in order to be more effective in counter-regulating inflammation while avoiding global immunosuppression. Here we highlight key findings and summarize recent advances in the clinical translation of low-dose IL-2 therapy for the treatment of autoimmune and rheumatic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jens Y. Humrich
- Department of Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein Lübeck, Lübeck, Germany
| |
Collapse
|
9
|
Russo F, Citro A, Squeri G, Sanvito F, Monti P, Gregori S, Roncarolo MG, Annoni A. InsB9-23 Gene Transfer to Hepatocyte-Based Combined Therapy Abrogates Recurrence of Type 1 Diabetes After Islet Transplantation. Diabetes 2021; 70:171-181. [PMID: 33122392 DOI: 10.2337/db19-1249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 10/22/2020] [Indexed: 11/13/2022]
Abstract
The induction of antigen (Ag)-specific tolerance represents a therapeutic option for autoimmune diabetes. We demonstrated that administration of a lentiviral vector enabling expression of insulin B chain 9-23 (InsB9-23) (LV.InsB) in hepatocytes arrests β-cell destruction in prediabetic NOD mice by generating InsB9-23-specific FoxP3+ T regulatory cells (Tregs). LV.InsB in combination with a suboptimal dose of anti-CD3 monoclonal antibody (combined therapy [CT], 1 × 5 μg [CT5]) reverts diabetes and prevents recurrence of autoimmunity after islet transplantation in ∼50% of NOD mice. We investigated whether CT optimization could lead to abrogation of recurrence of autoimmunity. Therefore, alloislets were transplanted after optimized CT tolerogenic conditioning (1 × 25 μg [CT25]). Diabetic NOD mice conditioned with CT25 when glycemia was <500 mg/dL remained normoglycemic for 100 days after alloislet transplantation and displayed reduced insulitis, but independently from the graft. Accordingly, cured mice showed T-cell unresponsiveness to InsB9-23 stimulation and increased Treg frequency in islet infiltration and pancreatic lymph nodes. Additional studies revealed a complex mechanism of Ag-specific immune regulation driven by CT25, in which both Tregs and PDL1 costimulation cooperate to control diabetogenic cells, while transplanted islets play a crucial role, although transient, recruiting diabetogenic cells. Therefore, CT25 before alloislet transplantation represents an Ag-specific immunotherapy to resolve autoimmune diabetes in the presence of residual endogenous β-cell mass.
Collapse
Affiliation(s)
- Fabio Russo
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Citro
- Diabetes Research Institute (DRI), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Giorgia Squeri
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sanvito
- Pathology Unit, Department of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Monti
- Diabetes Research Institute (DRI), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | | | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Peng Y, Wen D, Lin F, Mahato RI. Co-delivery of siAlox15 and sunitinib for reversing the new-onset of type 1 diabetes in non-obese diabetic mice. J Control Release 2018; 292:1-12. [DOI: 10.1016/j.jconrel.2018.10.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/16/2018] [Accepted: 10/25/2018] [Indexed: 01/12/2023]
|
11
|
Kuhn C, Weiner HL. Therapeutic anti-CD3 monoclonal antibodies: from bench to bedside. Immunotherapy 2016; 8:889-906. [DOI: 10.2217/imt-2016-0049] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The induction of tolerance is a major goal of immunotherapy. Investigations over the last 20 years have shown that anti-CD3 monoclonal antibodies (mAbs) effectively treat autoimmune disease in animal models and have also shown promise in clinical trials. Tolerance induction by anti-CD3 mAbs is related to the induction of Tregs that control pathogenic autoimmune responses. Here, we review preclinical and clinical studies in which intravenous or mucosal administration of anti-CD3 mAbs has been employed and provide an outlook on future developments to enhance the efficacy of this promising therapeutic approach.
Collapse
Affiliation(s)
- Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Gill RG, Pagni PP, Kupfer T, Wasserfall CH, Deng S, Posgai A, Manenkova Y, Bel Hani A, Straub L, Bernstein P, Atkinson MA, Herold KC, von Herrath M, Staeva T, Ehlers MR, Nepom GT. A Preclinical Consortium Approach for Assessing the Efficacy of Combined Anti-CD3 Plus IL-1 Blockade in Reversing New-Onset Autoimmune Diabetes in NOD Mice. Diabetes 2016; 65:1310-6. [PMID: 26718498 PMCID: PMC5860426 DOI: 10.2337/db15-0492] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
There is an ongoing need to develop strategic combinations of therapeutic agents to prevent type 1 diabetes (T1D) or to preserve islet β-cell mass in new-onset disease. Although clinical trials using candidate therapeutics are commonly based on preclinical studies, concern is growing regarding the reproducibility as well as the potential clinical translation of reported results using animal models of human disorders. In response, the National Institutes of Health Immune Tolerance Network and JDRF established a multicenter consortium of academic institutions designed to assess the efficacy and intergroup reproducibility of clinically applicable immunotherapies for reversing new-onset disease in the NOD mouse model of T1D. Predicated on prior studies, this consortium conducted coordinated, prospective studies, using joint standard operating procedures, fixed criteria for study entry, and common reagents, to optimize combined anti-CD3 treatment plus interleukin-1 (IL-1) blockade to reverse new-onset disease in NOD mice. We did not find that IL-1 blockade with anti-IL-1β monoclonal antibody or IL-1trap provided additional benefit for reversing new-onset disease compared with anti-CD3 treatment alone. These results demonstrate the value of larger, multicenter preclinical studies for vetting and prioritizing therapeutics for future clinical use.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/therapeutic use
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Biomedical Research/methods
- CD3 Complex/chemistry
- CD3 Complex/metabolism
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Drug Administration Schedule
- Drug Therapy, Combination
- Female
- Immunoglobulin Fab Fragments/administration & dosage
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/therapeutic use
- Immunotherapy/methods
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors
- Interleukin-1 Receptor Accessory Protein/metabolism
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Mice, Inbred NOD
- Multicenter Studies as Topic
- Pilot Projects
- Receptors, Interleukin-1 Type I/antagonists & inhibitors
- Receptors, Interleukin-1 Type I/metabolism
- Recombinant Fusion Proteins/therapeutic use
- Reproducibility of Results
- Research Design
- Specific Pathogen-Free Organisms
- United States
Collapse
Affiliation(s)
- Ronald G Gill
- Departments of Surgery and Immunology, University of Colorado Denver, Aurora, CO
| | | | - Tinalyn Kupfer
- Departments of Surgery and Immunology, University of Colorado Denver, Aurora, CO
| | | | - Songyan Deng
- Yale University School of Medicine, New Haven, CT
| | - Amanda Posgai
- Department of Pathology, University of Florida, Gainesville, FL
| | | | - Amira Bel Hani
- La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Laura Straub
- Immune Tolerance Network, University of California, San Francisco, San Francisco, CA
| | | | - Mark A Atkinson
- Department of Pathology, University of Florida, Gainesville, FL
| | | | | | | | - Mario R Ehlers
- Immune Tolerance Network, University of California, San Francisco, San Francisco, CA
| | | |
Collapse
|
13
|
Askenasy N. Less Is More: The Detrimental Consequences of Immunosuppressive Therapy in the Treatment of Type-1 Diabetes. Int Rev Immunol 2015; 34:523-37. [DOI: 10.3109/08830185.2015.1010723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Valle A, Barbagiovanni G, Jofra T, Stabilini A, Perol L, Baeyens A, Anand S, Cagnard N, Gagliani N, Piaggio E, Battaglia M. Heterogeneous CD3 expression levels in differing T cell subsets correlate with the in vivo anti-CD3-mediated T cell modulation. THE JOURNAL OF IMMUNOLOGY 2015; 194:2117-27. [PMID: 25646305 DOI: 10.4049/jimmunol.1401551] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The tolerogenic anti-CD3ε monoclonal Abs (anti-CD3) are promising compounds for the treatment of type 1 diabetes. Anti-CD3 administration induces transient T cell depletion both in preclinical and in clinical studies. Notably, the said depletion mainly affects CD4(+) but not CD8(+) T cells. Moreover, type 1 diabetes reversal in preclinical models is accompanied by the selective expansion of CD4(+)Foxp3(+) T regulatory (Treg) cells, which are fundamental for the long-term maintenance of anti-CD3-mediated tolerance. The mechanisms that lead to this immune-shaping by affecting mainly CD4(+) T effector cells while sparing CD4(+)Foxp3(+) Treg cells have still to be fully elucidated. This study shows that CD3 expression levels differ from one T cell subset to another. CD4(+)Foxp3(-) T cells contain higher amounts of CD3 molecules than do CD4(+)Foxp3(+) and CD8(+) T cells in both mice and humans. The said differences correlate with the anti-CD3-mediated immune resetting that occurs in vivo after anti-CD3 administration in diabetic NOD mice. Additionally, transcriptome analysis demonstrates that CD4(+)Foxp3(+) Treg cells are significantly less responsive than are CD4(+)Foxp3(-) T cells to anti-CD3 treatment at a molecular level. Thus, heterogeneity in CD3 expression seems to confer to the various T cell subsets differing susceptibility to the in vivo tolerogenic anti-CD3-mediated modulation. These data shed new light on the molecular mechanism that underlies anti-CD3-mediated immune resetting and thus may open new opportunities to improve this promising treatment.
Collapse
Affiliation(s)
- Andrea Valle
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Giulia Barbagiovanni
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Tatiana Jofra
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angela Stabilini
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Louis Perol
- Université Pierre et Marie Curie Université Paris 06, 75005 Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7211, 75013 Paris, France; INSERM Unité 959, Immunology-Immunopathology-Immunotherapy, 75013 Paris, France; INSERM Unité 932, 75005 Paris, France; Institut Curie, Section Recherche, 75005 Paris, France
| | - Audrey Baeyens
- Université Pierre et Marie Curie Université Paris 06, 75005 Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7211, 75013 Paris, France; INSERM Unité 959, Immunology-Immunopathology-Immunotherapy, 75013 Paris, France
| | - Santosh Anand
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy; Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Nicolas Cagnard
- INSERM Unité 580, 75015 Paris, France; Bioinformatics Platform, Faculty of Medicine Paris Descartes, Necker Hospital for Sick Children, 75015 Paris, France
| | - Nicola Gagliani
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Eliane Piaggio
- Université Pierre et Marie Curie Université Paris 06, 75005 Paris, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7211, 75013 Paris, France; INSERM Unité 959, Immunology-Immunopathology-Immunotherapy, 75013 Paris, France; INSERM Unité 932, 75005 Paris, France; Institut Curie, Section Recherche, 75005 Paris, France; INSERM Center of Clinical Investigation (CBT507 IGR-Curie), 75005 Paris, France; and
| | - Manuela Battaglia
- San Raffaele Diabetes Research Institute, San Raffaele Scientific Institute, 20132 Milan, Italy; TrialNet Clinical Center, San Raffaele Hospital, 20132 Milan, Italy
| |
Collapse
|
15
|
Rosenzwajg M, Churlaud G, Hartemann A, Klatzmann D. Interleukin 2 in the pathogenesis and therapy of type 1 diabetes. Curr Diab Rep 2014; 14:553. [PMID: 25344788 DOI: 10.1007/s11892-014-0553-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) play a major role in controlling effector T cells (Teffs) responding to self-antigens, which cause autoimmune diseases. An improper Treg/Teff balance contributes to most autoimmune diseases, including type 1 diabetes (T1D). To restore a proper balance, blocking Teffs with immunosuppressants has been the only option, which was partly effective and too toxic. It now appears that expanding/activating Tregs with low-dose interleukin-2 (IL-2) could provide immunoregulation without immunosuppression. This is particularly interesting in T1D as Tregs from T1D patients are reported as dysfunctional and a relative deficiency in IL-2 production and/or IL-2-mediated signaling could contribute to this phenotype. A clinical study of low-dose IL-2 showed a very good safety profile and good Treg expansion/activation in T1D patients. This opens the way for efficacy trials to test low-dose IL-2 in prevention and treatment of T1D and to establish in which condition restoration of a proper Treg/Teff balance would be beneficial in the field of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Michelle Rosenzwajg
- Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department (i2B), AP-HP, Hôpital Pitié-Salpêtrière, 83 Boulevard de l'Hôpital, 75651, Paris, France,
| | | | | | | |
Collapse
|
16
|
Low-dose interleukin-2 therapy: a driver of an imbalance between immune tolerance and autoimmunity. Int J Mol Sci 2014; 15:18574-92. [PMID: 25322151 PMCID: PMC4227233 DOI: 10.3390/ijms151018574] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 09/22/2014] [Accepted: 10/08/2014] [Indexed: 01/21/2023] Open
Abstract
For many years, the role of interleukin-2 (IL-2) in autoimmune responses was established as a cytokine possessing strong pro-inflammatory activity. Studies of the past few years have changed our knowledge on IL-2 in autoimmune chronic inflammation, suggesting its protective role, when administered at low-doses. The disrupted balance between regulatory and effector T cells (Tregs and Teffs, respectively) is a characteristic of autoimmune diseases, and is dependent on homeostatic cytokines, including IL-2. Actually, inherent defects in the IL-2 signaling pathway and/or levels leading to Treg compromised function and numbers as well as Th17 expansion have been attributed to autoimmune disorders. In this review, we discuss the role of IL-2 in the pathogenesis of autoimmune diseases. In particular, we highlight the impact of the dysregulated IL-2 pathway on disruption of the Treg/Th17 balance, reversal of which appears to be a possible mechanism of the low-dose IL-2 treatment. The negative effects of IL-2 on the differentiation of follicular helper T cells (Tfh) and pathogenic Th17 cells, both of which contribute to autoimmunity, is emphasized in the paper as well. We also compare the current IL-2-based therapies of animal and human subjects with immune-mediated diseases aimed at boosting the Treg population, which is the most IL-2-dependent cell subset desirable for sufficient control of autoimmunity. New perspectives of therapeutic approaches focused on selective delivery of IL-2 to inflamed tissues, thus allowing local activity of IL-2 to be combined with its reduced systemic and pleiotropic toxicity, are also proposed in this paper.
Collapse
|
17
|
Abstract
The mammalian target of rapamycin (mTOR), a phosphoinositide-3-kinase-related protein kinase, acts as a rheostat capable of integrating a variety of environmental cues in the form of nutrients, energy, and growth factors to modulate organismal processes and homeostasis. Recently, there is a growing appreciation of mTOR in adaptive immunity for its crucial roles in keeping a proper balance between T cell quiescence and activation. Under steady-state circumstances, mTOR is subtly inhibited by multiple mechanisms to maintain normal T cell homeostasis. Antigen recognition by naïve T cells leads to mTOR activation, which subsequently promotes the differentiation of these cells into distinct effector T cell subsets. This review focuses primarily on the recent literature with respect to the regulatory effects and mechanisms of mTOR signaling in dictating T cell fate, and discusses the therapeutic implications of mTOR modulation in T-cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Yu Liu
- 1School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan, P. R. China
| | | | | |
Collapse
|
18
|
Hartemann A, Bensimon G, Payan CA, Jacqueminet S, Bourron O, Nicolas N, Fonfrede M, Rosenzwajg M, Bernard C, Klatzmann D. Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2013; 1:295-305. [PMID: 24622415 DOI: 10.1016/s2213-8587(13)70113-x] [Citation(s) in RCA: 321] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND An improper balance of regulatory/effector T (Treg/Teff) cells is central to the development of autoimmune diseases, including type 1 diabetes. We previously showed that low-dose interleukin 2 (IL2) induced Treg cell expansion and activation and clinical improvement in patients with hepatitis-C-virus-induced vasculitis. We aimed to establish which low doses of IL2 would be safe and induce Treg cells in patients with type 1 diabetes, considering that: (1) type 1 diabetes might be linked to alteration of the IL2/IL2R activation pathway; (2) activation of pathogenic Teff cells by IL2 could exacerbate disease; and (3) the safety of low-dose IL2 is not known in type 1 diabetes. METHODS This was a single-centre phase 1/2 study. 24 adult patients (18-55 years) with established insulin-dependent type 1 diabetes and at least one diabetes-related autoantibody were enrolled and randomly assigned (in a 1:1:1:1 ratio, by computer-generated randomisation list, with block size four) to placebo or IL2 at 0.33 MIU/day, 1 MIU/day, or 3 MIU/day for a 5-day course and were followed up for 60 days. All investigators and participants were masked to assignment. The primary outcome was change in Treg cells, measured by flow cytometry, and expressed as a percentage of CD4+ T cells, from day 1 to day 60. This trial is registered with ClinicalTrials.gov, number NCT01353833. FINDINGS Six patients were assigned to each group between June 1, 2011, and Feb 3, 2012. IL2 was well tolerated at all doses, with no serious adverse events. However, there was a dose-response association for non-serious adverse events during the treatment phase (days 1-6); one patient in the placebo group, three patients in the 0.33 MIU group, five patients in the 1 MIU group, and six patients in the 3 MIU group had non-serious adverse events. The most common adverse events in the treatment phase were injection-site reaction (no patients with placebo vs three patients with 0.33 MIU and 1 MIU vs two patients with 3 MIU) and influenza-like syndrome (no patients with placebo vs one patient with 0.33 MIU and 1 MIU vs four patients with 3 MIU). After the treatment phase, adverse events did not differ between groups. IL2 did not induce deleterious changes in glucose-metabolism variables. IL2 induced a dose-dependent increase in the proportion of Treg cells, significant at all doses compared with placebo (placebo mean increase 0.5% [SD 0.4]; 0.33 MIU 2.8% [1.2], p=0.0039; 1 MIU 3.9% [1.8], p=0.0039; 3 MIU 4.8% [1.9] p=0.0039). INTERPRETATION We have defined a well-tolerated and immunologically effective dose range of IL2 for application to type 1 diabetes therapy and prevention, which could be relevant to other disorders in which a Treg cell increase would be desirable.
Collapse
Affiliation(s)
- Agnès Hartemann
- Department of Diabetology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Gilbert Bensimon
- Department of Clinical Pharmacology, Hôpital Pitié-Salpêtrière, Paris, France; Pharmacologie, Paris, France
| | - Christine A Payan
- Department of Clinical Pharmacology, Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Olivier Bourron
- Department of Diabetology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Nathalie Nicolas
- Clinical Investigation Center Paris-Est-CIC-9304, Hôpital Pitié-Salpêtrière, Paris, France
| | - Michèle Fonfrede
- Department of Biochemistry, Hôpital Pitié-Salpêtrière, Paris, France
| | - Michelle Rosenzwajg
- Clinical Investigation Center for Biotherapies and Inflammation-Immunopathology-Biotherapy Department AP-HP, Hôpital Pitié-Salpêtrière, Paris, France; Immunology-Immunopathology-Immunotherapy UPMC Univ Paris 06, Paris, France; INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy, Paris, France; CNRS, UMR 7211, Immunology-Immunopathology-Immunotherapy, Paris, France
| | - Claude Bernard
- Clinical Investigation Center for Biotherapies and Inflammation-Immunopathology-Biotherapy Department AP-HP, Hôpital Pitié-Salpêtrière, Paris, France; Immunology-Immunopathology-Immunotherapy UPMC Univ Paris 06, Paris, France; INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy, Paris, France; CNRS, UMR 7211, Immunology-Immunopathology-Immunotherapy, Paris, France
| | - David Klatzmann
- Clinical Investigation Center for Biotherapies and Inflammation-Immunopathology-Biotherapy Department AP-HP, Hôpital Pitié-Salpêtrière, Paris, France; Immunology-Immunopathology-Immunotherapy UPMC Univ Paris 06, Paris, France; INSERM, UMR_S 959, Immunology-Immunopathology-Immunotherapy, Paris, France; CNRS, UMR 7211, Immunology-Immunopathology-Immunotherapy, Paris, France.
| |
Collapse
|
19
|
Diaz-de-Durana Y, Lau J, Knee D, Filippi C, Londei M, McNamara P, Nasoff M, DiDonato M, Glynne R, Herman AE. IL-2 immunotherapy reveals potential for innate beta cell regeneration in the non-obese diabetic mouse model of autoimmune diabetes. PLoS One 2013; 8:e78483. [PMID: 24205242 PMCID: PMC3813455 DOI: 10.1371/journal.pone.0078483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/12/2013] [Indexed: 01/09/2023] Open
Abstract
Type-1 diabetes (T1D) is an autoimmune disease targeting insulin-producing beta cells, resulting in dependence on exogenous insulin. To date, significant efforts have been invested to develop immune-modulatory therapies for T1D treatment. Previously, IL-2 immunotherapy was demonstrated to prevent and reverse T1D at onset in the non-obese diabetic (NOD) mouse model, revealing potential as a therapy in early disease stage in humans. In the NOD model, IL-2 deficiency contributes to a loss of regulatory T cell function. This deficiency can be augmented with IL-2 or antibody bound to IL-2 (Ab/IL-2) therapy, resulting in regulatory T cell expansion and potentiation. However, an understanding of the mechanism by which reconstituted regulatory T cell function allows for reversal of diabetes after onset is not clearly understood. Here, we describe that Ab/IL-2 immunotherapy treatment, given at the time of diabetes onset in NOD mice, not only correlated with reversal of diabetes and expansion of Treg cells, but also demonstrated the ability to significantly increase beta cell proliferation. Proliferation appeared specific to Ab/IL-2 immunotherapy, as anti-CD3 therapy did not have a similar effect. Furthermore, to assess the effect of Ab/IL-2 immunotherapy well after the development of diabetes, we tested the effect of delaying treatment for 4 weeks after diabetes onset, when beta cells were virtually absent. At this late stage after diabetes onset, Ab/IL-2 treatment was not sufficient to reverse hyperglycemia. However, it did promote survival in the absence of exogenous insulin. Proliferation of beta cells could not account for this improvement as few beta cells remained. Rather, abnormal insulin and glucagon dual-expressing cells were the only insulin-expressing cells observed in islets from mice with established disease. Thus, these data suggest that in diabetic NOD mice, beta cells have an innate capacity for regeneration both early and late in disease, which is revealed through IL-2 immunotherapy.
Collapse
Affiliation(s)
- Yaiza Diaz-de-Durana
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Janet Lau
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Deborah Knee
- Biotherapeutics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Christophe Filippi
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Marco Londei
- Translational Medicine, Novartis Institutes of Biomedical Research, San Diego, California, United States of America
| | - Peter McNamara
- Pharmacology Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Marc Nasoff
- Biotherapeutics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Michael DiDonato
- Structural Biology Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Richard Glynne
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
- * E-mail:
| | - Ann E. Herman
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| |
Collapse
|
20
|
Baeyens A, Pérol L, Fourcade G, Cagnard N, Carpentier W, Woytschak J, Boyman O, Hartemann A, Piaggio E. Limitations of IL-2 and rapamycin in immunotherapy of type 1 diabetes. Diabetes 2013; 62:3120-31. [PMID: 23670972 PMCID: PMC3749335 DOI: 10.2337/db13-0214] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Administration of low-dose interleukin-2 (IL-2) alone or combined with rapamycin (RAPA) prevents hyperglycemia in NOD mice. Also, low-dose IL-2 cures recent-onset type 1 diabetes (T1D) in NOD mice, partially by boosting pancreatic regulatory T cells (Treg cells). These approaches are currently being evaluated in humans. Our objective was to study the effect of higher IL-2 doses (250,000-500,000 IU daily) as well as low-dose IL-2 (25,000 IU daily) and RAPA (1 mg/kg daily) (RAPA/IL-2) combination. We show that, despite further boosting of Treg cells, high doses of IL-2 rapidly precipitated T1D in prediabetic female and male mice and increased myeloid cells in the pancreas. Also, we observed that RAPA counteracted IL-2 effects on Treg cells, failed to control IL-2-boosted NK cells, and broke IL-2-induced tolerance in a reversible way. Notably, the RAPA/IL-2 combination failure to cure T1D was associated with an unexpected deleterious effect on glucose homeostasis at multiple levels, including β-cell division, glucose tolerance, and liver glucose metabolism. Our data help to understand the therapeutic limitations of IL-2 alone or RAPA/IL-2 combination and could lead to the design of improved therapies for T1D.
Collapse
Affiliation(s)
- Audrey Baeyens
- Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7211, Paris, France
- Department of Immunology-Immunopathology-Immunotherapy, INSERM U959, Paris, France
| | - Louis Pérol
- Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7211, Paris, France
- Department of Immunology-Immunopathology-Immunotherapy, INSERM U959, Paris, France
| | - Gwladys Fourcade
- Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7211, Paris, France
- Department of Immunology-Immunopathology-Immunotherapy, INSERM U959, Paris, France
| | - Nicolas Cagnard
- INSERM U580, Paris, France
- Bioinformatics Platform, Faculty of Medicine Paris Descartes, Hôpital Necker-Enfants Malades, Paris, France
| | - Wassila Carpentier
- Université Pierre et Marie Curie, Paris, France
- Plateforme Post-Génomique P3S, Université Pierre et Marie Curie, Faculty of Medicine, Paris, France
| | - Janine Woytschak
- Laboratory of Applied Immunobiology, University of Zurich, Zurich, Switzerland
| | - Onur Boyman
- Laboratory of Applied Immunobiology, University of Zurich, Zurich, Switzerland
- Allergy Unit, Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Agnès Hartemann
- Department of Endocrinology, Nutrition and Diabetes, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière-Charles Foix Hospital, Paris, France
- Department of Medicine Faculty, Université Pierre et Marie Curie, Paris, France
| | - Eliane Piaggio
- Université Pierre et Marie Curie, Paris, France
- Centre National de la Recherche Scientifique, UMR 7211, Paris, France
- Department of Immunology-Immunopathology-Immunotherapy, INSERM U959, Paris, France
- Corresponding author: Eliane Piaggio,
| |
Collapse
|
21
|
Goto R, You S, Zaitsu M, Chatenoud L, Wood KJ. Delayed anti-CD3 therapy results in depletion of alloreactive T cells and the dominance of Foxp3+ CD4+ graft infiltrating cells. Am J Transplant 2013; 13:1655-64. [PMID: 23750800 PMCID: PMC3790953 DOI: 10.1111/ajt.12272] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 03/08/2013] [Accepted: 03/31/2013] [Indexed: 01/25/2023]
Abstract
The engineered Fc-nonbinding (crystallizable fragment-nonbinding) CD3 antibody has lower mitogenicity and a precise therapeutic window for disease remission in patients with type 1 diabetes. Before anti-CD3 can be considered for use in transplantation, the most effective timing of treatment relative to transplantation needs to be elucidated. In this study anti-CD3F(ab')2 fragments or saline were administered intravenously for 5 consecutive days (early: d1-3 or delayed: d3-7) to mice transplanted with a cardiac allograft (H2(b)-to-H2(k); d0). Survival of allografts was prolonged in mice treated with the early protocol (MST = 48 days), but most were rejected by d100. In contrast, in mice treated with the delayed protocol allografts continued to survive long term. The delayed protocol significantly inhibited donor alloreactivity at d30 as compared to the early protocol. A marked increase in Foxp3(+) T cells (50.3 ± 1.6%) infiltrating the allografts in mice treated with the delayed protocol was observed (p < 0.0001 vs. early (24.9 ± 2.1%)) at d10; a finding that was maintained in the accepted cardiac allografts at d100. We conclude that the timing of treatment with anti-CD3 therapy is critical for inducing long-term graft survival. Delaying administration effectively inhibits the alloreactivity and promotes the dominance of intragraft Foxp3(+) T cells allowing long-term graft acceptance.
Collapse
Affiliation(s)
- R Goto
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordJohn Radcliffe Hospital, Oxford, United Kingdom
| | - S You
- Universite Paris Descartes, Institut National de la Santé et de la Recherche Médicale Unit 1013Paris, France
| | - M Zaitsu
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordJohn Radcliffe Hospital, Oxford, United Kingdom
| | - L Chatenoud
- Universite Paris Descartes, Institut National de la Santé et de la Recherche Médicale Unit 1013Paris, France
| | - KJ Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordJohn Radcliffe Hospital, Oxford, United Kingdom,* Kathryn J. Wood,
| |
Collapse
|
22
|
Perl S, Perlman J, Weitzel RP, Phang O, Hsieh MM, Tisdale J. Addition of rapamycin to anti-CD3 antibody improves long-term glycaemia control in diabetic NOD mice. PLoS One 2013; 8:e67189. [PMID: 23826229 PMCID: PMC3691209 DOI: 10.1371/journal.pone.0067189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 05/17/2013] [Indexed: 01/12/2023] Open
Abstract
Aims/Hypothesis Non-Fc-binding Anti CD3 antibody has proven successful in reverting diabetes in the non-obese diabetes mouse model of type 1 diabetes and limited efficacy has been observed in human clinical trials. We hypothesized that addition of rapamycin, an mTOR inhibitor capable of inducing operational tolerance in allogeneic bone marrow transplantation, would result in improved diabetes reversal rates and overall glycemia. Methods Seventy hyperglycemic non-obese diabetic mice were randomized to either a single injection of anti CD3 alone or a single injection of anti CD3 followed by 14 days of intra-peritoneal rapamycin. Mice were monitored for hyperglycemia and metabolic control. Results Mice treated with the combination of anti CD3 and rapamycin had similar rates of diabetes reversal compared to anti CD3 alone (25/35 vs. 22/35). Mice treated with anti CD3 plus rapamycin had a significant improvement in glycemia control as exhibited by lower blood glucose levels in response to an intra-peritoneal glucose challenge; average peak blood glucose levels 30 min post intra-peritoneal injection of 2 gr/kg glucose were 6.9 mmol/L in the anti CD3 plus rapamycin group vs. 10 mmo/L in the anti CD3 alone (P<0.05). Conclusions/Interpretation The addition of rapamycin to anti CD3 results in significant improvement in glycaemia control in diabetic NOD mice.
Collapse
Affiliation(s)
- Shira Perl
- Center for Human Immunology, NHLBI, NIH, Bethesda, Maryland, United States of America.
| | | | | | | | | | | |
Collapse
|
23
|
Immunotherapy with Tolerogenic Dendritic Cells Alone or in Combination with Rapamycin Does Not Reverse Diabetes in NOD Mice. ISRN ENDOCRINOLOGY 2013; 2013:346987. [PMID: 23555060 PMCID: PMC3608187 DOI: 10.1155/2013/346987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 02/22/2013] [Indexed: 11/17/2022]
Abstract
Type 1 diabetes is a metabolic disease caused by autoimmunity towards β -cells. Different strategies have been developed to restore β -cell function and to reestablish immune tolerance to prevent and cure the disease. Currently, there is no effective treatment strategy to restore endogenous insulin secretion in patients with type 1 diabetes. This study aims to restore insulin secretion in diabetic mice with experimental antigen-specific immunotherapy alone or in combination with rapamycin, a compound well known for its immunomodulatory effect. Nonobese diabetic (NOD) mice develop spontaneous type 1 diabetes after 12 weeks of age. Autologous tolerogenic dendritic cells-consisting in dendritic cells pulsed with islet apoptotic cells-were administered to diabetic NOD mice alone or in combination with rapamycin. The ability of this therapy to revert type 1 diabetes was determined by assessing the insulitis score and by measuring both blood glucose levels and C-peptide concentration. Our findings indicate that tolerogenic dendritic cells alone or in combination with rapamycin do not ameliorate diabetes in NOD mice. These results suggest that alternative strategies may be considered for the cure of type 1 diabetes.
Collapse
|
24
|
Ehlers MR, Nepom GT. Immune-directed therapy for type 1 diabetes at the clinical level: the Immune Tolerance Network (ITN) experience. Rev Diabet Stud 2012; 9:359-71. [PMID: 23804273 DOI: 10.1900/rds.2012.9.359] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Reestablishing immune tolerance in type 1 diabetes (T1D), a chronic autoimmune disease, is a major goal. The Immune Tolerance Network (ITN) has initiated eight clinical trials of immunomodulatory therapies in recent-onset T1D over the past decade. Results have been mixed in terms of clinical efficacy, but the studies have provided valuable mechanistic insight that are enhancing our understanding of the disease and guiding the design of future trials. Trials of non-Fc-binding anti-CD3 mAbs have revealed that modulation of this target leads to partial responses, and ITN's AbATE trial led to identification of a robust responder group that could be distinguished from non-responders by baseline metabolic and immunologic features. A pilot study of the combination of IL-2 and rapamycin gave the first demonstration that frequency and function of regulatory T cells (Tregs) can be enhanced in T1D subjects, although the therapy triggered the activation of effectors with transient β-cell dysfunction. Similarly, therapy with anti-thymocyte globulin led to substantial lymphocyte depletion, but also to the activation of the acute-phase response with no clinical benefit during preliminary analyses. These and other results provide mechanistic tools that can be used as biomarkers for safety and efficacy in future trials. Furthermore, our results, together with those of other organizations, notably TrialNet, delineate the roles of the major components of the immune response in T1D. This information is setting the stage for future combination therapy trials. The development of disease-relevant biomarkers will also enable the implementation of innovative trial designs, notably adaptive trials, which will increase efficiencies in terms of study duration and sample size, and which will expedite the conduct of trials in which there are uncertainties about dose response and effect size.
Collapse
Affiliation(s)
- Mario R Ehlers
- Clinical Trials Group, Immune Tolerance Network, San Francisco, CA, USA.
| | | |
Collapse
|
25
|
Affiliation(s)
- Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany.
| |
Collapse
|
26
|
Azzi J, Moore RF, Elyaman W, Mounayar M, El Haddad N, Yang S, Jurewicz M, Takakura A, Petrelli A, Fiorina P, Ruckle T, Abdi R. The novel therapeutic effect of phosphoinositide 3-kinase-γ inhibitor AS605240 in autoimmune diabetes. Diabetes 2012; 61:1509-18. [PMID: 22403300 PMCID: PMC3357271 DOI: 10.2337/db11-0134] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) remains a major health problem worldwide, with a steadily rising incidence yet no cure. Phosphoinositide 3-kinase-γ (PI3Kγ), a member of a family of lipid kinases expressed primarily in leukocytes, has been the subject of substantial research for its role in inflammatory diseases. However, the role of PI3Kγ inhibition in suppressing autoimmune T1D remains to be explored. We tested the role of the PI3Kγ inhibitor AS605240 in preventing and reversing diabetes in NOD mice and assessed the mechanisms by which this inhibition abrogates T1D. Our data indicate that the PI3Kγ pathway is highly activated in T1D. In NOD mice, we found upregulated expression of phosphorylated Akt (PAkt) in splenocytes. Notably, T regulatory cells (Tregs) showed significantly lower expression of PAkt compared with effector T cells. Inhibition of the PI3Kγ pathway by AS605240 efficiently suppressed effector T cells and induced Treg expansion through the cAMP response element-binding pathway. AS605240 effectively prevented and reversed autoimmune diabetes in NOD mice and suppressed T-cell activation and the production of inflammatory cytokines by autoreactive T cells in vitro and in vivo. These studies demonstrate the key role of the PI3Kγ pathway in determining the balance of Tregs and autoreactive cells regulating autoimmune diabetes.
Collapse
Affiliation(s)
- Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Robert F. Moore
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Wassim Elyaman
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Marwan Mounayar
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Najib El Haddad
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Sunmi Yang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Mollie Jurewicz
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Ayumi Takakura
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alessandra Petrelli
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Paolo Fiorina
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Thomas Ruckle
- Therapeutic Area Neurodegenerative Diseases, Merck Serono S.A., Geneva, Switzerland
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Reza Abdi,
| |
Collapse
|
27
|
Roep BO, Buckner J, Sawcer S, Toes R, Zipp F. The problems and promises of research into human immunology and autoimmune disease. Nat Med 2012; 18:48-53. [PMID: 22227672 DOI: 10.1038/nm.2626] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Bart O Roep
- Leiden University Medical Center, National Diabetes Expert Center, Department of Immunohaematology and Blood Transfusion, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
28
|
Battaglia M, Roncarolo MG. Immune intervention with T regulatory cells: past lessons and future perspectives for type 1 diabetes. Semin Immunol 2011; 23:182-94. [PMID: 21831659 DOI: 10.1016/j.smim.2011.07.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 07/10/2011] [Indexed: 01/11/2023]
Abstract
In type 1 diabetes (T1D), insulin-producing pancreatic β-cells are attacked and destroyed by the immune system. Although man-made insulin is life-saving, it is not a cure and it cannot prevent long-term complications. In addition, most T1D patients would do almost anything to achieve release from the burden of daily glucose monitoring and insulin injection. Despite the formation of very large and promising clinical trials, a means to prevent/cure T1D in humans remains elusive. This has led to an increasing interest in the possibility of using T cells with regulatory properties (Treg cells) as a biological therapy to preserve and restore tolerance to self-antigens. In the present review we will attempt to consolidate learning from the past and to describe what we now believe could in the future become a successful Treg-cell based immune intervention in T1D.
Collapse
Affiliation(s)
- Manuela Battaglia
- San Raffaele Diabetes Research Institute, via Olgettina 58, 20132 Milan, Italy.
| | | |
Collapse
|
29
|
Hedl M, Abraham C. Secretory mediators regulate Nod2-induced tolerance in human macrophages. Gastroenterology 2011; 140:231-41. [PMID: 20854823 PMCID: PMC3145247 DOI: 10.1053/j.gastro.2010.09.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 07/29/2010] [Accepted: 09/09/2010] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Nucleotide oligomerization domain 2 (Nod2) polymorphisms increase the risk of developing Crohn's disease, which is characterized by chronic intestinal inflammation. Bacterial peptidoglycan products chronically stimulate Nod2 in the intestine. Recent studies found that chronic Nod2 stimulation in human macrophages down-regulates proinflammatory cytokines on Nod2 or Toll-like receptor (TLR) restimulation. Therefore, an emerging hypothesis is that Nod2-mediated cytokine down-regulation is required for intestinal homeostasis, but the mechanisms mediating this down-regulation are incompletely understood. METHODS Utilizing primary human macrophages, we examined secretory mediators as a mechanism of Nod2-mediated tolerance by inhibiting their function and assessing tolerance reversal through cytokine secretion. Signaling pathways contributing to secretory mediator induction and Nod2-mediated tolerance were identified through pathway inhibition. RESULTS We found that chronic Nod2 stimulation cross-tolerizes not only to TLRs but also to the interleukin (IL)-1 receptor. Moreover, chronic IL-1β stimulation down-regulates Nod2 responses. Accordingly, IL-1β blockade partially reverses Nod2-mediated tolerance. We found that an additional essential mechanism for Nod2-mediated tolerance is the early secretion of the anti-inflammatory mediators IL-10, transforming growth factor β, and IL-1Ra. Importantly, the mammalian target of rapamycin (mTOR) pathway, involved in cell growth, differentiation, and activation, significantly contributes to Nod2-induced anti-inflammatory as opposed to proinflammatory cytokines and to Nod2-mediated tolerance. CONCLUSIONS Inflammatory responses through the IL-1R are down-regulated upon chronic Nod2 stimulation, secretory mediators are a critical mechanism for Nod2-mediated cytokine down-regulation, and the mTOR pathway is crucial for Nod2-mediated tolerance. These results further contribute to our understanding of the mechanisms through which Nod2, a protein critical to intestinal homeostasis, down-regulates cytokine responses.
Collapse
Affiliation(s)
- Matija Hedl
- Department of Medicine, Yale University, New Haven, CT 06511
| | - Clara Abraham
- Department of Medicine, Yale University, New Haven, CT 06511,To whom correspondence may be addressed: Clara Abraham, MD, Department of Internal Medicine, Section of Digestive Diseases, 333 Cedar Street (LMP 1080), New Haven, CT 06520,
| |
Collapse
|
30
|
Cooles FAH, Isaacs JD. Treating to re-establish tolerance in inflammatory arthritis - lessons from other diseases. Best Pract Res Clin Rheumatol 2010; 24:497-511. [PMID: 20732648 DOI: 10.1016/j.berh.2010.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapeutic tolerance embraces the concept of 'switching off' immunopathology by specifically targeting elements of the immune system. It has been achievable in preclinical models of transplantation and auto-immunity for more than two decades; however, previous attempts to translate to the clinic have been unsuccessful. Nonetheless, an improved understanding of tolerance mechanisms, along with novel therapeutic agents and strategies, are starting to bear fruit in a number of disease areas. True tolerance is achievable in transplantation settings, and long-term remissions can be induced in various auto-immune and atopic conditions. Equivalent outcomes should be achievable in inflammatory arthritis, although this may require an improved understanding of the immune dysregulation that is intrinsic to rheumatoid arthritis (RA), and better definitions of RA autoantigens. Biomarkers of tolerance induction would rapidly advance the field in all therapeutic areas. This article summarises the advances made in other therapeutic areas, and the lessons learned that we can now apply to RA.
Collapse
Affiliation(s)
- Faye A H Cooles
- Institute of Cellular Medicine, Musculoskeletal Research Group, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| | | |
Collapse
|
31
|
Driver JP, Serreze DV, Chen YG. Mouse models for the study of autoimmune type 1 diabetes: a NOD to similarities and differences to human disease. Semin Immunopathol 2010; 33:67-87. [DOI: 10.1007/s00281-010-0204-1] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/18/2010] [Indexed: 01/12/2023]
|
32
|
Surrogate end points in the design of immunotherapy trials: emerging lessons from type 1 diabetes. Nat Rev Immunol 2010; 10:145-52. [DOI: 10.1038/nri2705] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
33
|
Mehta DS, Christmas RA, Waldmann H, Rosenzweig M. Partial and transient modulation of the CD3-T-cell receptor complex, elicited by low-dose regimens of monoclonal anti-CD3, is sufficient to induce disease remission in non-obese diabetic mice. Immunology 2010; 130:103-13. [PMID: 20059577 DOI: 10.1111/j.1365-2567.2009.03217.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
It has been established that a total of 250 microg of monoclonal anti-mouse CD3 F(ab')(2) fragments, administered daily (50 microg per dose), induces remission of diabetes in the non-obese diabetic (NOD) mouse model of autoimmune diabetes by preventing beta cells from undergoing further autoimmune attack. We evaluated lower-dose regimens of monoclonal anti-CD3 F(ab')(2) in diabetic NOD mice for their efficacy and associated pharmacodynamic (PD) effects, including CD3-T-cell receptor (TCR) complex modulation, complete blood counts and proportions of circulating CD4(+), CD8(+) and CD4(+) FoxP3(+) T cells. Four doses of 2 microg (total dose 8 microg) induced 53% remission of diabetes, similarly to the 250 microg dose regimen, whereas four doses of 1 microg induced only 16% remission. While the 250 microg dose regimen produced nearly complete and sustained modulation of the CD3 -TCR complex, lower doses, spaced 3 days apart, which induced similar remission rates, elicited patterns of transient and partial modulation. In treated mice, the proportions of circulating CD4(+) and CD8(+) T cells decreased, whereas the proportions of CD4(+) FoxP3(+) T cells increased; these effects were transient. Mice with greater residual beta-cell function, estimated using blood glucose and C-peptide levels at the initiation of treatment, were more likely to enter remission than mice with more advanced disease. Thus, lower doses of monoclonal anti-CD3 that produced only partial and transient modulation of the CD3-TCR complex induced remission rates comparable to higher doses of monoclonal anti-CD3. Accordingly, in a clinical setting, lower-dose regimens may be efficacious and may also improve the safety profile of therapy with monoclonal anti-CD3, potentially including reductions in cytokine release-related syndromes and maintenance of pathogen-specific immunosurveillance during treatment.
Collapse
|
34
|
Abstract
Since their discovery by Steinman and Cohn in 1973, dendritic cells (DCs) have become increasingly recognized for their crucial role as regulators of innate and adaptive immunity. DCs are exquisitely adept at acquiring, processing, and presenting antigens to T cells. They also adjust the context (and hence the outcome) of antigen presentation in response to a plethora of environmental inputs that signal the occurrence of pathogens or tissue damage. Such signals generally boost DC maturation, which promotes their migration from peripheral tissues into and within secondary lymphoid organs and their capacity to induce and regulate effector T cell responses. Conversely, more recent observations indicate that DCs are also crucial to ensure immunological peace. Indeed, DCs constantly present innocuous self- and nonself-antigens in a fashion that promotes tolerance, at least in part, through the control of regulatory T cells (Tregs). Tregs are specialized T cells that exert their immunosuppressive function through a variety of mechanisms affecting both DCs and effector cells. Here, we review recent advances in our understanding of the relationship between tolerogenic DCs and Tregs.
Collapse
|
35
|
Current literature in diabetes. Diabetes Metab Res Rev 2009; 25:i-x. [PMID: 19790194 DOI: 10.1002/dmrr.1037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
36
|
Abstract
The potent immunosuppressive action of rapamycin is commonly ascribed to inhibition of growth factor-induced T cell proliferation. However, it is now evident that the serine/threonine protein kinase mammalian target of rapamycin (mTOR) has an important role in the modulation of both innate and adaptive immune responses. mTOR regulates diverse functions of professional antigen-presenting cells, such as dendritic cells (DCs), and has important roles in the activation of effector T cells and the function and proliferation of regulatory T cells. In this Review, we discuss our current understanding of the mTOR pathway and the consequences of mTOR inhibition, both in DCs and T cells, including new data on the regulation of forkhead box P3 expression.
Collapse
|