1
|
Parham KA, Tan XXS, Morelli DM, Chowdhury L, Craig HC, Kerfoot SM. Pre–Germinal Center Interactions with T Cells Are Natural Checkpoints to Limit Autoimmune B Cell Responses. THE JOURNAL OF IMMUNOLOGY 2022; 209:1703-1712. [DOI: 10.4049/jimmunol.2200534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/25/2022] [Indexed: 11/05/2022]
|
2
|
Wilson CS, Stocks BT, Hoopes EM, Rhoads JP, McNew KL, Major AS, Moore DJ. Metabolic preconditioning in CD4+ T cells restores inducible immune tolerance in lupus-prone mice. JCI Insight 2021; 6:e143245. [PMID: 34403367 PMCID: PMC8525586 DOI: 10.1172/jci.insight.143245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
Autoimmune disease has presented an insurmountable barrier to restoration of durable immune tolerance. Previous studies indicate that chronic therapy with metabolic inhibitors can reduce autoimmune inflammation, but it remains unknown whether acute metabolic modulation enables permanent immune tolerance to be established. In an animal model of lupus, we determined that targeting glucose metabolism with 2-deoxyglucose (2DG) and mitochondrial metabolism with metformin enables endogenous immune tolerance mechanisms to respond to tolerance induction. A 2-week course of 2DG and metformin, when combined with tolerance-inducing therapy anti-CD45RB, prevented renal deposition of autoantibodies for 6 months after initial treatment and restored tolerance induction to allografts in lupus-prone mice. The restoration of durable immune tolerance was linked to changes in T cell surface glycosylation patterns, illustrating a role for glycoregulation in immune tolerance. These findings indicate that metabolic therapy may be applied as a powerful preconditioning to reinvigorate tolerance mechanisms in autoimmune and transplant settings that resist current immune therapies.
Collapse
Affiliation(s)
| | | | - Emilee M. Hoopes
- Ian Burr Division of Endocrinology and Diabetes, Department of Pediatrics
| | | | | | - Amy S. Major
- Department of Pathology, Microbiology, and Immunology; and
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daniel J. Moore
- Ian Burr Division of Endocrinology and Diabetes, Department of Pediatrics
- Department of Pathology, Microbiology, and Immunology; and
| |
Collapse
|
3
|
Host Expression of the CD8 Treg/NK Cell Restriction Element Qa-1 is Dispensable for Transplant Tolerance. Sci Rep 2017; 7:11181. [PMID: 28894277 PMCID: PMC5593978 DOI: 10.1038/s41598-017-11780-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/30/2017] [Indexed: 11/08/2022] Open
Abstract
Disruption of the non-classical Major Histocompatibility Complex (MHC) Ib molecule Qa-1 impairs CD8 Treg and natural killer (NK) cell function and promotes a lupus-like autoimmune disease. This immune perturbation would be expected to enhance anti-transplant responses and impair tolerance induction, but the effect of Qa-1 deficiency on the transplant response has not been previously reported. Qa-1 deficiency enhanced CD4 TFH and germinal center (GC) B cell numbers in naïve mice and hastened islet allograft rejection. Despite enhanced immunity in B6.Qa-1-/- mice, these mice did not generate an excessive primary CD4 TFH cell response nor an enhanced alloantibody reaction. Both CD8 Tregs and NK cells, which often regulate other cells through host Qa-1 expression, were targets of anti-CD45RB therapy that had not been previously recognized. However, B6.Qa-1-/- mice remained susceptible to anti-CD45RB mediated suppression of the alloantibody response and transplant tolerance induction to mismatched islet allografts. Overall, despite enhanced immunity as demonstrated by augmented CD4 TFH/GC B cell numbers and hastened islet allograft rejection in naïve 12-week old Qa-1 deficient mice, the CD8 Treg/NK cell restriction element Qa-1 does not regulate the primary cellular or humoral alloresponse and is not required for long-term transplant tolerance.
Collapse
|
4
|
Reeves PL, Rudraraju R, Liu X, Wong FS, Hamilton-Williams EE, Steptoe RJ. APC-targeted proinsulin expression inactivates insulin-specific memory CD8 + T cells in NOD mice. Immunol Cell Biol 2017; 95:765-774. [PMID: 28611473 DOI: 10.1038/icb.2017.48] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) results from T-cell-mediated autoimmune destruction of pancreatic β cells. Effector T-cell responses emerge early in disease development and expand as disease progresses. Following β-cell destruction, a long-lived T-cell memory is generated that represents a barrier to islet transplantation and other cellular insulin-replacement therapies. Development of effective immunotherapies that control or ablate β-cell destructive effector and memory T-cell responses has the potential to prevent disease progression and recurrence. Targeting antigen expression to antigen-presenting cells inactivates cognate CD8+ effector and memory T-cell responses and has therapeutic potential. Here we investigated this in the context of insulin-specific responses in the non-obese diabetic mouse where genetic immune tolerance defects could impact on therapeutic tolerance induction. Insulin-specific CD8+ memory T cells transferred to mice expressing proinsulin in antigen-presenting cells proliferated in response to transgenically expressed proinsulin and the majority were rapidly deleted. A small proportion of transferred insulin-specific Tmem remained undeleted and these were antigen-unresponsive, exhibited reduced T cell receptor (TCR) expression and H-2Kd/insB15-23 tetramer binding and expressed co-inhibitory molecules. Expression of proinsulin in antigen-presenting cells also abolished the diabetogenic capacity of CD8+ effector T cells. Therefore, destructive insulin-specific CD8+ T cells are effectively inactivated by enforced proinsulin expression despite tolerance defects that exist in diabetes-prone NOD mice. These findings have important implications in developing immunotherapeutic approaches to T1D and other T-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Peta Ls Reeves
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Rajeev Rudraraju
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Xiao Liu
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - F Susan Wong
- Institute of Molecular &Experimental Medicine, Cardiff University School of Medicine, Cardiff, Wales
| | | | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| |
Collapse
|
5
|
Stocks BT, Thomas AB, Elizer SK, Zhu Y, Marshall AF, Wilson CS, Moore DJ. Hematopoietic Stem Cell Mobilization Is Necessary but Not Sufficient for Tolerance in Islet Transplantation. Diabetes 2017; 66:127-133. [PMID: 27797908 PMCID: PMC5204317 DOI: 10.2337/db16-0444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 10/21/2016] [Indexed: 11/13/2022]
Abstract
Overcoming the immune response to establish durable immune tolerance in type 1 diabetes remains a substantial challenge. The ongoing effector immune response involves numerous immune cell types but is ultimately orchestrated and sustained by the hematopoietic stem cell (HSC) niche. We therefore hypothesized that tolerance induction also requires these pluripotent precursors. In this study, we determined that the tolerance-inducing agent anti-CD45RB induces HSC mobilization in nonautoimmune B6 mice but not in diabetes-prone NOD mice. Ablation of HSCs impaired tolerance to allogeneic islet transplants in B6 recipients. Mobilization of HSCs resulted in part from decreasing osteoblast expression of HSC retention factors. Furthermore, HSC mobilization required a functioning sympathetic nervous system; sympathectomy prevented HSC mobilization and completely abrogated tolerance induction. NOD HSCs were held in their niche by excess expression of CXCR4, which, when blocked, led to HSC mobilization and prolonged islet allograft survival. Overall, these findings indicate that the HSC compartment plays an underrecognized role in the establishment and maintenance of immune tolerance, and this role is disrupted in diabetes-prone NOD mice. Understanding the stem cell response to immune therapies in ongoing human clinical studies may help identify and maximize the effect of immune interventions for type 1 diabetes.
Collapse
Affiliation(s)
- Blair T Stocks
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Analise B Thomas
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Sydney K Elizer
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Yuantee Zhu
- Departartment of Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Andrew F Marshall
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| | - Christopher S Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Daniel J Moore
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
6
|
Wilson CS, Elizer SK, Marshall AF, Stocks BT, Moore DJ. Regulation of B lymphocyte responses to Toll-like receptor ligand binding during diabetes prevention in non-obese diabetic (NOD) mice. J Diabetes 2016; 8:120-31. [PMID: 25564999 PMCID: PMC4598313 DOI: 10.1111/1753-0407.12263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/12/2014] [Accepted: 12/23/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Interactions between genetic risk factors and the environment drive type 1 diabetes (T1D). The system of Toll-like receptors (TLR) detects these environmental triggers; however, the target cell that intermediates these interactions to drive T1D remains unknown. METHODS We investigated the effect of TLR pathway activation (myeloid differentiation primary response 88 [MyD88] vs TIR-domain-containing adapter-inducing interferon-β [TRIF]) on B cell subsets via flow cytometry, including their activation, survival, proliferation, and cytoskeletal mobilization. The effect of polyinosinic-polycytidylic acid (poly(I:C)) on diabetes development was addressed, including the B cell-dependent activation of diabetes-protective DX5+ cells, using genetic models and adoptive transfer. RESULTS B lymphocytes from non-obese diabetic (NOD) mice expressed enhanced levels of TLR-responsive proteins. Ex vivo analysis of B lymphocyte subsets demonstrated that TLR3 stimulation via TRIF deletes cells exhibiting a marginal zone phenotype, whereas MyD88-dependent ligands enhance their survival. In vivo, marginal zone B cells were activated by poly(I:C) and were unexpectedly retained in the spleen of NOD mice, in contrast with the mobilization of these cells in non-autoimmune mice, a phenotype we traced to defective actin cytoskeletal dynamics. These activated B cells mediated TLR3-induced diabetes protection. CONCLUSIONS Immunotherapies must account for both B cell location and activation, and these properties may differ in autoimmune and healthy settings.
Collapse
Affiliation(s)
- Christopher S. Wilson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, School of Medicine, 1161 21st Ave South. Nashville, TN 37232-2363
| | - Sydney K. Elizer
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, School of Medicine, 2200 Children's Way. Nashville, TN 37232-2363
| | - Andrew F. Marshall
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, School of Medicine, 2200 Children's Way. Nashville, TN 37232-2363
| | - Blair T. Stocks
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, School of Medicine, 1161 21st Ave South. Nashville, TN 37232-2363
- Vanderbilt Medical Scientist Training Program
| | - Daniel J. Moore
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, School of Medicine, 1161 21st Ave South. Nashville, TN 37232-2363
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, School of Medicine, 2200 Children's Way. Nashville, TN 37232-2363
| |
Collapse
|
7
|
Stocks BT, Wilhelm AJ, Wilson CS, Marshall AF, Putnam NE, Major AS, Moore DJ. Lupus-Prone Mice Resist Immune Regulation and Transplant Tolerance Induction. Am J Transplant 2016; 16:334-41. [PMID: 26372909 PMCID: PMC4718751 DOI: 10.1111/ajt.13449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/26/2015] [Accepted: 07/09/2015] [Indexed: 01/25/2023]
Abstract
The strongly immunogenic environment in autoimmune diseases such as lupus may pose a stringent barrier to transplantation. Despite available murine models of lupus, transplant tolerance in this setting has yet to be fully investigated in highly penetrant genetic models of disease. Such studies are of clear clinical importance because lupus is a transplant indication in which transplanted kidneys have a substantially increased risk of rejection including a role for recurrent nephritis. In the fully penetrant B6.SLE123 mouse, we determined that CD4 T follicular helper and germinal center B cells were significantly expanded compared with healthy controls. We traced this expansion to resistance of effector CD4 T and B cells in B6.SLE123 mice to regulation by either CD4 T regulatory cells (CD4Tregs) or CD8 T regulatory cells (CD8Tregs), despite demonstrating normal function by Tregs in this strain. Finally, we determined that B6.SLE123 mice resist anti-CD45RB-mediated tolerance induction to foreign islet allografts, even in the absence of islet autoimmunity. Overall, B6.SLE123 lupus-prone mice are highly resistant to transplant tolerance induction, which provides a new model of failed tolerance in autoimmunity that may elucidate barriers to clinical transplantation in lupus through further cellular and genetic dissection.
Collapse
Affiliation(s)
- B. T. Stocks
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN
| | - A. J. Wilhelm
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN
| | - C. S. Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN
| | - A. F. Marshall
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, Nashville, TN
| | | | - A. S. Major
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN,Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN
| | - D. J. Moore
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN,Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt University, Nashville, TN,Corresponding author: Daniel J. Moore,
| |
Collapse
|
8
|
Zhao G, Moore DJ, Kim JI, Lee KM, O'Connor M, Yang M, Marshall AF, Lei J, Schuetz C, Markmann JF, Deng S. An immunosufficient murine model for the study of human islets. Xenotransplantation 2014; 21:567-73. [PMID: 25041432 DOI: 10.1111/xen.12126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 05/30/2014] [Indexed: 01/28/2023]
Abstract
For the sake of therapy of diabetes, it is critical to understand human beta cell function in detail in health and disease. Current studies of human beta cell physiology in vivo are mostly limited to immunodeficient mouse models, which possess significant technical limitations. This study aimed to create a new model for the study of human islets through induction of transplant tolerance in immunosufficient mice. B6 diabetic mice were transplanted with human islets and treated with anti-CD45RB. To assess whether anti-CD45RB-induced transplant tolerance requires B cells, B6 recipients received additional anti-CD20 or B6μMT-/- mice were used. For some anti-CD45RB-treated B6μMT-/- mice, additional anti-CD25 mAb was applied at the early or late stage post-transplant. Immunohistology was performed to show the Foxp3 cells in grafted anti-CD45RB/anti-CD20-treated Foxp3-GFP B6 mice. The results showed that anti-CD45RB alone allowed indefinite graft survival in 26.6% of B6 mice, however 100% of xenografts were accepted in mice treated simultaneously with anti-CD20, and 88.9% of xenografts accepted in anti-CD45RB-treated μMT-/- mice. These μMT-/- mice accepted the islets from another human donor but rejected the islets from baboon. Additional administration of anti-CD25 mAb at the time of transplantation resulted in 100% rejection, whereas 40% of grafts were rejected while the antibody was administrated at days 60 post-transplant. Immunohistologic examination showed Foxp3+ cells accumulated around grafts. We conclude that induction of tolerance to human islets in an immunosufficient mouse model could be generated by targeting murine CD45RB and CD20. This new system will facilitate study of human islets and accelerate the dissection of the critical mechanisms underlying islet health in human disease.
Collapse
Affiliation(s)
- Gaoping Zhao
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Sichuan Provincial Key Laboratory for Translational Medicine of Organ Transplantation, Department of Surgery, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Lee KM, Kim JI, Stott R, Soohoo J, O’Connor MR, Yeh H, Zhao G, Eliades P, Fox C, Cheng N, Deng S, Markmann JF. Anti-CD45RB/anti-TIM-1-induced tolerance requires regulatory B cells. Am J Transplant 2012; 12:2072-8. [PMID: 22494812 PMCID: PMC3396747 DOI: 10.1111/j.1600-6143.2012.04055.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The role of B cells in transplant tolerance remains unclear. Although B-cell depletion often prolongs graft survival, sometimes it results in more rapid rejection, suggesting that B cells may have regulatory activity. We previously demonstrated that tolerance induction by anti-CD45RB antibody requires recipient B cells. Here, we show that anti-CD45RB in combination with anti-TIM-1 antibody has a synergistic effect, inducing tolerance in all recipients in a mouse islet allograft model. This effect depends on the presence of recipient B cells, requires B-cell IL-10 activity, and is antigen-specific. These data suggest the existence of a regulatory B-cell population that promotes tolerance via an IL-10-dependent pathway.
Collapse
Affiliation(s)
- Kang Mi Lee
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - James I. Kim
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Ryan Stott
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Julie Soohoo
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Matthew R O’Connor
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Heidi Yeh
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Gaoping Zhao
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,Department of Surgery, Sichuan Provincial People’s Hospital and Sichuan Academy of Medical Sciences, Chengdu 610072, Sichuan Province, P. R. China
| | - Philip Eliades
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Courtney Fox
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Nan Cheng
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Shaoping Deng
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,Department of Surgery, Sichuan Provincial People’s Hospital and Sichuan Academy of Medical Sciences, Chengdu 610072, Sichuan Province, P. R. China
| | - James F. Markmann
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
10
|
Leuschner F, Dutta P, Gorbatov R, Novobrantseva TI, Donahoe JS, Courties G, Lee KM, Kim JI, Markmann JF, Marinelli B, Panizzi P, Lee WW, Iwamoto Y, Milstein S, Epstein-Barash H, Cantley W, Wong J, Cortez-Retamozo V, Newton A, Love K, Libby P, Pittet MJ, Swirski FK, Koteliansky V, Langer R, Weissleder R, Anderson DG, Nahrendorf M. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat Biotechnol 2011; 29:1005-10. [PMID: 21983520 PMCID: PMC3212614 DOI: 10.1038/nbt.1989] [Citation(s) in RCA: 664] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 08/29/2011] [Indexed: 12/24/2022]
Abstract
Inflammatory monocytes -- but not the non-inflammatory subset -- depend on the chemokine receptor CCR2 for distribution to injured tissue and stimulate disease progression. Precise therapeutic targeting of this inflammatory monocyte subset could spare innate immunity's essential functions for maintenance of homeostasis and thus limit unwanted effects. Here we developed siRNA nanoparticles targeting CCR2 expression in inflammatory monocytes. We identified an optimized lipid nanoparticle and silencing siRNA sequence that when administered systemically, had rapid blood clearance, accumulated in spleen and bone marrow and showed high cellular localization of fluorescently tagged siRNA inside monocytes. Efficient degradation of CCR2 mRNA in monocytes prevented their accumulation in sites of inflammation. Specifically, the treatment attenuated their number in atherosclerotic plaques, reduced infarct size following coronary artery occlusion, prolonged normoglycemia in diabetic mice after pancreatic islet transplantation and resulted in reduced tumor volumes and lower numbers of tumor-associated macrophages. Taken together, siRNA nanoparticle-mediated CCR2 gene silencing in leukocytes selectively modulates functions of innate immune cell subtypes and may allow for the development of specific anti-inflammatory therapy.
Collapse
Affiliation(s)
- Florian Leuschner
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhao G, Moore DJ, Kim JI, Lee KM, O'Connor MR, Duff PE, Yang M, Lei J, Markmann JF, Deng S. Inhibition of transplantation tolerance by immune senescence is reversed by endocrine modulation. Sci Transl Med 2011; 3:87ra52. [PMID: 21677198 DOI: 10.1126/scitranslmed.3002270] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The senescent immune system responds poorly to new stimuli; thymic involution, accumulation of memory cells against other specificities, and general refractoriness to antigen signaling all may contribute to poor resistance to infection. These same changes may pose a significant clinical barrier to organ transplantation, as transplantation tolerance requires thymic participation and integrated, tolerance-promoting responses to novel antigens. We found that after the age of 12 months, mice became resistant to the tolerance-inducing capacity of the monoclonal antibody therapy anti-CD45RB. This resistance to tolerance to cardiac allografts could be overcome by surgical castration of male mice, a procedure that led to thymic regeneration and long-term graft acceptance. The potential for clinical translation of this endocrine-immune interplay was confirmed by the ability of Lupron Depot injections, which temporarily disrupt gonadal function, to restore tolerance in aged mice. Furthermore, we demonstrated that the restoration of tolerance after surgical or chemical castration depended on thymic production of regulatory T cells (T(regs)); thymectomy or T(reg) depletion abrogated tolerance restoration. The aging of the immune system ("immune senescence") is a significant barrier to immune tolerance, but this barrier can be overcome by targeting sex steroid production with commonly used clinical therapeutics.
Collapse
Affiliation(s)
- Gaoping Zhao
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhao G, Moore DJ, Lee KM, Kim JI, Duff PE, O’Connor MR, Hirohashi T, Lei J, Yang M, Markmann JF, Deng S. An unexpected counter-regulatory role of IL-10 in B-lymphocyte-mediated transplantation tolerance. Am J Transplant 2010; 10:796-801. [PMID: 20199511 PMCID: PMC2934759 DOI: 10.1111/j.1600-6143.2010.03027.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Monoclonal antibody against the CD45RB protein induces stable transplantation tolerance to multiple types of allograft. We have previously established that this tolerance protocol relies on the regulatory function of B lymphocytes for its effect. B lymphocytes have also been reported to participate in immune regulation in several other settings. In most of these systems, the regulatory function of B lymphocytes depends on the production of IL-10. Therefore, we investigated the role of IL-10 in the anti-CD45RB model of B-cell-mediated transplantation tolerance. Surprisingly, using antibody-mediated neutralization of IL-10, IL-10-deficient recipients and adoptive transfer of IL-10-deficient B lymphocytes, we found that IL-10 actually counter-regulates tolerance induced by anti-CD45RB. Furthermore, neutralization of IL-10 reduced the development of chronic allograft vasculopathy compared to anti-CD45RB alone and reduced the production of graft reactive alloantibodies. These data suggest that the participation of regulatory B lymphocytes in transplantation tolerance may be distinct from how they operate in other systems. Identifying the specific B lymphocytes that mediate transplantation tolerance and defining their mechanism of action may yield new insights into the complex cellular network through which antigen-specific tolerance is established and maintained.
Collapse
Affiliation(s)
- G. Zhao
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Sichuan Provincial People’s Hospital & Sichuan Academy of Medical Sciences, Chengdu, Sichuan Province, China
| | - D. J. Moore
- Department of Pediatrics, Ian Burr Division of Endocrinology and Diabetes, Vanderbilt Children’s Hospital, Nashville, TN
| | - K. M. Lee
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - J. I. Kim
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - P. E. Duff
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - M. R. O’Connor
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - T. Hirohashi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - J. Lei
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - M. Yang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Sichuan Provincial People’s Hospital & Sichuan Academy of Medical Sciences, Chengdu, Sichuan Province, China
| | - J. F. Markmann
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - S. Deng
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Sichuan Provincial People’s Hospital & Sichuan Academy of Medical Sciences, Chengdu, Sichuan Province, China
| |
Collapse
|
13
|
Key developmental transitions in human germinal center B cells are revealed by differential CD45RB expression. Blood 2008; 113:3999-4007. [PMID: 19059880 DOI: 10.1182/blood-2008-03-145979] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously reported that RO(+) expression correlated with increased mutation, activation, and selection among human germinal center (GC) B cells. Here, we subdivided human tonsillar B cells, including IgD(-)CD38(+) GC B cells, into different fractions based on RB expression. Although each subset contained RB(+) cells, when used as an intrasubset marker, differential RB expression effectively discriminated between phenotypically distinct cells. For example, RB(+) GC B cells were enriched for activated cells with lower AID expression. RB inversely correlated with mutation frequency, demonstrating a key difference between RB- and RO-expressing GC B cells. Reduced RB expression during the transition from pre-GC (IgM(+)IgD(+)CD38(+)CD27(-)) to GCB cells was followed by a dramatic increase during the GC-to-plasmablast (IgD(-)CD38(++)CD27(+)) and memory (IgD(-)CD38(-)CD27(+)) transition. Interestingly, RB(+) GC B cells showed increased signs of terminal differentiation toward CD27(+) post-GC early plasmablast (increased CD38 and RO) or early memory (decreased CD38 and RO) B cells. We propose that as in T cells, differential RB expression directly correlates with development- and function-based transitions in tonsillar B cells. Application of this RB:RO system should advance our understanding of normal B-cell development and facilitate the isolation of more discrete B-cell populations with potentially different propensities in disease pathogenesis.
Collapse
|
14
|
Huang X, Moore DJ, Ketchum RJ, Nunemaker CS, Kovatchev B, McCall AL, Brayman KL. Resolving the conundrum of islet transplantation by linking metabolic dysregulation, inflammation, and immune regulation. Endocr Rev 2008; 29:603-30. [PMID: 18664617 PMCID: PMC2819735 DOI: 10.1210/er.2008-0006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Accepted: 05/29/2008] [Indexed: 02/08/2023]
Abstract
Although type 1 diabetes cannot be prevented or reversed, replacement of insulin production by transplantation of the pancreas or pancreatic islets represents a definitive solution. At present, transplantation can restore euglycemia, but this restoration is short-lived, requires islets from multiple donors, and necessitates lifelong immunosuppression. An emerging paradigm in transplantation and autoimmunity indicates that systemic inflammation contributes to tissue injury while disrupting immune tolerance. We identify multiple barriers to successful islet transplantation, each of which either contributes to the inflammatory state or is augmented by it. To optimize islet transplantation for diabetes reversal, we suggest that targeting these interacting barriers and the accompanying inflammation may represent an improved approach to achieve successful clinical islet transplantation by enhancing islet survival, regeneration or neogenesis potential, and tolerance induction. Overall, we consider the proinflammatory effects of important technical, immunological, and metabolic barriers including: 1) islet isolation and transplantation, including selection of implantation site; 2) recurrent autoimmunity, alloimmune rejection, and unique features of the autoimmune-prone immune system; and 3) the deranged metabolism of the islet transplant recipient. Consideration of these themes reveals that each is interrelated to and exacerbated by the other and that this connection is mediated by a systemic inflammatory state. This inflammatory state may form the central barrier to successful islet transplantation. Overall, there remains substantial promise in islet transplantation with several avenues of ongoing promising research. This review focuses on interactions between the technical, immunological, and metabolic barriers that must be overcome to optimize the success of this important therapeutic approach.
Collapse
Affiliation(s)
- Xiaolun Huang
- Department of Surgery, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Huang X, Moore DJ, Mohiuddin M, Lian MM, Kim JI, Sonawane S, Wang J, Gu Y, Yeh H, Markmann JF, Deng S. Inhibition of ICAM-1/LFA-1 interactions prevents B-cell-dependent anti-CD45RB-induced transplantation tolerance. Transplantation 2008; 85:675-80. [PMID: 18337659 PMCID: PMC2934773 DOI: 10.1097/tp.0b013e3181663422] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Allogeneic tolerance can be reliably obtained with monoclonal antibody therapy targeting CD45RB. Although regulatory T cells play an important role in the mechanism, we have recently demonstrated the active participation of host B lymphocytes. After anti-CD45RB therapy, B lymphocytes demonstrate phenotypic alterations that include up-regulation of CD54 (intercellular adhesion molecule [ICAM]-1). We have investigated the hypothesis that alteration in ICAM-1 expression is required for tolerance induction. MATERIALS AND METHODS Recipients of heterotopic allogeneic cardiac grafts (C3H donors into B6 recipients) were treated with anti-CD45RB, anti-ICAM, anti-lymphocyte function-associated antigen-1 (LFA), or the combination of these agents. These data were extended by performing allogeneic cardiac transplants into ICAM or LFA recipients treated with a 5-day course of anti-CD45RB. Finally, B-cell-deficient animals were reconstituted with ICAM splenocytes to create a recipient with a selective deficiency of ICAM-1 restricted to the B-cell compartment. RESULTS Anti-CD45RB alone or the combination of anti-LFA/anti-ICAM reliably induced transplantation tolerance. However, the triple combination was routinely unsuccessful and induced long-term graft survival in no recipients. ICAM-deficient or LFA-deficient recipients were also resistant to tolerance induced by anti-CD45RB. Finally, transfer of control splenocytes to B-cell-deficient recipients permitted anti-CD45RB-induced tolerance, whereas transfer of ICAM cells was unable to support tolerance induction. CONCLUSIONS Expression of ICAM-1 by B lymphocytes and interaction with LFA-1 form a central aspect of transplantation tolerance induced by anti-CD45RB therapy. These data further elucidate the cellular mechanisms used by B lymphocytes in the induction of transplantation tolerance.
Collapse
Affiliation(s)
- Xiaolun Huang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; D.J.M. currently, Department of Pediatrics, Division of Endocrinology, Vanderbilt University, Nashville, Tennessee; M.M. currently, NIH/NHLBI, Bethesda, Maryland
| | - Daniel J. Moore
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; D.J.M. currently, Department of Pediatrics, Division of Endocrinology, Vanderbilt University, Nashville, Tennessee; M.M. currently, NIH/NHLBI, Bethesda, Maryland
| | - Mohammad Mohiuddin
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; D.J.M. currently, Department of Pediatrics, Division of Endocrinology, Vanderbilt University, Nashville, Tennessee; M.M. currently, NIH/NHLBI, Bethesda, Maryland
| | - Moh-Moh Lian
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; D.J.M. currently, Department of Pediatrics, Division of Endocrinology, Vanderbilt University, Nashville, Tennessee; M.M. currently, NIH/NHLBI, Bethesda, Maryland
| | - James I. Kim
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Samsher Sonawane
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; D.J.M. currently, Department of Pediatrics, Division of Endocrinology, Vanderbilt University, Nashville, Tennessee; M.M. currently, NIH/NHLBI, Bethesda, Maryland
| | - Jing Wang
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; D.J.M. currently, Department of Pediatrics, Division of Endocrinology, Vanderbilt University, Nashville, Tennessee; M.M. currently, NIH/NHLBI, Bethesda, Maryland
| | - Yi Gu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Heidi Yeh
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - James F. Markmann
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Shaoping Deng
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Surgery, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
16
|
Deng S, Moore DJ, Huang X, Lian MM, Mohiuddin M, Velededeoglu E, Lee MK, Sonawane S, Kim J, Wang J, Chen H, Corfe SA, Paige C, Shlomchik M, Caton A, Markmann JF. Cutting edge: transplant tolerance induced by anti-CD45RB requires B lymphocytes. THE JOURNAL OF IMMUNOLOGY 2007; 178:6028-32. [PMID: 17475825 DOI: 10.4049/jimmunol.178.10.6028] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Selective interference with the CD45RB isoform by mAb (anti-CD45RB) reliably induces donor-specific tolerance. Although previous studies suggest participation of regulatory T cells, a mechanistic understanding of anti-CD45RB-induced tolerance is lacking. We report herein the unexpected finding that tolerance induced by this agent is not established in B cell-deficient mice but can be recovered by preemptive B lymphocyte transfer to B cell-deficient hosts. Using B cells from genetically modified donors to reconstitute B cell-deficient recipients, we evaluate the role of B lymphocyte-expressed CD45RB, T cell costimulatory molecules, and the production of Abs in this novel tolerance mechanism. Our data document an Ab-induced tolerance regimen that is uniquely B lymphocyte-dependent and suggest mechanistic contributions to tolerance development from the B cell compartment through interactions with T cells.
Collapse
Affiliation(s)
- Shaoping Deng
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Truong W, Hancock WW, Anderson CC, Merani S, Shapiro AMJ. Coinhibitory T-cell signaling in islet allograft rejection and tolerance. Cell Transplant 2006; 15:105-19. [PMID: 16719045 DOI: 10.3727/000000006783982160] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Autoaggressive T cells directed against insulin secreting pancreatic beta-cells mediate the development of type 1 diabetes. Islet transplantation offers superior glycemic control over exogenous insulin, but chronic immunosuppression limits its broad application. Pathogenic T cells are also important in allograft rejection. Inducing and maintaining antigen-specific peripheral T-cell tolerance toward beta-cells is an attractive strategy to prevent autoimmune disease, and to facilitate treatment of diabetes with islet allografts without long-term immunosuppression. Recent efforts have focused on blocking costimulatory T-cell signals for tolerance induction. Although costimulatory blockade can prolong graft survival, true immunological tolerance remains elusive. Costimulatory signals may even be required for the maintenance of peripheral tolerance. The discovery of novel coinhibitory T-cell pathways, including CTLA-4, PD-1, and BTLA, offers an alternative approach. Stimulating negative T cell cosignals alone or in combination may help induce tolerance. The focus of this review is to summarize the strategies directed at turning off the immune response by exploiting these negative cosignaling pathways in tolerance induction in islet transplantation. Activating several coinhibitory pathways together may be synergistic in preventing pathogenic T-cell responses. Tolerance induction will likely rely on understanding the balance of positive and negative signals affecting the state of T-cell activation.
Collapse
Affiliation(s)
- Wayne Truong
- Department of Surgery, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | |
Collapse
|
18
|
Deng S, Moore DJ, Huang X, Mohiuddin M, Lee MK, Velidedeoglu E, Lian MM, Chiaccio M, Sonawane S, Orlin A, Wang J, Chen H, Caton A, Zhong R, Markmann JF. Antibody-induced transplantation tolerance that is dependent on thymus-derived regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:2799-807. [PMID: 16493036 DOI: 10.4049/jimmunol.176.5.2799] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Targeting of the CD45RB isoform by mAb (anti-CD45RB) effectively induces donor-specific tolerance to allografts. The immunological mechanisms underlying the tolerant state remain unclear although some studies have suggested the involvement of regulatory T cells (T-regs). Although their generative pathway remains undefined, tolerance promoting T-regs induced by systemic anti-CD45RB treatment have been assumed to originate in the peripheral immune system. We demonstrate herein that separable effects on the peripheral and central immune compartments mediate graft survival induced by anti-CD45RB administration. In the absence of the thymus, anti-CD45RB therapy is not tolerogenic though it retains peripheral immunosuppressive activity. The thymus is required for anti-CD45RB to produce indefinite graft survival and donor-specific tolerance, and this effect is accomplished through thymic production of donor-specific T-regs. These data reveal for the first time an Ab-based tolerance regimen that relies on the central tolerance pathway.
Collapse
Affiliation(s)
- Shaoping Deng
- Harrison Department of Surgical Research, Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Woodward EJ, Thomas JW. Multiple germline kappa light chains generate anti-insulin B cells in nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2005; 175:1073-9. [PMID: 16002708 DOI: 10.4049/jimmunol.175.2.1073] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The highly selective nature of organ-specific autoimmune disease is consistent with a critical role for adaptive immune responses against specific autoantigens. In type 1 diabetes mellitus, autoantibodies to insulin are important markers of the disease process in humans and nonobese diabetic (NOD) mice; however, the Ag-specific receptors responsible for these autoantibodies are obscured by the polyclonal repertoire. NOD mice that harbor an anti-insulin transgene (Tg) (V(H)125Tg/NOD) circumvent this problem by generating a tractable population of insulin-binding B cells. The nucleotide structure and genetic origin of the endogenous kappa L chain (Vkappa or IgL) repertoire that pairs with the V(H)125Tg were analyzed. In contrast to oligoclonal expansion observed in systemic autoimmune disease models, insulin-binding B cells from V(H)125Tg/NOD mice use specific Vkappa genes that are clonally independent and germline encoded. When compared with homologous IgL genes from nonautoimmune strains, Vkappa genes from NOD mice are polymorphic. Analysis of the most frequently expressed Vkappa1 and Vkappa9 genes indicates these are shared with lupus-prone New Zealand Black/BINJ mice (e.g., Vkappa1-110*02 and 9-124) and suggests that NOD mice use the infrequent b haplotype. These findings show that a diverse repertoire of anti-insulin B cells is part of the autoimmune process in NOD mice and structural or regulatory elements within the kappa locus may be shared with a systemic autoimmune disease.
Collapse
Affiliation(s)
- Emily J Woodward
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|