1
|
Xie S, Xu C, Zhu Z, Qin C, Song X, Wang X, Xu W, Zhu M. Influence of CDC‑XM and HLA compatibility on clinical outcomes in kidney transplant recipients during the post‑operative recovery period: A retrospective analysis. Exp Ther Med 2025; 29:99. [PMID: 40165799 PMCID: PMC11955818 DOI: 10.3892/etm.2025.12849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/23/2025] [Indexed: 04/02/2025] Open
Abstract
Kidney transplantation remains the preferred treatment for patients with end-stage kidney failure. Complement-dependent cytotoxicity (CDC) crossmatch (CDC-XM) and human leukocyte antigen (HLA) typing are two important methods of donor-recipient matching prior to kidney transplantation. The purpose of the present study was to explore the effects of CDC-XM levels and HLA matching on early post-operative clinical outcomes in kidney transplant recipients. A total of 112 consecutive recipients who underwent allogeneic kidney transplantation were selected and their data collected, including pre-operative general information, indicators associated with renal function, red blood cell and white blood cell counts, the blood glucose level at each follow-up time point and both the incidence of adverse events following transplantation and their risk factors. During the follow-up period, statistical methods were used to compare and systematically analyze the differences in clinical indicators and adverse events between each groups. In each different groups that were assigned for the CDC-XM levels and HLA matching, the differences in the clinical indicators between the groups during the follow-up period were mainly centered on the first week post-transplantation, with the greatest differences being identified for the renal function-associated indicators, whereas the observed recovery was essentially comparable between 1-6 months. According to the multivariate analysis, recipients of age ≥40 years and with a BMI ≥25 tended to have an increased risk of delayed graft function (DGF), whereas the risk was reduced when the organ had been donated by a living donor and also when the number of HLA mismatches was 0-2. In conclusion, the present study showed that CDC-negativity and improved HLA matching help to promote the recovery of renal function in kidney transplant recipients during the early post-operative period. Patients who met the conditions of CDC-negativity and fewer HLA mismatches had faster and improved early postoperative recovery of renal function and a lower incidence of DGF. Furthermore, in a multifactorial analysis of DGF, recipient age, recipient BMI, donor type and HLA mismatch were found to be important risk factors for DGF.
Collapse
Affiliation(s)
- Siqi Xie
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Chonghe Xu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China
| | - Zhongqi Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Chao Qin
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xixi Song
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xin Wang
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Wei Xu
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Mei Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| |
Collapse
|
2
|
Pan W, Li S, Li K, Zhou P. Mesenchymal Stem Cells and Extracellular Vesicles: Therapeutic Potential in Organ Transplantation. Stem Cells Int 2024; 2024:2043550. [PMID: 38708382 PMCID: PMC11068458 DOI: 10.1155/2024/2043550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
At present, organ transplantation remains the most appropriate therapy for patients with end-stage organ failure. However, the field of organ transplantation is still facing many challenges, including the shortage of organ donors, graft function damage caused by organ metastasis, and antibody-mediated immune rejection. It is therefore urgently necessary to find new and effective treatment. Stem cell therapy has been regarded as a "regenerative medicine technology." Mesenchymal stem cells (MSCs), as the most common source of cells for stem cell therapy, play an important role in regulating innate and adaptive immune responses and have been widely used in clinical trials for the treatment of autoimmune and inflammatory diseases. Increasing evidence has shown that MSCs mainly rely on paracrine pathways to exert immunomodulatory functions. In addition, mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are the main components of paracrine substances of MSCs. Herein, an overview of the application of the function of MSCs and MSC-EVs in organ transplantation will focus on the progress reported in recent experimental and clinical findings and explore their uses for graft preconditioning and recipient immune tolerance regulation. Additionally, the limitations on the use of MSC and MSC-EVs are also discussed, covering the isolation of exosomes and preservation techniques. Finally, the opportunities and challenges for translating MSCs and MSC-EVs into clinical practice of organ transplantation are also evaluated.
Collapse
Affiliation(s)
- Wennuo Pan
- Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shaohan Li
- Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Kunsheng Li
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
3
|
Zhou AW, Jin J, Liu Y. Cellular strategies to induce immune tolerance after liver transplantation: Clinical perspectives. World J Gastroenterol 2024; 30:1791-1800. [PMID: 38659486 PMCID: PMC11036497 DOI: 10.3748/wjg.v30.i13.1791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/03/2024] [Accepted: 03/14/2024] [Indexed: 04/03/2024] Open
Abstract
Liver transplantation (LT) has become the most efficient treatment for pediatric and adult end-stage liver disease and the survival time after transplantation is becoming longer due to the development of surgical techniques and perioperative management. However, long-term side-effects of immunosuppressants, like infection, metabolic disorders and malignant tumor are gaining more attention. Immune tolerance is the status in which LT recipients no longer need to take any immunosuppressants, but the liver function and intrahepatic histology maintain normal. The approaches to achieve immune tolerance after transplantation include spontaneous, operational and induced tolerance. The first two means require no specific intervention but withdrawing immunosuppressant gradually during follow-up. No clinical factors or biomarkers so far could accurately predict who are suitable for immunosuppressant withdraw after transplantation. With the understanding to the underlying mechanisms of immune tolerance, many strategies have been developed to induce tolerance in LT recipients. Cellular strategy is one of the most promising methods for immune tolerance induction, including chimerism induced by hematopoietic stem cells and adoptive transfer of regulatory immune cells. The safety and efficacy of various cell products have been evaluated by prospective preclinical and clinical trials, while obstacles still exist before translating into clinical practice. Here, we will summarize the latest perspectives and concerns on the clinical application of cellular strategies in LT recipients.
Collapse
Affiliation(s)
- Ai-Wei Zhou
- Department of Liver Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jing Jin
- Department of Nursing, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yuan Liu
- Department of Liver Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Liver Transplantation, Shanghai Immune Therapy Institute, Shanghai 200127, China
| |
Collapse
|
4
|
Lindoso RS, Collino F, Kasai-Brunswick TH, Costa MR, Verdoorn KS, Einicker-Lamas M, Vieira-Beiral HJ, Wessely O, Vieyra A. Resident Stem Cells in Kidney Tissue. RESIDENT STEM CELLS AND REGENERATIVE THERAPY 2024:159-203. [DOI: 10.1016/b978-0-443-15289-4.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Chang SH, Park CG. Comparing the Benefits and Drawbacks of Stem Cell Therapy Based on the Cell Origin or Manipulation Process: Addressing Immunogenicity. Immune Netw 2023; 23:e44. [PMID: 38188600 PMCID: PMC10767552 DOI: 10.4110/in.2023.23.e44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/12/2023] [Indexed: 01/09/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are effective in treating autoimmune diseases and managing various conditions, such as engraftment of allogeneic islets. Additionally, autologous and HLA-matched allogeneic MSCs can aid in the engraftment of human allogeneic kidneys with or without low doses of tacrolimus, respectively. However, HLA alloantigens are problematic because cell therapy uses more HLA-mismatched allogeneic cells than autologous for convenience and standardization. In particular, HLA-mismatched MSCs showed increased Ag-specific T/B cells and reduced viability faster than HLA-matched MSCs. In CRISPR/Cas9-based cell therapy, Cas9 induce T cell activation in the recipient's immune system. Interestingly, despite their immunogenicity being limited to the cells with foreign Ags, the accumulation of HLA alloantigen-sensitized T/B cells may lead to allograft rejection, suggesting that alloantigens may have a greater scope of adverse effects than foreign Ags. To avoid alloantigen recognition, the β2-microglobulin knockout (B2MKO) system, eliminating class-I MHC, was able to avoid rejection by alloreactive CD8 T cells compared to controls. Moreover, universal donor cells in which both B2M and Class II MHC transactivator (CIITA) were knocked out was more effective in avoiding immune rejection than single KO. However, B2MKO and CIITA KO system remain to be controlled and validated for adverse effects such as the development of tumorigenicity due to deficient Ag recognition by CD8 T and CD4 T cells, respectively. Overall, better HLA-matching or depletion of HLA alloantigens prior to cell therapy can reduce repetitive transplantation through the long-term survival of allogeneic cell therapy, which may be especially important for patients seeking allogeneic transplantation.
Collapse
Affiliation(s)
- Sung-Ho Chang
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea
| | - Chung Gyu Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Transplantation Research Institute, Medical Research center, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
6
|
Kang H, Feng J, Peng Y, Liu Y, Yang Y, Wu Y, Huang J, Jie Y, Chen B, He Y. Human mesenchymal stem cells derived from adipose tissue showed a more robust effect than those from the umbilical cord in promoting corneal graft survival by suppressing lymphangiogenesis. Stem Cell Res Ther 2023; 14:328. [PMID: 37957770 PMCID: PMC10644560 DOI: 10.1186/s13287-023-03559-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown promising potential in allograft survival. However, few reports have focused on comparing the immunosuppressive capacity of MSCs from different sources and administered via different routes in inhibiting transplant rejection. Moreover, virtually nothing is known about the role of MSCs in the regulation of graft neovascularization and lymphangiogenesis. In this study, we compared the efficacy of human adipose MSCs (hAD-MSCs) and human umbilical cord MSCs (hUC-MSCs) in vitro and in corneal transplantation models to explore the underlying molecular mechanisms and provide a powerful strategy for future clinical applications. METHODS hAD-MSCs and hUC-MSCs were generated, and their self-renewal and multi-differentiation abilities were evaluated. The inhibitory effect of human MSCs (hMSCs) was examined by T-cell proliferation assays with or without transwell in vitro. Two MSCs from different sources were separately adoptively transferred in mice corneal transplantation (5 × 105 or 1 × 106/mouse) via topical subconjunctival or intravenous (IV) routes. Allograft survival was evaluated every other day, and angiogenesis and lymphomagenesis were quantitatively analyzed by immunofluorescence staining. The RNA expression profiles of hMSCs were revealed by RNA sequencing (RNA-seq) and verified by quantitative real-time PCR (qRT‒PCR), western blotting or ELISA. The function of the differentially expressed gene FAS was verified by a T-cell apoptosis assay. RESULTS hAD-MSCs induced stronger immunosuppression in vitro than hUC-MSCs. The inhibitory effect of hUC-MSCs but not hAD-MSCs was mediated by cell-cell contact-dependent mechanisms. Systemic administration of a lower dose of hAD-MSCs showed better performance in prolonging corneal allograft survival than hUC-MSCs, while subconjunctival administration of hMSCs was safer and further prolonged corneal allograft survival. Both types of hMSCs could inhibit corneal neovascularization, while hAD-MSCs showed greater superiority in suppressing graft lymphangiogenesis. RNA-seq analysis and confirmation experiments revealed the superior performance of hAD-MSCs in allografts based on the lower expression of vascular endothelial growth factor C (VEGF-C) and higher expression of FAS. CONCLUSIONS The remarkable inhibitory effects on angiogenesis/lymphangiogenesis and immunological transplantation effects support the development of hAD-MSCs as a cell therapy against corneal transplant rejection. Topical administration of hMSCs was a safer and more effective route for application than systemic administration.
Collapse
Affiliation(s)
- Huanmin Kang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Jianing Feng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
- Shanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital of Northwest University, Xi'an, 710004, China
| | - Yingqian Peng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Yingyi Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Yalei Yang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Ying Wu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Jian Huang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ying Jie
- Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Baihua Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, 410011, Hunan, China
| | - Yan He
- Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Bezstarosti S, Meziyerh S, Reinders MEJ, Voogt-Bakker K, Groeneweg KE, Roelen DL, Kers J, de Fijter JW, Heidt S. HLA-DQ eplet mismatch load may identify kidney transplant patients eligible for tacrolimus withdrawal without donor-specific antibody formation after mesenchymal stromal cell therapy. HLA 2023. [PMID: 36841928 DOI: 10.1111/tan.15008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Recently, the randomized phase-II Triton study demonstrated that mesenchymal stromal cell (MSC) therapy facilitated early tacrolimus withdrawal in living donor kidney transplant recipients. The current sub-study analyzed formation of de novo donor-specific HLA antibodies (dnDSA) in the context of the degree of HLA eplet mismatches. At the time of protocol biopsy at 6 months, 7/29 patients (24%) in the MSC group and 1/27 patient (3.7%) in the control group had developed dnDSA. In the MSC group, all dnDSA were anti-HLA-DQ; two patients had anti-DQ alone and five patients combined with anti-class I, HLA-DR or -DP. Despite excess dnDSA formation in the MSC-arm of the study, the evolution of eGFR (CKD-EPI) and proteinuria were comparable 2 years posttransplant. All dnDSA were complement-binding and three patients had antibody-mediated rejection in the protocol biopsy, but overall rejection episodes were not increased. Everolimus had to be discontinued in nine patients because of toxicity, and tacrolimus was reintroduced in six patients because of dnDSA formation. The HLA-DQ eplet mismatch load independently associated with dnDSA (adjusted hazard ratio = 1.07 per eplet mismatch, p = 0.008). A threshold of ≥11 HLA-DQ eplet mismatches predicted subsequent dnDSA in all 11 patients in the MSC group, but specificity was low (44%). Further research is warranted to explore HLA molecular mismatch load as a biomarker to guide personalized maintenance immunosuppression in kidney transplantation.
Collapse
Affiliation(s)
- Suzanne Bezstarosti
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands.,Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Soufian Meziyerh
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Kim Voogt-Bakker
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Koen E Groeneweg
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesper Kers
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Johan W de Fijter
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands.,Eurotransplant Reference Laboratory, Leiden, The Netherlands
| |
Collapse
|
8
|
Cheng HY, Anggelia MR, Lin CH, Wei FC. Toward transplantation tolerance with adipose tissue-derived therapeutics. Front Immunol 2023; 14:1111813. [PMID: 37187733 PMCID: PMC10175575 DOI: 10.3389/fimmu.2023.1111813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Solid organ and composite tissue allotransplanation have been widely applied to treat end-stage organ failure and massive tissue defects, respectively. Currently there are a lot of research endeavors focusing on induction of transplantation tolerance, to relieve the burden derived from long-term immunosuppressant uptake. The mesenchymal stromal cells (MSCs) have been demonstrated with potent immunomodulatory capacities and applied as promising cellular therapeutics to promote allograft survival and induce tolerance. As a rich source of adult MSCs, adipose tissue provides additional advantages of easy accessibility and good safety profile. In recent years, the stromal vascular fraction (SVF) isolated from adipose tissues following enzymatic or mechanical processing without in vitro culture and expansion has demonstrated immunomodulatory and proangiogenic properties. Furthermore, the secretome of AD-MSCs has been utilized in transplantation field as a potential "cell-free" therapeutics. This article reviews recent studies that employ these adipose-derived therapeutics, including AD-MSCs, SVF, and secretome, in various aspects of organ and tissue allotransplantation. Most reports validate their efficacies in prolonging allograft survival. Specifically, the SVF and secretome have performed well for graft preservation and pretreatment, potentially through their proangiogenic and antioxidative capacities. In contrast, AD-MSCs were suitable for peri-transplantation immunosuppression. The proper combination of AD-MSCs, lymphodepletion and conventional immunosuppressants could consistently induce donor-specific tolerance to vascularized composite allotransplants (VCA). For each type of transplantation, optimizing the choice of therapeutics, timing, dose, and frequency of administration may be required. Future progress in the application of adipose-derived therapeutics to induce transplantation tolerance will be further benefited by continued research into their mechanisms of action and the development of standardized protocols for isolation methodologies, cell culture, and efficacy evaluation.
Collapse
Affiliation(s)
- Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- *Correspondence: Hui-Yun Cheng,
| | - Madonna Rica Anggelia
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chan Wei
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
9
|
Večerić-Haler Ž, Sever M, Kojc N, Halloran PF, Boštjančič E, Mlinšek G, Oblak M, Poženel P, Švajger U, Hartman K, Kneževič M, Barlič A, Girandon L, Aleš Rigler A, Zver S, Buturović Ponikvar J, Arnol M. Autologous Mesenchymal Stem Cells for Treatment of Chronic Active Antibody-Mediated Kidney Graft Rejection: Report of the Phase I/II Clinical Trial Case Series. Transpl Int 2022; 35:10772. [PMID: 36484064 PMCID: PMC9722440 DOI: 10.3389/ti.2022.10772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022]
Abstract
Mesenchymal stem cell (MSCs) therapy has already been studied in kidney transplant recipients (KTRs), and the available data showed that it is safe and well tolerated. The aim of this study was to evaluate the safety and efficacy of autologous MSCs in combination with standard therapy in KTRs with biopsy-proven chronic active antibody-mediated rejection (AMR). Patients with biopsy-proven chronic active AMR received treatment with autologous bone marrow-derived MSCs (3 × 106 cells/kg iv) after completion of standard therapy and were followed for up to 12 months. The primary endpoints were safety by assessment of adverse events. Secondary endpoints included assessment of kidney graft function, immunological and histological changes related to AMR activity and chronicity assessed by conventional microscopy and molecular transcripts. A total of 3 patients were enrolled in the study before it was terminated prematurely because of adverse events. We found that AMR did not improve in any of the patients after treatment with MSCs. In addition, serious adverse events were observed in one case when autologous MSCs therapy was administered in the late phase after kidney transplantation, which requires further elucidation.
Collapse
Affiliation(s)
- Željka Večerić-Haler
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,*Correspondence: Željka Večerić-Haler,
| | - Matjaž Sever
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Nika Kojc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Philip F. Halloran
- Division of Nephrology and Transplant Immunology, Alberta Transplant Applied Genomics Centre, University of Alberta, Edmonton, AB, Canada
| | - Emanuela Boštjančič
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Mlinšek
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Manca Oblak
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Primož Poženel
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Urban Švajger
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Katrina Hartman
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | | | - Ariana Barlič
- Educell d.o.o Cell Therapy Service, Ljubljana, Slovenia
| | | | - Andreja Aleš Rigler
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Samo Zver
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jadranka Buturović Ponikvar
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Arnol
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
10
|
Deo D, Marchioni M, Rao P. Mesenchymal Stem/Stromal Cells in Organ Transplantation. Pharmaceutics 2022; 14:pharmaceutics14040791. [PMID: 35456625 PMCID: PMC9029865 DOI: 10.3390/pharmaceutics14040791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Organ transplantation is essential and crucial for saving and enhancing the lives of individuals suffering from end-stage organ failure. Major challenges in the medical field include the shortage of organ donors, high rates of organ rejection, and long wait times. To address the current limitations and shortcomings, cellular therapy approaches have been developed using mesenchymal stem/stromal cells (MSC). MSC have been isolated from various sources, have the ability to differentiate to important cell lineages, have anti-inflammatory and immunomodulatory properties, allow immunosuppressive drug minimization, and induce immune tolerance towards the transplanted organ. Additionally, rapid advances in the fields of tissue engineering and regenerative medicine have emerged that focus on either generating new organs and organ sources or maximizing the availability of existing organs. This review gives an overview of the various properties of MSC that have enabled its use as a cellular therapy for organ preservation and transplant. We also highlight emerging fields of tissue engineering and regenerative medicine along with their multiple sub-disciplines, underlining recent advances, widespread clinical applications, and potential impact on the future of tissue and organ transplantation.
Collapse
|
11
|
Yeo WS, Ng QX. Biomarkers of immune tolerance in kidney transplantation: an overview. Pediatr Nephrol 2022; 37:489-498. [PMID: 33712863 DOI: 10.1007/s00467-021-05023-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 11/30/2022]
Abstract
Kidney failure, one of the most prevalent diseases in the world and with increasing incidence, is associated with substantial morbidity and mortality. Currently available modes of kidney replacement therapy include dialysis and kidney transplantation. Though kidney transplantation is the preferred and ideal mode of kidney replacement therapy, this modality, however, is not without its risks. Kidney transplant recipients are constantly at risk of complications associated with immunosuppression, namely, opportunistic infections (e.g., Epstein-Barr virus and cytomegalovirus infections), post-transplant lymphoproliferative disorder, and complications associated with immunosuppressants (e.g., calcineurin inhibitor- and corticosteroid-associated new onset diabetes after transplantation and calcineurin inhibitor-associated nephrotoxicity). Transplantation tolerance, an acquired state in which immunocompetent recipients have developed donor-specific unresponsiveness, may be the Holy Grail in enabling optimal allograft survival and obviating the risks associated with immunosuppression in kidney transplant recipients. This review aims to discuss the biomarkers available to predict, identify, and define the transplant immune tolerant state and various tolerance induction strategies. Regrettably, pediatric patients have not been included in any tolerance studies and this should be the focus of future studies.
Collapse
Affiliation(s)
- Wee-Song Yeo
- Mount Elizabeth Hospital, 3 Mount Elizabeth, Singapore, 228510, Singapore.
| | - Qin Xiang Ng
- MOH Holdings Pte Ltd, 1 Maritime Square, Singapore, 099253, Singapore
| |
Collapse
|
12
|
Mesenchymal Stromal Cells Mediate Clinically Unpromising but Favourable Immune Responses in Kidney Transplant Patients. Stem Cells Int 2022; 2022:2154544. [PMID: 35211176 PMCID: PMC8863486 DOI: 10.1155/2022/2154544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/11/2022] [Indexed: 01/22/2023] Open
Abstract
Background Allograft rejection postkidney transplantation (KTx) is a major clinical challenge despite increased access to a healthcare system and improvement in immunosuppressive (IS) drugs. In recent years, mesenchymal stromal cells (MSCs) have aroused considerable interest in field of transplantation due to their immunomodulatory and regenerative properties. This study was aimed at investigating safety, feasibility, and immunological effects of autologous MSCs (auto-MSCs) and allogeneic MSCs (allo-MSCs) as a complement to IS drug therapy in KTx patients. Methods 10 patients undergoing KTx with a living-related donor were analysed along with 5 patients in the control group. Patients were given auto-MSCs or allo-MSCs at two time points, i.e., one day before transplant (D-0) and 30 days after transplant (D-30) at the rate of 1.0-1.5 × 106 MSCs per kg body weight in addition to immunosuppressants. Patients were followed up for 2 years, and 29 immunologically relevant lymphocyte subsets and 8 cytokines and important biomarkers were analysed at all time points. Results Patients displayed no signs of discomfort or dose-related toxicities in response to MSC infusion. Flow cytometric analysis revealed an increase in B regulatory lymphocyte populations and nonconventional T regulatory cells and a decrease in T effector lymphocyte proportions in auto-MSC-infused patients. No such favourable immune responses were observed in all MSC-infused patients. Conclusion This study provides evidence that auto-MSCs are safe and well tolerated. This is the first ever report to compare autologous and allogeneic MSC infusion in KTx patients. Importantly, our data demonstrated that MSC-induced immune responses in patients did not completely correlate with clinical outcomes. Our findings add to the current perspective of using MSCs in KTx and explore possibilities through which donor/recipient chimerism can be achieved to induce immune tolerance in KTx patients.
Collapse
|
13
|
Molecular Mechanisms of Kidney Injury and Repair. Int J Mol Sci 2022; 23:ijms23031542. [PMID: 35163470 PMCID: PMC8835923 DOI: 10.3390/ijms23031542] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) will become the fifth global cause of death by 2040, thus emphasizing the need to better understand the molecular mechanisms of damage and regeneration in the kidney. CKD predisposes to acute kidney injury (AKI) which, in turn, promotes CKD progression. This implies that CKD or the AKI-to-CKD transition are associated with dysfunctional kidney repair mechanisms. Current therapeutic options slow CKD progression but fail to treat or accelerate recovery from AKI and are unable to promote kidney regeneration. Unraveling the cellular and molecular mechanisms involved in kidney injury and repair, including the failure of this process, may provide novel biomarkers and therapeutic tools. We now review the contribution of different molecular and cellular events to the AKI-to-CKD transition, focusing on the role of macrophages in kidney injury, the different forms of regulated cell death and necroinflammation, cellular senescence and the senescence-associated secretory phenotype (SAPS), polyploidization, and podocyte injury and activation of parietal epithelial cells. Next, we discuss key contributors to repair of kidney injury and opportunities for their therapeutic manipulation, with a focus on resident renal progenitor cells, stem cells and their reparative secretome, certain macrophage subphenotypes within the M2 phenotype and senescent cell clearance.
Collapse
|
14
|
Zhang L, Lai X, Guo Y, Ma J, Fang J, Li G, Xu L, Yin W, Chen Z. Autologous bone marrow-derived mesenchymal stem cells for interstitial fibrosis and tubular atrophy: a pilot study. Ren Fail 2021; 43:1266-1275. [PMID: 34493167 PMCID: PMC8425735 DOI: 10.1080/0886022x.2021.1968432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs)-based therapy has shown promising results for renal injury. In this study, the efficacy and safety of autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) in treating nonspecific interstitial fibrosis and tubular atrophy (IFTA) were evaluated. Methods From March 2011 to January 2013, 11 renal transplanted patients with IFTA were recruited. At baseline, patients were given one intra-arterial infusion of BM-MSCs; 7 days and 1 month later, another two intravenous infusions of cells were followed. Serum creatinine, creatinine clearance rate, and serum cystatin-C at baseline and 7 days, 1 month, 3 months, 6 months, and 12 months after the intra-arterial infusion of BM-MSCs were used to assess renal function. At baseline and 6 months, histological examination based on hematoxylin-eosin, Masson’s trichrome and periodic acid-Schiff staining and immunohistochemistry for transforming growth factor β1 (TGF-β1) and connective tissue growth factor (CTGF) was performed. Adverse events were recorded to evaluate the safety of BM-MSCs treatment. Results At 12 months, the renal function of 6 patients (54.5%) was improved, 3 (27.3%) were stable and 2 (18.2%) were worsened. At 6 months, the mean IFTA scores of all participators were similar with the baseline (1.73 ± 0.41 vs.1.50 ± 0.0.77, p = 0.242); however, it was significantly decreased when only 6 patients with improved renal function were analyzed (1.67 ± 0.41 vs. 1.08 ± 0.20, p = 0.013). Besides, decreased expression of TGF-β1 and CTGF were also observed at 6 months. During 1 year follow-up period, only two minor complications including infection and allergy were observed. Conclusion Our results demonstrated that autologous BM-MSCs are safe and beneficial for IFTA patients. Abbreviations: MSCs: mesenchymal stem cells; BM-MSCs: marrow-derived mesenchymal stem cells; IFTA: interstitial fibrosis and tubular atrophy; CAN: chronic allograft nephropathy; CNIs: calcineurin inhibitors; Scr: serum creatinine; CCr: creatinine clearance rate; Cys-C: cystatin-C; TGF-β1: transforming growth factor β1; CTGF: connective tissue growth factor
Collapse
Affiliation(s)
- Lei Zhang
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Xingqiang Lai
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Yuhe Guo
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Junjie Ma
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Jiali Fang
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Guanghui Li
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Lu Xu
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Wei Yin
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| | - Zheng Chen
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University/The Second Clinical Medicine School of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Reinders MEJ, Groeneweg KE, Hendriks SH, Bank JR, Dreyer GJ, de Vries APJ, van Pel M, Roelofs H, Huurman VAL, Meij P, Moes DJAR, Fibbe WE, Claas FHJ, Roelen DL, van Kooten C, Kers J, Heidt S, Rabelink TJ, de Fijter JW. Autologous bone marrow-derived mesenchymal stromal cell therapy with early tacrolimus withdrawal: The randomized prospective, single-center, open-label TRITON study. Am J Transplant 2021; 21:3055-3065. [PMID: 33565206 PMCID: PMC8518640 DOI: 10.1111/ajt.16528] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 01/25/2023]
Abstract
After renal transplantation, there is a need for immunosuppressive regimens which effectively prevent allograft rejection, while preserving renal function and minimizing side effects. From this perspective, mesenchymal stromal cell (MSC) therapy is of interest. In this randomized prospective, single-center, open-label trial, we compared MSCs infused 6 and 7 weeks after renal transplantation and early tacrolimus withdrawal with a control tacrolimus group. Primary end point was quantitative evaluation of interstitial fibrosis in protocol biopsies at 4 and 24 weeks posttransplant. Secondary end points included acute rejection, graft loss, death, renal function, adverse events, and immunological responses. Seventy patients were randomly assigned of which 57 patients were included in the final analysis (29 MSC; 28 controls). Quantitative progression of fibrosis failed to show benefit in the MSC group and GFR remained stable in both groups. One acute rejection was documented (MSC group), while subclinical rejection in week 24 protocol biopsies occurred in seven patients (four MSC; three controls). In the MSC group, regulatory T cell numbers were significantly higher compared to controls (p = .014, week 24). In conclusion, early tacrolimus withdrawal with MSC therapy was safe and feasible without increased rejection and with preserved renal function. MSC therapy is a potentially useful approach after renal transplantation.
Collapse
Affiliation(s)
- Marlies E. J. Reinders
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Koen E. Groeneweg
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Sanne H. Hendriks
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands
| | - Jonna R. Bank
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Geertje J. Dreyer
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Aiko P. J. de Vries
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Melissa van Pel
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands,NECSTGENLeidenthe Netherlands
| | - Helene Roelofs
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands
| | - Volkert A. L. Huurman
- Department of Transplant Surgery and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Paula Meij
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenthe Netherlands
| | - Dirk J. A. R. Moes
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenthe Netherlands
| | - Willem E. Fibbe
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands
| | - Frans H. J. Claas
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands
| | - Dave L. Roelen
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Jesper Kers
- Department of PathologyLeiden University Medical CenterLeidenthe Netherlands,Department of PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands,Van ‘t Hoff Institute for Molecular Sciences (HIMS)University of AmsterdamAmsterdamthe Netherlands
| | - Sebastiaan Heidt
- Department of ImmunologyLeiden University Medical CenterLeidenthe Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Johan W. de Fijter
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
16
|
Shaw BI, Ord JR, Nobuhara C, Luo X. Cellular Therapies in Solid Organ Allotransplantation: Promise and Pitfalls. Front Immunol 2021; 12:714723. [PMID: 34526991 PMCID: PMC8435835 DOI: 10.3389/fimmu.2021.714723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/30/2022] Open
Abstract
Donor specific transfusions have been the basis of tolerance inducing protocols since Peter Medawar showed that it was experimentally feasible in the 1950s. Though trials of cellular therapies have become increasingly common in solid organ transplantation, they have not become standard practice. Additionally, whereas some protocols have focused on cellular therapies as a method for donor antigen delivery-thought to promote tolerance in and of itself in the correct immunologic context-other approaches have alternatively focused on the intrinsic immunosuppressive properties of the certain cell types with less emphasis on their origin, including mesenchymal stem cells, regulatory T cells, and regulatory dendritic cells. Regardless of intent, all cellular therapies must contend with the potential that introducing donor antigen in a new context will lead to sensitization. In this review, we focus on the variety of cellular therapies that have been applied in human trials and non-human primate models, describe their efficacy, highlight data regarding their potential for sensitization, and discuss opportunities for cellular therapies within our current understanding of the immune landscape.
Collapse
Affiliation(s)
- Brian I. Shaw
- Department of Surgery, Duke University, Durham, NC, United States
| | - Jeffrey R. Ord
- School of Medicine, Duke University, Durham, NC, United States
| | - Chloe Nobuhara
- School of Medicine, Duke University, Durham, NC, United States
| | - Xunrong Luo
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC, United States
| |
Collapse
|
17
|
Casiraghi F, Perico N, Podestà MA, Todeschini M, Zambelli M, Colledan M, Camagni S, Fagiuoli S, Pinna AD, Cescon M, Bertuzzo V, Maroni L, Introna M, Capelli C, Golay JT, Buzzi M, Mister M, Ordonez PYR, Breno M, Mele C, Villa A, Remuzzi G. Third-party bone marrow-derived mesenchymal stromal cell infusion before liver transplantation: A randomized controlled trial. Am J Transplant 2021; 21:2795-2809. [PMID: 33370477 DOI: 10.1111/ajt.16468] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/24/2020] [Accepted: 12/21/2020] [Indexed: 01/25/2023]
Abstract
Mesenchymal stromal cells (MSC) have emerged as a promising therapy to minimize the immunosuppressive regimen or induce tolerance in solid organ transplantation. In this randomized open-label phase Ib/IIa clinical trial, 20 liver transplant patients were randomly allocated (1:1) to receive a single pretransplant intravenous infusion of third-party bone marrow-derived MSC or standard of care alone. The primary endpoint was the safety profile of MSC administration during the 1-year follow-up. In all, 19 patients completed the study, and none of those who received MSC experienced infusion-related complications. The incidence of serious and non-serious adverse events was similar in the two groups. Circulating Treg/memory Treg and tolerant NK subset of CD56bright NK cells increased slightly over baseline, albeit not to a statistically significant extent, in MSC-treated patients but not in the control group. Graft function and survival, as well as histologic parameters and intragraft expression of tolerance-associated transcripts in 1-year protocol biopsies were similar in the two groups. In conclusion, pretransplant MSC infusion in liver transplant recipients was safe and induced mild positive changes in immunoregulatory T and NK cells in the peripheral blood. This study opens the way for a trial on possible tolerogenic efficacy of MSC in liver transplantation. ClinicalTrials.gov identifier: NCT02260375.
Collapse
Affiliation(s)
- Federica Casiraghi
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Norberto Perico
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Manuel A Podestà
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.,Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marta Todeschini
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marco Zambelli
- Department of Organ Failure and Transplantation, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Michele Colledan
- Department of Organ Failure and Transplantation, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Stefania Camagni
- Department of Organ Failure and Transplantation, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Stefano Fagiuoli
- Gastroenterology, Hepatology and Transplantation, Department of Medicine, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Antonio D Pinna
- General Surgery and Transplant Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria-Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Matteo Cescon
- General Surgery and Transplant Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria-Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Valentina Bertuzzo
- General Surgery and Transplant Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria-Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Lorenzo Maroni
- General Surgery and Transplant Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria-Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Martino Introna
- G. Lanzani Laboratory of Cell Therapy, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Chiara Capelli
- G. Lanzani Laboratory of Cell Therapy, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Josee T Golay
- G. Lanzani Laboratory of Cell Therapy, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Marina Buzzi
- Emilia Romagna Cord Blood Bank, Immunohematology and Transfusion Medicine, Azienda Ospedaliero-Universitaria-Policlinico S. Orsola-Malpighi, Bologna, Italy
| | - Marilena Mister
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Pamela Y R Ordonez
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Matteo Breno
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Caterina Mele
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Alessandro Villa
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | | |
Collapse
|
18
|
Wong CY. Current advances of stem cell-based therapy for kidney diseases. World J Stem Cells 2021; 13:914-933. [PMID: 34367484 PMCID: PMC8316868 DOI: 10.4252/wjsc.v13.i7.914] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Kidney diseases are a prevalent health problem around the world. Multidrug therapy used in the current routine treatment for kidney diseases can only delay disease progression. None of these drugs or treatments can reverse the progression to an end-stage of the disease. Therefore, it is crucial to explore novel therapeutics to improve patients’ quality of life and possibly cure, reverse, or alleviate the kidney disease. Stem cells have promising potentials as a form of regenerative medicine for kidney diseases due to their unlimited replication and their ability to differentiate into kidney cells in vitro. Mounting evidences from the administration of stem cells in an experimental kidney disease model suggested that stem cell-based therapy has therapeutic or renoprotective effects to attenuate kidney damage while improving the function and structure of both glomerular and tubular compartments. This review summarises the current stem cell-based therapeutic approaches to treat kidney diseases, including the various cell sources, animal models or in vitro studies. The challenges of progressing from proof-of-principle in the laboratory to widespread clinical application and the human clinical trial outcomes reported to date are also highlighted. The success of cell-based therapy could widen the scope of regenerative medicine in the future.
Collapse
Affiliation(s)
- Chee-Yin Wong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
- Research Department, Cytopeutics, Cyberjaya 63000, Selangor, Malaysia
| |
Collapse
|
19
|
Večerić-Haler Ž, Kojc N, Sever M, Zver S, Švajger U, Poženel P, Hartman K, Urdih T, Mlinšek G, Oblak M, Aleš Rigler A, Ihan A, Buturović Ponikvar J, Halloran PP, Arnol M. Case Report: Capillary Leak Syndrome With Kidney Transplant Failure Following Autologous Mesenchymal Stem Cell Therapy. Front Med (Lausanne) 2021; 8:708744. [PMID: 34368198 PMCID: PMC8334176 DOI: 10.3389/fmed.2021.708744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted great interest in the field of kidney transplantation due to their immunomodulatory and reparative properties. In registered clinical trials, MSCs have been used before, at the time of, or early after transplantation and have been reported to be well-tolerated with no serious safety concerns. No results are available on the use of MSCs in the late post-transplant period. Here, we present a case report of a severe systemic complication mimicking capillary leak syndrome with ultimate kidney transplant failure after autologous transplantation of MSCs used as rescue treatment of late antibody-mediated kidney allograft rejection.
Collapse
Affiliation(s)
- Željka Večerić-Haler
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nika Kojc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matjaž Sever
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Samo Zver
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.,Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Urban Švajger
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Primož Poženel
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Katrina Hartman
- Division for Cells and Tissue, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Tereza Urdih
- Department of Haematology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Gregor Mlinšek
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Manca Oblak
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Andreja Aleš Rigler
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jadranka Buturović Ponikvar
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Philip P Halloran
- Division of Nephrology and Transplant Immunology, University of Alberta, Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
| | - Miha Arnol
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
20
|
Chen Y, Yan G, Ma Y, Zhong M, Yang Y, Guo J, Wang C, Han W, Zhang L, Xu S, Huang J, Dai H, Qi Z. Combination of mesenchymal stem cells and FK506 prolongs heart allograft survival by inhibiting TBK1/IRF3-regulated-IFN-γ production. Immunol Lett 2021; 238:21-28. [PMID: 34228988 DOI: 10.1016/j.imlet.2021.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/04/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Lifelong immunosuppression use presents many serious side effects to transplant recipients. Previous studies have shown that mesenchymal stem cells (MSC) regulate the progress of inflammation and protect allograft function. However, the benefits of MSC combined with low-dose tacrolimus (FK506) has not been investigated in heart transplant recipients, and its mechanism deserves further investigation. SD Rat bone marrow-derived MSC were infused into recipient mouse (C57BL/6, B6) through the tail vein, followed by a BALB/c donor cervical ectopic heart transplantation on the next day of infusion. T-lymphocyte subsets and their functions were determined using flow cytometry, ELISA, and qPCR. Thereafter, in vitro and in vivo experiments were conducted to identify the mechanisms regarding MSC and FK506 combination (MF group) use in regulating IFN-γ signaling. MF group in the allogeneic heart transplantation mouse model inhibited acute rejection and prolonged mean survival time (MST) of grafts from 7 days (d) to 22d. Pathological examination of heart grafts suggested that inflammatory cell infiltration decreased, and tissue damage was significantly reduced in the MF group. IFN-γ mRNA expression levels in the grafts and recipients decreased, while IL-4 and TGF-β mRNA expression increased in the MF group. Phosphorylation of TBK1/IRF3 in recipient immune cells decreased under donor antigen stimulation. Combination use of MSC and FK506 can prolong graft survival, possibly by down-regulating TBK1/IRF3 phosphorylation, thus reducing IFN-γ production to prevent infiltration of inflammatory cells in the graft and extend graft survival. The findings provide a potential new approach to immunosuppression selection.
Collapse
Affiliation(s)
- Yingyu Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China; Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Guoliang Yan
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yunhan Ma
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Mengya Zhong
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yan Yang
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Junjun Guo
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chenxi Wang
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Weimin Han
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Liyi Zhang
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuangyue Xu
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinjin Huang
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Helong Dai
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China; Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China; Clinical Immunology Center, Central South University, Changsha, China.
| | - Zhongquan Qi
- Organ Transplantation institute, School of Medicine, Xiamen University, Xiamen, Fujian, China; Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen, Fujian, China; Medical College, Guangxi University, Nanning, Guangxi, China.
| |
Collapse
|
21
|
El Hennawy HM, Faifi ASA, El Nazer W, Mahedy A, Kamal A, Al Faifi IS, Abdulmalik H, Safar O, Zaitoun MF, Fahmy AE. Calcineurin Inhibitors Nephrotoxicity Prevention Strategies With Stress on Belatacept-Based Rescue Immunotherapy: A Review of the Current Evidence. Transplant Proc 2021; 53:1532-1540. [PMID: 34020797 DOI: 10.1016/j.transproceed.2021.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/10/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND A traditional narrative review was performed to evaluate clinical studies that have examined the clinical implications, risk factors, and prevention of calcineurin inhibitors (CNIs) nephrotoxicity with stress on a belatacept-based rescue regimen. METHODS The Cochrane Library, PubMed/MEDLINE, EBSCO (Academic Search Ultimate), ProQuest (Central), and Excerpta Medical databases and Google scholar were searched using the keywords (CNI AND Nephrotoxicity prevention) OR ("Calcineurin inhibitor" AND Nephrotoxicity) OR (Tacrolimus AND Nephrotoxicity) OR (Ciclosporin AND Nephrotoxicity) OR (cyclosporine AND Nephrotoxicity) OR (Belatacept) OR (CNI Conversion) for the period from 1990 to 2020. Fifty-five related articles and reviews were found. CONCLUSION A better understanding of the mechanisms underlying calcineurin inhibitor nephrotoxicity could help in the individualization of therapy for and prevention of CNI nephrotoxicity. Identification of high-risk patients for CNI nephrotoxicity before renal transplantation enables better use and selection of immunosuppression with reduced adverse effects and, eventually, successful treatment of the kidney recipients. Belatacept conversion is a good and safe option in patients with deteriorating renal function attributed to CNI nephrotoxicity.
Collapse
Affiliation(s)
- Hany M El Hennawy
- Transplant Surgery Section, Surgery Department, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia.
| | - Abdullah S Al Faifi
- Transplant Surgery Section, Surgery Department, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Weam El Nazer
- Nephrology Department, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Ahmed Mahedy
- Nephrology Department, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Ahmed Kamal
- Nephrology Department, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Ibrahim S Al Faifi
- Department of Family Medicine, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Hana Abdulmalik
- Department of Surgery, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Omar Safar
- Department of Urology, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Mohammad F Zaitoun
- Department of Pharmacy, Armed Forces Hospital-Southern Regions, Khamis Mushayt, Saudi Arabia
| | - Ahmed E Fahmy
- Department of Surgery, Division of Transplantation, North Shore University Hospital, Northwell Health, Manhasset, New York
| |
Collapse
|
22
|
Mansourabadi AH, Mohamed Khosroshahi L, Noorbakhsh F, Amirzargar A. Cell therapy in transplantation: A comprehensive review of the current applications of cell therapy in transplant patients with the focus on Tregs, CAR Tregs, and Mesenchymal stem cells. Int Immunopharmacol 2021; 97:107669. [PMID: 33965760 DOI: 10.1016/j.intimp.2021.107669] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Organ transplantation is a practical treatment for patients with end-stage organ failure. Despite the advances in short-term graft survival, long-term graft survival remains the main challenge considering the increased mortality and morbidity associated with chronic rejection and the toxicity of immunosuppressive drugs. Since a novel therapeutic strategy to induce allograft tolerance seems urgent, focusing on developing novel and safe approaches to prolong graft survival is one of the main goals of transplant investigators. Researchers in the field of organ transplantation are interested in suppressing or optimizing the immune responses by focusing on immune cells including mesenchymal stem cells (MSCs), polyclonal regulatory Tcells (Tregs), and antigen-specific Tregs engineered with chimeric antigen receptors (CAR Tregs). We review the mechanistic pathways, phenotypic and functional characteristics of these cells, and their promising application in organ transplantation.
Collapse
Affiliation(s)
- Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 009821 Tehran, Iran; Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 009821 Tehran, Iran
| | - Leila Mohamed Khosroshahi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran.
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran.
| |
Collapse
|
23
|
Mesenchymal stromal cells for corneal transplantation: Literature review and suggestions for successful clinical trials. Ocul Surf 2021; 20:185-194. [PMID: 33607323 PMCID: PMC9878990 DOI: 10.1016/j.jtos.2021.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 01/28/2023]
Abstract
Corneal transplantation is a routine procedure for patients with corneal blindness. Despite the streamlining of surgical techniques and deeper understanding of the cellular and molecular pathways mediating rejection, corticosteroids are still the main immunosuppressive regimen in corneal transplantation, and the 15-year survival of corneal transplants remains as low as 50%, which is poorer than that for most solid organ transplants. Recently, mesenchymal stromal cells (MSCs) with unique regenerative and immune-modulating properties have emerged as a promising cell therapy to promote transplant tolerance, minimize the use of immunosuppressants, and prevent chronic rejection. Here, we review the literature on preclinical studies of MSCs for corneal transplantation and summarize the key findings from clinical trials with MSCs in solid organ transplantation. Finally, we highlight current issues and challenges regarding MSC therapies and suggest strategies for safe and effective MSC-based therapies in clinical transplantation.
Collapse
|
24
|
Zhao L, Hu C, Han F, Chen D, Cheng J, Wu J, Peng W, Chen J. Induction therapy with mesenchymal stromal cells in kidney transplantation: a meta-analysis. Stem Cell Res Ther 2021; 12:158. [PMID: 33648596 PMCID: PMC7923637 DOI: 10.1186/s13287-021-02219-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/09/2021] [Indexed: 12/22/2022] Open
Abstract
Objective The aim of this meta-analysis was to evaluate the therapeutic effects of mesenchymal stromal cells (MSCs) versus traditional regimens for induction therapy in kidney transplantation (KT), especially the safety of MSC infusion, practicability of MSCs as induction therapy agents, and posttransplant complications. Methods PubMed, Embase, EBSCO, Ovid, and the Cochrane Library were searched for prospective clinical trials that compared MSCs with traditional regimens for induction therapy in KT. Results Four trials were included, including a total of 197 patients. The pooled results revealed that MSC therapy had a lower 1-year infection rate than did the traditional therapies (RR = 0.65, 95% CI: 0.46–0.9, P = 0.01). There were no significant differences between the two protocols regarding the 1-year acute rejection (AR) rate (RR = 0.77, 95% CI: 0.41–1.45, P = 0.42), 1-year graft survival rate (RR = 0.99, 95% CI: 0.95–1.03, P = 0.74), delayed graft function (DGF) rate (RR = 0.54, 95% CI: 0.21–1.38, P = 0.2) and renal graft function at 1 month (MD = −1.56, 95% CI: − 14.2–11.08, p = 0.81), 3 months (MD = 0.15, 95% CI: − 5.63–5.93, p = 0.96), 6 months (MD = − 1.95, 95% CI: − 9.87–5.97, p = 0.63), and 12 months (MD = − 1.13, 95% CI: − 7.16–4.89, p = 0.71) postsurgery. Subgroup analysis demonstrated that the 1-year AR rate, 1-year graft survival rate, DGF rate, and renal graft function at 12 months postsurgery did not significantly differ between the low-dose calcineurin inhibitor (CNI) group and the standard-dose CNI group, indicating the potential benefits of successful CNI sparing in combination with MSC treatment. Moreover, when MSCs were applied as an alternative therapy rather than an additional therapy or allogeneic MSCs were utilized instead of autologous MSCs, all of the outcomes mentioned above were comparable. Conclusion Induction therapy with MSCs is safe and has similar immune response modulation effects to those of traditional regimens in the short term in KT recipients. However, regarding the long-term effects, as suggested by the 1-year infection rate and the potential of CNI sparing, MSC therapy has significant advantages. However, these advantages should be further verified in more well-designed, multicenter randomized controlled trials (RCTs) with large sample sizes and long follow-up periods. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02219-7.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Institute of Nephrology, Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Dajin Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jun Cheng
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jianyong Wu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Wenhan Peng
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.
| |
Collapse
|
25
|
Podestà MA, Remuzzi G, Casiraghi F. Mesenchymal Stromal Cell Therapy in Solid Organ Transplantation. Front Immunol 2021; 11:618243. [PMID: 33643298 PMCID: PMC7902912 DOI: 10.3389/fimmu.2020.618243] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/29/2020] [Indexed: 12/29/2022] Open
Abstract
Transplantation is the gold-standard treatment for the failure of several solid organs, including the kidneys, liver, heart, lung and small bowel. The use of tailored immunosuppressive agents has improved graft and patient survival remarkably in early post-transplant stages, but long-term outcomes are frequently unsatisfactory due to the development of chronic graft rejection, which ultimately leads to transplant failure. Moreover, prolonged immunosuppression entails severe side effects that severely impact patient survival and quality of life. The achievement of tolerance, i.e., stable graft function without the need for immunosuppression, is considered the Holy Grail of the field of solid organ transplantation. However, spontaneous tolerance in solid allograft recipients is a rare and unpredictable event. Several strategies that include peri-transplant administration of non-hematopoietic immunomodulatory cells can safely and effectively induce tolerance in pre-clinical models of solid organ transplantation. Mesenchymal stromal cells (MSC), non-hematopoietic cells that can be obtained from several adult and fetal tissues, are among the most promising candidates. In this review, we will focus on current pre-clinical evidence of the immunomodulatory effect of MSC in solid organ transplantation, and discuss the available evidence of their safety and efficacy in clinical trials.
Collapse
Affiliation(s)
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Bergamo, Italy
| | - Federica Casiraghi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Bergamo, Italy
| |
Collapse
|
26
|
Hoogduijn MJ, Issa F, Casiraghi F, Reinders MEJ. Cellular therapies in organ transplantation. Transpl Int 2021; 34:233-244. [PMID: 33207013 PMCID: PMC7898347 DOI: 10.1111/tri.13789] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/15/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Cellular therapy is a promising tool for improving the outcome of organ transplantation. Various cell types with different immunoregulatory and regenerative properties may find application for specific transplant rejection or injury-related indications. The current era is crucial for the development of cellular therapies. Preclinical models have demonstrated the feasibility of efficacious cell therapy in transplantation, early clinical trials have shown safety of several of these therapies, and the first steps towards efficacy studies in humans have been made. In this review, we address the current state of the art of cellular therapies in clinical transplantation and discuss monitoring tools and endpoints for these studies.
Collapse
Affiliation(s)
- Martin J. Hoogduijn
- Nephrology and TransplantationDepartment of Internal MedicineErasmus University Medical CenterErasmus Medical CenterRotterdamThe Netherlands
| | - Fadi Issa
- Transplantation Research and Immunology GroupNuffield Department of Surgical SciencesJohn Radcliffe HospitalUniversity of OxfordOxfordUK
| | | | - Marlies E. J. Reinders
- Nephrology and TransplantationDepartment of Internal MedicineErasmus University Medical CenterErasmus Medical CenterRotterdamThe Netherlands
| |
Collapse
|
27
|
Tran LM, Thomson AW. Detection and Monitoring of Regulatory Immune Cells Following Their Adoptive Transfer in Organ Transplantation. Front Immunol 2020; 11:614578. [PMID: 33381125 PMCID: PMC7768032 DOI: 10.3389/fimmu.2020.614578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Application of cell-based immunotherapy in organ transplantation to minimize the burden of immunosuppressive medication and promote allograft tolerance has expanded significantly over the past decade. Adoptively transferred regulatory immune cells prolong allograft survival and transplant tolerance in pre-clinical models. Many cell products are currently under investigation in early phase human clinical trials designed to assess feasibility and safety. Despite rapid advances in manufacturing practices, defining the appropriate protocol that will optimize in vivo conditions for tolerance induction remains a major challenge and depends heavily on understanding the fate, biodistribution, functional stability and longevity of the cell product after administration. This review focuses on in vivo detection and monitoring of various regulatory immune cell types administered for allograft tolerance induction in both pre-clinical animal models and early human clinical trials. We discuss the current status of various non-invasive methods for tracking regulatory cell products in the context of organ transplantation and implications for enhanced understanding of the therapeutic potential of cell-based therapy in the broad context of control of immune-mediated inflammatory disorders.
Collapse
Affiliation(s)
- Lillian M Tran
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angus W Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
28
|
Abstract
Over the past decade, the clinical application of mesenchymal stromal cells (MSCs) has generated growing enthusiasm as an innovative cell-based approach in solid organ transplantation (SOT). These expectations arise from a significant number of both transplant- and non-transplant-related experimental studies investigating the complex anti-inflammatory, immunomodulatory, and tissue-repair properties of MSCs. Promising preclinical results have prompted clinical trials using MSC-based therapy in SOT. In the present review, the general properties of MSCs are summarized, with a particular emphasis on MSC-mediated impact on the immune system and in the ischemic conditioning strategy. Next, we chronologically detail all clinical trials using MSCs in the field of SOT. Finally, we envision the challenges and perspectives of MSC-based cell therapy in SOT.
Collapse
|
29
|
Liu D, Cheng F, Pan S, Liu Z. Stem cells: a potential treatment option for kidney diseases. Stem Cell Res Ther 2020; 11:249. [PMID: 32586408 PMCID: PMC7318741 DOI: 10.1186/s13287-020-01751-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
The prevalence of kidney diseases is emerging as a public health problem. Stem cells (SCs), currently considered as a promising tool for therapeutic application, have aroused considerable interest and expectations. With self-renewal capabilities and great potential for proliferation and differentiation, stem cell therapy opens new avenues for the development of renal function and structural repair in kidney diseases. Mounting evidence suggests that stem cells exert a therapeutic effect mainly by replacing damaged tissues and paracrine pathways. The benefits of various types of SCs in acute kidney disease and chronic kidney disease have been demonstrated in preclinical studies, and preliminary results of clinical trials present its safety and tolerability. This review will focus on the stem cell-based therapy approaches for the treatment of kidney diseases, including various cell sources used, possible mechanisms involved, and outcomes that are generated so far, along with prospects and challenges in clinical application.
Collapse
Affiliation(s)
- Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China
| | - Fei Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, People's Republic of China.
- Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, People's Republic of China.
| |
Collapse
|
30
|
Bulut Ö, GÜrsel İ. Mesenchymal stem cell derived extracellular vesicles: promising immunomodulators against autoimmune, autoinflammatory disorders and SARS-CoV-2 infection. ACTA ACUST UNITED AC 2020; 44:273-282. [PMID: 32595362 PMCID: PMC7314505 DOI: 10.3906/biy-2002-79] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Discovery of novel and broad-acting immunomodulators is of critical importance for the prevention and treatment of disorders occurring due to overexuberant immune responseincluding SARS-CoV-2 triggered cytokine storm leading to lung pathology and mortality during the ongoing viral pandemic. Mesenchymal stem/stromal cells (MSCs), highly regarded for their regenerative capacities, also possessesremarkable immunoregulatory functions affecting all types of innate and adaptive immune cells. Owing to that, MSCs have been heavily investigated in clinic for the treatment of autoimmune and inflammatory diseases along with transplant rejection. Extensive research in the last decaderevealed that MSCs carry out most of their functions through paracrine factors which are soluble mediators and extracellular vesicles (EVs). EVs, including exosomes and microvesicles, are an efficient way of intercellular communication due to their unique ability to carry biological messages such as transcription factors, growth factors, cytokines, mRNAs and miRNAs over long distances. EVs originate through direct budding of the cell membrane or the endosomal secretion pathway and they consist of the cytosolic and membrane components of their parent cell. Therefore, they are able to mimic the characteristics of the parent cell, affecting the target cells upon binding or internalization. EVs secreted by MSCs are emerging as a cell-free alternative to MSC-based therapies. MSC EVs are being tested in preclinical and clinical settings where they exhibit exceptional immunosuppressivecapacity. They regulate the migration, proliferation, activation and polarization of various immune cells, promoting a tolerogenic immune response while inhibiting inflammatory response. Being as effective immunomodulators as their parent cells, MSC EVs are also preferable over MSC-based therapies due to their lower risk of immunogenicity, tumorigenicity and overall superior safety. In this review, we present the outcomes of preclinical and clinical studies utilizing MSC EVs as therapeutic agents for the treatment of a wide variety of immunological disorders.
Collapse
Affiliation(s)
- Özlem Bulut
- Therapeutic Oligodeoxynucleotide Research Laboratory (THORLAB), Department of Molecular Biology and Genetics, Faculty of Science, İhsan Doğramacı Bilkent University, Ankara Turkey
| | - İhsan GÜrsel
- Therapeutic Oligodeoxynucleotide Research Laboratory (THORLAB), Department of Molecular Biology and Genetics, Faculty of Science, İhsan Doğramacı Bilkent University, Ankara Turkey
| |
Collapse
|
31
|
Gao C, Wang X, Lu J, Li Z, Jia H, Chen M, Chang Y, Liu Y, Li P, Zhang B, Du X, Qi F. Mesenchymal stem cells transfected with sFgl2 inhibit the acute rejection of heart transplantation in mice by regulating macrophage activation. Stem Cell Res Ther 2020; 11:241. [PMID: 32552823 PMCID: PMC7301524 DOI: 10.1186/s13287-020-01752-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have become a promising candidate for cell-based immune therapy for acute rejection (AR) after heart transplantation due to possessing immunomodulatory properties. In this study, we evaluated the efficacy of soluble fibronectin-like protein 2 (sFgl2) overexpressing mesenchymal stem cells (sFgl2-MSCs) in inhibiting AR of heart transplantation in mice by regulating immune tolerance through inducing M2 phenotype macrophage polarization. METHODS AND RESULTS The sFgl2, a novel immunomodulatory factor secreted by regulatory T cells, was transfected into MSCs to enhance their immunosuppressive functions. After being co-cultured for 72 h, the sFgl2-MSCs inhibited M1 polarization whereas promoted M2 of polarization macrophages through STAT1 and NF-κB pathways in vitro. Besides, the sFgl2-MSCs significantly enhanced the migration and phagocytosis ability of macrophages stimulated with interferon-γ (IFN-γ) and lipopolysaccharide (LPS). Further, the application potential of sFgl2-MSCs in AR treatment was demonstrated by heterotopic cardiac transplantation in mice. The tissue damage and macrophage infiltration were evaluated by H&E and immunohistochemistry staining, and the secretion of inflammatory cytokines was analyzed by ELISA. The results showed that sFgl2-MSCs injected intravenously were able to locate in the graft, promote the M2 polarization of macrophages in vivo, regulate the local and systemic immune response, significantly protect tissues from damaging, and finally prolonged the survival time of mice heart grafts. CONCLUSION sFgl2-MSCs ameliorate AR of heart transplantation by regulating macrophages, which provides a new idea for the development of anti-AR treatment methods after heart transplantation.
Collapse
Affiliation(s)
- Chao Gao
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Xiaodong Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310003, Zhejiang province, China
| | - Jian Lu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Zhilin Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Haowen Jia
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Minghao Chen
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Yuchen Chang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Yanhong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Peiyuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Baotong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Xuezhi Du
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Feng Qi
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin General Surgery Institute, Tianjin, 300052, China.
| |
Collapse
|
32
|
Sergeant E, Buysse M, Devos T, Sprangers B. Multipotent mesenchymal stromal cells in kidney transplant recipients: The next big thing? Blood Rev 2020; 45:100718. [PMID: 32507576 DOI: 10.1016/j.blre.2020.100718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 04/13/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Bone marrow-derived multipotent mesenchymal stromal cells (BM-MSCs) are non-haematopoietic cells present in the bone marrow stroma. They have the potential to modulate immune responses and exhibit a capacity to promote immune tolerance. Although the efficacy of immunosuppressive drugs has improved significantly, thereby ameliorating renal graft outcome, the use of these drugs still carries an increased risk of malignancies and opportunistic infections, and sometimes fail to prevent chronic allograft rejection or recurrence of the original kidney disease. As such, there is strong interest in ways to induce immune tolerance and thereby tempering or avoiding conventional immunosuppressive drugs. Cellular immunomodulation by MSCs can create a new way to induce transplant tolerance. This review will give a critical overview of the use of BM-MSCs as a cell-based immunosuppressive therapy in kidney transplant recipients. In vitro studies revealed several mechanisms that can clarify the immunomodulatory potential of BM-MSCs. Several clinical studies showed that BM-MSCs can modulate T-cell proliferation and can alter the ratio of T-cell subsets, favoring immune tolerance. However, this immunomodulation was often not associated with better clinical outcome during follow-up when compared to control groups. Some clinical studies found that BM-MSCs allow a reduction in dose of conventional immunosuppressive drugs and prevent acute graft dysfunction. Most clinical studies emphasized that BM-MSC infusion was safe. This review suggests that the use of BM-MSCs as cell-based immunosuppression therapy in kidney transplant recipients has potential, however some caution regarding their clinical use is appropriate. Mechanisms by which BM-MSCs induce transplant tolerance and factors that can alter their functionality need to be analyzed in more detail before clinical use.
Collapse
Affiliation(s)
- Elien Sergeant
- Division of Internal Medicine, University Hospitals Leuven, Leuven, Belgium.
| | - Malicorne Buysse
- Division of Hematology, University Hospitals Ghent, Ghent, Belgium.
| | - Timothy Devos
- Department of Microbiology and Immunology, Laboratory of Molecular Immunology (Rega Institute), KU Leuven, Leuven, Belgium; Division of Hematology, University Hospitals Leuven, Leuven, Belgium.
| | - Ben Sprangers
- Department of Microbiology and Immunology, Laboratory of Molecular Immunology (Rega Institute), KU Leuven, Leuven, Belgium; Division of Nephrology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
33
|
Zhao L, Hu C, Han F, Cai F, Wang J, Chen J. Preconditioning is an effective strategy for improving the efficiency of mesenchymal stem cells in kidney transplantation. Stem Cell Res Ther 2020; 11:197. [PMID: 32448356 PMCID: PMC7245776 DOI: 10.1186/s13287-020-01721-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/22/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022] Open
Abstract
The inevitable side effects caused by lifelong immunosuppressive agents in kidney transplantation patients spurred the exploration of novel immunosuppressive strategies with definite curative effects and minimal adverse effects. Mesenchymal stem cells (MSCs) have become a promising candidate due to their role in modulating the immune system. Encouraging results obtained from experimental models have promoted the translation of this strategy into clinical settings. However, the demonstration of only marginal or transient benefits by several recent clinical controlled studies has made physicians hesitant to adopt the routine utilization of this procedure in clinical settings. Impaired MSC function after infusion in vivo was thought to be the main reason for their limited effects. For this reason, some preconditioning methods were developed. In this review, we aim to outline the current understanding of the preconditioning methods being explored as a strategy to improve the therapeutic effects of MSCs in kidney transplantation and promote its clinical translation.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fei Han
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fanghao Cai
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Junni Wang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China. .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
34
|
Update on mesenchymal stromal cell studies in organ transplant recipients. Curr Opin Organ Transplant 2020; 25:27-34. [DOI: 10.1097/mot.0000000000000716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
35
|
Sávio-Silva C, Soinski-Sousa PE, Balby-Rocha MTA, Lira ÁDO, Rangel ÉB. Mesenchymal stem cell therapy in acute kidney injury (AKI): review and perspectives. REVISTA DA ASSOCIAÇÃO MÉDICA BRASILEIRA 2020; 66:s45-s54. [DOI: 10.1590/1806-9282.66.s1.45] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
36
|
Casiraghi F, Perico N, Gotti E, Todeschini M, Mister M, Cortinovis M, Portalupi V, Plati AR, Gaspari F, Villa A, Introna M, Longhi E, Remuzzi G. Kidney transplant tolerance associated with remote autologous mesenchymal stromal cell administration. Stem Cells Transl Med 2019; 9:427-432. [PMID: 31872574 PMCID: PMC7103624 DOI: 10.1002/sctm.19-0185] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Here we report the case of successful immune tolerance induction in a living‐donor kidney transplant recipient remotely treated with autologous bone marrow‐derived mesenchymal stromal cells (MSC). This case report, which to the best of our knowledge is the first in the world in this setting, provides evidence that the modulation of the host immune system with MSC can enable the safe withdrawal of maintenance immunosuppressive drugs while preserving optimal long‐term kidney allograft function.
Collapse
Affiliation(s)
| | - Norberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Eliana Gotti
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Marta Todeschini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marilena Mister
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Monica Cortinovis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Valentina Portalupi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Anna Rita Plati
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Flavio Gaspari
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Alessandro Villa
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Martino Introna
- G. Lanzani Laboratory of Cell Therapy, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Elena Longhi
- Laboratory of Transplant Immunology, UOC Coordinamento Trapianti IRCCS Fondazione Ca' Granda - Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
37
|
You Y, Wen DG, Gong JP, Liu ZJ. Research Status of Mesenchymal Stem Cells in Liver Transplantation. Cell Transplant 2019; 28:1490-1506. [PMID: 31512503 PMCID: PMC6923564 DOI: 10.1177/0963689719874786] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Liver transplantation has been deemed the best choice for end-stage liver disease
patients but immune rejection after surgery is still a serious problem. Patients have to
take immunosuppressive drugs for a long time after liver transplantation, and this often
leads to many side effects. Mesenchymal stem cells (MSCs) gradually became of interest to
researchers because of their powerful immunomodulatory effects. In the past, a large
number of in vitro and in vivo studies have demonstrated the great potential of MSCs for
participation in posttransplant immunomodulation. In addition, MSCs also have properties
that may potentially benefit patients undergoing liver transplantation. This article aims
to provide an overview of the current understanding of the immunomodulation achieved by
the application of MSCs in liver transplantation, to discuss the problems that may be
encountered when using MSCs in clinical practice, and to describe some of the underlying
capabilities of MSCs in liver transplantation. Cell–cell contact, soluble molecules, and
exosomes have been suggested to be critical approaches to MSCs’ immunoregulation in vitro;
however, the exact mechanism, especially in vivo, is still unclear. In recent years, the
clinical safety of MSCs has been proven by a series of clinical trials. The obstacles to
the clinical application of MSCs are decreasing, but large sample clinical trials
involving MSCs are still needed to further study their clinical effects.
Collapse
Affiliation(s)
- Yu You
- Hepatobiliary Surgery Department, Second Affiliated Hospital of Chongqing Medical University, China.,Yu You and Di-guang Wen are equal contributors and co-first authors of this article
| | - Di-Guang Wen
- Hepatobiliary Surgery Department, Second Affiliated Hospital of Chongqing Medical University, China.,Yu You and Di-guang Wen are equal contributors and co-first authors of this article
| | - Jian-Ping Gong
- Hepatobiliary Surgery Department, Second Affiliated Hospital of Chongqing Medical University, China
| | - Zuo-Jin Liu
- Hepatobiliary Surgery Department, Second Affiliated Hospital of Chongqing Medical University, China
| |
Collapse
|
38
|
Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci 2019; 76:3323-3348. [PMID: 31055643 PMCID: PMC11105258 DOI: 10.1007/s00018-019-03125-1] [Citation(s) in RCA: 355] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are isolated from multiple biological tissues-adult bone marrow and adipose tissues and neonatal tissues such as umbilical cord and placenta. In vitro, MSCs show biological features of extensive proliferation ability and multipotency. Moreover, MSCs have trophic, homing/migration and immunosuppression functions that have been demonstrated both in vitro and in vivo. A number of clinical trials are using MSCs for therapeutic interventions in severe degenerative and/or inflammatory diseases, including Crohn's disease and graft-versus-host disease, alone or in combination with other drugs. MSCs are promising for therapeutic applications given the ease in obtaining them, their genetic stability, their poor immunogenicity and their curative properties for tissue repair and immunomodulation. The success of MSC therapy in degenerative and/or inflammatory diseases might depend on the robustness of the biological functions of MSCs, which should be linked to their therapeutic potency. Here, we outline the fundamental and advanced concepts of MSC biological features and underline the biological functions of MSCs in their basic and translational aspects in therapy for degenerative and/or inflammatory diseases.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Benoit Favier
- CEA, DRF-IBFJ, IDMIT, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Paris-Sud University, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, Etablissement Français du Sang Occitanie, UMR 5273 CNRS, INSERM U1031, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Francois Jacob Institute, Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, IRSL, UMRS 976, Paris, France
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
39
|
The Anti-Inflammatory, Anti-Oxidative, and Anti-Apoptotic Benefits of Stem Cells in Acute Ischemic Kidney Injury. Int J Mol Sci 2019; 20:ijms20143529. [PMID: 31330934 PMCID: PMC6678402 DOI: 10.3390/ijms20143529] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/11/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) plays a significant role in the pathogenesis of acute kidney injury (AKI). The complicated interaction between injured tubular cells, activated endothelial cells, and the immune system leads to oxidative stress and systemic inflammation, thereby exacerbating the apoptosis of renal tubular cells and impeding the process of tissue repair. Stem cell therapy is an innovative approach to ameliorate IRI due to its antioxidative, immunomodulatory, and anti-apoptotic properties. Therefore, it is crucial to understand the biological effects and mechanisms of action of stem cell therapy in the context of acute ischemic AKI to improve its therapeutic benefits. The recent finding that treatment with conditioned medium (CM) derived from stem cells is likely an effective alternative to conventional stem cell transplantation increases the potential for future therapeutic uses of stem cell therapy. In this review, we discuss the recent findings regarding stem cell-mediated cytoprotection, with a focus on the anti-inflammatory effects via suppression of oxidative stress and uncompromised immune responses following AKI. Stem cell-derived CM represents a favorable approach to stem cell-based therapy and may serve as a potential therapeutic strategy against acute ischemic AKI.
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Mesenchymal stromal cells (MSC) have emerged as one of the most promising candidates for immunomodulatory cell therapy in kidney transplantation. Here we describe novel insights into the MSC mechanism of action and provide an overview of initial safety and feasibility studies with MSC in kidney transplantation. RECENT FINDINGS Clinical studies of MSC-based cell therapy in kidney transplant recipients demonstrated the safety and feasibility of cell therapy and provide the first encouraging evidence of the efficacy of MSC in enabling the minimization of immunosuppressive drugs. In our initial experience with MSC-based therapy in kidney transplant recipients we carried out extensive clinical and immunological monitoring of MSC-treated patients and found possible biomarkers of MSC immunomodulation in some of them. Based on these biomarkers we identified a patient in whom complete discontinuation of immunosuppression has been achieved safely and successfully. SUMMARY Many issues should be addressed before MSC-based therapy becomes a standard treatment protocol for kidney transplantation. A better understanding of the MSC mechanism of action and the identification of biomarkers of response to therapy will inform the rational design of the most effective clinical protocol and the selection of patients amenable to safe immunosuppressive drug withdrawal.
Collapse
|
41
|
Rota C, Morigi M, Imberti B. Stem Cell Therapies in Kidney Diseases: Progress and Challenges. Int J Mol Sci 2019; 20:ijms20112790. [PMID: 31181604 PMCID: PMC6600599 DOI: 10.3390/ijms20112790] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 12/15/2022] Open
Abstract
The prevalence of renal diseases is emerging as a public health problem. Despite major progress in supportive therapy, mortality rates among patients remain high. In an attempt to find innovative treatments to stimulate kidney regeneration, stem cell-based technology has been proposed as a potentially promising strategy. Here, we summarise the renoprotective potential of pluripotent and adult stem cell therapy in experimental models of acute and chronic kidney injury and we explore the different mechanisms at the basis of stem cell-induced kidney regeneration. Specifically, cell engraftment, incorporation into renal structures, or paracrine activities of embryonic or induced pluripotent stem cells as well as mesenchymal stem cells and renal precursors are analysed. We also discuss the relevance of stem cell secretome-derived bioproducts, including soluble factors and extracellular vesicles, and the option of using them as cell-free therapy to induce reparative processes. The translation of the experimental results into clinical trials is also addressed, highlighting the safety and feasibility of stem cell treatments in patients with kidney injury.
Collapse
Affiliation(s)
- Cinzia Rota
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy.
| | - Marina Morigi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy.
| | - Barbara Imberti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy.
| |
Collapse
|
42
|
Podestà MA, Remuzzi G, Casiraghi F. Mesenchymal Stromal Cells for Transplant Tolerance. Front Immunol 2019; 10:1287. [PMID: 31231393 PMCID: PMC6559333 DOI: 10.3389/fimmu.2019.01287] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/21/2019] [Indexed: 12/18/2022] Open
Abstract
In solid organ transplantation lifelong immunosuppression exposes transplant recipients to life-threatening complications, such as infections and malignancies, and to severe side effects. Cellular therapy with mesenchymal stromal cells (MSC) has recently emerged as a promising strategy to regulate anti-donor immune responses, allowing immunosuppressive drug minimization and tolerance induction. In this review we summarize preclinical data on MSC in solid organ transplant models, focusing on potential mechanisms of action of MSC, including down-regulation of effector T-cell response and activation of regulatory pathways. We will also provide an overview of available data on safety and feasibility of MSC therapy in solid organ transplant patients, highlighting the issues that still need to be addressed before establishing MSC as a safe and effective tolerogenic cell therapy in transplantation.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.,Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Remuzzi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Federica Casiraghi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
43
|
Wang S, Zhu R, Li H, Li J, Han Q, Zhao RC. Mesenchymal stem cells and immune disorders: from basic science to clinical transition. Front Med 2019; 13:138-151. [PMID: 30062557 DOI: 10.1007/s11684-018-0627-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023]
Abstract
As a promising candidate seed cell type in regenerative medicine, mesenchymal stem cells (MSCs) have attracted considerable attention. The unique capacity of MSCs to exert a regulatory effect on immunity in an autologous/allergenic manner makes them an attractive therapeutic cell type for immune disorders. In this review, we discussed the current knowledge of and advances in MSCs, including its basic biological properties, i.e., multilineage differentiation, secretome, and immunomodulation. Specifically, on the basis of our previous work, we proposed three new concepts of MSCs, i.e., "subtotipotent stem cell" hypothesis, MSC system, and "Yin and Yang" balance of MSC regulation, which may bring new insights into our understanding of MSCs. Furthermore, we analyzed data from the Clinical Trials database ( http://clinicaltrials.gov ) on registered clinical trials using MSCs to treat a variety of immune diseases, such as graft-versus-host disease, systemic lupus erythematosus, and multiple sclerosis. In addition, we highlighted MSC clinical trials in China and discussed the challenges and future directions in the field of MSC clinical application.
Collapse
Affiliation(s)
- Shihua Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Rongjia Zhu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hongling Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Qin Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Peking Union Medical College Hospital, Beijing, 100005, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
- Peking Union Medical College Hospital, Beijing, 100005, China.
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
44
|
Erpicum P, Weekers L, Detry O, Bonvoisin C, Delbouille MH, Grégoire C, Baudoux E, Briquet A, Lechanteur C, Maggipinto G, Somja J, Pottel H, Baron F, Jouret F, Beguin Y. Infusion of third-party mesenchymal stromal cells after kidney transplantation: a phase I-II, open-label, clinical study. Kidney Int 2019; 95:693-707. [DOI: 10.1016/j.kint.2018.08.046] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/25/2018] [Accepted: 08/23/2018] [Indexed: 02/08/2023]
|
45
|
Morsy M, Hossain MA, Bagul A. Exploring the Role of Mesenchymal Stem Cells During Normothermic Organ Perfusion: A New Paradigm to Enhance Outcome Following Allograft Transplantation. ACTA ACUST UNITED AC 2018. [DOI: 10.2174/1876893801805010047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background:
Normothermic Machine Perfusion (NMP) has been established in the field of solid organ transplantation for both liver and kidney allografts. The ability to perfuse organs at body temperature enables viability assessment as well as optimisation prior to implantation.
Discussion:
A recent in vitro report of the use of Mesenchymal Stem Cells (MSCs) in the use of a normothermic lung perfusion circuit has raised the possibility of their use in solid organ transplantation. The aim of this short review is to outline the potential uses of bone marrow derived MSCs for their use in renal allograft ex vivo NMP. An overview is provided of current literature of NMP as well as theorised uses for MSCs.
Collapse
|
46
|
Zhang Z, Wilson NA, Chinnadurai R, Panzer SE, Redfield RR, Reese SR, Galipeau J, Djamali A. Autologous Mesenchymal Stromal Cells Prevent Transfusion-elicited Sensitization and Upregulate Transitional and Regulatory B Cells. Transplant Direct 2018; 4:e387. [PMID: 30234156 PMCID: PMC6133404 DOI: 10.1097/txd.0000000000000827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/14/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We hypothesized that immunomodulatory properties of mesenchymal stromal cells (MSC) may be considered for desensitization. METHODS Autologous or allogeneic bone marrow derived MSC were infused via tail vein at 0.5 M (0.5 × 106), 1 M, or 2 M cells/dose on days -2, 3, 6, 9, 12 (prevention) or 14, 17, 20, 23, 26 (treatment) relative to transfusion in a Brown Norway to Lewis rat model (10 groups total, n = 6 per group). RESULTS At 4 weeks, pooled analyses demonstrated that autologous and allogeneic MSC were equally effective in reducing IgG1 and IgG2a de novo donor-specific antibody (dnDSA, P < 0.001). Dose-response studies indicated that moderate-dose MSC (5 M total) was most effective in reducing IgG1, IgG2a, and IgG2c dnDSA (P ≤ 0.01). Time course studies determined that preventive and treatment strategies were equally effective in reducing IgG1 and IgG2a dnDSA (P ≤ 0.01). However, individual group analyses determined that moderate-dose (5 M) treatment with autologous MSC was most effective in reducing IgG1, IgG2a, and IgG2c dnDSA (P ≤ 0.01). In this group, dnDSA decreased after 1 week of treatment; regulatory B cells increased in the spleen and peripheral blood mononuclear cells; and transitional B cells increased in the spleen, peripheral blood mononuclear cells, and bone marrow (P < 0.05 for all). CONCLUSIONS Our findings indicate that autologous MSC prevent transfusion-elicited sensitization and upregulate transitional, and regulatory B cells. Additional studies are needed to determine the biological relevance of these changes after kidney transplantation.
Collapse
Affiliation(s)
- Zijian Zhang
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI
- Department of Urology, Beijing Chao-Yang Hospital, China Capital Medical University, Beijing, China
| | - Nancy A. Wilson
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI
| | - Raghavan Chinnadurai
- Division of Hematology-Oncology, Department of Medicine, University of Wisconsin, Madison, WI
| | - Sarah E. Panzer
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI
| | - Robert R. Redfield
- Division of Transplant, Department of Surgery, University of Wisconsin, Madison, WI
| | - Shannon R. Reese
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI
| | - Jacques Galipeau
- Division of Hematology-Oncology, Department of Medicine, University of Wisconsin, Madison, WI
| | - Arjang Djamali
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI
- Division of Transplant, Department of Surgery, University of Wisconsin, Madison, WI
| |
Collapse
|
47
|
Wang D, Sun YQ, Gao WX, Fan XL, Shi JB, Fu QL. An in Vitro and in Vivo Study of the Effect of Dexamethasone on Immunoinhibitory Function of Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells. Cell Transplant 2018; 27:1340-1351. [PMID: 30056763 PMCID: PMC6168992 DOI: 10.1177/0963689718780194] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) represent a promising cell source for patient-specific cell therapy. We previously demonstrated that they display an immunomodulatory effect on allergic airway inflammation. Glucocorticoids are powerful anti-inflammatory compounds and widely used in the therapy of allergic diseases. However, the effect of glucocorticoids on the immunomodulatory function of iPSC-MSCs remains unknown. This study aimed to determine the effect of dexamethasone (Dex) on the immunomodulatory function of iPSC-MSCs in vitro and in vivo. A total of three human iPSC-MSC clones were generated from amniocyte-derived iPSCs. Anti-CD3/CD28-induced peripheral blood mononuclear cell (PBMC) proliferation was used to assess the effect of Dex on the immunoinhibitory function of iPSC-MSCs in vitro. Mouse models of contact hypersensitivity (CHS) and allergic airway inflammation were induced, and the levels of inflammation in mice were analyzed with the treatments of iPSC-MSCs and Dex, alone and combined. The results showed that Dex did not interfere with the immunoinhibitory effect of iPSC-MSCs on PBMC proliferation. In CHS mice, simultaneous treatment with Dex did not affect the effect of iPSC-MSCs on the inflammation, both in regional draining lymph nodes and in inflamed ear tissue. In addition, co-administration of iPSC-MSCs with Dex decreased the local expression of interferon (IFN)-γ and tumor necrosis factor (TNF)-α in the ears of CHS mice. In the mouse model of allergic airway inflammation, iPSC-MSC treatment combined with Dex resulted in a similar extent of reduction in pulmonary inflammation as iPSC-MSCs or Dex treatment alone. In conclusion, Dex does not significantly affect the immunomodulatory function of iPSC-MSCs both in vitro and in vivo. These findings may have implications when iPSC-MSCs and glucocorticoids are co-administered.
Collapse
Affiliation(s)
- Dan Wang
- 1 Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yue-Qi Sun
- 1 Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wen-Xiang Gao
- 1 Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xing-Liang Fan
- 1 Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian-Bo Shi
- 1 Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qing-Ling Fu
- 1 Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
48
|
Perico N, Casiraghi F, Todeschini M, Cortinovis M, Gotti E, Portalupi V, Mister M, Gaspari F, Villa A, Fiori S, Introna M, Longhi E, Remuzzi G. Long-Term Clinical and Immunological Profile of Kidney Transplant Patients Given Mesenchymal Stromal Cell Immunotherapy. Front Immunol 2018; 9:1359. [PMID: 29963053 PMCID: PMC6014158 DOI: 10.3389/fimmu.2018.01359] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 05/31/2018] [Indexed: 12/11/2022] Open
Abstract
We report here the long-term clinical and immunological results of four living-donor kidney transplant patients given autologous bone marrow-derived mesenchymal stromal cells (MSCs) as part of a phase 1 study focused on the safety and feasibility of this cell therapy. According to study protocols implemented over time, based on initial early safety findings, the patients were given MSC at day 7 posttransplant (n = 2) or at day −1 pretransplant (n = 2) and received induction therapy with basiliximab and low-dose rabbit anti-thymocyte globulin (RATG) or RATG alone, and were maintained on low-dose ciclosporin (CsA)/mycophenolate mofetil (MMF). All MSC-treated patients had stable graft function during the 5- to 7-year follow-up, without increased susceptibility to infections or neoplasm. In three MSC recipients, but not historical control patients, circulating memory CD8+ T cell percentages remained lower than basal, coupled with persistent reduction of ex vivo donor-specific cytotoxicity. Two patients showed a long-lasting increase in the regulatory T cell/memory CD8+ T cell ratio, paralleled by high circulating levels of naïve and transitional B cells. In one of these two patients, CsA was successfully discontinued, and currently the low-dose MMF monotherapy is on the tapering phase. The study shows that MSC therapy is safe in the long term and could promote a pro-tolerogenic environment in selected patients. Extensive immunomonitoring of MSC-treated kidney transplant recipients could help selection of patients for safe withdrawal of maintenance immunosuppressive drugs (NCT00752479 and NCT02012153).
Collapse
Affiliation(s)
- Norberto Perico
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | | | - Marta Todeschini
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Monica Cortinovis
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Eliana Gotti
- Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Valentina Portalupi
- Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Marilena Mister
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Flavio Gaspari
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Alessandro Villa
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Sonia Fiori
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Martino Introna
- G. Lanzani Laboratory of Cell Therapy, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Elena Longhi
- Laboratory of Transplant Immunology, UOC Coordinamento Trapianti IRCCS Fondazione Ca' Granda - Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Giuseppe Remuzzi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy.,Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
49
|
Meng X, Chen M, Su W, Tao X, Sun M, Zou X, Ying R, Wei W, Wang B. The differentiation of mesenchymal stem cells to vascular cells regulated by the HMGB1/RAGE axis: its application in cell therapy for transplant arteriosclerosis. Stem Cell Res Ther 2018; 9:85. [PMID: 29615103 PMCID: PMC5883535 DOI: 10.1186/s13287-018-0827-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/18/2018] [Accepted: 03/06/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation shows promise for treating transplant arteriosclerosis, at least partly via promoting endothelial regeneration. However, the efficacy and safety are still under investigation especially regarding recent findings that neointimal smooth muscle cells are derived from MSC-like cells. The high mobility group box 1 (HMGB1)/receptor for advanced glycation end-product (RAGE) axis is involved in regulating proliferation, migration, and differentiation of MSCs, and therefore it can be presumably applied to improve the outcome of cell therapy. The aim of the current study was to investigate this hypothesis. METHODS Rat MSCs were treated with HMGB1 or modified with HMGB1 vectors to activate the HMGB1/RAGE axis. RAGE was targeted and inhibited by specific short hairpin RNA vectors. We assessed the capacity for cell proliferation, migration, and differentiation after vector transfection in vitro and in a rat model of transplant arteriosclerosis. The expression of CD31 and α-smooth muscle actin (αSMA) was determined to evaluate the differentiation of MSCs to endothelial cells and smooth muscle cells. RESULTS Exogenous HMGB1 treatment and transfection with HMGB1 vectors promoted MSC migration and vascular endothelial growth factor (VEGF)-induced differentiation to CD31+ cells while inhibiting their proliferation and platelet-derived growth factor (PDGF)-induced differentiation to αSMA+ cells. Such an effect was blocked by RAGE knockdown. HMGB1-modified cells preferably migrated to graft neointima and differentiated to CD31+ cells along with significant relief of transplant arteriosclerosis and inhibition of HMGB1 and RAGE expression in graft vessels. RAGE knockdown inhibited cell migration to graft vessels. CONCLUSIONS HMGB1 stimulated MSCs to migrate and differentiate to endothelial cells via RAGE signaling, which we translated to successful application in cell therapy for transplant arteriosclerosis.
Collapse
Affiliation(s)
- Xiaohu Meng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China
| | - Min Chen
- Department of Gastroenterology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Wenjie Su
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital Affiliated to Nanjing Medical University, Hangzhou, China
| | - Xuan Tao
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China
| | - Mingyang Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Rongchao Ying
- Department of Gastroenterological Surgery, Hangzhou First People's Hospital Affiliated to Nanjing Medical University, Hangzhou, China
| | - Wei Wei
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China.
| | - Baolin Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan, Nanjing, 210011, China.
| |
Collapse
|
50
|
Kaundal U, Bagai U, Rakha A. Immunomodulatory plasticity of mesenchymal stem cells: a potential key to successful solid organ transplantation. J Transl Med 2018; 16:31. [PMID: 29448956 PMCID: PMC5815241 DOI: 10.1186/s12967-018-1403-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
Organ transplantation remains to be a treatment of choice for patients suffering from irreversible organ failure. Immunosuppressive (IS) drugs employed to maintain the allograft have shown excellent short-term graft survival, but, their long-term use could contribute to immunological and non-immunological risk factors, resulting in graft dysfunctionalities. Upcoming IS regimes have highlighted the use of cell-based therapies, which can eliminate the risk of drug-borne toxicities while maintaining efficacy of the treatment. Mesenchymal stem cells (MSCs) have been considered as an invaluable cell type, owing to their unique immunomodulatory properties, which makes them desirable for application in transplant settings, where hyper-activation of the immune system is evident. The immunoregulatory potential of MSCs holds true for preclinical studies while achieving it in clinical studies continues to be a challenge. Understanding the biological factors responsible for subdued responses of MSCs in vivo would allow uninhibited use of this therapy for countless conditions. In this review, we summarize the variations in the preclinical and clinical studies utilizing MSCs, discuss the factors which might be responsible for variability in outcome and propose the advancements likely to occur in future for using this as a "boutique/personalised therapy" for patient care.
Collapse
Affiliation(s)
- Urvashi Kaundal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, India
- Department of Zoology, Panjab University, Sector 14, Chandigarh, India
| | - Upma Bagai
- Department of Zoology, Panjab University, Sector 14, Chandigarh, India
| | - Aruna Rakha
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh, India
| |
Collapse
|