1
|
Rivera Orsini MA, Ozmen EB, Miles A, Newby SD, Springer N, Millis D, Dhar M. Isolation and Characterization of Canine Adipose-Derived Mesenchymal Stromal Cells: Considerations in Translation from Laboratory to Clinic. Animals (Basel) 2024; 14:2974. [PMID: 39457904 PMCID: PMC11503832 DOI: 10.3390/ani14202974] [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: 09/06/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
In allogeneic MSC implantation, the cells are isolated from a donor different from the recipient. When tested, allogeneic MSCs have several advantages over autologous ones: faster cell growth, sufficient cell concentration, and readily available cells for clinics. To ensure the safe and efficient use of allogeneic MSCs in clinics, the MSCs need to be first tested in vitro. With this study, we paved the way by addressing the in vitro aspects of canine adipose-derived MSCs, considering the limited studies on the clinical use of canine cells. We isolated cAD-MSCs from canine falciform ligament fat and evaluated their viability and proliferation using an MTS assay. Then, we characterized the MSC-specific antigens using immunophenotyping and immunofluorescence and demonstrated their potential for in vitro differentiation. Moreover, we established shipping and cryobanking procedures to lead the study to become an off-the-shelf therapy. During expansion, the cells demonstrated a linear increase in cell numbers, confirming their proliferation quantitatively. The cells showed viability before and after cryopreservation, demonstrating that cell viability can be preserved. From a clinical perspective, the established shipping conditions demonstrated that the cells retain their viability for up to 48 h. This study lays the groundwork for the potential use of allogeneic cAD-MSCs in clinical applications.
Collapse
Affiliation(s)
- Michael A. Rivera Orsini
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
| | - Emine Berfu Ozmen
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
- Genome Science and Technology, University of Tennessee Knoxville, Knoxville, TN 37996, USA
| | - Alyssa Miles
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (A.M.); (N.S.)
| | - Steven D. Newby
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
| | - Nora Springer
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (A.M.); (N.S.)
| | - Darryl Millis
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA;
| | - Madhu Dhar
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
| |
Collapse
|
2
|
Kumar N, Bidkhori HR, Yawno T, Lim R, Inocencio IM. Therapeutic potential of extracellular vesicles derived from human amniotic epithelial cells for perinatal cerebral and pulmonary injury. Stem Cells Transl Med 2024; 13:711-723. [PMID: 38895873 PMCID: PMC11328935 DOI: 10.1093/stcltm/szae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/21/2024] Open
Abstract
Lung and brain injury that occurs during the perinatal period leads to lifelong disability and is often driven and/or exacerbated by inflammation. Human amniotic epithelial cells (hAEC), which demonstrate immunomodulatory, anti-fibrotic, and regenerative capabilities, are being explored as a therapeutic candidate for perinatal injury. However, limitations regarding scalable manufacturing, storage, transport, and dose-related toxicity have impeded clinical translation. Isolated therapeutic extracellular vesicles (EVs) from stem and stem-like cells are thought to be key paracrine mediators of therapeutic efficacy. The unique characteristics of EVs suggest that they potentially circumvent the limitations of traditional cell-based therapies. However, given the novelty of EVs as a therapeutic, recommendations around ideal methods of production, isolation, storage, and delivery have not yet been created by regulatory agencies. In this concise review, we discuss the pertinence and limitations of cell-based therapeutics in perinatal medicine. We also review the preclinical evidence supporting the use of therapeutic EVs for perinatal therapy. Further, we summarize the arising considerations regarding adequate cell source, biodistribution, isolation and storage methods, and regulatory roadblocks for the development of therapeutic EVs.
Collapse
Affiliation(s)
- Naveen Kumar
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Hamid Reza Bidkhori
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton 3168, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Ishmael Miguel Inocencio
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| |
Collapse
|
3
|
Yudhawati R, Shimizu K. PGE2 Produced by Exogenous MSCs Promotes Immunoregulation in ARDS Induced by Highly Pathogenic Influenza A through Activation of the Wnt-β-Catenin Signaling Pathway. Int J Mol Sci 2023; 24:ijms24087299. [PMID: 37108459 PMCID: PMC10138595 DOI: 10.3390/ijms24087299] [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: 02/06/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Acute respiratory distress syndrome is an acute respiratory failure caused by cytokine storms; highly pathogenic influenza A virus infection can induce cytokine storms. The innate immune response is vital in this cytokine storm, acting by activating the transcription factor NF-κB. Tissue injury releases a danger-associated molecular pattern that provides positive feedback for NF-κB activation. Exogenous mesenchymal stem cells can also modulate immune responses by producing potent immunosuppressive substances, such as prostaglandin E2. Prostaglandin E2 is a critical mediator that regulates various physiological and pathological processes through autocrine or paracrine mechanisms. Activation of prostaglandin E2 results in the accumulation of unphosphorylated β-catenin in the cytoplasm, which subsequently reaches the nucleus to inhibit the transcription factor NF-κB. The inhibition of NF-κB by β-catenin is a mechanism that reduces inflammation.
Collapse
Affiliation(s)
- Resti Yudhawati
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga-Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
- Indonesia-Japan Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60286, Indonesia
| | - Kazufumi Shimizu
- Indonesia-Japan Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya 60286, Indonesia
- Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
4
|
The Role of Stem Cells Derived From the Mesenchyme of the Umbilical Cord in Reducing Immunosuppressive Drug Doses Used in Allogenic Transplantations. Ann Plast Surg 2022; 89:684-693. [DOI: 10.1097/sap.0000000000003314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
5
|
Clinical and preclinical tolerance protocols for vascularized composite allograft transplantation. Arch Plast Surg 2021; 48:703-713. [PMID: 34818720 PMCID: PMC8627932 DOI: 10.5999/aps.2021.00927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/06/2021] [Indexed: 12/01/2022] Open
Abstract
The field of vascularized composite allografts (VCAs) has undergone significant advancement in recent decades, and VCAs are increasingly common and accepted in the clinical setting, bringing hope of functional recovery to patients with debilitating injuries. A major obstacle facing the widespread application of VCAs is the side effect profile associated with the current immunosuppressive regimen, which can cause a wide array of complications such as infection, malignancy, and even death. Significant concerns remain regarding whether the treatment outweighs the risk. The potential solution to this dilemma would be achieving VCA tolerance, which would allow recipients to receive allografts without significant immunosuppression and its sequelae. Promising tolerance protocols are being studied in kidney transplantation; four major trials have attempted to withdraw immunosuppressive treatment with various successes. The common theme in all four trials is the use of radiation treatment and donor cell transplantation. The knowledge gained from these trials can provide valuable insight into the development of a VCA tolerance protocol. Despite similarities, VCAs present additional barriers compared to kidney allografts regarding tolerance induction. VCA donors are likely to be deceased, which limits the time for significant pre-conditioning. VCA donors are also more likely to be human leukocyte antigen–mismatched, which means that tolerance must be induced across major immunological barriers. This review also explores adjunct therapies studied in large animal models that could be the missing element in establishing a safe and stable tolerance induction method.
Collapse
|
6
|
Kolanko E, Grajoszek A, Czekaj P. Immunosuppressive Potential of Activated Human Amniotic Cells in an Experimental Murine Model of Skin Allo- and Xenotransplantation. Front Med (Lausanne) 2021; 8:715590. [PMID: 34631739 PMCID: PMC8494785 DOI: 10.3389/fmed.2021.715590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/26/2021] [Indexed: 11/25/2022] Open
Abstract
Isolated human amniotic cells (hAC) could be used as a source of immunomodulatory factors in regenerative medicine and transplantation. However, in previous experimental studies, native hAC administered to skin graft recipients did not induce graft immunotolerance. To strengthen the immunomodulatory properties of hAC prior to administration to the recipient, we activated them ex vivo using pro-inflammatory cytokines. In this study, we compared the transplantation efficiency of skin allografts (mouse to mouse) and xnografts (rat to mouse) in recipient mice divided into three main groups receiving: 1. Placebo (control group); 2. Cyclosporine A (CsA) [10 or 50 mg/kg body weight (bw)]; 3. suspension of hAC activated ex vivo by IL-1β and INFγ, administered into a tail vein or subcutaneously. During 15 days of observation, hAC administered intravenously or subcutaneously after allotransplantation appeared to be as safe and efficient as CsA at the dose of 10 mg/kg bw in preventing rejection of skin allo- and xenografts. After xenotransplantation, however, only hAC administered intravenously prevented rejection to an extent comparable to CsA. Both CsA (10 mg/kg bw) and activated hAC reduced inflammatory infiltration in the skin (after intravenous injection) and did not increase the concentration of the inflammation marker SAP in serum or percentage of leukocytes in blood. Finally, we concluded that administration of activated hAC is safe and efficient in the presented animal model of skin allo- and xenotransplantation in a route-dependent manner. Activated hAC injected intravenously exhibit an immunosuppressive effect comparable to CsA administered at the dose of 10 mg/kg bw in both allo- and xenotransplantation.
Collapse
Affiliation(s)
- Emanuel Kolanko
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Aniela Grajoszek
- Department of Experimental Medicine, Medical University of Silesia in Katowice, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
7
|
Hu JL, Kim BJ, Yu NH, Kwon ST. Impact of Injection Frequency of Adipose-Derived Stem Cells on Allogeneic Skin Graft Survival Outcomes in Mice. Cell Transplant 2021; 30:9636897211041966. [PMID: 34538121 PMCID: PMC8743972 DOI: 10.1177/09636897211041966] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previous studies indicated that mesenchymal stem cells (MSCs) exhibit immunomodulatory properties in composite tissue allotransplantation. However, due to the high immunogenicity of skin, although the single administration of MSCs improves survival of the skin allotransplant, immune rejection is still inevitable. The aim of our study was to evaluate whether multiple administrations of MSCs would improve immune tolerance in the allogeneic skin graft, compared to that with a single administration in a mouse model. After full-thickness skin allotransplantation on the backs of the mice, the recipient mice were infused with phosphate-buffered saline and isogenic 1.5 × 105/mL adipose-derived stem cells (ADSCs). ADSCs were transplanted into different mice according to the different injection frequencies such as single, once a week, and twice a week. Skin sections were taken on days 7 and 21 post-transplantation in all groups for gene expression and histological studies. ADSCs increased skin allograft survival compared to that in control mice (P < 0.05). Interleukin-6 and tumor necrosis factor-alpha messenger RNA levels were decreased, and the abundance of lymphocytes, based on immunohistochemistry, was also decreased in ADSC-infused mice (P < 0.05). However, among the different ADSC injection frequency groups, multiple ADSC infusion did not improve the survival rate and decreased proinflammatory cytokines and lymphocytes, compared to those with the single administration of ADSCs (P > 0.05). Conversely, the results with single administration were slightly better than those with multiple administrations. Our study demonstrated that ADSCs have the potential for immunomodulation in vivo. However, the results with multiple ADSC administration were not as good as those with single administration, which indicates the complexity of ADSCs in vivo and implying the need for adequate preclinical experimentation.
Collapse
Affiliation(s)
- Ju Long Hu
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Byung Jun Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Na Hee Yu
- Biomedical Research Institute, Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Tack Kwon
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
|
9
|
Torres Crigna A, Uhlig S, Elvers-Hornung S, Klüter H, Bieback K. Human Adipose Tissue-Derived Stromal Cells Suppress Human, but Not Murine Lymphocyte Proliferation, via Indoleamine 2,3-Dioxygenase Activity. Cells 2020; 9:E2419. [PMID: 33167329 PMCID: PMC7694333 DOI: 10.3390/cells9112419] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Over recent years, mesenchymal stromal cells (MSC) have gained immense attraction in immunotherapy, regenerative medicine and tissue engineering. MSC microenvironment modulation occurs through synergy of direct cell-cell contact, and secreted soluble factors and extracellular vesicles (EV). MSC-derived EV have been suggested as cell-free immunomodulatory alternative to MSC; however, previous findings have challenged this. Furthermore, recent data suggest that evaluating the mechanism of action of human MSC (hMSC) in animal models might promote adverse immune reactions or lack of functionality due to xeno-incompatibilities. In this study, we first assessed the immunomodulatory strength of different human MSC sources on in vitro stimulated T cells and compared this to interferon-gamma (IFNγ) primed MSC conditioned medium (CM) and EV. Second, we addressed the main molecular mechanisms, and third, we assessed the MSC in vitro immunosuppressive effect across interspecies barriers. We identified human adipose tissue-derived stromal cells (ASC) with strongest immunomodulatory strength, followed by bone marrow (BM) and cord blood-derived MSC (CB). Whilst CM from primed ASC managed to exert analogous effects as their cellular counterpart, EV derived thereof did not, reproducing previous findings. IFNγ-induced indoleamine 2,3-dioxygenase (IDO) activity was identified as key mechanism to suppress human lymphocyte proliferation, as in the presence of the IDO inhibitor epacadostat (Epac) a stimulation of proliferation was seen. In addition, we revealed MSC immunosuppressive effects to be species-specific, because human cells failed to suppress murine lymphocyte proliferation. In summary, ASC were the strongest immunomodulators with the IDO-kynurenine pathway being key within the human system. Importantly, the in vitro lack of interspecies immunomodulatory strength suggests that preclinical data need to be carefully interpreted especially when considering a possible translation to clinical field.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (A.T.C.); (S.E.-H.); (H.K.)
| | - Stefanie Uhlig
- FlowCore Mannheim Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Susanne Elvers-Hornung
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (A.T.C.); (S.E.-H.); (H.K.)
| | - Harald Klüter
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (A.T.C.); (S.E.-H.); (H.K.)
- Medical Faculty Mannheim, Mannheim Institute for Innate Immunoscience, Heidelberg University, 68167 Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (A.T.C.); (S.E.-H.); (H.K.)
- FlowCore Mannheim Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
- Medical Faculty Mannheim, Mannheim Institute for Innate Immunoscience, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
10
|
Cyclosporine A promotes the therapeutic effect of mesenchymal stem cells on transplantation reaction. Clin Sci (Lond) 2020; 133:2143-2157. [PMID: 31654074 DOI: 10.1042/cs20190294] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022]
Abstract
The successful application of mesenchymal stem cells (MSCs) remains a major challenge in stem cell therapy. Currently, several in vitro studies have indicated potentially beneficial interactions of MSCs with immunosuppressive drugs. These interactions can be even more complex in vivo, and it is in this setting that we investigate the effect of MSCs in combination with Cyclosporine A (CsA) on transplantation reaction and allogeneic cell survival. Using an in vivo mouse model, we found that CsA significantly promoted the survival of MSCs in various organs and tissues of the recipients. In addition, compared to treatment with CsA or MSCs alone, the survival of transplanted allogeneic cells was significantly improved after the combined application of MSCs with CsA. We further observed that the combinatory treatment suppressed immune response to the alloantigen challenge and modulated the immune balance by harnessing proinflammatory CD4+T-bet+ and CD4+RORγt+ cell subsets. These changes were accompanied by a significant decrease in IL-17 production along with an elevated level of IL-10. Co-cultivation of purified naive CD4+ cells with peritoneal macrophages isolated from mice treated with MSCs and CsA revealed that MSC-educated macrophages play an important role in the immunomodulatory effect observed on distinct T-cell subpopulations. Taken together, our findings suggest that CsA promotes MSC survival in vivo and that the therapeutic efficacy of the combination of MSCs with CsA is superior to each monotherapy. This combinatory treatment thus represents a promising approach to reducing immunosuppressant dosage while maintaining or even improving the outcome of therapy.
Collapse
|
11
|
Mesenchymal Stem Cells: A Trump Card for the Treatment of Diabetes? Biomedicines 2020; 8:biomedicines8050112. [PMID: 32384630 PMCID: PMC7277294 DOI: 10.3390/biomedicines8050112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
The advent of the new revolutionary approach based on regenerative medicine is progressively reshaping the therapeutic scenario of many different diseases, such as cardiovascular diseases and immune diseases, with encouraging results. During the last 10 years, many studies have also proposed the use of mesenchymal stem cells (MSCs), adult stem cells with several interesting properties already used in different experimental models, for the treatment of diabetes, however, reporting conflicting outcomes. These reasons have given rise to a question: are these cells a real trump card for the biomedical field? Are they really able to outclass the traditional therapies, or at least able to give an advantage over them? In this review, we will discuss the most promising results obtained with MSCs for the treatment of diabetes and its complications, we will compare the different therapeutic treatments applied as well as the most likely mechanisms of action, and overall we will give an in-depth overview of the pros and the cons of the use of MSCs for the therapy of both type-1 and type-2 diabetes.
Collapse
|
12
|
Reparative and Regenerative Effects of Mesenchymal Stromal Cells-Promising Potential for Kidney Transplantation? Int J Mol Sci 2019; 20:ijms20184614. [PMID: 31540361 PMCID: PMC6770554 DOI: 10.3390/ijms20184614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) possess reparative, regenerative and immunomodulatory properties. The current literature suggests that MSCs could improve kidney transplant outcome via immunomodulation. In many clinical domains, research has also focussed on the regenerative and reparative effects of therapies with MSCs. However, in the field of transplantation, data on this subject remain scarce. This review provides an overview of what is known about the regenerative and reparative effects of MSCs in various fields ranging from wound care to fracture healing and also examines the potential of these promising MSC properties to improve the outcome of kidney transplantations.
Collapse
|
13
|
Burgess JK, Heijink IH. The Safety and Efficiency of Addressing ARDS Using Stem Cell Therapies in Clinical Trials. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7121814 DOI: 10.1007/978-3-030-29403-8_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Janette K. Burgess
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Irene H. Heijink
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| |
Collapse
|
14
|
Soares MA, Massie JP, Rifkin WJ, Rao N, Duckworth AM, Park C, Kadle RL, David JA, Rabbani PS, Ceradini DJ. Ex vivo allotransplantation engineering: Delivery of mesenchymal stem cells prolongs rejection-free allograft survival. Am J Transplant 2018; 18:1657-1667. [PMID: 29359512 DOI: 10.1111/ajt.14668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/14/2018] [Accepted: 01/15/2018] [Indexed: 01/25/2023]
Abstract
Current pharmacologic regimens in transplantation prevent allograft rejection through systemic recipient immunosuppression but are associated with severe morbidity and mortality. The ultimate goal of transplantation is the prevention of allograft rejection while maintaining recipient immunocompetence. We hypothesized that allografts could be engineered ex vivo (after allotransplant procurement but before transplantation) by using mesenchymal stem cell-based therapy to generate localized immunomodulation without affecting systemic recipient immunocompetence. To this end, we evaluated the therapeutic efficacy of bone marrow-derived mesenchymal stem cells in vitro and activated them toward an immunomodulatory fate by priming in inflammatory or hypoxic microenvironments. Using an established rat hindlimb model for allotransplantation, we were able to significantly prolong rejection-free allograft survival with a single perioperative ex vivo infusion of bone marrow-derived mesenchymal stem cells through the allograft vasculature, in the absence of long-term pharmacologic immunosuppression. Critically, transplanted rats rejected a second, nonengineered skin graft from the same donor species to the contralateral limb at a later date, demonstrating that recipient systemic immunocompetence remained intact. This study represents a novel approach in transplant immunology and highlights the significant therapeutic opportunity of the ex vivo period in transplant engineering.
Collapse
Affiliation(s)
- Marc A Soares
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Jonathan P Massie
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - William J Rifkin
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Nakul Rao
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - April M Duckworth
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Chin Park
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Rohini L Kadle
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Joshua A David
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Piul S Rabbani
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| | - Daniel J Ceradini
- NYU Langone Health, Hansjörg Wyss Department of Plastic Surgery, New York, NY, USA
| |
Collapse
|
15
|
Mak WC, Magne B, Cheung KY, Atanasova D, Griffith M. Thermo-rheological responsive microcapsules for time-dependent controlled release of human mesenchymal stromal cells. Biomater Sci 2018; 5:2241-2250. [PMID: 28972602 DOI: 10.1039/c7bm00663b] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human mesenchymal stromal cells (hMSCs) are adult-source cells that have been extensively evaluated for cell-based therapies. hMSCs delivered by intravascular injection have been reported to accumulate at the sites of injury to promote tissue repair and can also be employed as vectors for the delivery of therapeutic genes. However, the full potential of hMSCs remains limited as the cells are lost after injection due to anoikis and the adverse pathologic environment. Encapsulation of cells has been proposed as a means of increasing cell viability. However, controlling the release of therapeutic cells over time to target tissue still remains a challenge today. Here, we report the design and development of thermo-rheological responsive hydrogels that allow for precise, time dependent controlled-release of hMSCs. The encapsulated hMSCs retained good viability from 76% to 87% dependent upon the hydrogel compositions. We demonstrated the design of different blended hydrogel composites with modulated strength (S parameter) and looseness of hydrogel networks (N parameter) to control the release of hMSCs from thermo-responsive hydrogel capsules. We further showed the feasibility for controlled-release of encapsulated hMSCs within 3D matrix scaffolds. We reported for the first time by a systematic analysis that there is a direct correlation between the thermo-rheological properties associated with the degradation of the hydrogel composite and the cell release kinetics. This work therefore provides new insights into the further development of smart carrier systems for stem cell therapy.
Collapse
Affiliation(s)
- W C Mak
- Department of Clinical and Experimental Medicine, Linköping University, SE58185, Linköping, Sweden.
| | | | | | | | | |
Collapse
|
16
|
Inhibition of Lymphatic Drainage With a Self-Designed Surgical Approach Prolongs the Vascularized Skin Allograft Survival in Rats. Ann Plast Surg 2018; 80:76-82. [DOI: 10.1097/sap.0000000000001210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
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: 8.1] [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
|
18
|
Dixit S, Baganizi DR, Sahu R, Dosunmu E, Chaudhari A, Vig K, Pillai SR, Singh SR, Dennis VA. Immunological challenges associated with artificial skin grafts: available solutions and stem cells in future design of synthetic skin. J Biol Eng 2017; 11:49. [PMID: 29255480 PMCID: PMC5729423 DOI: 10.1186/s13036-017-0089-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 11/17/2017] [Indexed: 12/29/2022] Open
Abstract
The repair or replacement of damaged skins is still an important, challenging public health problem. Immune acceptance and long-term survival of skin grafts represent the major problem to overcome in grafting given that in most situations autografts cannot be used. The emergence of artificial skin substitutes provides alternative treatment with the capacity to reduce the dependency on the increasing demand of cadaver skin grafts. Over the years, considerable research efforts have focused on strategies for skin repair or permanent skin graft transplantations. Available skin substitutes include pre- or post-transplantation treatments of donor cells, stem cell-based therapies, and skin equivalents composed of bio-engineered acellular or cellular skin substitutes. However, skin substitutes are still prone to immunological rejection, and as such, there is currently no skin substitute available to overcome this phenomenon. This review focuses on the mechanisms of skin rejection and tolerance induction and outlines in detail current available strategies and alternatives that may allow achieving full-thickness skin replacement and repair.
Collapse
Affiliation(s)
- Saurabh Dixit
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA.,Immunity, Inflammation, and Disease Laboratory, NIH/NIEHS, Durham, 27709 NC USA
| | - Dieudonné R Baganizi
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Rajnish Sahu
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Ejowke Dosunmu
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Atul Chaudhari
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Komal Vig
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Shreekumar R Pillai
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Shree R Singh
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Vida A Dennis
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| |
Collapse
|
19
|
Allogeneic Umbilical Cord-Derived Mesenchymal Stem Cells as a Potential Source for Cartilage and Bone Regeneration: An In Vitro Study. Stem Cells Int 2017; 2017:1732094. [PMID: 29358953 PMCID: PMC5735324 DOI: 10.1155/2017/1732094] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/02/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord (UC) may represent an attractive cell source for allogeneic mesenchymal stem cell (MSC) therapy. The aim of this in vitro study is to investigate the chondrogenic and osteogenic potential of UC-MSCs grown onto tridimensional scaffolds, to identify a possible clinical relevance for an allogeneic use in cartilage and bone reconstructive surgery. Chondrogenic differentiation on scaffolds was confirmed at 4 weeks by the expression of sox-9 and type II collagen; low oxygen tension improved the expression of these chondrogenic markers. A similar trend was observed in pellet culture in terms of matrix (proteoglycan) production. Osteogenic differentiation on bone-graft-substitute was also confirmed after 30 days of culture by the expression of osteocalcin and RunX-2. Cells grown in the hypertrophic medium showed at 5 weeks safranin o-positive stain and an increased CbFa1 expression, confirming the ability of these cells to undergo hypertrophy. These results suggest that the UC-MSCs isolated from minced umbilical cords may represent a valuable allogeneic cell population, which might have a potential for orthopaedic tissue engineering such as the on-demand cell delivery using chondrogenic, osteogenic, and endochondral scaffold. This study may have a clinical relevance as a future hypothetical option for allogeneic single-stage cartilage repair and bone regeneration.
Collapse
|
20
|
Moreira A, Kahlenberg S, Hornsby P. Therapeutic potential of mesenchymal stem cells for diabetes. J Mol Endocrinol 2017; 59:R109-R120. [PMID: 28739632 PMCID: PMC5570611 DOI: 10.1530/jme-17-0117] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are self-renewing multipotent cells that have the capacity to secrete multiple biologic factors that can restore and repair injured tissues. Preclinical and clinical evidence have substantiated the therapeutic benefit of MSCs in various medical conditions. Currently, MSCs are the most commonly used cell-based therapy in clinical trials because of their regenerative effects, ease of isolation and low immunogenicity. Experimental and clinical studies have provided promising results using MSCs to treat diabetes. This review will summarize the role of MSCs on tissue repair, provide emerging strategies to improve MSC function and describe how these processes translate to clinical treatments for diabetes.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of PediatricsUniversity of Texas Health Science Center-San Antonio, San Antonio, Texas, USA
| | - Samuel Kahlenberg
- Department of PediatricsUniversity of Texas Health Science Center-San Antonio, San Antonio, Texas, USA
| | - Peter Hornsby
- Department of PhysiologyTexas Research Park Campus, Barshop Institute for Longevity and Aging Studies, San Antonio, Texas, USA
| |
Collapse
|
21
|
Hong JW, Lim JH, Chung CJ, Kang TJ, Kim TY, Kim YS, Roh TS, Lew DH. Immune Tolerance of Human Dental Pulp-Derived Mesenchymal Stem Cells Mediated by CD4⁺CD25⁺FoxP3⁺ Regulatory T-Cells and Induced by TGF-β1 and IL-10. Yonsei Med J 2017; 58:1031-1039. [PMID: 28792150 PMCID: PMC5552631 DOI: 10.3349/ymj.2017.58.5.1031] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/20/2017] [Accepted: 06/27/2017] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Most studies on immune tolerance of mesenchymal stem cells (MSCs) have been performed using MSCs derived from bone marrow, cord blood, or adipose tissue. MSCs also exist in the craniofacial area, specifically in teeth. The purpose of this study was to evaluate the mechanisms of immune tolerance of dental pulp-derived MSC (DP-MSC) in vitro and in vivo. MATERIALS AND METHODS We isolated DP-MSCs from human dental pulp and co-cultured them with CD4⁺ T-cells. To evaluate the role of cytokines, we blocked TGF-β and IL-10, separately and together, in co-cultured DP-MSCs and CD4⁺ T-cells. We analyzed CD25 and FoxP3 to identify regulatory T-cells (Tregs) by fluorescence-activated cell sorting (FACS) and real-time PCR. We performed alloskin grafts with and without DP-MSC injection in mice. We performed mixed lymphocyte reactions (MLRs) to check immune tolerance. RESULTS Co-culture of CD4⁺ T-cells with DP-MSCs increased the number of CD4⁺CD25⁺FoxP3⁺ Tregs (p<0.01). TGF-β or/and IL-10 blocking suppressed Treg induction in co-cultured cells (p<0.05). TGF-β1 mRNA levels were higher in co-cultured DP-MSCs and in co-cultured CD4⁺ T-cells than in the respective monocultured cells. However, IL-10 mRNA levels were not different. There was no difference in alloskin graft survival rate and area between the DP-MSC injection group and the non-injection group. Nonetheless, MLR was reduced in the DP-MSC injected group (p<0.05). CONCLUSION DP-MSCs can modulate immune tolerance by increasing CD4⁺CD25⁺FoxP3⁺ Tregs. TGF-β1 and IL-10 are factors in the immune-tolerance mechanism. Pure DP-MSC therapy may not be an effective treatment for rejection, although it may module immune tolerance in vivo.
Collapse
Affiliation(s)
- Jong Won Hong
- Department of Plastic & Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Institute for Human Tissue Restoration, College of Medicine, Yonsei University, Seoul, Korea.
| | - Jung Hyun Lim
- Department of Plastic & Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Institute for Human Tissue Restoration, College of Medicine, Yonsei University, Seoul, Korea
| | - Chooryung J Chung
- Department of Orthodontics, College of Dentistry, Yonsei University, Seoul, Korea
| | - Tae Jo Kang
- Department of Plastic & Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Yujin Plastic Surgery, Seoul, Korea
| | - Tae Yeon Kim
- Department of Orthodontics, College of Dentistry, Yonsei University, Seoul, Korea
| | - Young Seok Kim
- Department of Plastic & Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Institute for Human Tissue Restoration, College of Medicine, Yonsei University, Seoul, Korea
| | - Tae Suk Roh
- Department of Plastic & Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Institute for Human Tissue Restoration, College of Medicine, Yonsei University, Seoul, Korea
| | - Dae Hyun Lew
- Department of Plastic & Reconstructive Surgery, College of Medicine, Yonsei University, Seoul, Korea
- Institute for Human Tissue Restoration, College of Medicine, Yonsei University, Seoul, Korea
| |
Collapse
|
22
|
Bao L, Meng Q, Li Y, Deng S, Yu Z, Liu Z, Zhang L, Fan H. C-Kit Positive Cardiac Stem Cells and Bone Marrow-Derived Mesenchymal Stem Cells Synergistically Enhance Angiogenesis and Improve Cardiac Function After Myocardial Infarction in a Paracrine Manner. J Card Fail 2017; 23:403-415. [PMID: 28284757 DOI: 10.1016/j.cardfail.2017.03.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/16/2017] [Accepted: 03/06/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Stem cell transplantation offers a promising treatment for heart failure. Recent studies show that both c-kit positive cardiac stem cells (CSCs) and bone marrow-derived mesenchymal stem cells (BM-MSCs) are good candidates for stem cell therapy to treat heart failure; however, the exact mechanism of stem cell therapy in improving cardiac function of ischemic cardiomyopathy is not fully known. Our objective was to test our hypothesis that CSCs and/or BM-MSCs repair the damaged heart by boosting post-myocardial infarction (MI) angiogenesis in a paracrine manner. METHODS AND RESULTS We isolated and purified CSCs and BM-MSCs from rats. Intramyocardial injections of CSCs and/or BM-MSCs were performed at 28 days after MI. We applied cardiac ultrasound and histological analysis to evaluate the effect of cell therapy on cardiac function and cardiac remodeling. In vivo donor cell transplantation experiments showed that CSCs and/or BM-MSCs improved cardiac function after MI and reduced infarct size. However, in vivo cell tracking experiments showed that minimal donor cells remained in the myocardium after cell transplantation. Our further in vitro and in vivo experiments showed that transplantation of CSCs enhanced the expression of pro-angiogenic factors and boosted post-MI angiogenesis in the myocardium in a paracrine manner, which in part contributed to the effect of CSCs on cardiac recovery after MI. CSCs and BM-MSCs synergistically inhibited CSC/BM-MSC apoptosis and enhanced their proliferation in a paracrine manner. This resulted in a larger number of transplanted cells remaining in the post-MI myocardium after coinjection of CSCs and BM-MSCs, and therefore the accumulation of more pro-angiogenic factors in the heart tissue compared to transplantation of CSCs or MSCs alone. Consequently, transplantation of both CSCs and BM-MSCs was superior to transplantation of either CSCs or BM-MSCs alone to boost post-MI angiogenesis and improve cardiac function after MI. CONCLUSION C-kit+ CSC and/or BM-MSC transplantation can improve cardiac function after MI in a paracrine manner. Coinjection of both CSCs and BM-MSCs improves cardiac function more significantly than CSC or BM-MSC transplantation alone in a paracrine manner by improving the engraftment of donor cells and boosting the expression of multiple pro-angiogenic factors.
Collapse
Affiliation(s)
- Luer Bao
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qingshu Meng
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuan Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Shengqiong Deng
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zuoren Yu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Lin Zhang
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Huimin Fan
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
23
|
Aktas E, Chamberlain CS, Saether EE, Duenwald-Kuehl SE, Kondratko-Mittnacht J, Stitgen M, Lee JS, Clements AE, Murphy WL, Vanderby R. Immune modulation with primed mesenchymal stem cells delivered via biodegradable scaffold to repair an Achilles tendon segmental defect. J Orthop Res 2017; 35:269-280. [PMID: 27061844 DOI: 10.1002/jor.23258] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/04/2016] [Indexed: 02/06/2023]
Abstract
Tendon healing is a complex coordinated series of events resulting in protracted recovery, limited regeneration, and scar formation. Mesenchymal stem cell (MSC) therapy has shown promise as a new technology to enhance soft tissue and bone healing. A challenge with MSC therapy involves the ability to consistently control the inflammatory response and subsequent healing. Previous studies suggest that preconditioning MSCs with inflammatory cytokines, such as IFN-γ, TNF-α, and IL-1β may accelerate cutaneous wound closure. The objective of this study was to therefore elucidate these effects in tendon. That is, the in vivo healing effects of TNF-α primed MSCs were studied using a rat Achilles segmental defect model. Rat Achilles tendons were subjected to a unilateral 3 mm segmental defect and repaired with either a PLG scaffold alone, MSC-seeded PLG scaffold, or TNF-α-primed MSC-seeded PLG scaffold. Achilles tendons were analyzed at 2 and 4 weeks post-injury. In vivo, MSCs, regardless of priming, increased IL-10 production and reduced the inflammatory factor, IL-1α. Primed MSCs reduced IL-12 production and the number of M1 macrophages, as well as increased the percent of M2 macrophages, and synthesis of the anti-inflammatory factor IL-4. Primed MSC treatment also increased the concentration of type I procollagen in the healing tissue and increased failure stress of the tendon 4 weeks post-injury. Taken together delivery of TNF-α primed MSCs via 3D PLG scaffold modulated macrophage polarization and cytokine production to further accentuate the more regenerative MSC-induced healing response. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:269-280, 2017.
Collapse
Affiliation(s)
- Erdem Aktas
- Department of Orthopedics, Ankara Oncology Research and Training Hospital, Ankara, Turkey
| | - Connie S Chamberlain
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705
| | - Erin E Saether
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Sarah E Duenwald-Kuehl
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | | | - Michael Stitgen
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Jae Sung Lee
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Anna E Clements
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| | - Ray Vanderby
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, 53705.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, 53705
| |
Collapse
|
24
|
Mesenchymal stem cells increase skin graft survival time and up-regulate PD-L1 expression in splenocytes of mice. Immunol Lett 2017; 182:39-49. [PMID: 28069488 DOI: 10.1016/j.imlet.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/26/2016] [Accepted: 01/05/2017] [Indexed: 02/07/2023]
Abstract
Recently, mesenchymal stem cells (MSCs) have gained considerable interests as hopeful therapeutic cells in transplantation due to their immunoregulatory functions. But exact mechanisms underlying MSCs immunoregulatory function is not fully understood. Herein, in addition to investigate the ability of MSCs to prolong graft survival time, the effects of them on the expression of PD-L1 and IDO immunomodulatory molecules in splenocytes of skin graft recipient mice was clarified. To achieve this goal, full-thickness skins were transplanted from C57BL/6 to BALB/c mice. MSCs were isolated from bone marrow of BALB/c mice and injected to the recipient mice. Skin graft survival was monitored daily to determine graft rejection time. On days 2, 5 and 10 post skin transplantation, serum cytokine levels and expression of PD-L1 and IDO mRNA and protein in the splenocytes of recipient mice were evaluated. The results showed that administration of MSCs prolonged skin graft survival time from 11 to 14 days. On days 2 and 5 post transplantation, splenocytes PD-L1 expression and IL-10 serum level in MSCs treated mice were higher than those in the controls, while IL-2 and IFN-γ levels were lower. Rejection in MSCs treated mice was accompanied by an increase in IL-2 and IFN-γ, and decrease in PD-L1 expression and IL-10 level. No difference in the expression of IDO between MSCs treated mice and controls was observed. In conclusion, we found that one of the mechanisms underlying MSCs immunomodulatory function could be up-regulating PD-L1 expression.
Collapse
|
25
|
Immunomodulatory Role of Mesenchymal Stem Cell Therapy in Vascularized Composite Allotransplantation. J Transplant 2016; 2016:6951693. [PMID: 27822384 PMCID: PMC5086388 DOI: 10.1155/2016/6951693] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 09/15/2016] [Indexed: 12/12/2022] Open
Abstract
This review aims to summarize contemporary evidence of the in vitro and in vivo immunomodulatory effects of mesenchymal stem cells (MSCs) in promoting vascularized composite allotransplant (VCA) tolerance. An extensive literature review was performed to identify pertinent articles of merit. Prospective preclinical trials in mammal subjects receiving VCA (or skin allograft) with administration of MSCs were reviewed. Prospective clinical trials with intravascular delivery of MSCs in human populations undergoing solid organ transplant were also identified and reviewed. Sixteen preclinical studies are included. Eleven studies compared MSC monotherapy to no therapy; of these, ten reported improved graft survival, which was statistically significantly prolonged in eight. Eight studies analyzed allograft survival with MSC therapy as an adjunct to proven immunosuppressive regimens. In these studies, daily immunosuppression was transiently delivered and then stopped. In all studies, treatment-free graft survival was statistically significantly prolonged in animals that received MSC therapy. MSCs have been safely administered clinically and their use in renal transplant clinical trials provides evidence that they improve allograft transplant tolerance in clinical practice. There is potential for MSC induction therapy to overcome many of the obstacles to widespread VCA in clinical practice. Preclinical studies are needed before MSC-induced VCA tolerance becomes a clinical reality.
Collapse
|
26
|
Hoornaert CJ, Luyckx E, Reekmans K, Dhainaut M, Guglielmetti C, Le Blon D, Dooley D, Fransen E, Daans J, Verbeeck L, Quarta A, De Vocht N, Lemmens E, Goossens H, Van der Linden A, Roobrouck VD, Verfaillie C, Hendrix S, Moser M, Berneman ZN, Ponsaerts P. In Vivo Interleukin-13-Primed Macrophages Contribute to Reduced Alloantigen-Specific T Cell Activation and Prolong Immunological Survival of Allogeneic Mesenchymal Stem Cell Implants. Stem Cells 2016; 34:1971-84. [PMID: 26992046 DOI: 10.1002/stem.2360] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 02/12/2016] [Indexed: 12/11/2022]
Abstract
Transplantation of mesenchymal stem cells (MSCs) into injured or diseased tissue-for the in situ delivery of a wide variety of MSC-secreted therapeutic proteins-is an emerging approach for the modulation of the clinical course of several diseases and traumata. From an emergency point-of-view, allogeneic MSCs have numerous advantages over patient-specific autologous MSCs since "off-the-shelf" cell preparations could be readily available for instant therapeutic intervention following acute injury. Although we confirmed the in vitro immunomodulatory capacity of allogeneic MSCs on antigen-presenting cells with standard coculture experiments, allogeneic MSC grafts were irrevocably rejected by the host's immune system upon either intramuscular or intracerebral transplantation. In an attempt to modulate MSC allograft rejection in vivo, we transduced MSCs with an interleukin-13 (IL13)-expressing lentiviral vector. Our data clearly indicate that prolonged survival of IL13-expressing allogeneic MSC grafts in muscle tissue coincided with the induction of an alternatively activated macrophage phenotype in vivo and a reduced number of alloantigen-reactive IFNγ- and/or IL2-producing CD8(+) T cells compared to nonmodified allografts. Similarly, intracerebral IL13-expressing MSC allografts also exhibited prolonged survival and induction of an alternatively activated macrophage phenotype, although a peripheral T cell component was absent. In summary, this study demonstrates that both innate and adaptive immune responses are effectively modulated in vivo by locally secreted IL13, ultimately resulting in prolonged MSC allograft survival in both muscle and brain tissue. Stem Cells 2016;34:1971-1984.
Collapse
Affiliation(s)
- Chloé J Hoornaert
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Evi Luyckx
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Kristien Reekmans
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Maxime Dhainaut
- Laboratory of Immunobiology, Department of Molecular Biology, Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Debbie Le Blon
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Dearbhaile Dooley
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Erik Fransen
- StatUa Centre for Statistics, University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Louca Verbeeck
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Nathalie De Vocht
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Evi Lemmens
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | | | - Valerie D Roobrouck
- Stem Cell Institute, Stem Cell Biology and Embryology Unit, KU Leuven, Leuven, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute, Stem Cell Biology and Embryology Unit, KU Leuven, Leuven, Belgium
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Muriel Moser
- Laboratory of Immunobiology, Department of Molecular Biology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
27
|
MicroRNA-27b Enhances the Hepatic Regenerative Properties of Adipose-Derived Mesenchymal Stem Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e285. [PMID: 26836372 PMCID: PMC4884788 DOI: 10.1038/mtna.2015.55] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 11/27/2015] [Indexed: 01/10/2023]
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are readily available multipotent mesenchymal progenitor cells and have become an attractive therapeutic tool for regenerative medicine. We herein investigated the mechanistic role of how miR-27b modulated regenerative capacities of ASCs. Intravenous administration of miR-27b-transfected ASCs (ASCs-miR-27b) was conducted after 70% partial hepatectomy (PH). After PH, rats injected with ASCs-miR-27b had decreased inflammatory cytokines and increased hepatocyte growth factor and other related growth factors. We showed that the nature of ASCs-miR-27b to inhibit hepatic stellate cell activation was dependent upon peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) in vitro. Moreover, expression of miR-27b in ASCs induced heme oxygenase-1 (HO-1), resulting in increased production of ATP, protective cytokines/growth factors, and genes involved in mitochondrial biogenesis in a PGC-1α-dependent manner. RNA sequencing (RNA-Seq) analysis revealed drastic transcriptional changes in livers treated with ASCs-miR-27b after PH. The differentially expressed genes classified into "regeneration," "fibrosis," and "mitochondrial biogenesis" clusters were mainly mitochondrial. The potential biological context reflecting the effects of PGC-1α by ASCs-miR-27b treatment was also observed by the subnetwork analysis with HO-1 and PGC-1α being the top-ranked regulatory genes. We demonstrate autologous ASCs-miR-27b enhances liver regeneration and, importantly, preserves hepatic function through paracrine actions which offers a viable therapeutic option to facilitate rapid recovery after liver injury.
Collapse
|
28
|
Hivelin M, Klimczak A, Cwykiel J, Sonmez E, Nasir S, Gatherwright J, Siemionow M. Immunomodulatory Effects of Different Cellular Therapies of Bone Marrow Origin on Chimerism Induction and Maintenance Across MHC Barriers in a Face Allotransplantation Model. Arch Immunol Ther Exp (Warsz) 2015; 64:299-310. [PMID: 26708158 DOI: 10.1007/s00005-015-0380-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/05/2015] [Indexed: 01/31/2023]
Abstract
Many more patients would benefit from vascularized composite allotransplantation if less toxic and safer immunosuppressive protocols will become available. Tolerance induction protocols with donor cells co-transplantation are one of the promising pathways to reduce maintenance immunosupressive regimens. We investigated the role of donor bone marrow cells (BMC), mesenchymal stromal cells (MSC) and in vivo created chimeric cells (CC) used as supportive therapies in a fully MHC-mismatched rat face transplantation model. Twenty-four fully MHC-mismatched hemiface transplantations were performed between ACI (RT1(a)) donors and Lewis (RT1(l)) recipients under combined seven-day immunosuppressive regimen of anti-αβ-T-cell receptor (TCR) monoclonal antibody and cyclosporin A. We studied four experimental groups-group 1: no cellular therapy; group 2: supportive therapy with BMC; group 3: supportive therapy with MSC; group 4: supportive therapy with CC generated in a primary chimera. We evaluated clinical and histological rejection grades, transplanted cells migration, donor-specific chimerism in the peripheral blood and bone marrow compartments, and CD4(+)/CD25(+) T-cell levels. Face allograft rejection was observed at 26.8 ± 0.6 days post-transplant (PT) in the absence of cellular therapy, at 34.5 ± 1.1 days for group 2, 29.3 ± 0.8 days for group 3, and 30.3 ± 1.38 PT for group 4. The longest survival was observed in allografts supported by co-transplantation of BMC. All support in cellular therapies delayed face allograft rejection by chimerism induction and/or immunomodulatory properties of co-transplanted cells. Survival time was comparable between groups, however, further studies, with different cell dosages, delivery routes and delivery times are required.
Collapse
Affiliation(s)
- Mikael Hivelin
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Aleksandra Klimczak
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.,L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Joanna Cwykiel
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA.,Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA
| | - Erhan Sonmez
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Serdar Nasir
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | | | - Maria Siemionow
- Department of Plastic Surgery, Cleveland Clinic, Cleveland, OH, USA. .,Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
29
|
The challenges and promises of allogeneic mesenchymal stem cells for use as a cell-based therapy. Stem Cell Res Ther 2015; 6:234. [PMID: 26620426 PMCID: PMC4665863 DOI: 10.1186/s13287-015-0240-9] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are ideal for cell-based therapy in various inflammatory diseases because of their immunosuppressive and tissue repair properties. Moreover, their immunosuppressive properties and low immunogenicity contribute to a reduced or weakened immune response elicited by the implantation of allogeneic MSCs compared with other cell types. Therefore, implantation of allogeneic MSCs may be a promising cell-based therapy. In this review, we first summarize the unique advantages of allogeneic MSCs for therapeutic applications. Second, we critically analyze the factors influencing their therapeutic effects, including administration routes, detection time-points, disease models, differentiation of MSCs in vivo, and timing and dosage of MSC administration. Finally, current approaches to allogeneic MSC application are discussed. In conclusion, allogeneic MSCs are a promising option because of their low immunogenicity and immunosuppressive and tissue repair capabilities. Further investigations are needed to enhance the consistency and efficacy of MSCs when used as a cell-based therapy in inflammatory diseases as well as for tissue repair.
Collapse
|
30
|
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: 5.9] [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
|
31
|
Tsuji W, Schnider JT, McLaughlin MM, Schweizer R, Zhang W, Solari MG, Rubin JP, Marra KG, Plock JA, Gorantla VS. Effects of immunosuppressive drugs on viability and susceptibility of adipose- and bone marrow-derived mesenchymal stem cells. Front Immunol 2015; 6:131. [PMID: 25932028 PMCID: PMC4399413 DOI: 10.3389/fimmu.2015.00131] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/10/2015] [Indexed: 01/22/2023] Open
Abstract
The immunomodulatory potential of cell therapies using adipose-derived stem cells (ASCs) and bone marrow-derived mesenchymal stem cells (BM-MSCs) has been studied in vascularized composite allotransplantation (VCA). Most cell therapy-based experimental and clinical protocols integrate some degree of recipient conditioning/induction with antibodies or other immunosuppressive agents. We investigated the susceptibility of ASCs and BM-MSCs to anti-lymphocyte serum (ALS) and tacrolimus. Rat ASCs and BM-MSCs were exposed to varying concentrations of tacrolimus and ALS in vitro. Serum from ALS-treated animals was added to cell cultures. Viability, susceptibility, and cytotoxicity parameters were evaluated. ALS inhibited ASC and BM-MSC viability and susceptibility in vitro in a dose-dependent manner. ASCs were more susceptible to both ALS and tacrolimus than BM-MSCs. Trypsinized and adherent ASCs were significantly smaller than BM-MSCs. This is the first report on the viability and susceptibility characteristics of BM-MSCs or ASCs to collateral effects of ALS and tacrolimus. These in vitro insights may impact choice of cell type as well as concomitant conditioning agents and the logistical coordination of the timing, dosing, and frequency of drug or cell therapy in solid organ transplantation or VCA protocols.
Collapse
Affiliation(s)
- Wakako Tsuji
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Surgery, Shiga Medical Center for Adults , Moriyama , Japan
| | - Jonas T Schnider
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA
| | - Meghan M McLaughlin
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA
| | - Riccardo Schweizer
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Plastic Surgery and Hand Surgery, University Hospital Zurich , Zurich , Switzerland
| | - Wensheng Zhang
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA
| | - Mario G Solari
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Bioengineering, University of Pittsburgh , Pittsburgh, PA , USA
| | - Kacey G Marra
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Bioengineering, University of Pittsburgh , Pittsburgh, PA , USA
| | - Jan A Plock
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Plastic Surgery and Hand Surgery, University Hospital Zurich , Zurich , Switzerland
| | - Vijay S Gorantla
- Department of Plastic Surgery, University of Pittsburgh , Pittsburgh, PA , USA ; McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, PA , USA
| |
Collapse
|
32
|
Yang JF, Cao HC, Pan QL, Yu J, Li J, Li LJ. Mesenchymal stem cells from the human umbilical cord ameliorate fulminant hepatic failure and increase survival in mice. Hepatobiliary Pancreat Dis Int 2015; 14:186-93. [PMID: 25865692 DOI: 10.1016/s1499-3872(15)60354-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cell therapy has been promising for various diseases. We investigated whether transplantation of human umbilical cord mesenchymal stem cells (hUCMSCs) has any therapeutic effects on D-galactosamine/lipopolysaccharide (GalN/LPS)-induced fulminant hepatic failure in mice. METHODS hUCMSCs isolated from human umbilical cord were cultured and transplanted via the tail vein into severe combined immune deficiency mice with GalN/LPS-induced fulminant hepatic failure. After transplantation, the localization and differentiation of hUCMSCs in the injured livers were investigated by immunohistochemical and genetic analyses. The recovery of the injured livers was evaluated histologically. The survival rate of experimental animals was analyzed by the Kaplan-Meier method and log-rank test. RESULTS hUCMSCs expressed high levels of CD29, CD73, CD13, CD105 and CD90, but did not express CD31, CD79b, CD133, CD34, and CD45. Cultured hUCMSCs displayed adipogenic and osteogenic differentiation potential. Hematoxylin and eosin staining revealed that transplantation of hUCMSCs reduced hepatic necrosis and promoted liver regeneration. Transplantation of hUCMSCs prolonged the survival rate of mice with fulminant hepatic failure. Polymerase chain reaction for human alu sequences showed the presence of human cells in mouse livers. Positive staining for human albumin, human alpha-fetoprotein and human cytokeratin 18 suggested the formation of hUCMSCs-derived hepatocyte-like cells in vivo. CONCLUSIONS hUCMSC was a potential candidate for stem cell based therapies. After transplantation, hUCMSCs partially repaired hepatic damage induced by GalN/LPS in mice. hUCMSCs engrafted into the injured liver and differentiated into hepatocyte-like cells.
Collapse
Affiliation(s)
- Jin-Feng Yang
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Hangzhou 310003, China.
| | | | | | | | | | | |
Collapse
|
33
|
Preclinical evaluation of the immunomodulatory properties of cardiac adipose tissue progenitor cells using umbilical cord blood mesenchymal stem cells: a direct comparative study. BIOMED RESEARCH INTERNATIONAL 2015; 2015:439808. [PMID: 25861626 PMCID: PMC4377370 DOI: 10.1155/2015/439808] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/20/2015] [Accepted: 02/16/2015] [Indexed: 01/14/2023]
Abstract
Cell-based strategies to regenerate injured myocardial tissue have emerged over the past decade, but the optimum cell type is still under scrutiny. In this context, human adult epicardial fat surrounding the heart has been characterized as a reservoir of mesenchymal-like progenitor cells (cardiac ATDPCs) with potential clinical benefits. However, additional data on the possibility that these cells could trigger a deleterious immune response following implantation are needed. Thus, in the presented study, we took advantage of the well-established low immunogenicity of umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) to comparatively assess the immunomodulatory properties of cardiac ATDPCs in an in vitro allostimulatory assay using allogeneic mature monocyte-derived dendritic cells (MDDCs). Similar to UCBMSCs, increasing amounts of seeded cardiac ATDPCs suppressed the alloproliferation of T cells in a dose-dependent manner. Secretion of proinflammatory cytokines (IL6, TNFα, and IFNγ) was also specifically modulated by the different numbers of cardiac ATDPCs cocultured. In summary, we show that cardiac ATDPCs abrogate T cell alloproliferation upon stimulation with allogeneic mature MDDCs, suggesting that they could further regulate a possible harmful immune response in vivo. Additionally, UCBMSCs can be considered as valuable tools to preclinically predict the immunogenicity of prospective regenerative cells.
Collapse
|
34
|
Sivanathan KN, Gronthos S, Rojas-Canales D, Thierry B, Coates PT. Interferon-gamma modification of mesenchymal stem cells: implications of autologous and allogeneic mesenchymal stem cell therapy in allotransplantation. Stem Cell Rev Rep 2014; 10:351-75. [PMID: 24510581 DOI: 10.1007/s12015-014-9495-2] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSC) have unique immunomodulatory and reparative properties beneficial for allotransplantation cellular therapy. The clinical administration of autologous or allogeneic MSC with immunosuppressive drugs is able to prevent and treat allograft rejection in kidney transplant recipients, thus supporting the immunomodulatory role of MSC. Interferon-gamma (IFN-γ) is known to enhance the immunosuppressive properties of MSC. IFN-γ preactivated MSC (MSC-γ) directly or indirectly modulates T cell responses by enhancing or inducing MSC inhibitory factors. These factors are known to downregulate T cell activation, enhance T cell negative signalling, alter T cells from a proinflammatory to an anti-inflammatory phenotype, interact with antigen-presenting cells and increase or induce regulatory cells. Highly immunosuppressive MSC-γ with increased migratory and reparative capacities may aid tissue repair, prolong allograft survival and induce allotransplant tolerance in experimental models. Nevertheless, there are contradictory in vivo observations related to allogeneic MSC-γ therapy. Many studies report that allogeneic MSC are immunogenic due to their inherent expression of major histocompatibility (MHC) molecules. Enhanced expression of MHC in allogeneic MSC-γ may increase their immunogenicity and this can negatively impact allograft survival. Therefore, strategies to reduce MSC-γ immunogenicity would facilitate "off-the-shelf" MSC therapy to efficiently inhibit alloimmune rejection and promote tissue repair in allotransplantation. In this review, we examine the potential benefits of MSC therapy in the context of allotransplantation. We also discuss the use of autologous and allogeneic MSC and the issues associated with their immunogenicity in vivo, with particular focus on the use of enhanced MSC-γ cellular therapy.
Collapse
Affiliation(s)
- Kisha Nandini Sivanathan
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005, South Australia, Australia,
| | | | | | | | | |
Collapse
|
35
|
Li C, Wang X, Tan J, Wang T, Wang Q. The immunomodulatory properties of periodontal ligament stem cells isolated from inflamed periodontal granulation. Cells Tissues Organs 2014; 199:256-65. [PMID: 25471814 DOI: 10.1159/000367986] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2014] [Indexed: 12/15/2022] Open
Abstract
Periodontitis is currently the main cause of tooth loss and as yet there is no appropriate method for establishing a functional and predictable periodontal regeneration. Tissue engineering involving seed cells provides a new prospect for periodontal regeneration. While periodontal ligament stem cells (PDLSCs) are a good choice for seed cells, it is not always possible to obtain the patients' own PDLSCs. We and others have found a type of stromal cells from inflamed periodontal granulation. These cells displayed similar differentiation properties to PDLSCs. Inflammation has a profound influence on the immunomodulatory properties of mesenchymal stem cells, which may affect therapeutic outcome. In this study, we assessed the immunomodulatory characteristics of these inflamed human (ih)PDLSCs. Along with the similarity in cell surface marker expressions, they also displayed immunomodulatory properties comparable to those in healthy human (hh)PDLSCs. Both hhPDLSCs and ihPDLSCs can suppress the proliferation and secretion of IFN-γ in peripheral blood mononuclear cells by indirect soluble mediators and direct cell-cell contact. Albeit with some quantitative variances, the gene expressions of inducible nitric oxide synthases, indoleamine 2,3 dioxygenase, cyclooxygenase-2, TNF-α-induced protein 6 and IL-10 in ihPDLSCs displayed similar patterns as those in hhPDLSCs. Taken together, our results suggest that ihPDLSCs can provide a promising alternative to hhPDLSCs in terms of evident similarities in immunomodulatory properties as well as their easier accessibility and availability.
Collapse
Affiliation(s)
- Chenghua Li
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, PR China
| | | | | | | | | |
Collapse
|
36
|
Vandermeulen M, Grégoire C, Briquet A, Lechanteur C, Beguin Y, Detry O. Rationale for the potential use of mesenchymal stromal cells in liver transplantation. World J Gastroenterol 2014; 20:16418-16432. [PMID: 25469010 PMCID: PMC4248185 DOI: 10.3748/wjg.v20.i44.16418] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and self-renewing cells that reside essentially in the bone marrow as a non-hematopoietic cell population, but may also be isolated from the connective tissues of most organs. MSCs represent a heterogeneous population of adult, fibroblast-like cells characterized by their ability to differentiate into tissues of mesodermal lineages including adipocytes, chondrocytes and osteocytes. For several years now, MSCs have been evaluated for their in vivo and in vitro immunomodulatory and ‘tissue reconstruction’ properties, which could make them interesting in various clinical settings, and particularly in organ transplantation. This paper aims to review current knowledge on the properties of MSCs and their use in pre-clinical and clinical studies in solid organ transplantation, and particularly in the field of liver transplantation. The first available clinical data seem to show that MSCs are safe to use, at least in the medium-term, but more time is needed to evaluate the potential adverse effects of long-term use. Many issues must be resolved on the correct use of MSCs. Intensive in vitro and pre-clinical research are the keys to a better understanding of the way that MSCs act, and to eventually lead to clinical success.
Collapse
|
37
|
Zhang FG, Tang XF. New advances in the mesenchymal stem cells therapy against skin flaps necrosis. World J Stem Cells 2014; 6:491-496. [PMID: 25258671 PMCID: PMC4172678 DOI: 10.4252/wjsc.v6.i4.491] [Citation(s) in RCA: 10] [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: 07/16/2014] [Revised: 08/06/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotential cells that reside within the bone marrow, can be induced to differentiate into various cells, such as osteoblasts, adipocytes, chondrocytes, vascular endothelial progenitor cells, and other cell types. MSCs are being widely studied as potential cell therapy agents due to their angiogenic properties, which have been well established by in vitro and in vivo researches. Within this context, MSCs therapy appears to hold substantial promise, particularly in the treatment of conditions involving skin grafts, pedicle flaps, as well as free flaps described in literatures. The purpose of this review is to report the new advances and mechanisms underlying MSCs therapy against skin flaps necrosis.
Collapse
|
38
|
Abrate A, Buono R, Canu T, Esposito A, Del Maschio A, Lucianò R, Bettiga A, Colciago G, Guazzoni G, Benigni F, Hedlund P, Altaner C, Montorsi F, Cavarretta IT. Mesenchymal stem cells expressing therapeutic genes induce autochthonous prostate tumour regression. Eur J Cancer 2014; 50:2478-88. [DOI: 10.1016/j.ejca.2014.06.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 06/08/2014] [Accepted: 06/16/2014] [Indexed: 01/14/2023]
|
39
|
Jiang X, Liu C, Hao J, Guo D, Guo J, Yao J, Jiang K, Cui Z, Zhu L, Sun W, Lin L, Liang J. CD4(+)CD25 (+) regulatory T cells are not required for mesenchymal stem cell function in fully MHC-mismatched mouse cardiac transplantation. Cell Tissue Res 2014; 358:503-14. [PMID: 25103227 DOI: 10.1007/s00441-014-1956-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 07/01/2014] [Indexed: 01/12/2023]
Abstract
Although the immunomodulative properties of mesenchymal stem cells (MSCs) open up attractive possibilities in solid-organ transplantation, information concerning the optimal dose, route, timing of administration, major histocompatibility complex (MHC)-restriction and relevant mechanisms is currently lacking. Therefore, better characterization of MSC immunoregulatory activity and elucidation of its mechanisms are crucial. In this study, we confirmed that MSCs did not elicit proliferation by allogeneic CD4(+) T cells, suggesting that MSCs were not immunogenic. By using C57BL/6 mouse MSCs as donor-derived or recipient-derived or as third-party MSCs, we discovered that MSCs suppressed CD4(+) T cell proliferation and prolonged mouse cardiac allograft survival in a dose-dependent and non-MHC-restricted manner. We also found that intraperitoneal administration favored survival prolongation, although this prolongation was weaker than that via the intravenous route. Only infusion at earlier time points favored survival prolongation. Depletion of CD4(+)CD25(+) T cells did not affect the immunosuppression of MSCs on CD4(+) T cells. Moreover, MSCs did not induce regulatory T cells. The in vivo data revealed that MSCs did not increase the percentage of CD4(+)CD25(+) T cells and FoxP3 expression. More importantly, we demonstrated for the first time that depletion of CD4(+)CD25(+) T cells did not hinder MSC-induced survival prolongation, indicating that CD4(+)CD25(+) regulatory T cells were not essential for the prolongation of MSC-mediated allograft survival.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, Liaoning Province, People's Republic of China,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Li F, Zhao SZ. Mesenchymal stem cells: Potential role in corneal wound repair and transplantation. World J Stem Cells 2014; 6:296-304. [PMID: 25126379 PMCID: PMC4131271 DOI: 10.4252/wjsc.v6.i3.296] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/06/2014] [Accepted: 04/11/2014] [Indexed: 02/06/2023] Open
Abstract
Corneal diseases are a major cause of blindness in the world. Although great progress has been achieved in the treatment of corneal diseases, wound healing after severe corneal damage and immunosuppressive therapy after corneal transplantation remain problematic. Mesenchymal stem cells (MSCs) derived from bone marrow or other adult tissues can differentiate into various types of mesenchymal lineages, such as osteocytes, adipocytes, and chondrocytes, both in vivo and in vitro. These cells can further differentiate into specific cell types under specific conditions. MSCs migrate to injury sites and promote wound healing by secreting anti-inflammatory and growth factors. In addition, MSCs interact with innate and acquired immune cells and modulate the immune response through their powerful paracrine function. Over the last decade, MSCs have drawn considerable attention because of their beneficial properties and promising therapeutic prospective. Furthermore, MSCs have been applied to various studies related to wound healing, autoimmune diseases, and organ transplantation. This review discusses the potential functions of MSCs in protecting corneal tissue and their possible mechanisms in corneal wound healing and corneal transplantation.
Collapse
|
41
|
Lett B, Sivanathan KN, Coates PT. Mesenchymal stem cells for kidney transplantation. World J Clin Urol 2014; 3:87-95. [DOI: 10.5410/wjcu.v3.i2.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/06/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
The long term consequence of immunosuppressive therapy in kidney transplantation has prompted investigation of alternative means to modify the immune response to the allograft. Cell based therapies are potentially attractive as they may provide a long lasting immunomodulatory effect, may repair tissues and reduce the necessity to take immunosuppressive drug therapy. Of the current cell therapies, mesenchymal stem cells have now been trialled in small numbers of human kidney transplantation with apparent safety and potential efficacy. Many issues however need to be resolved before these cells will become mainstays of transplant immunosuppression including ex vivo modification to enhance immunomodulatory properties, cell number, route and frequency of administration as well as cellular source of origin.
Collapse
|
42
|
Zhang W, Shen ZY, Song HL, Yang Y, Wu BJ, Fu NN, Liu T. Protective effect of bone marrow mesenchymal stem cells in intestinal barrier permeability after heterotopic intestinal transplantation. World J Gastroenterol 2014; 20:7442-7451. [PMID: 24966614 PMCID: PMC4064089 DOI: 10.3748/wjg.v20.i23.7442] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 04/23/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the protective effect of bone marrow mesenchymal stem cells (BM MSCs) in the small intestinal mucosal barrier following heterotopic intestinal transplantation (HIT) in a rat model.
METHODS: BM MSCs were isolated from male Lewis rats by density gradient centrifugation, cultured, and analyzed by flow cytometry. The HIT models were divided into a non-rejection group, saline-treated rejection group (via penile vein), and BM MSC–treated group (via penile vein). Intestinal mucosal barrier injury was estimated by diamine oxidase (DAO) and D-lactic acid (D-LA) expression levels. Tumor necrosis factor-α (TNF-α), interferon-γ (INF-γ), interleukin-10 (IL-10), and transforming growth factor-β (TGF-β) were detected by enzyme-linked immunosorbent assay. Ultrastructural change of tight junctions (TJs) was observed under transmission electron microscope. Expression levels of the TJ proteins occludin and zona occludens (ZO)-1, affected by the inflammatory factors, were measured using real-time polymerase chain reaction and Western blotting.
RESULTS: The pathological score at each time point after surgery indicated significantly less serious injury in the BM MSCs-treated group than in the rejection group (P < 0.05). In the former, graft levels of DAO and D-LA were reduced, and TNF-α and INF-γ production was inhibited (at day 7: 10.6473 ± 0.0710 vs 17.2128 ± 0.4991, P < 0.05; 545.1506 ± 31.9416 vs 810.2637 ± 25.1175, P < 0.05). IL-10 and TGF-β production was increased greatly (at day 7: 125.7773 ± 4.7719 vs 80.3756 ± 2.5866, P < 0.05; 234.5273 ± 9.3980 vs 545.1506 ± 31.9416, P < 0.05). There was increased expression of occludin and ZO-1 protein (at day 7: 0.2674 ± 0.0128 vs 0.1352 ± 0.0142, P < 0.05; at day 5: 0.7189 ± 0.0289 vs 0.4556 ± 0.0242, P < 0.05) and mRNA (at day 7: 0.3860 ± 0.0254 vs 0.1673 ± 0.0369, P < 0.05; at day 5: 0.5727 ± 0.0419 vs 0.3598 ± 0.0242, P < 0.05).
CONCLUSION: BM MSCs can improve intestinal barrier permeability, repair TJs, and increase occludin and ZO-1 protein expression. With altered cytokine levels, they can protect the intestinal mucosa after transplantation.
Collapse
MESH Headings
- Amine Oxidase (Copper-Containing)/metabolism
- Animals
- Bone Marrow Transplantation
- Cells, Cultured
- Cytokines/metabolism
- Graft Survival
- Inflammation Mediators/metabolism
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/transplantation
- Intestinal Mucosa/ultrastructure
- Intestine, Small/metabolism
- Intestine, Small/transplantation
- Intestine, Small/ultrastructure
- Lactic Acid/metabolism
- Male
- Mesenchymal Stem Cell Transplantation
- Microscopy, Electron, Transmission
- Occludin/genetics
- Occludin/metabolism
- Permeability
- RNA, Messenger/metabolism
- Rats, Inbred BN
- Rats, Inbred Lew
- Tight Junctions/metabolism
- Tight Junctions/ultrastructure
- Time Factors
- Transplantation, Heterotopic
- Zonula Occludens-1 Protein/genetics
- Zonula Occludens-1 Protein/metabolism
Collapse
|
43
|
Immunomodulatory effects of mesenchymal stromal cells in solid organ transplantation. Curr Opin Organ Transplant 2014; 15:731-7. [PMID: 20881495 DOI: 10.1097/mot.0b013e328340172c] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Multipotent mesenchymal stromal cells (MSCs) possess powerful immunomodulatory activity highlighting the potential for their clinical translation in solid organ transplantation. In this review, we summarize recent advances in understanding MSC immunomodulatory effect in vitro and in experimental transplant models and discuss topics of crucial importance for the future clinical use of MSCs as immunotherapy in solid organ transplantation. RECENT FINDINGS MSCs strongly inhibited T-cell activity in vitro and exerted similar inhibitory effects on other cells of the immune system. MSC-mediated immune suppression has been attributed mainly to the secretion of soluble factors; however, cell-contact mechanisms cannot be excluded. Available studies in animal transplant models raised variable results, but overall indicate that MSCs could be useful to modulate recipient immune cells. The timing of cell application and the origin of MSCs (autologous or allogeneic) seem to be the most crucial factors impacting the in-vivo efficacy of MSCs. SUMMARY A better understanding of the mechanisms underlying the immunomodulatory effects of MSCs in vitro and in vivo is needed to define the optimal condition for the use of MSCs as immunotherapy in solid organ transplantation.
Collapse
|
44
|
Lee SM, Lee SC, Kim SJ. Contribution of human adipose tissue-derived stem cells and the secretome to the skin allograft survival in mice. J Surg Res 2014; 188:280-9. [PMID: 24560349 DOI: 10.1016/j.jss.2013.10.063] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/16/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite considerable evidence showing the immunosuppressive properties of mesenchymal stem cells (MSCs) in vitro, such properties have not been fully demonstrated in vivo. The aim of this study was to evaluate the effect of MSCs and/or MSC secretome in inducing tolerance in a mouse skin transplantation model. METHODS After receiving full-thickness skin allotransplantation on the back of the mouse, the recipient mice were infused with phosphate-buffered saline, adipose tissue-derived stem cells (ASCs), conditioned media (CM), and control media. Specifically, ASCs (1.0 × 10(6)/0.1 mL) were transplanted to ASC-infused mice and 25-fold concentrated CM, which had been obtained from ASC culture were infused to CM-infused mice. Graft survival rates and the parameters reflecting immunologic consequences were assessed. RESULTS The serum level of proinflammatory cytokine interleukin 6 decreased in mice treated with ASCs or CM compared with the control groups after infusion (P < 0.05). Interferon gamma, interleukin 10, and tumor necrosis factor alpha messenger RNA levels in the skin graft seemed to be decreased in the ASC-infused mice and CM-infused mice. Hyporesponsiveness was identified in mixed lymphocyte reaction assay at 30-d posttransplantation in ASC- or CM-infused mice. And, administering ASCs and CM markedly increased skin allograft survival compared with control animals (P < 0.001). CONCLUSIONS These findings suggest that ASCs and their secretome have the potential to induce immunologic tolerance. Moreover, our results demonstrate that the immunosuppressive properties of ASCs are mediated by the ASC secretome. Our approach could provide insights into a promising strategy to avoid toxicities of chemical immunosuppressive regimen in solid organ transplantation.
Collapse
Affiliation(s)
- Sang Mook Lee
- Department of Anesthesiology, Daejeon St. Mary's Hospital, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Sang Chul Lee
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Say-June Kim
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea, Daejeon, Republic of Korea.
| |
Collapse
|
45
|
Lee SC, Sul H, Lee SM, Kim SJ. Effects of Human Adipose-Tissue Derived Stem Cell Infusion on the Immunological Consequences in Skin Allograft Mice. KOREAN JOURNAL OF TRANSPLANTATION 2013. [DOI: 10.4285/jkstn.2013.27.4.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Sang Chul Lee
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Daejeon, Korea
| | - Haejoung Sul
- Department of Pathology, Daejeon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Daejeon, Korea
| | - Sang-Mook Lee
- Department of Anesthesiology, Daejeon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Daejeon, Korea
| | - Say-June Kim
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Daejeon, Korea
| |
Collapse
|
46
|
Syngeneic adipose-derived stem cells with short-term immunosuppression induce vascularized composite allotransplantation tolerance in rats. Cytotherapy 2013; 16:369-80. [PMID: 24119648 DOI: 10.1016/j.jcyt.2013.06.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/23/2013] [Accepted: 06/27/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND AIMS A clinically applicable tolerance induction regimen that removes the requirement for lifelong immunosuppression would benefit recipients of vascularized composite allotransplantation (VCA). We characterized the immunomodulatory properties of syngeneic (derived from the recipient strain) adipocyte-derived stem cells (ADSCs) and investigated their potential to induce VCA tolerance in rats. METHODS ADSCs were isolated from Lewis (LEW, RT1A(l)) rats; their immunomodulatory properties were evaluated by means of mixed lymphocyte reactions in vitro and VCAs in vivo across a full major histocompatibility complex mismatch with the use of Brown-Norway (BN, RT1A(n)) donor rats. Two control and four experimental groups were designed to evaluate treatment effects of ADSCs and transient immunosuppressants (anti-lymphocyte globulin, cyclosporine) with or without low-dose (200 cGy) total body irradiation. Flow cytometry was performed to quantify levels of circulating CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs). RESULTS Cultured syngeneic ADSCs exhibited CD90.1(+)CD29(+)CD73(+)CD45(-)CD79a(-)CD11b/c(-) phenotype and the plasticity to differentiate to adipocytes and osteocytes. ADSCs dramatically suppressed proliferation of LEW splenocytes against BN antigen and mitogen, respectively, in a dose-dependent fashion, culminating in abrogation of allo- and mitogen-stimulated proliferation at the highest concentration tested. Accordingly, one infusion of syngeneic ADSCs markedly prolonged VCA survival in LEW recipients treated with transient immunosuppression; of these, 66% developed tolerance. Total body irradiation provided no additional VCA survival benefit. An important role for Tregs in tolerance induction/maintenance was suggested in vivo and in vitro. CONCLUSIONS Treatment comprising syngeneic ADSCs and transient immunosuppression (i) increased levels of circulating Tregs and (ii) induced tolerance in 66% of recipients of major histocompatibility complex-mismatched VCAs.
Collapse
|
47
|
Calloni R, Viegas GS, Türck P, Bonatto D, Pegas Henriques JA. Mesenchymal stromal cells from unconventional model organisms. Cytotherapy 2013; 16:3-16. [PMID: 24113426 DOI: 10.1016/j.jcyt.2013.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 07/22/2013] [Accepted: 07/23/2013] [Indexed: 12/23/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent, plastic, adherent cells able to differentiate into osteoblasts, chondroblasts and adipocytes. MSCs can be isolated from many different body compartments of adult and fetal individuals. The most commonly studied MSCs are isolated from humans, mice and rats. However, studies are also being conducted with the use of MSCs that originate from different model organisms, such as cats, dogs, guinea pigs, ducks, chickens, buffalo, cattle, sheep, goats, horses, rabbits and pigs. MSCs derived from unconventional model organisms all present classic fibroblast-like morphology, the expression of MSC-associated cell surface markers such as CD44, CD73, CD90 and CD105 and the absence of CD34 and CD45. Moreover, these MSCs have the ability to differentiate into osteoblasts, chondroblasts and adipocytes. The MSCs isolated from unconventional model organisms are being studied for their potential to heal different tissue defects and injuries and for the development of scaffold compositions that improve the proliferation and differentiation of MSCs for tissue engineering.
Collapse
Affiliation(s)
- Raquel Calloni
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Gabrihel Stumpf Viegas
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Patrick Türck
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Diego Bonatto
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil.
| | - João Antonio Pegas Henriques
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| |
Collapse
|
48
|
Barbeau DJ, La KT, Kim DS, Kerpedjieva SS, Shurin GV, Tamama K. Early growth response-2 signaling mediates immunomodulatory effects of human multipotential stromal cells. Stem Cells Dev 2013; 23:155-66. [PMID: 24007274 DOI: 10.1089/scd.2013.0194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
While most studies have suggested multipotential stromal cell or mesenchymal stem cell (MSC) therapies are useful for immune-mediated diseases, MSCs' immunomodulatory effects were not entirely reproduced in some studies, indicating the necessity to determine the underlying mechanism of MSCs' effects on immune response regulation to maximize their immunomodulatory effects. We have identified the transcription factor early growth response gene-2 (EGR2) as a novel molecular switch regulating known immunomodulatory molecules in human MSCs. EGR2 binds to the promoter regions of these genes, interleukin-6 (IL6), leukemia inhibitory factor (LIF), indoleamine dioxygenase-1 (IDO1), and cyclooxygenase-2/prostaglandin-endoperoxide synthase 2 (COX2/PTGS2), and siRNA against EGR2 was shown to downregulate these genes and reduce the production of prostaglandin E2, an immunomodulatory mediator produced downstream of COX2/PTGS2. Moreover, EGR2 knockdown restores T-lymphocyte proliferation reduced by MSC coculture. Therefore, EGR2 is a potential target for the optimization of immunomodulatory properties of MSC-based therapies.
Collapse
Affiliation(s)
- Dominique J Barbeau
- 1 Department of Pathology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | | | | | | | | | | |
Collapse
|
49
|
Lee JH, Jeon EJ, Kim N, Nam YS, Im KI, Lim JY, Kim EJ, Cho ML, Han KT, Cho SG. The synergistic immunoregulatory effects of culture-expanded mesenchymal stromal cells and CD4(+)25(+)Foxp3+ regulatory T cells on skin allograft rejection. PLoS One 2013; 8:e70968. [PMID: 23940676 PMCID: PMC3733648 DOI: 10.1371/journal.pone.0070968] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 06/26/2013] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are seen as an ideal source of cells to induce graft acceptance; however, some reports have shown that MSCs can be immunogenic rather than immunosuppressive. We speculate that the immunomodulatory effects of regulatory T cells (Tregs) can aid the maintenance of immunoregulatory functions of MSCs, and that a combinatorial approach to cell therapy can have synergistic immunomodulatory effects on allograft rejection. After preconditioning with Fludarabine, followed by total body irradiation and anti-asialo-GM-1(ASGM-1), tail skin grafts from C57BL/6 (H-2kb) mice were grafted onto the lateral thoracic wall of BALB/c (H-2kd) mice. Group A mice (control group, n = 9) did not receive any further treatment after preconditioning, whereas groups B and C (n = 9) received cell therapy with MSCs or Tregs, respectively, on days −1, +6 and +13 relative to the skin transplantation. Group D (n = 10) received cell therapy with MSCs and Tregs on days −1, +6 and +13. Cell suspensions were obtained from the spleens of five randomly chosen mice from each group on day +7, and the immunomodulatory effects of the cell therapy were evaluated by flow cytometry and real-time PCR. Our results show that allograft survival was significantly longer in group D compared to the control group (group A). Flow cytometric analysis and real-time PCR for splenocytes revealed that the Th2 subpopulation in group D increased significantly compared to the group B. Also, the expression of Foxp3 and STAT 5 increased significantly in group D compared to the conventional cell therapy groups (B and C). Taken together, these data suggest that a combined cell therapy approach with MSCs and Tregs has a synergistic effect on immunoregulatory function in vivo, and might provide a novel strategy for improving survival in allograft transplantation.
Collapse
Affiliation(s)
- Jung Ho Lee
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Eun-Joo Jeon
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Nayoun Kim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Young-Sun Nam
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Keon-Il Im
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jung-Yeon Lim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Eun-Jung Kim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Institutes of Medical Science, Seoul, Korea
| | - Ki Taik Han
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Seok-Goo Cho
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
- Catholic Blood and Marrow Transplantation Center, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
50
|
Plock JA, Schnider JT, Solari MG, Zheng XX, Gorantla VS. Perspectives on the use of mesenchymal stem cells in vascularized composite allotransplantation. Front Immunol 2013; 4:175. [PMID: 23888159 PMCID: PMC3719134 DOI: 10.3389/fimmu.2013.00175] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 06/18/2013] [Indexed: 12/13/2022] Open
Abstract
Reconstructive transplantation has emerged as clinical reality over the past decade. Long-term graft acceptance has been feasible in extremity and facial vascularized composite allotransplantation (VCA) under standard immunosuppression. Minimizing overall burden of lifelong immunosuppression is key to wider application of these non-life saving grafts. Allograft tolerance is the holy grail of many cell-based immunomodulatory strategies. Recent protocols using mesenchymal stem cells from bone marrow and adipose tissue offer promise and potential in VCA. This article provides an overview of the experimental basis, the scientific background and clinical applications of stem cell-based therapies in the field of reconstructive allotransplantation.
Collapse
Affiliation(s)
- Jan A Plock
- Department of Plastic Surgery, University of Pittsburgh Medical Center , Pittsburgh, PA , USA ; Division of Plastic and Hand Surgery, University Hospital Zurich , Zurich , Switzerland
| | | | | | | | | |
Collapse
|