1
|
Miller-Handley H, Harper G, Pham G, Turner LH, Shao TY, Russi AE, Erickson JJ, Ford ML, Araki K, Way SS. Immune suppression sustained allograft acceptance requires PD1 inhibition of CD8+ T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:192-198. [PMID: 40073258 PMCID: PMC11904129 DOI: 10.1093/jimmun/vkae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/24/2024] [Indexed: 03/14/2025]
Abstract
Organ transplant recipients require continual immune-suppressive therapies to sustain allograft acceptance. Although medication nonadherence is a major cause of rejection, the mechanisms responsible for graft loss in this clinically relevant context among individuals with preceding graft acceptance remain uncertain. Here, we demonstrate that skin allograft acceptance in mice maintained with clinically relevant immune-suppressive therapies, tacrolimus and mycophenolate, sensitizes hypofunctional PD1hi graft-specific CD8+ T cells. Uninterrupted immune-suppressive therapy is required because drug discontinuation triggers allograft rejection, replicating the requirement for immune-suppressive therapy adherence in transplant recipients. Graft-specific CD8+ T cells in allograft-accepted mice show diminished effector differentiation and cytokine production, with reciprocally increased PD1 expression. Allograft acceptance-induced PD1 expression is essential, as PDL1 blockade reinvigorates graft-specific CD8+ T cell activation with ensuing allograft rejection despite continual immune-suppressive therapy. Thus, PD1 sustained CD8+ T cell inhibition is essential for allograft acceptance maintained by tacrolimus plus mycophenolate. This necessity for PD1 in sustaining allograft acceptance explains the high rates of rejection in transplant recipients with cancer administered immune checkpoint inhibitors targeting PD1/PDL1, highlighting shared immune suppression pathways exploited by tumor cells and current therapies for averting allograft rejection.
Collapse
Affiliation(s)
- Hilary Miller-Handley
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
- Department of Medicine, University of Cincinnati College of Medicine
| | - Gavin Harper
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Lucien H. Turner
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Abigail E. Russi
- Division of Gastroenterology, Hepatology and Advanced Nutrition, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - John J. Erickson
- Division of Neonatology, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Mandy L. Ford
- Winship Cancer Institute, Emory University School of Medicine
| | - Koichi Araki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
2
|
Nicosia M, Valujskikh A. Recognizing Complexity of CD8 T Cells in Transplantation. Transplantation 2024; 108:2186-2196. [PMID: 38637929 PMCID: PMC11489323 DOI: 10.1097/tp.0000000000005001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
The major role of CD8 + T cells in clinical and experimental transplantation is well documented and acknowledged. Nevertheless, the precise impact of CD8 + T cells on graft tissue injury is not completely understood, thus impeding the development of specific treatment strategies. The goal of this overview is to consider the biology and functions of CD8 + T cells in the context of experimental and clinical allotransplantation, with special emphasis on how this cell subset is affected by currently available and emerging therapies.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
3
|
Ashraf MI, Mengwasser J, Reutzel-Selke A, Polenz D, Führer K, Lippert S, Tang P, Michaelis E, Catar R, Pratschke J, Witzel C, Sauer IM, Tullius SG, Kern B. Depletion of donor dendritic cells ameliorates immunogenicity of both skin and hind limb transplants. Front Immunol 2024; 15:1395945. [PMID: 38799435 PMCID: PMC11116604 DOI: 10.3389/fimmu.2024.1395945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Acute cellular rejection remains a significant obstacle affecting successful outcomes of organ transplantation including vascularized composite tissue allografts (VCA). Donor antigen presenting cells (APCs), particularly dendritic cells (DCs), orchestrate early alloimmune responses by activating recipient effector T cells. Employing a targeted approach, we investigated the impact of donor-derived conventional DCs (cDCs) and APCs on the immunogenicity of skin and skin-containing VCA grafts, using mouse models of skin and hind limb transplantation. By post-transplantation day 6, skin grafts demonstrated severe rejections, characterized by predominance of recipient CD4 T cells. In contrast, hind limb grafts showed moderate rejection, primarily infiltrated by CD8 T cells. Notably, the skin component exhibited heightened immunogenicity when compared to the entire VCA, evidenced by increased frequencies of pan (CD11b-CD11c+), mature (CD11b-CD11c+MHCII+) and active (CD11b-CD11c+CD40+) DCs and cDC2 subset (CD11b+CD11c+ MHCII+) in the lymphoid tissues and the blood of skin transplant recipients. While donor depletion of cDC and APC reduced frequencies, maturation and activation of DCs in all analyzed tissues of skin transplant recipients, reduction in DC activities was only observed in the spleen of hind limb recipients. Donor cDC and APC depletion did not impact all lymphocyte compartments but significantly affected CD8 T cells and activated CD4 T in lymph nodes of skin recipients. Moreover, both donor APC and cDC depletion attenuated the Th17 immune response, evident by significantly reduced Th17 (CD4+IL-17+) cells in the spleen of skin recipients and reduced levels of IL-17E and lymphotoxin-α in the serum samples of both skin and hind limb recipients. In conclusion, our findings underscore the highly immunogenic nature of skin component in VCA. The depletion of donor APCs and cDCs mitigates the immunogenicity of skin grafts while exerting minimal impact on VCA.
Collapse
Affiliation(s)
- Muhammad Imtiaz Ashraf
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joerg Mengwasser
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Dietrich Polenz
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Kirsten Führer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Steffen Lippert
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Peter Tang
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Edward Michaelis
- Department of Pathology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christian Witzel
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Igor M. Sauer
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Stefan G. Tullius
- Division of Transplant Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Einstein Berlin Institute of Health Visiting Fellow, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara Kern
- Department of Surgery, Experimental Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Department of Plastic Surgery, Charité – Universitätsmedizin Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Biomedical Innovation Academy, Berlin Institute of Health (BIH) Charité Clinician Scientist Program, Berlin, Germany
| |
Collapse
|
4
|
Li S, Zou D, Chen W, Cheng Y, Britz GW, Weng YL, Liu Z. Ablation of BATF Alleviates Transplant Rejection via Abrogating the Effector Differentiation and Memory Responses of CD8 + T Cells. Front Immunol 2022; 13:882721. [PMID: 35514970 PMCID: PMC9062028 DOI: 10.3389/fimmu.2022.882721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/25/2022] [Indexed: 02/02/2023] Open
Abstract
Allogeneic CD8+ T cells are prominently involved in allograft rejection, but how their effector differentiation and function are regulated at a transcriptional level is not fully understood. Herein, we identified the basic leucine zipper ATF-like transcription factor (BATF) as a key transcription factor that drives the effector program of allogeneic CD8+ T cells. We found that BATF is highly expressed in graft-infiltrating CD8+ T cells, and its ablation in CD8+ T cells significantly prolonged skin allograft survival in a fully MHC-mismatched transplantation model. To investigate how BATF dictates allogeneic CD8+ T cell response, BATF-/- and wild-type (WT) CD8+ T cells were mixed in a 1:1 ratio and adoptively transferred into B6.Rag1-/- mice 1 day prior to skin transplantation. Compared with WT CD8+ T cells at the peak of rejection response, BATF-/- CD8+ T cells displayed a dysfunctional phenotype, evident by their failure to differentiate into CD127-KLRG1+ terminal effectors, impaired proliferative capacity and production of pro-inflammatory cytokines/cytotoxic molecules, and diminished capacity to infiltrate allografts. In association with the failure of effector differentiation, BATF-/- CD8+ T cells largely retained TCF1 expression and expressed significantly low levels of T-bet, TOX, and Ki67. At the memory phase, BATF-deficient CD8+ T cells displayed impaired effector differentiation upon allogeneic antigen re-stimulation. Therefore, BATF is a critical transcriptional determinant that governs the terminal differentiation and memory responses of allogeneic CD8+ T cells in the transplantation setting. Targeting BATF in CD8+ T cells may be an attractive therapeutic approach to promote transplant acceptance.
Collapse
Affiliation(s)
- Shuang Li
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,Institute of Clinical Pharmacology, Central South University, Changsha, China,Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, United States,Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Dawei Zou
- Immunobiology & Transplant Science Center, Department of Surgery, Houston Methodist Research Institute & Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Wenhao Chen
- Immunobiology & Transplant Science Center, Department of Surgery, Houston Methodist Research Institute & Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Yating Cheng
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, United States,Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Gavin W. Britz
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, United States,Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Yi-Lan Weng
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, United States,Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, United States,*Correspondence: Zhaoqian Liu, ;Yi-Lan Weng,
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,Institute of Clinical Pharmacology, Central South University, Changsha, China,*Correspondence: Zhaoqian Liu, ;Yi-Lan Weng,
| |
Collapse
|
5
|
Balam S, Kesselring R, Eggenhofer E, Blaimer S, Evert K, Evert M, Schlitt HJ, Geissler EK, van Blijswijk J, Lee S, Reis e Sousa C, Brunner SM, Fichtner-Feigl S. Cross-presentation of dead-cell-associated antigens by DNGR-1 + dendritic cells contributes to chronic allograft rejection in mice. Eur J Immunol 2020; 50:2041-2054. [PMID: 32640051 DOI: 10.1002/eji.201948501] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 01/06/2023]
Abstract
The purpose of this study was to elucidate whether DC NK lectin group receptor-1 (DNGR-1)-dependent cross-presentation of dead-cell-associated antigens occurs after transplantation and contributes to CD8+ T cell responses, chronic allograft rejection (CAR), and fibrosis. BALB/c or C57BL/6 hearts were heterotopically transplanted into WT, Clec9a-/- , or Batf3-/- recipient C57BL/6 mice. Allografts were analyzed for cell infiltration, CD8+ T cell activation, fibrogenesis, and CAR using immunohistochemistry, Western blot, qRT2 -PCR, and flow cytometry. Allografts displayed infiltration by recipient DNGR-1+ DCs, signs of CAR, and fibrosis. Allografts in Clec9a-/- recipients showed reduced CAR (p < 0.0001), fibrosis (P = 0.0137), CD8+ cell infiltration (P < 0.0001), and effector cytokine levels compared to WT recipients. Batf3-deficiency greatly reduced DNGR-1+ DC-infiltration, CAR (P < 0.0001), and fibrosis (P = 0.0382). CD8 cells infiltrating allografts of cytochrome C treated recipients, showed reduced production of CD8 effector cytokines (P < 0.05). Further, alloreactive CD8+ T cell response in indirect pathway IFN-γ ELISPOT was reduced in Clec9a-/- recipient mice (P = 0.0283). Blockade of DNGR-1 by antibody, similar to genetic elimination of the receptor, reduced CAR (P = 0.0003), fibrosis (P = 0.0273), infiltration of CD8+ cells (p = 0.0006), and effector cytokine levels. DNGR-1-dependent alloantigen cross-presentation by DNGR-1+ DCs induces alloreactive CD8+ cells that induce CAR and fibrosis. Antibody against DNGR-1 can block this process and prevent CAR and fibrosis.
Collapse
Affiliation(s)
- Saidou Balam
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Rebecca Kesselring
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Elke Eggenhofer
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Stephanie Blaimer
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Katja Evert
- Department of Pathology, University Medical Center Regensburg, Regensburg, Germany
| | - Matthias Evert
- Department of Pathology, University Medical Center Regensburg, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | - Sonia Lee
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | | - Stefan M Brunner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Stefan Fichtner-Feigl
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany.,Department of General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Watson AR, Dai H, Diaz-Perez JA, Killeen ME, Mathers AR, Thomson AW. mTORC2 deficiency in cutaneous dendritic cells potentiates CD8 + effector T cell responses and accelerates skin graft rejection. Am J Transplant 2019; 19:646-661. [PMID: 30129283 PMCID: PMC6384165 DOI: 10.1111/ajt.15083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/27/2018] [Accepted: 08/14/2018] [Indexed: 01/25/2023]
Abstract
Mechanistic target of rapamycin (mTOR) complex (mTORC)1 and mTORC2 regulate the differentiation and function of immune cells. While inhibition of mTORC1 antagonizes dendritic cell (DC) differentiation and suppresses graft rejection, the role of mTORC2 in DCs in determining host responses to transplanted tissue remains undefined. Using a mouse model in which mTORC2 was deleted specifically in CD11c+ DCs (TORC2DC-/- ), we show that the transplant of minor histocompatibility Ag (HY)-mismatched skin grafts from TORC2DC-/- donors into wild-type recipients results in accelerated rejection characterized by enhanced CD8+ T cell responses in the graft and regional lymphoid tissue [Correction added on January 9, 2019, after first online publication: in the previous sentence, major was changed to minor]. Similar enhancement of CD8+ effector T cell responses was observed in MHC-mismatched recipients of TORC2DC-/- grafts. Augmented CD8+ T cell responses were also observed in a delayed-type hypersensitivity model in which mTORC2 was absent in cutaneous DCs. These elevated responses could be ascribed to an increased T cell stimulatory phenotype of TORC2DC-/- and not to enhanced lymph node homing of the cells. In contrast, rejection of ovalbumin transgenic skin grafts in TORC2DC-/- recipients was unaffected. These findings suggest that mTORC2 in skin DCs restrains effector CD8+ T cell responses and have implications for understanding of the influence of mTOR inhibitors that target mTORC2 in transplant.
Collapse
Affiliation(s)
- Alicia R. Watson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Helong Dai
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Julio A. Diaz-Perez
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Meaghan E. Killeen
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Alicia R. Mathers
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Angus W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA,Correspondence: Angus W. Thomson, PhD, DSc, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1540, Pittsburgh, PA 15261, , (412) 624-6392
| |
Collapse
|
7
|
Ablamunits V, Goldstein AJ, Tovbina MH, Gaetz HP, Klebanov S. Acute Rejection of White Adipose Tissue Allograft. Cell Transplant 2017; 16:375-90. [PMID: 17658128 DOI: 10.3727/000000007783464830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
White adipose tissue (WAT) transplantation, although widely used in humans, has been done for cosmetic and reconstructive purposes only. Accumulating evidence indicates, however, that WAT is an important endocrine organ and, therefore, WAT transplantation may become valuable as a replacement therapy for a number of hereditary human diseases. Because the most readily available source for such transplantations would be allogeneic tissue, the mechanisms involved in the rejection of WAT allograft should be explored. We have established a model in which leptin-producing allogeneic WAT is transplanted into leptin-deficient ob/ob mice. Because ob/ob mice are obese, hyperphagic, and hypothermic, WAT allograft function is monitored as the reversal of this leptin-deficient phenotype. Here we report that allografted WAT is primarily nonfunctional. However, when WAT is transplanted into immunodeficient (Rag1–/–) ob/ob mice, or into ob/ob mice depleted of T cells by anti-CD3 antibody, a long-term graft survival is achieved as indicated by the reversal of hyperphagia, weight loss, and normalization of body temperature. The symptoms of leptin deficiency rapidly recur when normal spleen cells of the recipient type are injected, or when the antibody treatment is terminated. In contrast, selective depletion of either CD4+ or CD8+ cells alone does not prevent WAT allograft rejection. Similarly, WAT allografts that do not express MHC class I or class II molecules are rapidly rejected, suggesting that both CD4+ and CD8+ T cells may independently mediate WAT allograft rejection.
Collapse
Affiliation(s)
- Vitaly Ablamunits
- Obesity Research Center, St. Luke's Hospital, New York, NY 10025, USA.
| | | | | | | | | |
Collapse
|
8
|
Deng G, Deng R, Yao J, Liao B, Chen Y, Wu Z, Hu H, Zhou X, Ma Y. Trichinella spiralis infection changes immune response in mice performed abdominal heterotopic cardiac transplantation and prolongs cardiac allograft survival time. Parasitol Res 2015; 115:407-14. [DOI: 10.1007/s00436-015-4762-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/28/2015] [Indexed: 10/22/2022]
|
9
|
Attenuation of Acute Rat Renal Allograft Rejection by Apolipoprotein E-Mimetic Peptide. Transplantation 2015; 99:925-34. [DOI: 10.1097/tp.0000000000000569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Abstract
Natural killer (NK) cells and cytotoxic T lymphocytes (CTL) use a highly toxic pore-forming protein perforin (PFN) to destroy cells infected with intracellular pathogens and cells with pre-cancerous transformations. However, mutations of PFN and defects in its expression can cause an abnormal function of the immune system and difficulties in elimination of altered cells. As discussed in this chapter, deficiency of PFN due to the mutations of its gene, PFN1, can be associated with malignancies and severe immune disorders such as familial hemophagocytic lymphohistiocytosis (FHL) and macrophage activation syndrome. On the other hand, overactivity of PFN can turn the immune system against autologous cells resulting in other diseases such as systemic lupus erythematosus, polymyositis, rheumatoid arthritis and cutaneous inflammation. PFN also has a crucial role in the cellular rejection of solid organ allografts and destruction of pancreatic β-cells resulting in type 1 diabetes. These facts highlight the importance of understanding the biochemical characteristics of PFN.
Collapse
Affiliation(s)
- Omar Naneh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | | | | |
Collapse
|
11
|
Patel SR, Zimring JC. Transfusion-induced bone marrow transplant rejection due to minor histocompatibility antigens. Transfus Med Rev 2013; 27:241-8. [PMID: 24090731 DOI: 10.1016/j.tmrv.2013.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 08/15/2013] [Accepted: 08/17/2013] [Indexed: 11/18/2022]
Abstract
Traditionally, alloimmunization to transfused blood products has focused exclusively on recipient antibodies recognizing donor alloantigens present on the cell surface. Accordingly, the immunologic sequelae of alloimmunization have been antibody mediated effects (ie, hemolytic transfusion reactions, platelet refractoriness, anti-HLA and anti-HNA effects, etc). However, in addition to the above sequelae, there is also a correlation between the number of antecedent transfusions in humans and the rate of bone marrow transplant (BMT) rejection-under reduced intensity conditioning with HLA-matched or HLA-identical marrow. Bone marrow transplant of this nature is the only existing cure for a series of nonmalignant hematologic diseases (eg, sickle cell disease, thalassemias, etc); however, rejection remains a clinical problem. It has been hypothesized that transfusion induces subsequent BMT rejection through immunization. Studies in animal models have observed the same effect and have demonstrated that transfusion-induced BMT rejection can occur in response to alloimmunization. However, unlike traditional antibody responses, sensitization in this case results in cellular immune effects, involving populations such as T cell or natural killer cells. In this case, rejection occurs in the absence of alloantibodies and would not be detected by existing immune-hematologic methods. We review human and animal studies in light of the hypothesis that, for distinct clinical populations, enhanced rejection of BMT may be an unappreciated adverse consequence of transfusion, which current blood bank methodologies are unable to detect.
Collapse
Affiliation(s)
- Seema R Patel
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | | |
Collapse
|
12
|
Marchant DJ, Boyd JH, Lin DC, Granville DJ, Garmaroudi FS, McManus BM. Inflammation in myocardial diseases. Circ Res 2012; 110:126-44. [PMID: 22223210 DOI: 10.1161/circresaha.111.243170] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammatory processes underlie a broad spectrum of conditions that injure the heart muscle and cause both structural and functional deficits. In this article, we address current knowledge regarding 4 common forms of myocardial inflammation: myocardial ischemia and reperfusion, sepsis, viral myocarditis, and immune rejection. Each of these pathological states has its own unique features in pathogenesis and disease evolution, but all reflect inflammatory mechanisms that are partially shared. From the point of injury to the mobilization of innate and adaptive immune responses and inflammatory amplification, the cellular and soluble mediators and mechanisms examined in this review will be discussed with a view that both beneficial and adverse consequences arise in these human conditions.
Collapse
Affiliation(s)
- David J Marchant
- James Hogg Research Centre and Institute for Heart + Lung Health, Department of Pathology and Laboratory Medicine, University of British Columbia, Providence Health Care, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
13
|
Benichou G, Yamada Y, Yun SH, Lin C, Fray M, Tocco G. Immune recognition and rejection of allogeneic skin grafts. Immunotherapy 2012; 3:757-70. [PMID: 21668313 DOI: 10.2217/imt.11.2] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The transplantation of allogeneic skin grafts is associated with a potent inflammatory immune response leading to the destruction of donor cells and the rejection of the graft. Shortly after transplantation, skin dendritic cells (DCs) migrate out of the graft through lymphatic vessels and infiltrate the recipient's draining lymph nodes where they present donor antigens via two mechanisms: the direct pathway, in which T cells recognize intact donor MHC antigens on donor DCs; and the indirect pathway, involving T-cell recognition of donor peptides bound to self-MHC molecules on recipient DCs. Some recent studies have suggested that T cells can become activated via recognition of donor MHC molecules transferred on recipient antigen-presenting cells (semidirect pathway). Activation of T cells via direct or indirect allorecognition is sufficient to trigger acute rejection of allogeneic skin grafts. In addition, allospecific antibodies contribute to the rejection process either by killing allogeneic targets in a complement-dependent fashion or by opsonizing donor cells and forming immune complexes. Finally, several studies demonstrate that NK cells, activated due to missing self-MHC class I molecules on allogeneic cells, are involved in allogeneic skin graft rejection via direct killing of donor cells and through the production of proinflammatory cytokines including IFN-γ and TNF-α.
Collapse
Affiliation(s)
- Gilles Benichou
- Department of Surgery, Transplant Unit & Wellman Photomedicine Center Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Shaw PJ, Feske S. Physiological and pathophysiological functions of SOCE in the immune system. Front Biosci (Elite Ed) 2012. [PMID: 22202035 DOI: 10.2741/540] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcium signals play a critical role in many cell-type specific effector functions during innate and adaptive immune responses. The predominant mechanism to raise intracellular (Ca²⁺) used by most immune cells is store-operated Ca²⁺ entry (SOCE), whereby the depletion of endoplasmic reticulum (ER) Ca²⁺ stores triggers the influx of extracellular Ca²⁺. SOCE in immune cells is mediated by the highly Ca²⁺ selective Ca²⁺-release-activated Ca²⁺ (CRAC) channel, encoded by ORAI1, ORAI2 and ORAI3 genes. ORAI proteins are activated by stromal interaction molecules (STIM) 1 and 2, which act as sensors of ER Ca²⁺ store depletion. The importance of SOCE mediated by STIM and ORAI proteins for immune function is evident from the immunodeficiency and autoimmunity in patients with mutations in STIM1 and ORAI1 genes. These patients and studies in gene-targeted mice have revealed an essential role for ORAI/STIM proteins in the function of several immune cells. This review focuses on recent advances made towards understanding the role of SOCE in immune cells with an emphasis on the immune dysregulation that results from defects in SOCE in human patients and transgenic mice.
Collapse
Affiliation(s)
- Patrick J Shaw
- Department of Pathology, New York University Langone Medical Center, New York, NY 10016, USA
| | | |
Collapse
|
15
|
McCarl CA, Khalil S, Ma J, Oh-hora M, Yamashita M, Roether J, Kawasaki T, Jairaman A, Sasaki Y, Prakriya M, Feske S. Store-operated Ca2+ entry through ORAI1 is critical for T cell-mediated autoimmunity and allograft rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:5845-58. [PMID: 20956344 PMCID: PMC2974040 DOI: 10.4049/jimmunol.1001796] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
ORAI1 is the pore-forming subunit of the Ca(2+) release-activated Ca(2+) (CRAC) channel, which is responsible for store-operated Ca(2+) entry in lymphocytes. A role for ORAI1 in T cell function in vivo has been inferred from in vitro studies of T cells from human immunodeficient patients with mutations in ORAI1 and Orai1(-/-) mice, but a detailed analysis of T cell-mediated immune responses in vivo in mice lacking functional ORAI1 has been missing. We therefore generated Orai1 knock-in mice (Orai1(KI/KI)) expressing a nonfunctional ORAI1-R93W protein. Homozygosity for the equivalent ORAI1-R91W mutation abolishes CRAC channel function in human T cells resulting in severe immunodeficiency. Homozygous Orai1(KI/KI) mice die neonatally, but Orai1(KI/KI) fetal liver chimeric mice are viable and show normal lymphocyte development. T and B cells from Orai1(KI/KI) mice display severely impaired store-operated Ca(2+) entry and CRAC channel function resulting in a strongly reduced expression of several key cytokines including IL-2, IL-4, IL-17, IFN-γ, and TNF-α in CD4(+) and CD8(+) T cells. Cell-mediated immune responses in vivo that depend on Th1, Th2, and Th17 cell function were severely attenuated in ORAI1-deficient mice. Orai1(KI/KI) mice lacked detectable contact hypersensitivity responses and tolerated skin allografts significantly longer than wild-type mice. In addition, T cells from Orai1(KI/KI) mice failed to induce colitis in an adoptive transfer model of inflammatory bowel disease. These findings reaffirm the critical role of ORAI1 for T cell function and provide important insights into the in vivo functions of CRAC channels for T cell-mediated immunity.
Collapse
Affiliation(s)
- Christie-Ann McCarl
- Department of Pathology, New York University Langone Medical Center, NY, NY 10016, USA
- Immune Disease Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Sara Khalil
- Department of Pathology, New York University Langone Medical Center, NY, NY 10016, USA
| | - Jian Ma
- Department of Pathology, New York University Langone Medical Center, NY, NY 10016, USA
| | - Masatsugu Oh-hora
- Immune Disease Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Megumi Yamashita
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jens Roether
- Immune Disease Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Takumi Kawasaki
- Department of Pathology, New York University Langone Medical Center, NY, NY 10016, USA
| | - Amit Jairaman
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yoshiteru Sasaki
- Immune Disease Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Murali Prakriya
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Stefan Feske
- Department of Pathology, New York University Langone Medical Center, NY, NY 10016, USA
- Immune Disease Institute and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Gorbacheva V, Fan R, Li X, Valujskikh A. Interleukin-17 promotes early allograft inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1265-73. [PMID: 20651239 DOI: 10.2353/ajpath.2010.091106] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Acute cellular rejection of organ transplants is executed by donor-reactive T cells, which are dominated by interferon-gamma-producing cells. As interferon-gamma is dispensable for graft destruction, we evaluated the contribution of interleukin-17A (IL-17) to intragraft inflammation in major histocompatibility complex-mismatched heart transplants. A/J (H-2(a)) cardiac allografts placed into wild-type BALB/c (H-2(d)) mice induced intragraft IL-17 production on day 2 after transplant. Allografts placed into BALB/c IL-17(-/-) recipients demonstrated diminished production of the chemokines CXCL1 and CXCL2 and delayed neutrophil and T cell recruitment. However, by day 7 after transplant, allografts from IL-17(-/-) and wild-type recipients had comparable levels of cellular infiltration. The priming of donor-specific T cells was not affected by the absence of IL-17, and the kinetics of cardiac allograft rejection were similar in wild-type and IL-17(-/-) recipients. In contrast, IL-17(-/-) mice depleted of CD8 T cells rejected A/J allografts in a delayed fashion compared with CD8-depleted wild-type recipients. Although donor-reactive CD4 T cells were efficiently activated in both groups, the infiltration of effector T cells into allografts was impaired in IL-17(-/-) recipients. Our data indicate that locally produced IL-17 amplifies intragraft inflammation early after transplantation and promotes tissue injury by facilitating T cell recruitment into the graft. Targeting the IL-17 signaling network in conjunction with other graft-prolonging therapies may decrease this injury and improve the survival of transplanted organs.
Collapse
|
17
|
Immunological tolerance induced by galectin-1 in rat allogeneic renal transplantation. Int Immunopharmacol 2010; 10:643-7. [DOI: 10.1016/j.intimp.2010.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 02/25/2010] [Accepted: 03/02/2010] [Indexed: 11/19/2022]
|
18
|
Reimers K, Radtke C, Choi CY, Allmeling C, Kall S, Kiefer P, Muehlberger T, Vogt PM. Expression of TNF-related apoptosis-inducing ligand (TRAIL) in keratinocytes mediates apoptotic cell death in allogenic T cells. ANNALS OF SURGICAL INNOVATION AND RESEARCH 2009; 3:13. [PMID: 19925644 PMCID: PMC2790448 DOI: 10.1186/1750-1164-3-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2009] [Accepted: 11/19/2009] [Indexed: 11/10/2022]
Abstract
The objective of the present study was to evaluate the aptitude of TRAIL gene expression for inducing apoptosis in co-cultivated T-cells. This should allow preparing a strategy for the development of a durable, allogenic skin substitute based on the induction of an immune-privileged transplant. In order to counteract the significant potential of rejection in transplanted allogenic keratinocytes, we created a murine keratinocyte cell line which expressed TRAIL through stable gene transfer. The exogenic protein was localized on the cellular surface and was not found in soluble condition as sTRAIL. Contact to TRAIL expressing cells in co-culture induced cell death in sensitive Jurkat-cells, which was further intensified by lymphocyte activation. This cytotoxic effect is due to the induction of apoptosis. We therefore assume that the de-novo expression of TRAIL in keratinocytes can trigger apoptosis in activated lymphocytes and thus prevent the rejection of keratinocytes in allogenic, immune-privileged transplants.
Collapse
Affiliation(s)
- Kerstin Reimers
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| | - Christine Radtke
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| | - Claudia Y Choi
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| | - Christina Allmeling
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| | - Susanne Kall
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| | - Paul Kiefer
- University Duesseldorf, Moorenstrasse 5, 40225 Duesseldorf, Germany
| | - Thomas Muehlberger
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover Podbielskistraße 380, 30659 Hannover, Germany
| |
Collapse
|
19
|
|
20
|
Krishnadas DK, Li W, Kumar R, Tyrrell LJ, Agrawal B. In vitro activation and differentiation of naïve CD4+ and CD8+ T cells into HCV Core- and NS3-specific armed effector cells: A new role for CD4+ T cells. Cell Immunol 2009; 259:141-9. [DOI: 10.1016/j.cellimm.2009.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 06/10/2009] [Accepted: 06/12/2009] [Indexed: 12/26/2022]
|
21
|
Pancreatic Islets Induce CD4+CD25−Foxp3+ T-Cell Regulated Tolerance to HY-Mismatched Skin Grafts. Transplantation 2008; 86:1352-60. [DOI: 10.1097/tp.0b013e31818aa43c] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
22
|
Lange V, Renner A, Sagstetter MR, Lazariotou M, Harms H, Gummert JF, Leyh RG, Elert O. Heterotopic rat heart transplantation (Lewis to F344): early ICAM-1 expression after 8 hours of cold ischemia. J Heart Lung Transplant 2008; 27:1031-5. [PMID: 18765197 DOI: 10.1016/j.healun.2008.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 05/07/2008] [Accepted: 06/03/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Primary graft dysfunction is a still poorly understood complication after cardiac transplantation. Ischemia/reperfusion injury contributes to different disorders resulting in impaired graft function. METHODS In a heterotopic rat heart transplantation model we extended graft ischemic time up to 8 hours. RESULTS Using immunohistochemistry we detected an up to 4-fold increase in intracellular adhesion molecule-1 (ICAM-1) expression during 4 hours of reperfusion, independent of ischemic time (30-minute ischemia: 7.65 +/- 2.15 without reperfusion, 19.46 +/- 4.6 after 4-hour reperfusion; 240-minute ischemia: 5.6 +/- 1.99 and 22.3 +/- 3.77; 480-minute ischemia: 3.7 +/- 1.56 and 13.1 +/- 2.2). Eight-hour ischemic allografts had an increase in CD8-positive cells (1.37 +/- 0.5 and 2.3 +/- 0.77) and a significant increase in MHC II expression (11.48 +/- 2.1 and 18.27 +/- 1.34) during 4 hours of reperfusion. CONCLUSIONS We hypothesize that these findings reflect an early inflammatory reaction in the allograft possibly triggered by oxidative stress. During therapeutic interventions, both of these pathways must be considered.
Collapse
Affiliation(s)
- Volkmar Lange
- Department of Cardiac, Thoracic and Thoracic Vascular Surgery, University of Wuerzburg, Wuerzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Activation of Tim-3-Galectin-9 pathway improves survival of fully allogeneic skin grafts. Transpl Immunol 2008; 19:12-9. [PMID: 18346632 DOI: 10.1016/j.trim.2008.01.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 01/07/2008] [Indexed: 01/18/2023]
Abstract
T cell immunoglobulin and mucin domain (Tim)-3 is a molecule expressed on terminally differentiated murine Th1 cells but not on Th2 cells. Identification of Galectin-9 as a ligand for Tim-3 has now firmly established the Tim-3-Galectin-9 pathway as an important regulator of Th1 immunity, which results in apoptosis of Th1 cells. Here, we demonstrate that engagement of Tim-3 by mouse recombinant Galectin-9 remarkably suppresses allograft rejection and improves survival of allogeneic skin grafts. Furthermore, administration of recombinant Galectin-9 decreases Tim-3 positive cells in draining lymph node and selectively inhibits production of IFN-gamma after skin transplantation. At last, even low dose of Galectin-9 (1 microg/ml) can obviously inhibit TCR crosslinking-induced primary T cell proliferation in vitro. These observations suggest that Tim-3-Galectin-9 pathway plays an important role in the termination of productive Th1-immune response and could lead to developing novel therapies in transplant medicine.
Collapse
|
24
|
Zhai Y, Wang Y, Wu Z, Kupiec-Weglinski JW. Defective alloreactive CD8 T cell function and memory response in allograft recipients in the absence of CD4 help. THE JOURNAL OF IMMUNOLOGY 2007; 179:4529-34. [PMID: 17878349 DOI: 10.4049/jimmunol.179.7.4529] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have shown that alloreactive CD8 T cell activation may proceed via CD4-dependent and CD4-independent pathways, and that CD8 T cell activation in Ag-primed animals is independent of CD154 costimulation. In this report, we further analyzed the activation and function of alloreactive CD8 CTL effectors in CD4 knockout (KO) skin/cardiac allograft recipients. FACS analysis showed that alloreactive CD8 T cells were activated at a significantly reduced level in CD4 KO mice. Importantly, these helpless CD8 T cells failed to develop CD154 blockade resistance following reactivation by the same alloantigen, indicative of defective memory formation. Only transient CD4 help was required, as short-term CD4 blockade at the time of first skin graft challenge only delayed alloreactive CD8 activation, without affecting the CD8 T cell memory response to a second skin graft. Moreover, postoperative CD4 blockade had no effect on alloreactive CD8 activation. Alloreactive CD8 cells generated in the absence of CD4 help exhibited decreased effector responses. Interestingly, intragraft induction of T cell-targeted chemokines early after transplant was also dependent on CD4 help, as the induction kinetics of CXCL9 and CCL5 in CD4 KO recipients was significantly delayed, coupled with similarly delayed infiltration by CD3/CD8 cells. Remarkably, helpless CD8 cells ultimately entering the graft still displayed significantly diminished T cell effector molecules (IFN-gamma, granzyme B). Thus, CD4 help is critical for alloreactive CD8 activation, function, and memory formation.
Collapse
Affiliation(s)
- Yuan Zhai
- Division of Liver and Pancreas Transplantation, Department of Surgery, Dumont-University of California Los Angeles Transplant Center, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
25
|
The Tim-3 ligand galectin-9 negatively regulates CD8+ alloreactive T cell and prolongs survival of skin graft. Cell Immunol 2007; 250:68-74. [DOI: 10.1016/j.cellimm.2008.01.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 12/21/2007] [Accepted: 01/16/2008] [Indexed: 12/14/2022]
|
26
|
Habicht A, Najafian N, Yagita H, Sayegh MH, Clarkson MR. New insights in CD28-independent allograft rejection. Am J Transplant 2007; 7:1917-26. [PMID: 17617854 DOI: 10.1111/j.1600-6143.2007.01886.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CD28 costimulatory blockade induces tolerance in most murine transplant models but fails to do so in stringent transplant models, such as skin transplantation. The precise immunological mechanisms of CD28-independent rejection remain to be fully defined. Using two novel mouse strains in which both CD28 and either CD4 or CD8 are knocked out (CD4(-/-)CD28(-/-) or CD8(-/-)CD28(-/-) mice), we examined mechanisms of CD28-independent CD4(+) or CD8(+) T-cell-mediated allograft rejection. CD4(-/-)CD28(-/-) and CD8(-/-)CD28(-/) deficient mice rejected fully allogeneic skin allografts at a tempo comparable with that in wild-type mice. Rejection proceeded despite significant reduction in alloreactive T-cell clone sizes suggesting the presence of a subset of T cells harnessing alternate CD28-independent costimulatory pathways. Blockade of CD40-CD154 and CD134-CD134L, but not ICOS-B7h pathways in combination significantly prolonged allograft survival in CD8(-/-)CD28(-/-) recipients and to a lesser extent in CD4(-/-)CD28(-/-) recipients. Prolongation in allograft survival was associated with reduced effector-memory T-cell generation, decreased allospecific Th1 cytokine generation and diminished alloreactive T-cell proliferation in vivo. In aggregate, the data identify these two pathways as critical mediators of CD28-independent rejection by CD4(+) and to a lesser extent CD8(+) T cells, and provide novel mechanistic insights into functions of novel T-cell co-stimulatory pathways in vivo.
Collapse
Affiliation(s)
- A Habicht
- Transplantation Research Center, Renal Division, Brigham & Women's Hospital and Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
The inherent ability of the host immune system to distinguish between self- and non-self forms the basis of allorecognition. T lymphocytes constitute the most important effector arm of allorecognition. Here we describe the fundamentals of direct and indirect pathways by which allopeptides are presented to effector T cells. The nature of allopeptides presented along with tolerogenic strategies like altered peptide ligands and intra- or extra-thymic allopeptide inoculation are discussed. In addition, we speculate on the potential of regulatory T cells to modulate alloimmune responses.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery, Washington University School of Medicine, Box 8109-3328 CSRB, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | | |
Collapse
|
28
|
Robertson NJ, Chai JG, Millrain M, Scott D, Hashim F, Manktelow E, Lemonnier F, Simpson E, Dyson J. Natural Regulation of Immunity to Minor Histocompatibility Antigens. THE JOURNAL OF IMMUNOLOGY 2007; 178:3558-65. [PMID: 17339452 DOI: 10.4049/jimmunol.178.6.3558] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
MHC-matched hemopoietic stem cell transplantation is commonly used for the treatment of some forms of leukemia. Conditioning regimens before transplant act to reduce the burden of leukemic cells and the graft-vs-leukemia (GvL) effect can eliminate residual disease. The GvL effect results largely from the recognition of minor histocompatibility Ags by donor T cells on recipient tissues. These Ags are generally widely expressed and also provoke graft-vs-host (GvH) disease. Manipulation of immunity to promote GvL while curtailing GvH would greatly improve clinical outcome. To develop strategies that may achieve this, the parameters which control immunity to minor histocompatibility Ags need to be defined. In this study, we have analyzed responses to the mouse HY minor histocompatibility Ag using hemopoietic cell and skin grafts as surrogate GvL and GvH targets, respectively. We show that natural regulation of CD8 T cell responses to HY operates at multiple levels. First, CD4 T cell help is required for primary CD8 responses directed at hemopoietic cells. However, although CD4 T cells of H2(k) mouse strains recognize HY, they provide ineffective help associated with a proportion of recipients developing tolerance. This was further investigated using TCR-transgenic mice which revealed H2(k)-restricted HY-specific CD4 T cells are highly susceptible to regulation by CD25(+) regulatory T cells which expand in tolerant recipients. A second level of regulation, operating in the context of skin grafts, involves direct inhibition of CD8 T cell responses by CD94/NKG2 engagement of the nonclassical MHC class I molecule Qa1.
Collapse
Affiliation(s)
- Nathan J Robertson
- Transplantation Biology Group, Department of Immunology, Imperial College London, Hammersmith Hospital, Du Cane Road, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Stewart H, Ravanan R, Smith R. In vivo models for the study of transplantation tolerance. Methods Mol Biol 2007; 380:337-46. [PMID: 17876103 DOI: 10.1007/978-1-59745-395-0_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Experimental models of transplantation remain essential tools for the study of immunological tolerance. The immunological mechanisms resulting in acute allograft rejection may differ depending on the tissue transplanted and the antigenic mismatch between donor and recipient. Murine skin grafting is a model frequently used to study transplantation tolerance because it is readily learned and is not time consuming. Second grafts can be performed easily to confirm the induction of tolerance and its antigenic specificity. However, these grafts are strongly immunogenic and secondarily revascularized and may not, therefore, reproduce the conditions operating in clinical transplantation of primarily revascularized organs, such as the kidney and heart. Experimental rodent models of revascularized solid organ transplantation have been established, but given the technical skills required to perform them, are beyond the scope of this chapter. However, cellular transplants, including the use of islets, are of interest as they are being developed clinically and can be reproduced experimentally. Here, we describe the technique of renal subcapsular transplantation of pancreatic islets of Langerhans.
Collapse
|
30
|
Garrod KR, Chang CK, Liu FC, Brennan TV, Foster RD, Kang SM. Targeted Lymphoid Homing of Dendritic Cells Is Required for Prolongation of Allograft Survival. THE JOURNAL OF IMMUNOLOGY 2006; 177:863-8. [DOI: 10.4049/jimmunol.177.2.863] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Mouse model for analysis of non-MHC genes that influence allogeneic response: recombinant congenic strains of OcB/Dem series that carry identical H2 locus. Open Life Sci 2006. [DOI: 10.2478/s11535-006-0002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractAlloreactivity is the strongest known primary immune response. Its clinical manifestations are graft rejection, graft-versus-host disease and graft-versus-leukemia effect. The strongest stimulation by allogeneic cells is due to incompatibility at the major histocompatibility complex (MHC) genes. However, the non-MHC genes also participate in allogeneic response. Here we present a mouse model for study of the role of non-MHC genes in regulation of alloreactivity and show that they besides encoding antigens also regulate the responsiveness. Recombinant congenic strains (RCS) of O20/A (O20)-c-B10.O20/Dem (OcB/Dem) series have been derived from the parental strains O20 and B10.O20, which carry identical MHC haplotypes (H2pz) and therefore their differences in alloantigen response depend only on non-MHC genes. We have tested a MLR response by spleen cells of the strains O20, B10.O20, and 16 OcB/Dem strains through stimulation by cells from strains C57BL/10 (H2b), BALB/c (H2d), CBA (H2k), and DBA/1 (H2q) alloantigens. Proliferative response of O20, B10.O20 and OcB/Dem strains to these four alloantigens exhibited a similar but not completely identical pattern of reactivity. The responses to different alloantigens were highly correlated: C57BL/10-BALB/c r = 0.87, C57BL/10-CBA r = 0.84, C57BL/10-DBA/1 r = 0.83. Cluster analysis of the responses by O20, B10.O20, and OcB mice identified groups of strains with distinct patterns of response. This data shows that two main types of genes influence MLR: 1. structural genes for major and minor alloantigens and 2. genes regulating T-cell receptor signal transduction or mediating costimulatory signals by antigen-presenting cells.
Collapse
|