1
|
Lv J, Chen L, Zhao L. Renoprotective anti-CD45RB antibody induces B cell production in systemic lupus erythematosus based on single-cell RNA-seq analysis. J Autoimmun 2023; 134:102949. [PMID: 36455384 DOI: 10.1016/j.jaut.2022.102949] [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: 07/20/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that commonly affects the kidney. Single-cell RNA sequencing (scRNA-seq) technology is a powerful tool for characterizing individual cells and elucidating biological mechanisms at the cellular level. The purpose of this study was to identify the mechanism underlying kidney injury in SLE using scRNA-seq technology. METHODS scRNA-seq data of peripheral blood mononuclear cells (PBMCs) in SLE were retrieved from the GEO database, followed by batch effect elimination, dimensionality reduction, cluster analysis, cell annotation and enrichment analysis. A model of SLE was developed in NZB/WF1 mice. Effects of anti-CD45RB antibody on the SLE-induced kidney injury were evaluated, and we measured the distribution of regulatory T cells and B cells in mouse spleen and kidney tissues, levels of kidney function-related indexes, deposition of IgG and C3 in the glomeruli, and the levels of inflammatory cytokines. RESULTS CD45RB was a specific marker gene of B cell clusters and had influence on the B cells. anti-CD45RB antibody treatment induced regulatory B cells and consequently arrested the kidney injury caused by SLE. In addition, depletion of regulatory T cells was found to partially undermine the alleviatory effect of anti-CD45RB antibody on SLE-induced kidney injury. CONCLUSION Collectively, our data suggest that anti-CD45RB antibody can prevent the SLE-induced kidney injury, pointing to anti-CD45RB antibody as a potential therapeutic strategy in kidney injury-related disease.
Collapse
Affiliation(s)
- Juan Lv
- Department of Rheumatology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China; Department of Critical Care Medicine, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China.
| | - Lu Chen
- Department of Rheumatology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China.
| | - Ling Zhao
- Department of Rheumatology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China.
| |
Collapse
|
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: 2.7] [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
|
Levine MA, Chin JL, Rasmussen A, Sener A, Luke PP. The history of renal transplantation in Canada: A urologic perspective. Can Urol Assoc J 2020; 14:372-379. [PMID: 32569569 DOI: 10.5489/cuaj.6744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
While the urologist's involvement in kidney transplantation varies from center to center and country to country, urologists remain integral to many programs across Canada. From the early days of kidney transplant to contemporary times, the leadership, vision, and skillset of Canadian urologists have helped progress the field. In this review of Canadian urologists' role in kidney transplantation, the achievements of this professional group are highlighted and celebrated. Original contributors to the field, as well as notable achievements are highlighted, with a focus on the impact of Canadian urologists.
Collapse
Affiliation(s)
- Max Alexander Levine
- Department of Surgery, Division of Urology, Multiorgan Transplant Program, Western University, London, ON, Canada
| | - Joseph L Chin
- Department of Surgery, Division of Urology, Western University, London, ON, Canada
| | - Andrew Rasmussen
- Department of Surgery, Division of Urology, Multiorgan Transplant Program, Western University, London, ON, Canada
| | - Alp Sener
- Department of Surgery, Division of Urology, Multiorgan Transplant Program, Western University, London, ON, Canada
| | - Patrick P Luke
- Department of Surgery, Division of Urology, Multiorgan Transplant Program, Western University, London, ON, Canada
| |
Collapse
|
4
|
Li DY, Xie SL, Wang GY, Dang XW. CD47 blockade alleviates acute rejection of allogeneic mouse liver transplantation by reducing ischemia/reperfusion injury. Biomed Pharmacother 2019; 123:109793. [PMID: 31884341 DOI: 10.1016/j.biopha.2019.109793] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Despite advances in immunosuppressive therapies, acute rejection response is still a serious concern especially in the early phase after liver transplantation. This study aimed to evaluate whether blocking the TSP1-CD47 signaling pathway could attenuate the acute rejection after liver transplantation. An allogeneic mouse orthotopic liver transplantation model (Balb/c→C3H) with prolonged cold ischemic phase was used to induce severe IRI and lethal acute rejection. CD47mAb or isotype matched-control IgG2a was administered to donor liver during graft perfusion. Recipients were sacrificed at 1d, 3d, 5d and 7d after reperfusion. Blood samples were collected to evaluate serum alanine aminotransferase, total bilirubin, HMGB-1,TNF-α, IL-2 and INF-γ level. Flow cytometric analysis was used to detect the strength of innate and adaptive immune response. Liver tissue was obtained for HE, TUNEL staining and F4/80 immumohistochemical staining. Moreover, we conducted a mixed lymphocyte reaction treated with IgG2a or CD47mAb. Mice in CD47mAb-treated group demonstrated improved survival and significantly lower increase in Suzuki score, apoptosis index, acute rejection index, serum alanine aminotransferase, total bilirubin, HMGB-1, TNF-α, IL-2, INF-γ level and the degree of Kupffer cells' activation especially in the early phase of acute rejection. In addition, Pearson's correlation analysis confirmed significant correlation between Suzuki score/ALT and acute rejection index. The in vitro inhibition assay showed that CD47 blockade couldn't directly inhibit recipient lymphocyte proliferation. Based on the evidence that TSP1-CD47 signaling blockade with CD47mAb could alleviate acute rejection by reducing the extent of IRI after liver transplantation indirectly, this study provided a basis for new interventions and management methods to support better transplant outcomes.
Collapse
Affiliation(s)
- Ding-Yang Li
- Department of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China
| | - Shu-Li Xie
- Department of Hepatobiliary& Pancreatic Surgery, The First Norman Bethune Hospital Affiliated to Jilin University, Changchun 130021, Jilin Province, China
| | - Guang-Yi Wang
- Department of Hepatobiliary& Pancreatic Surgery, The First Norman Bethune Hospital Affiliated to Jilin University, Changchun 130021, Jilin Province, China
| | - Xiao-Wei Dang
- Department of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan Province, China.
| |
Collapse
|
5
|
Zischke J, Mamareli P, Pokoyski C, Gabaev I, Buyny S, Jacobs R, Falk CS, Lochner M, Sparwasser T, Schulz TF, Kay-Fedorov PC. The human cytomegalovirus glycoprotein pUL11 acts via CD45 to induce T cell IL-10 secretion. PLoS Pathog 2017. [PMID: 28628650 PMCID: PMC5491327 DOI: 10.1371/journal.ppat.1006454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human Cytomegalovirus (HCMV) is a widespread pathogen, infection with which can cause severe disease for immunocompromised individuals. The complex changes wrought on the host's immune system during both productive and latent HCMV infection are well known. Infected cells are masked and manipulated and uninfected immune cells are also affected; peripheral blood mononuclear cell (PBMC) proliferation is reduced and cytokine profiles altered. Levels increase of the anti-inflammatory cytokine IL-10, which may be important for the establishment of HCMV infections and is required for the development of high viral titres by murine cytomegalovirus. The mechanisms by which HCMV affects T cell IL-10 secretion are not understood. We show here that treatment of PBMC with purified pUL11 induces IL-10 producing T cells as a result of pUL11 binding to the CD45 phosphatase on T cells. IL-10 production induced by HCMV infection is also in part mediated by pUL11. Supernatants from pUL11 treated cells have anti-inflammatory effects on untreated PBMC. Considering the mechanism, CD45 can be a positive or negative regulator of TCR signalling, depending on its expression level, and we show that pUL11 also has concentration dependent activating or inhibitory effects on T cell proliferation and on the kinase function of the CD45 substrate Lck. pUL11 is therefore the first example of a viral protein that can target CD45 to induce T cells with anti-inflammatory properties. It is also the first HCMV protein shown to induce T cell IL-10 secretion. Understanding the mechanisms by which pUL11-induced changes in signal strength influence T cell development and function may provide the basis for the development of novel antiviral treatments and therapies against immune pathologies.
Collapse
Affiliation(s)
- Jasmin Zischke
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Panagiota Mamareli
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Claudia Pokoyski
- Department of General, Visceral and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Ildar Gabaev
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sabine Buyny
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Roland Jacobs
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Christine S. Falk
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Thomas F. Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
| | - Penelope C. Kay-Fedorov
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF, TTU-IICH), Hannover-Braunschweig Site, Hannover, Germany
- * E-mail:
| |
Collapse
|
6
|
Picarda E, Bézie S, Boucault L, Autrusseau E, Kilens S, Meistermann D, Martinet B, Daguin V, Donnart A, Charpentier E, David L, Anegon I, Guillonneau C. Transient antibody targeting of CD45RC induces transplant tolerance and potent antigen-specific regulatory T cells. JCI Insight 2017; 2:e90088. [PMID: 28194440 DOI: 10.1172/jci.insight.90088] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Rat and human CD4+ and CD8+ Tregs expressing low levels of CD45RC have strong immunoregulatory properties. We describe here that human CD45 isoforms are nonredundant and identify distinct subsets of cells. We show that CD45RC is not expressed by CD4+ and CD8+ Foxp3+ Tregs, while CD45RA/RB/RO are. Transient administration of a monoclonal antibody (mAb) targeting CD45RC in a rat cardiac allotransplantation model induced transplant tolerance associated with inhibition of allogeneic humoral responses but maintained primary and memory responses against cognate antigens. Anti-CD45RC mAb induced rapid death of CD45RChigh T cells through intrinsic cell signaling but preserved and potentiated CD4+ and CD8+ CD45RClow/- Tregs, which are able to adoptively transfer donor-specific tolerance to grafted recipients. Anti-CD45RC treatment results in distinct transcriptional signature of CD4+ and CD8+ CD45RClow/- Tregs. Finally, we demonstrate that anti-human CD45RC treatment inhibited graft-versus-host disease (GVHD) in immune-humanized NSG mice. Thus, short-term anti-CD45RC is a potent therapeutic candidate to induce transplantation tolerance in human.
Collapse
Affiliation(s)
- Elodie Picarda
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Laetitia Boucault
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Elodie Autrusseau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Stéphanie Kilens
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Dimitri Meistermann
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Bernard Martinet
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Véronique Daguin
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Audrey Donnart
- INSERM UMR1087, CNRS UMR6291, Université de Nantes, l'institut du thorax, Nantes, France
| | - Eric Charpentier
- INSERM UMR1087, CNRS UMR6291, Université de Nantes, l'institut du thorax, Nantes, France
| | - Laurent David
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
7
|
Dun H, Song L, Ma A, Hu Y, Zeng L, Bai J, Zhang G, Zhang L, Koide K, Okada Y, Hanaoka K, Yamamoto R, Hirose J, Morokata T, Daloze P, Chen H. ASP0028 in combination with suboptimal-dose of tacrolimus in Cynomolgus monkey renal transplantation model. Transpl Immunol 2017; 40:57-65. [PMID: 28077266 DOI: 10.1016/j.trim.2017.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/06/2017] [Indexed: 12/19/2022]
Abstract
FTY720, a S1P-receptor modulator, has shown to be effective in several transplant and autoimmune disease models, via modulating lymphocyte homing into secondary lymphoid organs (SLOs), and thereby reducing these cells in peripheral blood. ASP0028, a newly developed S1P1/S1P5-selective agonist, presented comparable efficacy to FTY720 and wider safety margins than FTY720. In this study, we assessed the efficacy and safety of ASP0028 co-administered with suboptimal-dose of tacrolimus in the Cynomolgus monkey renal transplantation model. Seven animals in group-1 or group-2 received mono-tacrolimus 1.0mg/kg once a day (QD), or ASP0028 0.6mg/kg plus tacrolimus 1.0mg/kg QD, respectively. Eight animals in group-3 received ASP0028 1.2mg/kg plus tacrolimus 1.0mg/kg QD. The allograft median survival time (MST) in group-2 and group-3 were significantly extended to 41 and 61.5days, versus that of 28days in group-1 (p=0.036 and 0.001, respectively). ASP0028 administration remarkably reduced absolute numbers of peripheral lymphocytes, particularly subsets of CD4+/ or CD8+/naive and central memory cells, CD4+/Treg cells, and to a lesser extent on B cells, but not CD4+/ or CD8+/effector memory cells and NK cells. These data show ASP0028 combined with suboptimal-dose of tacrolimus effectively prolongs renal allograft survival in nonhuman primates (NHPs) with well tolerated safety, supporting its further investigation to optimize CNI-sparing regimens.
Collapse
Affiliation(s)
- Hao Dun
- Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Canada
| | - Lijun Song
- Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Canada
| | - Anlun Ma
- Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Canada
| | - Yanxin Hu
- Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Canada
| | - Lin Zeng
- Laboratory Animals Center, the Academy of Military Medical Sciences, Beijing, China
| | - Jieying Bai
- Laboratory Animals Center, the Academy of Military Medical Sciences, Beijing, China
| | - Guangzhou Zhang
- Laboratory Animals Center, the Academy of Military Medical Sciences, Beijing, China
| | - Liangyan Zhang
- Laboratory Animals Center, the Academy of Military Medical Sciences, Beijing, China
| | - Kumi Koide
- Drug Discovery Research, Astellas Pharma Inc., Japan
| | - Yohei Okada
- Drug Discovery Research, Astellas Pharma Inc., Japan
| | - Kaori Hanaoka
- Drug Discovery Research, Astellas Pharma Inc., Japan
| | - Rie Yamamoto
- Drug Discovery Research, Astellas Pharma Inc., Japan
| | - Jun Hirose
- Drug Discovery Research, Astellas Pharma Inc., Japan
| | | | - Pierre Daloze
- Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Canada
| | - Huifang Chen
- Department of Surgery, Research Center, CHUM, Notre-Dame Hospital, University of Montreal, Montreal, Canada.
| |
Collapse
|
8
|
Krummey SM, Martinez RJ, Andargachew R, Liu D, Wagener M, Kohlmeier JE, Evavold BD, Larsen CP, Ford ML. Low-Affinity Memory CD8+ T Cells Mediate Robust Heterologous Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:2838-46. [PMID: 26864034 DOI: 10.4049/jimmunol.1500639] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 01/12/2016] [Indexed: 12/30/2022]
Abstract
Heterologous immunity is recognized as a significant barrier to transplant tolerance. Whereas it has been established that pathogen-elicited memory T cells can have high or low affinity for cross-reactive allogeneic peptide-MHC, the role of TCR affinity during heterologous immunity has not been explored. We established a model with which to investigate the impact of TCR-priming affinity on memory T cell populations following a graft rechallenge. In contrast to high-affinity priming, low-affinity priming elicited fully differentiated memory T cells with a CD45RB(hi) status. High CD45RB status enabled robust secondary responses in vivo, as demonstrated by faster graft rejection kinetics and greater proliferative responses. CD45RB blockade prolonged graft survival in low affinity-primed mice, but not in high affinity-primed mice. Mechanistically, low affinity-primed memory CD8(+) T cells produced more IL-2 and significantly upregulated IL-2Rα expression during rechallenge. We found that CD45RB(hi) status was also a stable marker of priming affinity within polyclonal CD8(+) T cell populations. Following high-affinity rechallenge, low affinity-primed CD45RB(hi) cells became CD45RB(lo), demonstrating that CD45RB status acts as an affinity-based differentiation switch on CD8(+) T cells. Thus, these data establish a novel mechanism by which CD45 isoforms tune low affinity-primed memory CD8(+) T cells to become potent secondary effectors following heterologous rechallenge. These findings have direct implications for allogeneic heterologous immunity by demonstrating that despite a lower precursor frequency, low-affinity priming is sufficient to generate memory cells that mediate potent secondary responses against a cross-reactive graft challenge.
Collapse
Affiliation(s)
| | - Ryan J Martinez
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Rakieb Andargachew
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Danya Liu
- Emory Transplant Center, Atlanta, GA 30322; and
| | | | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | - Brian D Evavold
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322
| | | | | |
Collapse
|
9
|
Viral interference with functions of the cellular receptor tyrosine phosphatase CD45. Viruses 2015; 7:1540-57. [PMID: 25807057 PMCID: PMC4379584 DOI: 10.3390/v7031540] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/24/2022] Open
Abstract
The receptor tyrosine phosphatase CD45 is expressed on the surface of almost all cells of hematopoietic origin. CD45 functions are central to the development of T cells and determine the threshold at which T and B lymphocytes can become activated. Given this pivotal role of CD45 in the immune system, it is probably not surprising that viruses interfere with the activity of CD45 in lymphocytes to dampen the immune response and that they also utilize this molecule to accomplish their replication cycle. Here we report what is known about the interaction of viral proteins with CD45. Moreover, we debate putative interactions of viruses with CD45 in myeloid cells and the resulting consequences-subjects that remain to be investigated. Finally, we summarize the evidence that pathogens were the driving force for the evolution of CD45.
Collapse
|
10
|
Abstract
Large animal models have long served as the proving grounds for advances in transplantation, bridging the gap between inbred mouse experimentation and human clinical trials. Although a variety of species have been and continue to be used, the emergence of highly targeted biologic- and antibody-based therapies has required models to have a high degree of homology with humans. Thus, the nonhuman primate has become the model of choice in many settings. This article will provide an overview of nonhuman primate models of transplantation. Issues of primate genetics and care will be introduced, and a brief overview of technical aspects for various transplant models will be discussed. Finally, several prominent immunosuppressive and tolerance strategies used in primates will be reviewed.
Collapse
Affiliation(s)
- Douglas J Anderson
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia 30322
| | | |
Collapse
|
11
|
Chen Song S, Zhong S, Xiang Y, Li JH, Guo H, Wang WY, Xiong YL, Li XC, Chen Shi S, Chen XP, Chen G. Complement inhibition enables renal allograft accommodation and long-term engraftment in presensitized nonhuman primates. Am J Transplant 2011; 11:2057-66. [PMID: 21831160 DOI: 10.1111/j.1600-6143.2011.03646.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Protection against humoral injury mediated by donor-specific antibodies (DSA), also known as accommodation, may allow for long-term allograft survival in presensitized recipients. In the present study, we determined the role of complement in renal allograft accommodation in donor skin-presensitized nonhuman primates under conventional immunosuppression. Donor skin allografts were transplanted to presensitized recipients 14 days prior to renal transplantation. Renal allografts not receiving any immunosuppressive treatment developed accelerated rejection with predominantly humoral injury, which was not prevented using conventional cyclosporine (CsA) triple therapy. Inhibition of complement activation with the Yunnan-cobra venom factor (Y-CVF) successfully prevented accelerated antibody-mediated rejection and resulted in successful accommodation and long-term renal allograft survival in most presensitized recipients. Accommodation in this model was associated with the prevention of the early antibody responses induced against donor antigens by complement inhibition. Some antiapoptotic proteins and complement regulatory proteins, including Bcl-2, CD59, CD46 and clusterin, were upregulated in the surviving renal allografts. These results suggest that the complement inhibition-based strategy may be valuable alternative in future clinical cross-match positive or ABO-incompatible transplantation.
Collapse
Affiliation(s)
- S Chen Song
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
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: 1.1] [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
|
13
|
Azimzadeh AM, Lees JR, Ding Y, Bromberg JS. Immunobiology of transplantation: impact on targets for large and small molecules. Clin Pharmacol Ther 2011; 90:229-42. [PMID: 21716276 DOI: 10.1038/clpt.2011.106] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Organ transplantation is the preferred method of treatment for many forms of end-stage organ failure. However, immunosuppressive drugs that are used to avoid rejection can result in numerous undesirable effects (infection, malignancy, hypertension, diabetes, and accelerated arteriosclerosis). Moreover, they are not effective at preventing chronic rejection resulting in late graft loss. This review summarizes the fundamental concepts underlying the rejection of solid-organ allografts with the aim of highlighting potential new targets for therapeutics. Future improvement will depend on new therapeutic moieties, including biologics, to target various pathways of both the innate and adaptive arms of immunity. Results from some of the most recent clinical trials in transplantation and emerging new therapies are also discussed.
Collapse
Affiliation(s)
- A M Azimzadeh
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
14
|
Shin HM, Cho WD, Lee GK, Lee SH, Lee KM, Ji GY, Yoon SS, Koo JH, Lee HC, Lee KH, Song HG. Characterization of Monoclonal Antibodies against Human Leukocyte Common Antigen (CD45). Immune Netw 2011; 11:114-22. [PMID: 21637389 PMCID: PMC3100522 DOI: 10.4110/in.2011.11.2.114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 03/31/2011] [Accepted: 04/05/2011] [Indexed: 12/01/2022] Open
Abstract
Background The leukocyte common antigen (CD45) is a transmembrane-type protein tyrosine phosphatase that has five isoforms. Methods We generated seven murine mAbs against human CD45 by injecting cells from different origins, such as human thymocytes, PBMCs, and leukemic cell lines. By using various immunological methods including flow cytometry, immunohistochemistry, and immunoprecipitation, we evaluated the reactivity of those mAbs to CD45 of thymus as well as tonsil lysates. Furthermore, we transiently transfected COS-7 cells with each of gene constructs that express five human CD45 isoforms respectively, and examined the specificities of the mAbs against the transfected isoforms. Results In case of thymocytes, lymphocytes, and monocytes, all the seven mAbs demonstrated positive reactivities whereas none was reactive to erythrocytes and platelets. The majority of immune cells in formalin-fixed paraffin-embedded thymus and tonsil tissues displayed strong membranous immunoreactivity, and the main antigen was detected near 220 kDa in all cases. Among the mAbs, four mAbs (AP4, DN11, SHL-1, and P6) recognized a region commonly present in all the five isoforms. One mAb, YG27, recognized four isoforms (ABC, AB, BC, and O). Two mAbs, P1 and P14, recognized the isoforms that contain exon A encoded regions (ABC and AB). Conclusion In this study, we confirmed that AP4, DN11, SHL-1, YG27 and P6, are mAbs reactive with the CD45 antigen whereas P1 and P14 are reactive with the CD45RA antigen.
Collapse
Affiliation(s)
- Hyang-Mi Shin
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju 361-763, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kawai T, Benedict Cosimi A. Induction of tolerance in clinical kidney transplantation. Clin Transplant 2010; 24 Suppl 22:2-5. [PMID: 20590685 DOI: 10.1111/j.1399-0012.2010.01268.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Induction of donor-specific tolerance has been an ultimate goal in organ transplantation. Although numerous regimens for the induction of allograft tolerance have been developed in rodents, their application to primates has been limited. The approaches that have been successfully applied in primates can be divided into (i) use of total lymphoid irradiation, (ii) costimulatory blockade, (iii) profound depletion of recipient T cells, (iv) infusion of regulatory cells and (v) donor bone marrow (DBM) infusion/transplantation. Among these approaches, successful allograft tolerance has been achieved in clinical kidney transplantation using DBM transplantation.
Collapse
Affiliation(s)
- Tatsuo Kawai
- Massachusetts General Hospital, Transplant Center, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
16
|
Abstract
T-cell depletion strategies are an efficient therapy for the treatment of acute rejection after organ transplantation and have been successfully used as induction regimens. Although eliminating whole T cells blocks alloreactivity, this therapy challenges the development of regulatory mechanisms because it depletes regulatory cells and modifies the profile of T cells after homeostatic repopulation. Targeting T-cell subpopulations or selectively activated T cells, without modifying Treg cells, could constitute a pro-tolerogenic approach. However, the perfect molecular target that would be totally specific probably still needs to be identified. In this study, we have reviewed the biological activities of broad or specific T-cell depletion strategies as these contribute to the induction of regulatory cells and tolerance in organ transplantation.
Collapse
Affiliation(s)
- Thomas Haudebourg
- INSERM, U643, CHU Nantes, Institut de Transplantation et de Recherche en Transplantation, ITERT, Université de Nantes, Faculté de Médecine, Nantes, France
| | | | | |
Collapse
|
17
|
Extracellular ligation-dependent CD45RB enzymatic activity negatively regulates lipid raft signal transduction. Blood 2008; 113:594-603. [PMID: 18840711 DOI: 10.1182/blood-2008-04-150987] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD45 is the most prominent membrane protein on lymphocytes. The function and regulation of this protein tyrosine phosphatase remain largely obscure, mainly because of the lack of a known ligand, and it still remains unknown whether such tyrosine phosphatases are subject to extracellular control at all. We report that an anti-CD45RB antibody (Ab) that prevents rejection and induces tolerance activates CD45RB tyrosine phosphatase enzymatic activity in T lymphocytes, allowing us to directly monitor the effects of increased CD45RB activity on signal transduction. Using both kinase substrate peptide arrays as well as conventional biochemistry, we also provide evidence of the various kinases involved in bringing about the inhibitory effect of this Ab on CD3-induced T-cell receptor signaling. Furthermore, we report that activated CD45RB translocates to lipid rafts and interferes with lipid raft localization and activation state of CD45 substrate Lck. Thus, these findings indeed prove that CD45 is subject to extracellular control and also define a novel mechanism by which receptor tyrosine phosphatases control lymphocyte biology and provide further insight into the intracellular signaling pathways effected by anti-CD45RB monoclonal Ab treatment.
Collapse
|
18
|
Javeed A, Zhao Y. The effects of immunosuppression on regulatory CD4(+)CD25(+) T cells: impact on immunosuppression selection in transplantation. Mol Diagn Ther 2008; 12:171-81. [PMID: 18510380 DOI: 10.1007/bf03256281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
During immune response and T-cell activation, both effector T cells and regulatory T(T(reg)) cells are activated and regulated simultaneously by both positive and negative pathways. CD4(+)CD25(+) T(reg) cells play a critical role in immune tolerance to self antigens as well as to allografts in some transplant settings. Effective immunosuppressive regimens significantly reduced the incidence of acute allograft rejection in patients following organ transplantation. However, the impact of immunosuppressive treatment on the potential induction of transplant tolerance has not been well determined. In this review we summarize the effects of immunosuppressive reagents on CD4(+)CD25(+) T(reg) cells in order to bring attention to this issue, which may affect the choice of immunosuppressive regimen in the clinical setting.
Collapse
Affiliation(s)
- Aqeel Javeed
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
19
|
Donor bone marrow transplantation as an approach to tolerance induction for clinical kidney transplantation. Curr Opin Organ Transplant 2007. [DOI: 10.1097/mot.0b013e3282f1fc01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Rathanaswami P, Babcook J, Gallo M. High-affinity binding measurements of antibodies to cell-surface-expressed antigens. Anal Biochem 2007; 373:52-60. [PMID: 17910940 DOI: 10.1016/j.ab.2007.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2007] [Revised: 08/10/2007] [Accepted: 08/13/2007] [Indexed: 10/22/2022]
Abstract
A simple method that allows affinity measurements of antibodies to integral membrane proteins is described. Kinetic Exclusion Assay was used to determine the concentration of free antibody that remains in solution after equilibrium has been established between the antibody and the cell-surface-expressed antigen, from which the equilibrium dissociation constant (Kd) was determined. It eliminates the requirement for soluble antigen and modifications such as radio-labeling or fluorescent labeling of the antibody. For one of the cell-surface-expressed antigens, it was determined that the affinity of the antibody to the cell-surface-expressed antigen was similar to that of the purified, soluble form of the antigen. In addition to the simplicity of the approach, the method provides a true measure of the affinity/avidity of the antibody to the native form of cell-surface-expressed targets, including antigens that cannot be produced in soluble forms, and to unknown cell surface antigens.
Collapse
|