1
|
Mönch D, Reinders MEJ, Dahlke MH, Hoogduijn MJ. How to Make Sense out of 75,000 Mesenchymal Stromal Cell Publications? Cells 2022; 11:cells11091419. [PMID: 35563725 PMCID: PMC9101744 DOI: 10.3390/cells11091419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stromal cells have been the subject of an expanding number of studies over the past decades. Today, over 75,000 publications are available that shine light on the biological properties and therapeutic effects of these versatile cells in numerous pre-clinical models and early-phase clinical trials. The massive number of papers makes it hard for researchers to comprehend the whole field, and furthermore, they give the impression that mesenchymal stromal cells are wonder cells that are curative for any condition. It is becoming increasingly difficult to dissect how and for what conditions mesenchymal stromal cells exhibit true and reproducible therapeutic effects. This article tries to address the question how to make sense of 75,000, and still counting, publications on mesenchymal stromal cells.
Collapse
Affiliation(s)
- Dina Mönch
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany;
- University of Tübingen, 72074 Tübingen, Germany
| | - Marlies E. J. Reinders
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Marc H. Dahlke
- Department of Surgery, Robert-Bosch-Hospital, 70376 Stuttgart, Germany;
| | - Martin J. Hoogduijn
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
2
|
Chen HS, Yau YC, Ko PT, Yen BLJ, Ho CT, Hung SC. Mesenchymal Stem Cells From a Hypoxic Culture Can Improve Rotator Cuff Tear Repair. Cell Transplant 2022; 31:9636897221089633. [PMID: 35438571 PMCID: PMC9021471 DOI: 10.1177/09636897221089633] [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: 11/15/2022] Open
Abstract
A rotator cuff tear is an age-related common cause of pain and disability. Studies including our previously published ones have demonstrated that mesenchymal stem cells cultured under hypoxic conditions [hypoxic multipotent stromal cells (MSCs)] facilitate the retention of transplanted cells and promote wound healing. However, there are very few, if any, reports targeting the punctured supraspinatus tendons to create more or equally serous wounds as age-related tears of rotator cuff. It remains to be determined whether transplantation of bone-marrow-derived hypoxic MSCs into the punctured supraspinatus tendon improves tendon repair and, when combined with ultrasound-guided delivery, could be used for future clinical applications. In this study, we used a total of 33 Sprague-Dawley rats in different groups for normal no-punched control, hypoxic MSC treatment, nontreated vehicle control, and MSC preparation, and then evaluated treatment outcomes by biomechanical testing and histological analysis. We found that the ultimate failure load of the hypoxic MSC-treated group was close to that of the normal tendon and significantly greater than that of the nontreated vehicle control group. In vivo tracking of cells labeled with superparamagnetic iron oxide (SPIO) nanoparticles revealed an enhanced retention of transplanted cells at the tear site. Our study demonstrates that hypoxic MSCs improve rotator cuff tear repair in a rat model.
Collapse
Affiliation(s)
- Hsin-Shui Chen
- PhD Program for Aging, College of Medicine, China Medical University, Taichung.,Department of Physical Medicine & Rehabilitation, National Taiwan University Hospital Yunlin Branch, Yunlin
| | - Yun-Chain Yau
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei
| | - Pin-Tsou Ko
- Department of Pathology, An-Nan Hospital, China Medical University, Tainan
| | - Betty Lin-Ju Yen
- Institute of Cellular & System Medicine, Regenerative Medicine Research Group, National Health Research Institutes, Zhunan
| | - Chun-Te Ho
- Institute of New Drug Development, China Medical University, Taichung.,Integrative Stem Cell Center, China Medical University Hospital, Taichung
| | - Shih-Chieh Hung
- Institute of New Drug Development, China Medical University, Taichung.,Integrative Stem Cell Center, China Medical University Hospital, Taichung
| |
Collapse
|
3
|
Zhang YL, Qiao SK, Xing LN, Guo XN, Ren JH. Mesenchymal Stem Cells Enhance Chemotaxis of Activated T Cells through the CCL2-CCR2 Axis In Vitro. Bull Exp Biol Med 2021; 172:263-269. [PMID: 34855085 DOI: 10.1007/s10517-021-05373-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Indexed: 12/30/2022]
Abstract
Activation and migration of donor T cells to the host target organs are critical mechanisms in the pathogenesis of graft-versus-host disease (GVHD). The role of monocyte chemoattractant protein-1 (MCP-1/CCL2) and its receptor CCR2 in the recruitment of T cells during immune or inflammatory response is also well known. For elucidation of the mechanism of the therapeutic effect of human bone marrow derived-mesenchymal stem cells (MSC) in GVHD, we studied the effect of these cells on migration of activated donor T cells through the CCL2-CCR2 axis in vitro. MSC were expanded from donors' bone marrow mononuclear cells. After co-culturing of IL-2-activated T cells with allogeneic MSC at different ratios, the levels of CCL2 in supernatants were measured by ELISA, and CCR2 expression in CD4+/CD8+ T cells subsets were detected by flow cytometry. The effect of MSC on the migration of activated T cells in the Transwell system was studied in the absence or presence of CCL2. Our results show that CCL2 levels in supernatants of co-cultures were significantly higher than in MSC monoculture and this increase depended on the number of MSC. MSC inhibited proliferation of T cells, but did not change the percentages of CD4+ and CD8+ T cells subsets. MSC can up-regulate the CCR2 expression in CD8+ subsets rather than in CD4+ subsets; MSC enhanced migration of IL-2-activated T cells to CCL2 by increasing the expression of CCR2. The data demonstrate that MSC can enhance chemotaxis of cytokine-activated T cells through the CCL2-CCR2 axis in vitro.
Collapse
Affiliation(s)
- Y L Zhang
- Department of General Practice, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - S K Qiao
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - L N Xing
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - X N Guo
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - J H Ren
- Department of Hematology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
4
|
Saeedi P, Halabian R, Imani Fooladi AA. A revealing review of mesenchymal stem cells therapy, clinical perspectives and Modification strategies. Stem Cell Investig 2019; 6:34. [PMID: 31620481 DOI: 10.21037/sci.2019.08.11] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022]
Abstract
Multipotent mesenchymal stem cells (MSCs) have been considerably inspected as effective tool for cell-based therapy of inflammatory, immune-mediated, and degenerative diseases, attributed to their immunomodulatory, immunosuppressive, and regenerative potentials. In the present review, we focus on recent research findings of the clinical applications and therapeutic potential of this cell type, MSCs' mechanisms of therapy, strategies to improve their therapeutic potentials such as manipulations and preconditioning, and potential/unexpected risks which should be considered as a prerequisite step before clinical use. The potential risks would probably include undesirable immune responses, tumor formation and the transmission of incidental agents. Then, we also review some of the milestones in the field, briefly discuss challenges and highlight the new guideline suggested for future directions and perspectives.
Collapse
Affiliation(s)
- Pardis Saeedi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Zhang P, Zhang G, Liu X, Liu H, Yang P, Ma L. Mesenchymal stem cells improve platelet counts in mice with immune thrombocytopenia. J Cell Biochem 2019; 120:11274-11283. [PMID: 30775797 DOI: 10.1002/jcb.28405] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/27/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Immune thrombocytopenia (ITP) is a common autoimmune bleeding disorder. The breakdown of immune tolerance (regulatory T [Treg] cells and suppressor cytokines) plays an important role in ITP pathophysiology, especially in refractory ITP. Bone marrow-derived mesenchymal stem cells (BM-MSCs) show immunomodulatory properties and have been extensively utilized for autoimmune diseases. However, it has not been fully elucidated how BM-MSCs affect ITP. In this study, we explore the therapeutic mechanism of BM-MSCs on ITP in mice. Dose-escalation passive ITP mice were inducted by injection of MWReg30. BALB/c mice were randomly divided into two groups: ITP with BM-MSC transplantation and ITP controls. The serum levels of cytokines (interleukin 10 [IL-10] and transforming growth factor-β1 [TGF-β1]) were examined by enzyme-linked immunosorbent assays. The frequency of Treg cells in both peripheral blood and spleen mononuclear cells was analyzed by flow cytometry, and the forkhead box P3 (Foxp3) messenger RNA (mRNA) level was measured by real-time polymerase chain reaction. After BM-MSC treatment, the platelet (PLT) counts were significantly elevated. Meanwhile, cytokines (TGF-β1 and IL-10), the ratios of Treg cells, and the Foxp3 mRNA expression level were significantly higher in the BM-MSC group. Our results show that BM-MSCs can improve PLT counts mainly by secreting suppressive cytokines and upregulating Tregs, which may provide new therapeutic potential for human ITP.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Hematology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guoyang Zhang
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Liu
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongyun Liu
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pengfeng Yang
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liping Ma
- Department of Hematology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
6
|
Kim SH, Jung J, Cho KJ, Choi JH, Lee HS, Kim GJ, Lee SG. Immunomodulatory Effects of Placenta-derived Mesenchymal Stem Cells on T Cells by Regulation of FoxP3 Expression. Int J Stem Cells 2018; 11:196-204. [PMID: 30343549 PMCID: PMC6285290 DOI: 10.15283/ijsc18031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 12/17/2022] Open
Abstract
The immunomodulatory effects of mesenchymal stem cells (MSCs) are an important mediator of their therapeutic effects in stem cell therapy and regenerative medicine. The regulation mechanism of MSCs is orchestrated by several factors in both intrinsic and extrinsic events. Recent studies have shown that the dynamic expression of cytokines secreted from MSCs control T cell function and maturation by regulating the expression of FoxP3, which figures prominently in T cell differentiation. However, there is no evidence that placenta-derived mesenchymal stem cells (PD-MSCs) have strong immunomodulatory effects on T cell function and maturation via FoxP3 expression. Therefore, we compared the expression of FoxP3 in activated T cells isolated from peripheral blood and co-cultured with PD-MSCs or bone marrow-derived mesenchymal stem cells (BM-MSCs) and analyzed their effect on T cell proliferation and cytokine profiles. Additionally, we verified the immunomodulatory function of PD-MSCs by siRNA-mediated silencing of FoxP3. MSCs, including PD-MSCs and BM-MSCs, promoted differentiation of naive peripheral blood T cells into CD4+CD25+FoxP3+ regulatory T (Treg) cells. Intriguingly, the population of CD4+CD25+FoxP3+ Treg cells co-cultured with PD-MSCs was significantly expanded in comparison to those co-cultured with BM-MSCs or WI38 cells (p<0.05, p<0.001). Dynamic expression patterns of several cytokines, including anti- and pro-inflammatory cytokines and members of the transforming growth factor-beta (TGF-β) family secreted from PD-MSCs according to FoxP3 expression were observed. The results suggest that PD-MSCs have an immunomodulatory effect on T cells by regulating FoxP3 expression.
Collapse
Affiliation(s)
- Soo-Hwan Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul,
Korea
- Department of Biomedical Laboratory Science, Gimcheon University, Gimcheon,
Korea
| | - Jieun Jung
- Placenta Research Laboratory, Department of Biomedical Science, CHA University, Seongnam,
Korea
| | - Kyung Jin Cho
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul,
Korea
- Faculty of Health and Environmental Science, College of Health Science, Korea University, Seoul,
Korea
| | - Jong-Ho Choi
- Placenta Research Laboratory, Department of Biomedical Science, CHA University, Seongnam,
Korea
| | - Hyeong Seon Lee
- Department of Biomedical Laboratory Science, Jungwon University, Goesan,
Korea
| | - Gi Jin Kim
- Placenta Research Laboratory, Department of Biomedical Science, CHA University, Seongnam,
Korea
| | - Seung Gwan Lee
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul,
Korea
- Faculty of Health and Environmental Science, College of Health Science, Korea University, Seoul,
Korea
| |
Collapse
|
7
|
Abstract
The goal of this chapter is to provide an overview of the different purposes for which the cell microencapsulation technology can be used. These include immunoisolation of non-autologous cells used for cell therapy; immobilization of cells for localized (targeted) delivery of therapeutic products to ablate, repair, or regenerate tissue; simultaneous delivery of multiple therapeutic agents in cell therapy; spatial compartmentalization of cells in complex tissue engineering; expansion of cells in culture; and production of different probiotics and metabolites for industrial applications. For each of these applications, specific examples are provided to illustrate how the microencapsulation technology can be utilized to achieve the purpose. However, successful use of the cell microencapsulation technology for whatever purpose will ultimately depend upon careful consideration for the choice of the encapsulating polymers, the method of fabrication (cross-linking) of the microbeads, which affects the permselectivity, the biocompatibility and the mechanical strength of the microbeads as well as environmental parameters such as temperature, humidity, osmotic pressure, and storage solutions.The various applications discussed in this chapter are illustrated in the different chapters of this book and where appropriate relevant images of the microencapsulation products are provided. It is hoped that this outline of the different applications of cell microencapsulation would provide a good platform for tissue engineers, scientists, and clinicians to design novel tissue constructs and products for therapeutic and industrial applications.
Collapse
Affiliation(s)
- Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA. .,Virginia Tech-Wake Forest School of Biomedical Engineering & Sciences (SBES), Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
8
|
Boieri M, Shah P, Dressel R, Inngjerdingen M. The Role of Animal Models in the Study of Hematopoietic Stem Cell Transplantation and GvHD: A Historical Overview. Front Immunol 2016; 7:333. [PMID: 27625651 PMCID: PMC5003882 DOI: 10.3389/fimmu.2016.00333] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow transplantation (BMT) is the only therapeutic option for many hematological malignancies, but its applicability is limited by life-threatening complications, such as graft-versus-host disease (GvHD). The last decades have seen great advances in the understanding of BMT and its related complications; in particular GvHD. Animal models are beneficial to study complex diseases, as they allow dissecting the contribution of single components in the development of the disease. Most of the current knowledge on the therapeutic mechanisms of BMT derives from studies in animal models. Parallel to BMT, the understanding of the pathophysiology of GvHD, as well as the development of new treatment regimens, has also been supported by studies in animal models. Pre-clinical experimentation is the basis for deep understanding and successful improvements of clinical applications. In this review, we retrace the history of BMT and GvHD by describing how the studies in animal models have paved the way to the many advances in the field. We also describe how animal models contributed to the understanding of GvHD pathophysiology and how they are fundamental for the discovery of new treatments.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen , Göttingen , Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
9
|
Darzi S, Werkmeister JA, Deane JA, Gargett CE. Identification and Characterization of Human Endometrial Mesenchymal Stem/Stromal Cells and Their Potential for Cellular Therapy. Stem Cells Transl Med 2016; 5:1127-32. [PMID: 27245365 DOI: 10.5966/sctm.2015-0190] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 03/23/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED SummaryHuman endometrium is a highly regenerative tissue, undergoing more than 400 cycles of proliferation, differentiation, and shedding during a woman's reproductive life. Adult stem cells, including mesenchymal stem/stromal cells (MSCs), are likely responsible for the immense cellular turnover in human endometrium. The unique properties of MSCs, including high proliferative ability, self-renewal, differentiation to mesodermal lineages, secretion of angiogenic factors, and many other growth-promoting factors make them useful candidates for cellular therapy and tissue engineering. In this review, we summarize the identification and characterization of newly discovered MSCs from the human endometrium: their properties, the surface markers used for their prospective isolation, their perivascular location in the endometrium, and their potential application in cellular therapies. SIGNIFICANCE The endometrium, or the lining of uterus, has recently been identified as a new and accessible source of mesenchymal stem cells, which can be obtained without anesthesia. Endometrial mesenchymal stem cells have comparable properties to bone marrow and adipose tissue mesenchymal stem cells. Endometrial mesenchymal stem cells are purified with known and novel perivascular surface markers and are currently under investigation for their potential use in cellular therapy for several clinical conditions with significant burden of disease.
Collapse
Affiliation(s)
- Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Jerome A Werkmeister
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia Commonwealth Scientific and Industrial Research Organisation, Clayton, Victoria, Australia
| | - James A Deane
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
10
|
RUSU E, NECULA LG, NEAGU AI, ALECU M, STAN C, ALBULESCU R, TANASE CP. Current status of stem cell therapy: opportunities and limitations. Turk J Biol 2016; 40:955-967. [DOI: 10.3906/biy-1506-95] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
11
|
Vertès AA. The potential of cytotherapeutics in hematologic reconstitution and in the treatment and prophylaxis of graft-versus-host disease. Chapter II: emerging transformational cytotherapies. Regen Med 2015; 10:345-73. [DOI: 10.2217/rme.15.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a life-saving treatment for inherited anemias, immunodeficiencies or hematologic malignancies. A major complication of allo-HSCT associated with high transplant-related mortality rates is graft-versus-host disease (GvHD). Current and future clinical benefits in HSCT enabled by advances in hematopoietic stem cells, mesenchymal stem cells, Tregs and natural killer cells technologies are reviewed here and discussed. Among these evolutions, based on the need for mesenchymal stem cells to be recruited by an inflammatory environment, the development and use of novel GvHD biomarkers could be explored further to deliver the right pharmaceutical to the right patient at the right time. The successful commercialization of cytotherapeutics to efficiently manage GvHD will create a virtuous ‘halo’ effect for regenerative medicine.
Collapse
Affiliation(s)
- Alain A Vertès
- Sloan Fellow, London Business School, London, UK
- NxR Biotechnologies GmbH, Basel, Switzerland
| |
Collapse
|
12
|
Peng Y, Chen X, Liu Q, Xu D, Zheng H, Liu L, Liu Q, Liu M, Fan Z, Sun J, Li X, Zou R, Xiang AP. Alteration of naïve and memory B-cell subset in chronic graft-versus-host disease patients after treatment with mesenchymal stromal cells. Stem Cells Transl Med 2014; 3:1023-31. [PMID: 25015640 DOI: 10.5966/sctm.2014-0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although mesenchymal stromal cells (MSCs) possess immunomodulatory properties and exhibit promising efficacy against chronic graft-versus-host disease (cGVHD), little is known about the immune changes by which MSCs ameliorate cGVHD in vivo. Recent studies have suggested that B lymphocytes might play an important role in the pathogenesis of cGVHD. In this study, we investigated changes in the numbers, phenotypes, and subpopulations of B lymphocytes in cGVHD patients who showed a complete response (CR), partial response (PR), or no response (NR) after MSC treatment. We found that the frequencies and numbers of CD27+ memory and pre-germinal center B lymphocytes were significantly increased in the CR and PR cGVHD patients after MSC treatment but decreased in the NR patients. A further analysis of CR/PR cGVHD patients showed that MSC treatment led to a decrease in the plasma levels of B cell-activating factor (BAFF) and increased expression of the BAFF receptor (BAFF-R) on peripheral B lymphocytes but no changes in plasma BAFF levels or BAFF-R expression on B lymphocytes in NR patients. Overall, our findings imply that MSCs might exert therapeutic effects in cGVHD patients, accompanied by alteration of naïve and memory B-cell subsets, modulating plasma BAFF levels and BAFF-R expression on B lymphocytes.
Collapse
Affiliation(s)
- Yanwen Peng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qifa Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dijing Xu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Haiqing Zheng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Longshan Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qiuli Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Muyun Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhiping Fan
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Sun
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaobo Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ruifeng Zou
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Medical Science Experimentation Center, and Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; Department of Rehabilitation Medicine Science and Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China; Organ Transplant Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
13
|
Wang X, Xi WC, Wang F. The beneficial effects of intracoronary autologous bone marrow stem cell transfer as an adjunct to percutaneous coronary intervention in patients with acute myocardial infarction. Biotechnol Lett 2014; 36:2163-8. [PMID: 24975729 DOI: 10.1007/s10529-014-1589-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/12/2014] [Indexed: 11/24/2022]
Abstract
The efficacy of post-percutaneous coronary intervention (PCI) intracoronary injection with bone marrow mesenchymal stem cells (BMSCs) in patients with acute myocardial infarction (AMI) remains controversial. Here, 58 patients with AMI undergoing PCI were randomly divided into two groups: BMSC and control groups. Autologous BSMCs were then generated in vitro from the BMSC patients. After transplantation, left ventricular ejection fraction (LVEF), left ventricular end-diastolic dimensions (LVDd), and infarct size (IS) were evaluated in both groups. LVEF, LVDd, and IS improved after BMSC transplantation but the changes were not significantly different from those in the controls. The number of adverse events and rehospitalization rates after 1 month were significantly higher in the control group than in the BMSC group. BMSC transplantation thus benefits patients by decreasing the number of adverse events and reducing the rehospitalization rate in the early stages following PCI.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Cardiology, Shanghai First People's Hospital, Affiliated Shanghai Jiaotong University, Shanghai, 200080, China
| | | | | |
Collapse
|
14
|
Choi J, Hwang MP, Lee JW, Lee KH. A glimpse into the interactions of cells in a microenvironment: the modulation of T cells by mesenchymal stem cells. Int J Nanomedicine 2014; 9 Suppl 1:127-39. [PMID: 24872708 PMCID: PMC4024981 DOI: 10.2147/ijn.s50767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been thought to hold potential as a mode of therapy for immuno-related pathologies, particularly for autoimmune diseases. Despite their potential, the interaction between MSCs and T cells, key players in the pathophysiology of autoimmune diseases, is not yet well understood, thereby preventing further clinical progress. A major obstacle is the highly heterogeneous nature of MSCs in vitro. Unfortunately, bulk assays do not provide information with regard to cell-cell contributions that may play a critical role in the overall cellular response. To address these issues, we investigated the interaction between smaller subsets of MSCs and CD4 T cells in a microwell array. We demonstrate that MSCs appear capable of modulating the T cell proliferation rate in response to persistent cell-cell interactions, and we anticipate the use of our microwell array in the classification of subpopulations within MSCs, ultimately leading to specific therapeutic interventions.
Collapse
Affiliation(s)
- Jonghoon Choi
- Institute of Research Strategy and Development (IRSD), Seoul, Republic of Korea ; Department of Bionano Engineering, Hanyang University, Ansan, Republic of Korea
| | - Mintai P Hwang
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jong-Wook Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kwan Hyi Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea ; Department of Biomedical Engineering, University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
15
|
Optimization of the therapeutic efficacy of human umbilical cord blood-mesenchymal stromal cells in an NSG mouse xenograft model of graft-versus-host disease. Cytotherapy 2014; 16:298-308. [PMID: 24418403 DOI: 10.1016/j.jcyt.2013.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/28/2013] [Accepted: 10/22/2013] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS Although in vitro studies have demonstrated the immunosuppressive capacity of mesenchymal stromal cells (MSCs), most in vivo studies on graft-versus-host disease (GVHD) have focused on prevention, and the therapeutic effect of MSCs is controversial. Moreover, optimal time intervals for infusing MSCs have not been established. METHODS We attempted to evaluate whether human umbilical cord blood-MSCs (hUCB-MSCs) could either prevent or treat GVHD in an NSG mouse xenograft model by injection of MSCs before or after in vivo clearance. Mice were infused with either a single dose or multiple doses of 5 × 10(5) hUCB-MSCs (3- or 7-day intervals) before or after GVHD onset. RESULTS Before onset, hUCB-MSCs significantly improved the survival rate only when repeatedly injected at 3-day intervals. In contrast, single or repeated injections after GVHD onset significantly increased the survival rate and effectively attenuated tissue damage and inflammation. Furthermore, the levels of prostaglandin E2 and transforming growth factor-β1 increased significantly, whereas the level of interferon-γ decreased significantly in all MSC treatment groups. CONCLUSIONS These data establish the optimal time intervals for preventing GVHD and show that hUCB-MSCs effectively attenuated symptoms and improved survival rate when administered after the onset of GVDH.
Collapse
|
16
|
Kudo K, Imai C, Lorenzini P, Kamiya T, Kono K, Davidoff AM, Chng WJ, Campana D. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res 2013; 74:93-103. [PMID: 24197131 DOI: 10.1158/0008-5472.can-13-1365] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To expand applications for T-cell-based immunotherapy in cancer, we designed a receptor that binds the Fc portion of human immunoglobulins and delivers activation signals. The construct included the high-affinity CD16 (FCGR3A) V158 variant, CD8α hinge, and transmembrane domains, along with signaling domains from CD3ζ and 4-1BB (TNFRSF9), forming a chimeric receptor termed CD16V-BB-ζ. After retrovirus-mediated expression in human T cells, CD16V-BB-ζ bound humanized antibodies with higher affinity than a control receptor containing the more common F158 variant. Engagement of CD16V-BB-ζ provoked T-cell activation, exocytosis of lytic granules, and sustained proliferation, with a mean cell recovery after 4-week coculture with Daudi lymphoma cells and rituximab of nearly 70-fold relative to input cells. In contrast, unbound antibody alone produced no effect. CD16V-BB-ζ T cells specifically killed lymphoma cells and primary chronic lymphocytic leukemia cells in combination with rituximab at a low effector:target ratio, even when assayed on mesenchymal cells. Trastuzumab triggered CD16V-BB-ζ-mediated killing of HER2 (ERBB2)(+) breast and gastric cancer cells; similar results were obtained with an anti-GD2 antibody in neuroblastoma and osteosarcoma cells. Furthermore, coadministration of CD16V-BB-ζ T cells with immunotherapeutic antibodies exerted considerable antitumor activity in vivo. Signaling mediated by 4-1BB-CD3ζ induced higher T-cell activation, proliferation, and cytotoxicity than CD3ζ or FcεRIγ, and the receptor was expressed effectively after mRNA electroporation without viral vectors, facilitating clinical translation. Our results offer preclinical proof of concept for CD16V-BB-ζ as a universal, next-generation chimeric receptor with the potential to augment the efficacy of antibody therapies for cancer.
Collapse
Affiliation(s)
- Ko Kudo
- Authors' Affiliations: Departments of Pediatrics and Surgery, and Cancer Science Institute, National University of Singapore, Singapore; Department of Pediatrics, Niigata University, Niigata, Japan; and Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Pritz T, Landgraf-Rauf K, Herndler-Brandstetter D, Rauf R, Lair J, Gassner R, Weinberger B, Krismer M, Grubeck-Loebenstein B. Bone marrow T cells from the femur are similar to iliac crest derived cells in old age and represent a useful tool for studying the aged immune system. IMMUNITY & AGEING 2013; 10:17. [PMID: 23642143 PMCID: PMC3648417 DOI: 10.1186/1742-4933-10-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/25/2013] [Indexed: 11/10/2022]
Abstract
Background CD4+ and CD8+ T cells reside in the human bone marrow (BM) and show a heightened activation state. However, only small sample sizes are available from sources such as the iliac crest. Larger samples can be obtained from the femur in the course of hip replacement surgery. It was therefore the goal of the present study to compare the phenotype and function of BM T cells from different sources from elderly persons and to investigate how femur derived bone marrow T cells can serve as a tool to gain a better understanding of the role of adaptive immune cells in the BM in old age. Results Bone marrow mononuclear cells (BMMC) were isolated from either the iliac crest or the femur shaft. As expected the yield of mononuclear cells was higher from femur than from iliac crest samples. There were no phenotypic differences between BMMC from the two sources. Compared to PBMC, both BM sample types contained fewer naïve and more antigen experienced CD4+ as well as CD8+ T cells, which, in contrast to peripheral cells, expressed CD69. Cytokine production was also similar in T cells from both BM types. Larger sample sizes allowed the generation of T cell lines from femur derived bone marrow using non-specific as well as specific stimulation. The phenotype of T cell lines generated by stimulation with OKT-3 and IL-2 for two weeks was very similar to the one of ex vivo BM derived T cells. Such lines can be used for studies on the interaction of different types of BM cells as shown by co-culture experiments with BM derived stromal cells. Using CMVNLV specific T cell lines we additionally demonstrated that BM samples from the femur are suitable for the generation of antigen specific T cell lines, which can be used in studies on the clonal composition of antigen specific BM T cells. Conclusion In conclusion, our results demonstrate that BMMC from the femur shaft are a useful tool for studies on the role of T cells in the BM in old age.
Collapse
Affiliation(s)
- Theresa Pritz
- Institute for Biomedical Aging Research, University Innsbruck, Rennweg 10, Innsbruck, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Huang TF, Yew TL, Chiang ER, Ma HL, Hsu CY, Hsu SH, Hsu YT, Hung SC. Mesenchymal stem cells from a hypoxic culture improve and engraft Achilles tendon repair. Am J Sports Med 2013; 41:1117-25. [PMID: 23539044 DOI: 10.1177/0363546513480786] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Bone marrow-derived mesenchymal stem cells (MSCs) from humans cultured under hypoxic conditions increase bone healing capacity. HYPOTHESIS Rat MSCs cultured under hypoxic conditions increase the tendon healing potential after transplantation into injured Achilles tendons. STUDY DESIGN Controlled laboratory study. METHODS Biomechanical testing, histological analysis, and bromodeoxyuridine (BrdU) labeling/collagen immunohistochemistry were performed to demonstrate that augmentation of an Achilles tendon rupture site with hypoxic MSCs increases healing capacity compared with normoxic MSCs and controls. Fifty Sprague-Dawley rats were used for the experiments, with 2 rats as the source of bone marrow MSCs. The cut Achilles tendons in the rats were equally divided into 3 groups: hypoxic MSC, normoxic MSC, and nontreated (vehicle control). The uncut tendons served as normal uncut controls. Outcome measures included mechanical testing in 24 rats, histological analysis, and BrdU labeling/collagen immunohistochemistry in another 24 rats. RESULTS The ultimate failure load in the hypoxic MSC group was significantly greater than that in the nontreated or normoxic MSC group at 2 weeks after incision (2.1 N/mm(2) vs 1.1 N/mm(2) or 1.9 N/mm(2), respectively) and at 4 weeks after incision (5.5 N/mm(2) vs 1.7 N/mm(2) or 2.7 N/mm(2), respectively). The ultimate failure load in the hypoxic MSC group at 4 weeks after incision (5.5 N/mm(2)) was close to but still significantly less than that of the uncut tendon (7.2 N/mm(2)). Histological analysis as determined by the semiquantitative Bonar histopathological grading scale revealed that the hypoxic MSC group underwent a significant improvement in Achilles tendon healing both at 2 and 4 weeks when compared with the nontreated or normoxic MSC group via statistical analysis. Immunohistochemistry further demonstrated that the hypoxic and normoxic MSC groups had stronger immunostaining for type I and type III collagen than did the nontreated group both at 2 and 4 weeks after incision. Moreover, BrdU labeling of MSCs before injection further determined the incorporation and retention of transplanted cells at the rupture site. CONCLUSION Transplantation of hypoxic MSCs may be a better and more readily available treatment than normoxic MSCs for Achilles tendon ruptures. CLINICAL RELEVANCE The present study provides evidence that transplantation of hypoxic MSCs may be a promising therapy for the treatment of Achilles tendon ruptures.
Collapse
Affiliation(s)
- Tung-Fu Huang
- Department of Surgery, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Characterization of the conditioned medium from amniotic membrane cells: prostaglandins as key effectors of its immunomodulatory activity. PLoS One 2012; 7:e46956. [PMID: 23071674 PMCID: PMC3468614 DOI: 10.1371/journal.pone.0046956] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 09/06/2012] [Indexed: 12/11/2022] Open
Abstract
We previously demonstrated that cells isolated from the mesenchymal region of the human amniotic membrane (human amniotic mesenchymal tissue cells, hAMTC) possess immunoregulatory roles, such as inhibition of lymphocyte proliferation and cytokine production, and suppression of generation and maturation of monocyte-derived dendritic cells, as reported for MSC from other sources. The precise factors and mechanisms responsible for the immunoregulatory roles of hAMTC remain unknown. In this study, we aimed to identify the soluble factors released by hAMTC and responsible for the anti-proliferative effect on lymphocytes, and the mechanisms underlying their actions, in vitro. Conditioned medium (CM) was prepared under routine culture conditions from hAMTC (CM-hAMTC) and also from fragments of the whole human amniotic membrane (CM-hAM). We analyzed the thermostability, chemical nature, and the molecular weight of the factors likely responsible for the anti-proliferative effects. We also evaluated the participation of cytokines known to be involved in the immunomodulatory actions of MSC from other sources, and attempted to block different synthetic pathways. We demonstrate that the inhibitory factors are temperature-stable, have a small molecular weight, and are likely of a non-proteinaceous nature. Only inhibition of cyclooxygenase pathway partially reverted the anti-proliferative effect, suggesting prostaglandins as key effector molecules. Factors previously documented to take part in the inhibitory effects of MSCs from other sources (HGF, TGF-β, NO and IDO) were not involved. Furthermore, we prove for the first time that the anti-proliferative effect is intrinsic to the amniotic membrane and cells derived thereof, since it is manifested in the absence of stimulating culture conditions, as opposed to MSC derived from the bone marrow, which possess an anti-proliferative ability only when cultured in the presence of activating stimuli. Finally, we show that the amniotic membrane could be an interesting source of soluble factors, without referring to extensive cell preparation.
Collapse
|
20
|
Oviedo A, Yañez R, Colmenero I, Aldea M, Rubio A, Bueren JA, Lamana ML. Reduced efficacy of mesenchymal stromal cells in preventing graft-versus-host disease in an in vivo model of haploidentical bone marrow transplant with leukemia. Cell Transplant 2012; 22:1381-94. [PMID: 23044223 DOI: 10.3727/096368912x657666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cell (MSC) immunosuppressive properties have been applied to treat graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplants (HSCTs). We have previously demonstrated that MSC infusions early after haplo-HSCT prevent GVHD in a haploidentical-HSCT mouse model. Now, we investigated the impact that MSCs' immunosuppressive properties have on the graft-versus-leukemia (GVL) effect. First, to mimic a chronic myeloid leukemia (CML) relapse after a haploidentical HSCT, lethally irradiated mice were coinfused with haploidentical donor bone marrow cells plus syngenic hematopoietic progenitors transduced with a retroviral vector encoding both the BCR/ABL oncogene and the ΔNGFR marker gene. As expected, a CML-like myeloproliferative syndrome developed in all the recipient animals. The addition of haploidentical splenocytes to the transplanted graft prevented CML development by a GVL effect, and all transplanted recipients died of GVHD. This GVL mouse model allowed us to investigate the impact of MSCs infused to prevent GVHD on days 0, 7, and 14 after HSCT, on the GVL effect, expecting an increase in leukemic relapse. Strikingly, a high mortality of the recipients was observed, caused by GVHD, and only few leukemic cells were detected in the recipient animals. In contrast, GVHD prevention by MSCs in the absence of BCR/ABL leukemic cells resulted in a significant survival of the recipients. In vitro data pointed to an inability of MSCs to control strong CTLs responses against BCR/ABL. Our results show that, although an evident increase in leukemic relapses induced by MSCs could not be detected, they showed a reduced efficacy in preventing GVHD that precluded us to draw clear conclusions on MSCs' impact over GVL effect.
Collapse
Affiliation(s)
- Alberto Oviedo
- Hematopoiesis and Gene Therapy Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
21
|
Tolar J, Wagner JE. Management of severe epidermolysis bullosa by haematopoietic transplant: principles, perspectives and pitfalls. Exp Dermatol 2012; 21:896-900. [PMID: 23016552 DOI: 10.1111/exd.12014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2012] [Indexed: 12/17/2022]
Abstract
People with severe forms of epidermolysis bullosa (EB) develop widespread blistering and progressively debilitating multisystem complications that may result in a shortened lifespan. As some wounds in EB individuals are difficult or impossible to access with topical therapy, we examined the potential of systemic therapy with normal haematopoietic stem cells. In both animal models and children with EB, healthy donor cells from the haematopoietic graft migrated to the injured skin; simultaneously, there was an increase in the production of skin-specific structural proteins deficient in EB, increased skin integrity and reduced tendency to blister formation. Even though the majority of evaluable individuals have had a positive response in skin healing, frequently changing their quality of life, the improvement in lifestyle has been varied and the overall clinical response incomplete. To change the current amelioration of disease into a full cure, we propose to (i) increase safety as well as efficacy of haematopoietic cell transplant (HCT) using co-infusion of mesenchymal stromal/stem cells with haematopoietic stem cells and non-myeloablative conditioning for transplant; (ii) optimize homing of donor cells into the skin erosions in animal models of EB; and (iii) discover and test new drugs for EB therapy using patient-specific induced pluripotent stem cells. We conclude that although HCT has always been a risky treatment restricted to those with serious life-threatening or debilitating diseases, by most benchmarks, the results of HCT in EB have shown that HCT has the potential of being a durable, systemic therapy for people with severe forms of EB.
Collapse
Affiliation(s)
- Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
22
|
Rodgers CJ, Burge S, Scarisbrick J, Peniket A. More than skin deep? Emerging therapies for chronic cutaneous GVHD. Bone Marrow Transplant 2012; 48:323-37. [DOI: 10.1038/bmt.2012.96] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
23
|
Yew TL, Huang TF, Ma HL, Hsu YT, Tsai CC, Chiang CC, Chen WM, Hung SC. Scale-up of MSC under hypoxic conditions for allogeneic transplantation and enhancing bony regeneration in a rabbit calvarial defect model. J Orthop Res 2012; 30:1213-20. [PMID: 22278907 DOI: 10.1002/jor.22070] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 12/20/2011] [Indexed: 02/04/2023]
Abstract
To realize the therapeutic potential of mesenchymal stem cells (MSCs), we aimed to develop a method for isolating and expanding New Zealand rabbit MSCs in a great scale. Rabbit MSCs expanded under hypoxic and normoxic conditions were compared in terms of replication capacity, differentiation potential, and the capacity for allogeneic transplantation in a calvarial defect model. The cells from all tested rabbits were expanded more rapidly when plated at low-density under hypoxic conditions compared to under normoxic conditions. Moreover, cells expanded under hypoxic conditions increased in the potential of osteoblastic, adipocytic, and chondrocytic differentiation. More importantly, radiographic analysis and micro-CT measurement of bone volume revealed the hypoxic cells when transplanted in the calvarial defects of another rabbit increased in the ability to repair bone defect compared to the normoxic cells. Six weeks after allogeneic transplantation of hypoxic MSCs, histological analysis revealed a callus spanned the length of the defect, and several bone tissues spotted in the implant. At 12 weeks, new bone had formed throughout the implant. Using BrdU labeling to track the transplanted cells, the hypoxic cells were more detected in the newly formed bone compared to the normoxic cells. For defects treated with allogeneic MSCs, no adverse host response could be detected at any time-point. In conclusion, we have developed a robust method for isolation and expansion of rabbit MSCs by combining low-density with hypoxic culture, which can be applied for the design of clinical trials in allogeneic transplantation of MSCs for bone healing.
Collapse
Affiliation(s)
- Tu-Lai Yew
- Faculty of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei 112, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Zanotti L, Sarukhan A, Dander E, Castor M, Cibella J, Soldani C, Trovato AE, Ploia C, Luca G, Calvitti M, Mancuso F, Arato I, Golemac M, Jonjic N, Biondi A, Calafiore R, Locati M, D'Amico G, Viola A. Encapsulated mesenchymal stem cells for in vivo immunomodulation. Leukemia 2012; 27:500-3. [PMID: 22878604 DOI: 10.1038/leu.2012.202] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
25
|
Shi C. Recent progress toward understanding the physiological function of bone marrow mesenchymal stem cells. Immunology 2012; 136:133-8. [PMID: 22321024 DOI: 10.1111/j.1365-2567.2012.03567.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that are being clinically explored as regenerative therapeutics. Cultured MSCs secrete various modulatory factors, which contribute to the immunosuppressive effects of transplanted MSCs as a therapy. Although the in vitro phenotype of MSCs has been well characterized, identification of MSCs in vivo is made difficult by the lack of specific markers. Current advances in murine MSC research provide valuable tools for studying the localization and function of MSCs in vivo. Recent findings suggest that MSCs exert diverse functions depending on tissue context and physiological conditions. This review focuses on bone marrow MSCs and their roles in haematopoiesis and immune responses.
Collapse
Affiliation(s)
- Chao Shi
- Immunology Program, Sloan-Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
26
|
Mesenchymal stem cells for cardiac regeneration: translation to bedside reality. Stem Cells Int 2012; 2012:646038. [PMID: 22754578 PMCID: PMC3382381 DOI: 10.1155/2012/646038] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/03/2012] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. According to the World Health Organization (WHO), an estimate of 17.3 million people died from CVDs in 2008 and by 2030, the number of deaths is estimated to reach almost 23.6 million. Despite the development of a variety of treatment options, heart failure management has failed to inhibit myocardial scar formation and replace the lost cardiomyocyte mass with new functional contractile cells. This shortage is complicated by the limited ability of the heart for self-regeneration. Accordingly, novel management approaches have been introduced into the field of cardiovascular research, leading to the evolution of gene- and cell-based therapies. Stem cell-based therapy (aka, cardiomyoplasty) is a rapidly growing alternative for regenerating the damaged myocardium and attenuating ischemic heart disease. However, the optimal cell type to achieve this goal has not been established yet, even after a decade of cardiovascular stem cell research. Mesenchymal stem cells (MSCs) in particular have been extensively investigated as a potential therapeutic approach for cardiac regeneration, due to their distinctive characteristics. In this paper, we focus on the therapeutic applications of MSCs and their transition from the experimental benchside to the clinical bedside.
Collapse
|
27
|
Kim J, Breunig MJ, Escalante LE, Bhatia N, Denu RA, Dollar BA, Stein AP, Hanson SE, Naderi N, Radek J, Haughy D, Bloom DD, Assadi-Porter FM, Hematti P. Biologic and immunomodulatory properties of mesenchymal stromal cells derived from human pancreatic islets. Cytotherapy 2012; 14:925-35. [PMID: 22571381 DOI: 10.3109/14653249.2012.684376] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) have now been shown to reside in numerous tissues throughout the body, including the pancreas. Ex vivo culture-expanded MSC derived from many tissues display important interactions with different types of immune cells in vitro and potentially play a significant role in tissue homeostasis in vivo. In this study, we investigated the biologic and immunomodulatory properties of human pancreatic islet-derived MSC. METHODS We culture-expanded MSC from cadaveric human pancreatic islets and characterized them using flow cytometry, differentiation assays and nuclear magnetic resonance-based metabolomics. We also investigated the immunologic properties of pancreatic islet-derived MSC compared with bone marrow (BM) MSC. RESULTS Pancreatic islet and BM-derived MSC expressed the same cell-surface markers by flow cytometry, and both could differentiate into bone, fat and cartilage. Metabolomics analysis of MSC from BM and pancreatic islets also showed a similar set of metabolic markers but quantitative polymerase chain reactions showed that pancreatic islet MSC expressed more interleukin(IL)-1b, IL-6, STAT3 and FGF9 compared with BM MSC, and less IL-10. However, similar to BM MSC, pancreatic islet MSC were able to suppress proliferation of allogeneic T lymphocytes stimulated with anti-CD3 and anti-CD28 antibodies. CONCLUSIONS Our in vitro analysis shows pancreatic islet-derived MSC have phenotypic, biologic and immunomodulatory characteristics similar, but not identical, to BM-derived MSC. We propose that pancreatic islet-derived MSC could potentially play an important role in improving the outcome of pancreatic islet transplantation by promoting engraftment and creating a favorable immune environment for long-term survival of islet allografts.
Collapse
Affiliation(s)
- Jaehyup Kim
- University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jung Y, Bauer G, Nolta JA. Concise review: Induced pluripotent stem cell-derived mesenchymal stem cells: progress toward safe clinical products. Stem Cells 2012; 30:42-7. [PMID: 21898694 DOI: 10.1002/stem.727] [Citation(s) in RCA: 191] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adult stem cell therapies have provided success for more than 50 years, through reconstitution of the hematopoietic system using bone marrow, umbilical cord blood, and mobilized peripheral blood transplantation. Mesenchymal stem cell (MSC)-mediated therapy is a fast-growing field that has proven safe and effective in the treatment of various degenerative diseases and tissue injuries. Since the first derivation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), there has been impressive progress toward developing safe clinical applications from PSCs. Recent successes in transgene-free iPSC reprogramming have brought attention to the potential of clinical applications of these pluripotent cells, but key hurdles must be overcome, which are discussed in this review. Looking to the future, it could be advantageous to derive MSC from iPSC or human ESC in cases where genetic engineering is needed, since in the PSCs, clones with "safe harbor" vector integration could be selected, expanded, and differentiated. Here, we describe the status of the progress of the use of MSC and PSCs in clinical trials and analyze the challenges that should be overcome before iPSC-derived MSC therapy can be used widely in the clinic.
Collapse
Affiliation(s)
- Yunjoon Jung
- Department of Biomedical Engineering, University of California, Davis, Sacramento, California 95817, USA
| | | | | |
Collapse
|
29
|
Zinöcker S, Vaage JT. Rat mesenchymal stromal cells inhibit T cell proliferation but not cytokine production through inducible nitric oxide synthase. Front Immunol 2012; 3:62. [PMID: 22566943 PMCID: PMC3341954 DOI: 10.3389/fimmu.2012.00062] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 03/12/2012] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stromal cells (MSC) have important immunomodulatory properties, they inhibit T lymphocyte allo-activation and have been used to treat graft-versus-host disease. How MSC exert their immunosuppressive functions is not completely understood but species specific mechanisms have been implicated. In this study we have investigated the mechanisms for rat MSC mediated inhibition of T lymphocyte proliferation and secretion of inflammatory cytokines in response to allogeneic and mitogenic stimuli in vitro. MSC inhibited the proliferation of T cells in allogeneic mixed lymphocyte reactions and in response to mitogen with similar efficacy. The anti-proliferative effect was mediated by the induced expression of nitric oxide (NO) synthase and production of NO by MSC. This pathway was required and sufficient to fully suppress lymphocyte proliferation and depended on proximity of MSC and target cells. Expression of inducible NO synthase by MSC was induced through synergistic stimulation with tumor necrosis factor α and interferon γ secreted by activated lymphocytes. Conversely, MSC had a pronounced inhibitory effect on the secretion of these cytokines by T cells which did not depend on NO synthase activity or cell contact, but was partially reversed by addition of the cyclooxygenase (COX) inhibitor indomethacin. In conclusion, rat MSC use different mechanisms to inhibit proliferative and inflammatory responses of activated T cells. While proliferation is suppressed by production of NO, cytokine secretion appears to be impaired at least in part by COX-dependent production of prostaglandin E2.
Collapse
Affiliation(s)
- Severin Zinöcker
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | |
Collapse
|
30
|
Electric impedance sensing in cell-substrates for rapid and selective multipotential differentiation capacity monitoring of human mesenchymal stem cells. Biosens Bioelectron 2012; 34:63-9. [DOI: 10.1016/j.bios.2012.01.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 01/11/2012] [Accepted: 01/13/2012] [Indexed: 01/18/2023]
|
31
|
Jamnig A, Lepperdinger G. From tendon to nerve: an MSC for all seasons. Can J Physiol Pharmacol 2012; 90:295-306. [DOI: 10.1139/y11-109] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The potential of mesenchymal stem cells (MSCs) to regenerate damaged tissue is well documented, as this specialized progenitor cell type exhibits superior cellular properties, and would allow medical as well as ethical limitations to be overcome. By now, MSCs have been successfully introduced in manifold experimental approaches within the newly defined realm of Regenerative Medicine. Advanced methods for in vitro cell expansion, defined induction of distinct differentiation processes, 3-dimensional culture on specific scaffold material, and tissue engineering approaches have been designed, and many clinical trials not only have been launched, but recently could be completed. To date, most of the MSC-based therapeutic approaches have been executed to address bone, cartilage, or heart regeneration; further, prominent studies have shown the efficacy of ex vivo expanded and infused MSCs to countervail graft-versus-host disease. Yet more fields of application emerge in which MSCs unfold beneficial effects, and presently, therapies that effectively ameliorate nonhealing conditions after tendon or spinal cord injury are, courtesy of scientific research, forging ahead to the clinical trial stage.
Collapse
Affiliation(s)
- Angelika Jamnig
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, Innsbruck A-6020, Austria
| | - Günter Lepperdinger
- Extracellular Matrix Research, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, Innsbruck A-6020, Austria
| |
Collapse
|
32
|
Nixon AJ, Watts AE, Schnabel LV. Cell- and gene-based approaches to tendon regeneration. J Shoulder Elbow Surg 2012; 21:278-94. [PMID: 22244071 DOI: 10.1016/j.jse.2011.11.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 11/14/2011] [Accepted: 11/15/2011] [Indexed: 02/06/2023]
Abstract
Repair of rotator cuff tears in experimental models has been significantly improved by the use of enhanced biologic approaches, including platelet-rich plasma, bone marrow aspirate, growth factor supplements, and cell- and gene-modified cell therapy. Despite added complexity, cell-based therapies form an important part of enhanced repair, and combinations of carrier vehicles, growth factors, and implanted cells provide the best opportunity for robust repair. Bone marrow-derived mesenchymal stem cells provide a stimulus for repair in flexor tendons, but application in rotator cuff repair has not shown universally positive results. The use of scaffolds such as platelet-rich plasma, fibrin, and synthetic vehicles and the use of gene priming for stem cell differentiation and local anabolic and anti-inflammatory impact have both provided essential components for enhanced tendon and tendon-to-bone repair in rotator cuff disruption. Application of these research techniques in human rotator cuff injury has generally been limited to autologous platelet-rich plasma, bone marrow concentrate, or bone marrow aspirates combined with scaffold materials. Cultured mesenchymal progenitor therapy and gene-enhanced function have not yet reached clinical trials in humans. Research in several animal species indicates that the concept of gene-primed stem cells, particularly embryonic stem cells, combined with effective culture conditions, transduction with long-term integrating vectors carrying anabolic growth factors, and development of cells conditioned by use of RNA interference gene therapy to resist matrix metalloproteinase degradation, may constitute potential advances in rotator cuff repair. This review summarizes cell- and gene-enhanced cell research for tendon repair and provides future directions for rotator cuff repair using biologic composites.
Collapse
Affiliation(s)
- Alan J Nixon
- Comparative Orthopaedics Laboratory, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA.
| | | | | |
Collapse
|
33
|
Cuthbert R, Boxall SA, Tan HB, Giannoudis PV, McGonagle D, Jones E. Single-platform quality control assay to quantify multipotential stromal cells in bone marrow aspirates prior to bulk manufacture or direct therapeutic use. Cytotherapy 2012; 14:431-40. [PMID: 22268519 DOI: 10.3109/14653249.2011.651533] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND AIMS The manufacture of multipotential stromal cell (MSC)-based products is costly; therefore, a rapid evaluation of bone marrow (BM) 'quality' with respect to MSC content is desirable. The aim of this study was to develop a rapid single-platform assay to quantify MSC in BM aspirates. METHODS Aspirated MSC were enumerated using the CD45(-/low) CD271(bright) phenotype and AccuCheck counting beads and compared with a classic colony-forming unit-fibroblast (CFU-F) assay. The phenotype of CD45(-/low) CD271(bright) cells was defined using a range of MSC (CD73, CD105, CD90) and non-MSC (CD31, CD33, CD34, CD19) markers. The effect of aspirated BM volume on MSC yield was also determined. RESULTS CD45(-/low) CD271(bright) cells had a classic MSC phenotype (CD73(+) CD105(+) CD90(+)). Their numbers correlated positively with CFU-F counted manually (R = 0.81, P < 0.001) or using automatic measurements of surface area occupied by colonies (R = 0.66, P < 0.001). Simultaneous enumeration of CD34(+) cells revealed donor variability ranges compatible with standard International Society of Hematotherapy and Graft Engineering (ISHGE) protocols. Aspirating larger marrow volumes gave a significant several-fold reduction in the frequency of CFU-F and CD45(-/low) CD271(bright) cells per milliliter. Therefore aspirated MSC yields can be maximized through a standardized, low-volume harvesting technique. CONCLUSIONS Absolute quantification of CD45(-/low) CD271(bright) cells was found to be a reliable method of predicting CFU-F yields in BM aspirates. This rapid (< 40 min) procedure could be suitable for intra-operative quality control of BM aspirates prior to volume reduction/direct injection in orthopedics. In the production of culture-expanded MSC, this assay could be used to exclude samples containing low numbers of MSC, resulting in improved consistency and quality of manufactured MSC batches.
Collapse
Affiliation(s)
- Richard Cuthbert
- Academic Unit of Musculoskeletal Disease, Leeds Institute of Molecular Medicine, University of Leeds, UK
| | | | | | | | | | | |
Collapse
|
34
|
Yarborough M, Tempkin T, Nolta J, Joyce N. The Complex Ethics of First In Human Stem Cell Clinical Trials. AJOB Neurosci 2012; 3:10.1080/21507740.2012.675010. [PMID: 24273680 PMCID: PMC3835337 DOI: 10.1080/21507740.2012.675010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
35
|
Beksac M, Preffer F. Is it time to revisit our current hematopoietic progenitor cell quantification methods in the clinic? Bone Marrow Transplant 2011; 47:1391-6. [PMID: 22139068 DOI: 10.1038/bmt.2011.240] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In the clinical practice of hematopoietic SCT, the minimum numbers of cells required for a successful engraftment are defined on the basis of their CD45 and CD34 expression profiles. However, the quantity of earlier progenitors or CD34-positive cells at different differentiation stages within stem cell grafts is not generally taken into consideration. During the last decade, various teams have quantified the number of cells expressing various combinations of CD34, CD38, CD133, CD90 co-expression and/or aldehyde dehydrogenase functional capacity using flow cytometry. Some of these studies resulted in the greater appreciation that combinations of these Ags were associated with varied myeloid, erythroid and platelet engraftment rates whereas others showed that the relative absence or presence of these markers could define cells responsible for either short- or long-term engraftment. These findings were also extended to differences between progenitor cell populations found within BM vs peripheral or cord-blood grafts. Cells harvested from donors are also generally frozen and stored; thawed cells have variable levels of viability and functional capacity based on the time tested post thaw, which also can be assessed by flow cytometry. Finally, flow cytometry has the potential for analysis of cells carrying a mesenchymal stem cell phenotype, which may be quiescent within some of the stem cell products. This review will address the need for stem cell subpopulation quantification and summarize existing published data to identify some Ags and functional characteristics that can be applicable to daily clinical practice.
Collapse
Affiliation(s)
- M Beksac
- Ankara University School of Medicine, Department of Hematology, Ankara, Turkey.
| | | |
Collapse
|
36
|
Gregoire-Gauthier J, Selleri S, Fontaine F, Dieng MM, Patey N, Despars G, Beauséjour CM, Haddad E. Therapeutic efficacy of cord blood-derived mesenchymal stromal cells for the prevention of acute graft-versus-host disease in a xenogenic mouse model. Stem Cells Dev 2011; 21:1616-26. [PMID: 21910645 DOI: 10.1089/scd.2011.0413] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stromal cells (MSCs) have been successfully utilized for the treatment of refractory graft-versus-host disease (GvHD). Despite the large number of in vitro and in vivo models developed for clarifying their immunomodulatory properties, the mechanism of action of MSCs remains elusive and their efficacy controversial. Here, we tested the ability of cord blood-derived MSCs to alleviate the symptoms of GvHD induced by the injection of human peripheral blood mononuclear cells into NOD/SCID/γc(-) mice. In this in vivo xeno-GvHD model, we demonstrate that a single MSC injection is able to inhibit GvHD in terms of clinical signs and related mortality. We also show that in this model MSCs act by both immunomodulating T-cells and fostering recovery after irradiation. The translational impact of these findings could provide a reliable preclinical model for studying the efficacy, dosage, and time of administration of human MSCs for the prevention of acute GvHD.
Collapse
|
37
|
|
38
|
Abstract
Recent observations have demonstrated that one of the functions of mesenchymal stem/stromal cells (MSCs) is to serve as guardians against excessive inflammatory responses. One mode of action of the cells is that they are activated to express the interleukin (IL)-1 receptor antagonist. A second mode of action is to create a negative feedback loop in which tumor necrosis factor-α (TNF-α) and other proinflammatory cytokines from resident macrophages activate MSCs to secrete the multifunctional anti-inflammatory protein TNF-α stimulated gene/protein 6 (TSG-6). The TSG-6 then reduces nuclear factor-κB (NF-κB) signaling in the resident macrophages and thereby modulates the cascade of proinflammatory cytokines. A third mode of action is to create a second negative feedback loop whereby lipopolysaccharide, TNF-α, nitric oxide, and perhaps other damage-associated molecular patterns (DAMPs) from injured tissues and macrophages activate MSCs to secrete prostaglandin E(2) (PGE(2)). The PGE(2) converts macrophages to the phenotype that secretes IL-10. There are also suggestions that MSCs may produce anti-inflammatory effects through additional modes of action including activation to express the antireactive oxygen species protein stanniocalcin-1.
Collapse
|
39
|
Wernicke CM, Grunewald TG, Hendrik J, Kuci S, Kuci Z, Koehl U, Mueller I, Doering M, Peters C, Lawitschka A, Kolb HJ, Bader P, Burdach S, von Luettichau I. Mesenchymal stromal cells for treatment of steroid-refractory GvHD: a review of the literature and two pediatric cases. Int Arch Med 2011; 4:27. [PMID: 21843360 PMCID: PMC3169455 DOI: 10.1186/1755-7682-4-27] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Accepted: 08/15/2011] [Indexed: 12/11/2022] Open
Abstract
Severe acute graft versus host disease (GvHD) is a life-threatening complication after allogeneic hematopoietic stem cell transplantation. Human mesenchymal stromal cells (MSCs) play an important role in endogenous tissue repair and possess strong immune-modulatory properties making them a promising tool for the treatment of steroid-refractory GvHD. To date, a few reports exist on the use of MSCs in treatment of GvHD in children indicating that children tend to respond better than adults, albeit with heterogeneous results. We here present a review of the literature and the clinical course of two instructive pediatric patients with acute steroid-refractory GvHD after haploidentical stem cell transplantation, which exemplify the beneficial effects of third-party transplanted MSCs in treatment of acute steroid-refractory GvHD. Moreover, we provide a meta-analysis of clinical studies addressing the outcome of patients with steroid-refractory GvHD and treatment with MSCs in adults and in children (n = 183; 122 adults, 61 children). Our meta-analysis demonstrates that the overall response-rate is high (73.8%) and confirms, for the first time, that children indeed respond better to treatment of GvHD with MSCs than adults (complete response 57.4% vs. 45.1%, respectively). These data emphasize the significance of this therapeutic approach especially in children and indicate that future prospective studies are needed to assess the reasons for the observed differential response-rates in pediatric and adult patients.
Collapse
Affiliation(s)
- Caroline M Wernicke
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| | - Thomas Gp Grunewald
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany.,Medical Life Science and Technology Center, TUM Graduate School, Technische Universität München, Boltzmannstrasse 17, 85748 Garching, Germany
| | - Juenger Hendrik
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| | - Selim Kuci
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Zyrafete Kuci
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ulrike Koehl
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ingo Mueller
- University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Michaela Doering
- University Children's Hospital, Hoppe-Seyler-Strasse 1, 72076 Tuebingen, Germany
| | - Christina Peters
- St. Anna Children's Hospital, Kinderspitalgasse 6, 1090 Vienna, Austria
| | - Anita Lawitschka
- St. Anna Children's Hospital, Kinderspitalgasse 6, 1090 Vienna, Austria
| | - Hans-Jochem Kolb
- Division for Stem Cell Transplantation, Department of Medicine III, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation, Department of Hematology, Oncology and Hemostasis, Hospital for Children and Adolescents, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Stefan Burdach
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| | - Irene von Luettichau
- Children's Cancer Research and Roman Herzog Comprehensive Cancer Center, Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Kölner Platz 1, 80804 Munich, Germany
| |
Collapse
|
40
|
Combination therapy with bone marrow stromal cells and FK506 enhanced amelioration of ischemic brain damage in rats. Life Sci 2011; 89:50-6. [DOI: 10.1016/j.lfs.2011.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 04/28/2011] [Accepted: 04/30/2011] [Indexed: 11/19/2022]
|
41
|
Landgraf K, Brunauer R, Lepperdinger G, Grubeck-Loebenstein B. The suppressive effect of mesenchymal stromal cells on T cell proliferation is conserved in old age. Transpl Immunol 2011; 25:167-72. [PMID: 21726642 DOI: 10.1016/j.trim.2011.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a useful tool in curing graft versus host disease (GVHD) after transplantation. No information is presently available whether the immunosuppressive properties of this cell type are maintained in old age. It was therefore the aim of our study to analyze the immunoregulatory effect of MSC on peripheral blood mononuclear cells (PBMC) in old age. We studied the proliferation, activation and cytokine production of PBMC following co-culture with MSC from young (<30 years) and old (>61 years) donors. Our results demonstrate that MSC from elderly donors exhibit the same suppressive effects on T cell proliferation as their young counterparts. In both age groups T cell activation was not influenced by co-culture with MSC from young and elderly donors. With the exception of IL-6, cytokine production by unstimulated or stimulated PBMC was also not affected by MSC from either age group. IL-6 production was increased during co-culture of PBMC and MSC and was higher when MSC from elderly donors were used. After PHA stimulation, however, this age-specific difference was balanced and appeared even. As high IL-6 production is a prerequisite for an effective suppression of T cell proliferation, MSC can be considered a powerful tool for immunoregulatory therapies in old age.
Collapse
Affiliation(s)
- Katja Landgraf
- Institute for Biomedical Aging Research, Austrian Academy of Sciences, Rennweg 10, 6020 Innsbruck, Austria.
| | | | | | | |
Collapse
|