1
|
Xu Q, Gu L, Li Z, Gao L, Wei L, Shafiq Z, Chen S, Cai Q. Current Status of Research on Nanomaterials Combined with Mesenchymal Stem Cells for the Treatment of Ischemic Stroke. Neuromolecular Med 2024; 26:51. [PMID: 39644405 DOI: 10.1007/s12017-024-08819-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024]
Abstract
Ischemic stroke (IS) is a disease with high mortality and disability rates worldwide and is a serious threat to patient health. Owing to the narrow therapeutic window, effective treatments during the recovery period are limited. However, in recent years, mesenchymal stem cells (MSCs) have attracted attention and have shown therapeutic potential in IS treatment because of their abilities to home and secrete multiple bioactive substances and potential for differentiation and substitution. The therapeutic mechanisms of MSCs in IS include the regulatory effects of MSCs on microglia, the dual role of MSCs in astrocytes, how MSCs connect innate and adaptive immunity, the secretion of cytokines by MSCs to counteract apoptosis and MSC apoptosis, the promotion of angiogenesis by MSCs to favor the restoration of the blood‒brain barrier (BBB), and the potential function of local neural replacement by MSCs. However, the low graft survival rate, insufficient homing, poor targeting, and inability to achieve directional differentiation of MSCs limit their wide application. As an approach to compensate for the shortcomings of MSCs, scientists have used nanomaterials to assist MSCs in homing, survival and proliferation. In addition, the unique material of nanomaterials adds tracking, imaging and real-time monitoring to stroke treatment. The identification of effective treatments for stroke is urgently needed; thus, an understanding of how MSCs treat stroke and further improvements in the use of nanomaterials are necessary.
Collapse
Affiliation(s)
- Qingxue Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhiyang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lu Wei
- Department of Anesthesiology, Eastern Campus, Renmin Hospital of Wuhan University, Wuhan, 430200, China
| | - Zohaib Shafiq
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shigui Chen
- The Institute for Advanced Studies, Wuhan University, 299 Bayi Road, Wuhan, 430072, Hubei, China.
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
Hendriks SH, Heidt S, Krop J, IJsselsteijn ME, Eggermont J, Kers J, Reinders ME, Koning F, van Kooten C. IDO + Endothelial Cells in Glomeruli of Kidney Transplantation Patients With Glomerulitis. Transplant Direct 2024; 10:e1674. [PMID: 38988690 PMCID: PMC11230740 DOI: 10.1097/txd.0000000000001674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/14/2024] [Indexed: 07/12/2024] Open
Abstract
Background Kidney transplantation is the preferred treatment option for patients with end-stage renal disease. However, long-term graft survival remains a challenge. The enzyme indoleamine 2,3 dioxygenase (IDO) has been reported to have immunomodulatory effects with IDO transcripts being elevated in both antibody-mediated rejection and T cell-mediated rejection. Methods A metal-conjugated antibody panel for the staining of kidney biopsies was developed, allowing the visualization of 41 structural and immune markers on a single tissue slide to gain in-depth insight into the composition and localization of the immune cell compartment. Staining was applied to week 4 and 24 protocol biopsies of 49 patients as well as on 15 indication biopsies of the TRITON study and 4 additional transplantation biopsies with glomerulitis. Results A highly distinctive and specific glomerular IDO expression was observed in biopsies from 3 of 49 patients in imaging mass cytometry. Immunohistochemistry confirmed IDO expression in glomeruli of 10 of 10 cases with glomerulitis. IDO was found to be expressed by CD31+ glomerular endothelial cells, accompanied by the presence of granzyme-B+Tbet+CD7+CD45RA+ natural killer cells and CD68+ macrophages. Furthermore, a proportion of both the immune cells and endothelial cells expressed Ki-67, indicative of cell proliferation, which was not observed in control glomeruli. Conclusions Our results show glomerular IDO expression in transplanted kidneys with glomerulitis, which is accompanied by increased numbers of natural killer cells and macrophages and likely reflects local immune activation.
Collapse
Affiliation(s)
- Sanne H. Hendriks
- Department of Immunology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Juliette Krop
- Department of Immunology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Marieke E. IJsselsteijn
- Department of Pathology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Jeroen Eggermont
- Department of LKEB Radiology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Jesper Kers
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marlies E.J. Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| |
Collapse
|
3
|
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
|
4
|
Faria J, Calcat-I-Cervera S, Skovronova R, Broeksma BC, Berends AJ, Zaal EA, Bussolati B, O'Brien T, Mihăilă SM, Masereeuw R. Mesenchymal stromal cells secretome restores bioenergetic and redox homeostasis in human proximal tubule cells after ischemic injury. Stem Cell Res Ther 2023; 14:353. [PMID: 38072933 PMCID: PMC10712181 DOI: 10.1186/s13287-023-03563-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Ischemia/reperfusion injury is the leading cause of acute kidney injury (AKI). The current standard of care focuses on supporting kidney function, stating the need for more efficient and targeted therapies to enhance repair. Mesenchymal stromal cells (MSCs) and their secretome, either as conditioned medium (CM) or extracellular vesicles (EVs), have emerged as promising options for regenerative therapy; however, their full potential in treating AKI remains unknown. METHODS In this study, we employed an in vitro model of chemically induced ischemia using antimycin A combined with 2-deoxy-D-glucose to induce ischemic injury in proximal tubule epithelial cells. Afterwards we evaluated the effects of MSC secretome, CM or EVs obtained from adipose tissue, bone marrow, and umbilical cord, on ameliorating the detrimental effects of ischemia. To assess the damage and treatment outcomes, we analyzed cell morphology, mitochondrial health parameters (mitochondrial activity, ATP production, mass and membrane potential), and overall cell metabolism by metabolomics. RESULTS Our findings show that ischemic injury caused cytoskeletal changes confirmed by disruption of the F-actin network, energetic imbalance as revealed by a 50% decrease in the oxygen consumption rate, increased oxidative stress, mitochondrial dysfunction, and reduced cell metabolism. Upon treatment with MSC secretome, the morphological derangements were partly restored and ATP production increased by 40-50%, with umbilical cord-derived EVs being most effective. Furthermore, MSC treatment led to phenotype restoration as indicated by an increase in cell bioenergetics, including increased levels of glycolysis intermediates, as well as an accumulation of antioxidant metabolites. CONCLUSION Our in vitro model effectively replicated the in vivo-like morphological and molecular changes observed during ischemic injury. Additionally, treatment with MSC secretome ameliorated proximal tubule damage, highlighting its potential as a viable therapeutic option for targeting AKI.
Collapse
Affiliation(s)
- João Faria
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Sandra Calcat-I-Cervera
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Renata Skovronova
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | - Alinda J Berends
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Esther A Zaal
- Division of Cell Biology, Metabolism and Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Timothy O'Brien
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Silvia M Mihăilă
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Siemionow M, Kulahci Y, Zor F. Novel cell-based strategies for immunomodulation in vascularized composite allotransplantation. Curr Opin Organ Transplant 2023; 28:431-439. [PMID: 37800652 DOI: 10.1097/mot.0000000000001109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
PURPOSE OF REVIEW Vascularized composite allotransplantation (VCA) has become a clinical reality in the past two decades. However, its routine clinical applications are limited by the risk of acute rejection, and the side effects of the lifelong immunosuppression. Therefore, there is a need for new protocols to induce tolerance and extend VCA survival. Cell- based therapies have emerged as an attractive strategy for tolerance induction in VCA. This manuscript reviews the current strategies and applications of cell-based therapies for tolerance induction in VCA. RECENT FINDINGS Cellular therapies, including the application of bone marrow cells (BMC), mesenchymal stem cells (MSC), adipose stem cells, regulatory T cells (Treg) cells, dendritic cells and donor recipient chimeric cells (DRCC) show promising potential as a strategy to induce tolerance in VCA. Ongoing basic science research aims to provide insights into the mechanisms of action, homing, functional specialization and standardization of these cellular therapies. Additionally, translational preclinical and clinical studies are underway, showing encouraging outcomes. SUMMARY Cellular therapies hold great potential and are supported by preclinical studies and clinical trials demonstrating safety and efficacy. However, further research is needed to develop novel cell-based immunosuppressive protocol for VCA.
Collapse
Affiliation(s)
- Maria Siemionow
- Department of Orthopeadics, University of Illinois at Chicago, Chicago, Illinois
| | - Yalcin Kulahci
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Fatih Zor
- Department of Plastic Surgery, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Dreyer GJ, Drabbels JJM, de Fijter JW, van Kooten C, Reinders MEJ, Heidt S. Cell-free DNA measurement of three genomes after allogeneic MSC therapy in kidney transplant recipients indicates early cell death of infused MSC. Front Immunol 2023; 14:1240347. [PMID: 38022634 PMCID: PMC10652747 DOI: 10.3389/fimmu.2023.1240347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Mesenchymal stromal cell (MSC) therapy is a promising treatment that allows for drug minimization in clinical kidney transplantation. While it is thought that MSCs rapidly go into apoptosis after infusion, clinical evidence for this is scarce since methods to detect cell death of infused cells in vivo are lacking. Cell-free DNA (cfDNA) has recently gained attention as a biomarker for cell death. Methods In this study, we longitudinally measured cfDNA in plasma samples of the recipient, kidney donor, and allogeneic third-party MSC in the context of the Neptune study. cfDNA levels were measured at several time points before and after allogeneic MSC infusion in the 10 recipients who participated in the Neptune study. cfDNA ratios between the recipient, kidney graft, and MSC were determined. Results We observed a peak in MSC-derived cfDNA 4 h after the first and second infusions, after which MSC-derived cfDNA became undetectable. Generally, kidney graft-derived cfDNA remained in the baseline-level range. Discussion Our results support preclinical data that MSC are short-lived after infusion, also in a clinical in vivo setting, and are relevant for further research into the mechanism of action of MSC therapy.
Collapse
Affiliation(s)
- Geertje J. Dreyer
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
| | - Jos JM. Drabbels
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Johan W. de Fijter
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
| | - Marlies EJ. Reinders
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
7
|
Klinkhammer BM, Boor P. Kidney fibrosis: Emerging diagnostic and therapeutic strategies. Mol Aspects Med 2023; 93:101206. [PMID: 37541106 DOI: 10.1016/j.mam.2023.101206] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
An increasing number of patients worldwide suffers from chronic kidney disease (CKD). CKD is accompanied by kidney fibrosis, which affects all compartments of the kidney, i.e., the glomeruli, tubulointerstitium, and vasculature. Fibrosis is the best predictor of progression of kidney diseases. Currently, there is no specific anti-fibrotic therapy for kidney patients and invasive renal biopsy remains the only option for specific detection and quantification of kidney fibrosis. Here we review emerging diagnostic approaches and potential therapeutic options for fibrosis. We discuss how translational research could help to establish fibrosis-specific endpoints for clinical trials, leading to improved patient stratification and potentially companion diagnostics, and facilitating and optimizing development of novel anti-fibrotic therapies for kidney patients.
Collapse
Affiliation(s)
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany; Division of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
8
|
Hendriks SH, Heidt S, Schulz AR, de Fijter JW, Reinders MEJ, Koning F, van Kooten C. Peripheral Blood Immune Cell Composition After Autologous MSC Infusion in Kidney Transplantation Recipients. Transpl Int 2023; 36:11329. [PMID: 37426430 PMCID: PMC10326287 DOI: 10.3389/ti.2023.11329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
Tacrolimus is the backbone of immunosuppressive agents to prevent transplant rejection. Paradoxically, tacrolimus is nephrotoxic, causing irreversible tubulointerstitial damage. Therefore, infusion of mesenchymal stromal cells (MSC) 6 and 7 weeks post-transplantation was assessed to facilitate withdrawal of tacrolimus in the randomized phase II TRITON trial. Here, we performed detailed analysis of the peripheral blood immune composition using mass cytometry to assess potential effects of MSC therapy on the immune system. We developed two metal-conjugated antibody panels containing 40 antibodies each. PBMC samples from 21 MSC-treated patients and 13 controls, obtained pre-transplant and at 24 and 52 weeks post-transplantation, were analyzed. In the MSC group at 24 weeks, 17 CD4+ T cell clusters were increased of which 14 Th2-like clusters and three Th1/Th2-like clusters, as well as CD4+FoxP3+ Tregs. Additionally, five B cell clusters were increased, representing either class switched memory B cells or proliferating B cells. At 52 weeks, CCR7+CD38+ mature B cells were decreased. Finally, eight Tc1 (effector) memory cytotoxic T cell clusters were increased. Our work provides a comprehensive account of the peripheral blood immune cell composition in kidney transplant recipients after MSC therapy and tacrolimus withdrawal. These results may help improving therapeutic strategies using MSCs with the aim to reduce the use of calcineurin inhibitors. Clinical Trial Registration: ClinicalTrials.gov, identifier NCT02057965.
Collapse
Affiliation(s)
- Sanne H. Hendriks
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Axel R. Schulz
- German Rheumatism Research Center (DRFZ), Berlin, Germany
| | - Johan W. de Fijter
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
| | - Marlies E. J. Reinders
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC Transplant Institute, Erasmus University Medical Center, Leiden, Netherlands
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant Center, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
9
|
Al-Azab M, Idiiatullina E, Safi M, Hezam K. Enhancers of mesenchymal stem cell stemness and therapeutic potency. Biomed Pharmacother 2023; 162:114356. [PMID: 37040673 DOI: 10.1016/j.biopha.2023.114356] [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: 12/08/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 04/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that can differentiate into a range of cell types, including osteoblasts, chondrocytes, myocytes, and adipocytes. Multiple preclinical investigations and clinical trials employed enhanced MSCs-dependent therapies in treatment of inflammatory and degenerative diseases. They have demonstrated considerable and prospective therapeutic potentials even though the large-scale use remains a problem. Several strategies have been used to improve the therapeutic potency of MSCs in cellular therapy. Treatment of MSCs utilizing pharmaceutical compounds, cytokines, growth factors, hormones, and vitamins have shown potential outcomes in boosting MSCs' stemness. In this study, we reviewed the current advances in enhancing techniques that attempt to promote MSCs' therapeutic effectiveness in cellular therapy and stemness in vivo with potential mechanisms and applications.
Collapse
Affiliation(s)
- Mahmoud Al-Azab
- Department of Immunology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Elina Idiiatullina
- Department of Immunology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China; Department of Therapy and Nursing, Bashkir State Medical University, Ufa 450008, Russia
| | - Mohammed Safi
- Department of Respiratory Diseases, Shandong Second Provincial General Hospital, Shandong University, Shandong, China
| | - Kamal Hezam
- Nankai University School of Medicine, Tianjin 300071, China; Department of Microbiology, Faculty of Applied Science, Taiz University, 6350 Taiz, Yemen
| |
Collapse
|
10
|
Tao R, Qu Z, Zhang K, Chen J, Wang X, Deng Y. Substance P modulates BMSCs migration for tissue repair through NK-1R/CXCR4/p-Akt signal activation. Mol Biol Rep 2022; 49:2227-2236. [PMID: 35034285 DOI: 10.1007/s11033-021-07044-y] [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: 09/02/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND The migration of bone marrow-derived mesenchymal stem cells (BMSCs) to the wound site played an important role in tissue repair. Substance P (SP) has been studied and reported to be involved in tissue repair by promoting the growth of endothelial cells and the migration of BMSCs. However, the complicated process and the molecular mechanisms were not fully understood. Thus, we aimed to investigate the effect of SP-induced BMSCs migration on tissue repair and its possible mechanism. METHODS AND RESULTS Western blot and q-PCR assay revealed that SP could induce the BMSCs migration through overexpression of CXCR4 and upregulation of Akt phosphorylation. And the upregulation was related to the activation of neurokinin-1 receptor (NK-1R). Besides, we found that the increased phosphorylation Akt caused by SP could be canceled by the inhibition of CXCR4 both in vitro and in vivo. Furthermore, a skin-injury animal model was established and used to observe the tissue repair process. Results showed that SP could accelerate wound closure, gain more granulation tissue accumulation, and more collagen deposition through the promotion of angiogenesis and induction of the BMSCs migration to the wound site. And these effects could be impaired by inhibition of CXCR4 and p-Akt. CONCLUSIONS Our results suggested that SP promoted tissue repair through BMSCs migration via upregulation of CXCR4 and p-Akt. The expression of CXCR4 and p-Akt were regulated by NK-1R activation. These findings add more evidence in understanding the mechanisms of SP-induced BMSCs migration and highlight the potential for clinical implementation of SP in tissue repair.
Collapse
Affiliation(s)
- Ran Tao
- Day Surgery Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China
| | - Zhan Qu
- Department of Essential Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, People's Republic of China
| | - Ke Zhang
- Day Surgery Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China
| | - Jie Chen
- Day Surgery Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China
| | - Xinyu Wang
- Department of Gastrointestinal Surgery, The First Hospital of Changsha, Changsha, 410008, Hunan Province, People's Republic of China
| | - Youming Deng
- Department of Essential Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, People's Republic of China.
| |
Collapse
|
11
|
Bernaldo-de-Quirós E, Pion M, Martínez-Bonet M, Correa-Rocha R. A New Generation of Cell Therapies Employing Regulatory T Cells (Treg) to Induce Immune Tolerance in Pediatric Transplantation. Front Pediatr 2022; 10:862807. [PMID: 35633970 PMCID: PMC9130702 DOI: 10.3389/fped.2022.862807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Kidney transplantation is the most common solid organ transplant and the preferred treatment for pediatric patients with end-stage renal disease, but it is still not a definitive solution due to immune graft rejection. Regulatory T cells (Treg) and their control over effector T cells is a crucial and intrinsic tolerance mechanism in limiting excessive immune responses. In the case of transplants, Treg are important for the survival of the transplanted organ, and their dysregulation could increase the risk of rejection in transplanted children. Chronic immunosuppression to prevent rejection, for which Treg are especially sensitive, have a detrimental effect on Treg counts, decreasing the Treg/T-effector balance. Cell therapy with Treg cells is a promising approach to restore this imbalance, promoting tolerance and thus increasing graft survival. However, the strategies used to date that employ peripheral blood as a Treg source have shown limited efficacy. Moreover, it is not possible to use this approach in pediatric patients due to the limited volume of blood that can be extracted from children. Here, we outline our innovative strategy that employs the thymus removed during pediatric cardiac surgeries as a source of therapeutic Treg that could make this therapy accessible to transplanted children. The advantageous properties and the massive amount of Treg cells obtained from pediatric thymic tissue (thyTreg) opens a new possibility for Treg therapies to prevent rejection in pediatric kidney transplants. We are recruiting patients in a clinical trial to prevent rejection in heart-transplanted children through the infusion of autologous thyTreg cells (NCT04924491). If its efficacy is confirmed, thyTreg therapy may establish a new paradigm in preventing organ rejection in pediatric transplants, and their allogeneic use would extend its application to other solid organ transplantation.
Collapse
Affiliation(s)
- Esther Bernaldo-de-Quirós
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
12
|
Li J, Thomson AW, Rogers NM. Myeloid and Mesenchymal Stem Cell Therapies for Solid Organ Transplant Tolerance. Transplantation 2021; 105:e303-e321. [PMID: 33756544 PMCID: PMC8455706 DOI: 10.1097/tp.0000000000003765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transplantation is now performed globally as a routine procedure. However, the increased demand for donor organs and consequent expansion of donor criteria has created an imperative to maximize the quality of these gains. The goal is to balance preservation of allograft function against patient quality-of-life, despite exposure to long-term immunosuppression. Elimination of immunosuppressive therapy to avoid drug toxicity, with concurrent acceptance of the allograft-so-called operational tolerance-has proven elusive. The lack of recent advances in immunomodulatory drug development, together with advances in immunotherapy in oncology, has prompted interest in cell-based therapies to control the alloimmune response. Extensive experimental work in animals has characterized regulatory immune cell populations that can induce and maintain tolerance, demonstrating that their adoptive transfer can promote donor-specific tolerance. An extension of this large body of work has resulted in protocols for manufacture, as well as early-phase safety and feasibility trials for many regulatory cell types. Despite the excitement generated by early clinical trials in autoimmune diseases and organ transplantation, there is as yet no clinically validated, approved regulatory cell therapy for transplantation. In this review, we summarize recent advances in this field, with a focus on myeloid and mesenchymal cell therapies, including current understanding of the mechanisms of action of regulatory immune cells, and clinical trials in organ transplantation using these cells as therapeutics.
Collapse
Affiliation(s)
- Jennifer Li
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Angus W Thomson
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Natasha M Rogers
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
13
|
Wang Z, Xu H, Cheng F, Zhang J, Feng Y, Liu D, Shang W, Feng G. Donor BMSC-derived small extracellular vesicles relieve acute rejection post-renal allograft through transmitting Loc108349490 to dendritic cells. Aging Cell 2021; 20:e13461. [PMID: 34499402 PMCID: PMC8520728 DOI: 10.1111/acel.13461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cell (BMSC)-derived small extracellular vesicles (sEVs) are potent candidates for the suppression of acute rejection post-renal allograft and have been reported to halt dendritic cells (DCs) maturation. However, whether BMSC-derived sEVs mitigate acute rejection post-renal allograft by targeting DCs is still unclear. In this study, donor BMSC-derived sEVs (sEVs) relieved the inflammatory response and suppressed mature DCs (mDCs) location in kidney grafts, and increased regulatory T (Treg) cell population in the spleens of the rats that underwent kidney allograft. In lipopolysaccharide (LPS)-stimulated immature DCs (imDCs), sEVs suppressed the maturation and migration of DCs and inactivated toll-like receptor 4 (TLR4) signaling. Compared with LPS-treated imDCs, imDCs treated with LPS+sEVs promoted CD4+ T cells differentiated toward Treg cells. Subsequently, we found that Loc108349490, a long non-coding RNA (lncRNA) abundant in sEVs, mediated the inhibitory effect of sEVs on DC maturation and migration by promoting TLR4 ubiquitination. In rats that underwent an allograft, Loc108349490 deficiency weakened the therapeutic effect of sEVs on acute rejection. The present study firstly found that sEVs alleviated acute rejection post-renal allograft by transferring lncRNA to DCs and screened out the functional lncRNA loaded in sEVs was Loc108349490.
Collapse
Affiliation(s)
- Zhi‐gang Wang
- Department of Kidney TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hong‐en Xu
- Precision Medicine Center of Zhengzhou UniversityAcademy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Fu‐min Cheng
- Department of Kidney TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jie Zhang
- Department of Kidney TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yong‐hua Feng
- Department of Kidney TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dan‐hua Liu
- Precision Medicine Center of Zhengzhou UniversityAcademy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Wen‐jun Shang
- Department of Kidney TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Gui‐wen Feng
- Department of Kidney TransplantationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
14
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
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: 23] [Impact Index Per Article: 5.8] [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
|
Liu Y, Su YY, Yang Q, Zhou T. Stem cells in the treatment of renal fibrosis: a review of preclinical and clinical studies of renal fibrosis pathogenesis. Stem Cell Res Ther 2021; 12:333. [PMID: 34112221 PMCID: PMC8194041 DOI: 10.1186/s13287-021-02391-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Renal fibrosis commonly leads to glomerulosclerosis and renal interstitial fibrosis and the main pathological basis involves tubular atrophy and the abnormal increase and excessive deposition of extracellular matrix (ECM). Renal fibrosis can progress to chronic kidney disease. Stem cells have multilineage differentiation potential under appropriate conditions and are easy to obtain. At present, there have been some studies showing that stem cells can alleviate the accumulation of ECM and renal fibrosis. However, the sources of stem cells and the types of renal fibrosis or renal fibrosis models used in these studies have differed. In this review, we summarize the pathogenesis (including signaling pathways) of renal fibrosis, and the effect of stem cell therapy on renal fibrosis as described in preclinical and clinical studies. We found that stem cells from various sources have certain effects on improving renal function and alleviating renal fibrosis. However, additional clinical studies should be conducted to confirm this conclusion in the future.
Collapse
Affiliation(s)
- Yiping Liu
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Yan-Yan Su
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Qian Yang
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, No. 69 Dongsha Road, Shantou, 515041, China.
| |
Collapse
|
17
|
Wang LT, Liu KJ, Sytwu HK, Yen ML, Yen BL. Advances in mesenchymal stem cell therapy for immune and inflammatory diseases: Use of cell-free products and human pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Transl Med 2021; 10:1288-1303. [PMID: 34008922 PMCID: PMC8380447 DOI: 10.1002/sctm.21-0021] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell therapy (MSCT) for immune and inflammatory diseases continues to be popular based on progressive accumulation of preclinical mechanistic evidence. This has led to further expansion in clinical indications from graft rejection, autoimmune diseases, and osteoarthritis, to inflammatory liver and pulmonary diseases including COVID‐19. A clear trend is the shift from using autologous to allogeneic MSCs, which can be immediately available as off‐the‐shelf products. In addition, new products such as cell‐free exosomes and human pluripotent stem cell (hPSC)‐derived MSCs are exciting developments to further prevalent use. Increasing numbers of trials have now published results in which safety of MSCT has been largely demonstrated. While reports of therapeutic endpoints are still emerging, efficacy can be seen for specific indications—including graft‐vs‐host‐disease, strongly Th17‐mediated autoimmune diseases, and osteoarthritis—which are more robustly supported by mechanistic preclinical evidence. In this review, we update and discuss outcomes in current MSCT clinical trials for immune and inflammatory disease, as well as new innovation and emerging trends in the field.
Collapse
Affiliation(s)
- Li-Tzu Wang
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, Taipei, Taiwan, Republic of China
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes (NHRI), Tainan, Taiwan, Republic of China
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases & Vaccinology, NHRI, Zhunan, Taiwan, Republic of China.,Department & Graduate Institute of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Men-Luh Yen
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, Taipei, Taiwan, Republic of China
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, NHRI, Zhunan, Taiwan, Republic of China
| |
Collapse
|
18
|
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: 4] [Impact Index Per Article: 1.0] [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
|
19
|
Dreyer GJ, Groeneweg KE, Heidt S, Roelen DL, van Pel M, Roelofs H, Huurman VAL, Bajema IM, Moes DJAR, Fibbe WE, Claas FHJ, van Kooten C, Rabelink RJ, de Fijter JW, Reinders MEJ. Human leukocyte antigen selected allogeneic mesenchymal stromal cell therapy in renal transplantation: The Neptune study, a phase I single-center study. Am J Transplant 2020; 20:2905-2915. [PMID: 32277568 PMCID: PMC7586810 DOI: 10.1111/ajt.15910] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/10/2020] [Accepted: 03/29/2020] [Indexed: 01/25/2023]
Abstract
Mesenchymal stromal cells (MSC) hold promise as a novel immune-modulatory therapy in organ transplantation. First clinical studies have used autologous MSCs; however, the use of allogeneic "off-the-shelf" MSCs is more sustainable for broad clinical implementation, although with the risk of causing sensitization. We investigated safety and feasibility of allogeneic MSCs in renal transplantation, using a matching strategy that prevented repeated mismatches. Ten patients received two doses of 1.5 × 106 /kg allogeneic MSCs 6 months after transplantation in a single-center nonrandomized phase Ib trial, followed by lowering of tacrolimus (trough level 3 ng/mL) in combination with everolimus and prednisone. Primary end point was safety, measured by biopsy proven acute rejection (BPAR) and graft loss 12 months after transplantation. Immune monitoring was performed before and after infusion. No BPAR or graft loss occurred and renal function remained stable. One patient retrospectively had DSAs against MSCs, formed before infusion. No major alterations in T and B cell populations or plasma cytokines were observed upon MSC infusion. Administration of HLA selected allogeneic MSCs combined with low-dose tacrolimus 6 months after transplantation is safe at least in the first year after renal transplantation. This sets the stage to further explore the efficacy of third-party MSCs in renal transplantation.
Collapse
Affiliation(s)
- Geertje J. Dreyer
- 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
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood TransfusionLeiden University Medical CenterLeidenthe Netherlands
| | - Dave L. Roelen
- Department of Immunohematology and Blood TransfusionLeiden University Medical CenterLeidenthe Netherlands
| | - Melissa van Pel
- Department of Immunohematology and Blood TransfusionLeiden University Medical CenterLeidenthe Netherlands
| | - Helene Roelofs
- Department of Immunohematology and Blood TransfusionLeiden University Medical CenterLeidenthe Netherlands
| | - Volkert A. L. Huurman
- Department of Transplant Surgery and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Ingeborg M. Bajema
- Department of PathologyLeiden University Medical CenterLeidenthe Netherlands
| | - Dirk Jan A. R. Moes
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenthe Netherlands
| | - Willem E. Fibbe
- Department of Immunohematology and Blood TransfusionLeiden University Medical CenterLeidenthe Netherlands
| | - Frans H. J. Claas
- Department of Immunohematology and Blood TransfusionLeiden University Medical CenterLeidenthe Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| | - Rabelink 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
| | - Marlies E. J. Reinders
- Department of Internal Medicine (Nephrology) and Transplant CenterLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
20
|
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] [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
|
21
|
Hoogduijn MJ, Montserrat N, van der Laan LJW, Dazzi F, Perico N, Kastrup J, Gilbo N, Ploeg RJ, Roobrouck V, Casiraghi F, Johnson CL, Franquesa M, Dahlke MH, Massey E, Hosgood S, Reinders MEJ. The emergence of regenerative medicine in organ transplantation: 1st European Cell Therapy and Organ Regeneration Section meeting. Transpl Int 2020; 33:833-840. [PMID: 32237237 PMCID: PMC7497223 DOI: 10.1111/tri.13608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/06/2020] [Accepted: 03/20/2020] [Indexed: 12/19/2022]
Abstract
Regenerative medicine is emerging as a novel field in organ transplantation. In September 2019, the European Cell Therapy and Organ Regeneration Section (ECTORS) of the European Society for Organ Transplantation (ESOT) held its first meeting to discuss the state‐of‐the‐art of regenerative medicine in organ transplantation. The present article highlights the key areas of interest and major advances in this multidisciplinary field in organ regeneration and discusses its implications for the future of organ transplantation.
Collapse
Affiliation(s)
- Martin J Hoogduijn
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Francesco Dazzi
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Norberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Jens Kastrup
- Cardiology Stem Cell Center, Rigshospitalet University Hospital Copenhagen, Copenhagen, Denmark
| | - Nicholas Gilbo
- Lab of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.,Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences and Oxford Transplant Centre, University of Oxford and Oxford University Hospitals NHS Trust, Oxford, UK
| | | | | | - Christian L Johnson
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marcella Franquesa
- REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Badalona, Spain
| | - Marc H Dahlke
- Department of Surgery, Robert-Bosch-Health-Campus, Stuttgart, Germany
| | - Emma Massey
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sarah Hosgood
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Marlies E J Reinders
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
22
|
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. Rev Assoc Med Bras (1992) 2020; 66Suppl 1: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] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
23
|
Wilson A, Hodgson-Garms M, Frith JE, Genever P. Multiplicity of Mesenchymal Stromal Cells: Finding the Right Route to Therapy. Front Immunol 2019; 10:1112. [PMID: 31164890 PMCID: PMC6535495 DOI: 10.3389/fimmu.2019.01112] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/01/2019] [Indexed: 12/26/2022] Open
Abstract
Over the last decade, the acceleration in the clinical use of mesenchymal stromal cells (MSCs) has been nothing short of spectacular. Perhaps most surprising is how little we know about the "MSC product." Although MSCs are being delivered to patients at an alarming rate, the regulatory requirements for MSC therapies (for example in terms of quality assurance and quality control) are nowhere near the expectations of traditional pharmaceuticals. That said, the standards that define a chemical compound or purified recombinant protein cannot be applied with the same stringency to a cell-based therapy. Biological processes are dynamic, adaptive and variable. Heterogeneity will always exist or emerge within even the most rigorously sorted clonal cell populations. With MSCs, perhaps more so than any other therapeutic cell, heterogeneity pervades at multiple levels, from the sample source to the single cell. The research and clinical communities collectively need to recognize and take steps to address this troublesome truth, to ensure that the promise of MSC-based therapies is fulfilled.
Collapse
Affiliation(s)
- Alison Wilson
- Department of Biology, University of York, York, United Kingdom
| | | | - Jessica E Frith
- Materials Science and Engineering, Monash University, Clayton, VIC, Australia
| | - Paul Genever
- Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
24
|
Chen D. Dually Efficacious Medicine Against Fibrosis and Cancer. Med Sci (Basel) 2019; 7:medsci7030041. [PMID: 30836705 PMCID: PMC6473536 DOI: 10.3390/medsci7030041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/18/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
Although there is a contemporary consensus of managing a severe disease with multi-targeted approach-based therapeutic combinations, it should not be ignored that certain patho-biological pathways are shared by distinct medical conditions and can be exploited to develop an exceptional type of medication conferring a dual efficacy. This article thus presents a spectrum of emerging molecular targets that substantially contribute to the pathogenesis of both fibrotic and neoplastic disorders, including kinase activities, cytokine cascades, and protein dynamics among others. Moreover, recently approved therapeutic agents in this regard have been sorted out to corroborate the drug’s ability upon targeting each one of these molecular pathways to treat fibrosis and cancer simultaneously. It not only streamlines an overlapping mechanistic profile in the pathogenesis across these two medical conditions, but also inspires clinicians and pharmaceutical innovation to tackle concomitant diseases, such as fibrosis and cancer, with an optimally efficacious medication.
Collapse
Affiliation(s)
- Daohong Chen
- Research Institute of Biological Medicine, Yiling Pharmaceutical; Beijing 102600, China.
| |
Collapse
|
25
|
Zhuang Q, Ma R, Yin Y, Lan T, Yu M, Ming Y. Mesenchymal Stem Cells in Renal Fibrosis: The Flame of Cytotherapy. Stem Cells Int 2019; 2019:8387350. [PMID: 30766607 PMCID: PMC6350586 DOI: 10.1155/2019/8387350] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/27/2018] [Indexed: 12/24/2022] Open
Abstract
Renal fibrosis, as the fundamental pathological process of chronic kidney disease (CKD), is a pathologic extension of the normal wound healing process characterized by endothelium injury, myofibroblast activation, macrophage migration, inflammatory signaling stimulation, matrix deposition, and remodelling. Yet, the current method of treating renal fibrosis is fairly limited, including angiotensin-converting enzyme inhibition, angiotensin receptor blockade, optimal blood pressure control, and sodium bicarbonate for metabolic acidosis. MSCs are pluripotent adult stem cells that can differentiate into various types of tissue lineages, such as the cartilage (chondrocytes), bone (osteoblasts), fat (adipocytes), and muscle (myocytes). Because of their many advantages like ubiquitous sources, convenient procurement and collection, low immunogenicity, and low adverse effects, with their special identification markers, mesenchymal stem MSC-based therapy is getting more and more attention. Based on the mechanism of renal fibrosis, MSCs mostly participate throughout the renal fibrotic process. According to the latest and overall literature reviews, we aim to elucidate the antifibrotic mechanisms and effects of diverse sources of MSCs on renal fibrosis, assess their efficacy and safety in preliminarily clinical application, answer the controversial questions, and provide novel ideas into the MSC cellular therapy of renal fibrosis.
Collapse
Affiliation(s)
- Quan Zhuang
- Transplantation Center of The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, Hunan 410013, China
| | - Ruoyu Ma
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Yanshuang Yin
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Tianhao Lan
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Meng Yu
- Transplantation Center of The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, Hunan 410013, China
| | - Yingzi Ming
- Transplantation Center of The 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Research Center of National Health Ministry on Transplantation Medicine, Changsha, Hunan 410013, China
| |
Collapse
|
26
|
|
27
|
Pan GH, Chen Z, Xu L, Zhu JH, Xiang P, Ma JJ, Peng YW, Li GH, Chen XY, Fang JL, Guo YH, Zhang L, Liu LS. Low-dose tacrolimus combined with donor-derived mesenchymal stem cells after renal transplantation: a prospective, non-randomized study. Oncotarget 2017; 7:12089-101. [PMID: 26933811 PMCID: PMC4914271 DOI: 10.18632/oncotarget.7725] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/29/2016] [Indexed: 12/22/2022] Open
Abstract
Calcineurin inhibitors, including tacrolimus, are largely responsible for advances in allotransplantation. However, the nephrotoxicity associated with these immunosuppressants impairs patients' long-term survival after renal allograft. Therefore, novel regimens that minimize or even eliminate calcineurin inhibitors could improve transplantation outcomes. In this pilot study, we investigated the use of low-dose tacrolimus in combination with mesenchymal stem cells (MSCs), which are immunosuppressive and prolong allograft survival in experimental organ transplant models. Donor-derived, bone marrow MSCs combined with a sparing dose of tacrolimus (0.04-0.05 mg/kg/day) were administered to 16 de novo living-related kidney transplant recipients; 16 other patients received a standard dose of tacrolimus (0.07-0.08 mg/kg/day). The safety of MSC infusion, acute rejection, graft function, graft survival, and patient survival were evaluated over ≥24 months following kidney transplantation. All patients survived and had stable renal function at the 24 month follow-up. The combination of low-dose tacrolimus and MSCs was as effective as standard dose tacrolimus in maintaining graft survival at least 2 years after transplantation. In addition, both groups had similar urea, urine protein, urinary RBC, urinary WBC, 24-h urine protein, and creatinine clearance rates from 7 days to 24 months after transplantation. Furthermore, no differences in the proportion of lymphocytes, CD19, CD3, CD34, CD38, and natural killer cells were detected between the control and experimental groups. None of the MSC recipients experienced immediate or long-term toxicity from the treatment. This preliminary data suggests that the addition of MSCs permits the use of lower dosages of nephrotoxic calcineurin inhibitors following renal transplantation.
Collapse
Affiliation(s)
- Guang-Hui Pan
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zheng Chen
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lu Xu
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jing-Hui Zhu
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, SunYat-sen University, Guangzhou, Guangdong, China
| | - Jun-Jie Ma
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan-Wen Peng
- Center for Stem Cell Biology and Tissue Engineering, SunYat-sen University, Guangzhou, Guangdong, China
| | - Guang-Hui Li
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Yong Chen
- Center for Stem Cell Biology and Tissue Engineering, SunYat-sen University, Guangzhou, Guangdong, China
| | - Jia-Li Fang
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yu-He Guo
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lei Zhang
- The Transplantation Centre, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Long-Shan Liu
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Mesenchymal stromal cells in clinical kidney transplantation: how tolerant can it be? Curr Opin Organ Transplant 2017; 21:550-558. [PMID: 27755168 DOI: 10.1097/mot.0000000000000364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Progress in the improvement of short-term and long-term outcomes of kidney transplantation seems to have reached a plateau, partially due to consequences of very efficient, but nonspecific immunosuppressive drugs. In recent years, various forms of cell therapy, including the use of mesenchymal stromal cells, have been put forward as an alternative strategy for more defined therapy. It is thought that these therapies will not only allow controlled tapering of immunosuppressive medication, but might bring us also closer to the ambition of generating donor-specific immune regulation and tolerance. RECENT FINDINGS Different forms of alloimmunity, including direct, indirect and semi-direct alloantigen presentation have to be controlled before donor-specific immune regulation can be reached. Several mechanisms have been described how mesenchymal stromal cells can affect alloimmunity. Especially, the interaction with professional antigen presenting cells, like dendritic cells, is of critical importance. SUMMARY This review will discuss the current status of ongoing clinical trials with mesenchymal stromal cells in kidney transplantation and specifically concentrate on the possibilities and impossibilities of how these therapeutic strategies can contribute to control of the different forms of alloreactivity operation in organ transplantation.
Collapse
|
29
|
Roushandeh AM, Bahadori M, Roudkenar MH. Mesenchymal Stem Cell-based Therapy as a New Horizon for Kidney Injuries. Arch Med Res 2017. [PMID: 28625316 DOI: 10.1016/j.arcmed.2017.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Today, the prevalence of kidney diseases is increasing around the world, but there has still been no effective medical treatment. The therapeutic choices are confined to supportive cares and preventive strategies. Currently, mesenchymal stem cells (MSCs)-based cell therapy was proposed for the treatment of kidney injuries. However, after the transplantation of MSCs, they are exposed to masses of cytotoxic factors involving an inflammatory cytokine storm, a nutritionally-poor hypoxic environment and oxidative stresses that finally lead to minimize the efficacy of MSCs based cell therapy. Therefore, several innovative strategies were developed in order to potentiate MSCs to withstand the unfavorable microenvironments of the injured kidney tissues and improve their therapeutic potentials. This review aims to introduce MSCs as a new modality in the treatment of renal failure. Here, we discuss the clinical trials of MSCs-based therapy in kidney diseases as well as the in vivo studies dealing with MSCs application in kidney injuries mainly from the proliferation, differentiation, migration and survival points of view. The obstacles and challenges of this new modality in kidney injuries are also discussed.
Collapse
Affiliation(s)
| | - Marzie Bahadori
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehryar Habibi Roudkenar
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences Rasht, Iran.
| |
Collapse
|
30
|
Zhang YC, Liu W, Fu BS, Wang GY, Li HB, Yi HM, Jiang N, Wang G, Zhang J, Yi SH, Li H, Zhang Q, Yang Y, Chen GH. Therapeutic potentials of umbilical cord–derived mesenchymal stromal cells for ischemic-type biliary lesions following liver transplantation. Cytotherapy 2017; 19:194-199. [DOI: 10.1016/j.jcyt.2016.11.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023]
|
31
|
Mesenchymal Stem Cell-Based Therapy for Kidney Disease: A Review of Clinical Evidence. Stem Cells Int 2016; 2016:4798639. [PMID: 27721835 PMCID: PMC5046016 DOI: 10.1155/2016/4798639] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/15/2016] [Accepted: 08/18/2016] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells form a population of self-renewing, multipotent cells that can be isolated from several tissues. Multiple preclinical studies have demonstrated that the administration of exogenous MSC could prevent renal injury and could promote renal recovery through a series of complex mechanisms, in particular via immunomodulation of the immune system and release of paracrine factors and microvesicles. Due to their therapeutic potentials, MSC are being evaluated as a possible player in treatment of human kidney disease, and an increasing number of clinical trials to assess the safety, feasibility, and efficacy of MSC-based therapy in various kidney diseases have been proposed. In the present review, we will summarize the current knowledge on MSC infusion to treat acute kidney injury, chronic kidney disease, diabetic nephropathy, focal segmental glomerulosclerosis, systemic lupus erythematosus, and kidney transplantation. The data obtained from these clinical trials will provide further insight into safety, feasibility, and efficacy of MSC-based therapy in renal pathologies and allow the design of consensus protocol for clinical purpose.
Collapse
|
32
|
Cameron AM, Wesson RN, Ahmadi AR, Singer AL, Hu X, Okabayashi T, Wang Y, Shigoka M, Fu Y, Gao W, Raccusen LC, Montgomery RA, Williams GM, Sun Z. Chimeric Allografts Induced by Short-Term Treatment With Stem Cell Mobilizing Agents Result in Long-Term Kidney Transplant Survival Without Immunosuppression: II, Study in Miniature Swine. Am J Transplant 2016; 16:2066-76. [PMID: 26748958 DOI: 10.1111/ajt.13703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/21/2015] [Accepted: 12/27/2015] [Indexed: 01/25/2023]
Abstract
Transplantation is now lifesaving therapy for patients with end-stage organ failure but requires lifelong immunosuppression with resultant morbidity. Current immunosuppressive strategies inhibit T cell activation and prevent donor-recipient engagement. Therefore, it is not surprising that few host cells are demonstrated in donor grafts. However, our recent small animal studies found large numbers of recipient stem cells present after transplantation and pharmacological mobilization, resulting in a chimeric, repopulated organ. We now confirm these findings in a well-characterized large animal preclinical model. Here, we show that AMD3100 and FK506 mobilization of endogenous stem cells immediately post kidney transplantation combined with repeat therapy at 1, 2, and 3 months led to drug-free long-term survival in maximally immunologically mismatched swine. Three long-term recipients have stable chimeric transplants, preserved antidonor skin graft responses, and normal serum creatinine levels despite withdrawal of all medication for 3 years.
Collapse
Affiliation(s)
- A M Cameron
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - R N Wesson
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - A R Ahmadi
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - A L Singer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD.,Transplant Center, Mayo Clinic, Phoenix, AZ, USA
| | - X Hu
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - T Okabayashi
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Surgery, Kochi Health Center, Kochi University, Kochi, Japan
| | - Y Wang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - M Shigoka
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Y Fu
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD.,Transplant Center, Tianjin First Central Hospital, Tianjin, China
| | - W Gao
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD.,Transplant Center, Tianjin First Central Hospital, Tianjin, China
| | - L C Raccusen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - R A Montgomery
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - G M Williams
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Z Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
33
|
Abstract
Clinical investigations using stem cell products in regenerative medicine are addressing a wide spectrum of conditions using a variety of stem cell types. To date, there have been few reports of safety issues arising from autologous or allogeneic transplants. Many cells administered show transient presence for a few days with trophic influences on immune or inflammatory responses. Limbal stem cells have been registered as a product for eye burns in Europe and mesenchymal stem cells have been approved for pediatric graft versus host disease in Canada and New Zealand. Many other applications are progressing in trials, some with early benefits to patients.
Collapse
Affiliation(s)
- Alan Trounson
- Hudson Institute for Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia.
| | - Courtney McDonald
- Hudson Institute for Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|
34
|
Premise and promise of mesenchymal stem cell-based therapies in clinical vascularized composite allotransplantation. Curr Opin Organ Transplant 2016; 20:608-14. [PMID: 26536421 DOI: 10.1097/mot.0000000000000247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Over the past decade, clinical vascularized composite allotransplantation (VCA) has enabled functional and quality of life restoration in a wide range of indications secondary to devastating tissue loss. However, the spectre of toxicity and long-term complications of chronic immunosuppression has curtailed the momentum of VCA. This study summarizes the literature evidence behind successful mesenchymal stem cell (MSC)-based cell therapies highlighting their multipronged immunomodulatory, restorative and regenerative characteristics with special emphasis towards VCA applications. RECENT FINDINGS Experimental and clinical studies in solid organs and VCA have confirmed that MSCs facilitate immunosuppression-free allograft survival or tolerance, stimulate peripheral nerve regeneration, attenuate ischaemia-reperfusion injury, and improve tissue healing after surgery. It has been hypothesized that MSC-induced long-term operational tolerance in experimental VCA is mediated by induction of mixed donor-specific chimerism and regulatory T-cell mechanisms. All these characteristics of MSCs could thus help expand the scope and clinical feasibility of VCA. SUMMARY Cellular therapies, especially those focusing on MSCs, are emerging in solid organ transplantation including VCA. Although some clinical trials have begun to assess the effects of MSCs in solid organ transplantation, much scientific domain remains uncharted, especially for VCA.
Collapse
|
35
|
Reinders MEJ, Dreyer GJ, Bank JR, Roelofs H, Heidt S, Roelen DL, Zandvliet ML, Huurman VAL, Fibbe WE, van Kooten C, Claas FHJ, Rabelink TJ, de Fijter JW. Safety of allogeneic bone marrow derived mesenchymal stromal cell therapy in renal transplant recipients: the neptune study. J Transl Med 2015; 13:344. [PMID: 26537851 PMCID: PMC4632480 DOI: 10.1186/s12967-015-0700-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/16/2015] [Indexed: 01/05/2023] Open
Abstract
Background Mesenchymal stromal cells (MSC) may serve as an attractive therapy in renal transplantation due to their immunosuppressive and reparative properties. While most studies have used autologous MSCs, allogeneic MSCs offer the advantage of immediate availability for clinical use. This is of major importance for indications where instant treatment is needed, for example allograft rejection or calcineurin inhibitor toxicity. Clinical studies using allogeneic MSCs are limited in number. Although these studies showed no adverse reactions, allogeneic MSCs could possibly elicit an anti-donor immune response, which may increase the incidence of rejection and impact the allograft survival in the long term. These safety issues should be addressed before further studies are planned with allogeneic MSCs in the solid organ transplant setting. Methods/design 10 renal allograft recipients, 18–75 years old, will be included in this clinical phase Ib, open label, single center study. Patients will receive two doses of 1.5 × 106 per/kg body weight allogeneic bone marrow derived MSCs intravenously, at 25 and 26 weeks after transplantation, when immune suppression levels are reduced. The primary end point of this study is safety by assessing biopsy proven acute rejection (BPAR)/graft loss after MSC treatment. Secondary end points, all measured before and after MSC infusions, include: comparison of fibrosis in renal biopsy by quantitative Sirius Red scoring; de novo HLA antibody development and extensive immune monitoring; renal function measured by cGFR and iohexol clearance; CMV and BK infection and other opportunistic infections. Discussion This study will provide information on the safety of allogeneic MSC infusion and its effect on the incidence of BPAR/graft loss. Trial registration: NCT02387151
Collapse
Affiliation(s)
- Marlies E J Reinders
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Geertje J Dreyer
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Jonna R Bank
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Helene Roelofs
- Department of Immuno-Haematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Sebastiaan Heidt
- Department of Immuno-Haematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Dave L Roelen
- Department of Immuno-Haematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Maarten L Zandvliet
- Department of Clinical Parmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Volkert A L Huurman
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Wim E Fibbe
- Department of Immuno-Haematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Cees van Kooten
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Frans H J Claas
- Department of Immuno-Haematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Ton J Rabelink
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands. .,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Johan W de Fijter
- Department of Nephrology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
36
|
Safety and Efficacy Endpoints for Mesenchymal Stromal Cell Therapy in Renal Transplant Recipients. J Immunol Res 2015; 2015:391797. [PMID: 26258149 PMCID: PMC4518147 DOI: 10.1155/2015/391797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/26/2015] [Indexed: 02/07/2023] Open
Abstract
Despite excellent short-term graft survival after renal transplantation, the long-term graft outcome remains compromised. It has become evident that a combination of sustained alloreactivity and calcineurin-inhibitor- (CNI-) related nephrotoxicity results in fibrosis and consequently dysfunction of the graft. New immunosuppressive regimens that can minimize or eliminate side effects, while maintaining efficacy, are required to improve long-term graft survival. In this perspective mesenchymal stromal cells (MSCs) are an interesting candidate, since MSCs have immunosuppressive and regenerative properties. The first clinical trials with MSCs in renal transplantation showed safety and feasibility and displayed promising results. Recently, the first phase II studies have been started. One of the most difficult and challenging aspects in those early phase trials is to define accurate endpoints that can measure safety and efficacy of MSC treatment. Since both graft losses and acute rejection rates declined, alternative surrogate markers such as renal function, histological findings, and immunological markers are used to measure efficacy and to provide mechanistic insight. In this review, we will discuss the current status of MSCs in renal transplantation with a focus on the endpoints used in the different experimental and clinical studies.
Collapse
|