1
|
Kouyoumdjian A, Tchervenkov J, Paraskevas S. TFNR2 in Ischemia-Reperfusion Injury, Rejection, and Tolerance in Transplantation. Front Immunol 2022; 13:903913. [PMID: 35874723 PMCID: PMC9300818 DOI: 10.3389/fimmu.2022.903913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022] Open
Abstract
Tumor necrosis factor receptor 2 (TNFR2) has been shown to play a crucial role in CD4+ T regulatory cells (CD4+Tregs) expansion and suppressive function. Increasing evidence has also demonstrated its role in a variety of immune regulatory cell subtypes such as CD8+ T regulatory cells (CD8+ Tregs), B regulatory cells (Bregs), and myeloid-derived suppressor cells (MDSCs). In solid organ transplantation, regulatory immune cells have been associated with decreased ischemia-reperfusion injury (IRI), improved graft survival, and improved overall outcomes. However, despite TNFR2 being studied in the context of autoimmune diseases, cancer, and hematopoietic stem cell transplantation, there remains paucity of data in the context of solid organ transplantation and islet cell transplantation. Interestingly, TNFR2 signaling has found a clinical application in islet transplantation which could guide its wider use. This article reviews the current literature on TNFR2 expression in immune modulatory cells as well as IRI, cell, and solid organ transplantation. Our results highlighted the positive impact of TNFR2 signaling especially in kidney and islet transplantation. However, further investigation of TNFR2 in all types of solid organ transplantation are required as well as dedicated studies on its therapeutic use during induction therapy or treatment of rejection.
Collapse
Affiliation(s)
- Araz Kouyoumdjian
- Division of Experimental Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of General Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- *Correspondence: Araz Kouyoumdjian,
| | - Jean Tchervenkov
- Division of Experimental Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of General Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
| | - Steven Paraskevas
- Division of Experimental Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Division of General Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Zimmerer JM, Ringwald BA, Chaudhari SR, Han J, Peterson CM, Warren RT, Hart MM, Abdel-Rasoul M, Bumgardner GL. Invariant NKT Cells Promote the Development of Highly Cytotoxic Multipotent CXCR3 +CCR4 +CD8 + T Cells That Mediate Rapid Hepatocyte Allograft Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:3107-3121. [PMID: 34810223 PMCID: PMC9124232 DOI: 10.4049/jimmunol.2100334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/13/2021] [Indexed: 12/22/2022]
Abstract
Hepatocyte transplant represents a treatment for metabolic disorders but is limited by immunogenicity. Our prior work identified the critical role of CD8+ T cells, with or without CD4+ T cell help, in mediating hepatocyte rejection. In this study, we evaluated the influence of invariant NKT (iNKT) cells, uniquely abundant in the liver, upon CD8-mediated immune responses in the presence and absence of CD4+ T cells. To investigate this, C57BL/6 (wild-type) and iNKT-deficient Jα18 knockout mice (cohorts CD4 depleted) were transplanted with allogeneic hepatocytes. Recipients were evaluated for alloprimed CD8+ T cell subset composition, allocytotoxicity, and hepatocyte rejection. We found that CD8-mediated allocytotoxicity was significantly decreased in iNKT-deficient recipients and was restored by adoptive transfer of iNKT cells. In the absence of both iNKT cells and CD4+ T cells, CD8-mediated allocytotoxicity and hepatocyte rejection was abrogated. iNKT cells enhance the proportion of a novel subset of multipotent, alloprimed CXCR3+CCR4+CD8+ cytolytic T cells that develop after hepatocyte transplant and are abundant in the liver. Alloprimed CXCR3+CCR4+CD8+ T cells express cytotoxic effector molecules (perforin/granzyme and Fas ligand) and are distinguished from alloprimed CXCR3+CCR4-CD8+ T cells by a higher proportion of cells expressing TNF-α and IFN-γ. Furthermore, alloprimed CXCR3+CCR4+CD8+ T cells mediate higher allocytotoxicity and more rapid allograft rejection. Our data demonstrate the important role of iNKT cells in promoting the development of highly cytotoxic, multipotent CXCR3+CCR4+CD8+ T cells that mediate rapid rejection of allogeneic hepatocytes engrafted in the liver. Targeting iNKT cells may be an efficacious therapy to prevent rejection of intrahepatic cellular transplants.
Collapse
Affiliation(s)
- Jason M Zimmerer
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Bryce A Ringwald
- Medical Student Research Program, The Ohio State University College of Medicine, Columbus, OH
| | - Sachi R Chaudhari
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Jing Han
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH; and
| | - Chelsea M Peterson
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Robert T Warren
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | - Madison M Hart
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH
| | | | - Ginny L Bumgardner
- Comprehensive Transplant Center, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH;
| |
Collapse
|
3
|
Carrión B, Liu Y, Hadi M, Lundstrom J, Christensen JR, Ammitzbøll C, Dziegiel MH, Sørensen PS, Comabella M, Montalban X, Sellebjerg F, Issazadeh-Navikas S. Transcriptome and Function of Novel Immunosuppressive Autoreactive Invariant Natural Killer T Cells That Are Absent in Progressive Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1065. [PMID: 34385365 PMCID: PMC8362604 DOI: 10.1212/nxi.0000000000001065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/16/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE The aim of this study was to determine whether natural killer T (NKT) cells, including invariant (i) NKT cells, have clinical value in preventing the progression of multiple sclerosis (MS) by examining the mechanisms by which a distinct self-peptide induces a novel, protective invariant natural killer T cell (iNKT cell) subset. METHODS We performed a transcriptomic and functional analysis of iNKT cells that were reactive to a human collagen type II self-peptide, hCII707-721, measuring differentially induced genes, cytokines, and suppressive capacity. RESULTS We report the first transcriptomic profile of human conventional vs novel hCII707-721-reactive iNKT cells. We determined that hCII707-721 induces protective iNKT cells that are found in the blood of healthy individuals but not progressive patients with MS (PMS). By transcriptomic analysis, we observed that hCII707-721 promotes their development and proliferation, favoring the splicing of full-length AKT serine/threonine kinase 1 (AKT1) and effector function of this unique lineage by upregulating tumor necrosis factor (TNF)-related genes. Furthermore, hCII707-721-reactive iNKT cells did not upregulate interferon (IFN)-γ, interleukin (IL)-4, IL-10, IL-13, or IL-17 by RNA-seq or at the protein level, unlike the response to the glycolipid alpha-galactosylceramide. hCII707-721-reactive iNKT cells increased TNFα only at the protein level and suppressed autologous-activated T cells through FAS-FAS ligand (FAS-FASL) and TNFα-TNF receptor I signaling but not TNF receptor II. DISCUSSION Based on their immunomodulatory properties, NKT cells have a potential value in the treatment of autoimmune diseases, such as MS. These significant findings suggest that endogenous peptide ligands can be used to expand iNKT cells, without causing a cytokine storm, constituting a potential immunotherapy for autoimmune conditions, including PMS.
Collapse
Affiliation(s)
- Belinda Carrión
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Yawei Liu
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Mahdieh Hadi
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Jon Lundstrom
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Jeppe Romme Christensen
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Cecilie Ammitzbøll
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Morten Hanefeld Dziegiel
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Per Soelberg Sørensen
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Manuel Comabella
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Xavier Montalban
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Finn Sellebjerg
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark
| | - Shohreh Issazadeh-Navikas
- From the Biotech Research and Innovation Centre (BRIC) (B.C., Y.L., M.H., J.L., S.I.-N.), University of Copenhagen; Danish Multiple Sclerosis Center (J.R.C., C.A., P.S.S.), University of Copenhagen and Department of Neurology, Rigshospitalet; Blood Bank (M.H.D.), Copenhagen University Hospital, Denmark; Centre d'Esclerosi Múltiple de Catalunya (M.C.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Barcelona, Spain; and Centre d'Esclerosi Múltiple de Catalunya (X.M.), Cemcat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d´Hebron (HUVH) - Universitat Autònoma de Barcelona, Spain; Danish Multiple Sclerosis Center, University of Copenhagen and Department of Neurology, Rigshospitalet, Denmark.
| |
Collapse
|
4
|
Eid A, Issa Y, Mohamed A, Badran F. Interleukin-9 and soluble tumor necrosis factor-like weak inducer of apoptosis in serum and suction blister fluid of nonsegmental vitiligo patients: Relation to disease severity. DERMATOL SIN 2021. [DOI: 10.4103/ds.ds_44_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
5
|
Bansal R, Reshef R. Revving the CAR - Combination strategies to enhance CAR T cell effectiveness. Blood Rev 2021; 45:100695. [PMID: 32402724 DOI: 10.1016/j.blre.2020.100695] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/22/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is currently approved for treatment of refractory B-cell malignancies. Response rates in these diseases are impressive by historical standards, but most patients do not have a durable response and there remains room for improvement. To date, CAR T cell activity has been even more limited in solid malignancies. These limitations are thought to be due to several pathways of resistance to CAR T cells, including cell-intrinsic mechanisms and the immunosuppressive tumor microenvironment. In this review, we discuss current experimental strategies that combine small molecules and monoclonal antibodies with CAR T cells to overcome these resistance mechanisms. We describe the biological rationale, pre-clinical data and clinical trials in progress that test the efficacy and safety of these combinations.
Collapse
Affiliation(s)
- Rajat Bansal
- Division of Hematology/Oncology, Columbia University Irving Medical Center, 177 Ft. Washington Ave, Floor: 6GN-435, New York, NY 10032, USA.
| | - Ran Reshef
- Division of Hematology/Oncology, Columbia University Irving Medical Center, 630 W. 168(th) Street Mailbox 127, New York, NY 10032, USA.
| |
Collapse
|
6
|
Zimmerer JM, Liu XL, Blaszczak A, Avila CL, Pham TA, Warren RT, Bumgardner GL. Critical Role of Macrophage FcγR Signaling and Reactive Oxygen Species in Alloantibody-Mediated Hepatocyte Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:3731-3740. [PMID: 30397035 PMCID: PMC6289737 DOI: 10.4049/jimmunol.1800333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/03/2018] [Indexed: 12/24/2022]
Abstract
Humoral alloimmunity negatively impacts both short- and long-term cell and solid organ transplant survival. We previously reported that alloantibody-mediated rejection of transplanted hepatocytes is critically dependent on host macrophages. However, the effector mechanism(s) of macrophage-mediated injury to allogeneic liver parenchymal cells is not known. We hypothesized that macrophage-mediated destruction of allogeneic hepatocytes occurs by cell-cell interactions requiring FcγRs. To examine this, alloantibody-dependent hepatocyte rejection in CD8-depleted wild-type (WT) and Fcγ-chain knockout (KO; lacking all functional FcγR) transplant recipients was evaluated. Alloantibody-mediated hepatocellular allograft rejection was abrogated in recipients lacking FcγR compared with WT recipients. We also investigated anti-FcγRI mAb, anti-FcγRIII mAb, and inhibitors of intracellular signaling (to block phagocytosis, cytokines, and reactive oxygen species [ROS]) in an in vitro alloantibody-dependent, macrophage-mediated hepatocytoxicity assay. Results showed that in vitro alloantibody-dependent, macrophage-mediated hepatocytotoxicity was critically dependent on FcγRs and ROS. The adoptive transfer of WT macrophages into CD8-depleted FcγR-deficient recipients was sufficient to induce alloantibody-mediated rejection, whereas adoptive transfer of macrophages from Fcγ-chain KO mice or ROS-deficient (p47 KO) macrophages was not. These results provide the first evidence, to our knowledge, that alloantibody-dependent hepatocellular allograft rejection is mediated by host macrophages through FcγR signaling and ROS cytotoxic effector mechanisms. These results support the investigation of novel immunotherapeutic strategies targeting macrophages, FcγRs, and/or downstream molecules, including ROS, to inhibit humoral immune damage of transplanted hepatocytes and perhaps other cell and solid organ transplants.
Collapse
Affiliation(s)
- Jason M Zimmerer
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Xin L Liu
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Alecia Blaszczak
- Medical Scientist Training Program, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Christina L Avila
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Thomas A Pham
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Robert T Warren
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Ginny L Bumgardner
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| |
Collapse
|
7
|
He R, Li Y, Zhou L, Su X, Li Y, Pan P, Hu C. miR-146b overexpression ameliorates lipopolysaccharide-induced acute lung injury in vivo and in vitro. J Cell Biochem 2018; 120:2929-2939. [PMID: 30500983 DOI: 10.1002/jcb.26846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a type of acute lung injury (ALI), which causes high morbidity and mortality. So far, effective clinical treatment of ARDS is still limited. Recently, miR-146b has been reported to play a key role in inflammation. In the present study, we evaluated the functional role of miR-146b in ARDS using the murine model of lipopolysaccharide (LPS)-induced ALI. The miR-146b expression could be induced by LPS stimulation, and miR-146b overexpression was required in the maintenance of body weight and survival of ALI mice; after miR-146b overexpression, LPS-induced lung injury, pulmonary inflammation, total cell and neutrophil counts, proinflammatory cytokines, and chemokines in bronchial alveolar lavage (BAL) fluid were significantly reduced. The promotive effect of LPS on lung permeability through increasing total protein, albumin and IgM in BAL fluid could be partially reversed by miR-146b overexpression. Moreover, in murine alveolar macrophages, miR-146b overexpression reduced LPS-induced TNF-α and interleukin (IL)-1β releasing. Taken together, we demonstrated that miR-146b overexpression could effectively improve the LPS-induced ALI; miR-146b is a promising target in ARDS treatment.
Collapse
Affiliation(s)
- Ruoxi He
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Li Zhou
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoli Su
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
Shivakumar P, Mizuochi T, Mourya R, Gutta S, Yang L, Luo Z, Bezerra JA. Preferential TNF α signaling via TNFR2 regulates epithelial injury and duct obstruction in experimental biliary atresia. JCI Insight 2017; 2:e88747. [PMID: 28289704 DOI: 10.1172/jci.insight.88747] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Biliary atresia is an obstructive cholangiopathy of infancy that progresses to end-stage cirrhosis. Although the pathogenesis of the disease is not completely understood, previous reports link TNFα to apoptosis of the bile duct epithelium in the presence of IFNγ. Here, we investigate if TNFα signaling regulates pathogenic mechanisms of biliary atresia. First, we quantified the expression of TNFA and its receptors TNFR1 and TNFR2 in human livers and found an increased expression of the receptors at the time of diagnosis. In mechanistic experiments using a neonatal mouse model of rhesus rotavirus-induced (RRV-induced) biliary atresia, the expression of the ligand and both receptors increased 6- to 8-fold in hepatic DCs and NK lymphocytes above controls. The activation of tissue NK cells by RRV-primed DCs was independent of TNFα-TNFR signaling. Once activated, the expression of TNFα by NK cells induced lysis of 55% ± 2% of bile duct epithelial cells, which was completely prevented by blocking TNFα or TNFR2, but not TNFR1. More notably, antibody-mediated or genetic disruption of TNFα-TNFR2 signaling in vivo decreased apoptosis and epithelial injury; suppressed the infiltration of livers by T cells, DCs, and NK cells; prevented extrahepatic bile duct obstruction; and promoted long-term survival. These findings point to a key role for the TNFα/TNFR2 axis on pathogenesis of experimental biliary atresia and identify new therapeutic targets to suppress the disease phenotype.
Collapse
Affiliation(s)
- Pranavkumar Shivakumar
- Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tatsuki Mizuochi
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
| | - Reena Mourya
- Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sridevi Gutta
- Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Li Yang
- Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zhenhua Luo
- Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jorge A Bezerra
- Cincinnati Children's Hospital Medical Center and the Department of Pediatrics of the University Of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
9
|
Ko EJ, Lee YT, Kim KH, Lee Y, Jung YJ, Kim MC, Lee YN, Kang T, Kang SM. Roles of Aluminum Hydroxide and Monophosphoryl Lipid A Adjuvants in Overcoming CD4+ T Cell Deficiency To Induce Isotype-Switched IgG Antibody Responses and Protection by T-Dependent Influenza Vaccine. THE JOURNAL OF IMMUNOLOGY 2016; 198:279-291. [PMID: 27881702 DOI: 10.4049/jimmunol.1600173] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/24/2016] [Indexed: 01/09/2023]
Abstract
Vaccine adjuvant effects in the CD4-deficient condition largely remain unknown. We investigated the roles of combined monophosphoryl lipid A (MPL) and aluminum hydroxide (Alum) adjuvant (MPL+Alum) in inducing immunity after immunization of CD4 knockout (CD4KO) and wild-type (WT) mice with T-dependent influenza vaccine. MPL+Alum adjuvant mediated IgG isotype-switched Abs, IgG-secreting cell responses, and protection in CD4KO mice, which were comparable to those in WT mice. In contrast, Alum adjuvant effects were dependent on CD4+ T cells. MPL+Alum adjuvant was effective in recruiting monocytes and neutrophils as well as in protecting macrophages from Alum-mediated cell loss at the injection site in CD4KO mice. MPL+Alum appeared to attenuate MPL-induced inflammatory responses in WT mice, likely improving the safety. Additional studies in CD4-depleted WT mice and MHC class II KO mice suggest that MHC class II+ APCs contribute to providing alternative B cell help in the CD4-deficient condition in the context of MPL+Alum-adjuvanted vaccination.
Collapse
Affiliation(s)
- Eun-Ju Ko
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Young-Tae Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Youri Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Yu-Jin Jung
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Min-Chul Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and.,Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and.,Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea
| | - Taeuk Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| |
Collapse
|
10
|
Jiang LF, Chen O, Chu TG, Ding J, Yu Q. T Lymphocyte Subsets and Cytokines in Rats Transplanted with Adipose-Derived Mesenchymal Stem Cells and Acellular Nerve for Repairing the Nerve Defects. J Korean Neurosurg Soc 2015; 58:101-6. [PMID: 26361524 PMCID: PMC4564740 DOI: 10.3340/jkns.2015.58.2.101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/13/2015] [Accepted: 03/23/2015] [Indexed: 11/27/2022] Open
Abstract
Objective The aim of this study was to explore the immunity in rats transplanted with adipose-derived mesenchymal stem cells (ADSCs) and acellular nerve (ACN) for repairing sciatic nerve defects. Methods ADSCs were isolated from the adipose tissues of Wistar rats. Sprague-Dawley rats were used to establish a sciatic nerve defect model and then divided into four groups, according to the following methods : Group A, allogenic nerve graft; Group B, allograft with ACN; Group C, allograft ADSCs+ACN, and Group D, nerve autograft. Results At the day before transplantation and 3, 7, 14, and 28 days after transplantation, orbital venous blood of the Sprague-Dawley rats in each group was collected to detect the proportion of CD3+, CD4+, and CD8+ subsets using flow cytometry and to determine the serum concentration of interleukin-2 (IL-2), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) using enzyme-linked immunosorbent assay (ELISA). At each postoperative time point, the proportion of CD3+, CD4+, and CD8+ subsets and the serum concentration of IL-2, TNF-α, and IFN-γ in group C were all near to those in group B and group D, in which no statistically significant difference was observed. As compared with group A, the proportion of CD3+, CD4+, and CD8+ subsets and the serum concentration of IL-2, TNF-α, and IFN-γ were significantly reduced in group C (p<0.05). Conclusion The artificial nerve established with ADSCs and ACN has no obvious allograft rejection for repairing rat nerve defects.
Collapse
Affiliation(s)
- Liang-Fu Jiang
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Ou Chen
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang, China
| | - Ting-Gang Chu
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jian Ding
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qing Yu
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
11
|
TNF-α promoter single nucleotide polymorphisms and haplotypes associate with susceptibility of immune thrombocytopenia in Chinese adults. Hum Immunol 2014; 75:980-5. [PMID: 25158149 DOI: 10.1016/j.humimm.2014.08.197] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 06/12/2014] [Accepted: 08/14/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Tumor necrosis factor-alpha (TNF-α) participates as a candidate susceptibility factor for immune thrombocytopenia (ITP). This study attempted to investigate the association between five single nucleotide polymorphisms (SNPs) spanning the TNF-α promoter and the susceptibility of primary ITP in Chinese Han adults. METHODS In 215 adult primary ITP patients and 206 healthy controls, SNPs were detected by PCR-RFLP and PCR-SSP. The χ(2) test or fisher's exact test was used to compare frequencies of genotypes and alleles between patients and controls. Haplotypes were analyzed with the SHEsis online program. TNF-α, IFN-γ and Galectin-9 mRNA of 35 newly diagnosed adult ITP patients and 35 healthy controls were detected by qRT-PCR. RESULTS The haplotype GGC (-238G/-308G/-857C) of TNF-α promoter was significantly associated with a decreased susceptibility of primary ITP, especially in males. The relative levels of mRNA expression of TNF-α, IFN-γ and Gal-9 in adult active primary ITP patients was significantly up-regulated compared with patients in remission and controls. CONCLUSIONS This study represented the first report that the haplotype GGC of TNF-α was differentially associated with the susceptibility of primary ITP in Chinese Han adults. The up-regulation of TNF-α, IFN-γ and Galectin-9 was significantly correlated with active primary ITP in adult patients.
Collapse
|
12
|
Zimmerer J, Pham T, Wright C, Tobin K, Sanghavi P, Elzein S, Sanders V, Bumgardner G. Alloprimed CD8(+) T cells regulate alloantibody and eliminate alloprimed B cells through perforin- and FasL-dependent mechanisms. Am J Transplant 2014; 14:295-304. [PMID: 24472191 PMCID: PMC4018729 DOI: 10.1111/ajt.12565] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 10/07/2013] [Accepted: 10/27/2013] [Indexed: 01/25/2023]
Abstract
While it is well known that CD4(+) T cells and B cells collaborate for antibody production, our group previously reported that CD8(+) T cells down-regulate alloantibody responses following transplantation. However, the exact mechanism involved in CD8(+) T cell-mediated down-regulation of alloantibody remains unclear. We also reported that alloantibody production is enhanced when either perforin or FasL is deficient in transplant recipients. Here, we report that CD8(+) T cell-deficient transplant recipient mice (high alloantibody producers) exhibit an increased number of primed B cells compared to WT transplant recipients. Furthermore, CD8(+) T cells require FasL, perforin and allospecificity to down-regulate posttransplant alloantibody production. In vivo CD8-mediated clearance of alloprimed B cells was also FasL- and perforin-dependent. In vitro data demonstrated that recipient CD8(+) T cells directly induce apoptosis of alloprimed IgG1(+) B cells in co-culture in an allospecific and MHC class I-dependent fashion. Altogether these data are consistent with the interpretation that CD8(+) T cells down-regulate posttransplant alloantibody production by FasL- and perforin-dependent direct elimination of alloprimed IgG1(+) B cells.
Collapse
Affiliation(s)
- J.M. Zimmerer
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University, Columbus, OH
| | - T.A. Pham
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University, Columbus, OH
| | - C.L. Wright
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University, Columbus, OH
| | - K.J. Tobin
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University, Columbus, OH
| | - P.B. Sanghavi
- Medical Student Research Program, College of Medicine, The Ohio State University, Columbus, OH
| | - S.M. Elzein
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University, Columbus, OH
| | - V.M. Sanders
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Wexner Medical Center, Columbus, OH
| | - G.L. Bumgardner
- Department of Surgery, Comprehensive Transplant Center, The Ohio State University, Columbus, OH
| |
Collapse
|
13
|
Trebicka J, Krag A, Gansweid S, Schiedermaier P, Strunk HM, Fimmers R, Strassburg CP, Bendtsen F, Møller S, Sauerbruch T, Spengler U. Soluble TNF-alpha-receptors I are prognostic markers in TIPS-treated patients with cirrhosis and portal hypertension. PLoS One 2013; 8:e83341. [PMID: 24386183 PMCID: PMC3873322 DOI: 10.1371/journal.pone.0083341] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/02/2013] [Indexed: 12/14/2022] Open
Abstract
Background TNFα levels are increased in liver cirrhosis even in the absence of infection, most likely owing to a continuous endotoxin influx into the portal blood. Soluble TNFα receptors (sTNFR type I and II) reflect release of the short-lived TNFα, because they are cleaved from the cells after binding of TNFα. The aims were to investigate the circulating levels of soluble TNFR-I and -II in cirrhotic patients receiving TIPS. Methods Forty-nine patients with liver cirrhosis and portal hypertension (12 viral, 37 alcoholic) received TIPS for prevention of re-bleeding (n = 14), therapy-refractory ascites (n = 20), or both (n = 15). Portal and hepatic venous blood was drawn in these patients during the TIPS procedure and during the control catheterization two weeks later. sTNFR-I and sTNFR-II were measured by ELISA, correlated to clinical and biochemical characteristics. Results Before TIPS insertion, sTNFR-II levels were lower in portal venous blood than in the hepatic venous blood, as well as in portal venous blood after TIPS insertion. No significant differences were measured in sTNFR-I levels. Hepatic venous levels of sTNFR-I above 4.5 ng/mL (p = 0.036) and sTNFR-II above 7 ng/mL (p = 0.05) after TIPS insertion were associated with decreased survival. A multivariate Cox-regression survival analysis identified the hepatic venous levels of sTNFR-I (p = 0.004) two weeks after TIPS, and Child score (p = 0.002) as independent predictors of mortality, while MELD-score was not. Conclusion Hepatic venous levels of sTNFR-I after TIPS insertion may predict mortality in patients with severe portal hypertension.
Collapse
Affiliation(s)
- Jonel Trebicka
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Aleksander Krag
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Gastroenterology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Stefan Gansweid
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | | | | | - Rolf Fimmers
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | | | - Fleming Bendtsen
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Søren Møller
- Centre of Functional Imaging and Research, Department of Clinical Physiology and Nuclear Medicine, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Ulrich Spengler
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| |
Collapse
|
14
|
Masutani K, Ninomiya T, Randhawa P. HLA-A2, HLA-B44 and HLA-DR15 are associated with lower risk of BK viremia. Nephrol Dial Transplant 2013. [PMID: 24084328 DOI: 10.1093/ndt/fgt298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Human leucocyte antigens (HLAs) modulate immunity to polyomavirus BK (BKV). Identification of HLAs that alter the course of infection will facilitate risk stratification, and customization of pre-emptive intervention strategies. METHODS We performed a retrospective cohort study with 998 kidney transplant patients with BKV infection status confirmed by polymerase chain reaction (PCR). Clinical parameters and donor-recipient matching for specific HLAs were examined in relation to occurrence of viremia. An emphasis was placed on donor-recipient matching rather than the actual frequency of specific HLA-alleles, since a successful immune response requires sharing of HLAs between a virus-infected target cell and the anti-viral effector cell. RESULTS Using multivariate statistics, low risk of BK viremia was associated with matching of HLA-A2 [hazard ratio (HR) 0.51, 95% confidence interval (CI) 0.28-0.85], HLA-B44 (HR 0.31, 95% CI 0.076-0.85) and HLA-DR15 (HR 0.35, 95% CI 0.084-0.93) (P < 0.05), whereas high risk of viremia was associated with male gender (HR 2.38, 95% CI 1.46-4.09, P < 0.001). CONCLUSIONS HLAs that associated with a lower predisposition to the development of BK viremia have been identified. Evaluation of donor-recipient mismatching for these HLAs could potentially be used to (i) fine tune virus screening strategies for BKV in individual patients and (ii) facilitate discovery of major histocompatibility complex (MHC) class I and II binding peptides that can elicit clinically meaningful BKV-specific immunity.
Collapse
Affiliation(s)
- Kosuke Masutani
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | |
Collapse
|
15
|
Masutani K, Ninomiya T, Randhawa P. HLA-A2, HLA-B44 and HLA-DR15 are associated with lower risk of BK viremia. Nephrol Dial Transplant 2013; 28:3119-26. [PMID: 24084328 DOI: 10.1093/ndt/gft298] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human leucocyte antigens (HLAs) modulate immunity to polyomavirus BK (BKV). Identification of HLAs that alter the course of infection will facilitate risk stratification, and customization of pre-emptive intervention strategies. METHODS We performed a retrospective cohort study with 998 kidney transplant patients with BKV infection status confirmed by polymerase chain reaction (PCR). Clinical parameters and donor-recipient matching for specific HLAs were examined in relation to occurrence of viremia. An emphasis was placed on donor-recipient matching rather than the actual frequency of specific HLA-alleles, since a successful immune response requires sharing of HLAs between a virus-infected target cell and the anti-viral effector cell. RESULTS Using multivariate statistics, low risk of BK viremia was associated with matching of HLA-A2 [hazard ratio (HR) 0.51, 95% confidence interval (CI) 0.28-0.85], HLA-B44 (HR 0.31, 95% CI 0.076-0.85) and HLA-DR15 (HR 0.35, 95% CI 0.084-0.93) (P < 0.05), whereas high risk of viremia was associated with male gender (HR 2.38, 95% CI 1.46-4.09, P < 0.001). CONCLUSIONS HLAs that associated with a lower predisposition to the development of BK viremia have been identified. Evaluation of donor-recipient mismatching for these HLAs could potentially be used to (i) fine tune virus screening strategies for BKV in individual patients and (ii) facilitate discovery of major histocompatibility complex (MHC) class I and II binding peptides that can elicit clinically meaningful BKV-specific immunity.
Collapse
Affiliation(s)
- Kosuke Masutani
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | |
Collapse
|
16
|
Dhama K, Latheef SK, Samad HA, Chakrabort S, Tiwari R, Kumar A, Rahal A. Tumor Necrosis Factor as Mediator of Inflammatory Diseases and its Therapeutic Targeting: A Review. JOURNAL OF MEDICAL SCIENCES 2013. [DOI: 10.3923/jms.2013.226.235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|