1
|
Rubino G, Yörük E. Immunosenescence, immunotolerance and rejection: clinical aspects in solid organ transplantation. Transpl Immunol 2024; 86:102068. [PMID: 38844001 DOI: 10.1016/j.trim.2024.102068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 07/21/2024]
Abstract
As a consequence of increased lifespan and rising number of elderly individuals developing end-stage organ disease, the higher demand for organs along with a growing availability for organs from older donors pose new challenges for transplantation. During aging, dynamic adaptations in the functionality and structure of the biological systems occur. Consistently, immunosenescence (IS) accounts for polydysfunctions within the lymphocyte subsets, and the onset of a basal but persistent systemic inflammation characterized by elevated levels of pro-inflammatory mediators. There is an emerging consensus about a causative link between such hallmarks and increased susceptibility to morbidities and mortality, however the role of IS in solid organ transplantation (SOT) remains loosely addressed. Dissecting the immune-architecture of immunologically-privileged sites may prompt novel insights to extend allograft survival. A deeper comprehension of IS in SOT might unveil key standpoints for the clinical management of transplanted patients.
Collapse
Affiliation(s)
- Graziella Rubino
- University Hospital Tübingen, Department of Tropical Medicine, Wilhelmstraße 27, 72074 Tübingen, Germany; Institute for Transfusion Medicine, University Ulm and Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, 89081 Ulm, Germany.
| | - Efdal Yörük
- Berit Klinik, Gastrointestinal Center, Florastrasse 1, 9403 Goldach, Switzerland; University Hospital Tübingen, Department of Ophthalmology, Elfriede-Alhorn-Straße 7, 72076 Tübingen, Germany
| |
Collapse
|
2
|
Xu J, Ma T, Deng G, Zhuang J, Li C, Wang S, Dai C, Zhou X, Shan Z, Qi Z. Inhibition of C-X-C motif chemokine 10 reduces graft loss mediated by memory CD8 + T cells in a rat cardiac re-transplant model. Exp Ther Med 2017; 15:1560-1567. [PMID: 29434741 DOI: 10.3892/etm.2017.5585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 06/08/2017] [Indexed: 11/05/2022] Open
Abstract
The interaction of chemokine (C-X-C motif) ligand 10 (CXCL10) with its receptor (CXCR3) is a critical process in recruiting donor reactive T cells to a graft and alloantigen-specific memory T (Tm) cells exert a principal function in promoting graft dysfunction during accelerated cardiac rejection. However, whether CXCL10 chemokine exerts any effects on acute accelerated rejection mediated by CD8+ Tm cells in a re-transplant model has remained elusive. The present study established a cardiac transplant model by advanced microsurgery technology and improved organ storage. A novel rat model of cardiac re-transplantation was established at 40 days following primary heart transplant. The experiment included two parts, and when models were established, the rats were divided into two groups: Primary cardiac transplant (HTx) and re-transplantation without treatment (HRTx). In part 1, recipients from part 2, including re-transplantation without treatment (HRTx+NS) and re-transplantation treated with anti-CXCL10 antibodies (500 µg every other day by intraperitoneal injection; HRTx+CXCL10 Abs group). The graft survival time was observed and graft infiltration by inflammatory cells was assessed via histology of cardiac graft sections; in addition, the gene expression and the serum concentration of CXCL10 in each group was assessed. Indexes such as rejection-associated cytokines were assayed by reverse-transcription quantitative PCR and ELISA kits, and flow cytometry of splenocytes was used to detect Tm cells in the re-transplantation groups. The results demonstrated that level of CXCL10 was significantly increased and the graft mean survival time was shortened accompanied with aggravated lymphocyte cell infiltration in the HRTx group when compared that in the HTx group; in addition, the serum levels and mRNA expression of interleukin (IL)-2 and interferon (IFN)-γ were increased, while transforming growth factor (TGF)-β was decreased in the HRTx group. Furthermore, neutralization of CXCL10 prolonged the graft mean survival time and delayed accelerated rejection. Compared with that in the HRTx+NS group, serum levels and graft tissue mRNA expression of IFN-γ and IL-2 were decreased in the HRTx+CXCL10 Abs group, while TGF-β mRNA was significantly increased but the serum concentration was not significantly affected. In addition, there was no difference in IL-10 between the two groups, while delayed accelerated rejection paralleled with inflammatory cell infiltration decreased and the proliferation and differentiation of CD8+ Tm cells in secondary lymphoid organs were reduced in the HRTx+CXCL10 Abs group vs. those in the HRTx+NS group. The present study demonstrated that CXCL10 had a crucial role in cardiac transplantation and re-transplantation, and that treatment with CXCL10 antibodies delays accelerated acute rejection mediated by Tm cells in a rat model of cardiac re-transplantation.
Collapse
Affiliation(s)
- Jiacheng Xu
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Teng Ma
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Guorong Deng
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Jiawei Zhuang
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Cheng Li
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Shaohu Wang
- Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Chen Dai
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Xiaobiao Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhonggui Shan
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhongquan Qi
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| |
Collapse
|
3
|
Luan J, Zhao Y, Zhang Y, Miao J, Li J, Chen ZN, Zhu P. CD147 blockade as a potential and novel treatment of graft rejection. Mol Med Rep 2017; 16:4593-4602. [PMID: 28849101 PMCID: PMC5647014 DOI: 10.3892/mmr.2017.7201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/01/2017] [Indexed: 11/25/2022] Open
Abstract
Cluster of differentiation (CD)147 is highly involved in the T cell activation process. High CD147 expression is observed on the surfaces of activated T cells, particularly CD4+ T cells. In organ transplantation, it is important to prevent graft rejection resulting from the excessive activation of T cells, particularly CD4+ T cells, which exhibit a key role in amplifying the immune response. The present study aimed to investigate the effects of CD147 blockade in vitro and in vivo and used a transplant rejection system to assess the feasibility of utilizing CD147 antibody-based immunosuppressant drugs for the treatment of graft rejection. The effects of CD147 antibodies were evaluated on lymphocyte proliferation stimulated by phytohemagglutinin or CD3/CD28 magnetic beads and in a one-way mixed lymphocyte reaction (MLR) system in vitro. For the in vivo analysis, an allogeneic skin transplantation mouse model was used. CD147 antibodies were effective against lymphocytes, particularly CD4+T lymphocytes, and were additionally effective in the one-way MLR system. In the allogeneic skin transplantation mouse model, the survival of transplanted skin was extended in the CD147 antibody-treated group. Furthermore, the level of inflammatory cell infiltration in transplanted skin was reduced. CD147 blockade decreased the serum levels of interleukin (IL)-17 and the proportions of peripheral blood CD4+ and CD8+ memory T cells. The data demonstrated that CD147 blockade suppressed skin graft rejection, primarily by suppressing CD4+T and memory T cell proliferation, indicating that CD147 exhibits great potential as a target of immunosuppressant drugs.
Collapse
Affiliation(s)
- Jing Luan
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yu Zhao
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yang Zhang
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jinlin Miao
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jia Li
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhi-Nan Chen
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ping Zhu
- National Translational Science Center for Molecular Medicine, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
4
|
Du G, Yang N, Gong W, Fang Y, He J, Zhou N, Lu X, Zhao Y. CD8 + effector memory T cells induce acute rejection of allogeneic heart retransplants in mice possibly through activating expression of inflammatory cytokines. Exp Cell Res 2017; 355:1-8. [PMID: 28232114 DOI: 10.1016/j.yexcr.2017.02.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/15/2017] [Accepted: 02/20/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND To investigate the effects of CD8+ memory T (Tm) cells and CD8+ effector memory T (Tem) cells on the results of allogeneic heart retransplantations performed in mice. METHODS A skin transplantation model was used to generate sensitized splenic CD8+ Tem cells for infusion into BALB/c mice. One week after infusion, the BALB/c mice underwent allogeneic heart transplantation in the abdominal cavity. Cyclosporin A was administered via intraperitoneal injection starting one day prior to transplantation to arrest immunological rejection of the transplanted heart. The effects of sensitized CD8+ Tem cells on allogeneic heart graft rejection were examined by monitoring survival of the transplanted hearts, the infiltration of effector memory CD8+ T cells into myocardium, and expressions of inflammatory cytokines in blood serum. RESULTS Adoptive transfer of sensitized CD8+ Tem cells prior to transplantation induced an acute rejection response which decreased the survival of transplanted hearts. The rejection response was accompanied by an infiltration of CD8+ Tem cells into the transplanted myocardial tissue. Additionally, infusion of sensitized CD8+ Tem cells induced markedly increased expressions of IL-2 and IFN-γ, and decreased expression of TGF-β in the transplanted hearts, as well as higher levels of IFN-γ and CXCL-9 in blood serum. CONCLUSIONS The infusion of sensitized CD8+ Tem cells induced an acute graft rejection response and decreased the survival of grafted hearts by regulating the expressions of inflammatory cytokines including CXCL-9, IL-2, and INF-γ. Cyclosporin A had no therapeutic effect on the graft rejection response induced by sensitized CD8+ Tem cells.
Collapse
Affiliation(s)
- Gang Du
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China; The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; The Department of Immunology, Guangxi Medical University, Nanning 530021, China
| | - Nuo Yang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China; The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; The Department of Immunology, Guangxi Medical University, Nanning 530021, China
| | - Wenlin Gong
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China
| | - Yuan Fang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China; The Department of Immunology, Guangxi Medical University, Nanning 530021, China
| | - Jian He
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China; The Department of Immunology, Guangxi Medical University, Nanning 530021, China
| | - Nuo Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China
| | - Xiaoling Lu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China; The Department of Immunology, Guangxi Medical University, Nanning 530021, China.
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, China; Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Guangxi Medical University, Nanning 530021, China; The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; The Department of Immunology, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
5
|
Merino D, San Segundo D, Medina JM, Rodrigo E, Asensio E, Irure J, Fernández-Fresnedo G, Arias MA, López-Hoyos M. Different in vitro proliferation and cytokine-production inhibition of memory T-cell subsets after calcineurin and mammalian target of rapamycin inhibitors treatment. Immunology 2016; 148:206-15. [PMID: 26931075 DOI: 10.1111/imm.12603] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/10/2016] [Accepted: 02/24/2016] [Indexed: 12/14/2022] Open
Abstract
Calcineurin inhibitors (CNI) and mammalian target of rapamycin inhibitors (mTORi) are the main immunosuppressants used for long-term maintenance therapy in transplant recipients to avoid acute rejection episodes. Both groups of immunosuppressants have wide effects and are focused against the T cells, although different impacts on specific T-cell subsets, such as regulatory T cells, have been demonstrated. A greater knowledge of the impact of immunosuppression on the cellular components involved in allograft rejection could facilitate decisions for individualized immunosuppression when an acute rejection event is suspected. Memory T cells have recently gained focus because they might induce a more potent response compared with naive cells. The impact of immunosuppressants on different memory T-cell subsets remains unclear. In the present study, we have studied the specific impact of CNI (tacrolimus) and mTORi (rapamycin and everolimus) over memory and naive CD4(+) T cells. To do so, we have analysed the proliferation, phenotypic changes and cytokine synthesis in vitro in the presence of these immunosuppressants. The present work shows a more potent effect of CNI on proliferation and cytokine production in naive and memory T cells. However, the mTORi permit the differentiation of naive T cells to the memory phenotype and allow the production of interleukin-2. Taken together, our data show evidence to support the combined use of CNI and mTORi in transplant immunosuppression.
Collapse
Affiliation(s)
- David Merino
- Autoimmunity and Transplant Group-IDIVAL, Santander, Spain
| | - David San Segundo
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Juan M Medina
- Autoimmunity and Transplant Group-IDIVAL, Santander, Spain
| | - Emilio Rodrigo
- Nephrology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Esther Asensio
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Juan Irure
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | | | - Manuel A Arias
- Nephrology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| |
Collapse
|
6
|
Scottà C, Fanelli G, Hoong SJ, Romano M, Lamperti EN, Sukthankar M, Guggino G, Fazekasova H, Ratnasothy K, Becker PD, Afzali B, Lechler RI, Lombardi G. Impact of immunosuppressive drugs on the therapeutic efficacy of ex vivo expanded human regulatory T cells. Haematologica 2015; 101:91-100. [PMID: 26471483 DOI: 10.3324/haematol.2015.128934] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 10/09/2015] [Indexed: 12/21/2022] Open
Abstract
Immunosuppressive drugs in clinical transplantation are necessary to inhibit the immune response to donor antigens. Although they are effective in controlling acute rejection, they do not prevent long-term transplant loss from chronic rejection. In addition, immunosuppressive drugs have adverse side effects, including increased rate of infections and malignancies. Adoptive cell therapy with human Tregs represents a promising strategy for the induction of transplantation tolerance. Phase I/II clinical trials in transplanted patients are already underway, involving the infusion of Tregs alongside concurrent immunosuppressive drugs. However, it remains to be determined whether the presence of immunosuppressive drugs negatively impacts Treg function and stability. We tested in vitro and in vivo the effects of tacrolimus, mycophenolate and methylprednisolone (major ISDs used in transplantation) on ex vivo expanded, rapamycin-treated human Tregs. The in vitro results showed that these drugs had no effect on phenotype, function and stability of Tregs, although tacrolimus affected the expression of chemokine receptors and IL-10 production. However, viability and proliferative capacity were reduced in a dose-dependent manner by all the three drugs. The in vivo experiments using a humanized mouse model confirmed the in vitro results. However, treatment of mice with only rapamycin maintained the viability, function and proliferative ability of adoptively transferred Tregs. Taken together, our results suggest that the key functions of ex vivo expanded Tregs are not affected by a concurrent immunosuppressive therapy. However, the choice of the drug combination and their timing and dosing should be considered as an essential component to induce and maintain tolerance by Treg.
Collapse
Affiliation(s)
- Cristiano Scottà
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giorgia Fanelli
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Sec Julie Hoong
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Marco Romano
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, Italy
| | - Estefania Nova Lamperti
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Mitalee Sukthankar
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giuliana Guggino
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Italy
| | - Henrieta Fazekasova
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Kulachelvy Ratnasothy
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Pablo D Becker
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Behdad Afzali
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert I Lechler
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giovanna Lombardi
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| |
Collapse
|
7
|
Arsenic trioxide inhibits accelerated allograft rejection mediated by alloreactive CD8(+) memory T cells and prolongs allograft survival time. Transpl Immunol 2015; 33:30-6. [PMID: 26044521 DOI: 10.1016/j.trim.2015.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/28/2015] [Accepted: 05/26/2015] [Indexed: 11/20/2022]
Abstract
CD8(+) memory T (Tm) cells are a significant barrier to transplant tolerance induction in alloantigen-primed recipients, and are insensitive to existing clinical immunosuppressants. Here, we studied the inhibition of CD8(+) Tm cells by arsenic trioxide (As2O3) for the first time. Alloantigen-primed CD8(+) Tm cells were transferred to T cell immunodeficient nude mice. The mice were subjected to heart allotransplantation, and treated with As2O3. The transplant survival time was determined, and the inhibitory effects of As2O3 on CD8(+) Tm cell-mediated immune rejection were assessed through serological studies and inspection of the transplanted heart and lymphoid organs. We found that As2O3 treatment prolonged the mean survival time of the graft and reduced the number of CD8(+) Tm cells in the spleen and lymph nodes. The expression of the genes encoding interleukin (IL)-2, and IFN-γ was reduced, while expression of IL-10 and transforming growth factor-β was increased in the transplant. Our findings show that As2O3 treatment inhibits allograft rejection mediated by alloreactive CD8(+) Tm cells in the mouse heart transplantation model.
Collapse
|
8
|
Ma T, Xu J, Zhuang J, Zhou X, Lin L, Shan Z, Qi Z. Combination of C-X-C motif chemokine 9 and C-X-C motif chemokine 10 antibodies with FTY720 prolongs the survival of cardiac retransplantation allografts in a mouse model. Exp Ther Med 2015; 9:1006-1012. [PMID: 25667668 PMCID: PMC4316950 DOI: 10.3892/etm.2015.2204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 11/17/2014] [Indexed: 12/14/2022] Open
Abstract
The upregulation of chemokine genes and the subsequent T-lymphocyte recruitment to the graft are early events in the development of acute cardiac transplant rejection or cardiac allograft vasculopathy. In the present study, a combined immunosuppressive regimen of C-X-C motif chemokine 9 (CXCL9) antibody (Ab), CXCL10 Ab and FTY720 was used in order to reduce the infiltration of memory T lymphocytes and prolong graft survival in a retransplantation murine model. BALB/c donor hearts were transplanted heterotopically into C57BL/6 mice at day 28 after skin transplantation. The mice were divided into four groups: i) Control (normal saline), ii) CXCL9 Ab and CXCL10 Ab [150 μg; once daily (qd); intraperitoneal (ip)], iii) FTY720 (0.2 mg/day; qd; ip) and iv) combined (2 mg/kg/day; qd; ip). Measurements of the median survival time of the cardiac grafts, histological examination, reverse transcription-quantitative polymerase chain reaction analysis, enzyme-linked immunosorbent assay and a mixed lymphocyte reaction were performed. The median graft survival time of the combined group was prolonged (9.3 days) compared with that of the control group (3.5 days) (P<0.001). Histological examination revealed that the combined treatment group graft rejection pathological score was 0.50, while the control group score was 3.62 (P<0.001). In addition, the gene expression level of interleukin (IL)-2 was significantly lower and the levels of IL-10 and transforming growth factor-β (TGF-β) were significantly higher in the combined group compared with those in the control group (P<0.001). Furthermore, the serum concentration levels of IL-2 and interferon-γ (IFN-γ) were significantly lower (P<0.001) and the concentration of IL-10 was significantly higher (P<0.05) in the combined group compared with those in the control group. In the mixed lymphocyte reaction, T-cell proliferation was found to be significantly lower in the combined treatment group than that in the control group (P<0.001). In conclusion, treatment with CXCL9 Ab and CXCL10 Ab or FTY720 reduced the graft infiltration of inflammatory cells, inhibited T-cell proliferation and prolonged graft survival. The combined treatment regimen of CXCL9 Ab, CXCL10 Ab and FTY720 was found to significantly reduce the infiltration of inflammatory cells in the graft and prolong graft survival.
Collapse
Affiliation(s)
- Teng Ma
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Jiacheng Xu
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Jiawei Zhuang
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Xiaobiao Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Lianfeng Lin
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhonggui Shan
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhongquan Qi
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| |
Collapse
|
9
|
Zhuang J, Shan Z, Ma T, Li C, Qiu S, Zhou X, Lin L, Qi Z. CXCL9 and CXCL10 accelerate acute transplant rejection mediated by alloreactive memory T cells in a mouse retransplantation model. Exp Ther Med 2014; 8:237-242. [PMID: 24944628 PMCID: PMC4061216 DOI: 10.3892/etm.2014.1714] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 05/02/2014] [Indexed: 11/21/2022] Open
Abstract
C-X-C motif chemokine ligand (CXCL) 9 and CXCL10 play key roles in the initiation and development of acute transplant rejection. Previously, higher levels of RANTES expression and secretion were demonstrated in retransplantation or T-cell memory-transfer models. In the present study, the effect of the chemokines, CXCL9 and CXCL10, were investigated in a mouse retransplantation model. BALB/c mice were used as donors, while C57BL/6 mice were used as recipients. In the experimental groups, a heterotopic heart transplantation was performed six weeks following skin grafting. In the control groups, a heterotopic heart transplantation was performed without skin grafting. Untreated mice served as blank controls. The mean graft survival time of the heterotopic heart transplantations was 7.7 days in the experimental group (n=6), as compared with 3.25 days in the control group (n=6; P<0.001). On day three following cardiac transplantation, histological evaluation of the grafts revealed a higher International Society for Heart & Lung Transplantation grade in the experimental group as compared with the control group. In addition, gene expression and serum concentrations of CXCL9, CXCL10, interferon-γ, and interleukin-2 were markedly higher in the experimental group when compared with the control group. Differences between the levels of CXCL9 and CXCL10 in the pre- and post-transplant mice indicated that the chemokines may serve as possible biomarkers to predict acute rejection. The results of the present study demonstrated that CXCL9 and CXCL10 play a critical role in transplantation and retransplantation. High levels of these cytokines during the pre-transplant period may lead to extensive acute rejection. Thus, the observations enhance the understanding of the mechanism underlying the increased expression and secretion of CXCL9 and CXCL10 by alloreactive memory T cells.
Collapse
Affiliation(s)
- Jiawei Zhuang
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhonggui Shan
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Teng Ma
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Chun Li
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Shuiwei Qiu
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Xiaobiao Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Lianfeng Lin
- Department of Cardiac Surgery, The First Affiliated Hospital, Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhongquan Qi
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen, Fujian 361005, P.R. China
| |
Collapse
|
10
|
Substantial proliferation of human renal tubular epithelial cell-reactive CD4+CD28null memory T cells, which is resistant to tacrolimus and everolimus. Transplantation 2014; 97:47-55. [PMID: 24157471 DOI: 10.1097/01.tp.0000435697.31148.b2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND In spite of maintenance treatment with immunosuppressive drugs, tubulitis still occurs and can lead to structural kidney graft damage. We hypothesize that human renal tubular epithelial cells (TECs) trigger selective proliferation of recipient T-cell subsets with variable sensitivity to immunosuppressive drugs. METHODS Recipient peripheral blood mononuclear cells were cocultured with donor-derived TECs for 7 days. The proliferation of the total CD4 T-cell pool was assessed. Next, we analyzed which CD4 T-cell subset proliferated and how this response was affected by tacrolimus, everolimus, prednisolone, and mycophenolic acid (MPA) in clinically relevant concentrations. RESULTS CD4 T-cell proliferation upon TEC encounter was mainly executed by memory T cells. Interestingly, 38%±7% of the proliferating CD4 T-cell pool showed a CD28 phenotype. These proliferating CD4CD28 memory T cells produced high levels of interferon-γ, tumor necrosis factor-α, and the cytolitic protease granzyme B. TEC-reactive CD4 T-cell proliferation was significantly suppressed by tacrolimus, everolimus, prednisolone, and MPA (P<0.05). Surprisingly and in contrast to prednisolone and MPA, neither tacrolimus nor everolimus could inhibit the CD4CD28 T-cell proliferative response. CONCLUSION Our data show substantial proliferation of TEC-reactive CD4CD28 memory T cells, which are resistant to tacrolimus and everolimus. This phenomenon might play an important mechanistic role during cellular rejection under full immunosuppression.
Collapse
|
11
|
Yan G, Xi Y, Xu S, Lin Y, Chen J, Dai H, Xia J, Li C, Li Q, Li Z, Qi Z. Inhibition of accelerated rejection mediated by alloreactive CD4⁺ memory T cells and prolonged allograft survival by arsenic trioxide. Immunol Invest 2014; 42:438-54. [PMID: 23802174 DOI: 10.3109/08820139.2013.801986] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The aim of this study was to evaluate and determine the potential mechanisms of As₂O₃ in accelerated rejection mediated by alloreactive CD4⁺ memory T cells. Vascularized heterotopic cardiac transplantation from C57BL/6 mice to nude mice (pre-transferred CD4⁺ memory T cells) was performed on Day 0, and As₂O₃ was administered to recipient mice from Day 0 to 10. As a result, As₂O₃ could reduce the proliferation of allo-primed CD4⁺ memory T cells in vitro in MLR and the baseline rate of proliferation was restored by the addition of exogenous IL-2. In vivo, compared with the control[+] group, the mean survival time of cardiac allografts in the As₂O₃ group was prolonged from 5.8 ± 0.7 to 14.2 ± 2.5 days. Five days after transplantation, the relative gene expression of IL-2, IFN-γ and Foxp3 was reduced in the grafts by As₂O₃ treatment, but the expression of IL-10 and TGF-β was increased. Correspondingly, the proportions of CD4⁺ T cells, CD4⁺ memory T cells and regulatory T cells (Tregs), both in recipient spleens and lymph nodes, were lowered. These results indicate the potential of As2O3 as a novel immunosuppressant targeting CD4⁺ memory T cells.
Collapse
Affiliation(s)
- Guoliang Yan
- Basic Medical Department of Medical College, Xiamen University, Xiamen City, Fujian Province, P. R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Role of regulated upon activation normal T-cell expressed and secreted in a model of retransplantation acute rejection mediated by alloreactive memory CD4+ T cells. Transplant Proc 2013; 45:546-51. [PMID: 23498790 DOI: 10.1016/j.transproceed.2012.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/13/2012] [Indexed: 11/23/2022]
Abstract
BACKGROUND It is unknown what role Regulated Upon Activation Normal T-Cell Exposed and Secreted may play in retransplantation or T-cell memory-transfer models. The experiment observed the influence of the chemokine RANTES in a mouse model of acute cardiac allograft rejection induced by adoptive transfer of alloreactive CD4(+) memory T (Tm) cells. METHODS Alloreactive CD4(+) Tm cells from spleens of skin-grafted C57BL/6 were adoptively transferred to naïve C57BL/6 recipients prior to heterotopic heart transplantation. We measured the median survival time of cardiac grafts and performed some tests. RESULTS Spleens from skin-grafted C57BL/6 contained 26.83% CD4(+) Tm cells. The median graft survival time of heterotopic heart transplantations (n = 6) was 5.17 ± 0.17 days for hosts receiving CD4(+) Tm cells compared with 7.76 ± 0.21 days among controls (n = 6; P < .001). The mean rejection activity in histological sections of cardiac allografts at day 5 postgrafting was 3.92 ± 0.08 in the CD4(+) Tm cell recipient group (n = 6) compared with 2.67 ± 0.14 in the controls (n = 6; P < .001). Gene expression of Ccl5, interferon (IFN)-γ and interleukin2 was significantly higher among CD4(+) Tm recipients compared with controls. Serum concentrations of RANTES and IFN-γ were higher in the heterotopic heart transplantation group receiving CD4(+) Tm compared with controls. CONCLUSIONS Alloreactive CD4(+) Tm cells contribute to increased expression and secretion of RANTES, and to the Tm and other inflammatory cells migration into the graft.
Collapse
|
13
|
|
14
|
Huang H, He J, Teng X, Yu Y, Ye W, Hu Y, Shen Z. Combined intrathymic and intravenous injection of mesenchymal stem cells can prolong the survival of rat cardiac allograft associated with decrease in miR-155 expression. J Surg Res 2013; 185:896-903. [PMID: 23870834 DOI: 10.1016/j.jss.2013.06.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/08/2013] [Accepted: 06/06/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have the potential to improve graft outcomes and promote allograft tolerance. In this study, we examined the effects and mechanism of combined intrathymic (i.t.) and intravenous (i.v.) injection of MSCs on the survival of transplanted hearts in a rat allograft model. METHODS Recipient Sprague-Dawley rats were transplanted with hearts from Wistar rats. Wistar rat MSCs were infused via i.t. or i.v. or combined i.t. and i.v. (i.t./i.v.) injection at designated intervals. In vitro mixed lymphocyte reaction assays were performed to assess the immunosuppressive capacity of MSCs. Mesenchymal stem cell surface markers and CD4+, CD25+, and Foxp3+ T-cells in the peripheral blood were detected using flow cytometry analysis. The expression of microRNAs and cytokines in graft infiltrating lymphocytes was analyzed by real-time polymerase chain reaction. RESULTS The MSCs cultured in vitro had multipotential differentiation capacity. Mixed lymphocyte reaction assays showed that donor-derived MSCs could not stimulate a proliferative response of recipient lymphocytes and could markedly suppress T-cell responses. Survival of the allografts was significantly prolonged by administration of i.t./i.v. injection of MSCs compared with controls, with a mean survival of 32.2 versus 6.5 d, respectively. Compared with the syngeneic groups posttransplant, miR-155 expression was significantly increased in the allogeneic group, and could be restored by injection of MSCs, especially i.t./i.v. injection of MSCs. Moreover, i.t./i.v. injection of MSCs decreased the level of interleukin (IL)-2 and interferon-gamma, but increased the levels of IL-4 and IL-10 in the allogeneic group. More important, i.t./i.v. injection of MSCs was the best way to increase the percentage of CD4+, CD25+, and Foxp3+ T-cell peripheral blood. CONCLUSIONS Our results indicated that i.t./i.v. injection of MSCs can prolong the survival of rat cardiac allograft, which may be associated with down-regulating miR-155 expression, a shift in the Th1/Th2 balance, and up-regulation of Treg cells expression.
Collapse
Affiliation(s)
- Haoyue Huang
- Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Zhang G, Chen G, Liu L, Qiu J, Chen L. A novel accelerated rejection model for mouse cardiac transplantation involving presensitization with donor splenocytes. J Surg Res 2013; 181:146-55. [PMID: 22683077 DOI: 10.1016/j.jss.2012.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Revised: 04/27/2012] [Accepted: 05/08/2012] [Indexed: 11/16/2022]
Abstract
BACKGROUND Accelerated rejection is a type of refractory rejection. Animal models of accelerated rejection are widely employed in research on transplant immunity. METHODS We divided 36 C57BL/6 mice into six groups that underwent heart transplant. To select the ideal number of splenocytes for the presensitization to induce accelerated rejection, were transferred 0.5 × 10(7), 1 × 10(7), 5 × 10(7), or 10 × 10(7) donor splenocytes 7 d before transplantation. We confirmed successful presensitization by increases in donor-reactive antibodies. We performed 12 additional heart transplants in the accelerated rejection group and the control groups for a histological examination, immunohistochemical staining for C3d, and a splenocyte test using flow cytometry. RESULTS The transfer of 5 × 10(7) donor splenocytes effectively and efficiently induced an accelerated rejection in the BALB/c→C57BL/6 heart transplant, with an allograft survival time that was decreased from 7.4 ± 0.5 d to 3.5 ± 0.8 d compared with the allogenic controls (P < 0.05, log-rank test). An analysis of this model indicated that compared with acute rejection, the number of donor-reactive antibodies was significantly increased, and the proportions of effector memory CD8(+) T cells and plasmacytes in the spleen were significantly increased (7.81% ± 1.2% versus 2.96% ± 1.0%, P = 0.006; 1.27% ± 0.13% versus 0.71% ± 0.22%, P = 0.018, respectively). We found the histological characteristics of both cellular and humoral rejection in the accelerated rejection model. CONCLUSIONS Presensitization via the transfer of donor splenocytes facilitates the establishment of an accelerated rejection model. Our findings with this model indicate that humoral rejection and cellular rejection are coexistent, and that the proportions of effector memory CD8(+) T cells and plasmacytes in the spleen increase significantly during accelerated rejection.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Organ Transplantation, First Affiliated Hospital of Sun Yat-sen University, Guang Zhou, Guang Dong Province, China
| | | | | | | | | |
Collapse
|
16
|
Yan G, Xi Y, Xu S, Chen J, Lin Y, Dai H, Cheng P, Xiao H, Liu Z, Qi Z. Inhibiting accelerated rejection mediated by alloreactive CD4+ memory T cells and prolonging allograft survival by 1α,25-dihydroxyvitamin D3 in nude mice. Immunol Lett 2013; 149:54-61. [DOI: 10.1016/j.imlet.2012.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 10/30/2012] [Accepted: 11/01/2012] [Indexed: 01/13/2023]
|
17
|
Functional Analysis of Alloreactive Memory CD4+ T Cells Derived from Skin Transplantation Recipient and Naïve CD4+ T Cells Derived from Untreated Mice. J Surg Res 2012; 176:649-56. [DOI: 10.1016/j.jss.2011.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 10/24/2011] [Accepted: 11/01/2011] [Indexed: 11/22/2022]
|
18
|
Sun Y, Chen X, Zhao J, Zou X, Li G, Li X, Shen B, Sun S. Combined use of rapamycin and leflunomide in prevention of acute cardiac allografts rejection in rats. Transpl Immunol 2012; 27:19-24. [DOI: 10.1016/j.trim.2012.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/31/2012] [Accepted: 04/08/2012] [Indexed: 01/11/2023]
|
19
|
Luo L, Sun Z, Luo G. Rapamycin is less fibrogenic than Cyclosporin A as demonstrated in a rat model of chronic allograft nephropathy. J Surg Res 2012; 179:e255-63. [PMID: 22480837 DOI: 10.1016/j.jss.2012.02.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 02/14/2012] [Accepted: 02/28/2012] [Indexed: 11/27/2022]
Abstract
BACKGROUND Cyclosporin A (CsA) is associated with significant chronic nephrotoxicity, which typically manifests as renal fibrosis. In contrast, rapamycin (RAPA) has been shown to inhibit fibrosis. This study sought to determine the effect of CsA and RAPA on the expression of connective tissue growth factor (CTGF) and E-cadherin in a rat kidney model of chronic allograft nephropathy. MATERIALS AND METHODS Left renal grafts from male Fisher (F344, RT1(1v1)) rats were orthotopically transplanted into Lewis (LEW, RT1(1)) rats. After transplantation, all recipients were given CsA 10 mg/kg(-1) d(-1) for 10 d and divided into three groups (n = 9/group): (1) vehicle, administered orally; (2) CsA, 6 mg/kg(-1) d(-1); (3) RAPA, 0.8 mg/kg(-1) d(-1). At 4, 8, and 12 wk posttransplantation, the kidney allografts were harvested and serum creatinine levels were measured. Connective tissue growth factor expression was determined using real-time polymerase chain reaction and Western blot. Kidney allografts sections also underwent hematoxylin-eosin and Masson trichrome staining, in addition to CTGF and E-cadherin immunostaining. RESULTS The serum creatinine levels were increased at 8 and 12 wk posttransplantation and were significantly lower in the RAPA group (P < 0.05). The Banff score also showed a significant decrease at 4, 8, and 12 wk (P < 0.05). CTGF messenger ribonucleic acid and protein levels were significantly lower in the RAPA group (P < 0.05), whereas E-cadherin expression was higher in the RAPA group at 4, 8, and 12 wk (P < 0.05). Masson's trichrome staining showed a significant decrease in collagen deposition at 8 and 12 wk after RAPA treatment. CONCLUSION RAPA can ameliorate fibrogenesis in kidney allografts by inhibiting epithelial-mesenchymal transition process, whereas CsA did not have this effect.
Collapse
Affiliation(s)
- Lei Luo
- Department of Research and Education, Guizhou Province People's Hospital, Guiyang, China
| | | | | |
Collapse
|
20
|
Bouchlaka MN, Redelman D, Murphy WJ. Immunotherapy following hematopoietic stem cell transplantation: potential for synergistic effects. Immunotherapy 2010; 2:399-418. [PMID: 20635904 DOI: 10.2217/imt.10.20] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a particularly important treatment for hematologic malignancies. Unfortunately, following allogeneic HSCT, graft-versus-host disease, immunosuppression and susceptibility to opportunistic infections remain among the most substantial problems restricting the efficacy and use of this procedure, particularly for cancer. Adoptive immunotherapy and/or manipulation of the graft offer ways to attack residual cancer as well as other transplant-related complications. Recent exciting discoveries have demonstrated that HSCT could be expanded to solid tissue cancers with profound effects on the effectiveness of adoptive immunotherapy. This review will provide a background regarding HSCT, discuss the complications that make it such a complex treatment procedure following up with current immunotherapeutic strategies and discuss emerging approaches in applying immunotherapy in HSCT for cancer.
Collapse
Affiliation(s)
- Myriam N Bouchlaka
- Department of Microbiology & Immunology, University of Nevada, Reno, NV, USA
| | | | | |
Collapse
|