1
|
Nguyen H, Hsu CC, Meeson A, Oldershaw R, Richardson G, Czosseck A, Lundy DJ. Differentiation, Metabolism, and Cardioprotective Secretory Functions of Human Cardiac Stromal Cells from Ischemic and Endocarditis Patients. Stem Cells Dev 2024; 33:484-495. [PMID: 38940748 DOI: 10.1089/scd.2024.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024] Open
Abstract
This study investigates the characteristics of cardiac mesenchymal stem cell-like cells (CMSCLCs) isolated from the right atrial appendage of human donors with ischemia and a young patient with endocarditis (NE-CMSCLCs). Typical CMSCLCs from ischemic heart patients were derived from coronary artery bypass grafting procedures and compared against bone marrow mesenchymal stromal cells (BM-MSCs). NE-CMSCLCs had a normal immunophenotype, but exhibited enhanced osteogenic differentiation potential, rapid proliferation, reduced senescence, reduced glycolysis, and lower reactive oxygen species generation after oxidative stress compared with typical ischemic CMSCLCs. These differences suggest a unique functional status of NE-CMSCLCs, influenced by the donor health condition. Despite large variances in their paracrine secretome, NE-CMSCLCs retained therapeutic potential, as indicated by their ability to protect hypoxia/reoxygenation-injured human cardiomyocytes, albeit less effectively than typical CMSCLCs. This research describes a unique cell phenotype and underscores the importance of donor health status in the therapeutic efficacy of autologous cardiac cell therapy.
Collapse
Affiliation(s)
- Helen Nguyen
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chuan-Chih Hsu
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Annette Meeson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rachel Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Gavin Richardson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andreas Czosseck
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - David J Lundy
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
- International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- Center for Cell Therapy, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Vadakke‐Madathil S, Chaudhry HW. Concepts of Cell Therapy and Myocardial Regeneration. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
3
|
Park YS, Park BW, Choi H, Lee SH, Kim M, Park HJ, Kim IB. Chorion-derived perinatal mesenchymal stem cells improve cardiac function and vascular regeneration: preferential treatment for ischemic heart disease. Hellenic J Cardiol 2022; 66:52-58. [DOI: 10.1016/j.hjc.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/04/2022] Open
|
4
|
Beliën H, Evens L, Hendrikx M, Bito V, Bronckaers A. Combining stem cells in myocardial infarction: The road to superior repair? Med Res Rev 2021; 42:343-373. [PMID: 34114238 DOI: 10.1002/med.21839] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/04/2021] [Accepted: 05/29/2021] [Indexed: 12/25/2022]
Abstract
Myocardial infarction irreversibly destroys millions of cardiomyocytes in the ventricle, making it the leading cause of heart failure worldwide. Over the past two decades, many progenitor and stem cell types were proposed as the ideal candidate to regenerate the heart after injury. The potential of stem cell therapy has been investigated thoroughly in animal and human studies, aiming at cardiac repair by true tissue replacement, by immune modulation, or by the secretion of paracrine factors that stimulate endogenous repair processes. Despite some successful results in animal models, the outcome from clinical trials remains overall disappointing, largely due to the limited stem cell survival and retention after transplantation. Extensive interest was developed regarding the combinational use of stem cells and various priming strategies to improve the efficacy of regenerative cell therapy. In this review, we provide a critical discussion of the different stem cell types investigated in preclinical and clinical studies in the field of cardiac repair. Moreover, we give an update on the potential of stem cell combinations as well as preconditioning and explore the future promises of these novel regenerative strategies.
Collapse
Affiliation(s)
- Hanne Beliën
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Lize Evens
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Virginie Bito
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| |
Collapse
|
5
|
Raposo L, Lourenço AP, Nascimento DS, Cerqueira R, Cardim N, Leite-Moreira A. Human umbilical cord tissue-derived mesenchymal stromal cells as adjuvant therapy for myocardial infarction: a review of current evidence focusing on pre-clinical large animal models and early human trials. Cytotherapy 2021; 23:974-979. [PMID: 34112613 DOI: 10.1016/j.jcyt.2021.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022]
Abstract
Although biologically appealing, the concept of tissue regeneration underlying first- and second-generation cell therapies has failed to translate into consistent results in clinical trials. Several types of cells from different origins have been tested in pre-clinical models and in patients with acute myocardial infarction (AMI). Mesenchymal stromal cells (MSCs) have gained attention because of their potential for immune modulation and ability to promote endogenous tissue repair, mainly through their secretome. MSCs can be easily obtained from several human tissues, the umbilical cord being the most abundant source, and further expanded in culture, making them attractive as an allogeneic "of-the-shelf" cell product, suitable for the AMI setting. The available evidence concerning umbilical cord-derived MSCs in AMI is reviewed, focusing on large animal pre-clinical studies and early human trials. Molecular and cellular mechanisms as well as current limitations and possible translational solutions are also discussed.
Collapse
Affiliation(s)
- Luís Raposo
- Cardiology Department, Santa Cruz Hospital, West Lisbon Hospital Center, Lisbon, Portugal; Hospital da Luz Lisboa, Luz Saúde, Lisbon, Portugal; Nova Medical School, Lisbon, Portugal.
| | - André P Lourenço
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diana S Nascimento
- Institute for Research and Innovation in Health, University of Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Portugal; Instituto Nacional de Engenharia Biomédica, University of Porto, Portugal
| | - Rui Cerqueira
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Nuno Cardim
- Hospital da Luz Lisboa, Luz Saúde, Lisbon, Portugal; Nova Medical School, Lisbon, Portugal
| | - Adelino Leite-Moreira
- Department of Cardiac Surgery, University Hospital Centre São João, Porto, Portugal; Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
6
|
Mesenchymal Stem Cells for Cardiac Regeneration: from Differentiation to Cell Delivery. Stem Cell Rev Rep 2021; 17:1666-1694. [PMID: 33954876 DOI: 10.1007/s12015-021-10168-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are so far the most widely researched stem cells in clinics and used as an experimental cellular therapy module, particularly in cardiac regeneration and repair. Ever since the discovery of cardiomyogenesis induction in MSCs, a wide variety of differentiation protocols have been extensively used in preclinical models. However, pre differentiated MSC-derived cardiomyocytes have not been used in clinical trials; highlighting discrepancies and limitations in its use as a source of derived cardiomyocytes for transplantation to improve the damaged heart function. Therefore, this review article focuses on the strategies used to derive cardiomyocytes-like cells from MSCs isolated from three widely used tissue sources and their differentiation efficiencies. We have further discussed the role of MSCs in inducing angiogenesis as a cellular precursor to endothelial cells and its secretory aspects including exosomes. We have then discussed the strategies used for delivering cells in the damaged heart and how its retention plays a critical role in the overall outcome of the therapy. We have also conversed about the scope of the local and systemic modes of delivery of MSCs and the application of biomaterials to improve the overall delivery efficacy and function. We have finally discussed the advantages and limitations of cell delivery to the heart and the future scope of MSCs in cardiac regenerative therapy.
Collapse
|
7
|
Ghodrat S, Hoseini SJ, Asadpour S, Nazarnezhad S, Alizadeh Eghtedar F, Kargozar S. Stem cell-based therapies for cardiac diseases: The critical role of angiogenic exosomes. Biofactors 2021; 47:270-291. [PMID: 33606893 DOI: 10.1002/biof.1717] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
Finding effective treatments for cardiac diseases is among the hottest subjects in medicine; cell-based therapies have brought great promises for managing a broad range of life-threatening heart complications such as myocardial infarction. After clarifying the critical role of angiogenesis in tissue repair and regeneration, various stem/progenitor cell were utilized to accelerate the healing of injured cardiac tissue. Embryonic, fetal, adult, and induced pluripotent stem cells have shown the appropriate proangiogenic potential for tissue repair strategies. The capability of stem cells for differentiating into endothelial lineages was initially introduced as the primary mechanism involved in improving angiogenesis and accelerated heart tissue repair. However, recent studies have demonstrated the leading role of paracrine factors secreted by stem cells in advancing neo-vessel formation. Genetically modified stem cells are also being applied for promoting angiogenesis regarding their ability to considerably overexpress and secrete angiogenic bioactive molecules. Yet, conducting further research seems necessary to precisely identify molecular mechanisms behind the proangiogenic potential of stem cells, including the signaling pathways and regulatory molecules such as microRNAs. In conclusion, stem cells' pivotal roles in promoting angiogenesis and consequent improved cardiac healing and remodeling processes should not be ignored, especially in the case of stem cell-derived extracellular vesicles.
Collapse
Affiliation(s)
- Sara Ghodrat
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Javad Hoseini
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shiva Asadpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Alizadeh Eghtedar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Haller C, Friedberg MK, Laflamme MA. The role of regenerative therapy in the treatment of right ventricular failure: a literature review. Stem Cell Res Ther 2020; 11:502. [PMID: 33239066 PMCID: PMC7687832 DOI: 10.1186/s13287-020-02022-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/09/2020] [Indexed: 01/13/2023] Open
Abstract
Right ventricular (RV) failure is a commonly encountered problem in patients with congenital heart disease but can also be a consequence of left ventricular disease, primary pulmonary hypertension, or RV-specific cardiomyopathies. Improved survival of the aforementioned pathologies has led to increasing numbers of patients suffering from RV dysfunction, making it a key contributor to morbidity and mortality in this population. Currently available therapies for heart failure were developed for the left ventricle (LV), and there is clear evidence that LV-specific strategies are insufficient or inadequate for the RV. New therapeutic strategies are needed to address this growing clinical problem, and stem cells show significant promise. However, to properly evaluate the prospects of a potential stem cell-based therapy for RV failure, one needs to understand the unique pathophysiology of RV dysfunction and carefully consider available data from animal models and human clinical trials. In this review, we provide a comprehensive overview of the molecular mechanisms involved in RV failure such as hypertrophy, fibrosis, inflammation, changes in energy metabolism, calcium handling, decreasing RV contractility, and apoptosis. We also summarize the available preclinical and clinical experience with RV-specific stem cell therapies, covering the broad spectrum of stem cell sources used to date. We describe two different scientific rationales for stem cell transplantation, one of which seeks to add contractile units to the failing myocardium, while the other aims to augment endogenous repair mechanisms and/or attenuate harmful remodeling. We emphasize the limitations and challenges of regenerative strategies, but also highlight the characteristics of the failing RV myocardium that make it a promising target for stem cell therapy.
Collapse
Affiliation(s)
- Christoph Haller
- Division of Cardiovascular Surgery, The Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,McEwen Stem Cell Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Canada
| | - Mark K Friedberg
- Division of Cardiology, The Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada. .,McEwen Stem Cell Institute, Toronto Medical Discovery Tower, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.
| |
Collapse
|
9
|
White SJ, Chong JJH. Mesenchymal Stem Cells in Cardiac Repair: Effects on Myocytes, Vasculature, and Fibroblasts. Clin Ther 2020; 42:1880-1891. [PMID: 32938532 DOI: 10.1016/j.clinthera.2020.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/09/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Cardiac pathologies remain a dominant cause of morbidity and mortality within the community. The drive to develop therapies capable of repairing damaged heart tissue to achieve clinically significant restoration of function has motivated the pursuit of novel approaches such as cell therapy. To this end, evidence of therapeutic benefits achieved by using mesenchymal stem cells (MSCs) has captured considerable interest despite a relative lack of information regarding the mechanisms involved. This narrative review synthesizes and interprets the current literature describing mechanisms by which MSCs can elicit cardiac repair, thereby directing attention to avenues of further inquiry. METHODS OVID versions of MEDLINE and EMBASE were searched for studies describing the role of MSCs in mammalian cardiac repair. Additional studies were sourced from the reference lists of relevant articles and other personal files. FINDINGS MSCs elicit cardiac repair in a range of in vitro systems and animal models of diseases such as myocardial infarction and heart failure. Important mechanisms include the preservation of myocardial contractility, the promotion of angiogenesis, and the modulation of fibrosis. Exposing in vitro MSCs to a microenvironment reflective of that encountered in the injured heart seems to potentiate these therapeutic mechanisms. IMPLICATIONS Promising results in animal studies warrant continuation of clinical MSC cardiac therapy studies. Paracrine functions of MSCs seem to be the dominant mechanism of cardiac repair over direct cellular effects. Although integral, the MSC secretome remains poorly defined. In addition, most of the mechanistic data within the literature have been derived from animal MSC research, necessitating more human MSC-based work.
Collapse
Affiliation(s)
- Samuel J White
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
10
|
Sokolowska P, Zukowski K, Lasocka I, Szulc-Dabrowska L, Jastrzebska E. Human mesenchymal stem cell (hMSC) differentiation towards cardiac cells using a new microbioanalytical method. Analyst 2020; 145:3017-3028. [PMID: 32133460 DOI: 10.1039/c9an02366f] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells (SCs) are more and more often applied in tissue engineering and cell therapies, e.g. in regenerative medicine. Standard methods of SC differentiation are time consuming and ineffective. Therefore, new bioanalytical methods (i.e. Lab-on-a-Chip systems) are develop to improve such type of studies. Although, microtechnology is a rapidly growing research area, there are so far not too many works which present SC differentiation into cardiomyocytes in the microsystems. Therefore, we present new microbioanalytical method of SC differentiation towards cardiac cells using a newly developed digitally controlled microdispenser integrated with a Heart-on-a-chip system. Seven-day culture of human mesenchymal stem cells (hMSCs) and their differentiation using biochemical factors such as 5-AZA (2 μM, 24 h) and VEGF (20 ng ml-1, 72 h) were investigated in the microsystem which was automatically operated using smartphone software. hMSC differentiation into the cardiac cells was confirmed using immunostaining of cardiac markers (α-actinin and troponin T). The usage of the microsystem allowed shortening the time of hMSC differentiation in comparison to macroscale method. We showed that the microsystem, in which the in vivo microenvironment is mimicked and dynamic conditions are provided by a microdispenser, favorably affect hMSC differentiation towards cardiac cells. Based on the presented research we can conclude that the developed digitally controlled microsystem could be successfully utilized as a new microbioanalytical method for stem cells differentiation and analysis of their function under dynamic conditions. In the future, this could be a helpful tool for scientists working on regenerative medicine.
Collapse
Affiliation(s)
- Patrycja Sokolowska
- Chair of Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Poland.
| | | | | | | | | |
Collapse
|
11
|
Carotenuto F, Teodori L, Maccari AM, Delbono L, Orlando G, Di Nardo P. Turning regenerative technologies into treatment to repair myocardial injuries. J Cell Mol Med 2019; 24:2704-2716. [PMID: 31568640 PMCID: PMC7077550 DOI: 10.1111/jcmm.14630] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Regenerative therapies including stem cell treatments hold promise to allow curing patients affected by severe cardiac muscle diseases. However, the clinical efficacy of stem cell therapy remains elusive, so far. The two key roadblocks that still need to be overcome are the poor cell engraftment into the injured myocardium and the limited knowledge of the ideal mixture of bioactive factors to be locally delivered for restoring heart function. Thus, therapeutic strategies for cardiac repair are directed to increase the retention and functional integration of transplanted cells in the damaged myocardium or to enhance the endogenous repair mechanisms through cell‐free therapies. In this context, biomaterial‐based technologies and tissue engineering approaches have the potential to dramatically impact cardiac translational medicine. This review intends to offer some consideration on the cell‐based and cell‐free cardiac therapies, their limitations and the possible future developments.
Collapse
Affiliation(s)
- Felicia Carotenuto
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy.,Diagnostics and Metrology (FSN-TECFIS-DIM), ENEA, C.R. Frascati, Rome, Italy
| | - Laura Teodori
- Diagnostics and Metrology (FSN-TECFIS-DIM), ENEA, C.R. Frascati, Rome, Italy
| | - Anna Maria Maccari
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy
| | - Luciano Delbono
- Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Giuseppe Orlando
- Wake Forest University School of Medicine, Winston Salem, NC, USA.,Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Paolo Di Nardo
- Centro Interdipartimentale di Medicina Rigenerativa, Università di Roma Tor Vergata, Rome, Italy.,Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Rome, Italy.,I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
12
|
Mazini L, Rochette L, Amine M, Malka G. Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). Int J Mol Sci 2019; 20:ijms20102523. [PMID: 31121953 PMCID: PMC6566837 DOI: 10.3390/ijms20102523] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is now on the top one of stem cell sources regarding its accessibility, abundance, and less painful collection procedure when compared to other sources. The adipose derived stem cells (ADSCs) that it contains can be maintained and expanded in culture for long periods of time without losing their differentiation capacity, leading to large cell quantities being increasingly used in cell therapy purposes. Many reports showed that ADSCs-based cell therapy products demonstrated optimal efficacy and efficiency in some clinical indications for both autologous and allogeneic purposes, hence becoming considered as potential tools for replacing, repairing, and regenerating dead or damaged cells. In this review, we analyzed the therapeutic advancement of ADSCs in comparison to bone marrow (BM) and umbilical cord (UC)-mesenchymal stem cells (MSCs) and designed the specific requirements to their best clinical practices and safety. Our analysis was focused on the ADSCs, rather than the whole stromal vascular fraction (SVF) cell populations, to facilitate characterization that is related to their source of origins. Clinical outcomes improvement suggested that these cells hold great promise in stem cell-based therapies in neurodegenerative, cardiovascular, and auto-immunes diseases.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| | - Luc Rochette
- Equipe d'Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Mohamed Amine
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Département de Santé Publique et de Médecine Communautaire, Faculté de Médecine et de Pharmacie, Université Cadi Ayyad, Marrakech 40000, Morocco.
| | - Gabriel Malka
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| |
Collapse
|
13
|
Mazini L, Rochette L, Amine M, Malka G. Regenerative Capacity of Adipose Derived Stem Cells (ADSCs), Comparison with Mesenchymal Stem Cells (MSCs). Int J Mol Sci 2019. [PMID: 31121953 DOI: 10.3390/ijms20102523.pmid:31121953;pmcid:pmc6566837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Adipose tissue is now on the top one of stem cell sources regarding its accessibility, abundance, and less painful collection procedure when compared to other sources. The adipose derived stem cells (ADSCs) that it contains can be maintained and expanded in culture for long periods of time without losing their differentiation capacity, leading to large cell quantities being increasingly used in cell therapy purposes. Many reports showed that ADSCs-based cell therapy products demonstrated optimal efficacy and efficiency in some clinical indications for both autologous and allogeneic purposes, hence becoming considered as potential tools for replacing, repairing, and regenerating dead or damaged cells. In this review, we analyzed the therapeutic advancement of ADSCs in comparison to bone marrow (BM) and umbilical cord (UC)-mesenchymal stem cells (MSCs) and designed the specific requirements to their best clinical practices and safety. Our analysis was focused on the ADSCs, rather than the whole stromal vascular fraction (SVF) cell populations, to facilitate characterization that is related to their source of origins. Clinical outcomes improvement suggested that these cells hold great promise in stem cell-based therapies in neurodegenerative, cardiovascular, and auto-immunes diseases.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| | - Luc Rochette
- Equipe d'Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Université de Bourgogne Franche Comté, Faculté des Sciences de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France.
| | - Mohamed Amine
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Département de Santé Publique et de Médecine Communautaire, Faculté de Médecine et de Pharmacie, Université Cadi Ayyad, Marrakech 40000, Morocco.
| | - Gabriel Malka
- Laboratoire Cellules Souches et Ingénierie Tissulaire, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
- Laboratoire d'Epidémiologie et de Biostatique, Centre Interface Applications Médicales CIAM, Université Mohammed VI polytechnique, Ben Guérir 43150, Morocco.
| |
Collapse
|
14
|
Abstract
There is a critical need to identify accessible stem cells that can form spontaneously beating cardiomyocytes (CMs) and enable regeneration. Here, we establish that intravenous delivery of placental Cdx2 cells resulted in directed homing, sustained engraftment, and differentiation into CMs and vascular cells in damaged hearts, significantly improving cardiac function. This study unveils a distinctive functional significance of Cdx2 beyond its established role in embryonic patterning. Therapeutic use of Cdx2 cells may represent a vital advance, as these cells are multipotent and immunologically naive, with a unique proteome, compared with embryonic stem cells. Moreover, they exhibit the ability to selectively home to sites of injury. These characteristics pave the way for novel allogeneic stem cell therapy for cardiac disease. The extremely limited regenerative potential of adult mammalian hearts has prompted the need for novel cell-based therapies that can restore contractile function in heart disease. We have previously shown the regenerative potential of mixed fetal cells that were naturally found migrating to the injured maternal heart. Exploiting this intrinsic mechanism led to the current hypothesis that Caudal-type homeobox-2 (Cdx2) cells in placenta may represent a novel cell type for cardiac regeneration. Using a lineage-tracing strategy, we specifically labeled fetal-derived Cdx2 cells with enhanced green fluorescent protein (eGFP). Cdx2-eGFP cells from end-gestation placenta were assayed for cardiac differentiation in vitro and in vivo using a mouse model of myocardial infarction. We observed that these cells differentiated into spontaneously beating cardiomyocytes (CMs) and vascular cells in vitro, indicating multipotentiality. When administered via tail vein to infarcted wild-type male mice, they selectively and robustly homed to the heart and differentiated to CMs and blood vessels, resulting in significant improvement in contractility as noted by MRI. Proteomics and immune transcriptomics studies of Cdx2-eGFP cells compared with embryonic stem (ES) cells reveal that they appear to retain “stem”-related functions of ES cells but exhibit unique signatures supporting roles in homing and survival, with an ability to evade immune surveillance, which is critical for cell-based therapy. Cdx2-eGFP cells may potentially represent a therapeutic advance in allogeneic cell therapy for cardiac repair.
Collapse
|
15
|
Schäfer R. Advanced cell therapeutics are changing the clinical landscape: will mesenchymal stromal cells be a part of it? BMC Med 2019; 17:53. [PMID: 30832655 PMCID: PMC6399823 DOI: 10.1186/s12916-019-1289-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/28/2022] Open
Abstract
During the past 15 years there have been dramatic changes in the medical landscape, particularly in oncology and regenerative medicine. Cell therapies have played a substantial part in this progress. Cellular immunotherapies can use immune cells, such as T cells or natural killer cells that, after functional modification ex vivo, exert powerful anti-cancer effects when given to the patient. Innovative technologies, such as re-programming terminally differentiated cells into pluripotent stem cells or into other cell types and applying specific enzymes to more precisely edit the human genome, are paving the way towards more potent cell and gene therapies.Mesenchymal stromal cells are promising cellular immunotherapeutics, which also have potential for use in tissue engineering strategies and other regenerative medicine applications. However, substantial gaps in our knowledge of their biology and therapeutic efficacy present major challenges to their sustainable implementation in the clinical routine.In this article, progress in the field of cell therapeutics during the past 15 years will be briefly discussed, with a focus on mesenchymal stromal cells, highlighting the impact of this field on patient care.
Collapse
Affiliation(s)
- Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital, Sandhofstrasse 1, 60528, Frankfurt am Main, Germany. .,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, 655 W Baltimore St, BRB 14-021, Baltimore, MD, 21201, USA.
| |
Collapse
|
16
|
Amin M, Kushida Y, Wakao S, Kitada M, Tatsumi K, Dezawa M. Cardiotrophic Growth Factor-Driven Induction of Human Muse Cells Into Cardiomyocyte-Like Phenotype. Cell Transplant 2019; 27:285-298. [PMID: 29637816 PMCID: PMC5898685 DOI: 10.1177/0963689717721514] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are endogenous nontumorigenic stem cells collectable as stage-specific embryonic antigen 3 (SSEA-3) + from various organs including the bone marrow and are pluripotent-like. The potential of human bone marrow-derived Muse cells to commit to cardiac lineage cells was evaluated. We found that (1) initial treatment of Muse cells with 5'-azacytidine in suspension culture successfully accelerated demethylation of cardiac marker Nkx2.5 promoter; (2) then transferring the cells onto adherent culture and treatment with early cardiac differentiation factors including wingless-int (Wnt)-3a, bone morphogenetic proteins (BMP)-2/4, and transforming growth factor (TGF) β1; and (3) further treatment with late cardiac differentiation cytokines including cardiotrophin-1 converted Muse cells into cardiomyocyte-like cells that expressed α-actinin and troponin-I with a striation-like pattern. MLC2a expression in the final step suggested differentiation of the cells into an atrial subtype. MLC2v, a marker for a mature ventricular subtype, was expressed when cells were treated with Dickkopf-related protein 1 (DKK-1) and Noggin, inhibitors of Wnt3a and BMP-4, respectively, between steps (2) and (3). None of the steps included exogenous gene transfection, making induced cells feasible for future clinical application.
Collapse
Affiliation(s)
- Mohamed Amin
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,2 Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia, Egypt
| | - Yoshihiro Kushida
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shohei Wakao
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaaki Kitada
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuki Tatsumi
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,3 Life Science Institute Inc., Regenerative Medicine Division, Nagoya, Japan
| | - Mari Dezawa
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
17
|
Miklíková M, Jarkovská D, Čedíková M, Švíglerová J, Kuncová J, Nalos L, Kubíková T, Liška V, Holubová M, Lysák D, Králíčková M, Vištejnová L, Štengl M. Beneficial effects of mesenchymal stem cells on adult porcine cardiomyocytes in non-contact co-culture. Physiol Res 2018; 67:S619-S631. [PMID: 30607969 DOI: 10.33549/physiolres.934051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been reported to improve survival of cardiomyocytes (CMCs) and overall regeneration of cardiac tissue. Despite promising preclinical results, interactions of MSCs and CMCs, both direct and indirect, remain unclear. In this study, porcine bone marrow MSCs and freshly isolated porcine primary adult CMCs were used for non-contact co-culture experiments. Morphology, viability and functional parameters of CMCs were measured over time and compared between CMCs cultured alone and CMCs co-cultured with MSCs. In non-contact co-culture, MSCs improved survival of CMCs. CMCs co-cultured with MSCs maintained CMCs morphology and viability in significantly higher percentage than CMCs cultured alone. In viable CMCs, mitochondrial respiration was preserved in both CMCs cultured alone and in CMCs co-cultured with MSCs. Comparison of cellular contractility and calcium handling, measured in single CMCs, revealed no significant differences between viable CMCs from co-culture and CMCs cultured alone. In conclusion, non-contact co-culture of porcine MSCs and CMCs improved survival of CMCs with a sufficient preservation of functional and mitochondrial parameters.
Collapse
Affiliation(s)
- M Miklíková
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Continuous zebularine treatment enhances hepatic differentiation of mesenchymal stem cells under liver-specific factors induction in vitro. Life Sci 2018; 215:57-63. [PMID: 30473025 DOI: 10.1016/j.lfs.2018.10.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 11/20/2022]
Abstract
AIMS To investigate the effect of zebularine, a stable inhibitor of DNA methylation, on hepatic differentiation of bone marrow-derived mesenchymal stem cells (BM-MSCs) under liver-specific factors induction in vitro. MAIN METHODS BM-MSCs were isolated from the mononuclear cell fraction of rabbit bone marrow samples. The identification of these cells was carried out by immunophenotype analysis. The three hepatic differentiation protocols of BM-MSCs were as follows: liver-specific factors (hepatocyte growth factor and epidermal growth factor) without zebularine, liver-specific factors combined with a 24 h zebularine pre-treatment, and liver-specific factors combined with continuous zebularine treatment. BM-MSCs cultured in basic medium without the differentiation stimuli were set as the control. Morphological features, liver-specific gene and protein expression, and functional analyses were assessed to evaluate hepatic differentiation of BM-MSCs. Global DNA methylation status was tested for investigating the underlying mechanism. KEY FINDINGS Flow cytometry immunophenotyping proved the isolated cells with plastic adherence and a spindle shape were CD29, CD90 positive and CD34, CD45 negative. Albumin (ALB) and alpha-fetoprotein (AFP) messenger RNA and protein expression, glycogen storage and urea production were significantly higher in the continuous zebularine-treated group than the other groups while the differences between the zebularine-untreated group and 24 h zebularine pre-treated group were not significant. Meanwhile, significant decrease of global DNA methylation was observed in the continuous zebularine-treated group. SIGNIFICANCE We conclude that continuous zebularine treatment can improve hepatic differentiation of BM-MSCs under liver-specific factors induction in vitro, and the decrease of global DNA methylation maybe involved in this process.
Collapse
|
19
|
Brychtova M, Thiele JA, Lysak D, Holubova M, Kralickova M, Vistejnova L. Mesenchymal stem cells as the near future of cardiology medicine - truth or wish? Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2018; 163:8-18. [PMID: 30439932 DOI: 10.5507/bp.2018.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/28/2018] [Indexed: 12/31/2022] Open
Abstract
Cardiac damage is one of major cause of worldwide morbidity and mortality. Despite the development in pharmacotherapy, cardiosurgery and interventional cardiology, many patients remain at increased risk of developing adverse cardiac remodeling. An alternative treatment approach is the application of stem cells. Mesenchymal stem cells are among the most promising cell types usable for cardiac regeneration. Their homing to the damaged area, differentiation into cardiomyocytes, paracrine and/or immunomodulatory effect on cardiac tissue was investigated extensively. Despite promising preclinical reports, clinical trials on human patients are not convincing. Meta-analyses of these trials open many questions and show that routine clinical application of mesenchymal stem cells as a cardiac treatment may be not as helpful as expected. This review summarizes contemporary knowledge about mesenchymal stem cells role in cardiac tissue repair and discusses the problems and perspectives of this experimental therapeutical approach.
Collapse
Affiliation(s)
- Michaela Brychtova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Jana-Aletta Thiele
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Daniel Lysak
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Monika Holubova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Milena Kralickova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Lucie Vistejnova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| |
Collapse
|
20
|
Ng WH, Umar Fuaad MZ, Azmi SM, Leong YY, Yong YK, Ng AMH, Tan JJ. Guided evaluation and standardisation of mesenchymal stem cell culture conditions to generate conditioned medium favourable to cardiac c-kit cell growth. Cell Tissue Res 2018; 375:383-396. [PMID: 30232595 DOI: 10.1007/s00441-018-2918-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 09/03/2018] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are known to secrete cardioprotective paracrine factors that can potentially activate endogenous cardiac c-kit cells (CCs). This study aims to optimise MSC growth conditions and medium formulation for generating the conditioned medium (CdM) to facilitate CC growth and expansion in vitro. The quality of MSC-CdM after optimisation of seeding density during MSC stabilisation and medium formulation used during MSC stimulation including glucose, ascorbic acid, serum and oxygen levels and the effects of treatment concentration and repeated CdM harvesting were assessed based on CC viability in vitro under growth factor- and serum-deprived condition. Our data showed that functional CdM can be produced from MSCs with a density of 20,000 cells/cm2, which were stimulated using high glucose (25 mM), ascorbic acid supplemented, serum-free medium under normoxic condition. The generated CdM, when applied to growth factor- and serum-deprived medium at 1:1 ratio, improved CC viability, migration and proliferation in vitro. Such an effect could further be augmented by generating CdM concentrates without compromising CC gene and protein expressions, while retaining its capability to undergo differentiation to form endothelial, smooth muscle and cardiomyocytes. Nevertheless, CdM could not be repeatedly harvested from the same MSC culture, as the protein content and its effect on CC viability deteriorated after the first harvest. In conclusion, this study provides a proof-of-concept strategy to standardise the production of CdM from MSCs based on rapid, stepwise assessment of CC viability, thus enabling production of CdM favourable to CC growth for in vitro or clinical applications.
Collapse
Affiliation(s)
- Wai Hoe Ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Mimi Zulaikha Umar Fuaad
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Siti Maisura Azmi
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Yin Yee Leong
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Angela Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, 56000, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Jun Jie Tan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia.
| |
Collapse
|
21
|
Vaez SA, Ebrahimi-Barough S, Soleimani M, Kolivand S, Farzamfar S, Ahmadi Tafti SH, Azami M, Noorbakhsh F, Ai J. The cardiac niche role in cardiomyocyte differentiation of rat bone marrow-derived stromal cells: comparison between static and microfluidic cell culture methods. EXCLI JOURNAL 2018; 17:762-774. [PMID: 30190666 PMCID: PMC6123612 DOI: 10.17179/excli2018-1539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022]
Abstract
Due to the restricted potential of the heart to regenerate its damaged region, stem cell therapy is a promising treatment modality for myocardial infarction. It has been shown that incubation of bone marrow-derived stromal cells (BMSCs) with initial steps of cardiac differentiation in vitro, can have a significant effect on their therapeutic potential to treat myocardial infarction. Based on these well-established principals we were encouraged to study the direct co-culture of rat BMSCs with neonatal mouse almost pure cardiomyocytes (APCs) and cardiac niche cells (CNCs) in static 2D and microfluidic cell culture systems. Our results showed that the difference regarding the beating rate in isolated APCs and CNCs in both 2D and the microfluidic system was not statistically significant for 30 days. No beat rate could be observed in induced BMSCs in all groups during experiment time. Except for BMSCs cultured alone in both experimental culture conditions, data obtained from Real-time PCR analysis showed that differentiated BMSCs in all co-cultured groups expressed GATA4, Nkx2.5, CX43, cTnI, cTnT, and β-MHC during 4 weeks. BMSCs demonstrated a higher expression of these cardiac factors in microfluidic chips than those co-cultured in 24 well plates. Moreover, immunocytochemistry (ICC), also revealed the GATA4 expression in differentiated BMSCs in all co-cultured groups. It was found that, when combined with shear stress, co-culture with cardiomyocyte can differentiate BMSCs significantly toward cardiomyocyte rather than co-culture alone.
Collapse
Affiliation(s)
- Seyed Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sedighe Kolivand
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Farzamfar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, Faculty of Medicine, Tehran University of Medical, Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|
23
|
Aspirin inhibits growth and enhances cardiomyocyte differentiation of bone marrow mesenchymal stem cells. Eur J Pharmacol 2018; 827:198-207. [PMID: 29551657 DOI: 10.1016/j.ejphar.2018.03.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 11/20/2022]
Abstract
This study aimed to examine the effects of aspirin on the growth and cardiomyocyte differentiation grade of bone marrow mesenchymal stem cells (BMMSCs). BMMSCs were divided into five differentiation groups with different concentrations of aspirin (0 mM, 0.5 mM, 1 mM, 2 mM, or 5 mM), and a undifferentiated control group. Cell growth was measured by cell proliferation, apoptosis assays and DNA cycle analysis. The differentiation grade of BMMSC-derived cardiomyocyte-like cells was examined by measuring the levels of cardiac-specific proteins with cyto-immunofluorescence staining, flow cytometry, and Western blotting. Electrophysiological analyses were performed by patch-clamp experiments and calcium transients were measured by a laser scanning confocal microscope. Cell proliferation decreased as the concentration of aspirin increased. Cell apoptosis increased with increasing aspirin concentration. DNA replication was inhibited in the high dose-aspirin group compared to the low dose- or non-aspirin groups. The number of α-myosin heavy chain (α-MHC) and cardiac troponin I (cTnI) positive cells, cardiac troponin T (cTnT) and connexin 43 (Cx43) positive rates, expression levels of Cx43, Nkx2.5, GATA4 and β1 adrenoceptor increased with increasing aspirin concentration. No sarcomeric cross-striations, spontaneous or induced beating activity or action potentials was observed in each group. Calcium transients were measured in small number cells in 2 mM aspirin group, but the features are atypical. Consequently, aspirin inhibits proliferation and survival of BMMSCs and enhances cardiomyocyte differentiation of BMMSCs.
Collapse
|
24
|
Greenberg JM, Lumbreras V, Pelaez D, Rajguru SM, Cheung HS. Neural Crest Stem Cells Can Differentiate to a Cardiomyogenic Lineage with an Ability to Contract in Response to Pulsed Infrared Stimulation. Tissue Eng Part C Methods 2017; 22:982-990. [PMID: 28192031 DOI: 10.1089/ten.tec.2016.0232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Cellular cardiomyoplasty has rapidly risen to prominence in the clinic following a myocardial infarction; however, low engraftment of transplanted cells limits the therapeutic benefit to these procedures. Recently, lineage-specific stem cells differentiated into cardiomyocytes have gained much attention to assist in the repair of an injured heart tissue; however, questions regarding the ideal cell source remain. In the present study, we have identified a source that is easy to extract stem cells from and show that the cells present have a high plasticity toward the cardiomyogenic lineage. We focused on the recently discovered neural crest stem cells residing in the periodontal ligament that can be easily obtained through dental procedures. MATERIALS AND METHODS Neural crest stem cells were obtained from human excised third molars and differentiated in culture using a protocol for directed differentiation into cardiomyocytes. Differentiation of cells was assessed through gene expression and immunostaining studies. Optical stimulation using pulsed infrared radiation (IR) (λ = 1863 nm) was delivered to cell aggregates to study their contractile ability. RESULTS We show that neural crest stem cells can be differentiated to a cardiomyogenic lineage, which was verified through immunostaining and gene expression. We observed a significant increase in cardiomyocyte-specific markers, NK2 homeobox 5 (NKX2.5) and troponin T type 2 (TNNT2), with positive changes in tropomyosin I (TPM1), gap junction protein alpha 1/Cx43 (GJA1/Cx43), and myocyte enhancement factor 2C (MEF2C). Furthermore, we were able to elicit and maintain pulse-by-pulse contractile responses in the derived cells, including in cardiospheres, with pulsed IR delivered at various radiant energies. The contractility in responses to IR could be maintained at different frequencies (0.25-2 Hz) and up to 10-min durations. While these cells did not maintain their contractility following cessation of IR, these cells demonstrated responses to the optical stimuli that are consistent with previous reports. We also found no evidence for irreversible mitochondrial depolarization in these cells following the long duration of infrared stimulation, suggesting the robustness of these cells. CONCLUSIONS Overall, these results suggest the merit of neural crest-derived stem cells for cardiomyogenic applications and a potential cell source for repair that should contribute to efforts to translate cell-based strategies to the clinic.
Collapse
Affiliation(s)
- Jordan M Greenberg
- 1 Department of Biomedical Engineering, College of Engineering, University of Miami , Coral Gables, Florida
| | - Vicente Lumbreras
- 1 Department of Biomedical Engineering, College of Engineering, University of Miami , Coral Gables, Florida
| | - Daniel Pelaez
- 2 Geriatric Research, Education and Clinical Center (GRECC), Miami Veterans Affairs Medical Center , Miami, Florida
| | - Suhrud M Rajguru
- 1 Department of Biomedical Engineering, College of Engineering, University of Miami , Coral Gables, Florida.,3 Department of Otolaryngology, Miller School of Medicine, University of Miami , Miami, Florida
| | - Herman S Cheung
- 1 Department of Biomedical Engineering, College of Engineering, University of Miami , Coral Gables, Florida.,2 Geriatric Research, Education and Clinical Center (GRECC), Miami Veterans Affairs Medical Center , Miami, Florida
| |
Collapse
|
25
|
Stem cells in cardiovascular diseases: turning bad days into good ones. Drug Discov Today 2017; 22:1730-1739. [DOI: 10.1016/j.drudis.2017.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 06/28/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022]
|
26
|
Unraveling the mechanistic effects of electric field stimulation towards directing stem cell fate and function: A tissue engineering perspective. Biomaterials 2017; 150:60-86. [PMID: 29032331 DOI: 10.1016/j.biomaterials.2017.10.003] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
Electric field (EF) stimulation can play a vital role in eliciting appropriate stem cell response. Such an approach is recently being established to guide stem cell differentiation through osteogenesis/neurogenesis/cardiomyogenesis. Despite significant recent efforts, the biophysical mechanisms by which stem cells sense, interpret and transform electrical cues into biochemical and biological signals still remain unclear. The present review critically analyses the variety of EF stimulation approaches that can be employed to evoke appropriate stem cell response and also makes an attempt to summarize the underlying concepts of this notion, placing special emphasis on stem cell based tissue engineering and regenerative medicine. This review also discusses the major signaling pathways and cellular responses that are elicited by electric stimulation, including the participation of reactive oxygen species and heat shock proteins, modulation of intracellular calcium ion concentration, ATP production and numerous other events involving the clustering or reassembling of cell surface receptors, cytoskeletal remodeling and so on. The specific advantages of using external electric stimulation in different modalities to regulate stem cell fate processes are highlighted with explicit examples, in vitro and in vivo.
Collapse
|
27
|
Mechanisms of stem cell based cardiac repair-gap junctional signaling promotes the cardiac lineage specification of mesenchymal stem cells. Sci Rep 2017; 7:9755. [PMID: 28852100 PMCID: PMC5574972 DOI: 10.1038/s41598-017-10122-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023] Open
Abstract
Different subtypes of bone marrow-derived stem cells are characterized by varying functionality and activity after transplantation into the infarcted heart. Improvement of stem cell therapeutics requires deep knowledge about the mechanisms that mediate the benefits of stem cell treatment. Here, we demonstrated that co-transplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) led to enhanced synergistic effects on cardiac remodeling. While HSCs were associated with blood vessel formation, MSCs were found to possess transdifferentiation capacity. This cardiomyogenic plasticity of MSCs was strongly promoted by a gap junction-dependent crosstalk between myocytes and stem cells. The inhibition of cell-cell coupling significantly reduced the expression of the cardiac specific transcription factors NKX2.5 and GATA4. Interestingly, we observed that small non-coding RNAs are exchanged between MSCs and cardiomyocytes in a GJ-dependent manner that might contribute to the transdifferentiation process of MSCs within a cardiac environment. Our results suggest that the predominant mechanism of HSCs contribution to cardiac regeneration is based on their ability to regulate angiogenesis. In contrast, transplanted MSCs have the capability for intercellular communication with surrounding cardiomyocytes, which triggers the intrinsic program of cardiogenic lineage specification of MSCs by providing cardiomyocyte-derived cues.
Collapse
|
28
|
Kim YH, Cho KA, Park M, Webster JA, Woo SY, Ryu KH. Conditioned medium from tonsil‑derived mesenchymal stem cells promotes adiponectin production. Mol Med Rep 2017; 16:6170-6177. [PMID: 28849047 DOI: 10.3892/mmr.2017.7335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/15/2017] [Indexed: 11/05/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are often considered to be a good source for the development of regenerative medicine. Previously, we reported that tonsil‑derived MSC conditioned medium (T‑MSC CM) produces visceral fat reducing effects. As reduced visceral adiposity is closely associated with an increase in circulating adiponectin, the present study investigated the effects of T‑MSC CM on adiponectin production. T‑MSC CM was collected from previously isolated and characterized T‑MSCs and injected into senescence‑accelerated mouse prone 6 mice, which exhibit characteristics of aging and obesity. The results demonstrated a reduction in mouse weight and epididymal adipose tissue (eAT) mass following injection of T‑MSC CM. Significant increases in adiponectin expression in the eAT, and total and high molecular weight (HMW) adiponectin in the circulation were observed in the T‑MSC CM‑injected mice compared with control mice using reverse transcription‑quantitative polymerase chain reaction, western blot analysis and ELISA. In 3T3‑L1 adipocytes, T‑MSC CM treatment increased adiponectin secretion and multimerization, as detected using western blotting under non‑reducing and non‑heat‑denaturing conditions. Furthermore, glucose oxidase was used to induce oxidative stress in 3T3‑L1 adipocytes and it was observed that T‑MSC CM reduced reactive oxygen species production and the expression of certain oxidative stress markers. In addition, the results also demonstrated that the production of HMW adiponectin was increased, which indicates that T‑MSC CM may enhance adiponectin multimerization via amelioration of oxidative stress. Further studies are required to elucidate anti‑oxidant molecules secreted from T‑MSCs, and these results highlight the potential therapeutic relevance of T‑MSC CM for the treatment of obesity or obesity‑associated diseases.
Collapse
Affiliation(s)
- Yu-Hee Kim
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Kyung-Ah Cho
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Minhwa Park
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Julie A Webster
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - So-Youn Woo
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| |
Collapse
|
29
|
Zeng B, Liu L, Wang S, Dai Z. ILK regulates MSCs survival and angiogenesis partially through AKT and mTOR signaling pathways. Acta Histochem 2017; 119:400-406. [PMID: 28457660 DOI: 10.1016/j.acthis.2017.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 04/18/2017] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSCs) exert therapeutic effects on treating acute myocardial infarction (AMI). Angiogenesis in ischemic heart can promote the supply of oxygen and nutrients to both ischemic myocardium and transplanted stem cells. Focus is then given to the evolving strategies amied at angiogenesis. ILK has been reported to be an important factor regulating apoptosis and angiogenesis. This study examined the role and mechanism of ILK in MSCs survival and angiogenesis. In hypoxic condition, upregulation of ILK expression increased the phosphorylation of Akt and mTOR, resulting in markedly enchanced MSCs survival and VEGF expression; while significantly inhibited MSCs survival and VEGF expression was detected in MSCs with ILK kinase inactivation, which was associated with a reduction of phosphorylation of Akt and mTOR. In addition, it also caused an inhibitory effects of ILK on MSCs survival and VEGF expression, which was abolished by Akt or mTOR inhibitor. Furthermore, it was observed that ILK-overexpressed MSCs increased MSCs survival at 4days and angiogenesis at 3 weeks after transplantation into infracted myocardium as compared with GFP-MSCs group and ILK-SiRNA-MSCs group. This enhanced response was associated with attenuated left ventricular (LV) chamber dilation, reduced LV fibrosis, decreased infarct size and improved LV function. These findings reveal ILK play a pivotal role in regulating MSCs survival and VEGF expression partially through Akt and mTOR signaling pathway. In addition, transplantation of ILK-overexpressed MSCs into infracted myocardium resulted in reduced fibrosis, improved cardiac function and remodeling, which mainly medicated through increased MSCs survival and angiogenesis.
Collapse
Affiliation(s)
- Bin Zeng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China.
| | - Lei Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Shaofeng Wang
- Department of Internal Medicine, Jiangxia District Hospital of Traditional Chinese Medicine, Jiangxia, Hubei, PR China
| | - Zhiguo Dai
- The First People's Hospital of Cardiology, Jinmen, Hubei, PR China
| |
Collapse
|
30
|
Schäfer R, Spohn G, Baer PC. Mesenchymal Stem/Stromal Cells in Regenerative Medicine: Can Preconditioning Strategies Improve Therapeutic Efficacy? Transfus Med Hemother 2016; 43:256-267. [PMID: 27721701 DOI: 10.1159/000447458] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/01/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are becoming increasingly important for the development of cell therapeutics in regenerative medicine. Featuring immunomodulatory potential as well as secreting a variety of trophic factors, MSCs showed remarkable therapeutic effects in numerous preclinical disease models. However, sustainable translation of MSC therapies to the clinic is hampered by heterogeneity of MSCs and non-standardized in vitro culture technologies. Moreover, potent MSC therapeutics require MSCs with maximum regenerative capacity. There is growing evidence that in vitro preconditioning strategies of MSCs can optimize their therapeutic potential. In the following we will discuss achievements and challenges of the development of MSC therapies in regenerative medicine highlighting specific in vitro preconditioning strategies prior to cell transplantation to increase their therapeutic efficacy.
Collapse
Affiliation(s)
- Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital, Frankfurt/M., Germany
| | - Gabriele Spohn
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital, Frankfurt/M., Germany
| | - Patrick C Baer
- Division of Nephrology, Department of Internal Medicine III, Goethe University, Frankfurt/M., Germany
| |
Collapse
|
31
|
Schäfer R, Bieback K. Characterization of mesenchymal stem or stromal cells: tissue sources, heterogeneity, and function. Transfusion 2016; 56:2S-5S. [DOI: 10.1111/trf.13561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Richard Schäfer
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Goethe University Hospital; Frankfurt am Main
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH; Mannheim Germany
| |
Collapse
|
32
|
Overath JM, Gauer S, Obermüller N, Schubert R, Schäfer R, Geiger H, Baer PC. Short-term preconditioning enhances the therapeutic potential of adipose-derived stromal/stem cell-conditioned medium in cisplatin-induced acute kidney injury. Exp Cell Res 2016; 342:175-83. [PMID: 26992633 DOI: 10.1016/j.yexcr.2016.03.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/02/2016] [Accepted: 03/06/2016] [Indexed: 11/17/2022]
Abstract
The development of new strategies to preserve renal function after acute kidney injury (AKI) is necessary due to limited clinical intervention options. The organ-protective effects of mesenchymal stromal/stem cells (MSCs) and their conditioned medium (CM) have been investigated demonstrating that both separately promoted tubular recovery and ameliorated the outcome of AKI. Nevertheless, strategies to optimise the regenerative potential of both are highly needed. Here we investigated the effects of CM from adipose-derived MSCs (ASCs) preincubated in a hypoxic environment (Hyp). Protective factors were investigated by PCR analysis and a protein array in vitro. The expression of 64 of the 308 proteins assayed was found to be more than two-fold increased after Hyp. CM of Hyp-pretreated ASCs (pCM) was used to enhance regeneration in a mouse model of cisplatin-induced AKI (cisAKI). Renal function was assessed by measurements of markers for AKI and serum cytokine levels. The pCM significantly ameliorated serum creatinine and neutrophil gelatinase-associated lipocalin values, and also the levels of inflammatory cytokines IL-1β and IL-6 in the serum of mice with AKI. Our work clearly showed that a Hyp preconditioning significantly increases the release of protective factors in ASCs and enhances the therapeutic effects of CM in cisAKI in mice.
Collapse
Affiliation(s)
- Jürgen M Overath
- Division of Nephrology, Department of Internal Medicine III, Goethe-University, 60590 Frankfurt/M., Germany.
| | - Stefan Gauer
- Division of Nephrology, Department of Internal Medicine III, Goethe-University, 60590 Frankfurt/M., Germany.
| | - Nicholas Obermüller
- Division of Nephrology, Department of Internal Medicine III, Goethe-University, 60590 Frankfurt/M., Germany.
| | - Ralf Schubert
- Department of Paediatrics, Goethe-University, 60590 Frankfurt/M., Germany.
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, Cell Therapeutics & Cell Processing, Goethe University, 60528 Frankfurt/M., Germany.
| | - Helmut Geiger
- Division of Nephrology, Department of Internal Medicine III, Goethe-University, 60590 Frankfurt/M., Germany.
| | - Patrick C Baer
- Division of Nephrology, Department of Internal Medicine III, Goethe-University, 60590 Frankfurt/M., Germany.
| |
Collapse
|
33
|
Sung IY, Son HN, Ullah I, Bharti D, Park JM, Cho YC, Byun JH, Kang YH, Sung SJ, Kim JW, Rho GJ, Park BW. Cardiomyogenic Differentiation of Human Dental Follicle-derived Stem Cells by Suberoylanilide Hydroxamic Acid and Their In Vivo Homing Property. Int J Med Sci 2016; 13:841-852. [PMID: 27877076 PMCID: PMC5118755 DOI: 10.7150/ijms.16573] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/01/2016] [Indexed: 12/29/2022] Open
Abstract
The purpose of the present study was to investigate the in vitro cardiomyogenic differentiation potential of human dental follicle-derived stem cells (DFCs) under the influence of suberoylanilide hydroxamic acid (SAHA), a member of the histone deacetylase inhibitor family, and analyze the in vivo homing capacity of induced cardiomyocytes (iCMs) when transplanted systemically. DFCs from extracted wisdom teeth showed mesenchymal stem cell (MSC) characteristics such as plate adherent growing, expression of MSC markers (CD44, CD90, and CD105), and mesenchymal lineage-specific differentiation potential. Adding SAHA to the culture medium induced the successful in vitro differentiation of DFCs into cardiomyocytes. These iCMs expressed cardiomyogenic markers, including alpha-smooth muscle actin (α-SMA), cardiac muscle troponin T (TNNT2), Desmin, and cardiac muscle alpha actin (ACTC1), at both the mRNA and protein level. For the assessment of homing capacity, PKH26 labeled iCMs were intraperitoneally injected (1×106 cells in 100 µL of PBS) into the experimental mice, and the ratios of PKH26 positive cells to the total number of injected cells, in multiple organs were determined. The calculated homing ratios, 14 days after systemic cell transplantation, were 5.6 ± 1.0%, 3.6 ± 1.1%, and 11.6 ± 2.7% in heart, liver, and kidney respectively. There was no difference in the serum levels of interleukin-2 and interleukin-10 at 14 days after transplantation, between the experimental (iCM injected) and control (no injection or PBS injection) groups. These results demonstrate that DFCs can be an excellent source for cardiomyocyte differentiation and regeneration. Moreover, the iCMs can be delivered into heart muscle via systemic administration without eliciting inflammatory or immune response. This can serve as the pilot study for further investigations into the in vitro cardiomyogenic differentiation potential of DFCs under the influence of SAHA and the in vivo homing capacity of the iCMs into the heart muscle, when injected systemically.
Collapse
Affiliation(s)
- Iel-Yong Sung
- Department of Oral and Maxillofacial Surgery, College of Medicine, Ulsan University, Ulsan, Republic of Korea
| | - Han-Na Son
- Department of Oral and Maxillofacial Surgery, College of Medicine, Ulsan University, Ulsan, Republic of Korea
| | - Imran Ullah
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Dinesh Bharti
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Ju-Mi Park
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Yeong-Cheol Cho
- Department of Oral and Maxillofacial Surgery, College of Medicine, Ulsan University, Ulsan, Republic of Korea
| | - June-Ho Byun
- Department of Dentistry, Gyeongsang National University School of Medicine and Institute of Health Science, Jinju, Republic of Korea
| | - Young-Hoon Kang
- Department of Dentistry, Gyeongsang National University School of Medicine and Institute of Health Science, Jinju, Republic of Korea; Department of Oral and Maxillofacial Surgery, Changwon Gyeongsang National University Hospital, Changwon, Republic of Korea
| | - Su-Jin Sung
- Department of Oral and Maxillofacial Surgery, Changwon Gyeongsang National University Hospital, Changwon, Republic of Korea
| | - Jong-Woo Kim
- Department of Thoracic and Cardiovascular Surgery, Gyeongsang National University School of Medicine and Changwon Gyeongsang National University Hospital, Changwon, Republic of Korea
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Bong-Wook Park
- Department of Dentistry, Gyeongsang National University School of Medicine and Institute of Health Science, Jinju, Republic of Korea; Department of Oral and Maxillofacial Surgery, Changwon Gyeongsang National University Hospital, Changwon, Republic of Korea
| |
Collapse
|
34
|
Scaffold-free and scaffold-assisted 3D culture enhances differentiation of bone marrow stromal cells. In Vitro Cell Dev Biol Anim 2015; 52:204-17. [PMID: 26542170 DOI: 10.1007/s11626-015-9971-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023]
Abstract
3D cultures of stem cells can preserve differentiation potential or increase the efficiency of methods that induce differentiation. Mouse bone marrow-derived stromal cells (BMSCs) were cultured in 3D as scaffold-free spheroids or "mesoid bodies" (MBs) and as aggregates on poly(lactic) acid microspheres (MB/MS). 3D cultures demonstrated viable cells, interaction on multiple planes, altered cell morphology, and the formation of structures similar to epithelial cell bridges. Cell proliferation was limited in suspension cultures of MB and MB/MS; however, cells regained proliferative capacity when transferred to flat substrates of tissue culture plates (TCPs). Expanded as monolayer, cells retained expression of Sca-1 and CD44 stem cell markers. 3D cultures demonstrated enhanced potential for adipogenic and osteogenic differentiation showing higher triglyceride accumulation and robust mineralization in comparison with TCP cultures. Enhanced and efficient adipogenesis was also observed in 3D cultures generated in a rotating cell culture system. Preservation of multilineage potential of BMSC was demonstrated in 5-azacytidine treatment of 3D cultures and TCP by expression of cardiac markers GATA4 and ACTA1 although functioning cardiomyocytes were not derived.
Collapse
|
35
|
Russo V, Omidi E, Samani A, Hamilton A, Flynn LE. Porous, Ventricular Extracellular Matrix-Derived Foams as a Platform for Cardiac Cell Culture. Biores Open Access 2015; 4:374-88. [PMID: 26487982 PMCID: PMC4598938 DOI: 10.1089/biores.2015.0030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To more closely mimic the native cellular microenvironment, 3D scaffolds derived from the extracellular matrix (ECM) are being developed as alternatives to conventional 2D culture systems. In the present study, we established methods to fabricate nonchemically cross-linked 3D porous foams derived entirely from decellularized porcine left ventricle (DLV) for use as an in vitro cardiac cell culture platform. Furthermore, we explored the effects of physically preprocessing the DLV through mechanical mincing versus cryomilling, as well as varying the ECM concentration on the structure, composition, and physical properties of the foams. Our results indicate that the less highly processed minced foams had a more cohesive and complex network of ECM components, enhanced mechanical properties, and improved stability under simulated culturing conditions. To validate the DLV foams, a proof-of-concept study was conducted to explore the early cardiomyogenic differentiation of pericardial fat adipose-derived stem/stromal cells (pfASCs) on the minced DLV foams relative to purified collagen I gel controls. Differentiation was induced using a modified cardiomyogenic medium (MCM) or through stimulation with 5-azacytidine (5-aza), and cardiomyocyte marker expression was characterized by immunohistochemistry and real-time reverse transcriptase-polymerase chain reaction. Our results indicate that early markers of cardiomyogenic differentiation were significantly enhanced on the DLV foams cultured in MCM, suggesting a synergistic effect of the cardiac ECM-derived scaffolds and the culture medium on the induction of pfASC differentiation. Furthermore, in analyzing the response in the noninduced control groups, the foams were observed to provide a mildly inductive microenvironment for pfASC cardiomyogenesis, supporting the rationale for using tissue-specific ECM as a substrate for cardiac cell culture applications.
Collapse
Affiliation(s)
- Valerio Russo
- Department of Chemical Engineering, Queen's University , Kingston, Ontario, Canada . ; Human Mobility Research Centre, Kingston General Hospital , Kingston, Ontario, Canada
| | - Ehsan Omidi
- Biomedical Engineering Graduate Program, The University of Western Ontario , London, Ontario, Canada
| | - Abbas Samani
- Biomedical Engineering Graduate Program, The University of Western Ontario , London, Ontario, Canada . ; Department of Electrical and Computer Engineering, The University of Western Ontario , London, Ontario, Canada
| | - Andrew Hamilton
- Department of Surgery, Kingston General Hospital , Kingston, Ontario, Canada
| | - Lauren E Flynn
- Department of Chemical Engineering, Queen's University , Kingston, Ontario, Canada . ; Biomedical Engineering Graduate Program, The University of Western Ontario , London, Ontario, Canada . ; Department of Chemical and Biochemical Engineering, The University of Western Ontario , London, Ontario, Canada . ; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario , London, Ontario, Canada
| |
Collapse
|
36
|
van der Spoel TIG, Gathier WA, Koudstaal S, van Slochteren F, Of Lorkeers SJ, Sluijter JPG, Hoefer IE, Steendijk P, Cramer MJM, Doevendans PA, van Belle E, Chamuleau SAJ. Autologous Mesenchymal Stem Cells Show More Benefit on Systolic Function Compared to Bone Marrow Mononuclear Cells in a Porcine Model of Chronic Myocardial Infarction. J Cardiovasc Transl Res 2015; 8:393-403. [PMID: 26382088 PMCID: PMC4623074 DOI: 10.1007/s12265-015-9643-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 07/06/2015] [Indexed: 12/15/2022]
Abstract
Cardiac cell therapy is a strategy to treat patients with chronic myocardial infarction (MI). No consensus exists regarding the optimal cell type. First, a comparison between autologous bone marrow-derived mononuclear cells (BMMNC) and mesenchymal stem cells (MSC) on therapeutic efficacy after MI was performed. Next, the effect of repetitive, NOGA-guided transendocardial injection was determined via a crossover design. Nineteen pigs were allocated in three groups: (1) placebo (at 4 and 8 weeks), (2) MSC (followed by placebo at 8 weeks), or (3) BMMNC (followed by MSC at 8 weeks) delivery including a priming strategy to enhance MSC effect. At 4 weeks, ejection fraction (EF) was significantly improved after MSC injection and not by BMMNC injection. After 8 weeks, no difference was observed in EF between cell-treated groups demonstrating the positive systolic effect of MSC. This study showed that MSC rather than BMMNC injection improves systolic function in chronic MI.
Collapse
Affiliation(s)
- T I G van der Spoel
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - W A Gathier
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - S Koudstaal
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - F van Slochteren
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - S Jansen Of Lorkeers
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - J P G Sluijter
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, The Netherlands
| | - I E Hoefer
- Department of Experimental Cardiology, Utrecht, The Netherlands
| | - P Steendijk
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - M J M Cramer
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - P A Doevendans
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, The Netherlands
| | - E van Belle
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - S A J Chamuleau
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Rm E03.511, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
37
|
LeBlon CE, Casey ME, Fodor CR, Zhang T, Zhang X, Jedlicka SS. Correlation between in vitro expansion-related cell stiffening and differentiation potential of human mesenchymal stem cells. Differentiation 2015; 90:1-15. [PMID: 26381795 DOI: 10.1016/j.diff.2015.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/10/2015] [Accepted: 08/20/2015] [Indexed: 12/28/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive cell source for tissue regeneration, given their self-renewal and multilineage potential. However, they are present in only small percentages in human bone marrow, and are generally propagated in vitro prior to downstream use. Previous work has shown that hMSC propagation can lead to alterations in cell behavior and differentiation potency, yet optimization of differentiation based on starting cell elastic modulus is an area still under investigation. To further advance the knowledge in this field, hMSCs were cultured and routinely passaged on tissue-culture polystyrene to investigate the correlation between cell stiffening and differentiation potency during in vitro aging. Local cell elastic modulus was measured at every passage using atomic force microscopy indentation. At each passage, cells were induced to differentiate down myogenic and osteogenic paths. Cells induced to differentiate, as well as undifferentiated cells were assessed for gene and protein expression using quantitative polymerase chain reaction and immunofluorescent staining, respectively, for osteogenic and myogenic markers. Myogenic and osteogenic cell potential are highly reliant on the elastic modulus of the starting cell population (of undifferentiated cells), and this potential appears to peak when the innate cell elastic modulus is close to that of differentiated tissue. However, the latent expression of the same markers in undifferentiated cells also appears to undergo a correlative relationship with cell elastic modulus, indicating some endogenous effects of cell elastic modulus and gene/protein expression. Overall, this study correlates age-related changes with regards to innate cell stiffening and gene/protein expression in commercial hMSCs, providing some guidance as to maintenance and future use of hMSCs in future tissue engineering applications.
Collapse
Affiliation(s)
- Courtney E LeBlon
- Mechanical Engineering & Mechanics, Packard Laboratory, Lehigh University, 19 Memorial Drive, Bethlehem, PA 18015, United States
| | - Meghan E Casey
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Caitlin R Fodor
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Tony Zhang
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Xiaohui Zhang
- Mechanical Engineering & Mechanics, Packard Laboratory, Lehigh University, 19 Memorial Drive, Bethlehem, PA 18015, United States; Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Sabrina S Jedlicka
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States; Materials Science and Engineering, Whitaker Laboratory, Lehigh University, 5 East Packer Ave., Bethlehem, PA 18015, United States; Center for Advanced Materials & Nanotechnology, Whitaker Laboratory, Lehigh University, 5 East Packer Ave., Bethlehem, PA 18015, United States.
| |
Collapse
|
38
|
Ding R, Jiang X, Ha Y, Wang Z, Guo J, Jiang H, Zheng S, Shen Z, Jie W. Activation of Notch1 signalling promotes multi-lineage differentiation of c-Kit(POS)/NKX2.5(POS) bone marrow stem cells: implication in stem cell translational medicine. Stem Cell Res Ther 2015; 6:91. [PMID: 25956503 PMCID: PMC4446115 DOI: 10.1186/s13287-015-0085-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 07/06/2014] [Accepted: 04/29/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction Transplantation of bone marrow mesenchymal stem cells (BMSCs) can repair injured hearts. However, whether BMSC populations contain cells with cardiac stem cell characteristics is ill-defined. We report here that Notch signalling can promote differentiation of c-KitPOS/NKX2.5POS BMSCs into cardiomyocyte-like cells. Methods Total BMSCs were isolated from Sprague–Dawley rat femurs and c-KitPOS cells were purified. c-KitPOS/NKX2.5POS cells were isolated by single-cell cloning, and the presence of cardiomyocyte, smooth muscle cell (SMC), and endothelial cell differentiation markers assessed by immunofluorescence staining and semi-quantitative reverse-transcription polymerase chain reaction (RT-PCR) analysis. Levels of c-Kit and Notch1–4 in total BMSCs and c-KitPOS/NKX2.5POS BMSCs were quantitated by flow cytometry. Following infection with an adenovirus over-expressing Notch1 intracellular domain (NICD), total BMSCs and c-KitPOS/NKX2.5POS cells were assessed for differentiation to cardiomyocyte, SMC, and endothelial cell lineages by immunofluorescence staining and real-time quantitative RT-PCR. Total BMSCs and c-KitPOS/NKX2.5POS cells were treated with the Notch1 ligand Jagged1 and markers of cardiomyocyte, SMC, and endothelial cell differentiation were examined by immunofluorescence staining and real-time quantitative RT-PCR analysis. Results c-KitPOS/NKX2.5POS cells were present among total BMSC populations, and these cells did not express markers of adult cardiomyocyte, SMC, or endothelial cell lineages. c-KitPOS/NKX2.5POS BMSCs exhibited a multi-lineage differentiation potential similar to total BMSCs. Following sorting, the c-Kit level in c-KitPOS/NKX2.5POS BMSCs was 84.4%. Flow cytometry revealed that Notch1 was the predominant Notch receptor present in total BMSCs and c-KitPOS/NKX2.5POS BMSCs. Total BMSCs and c-KitPOS/NKX2.5POS BMSCs overexpressing NICD had active Notch1 signalling accompanied by differentiation into cardiomyocyte, SMC, and endothelial cell lineages. Treatment of total BMSCs and c-KitPOS/NKX2.5POS BMSCs with exogenous Jagged1 activated Notch1 signalling and drove multi-lineage differentiation, with a tendency towards cardiac lineage differentiation in c-KitPOS/NKX2.5POS BMSCs. Conclusions c-KitPOS/NKX2.5POS cells exist in total BMSC pools. Activation of Notch1 signalling contributed to multi-lineage differentiation of c-KitPOS/NKX2.5POS BMSCs, favouring differentiation into cardiomyocytes. These findings suggest that modulation of Notch1 signalling may have potential utility in stem cell translational medicine. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0085-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ranran Ding
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Xiaofan Jiang
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Yanping Ha
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Zhenliang Wang
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Junli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou, 571199, China.
| | - Hanguo Jiang
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Shaojiang Zheng
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou, 571199, China.
| | - Zhihua Shen
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Wei Jie
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
| |
Collapse
|
39
|
Young PP, Schäfer R. Cell-based therapies for cardiac disease: a cellular therapist's perspective. Transfusion 2015; 55:441-51; quiz 440. [PMID: 25145464 PMCID: PMC5771653 DOI: 10.1111/trf.12826] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 12/14/2022]
Abstract
Cell-based therapy is an exciting, promising, and a developing new treatment for cardiac diseases. Stem cell-based therapies have the potential to fundamentally transform the treatment of ischemic cardiac injury and heart failure by achieving what would have been unthinkable only a few years ago-the Holy Grail of myocardial regeneration. Recent therapeutic approaches involve bone marrow (BM)-derived mononuclear cells and their subsets such as mesenchymal stem/stromal cells (MSCs), endothelial progenitor cells as well as adipose tissue-derived MSCs, cardiac tissue-derived stem cells, and cell combinations. Clinical trials employing these cells have demonstrated that cellular therapy is feasible and safe. Regarding delivery methods, the safety of catheter-based, transendocardial and -epicardial stem cell injection has been established. However, the results, while variable, suggest rather modest clinical efficacy overall in both heart failure and ischemic heart disease, such as in acute myocardial infarction. Future studies will focus on determining the most efficacious cell type(s) and/or cell combinations and the most reasonable indications and optimal timing of transplantation, as well as the mechanisms underlying their therapeutic effects. We will review and summarize the clinical trial results to date. In addition, we discuss challenges and operational issues in cell processing for cardiac applications.
Collapse
Affiliation(s)
- Pampee P. Young
- Department of Pathology, Microbiology and Immunology, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Nashville, Tennessee
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Johann-Wolfgang-Goethe-University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
40
|
Fatkhudinov T, Bolshakova G, Arutyunyan I, Elchaninov A, Makarov A, Kananykhina E, Khokhlova O, Murashev A, Glinkina V, Goldshtein D, Sukhikh G. Bone marrow-derived multipotent stromal cells promote myocardial fibrosis and reverse remodeling of the left ventricle. Stem Cells Int 2015; 2015:746873. [PMID: 25685158 PMCID: PMC4320796 DOI: 10.1155/2015/746873] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/28/2014] [Accepted: 12/28/2014] [Indexed: 02/07/2023] Open
Abstract
Cell therapy is increasingly recognized as a beneficial practice in various cardiac conditions, but its fundamentals remain largely unclear. The fates of transplanted multipotent stromal cells in postinfarction cardiac microenvironments are particularly understudied. To address this issue, labeled multipotent stromal cells were infused into rat myocardium at day 30 after myocardial infarction, against the background of postinfarction cardiosclerosis. Therapeutic effects of the transplantation were assessed by an exercise tolerance test. Histological examination at 14 or 30 days after the transplantation was conducted by means of immunostaining and quantitative image analysis. An improvement in the functional status of the cardiovascular system was observed after both the autologous and the allogeneic transplantations. Location of the label-positive cells within the heart was restricted to the affected part of myocardium. The transplanted cells could give rise to fibroblasts or myofibroblasts but not to cardiac myocytes or blood vessel cells. Both types of transplantation positively influenced scarring processes, and no expansion of fibrosis to border myocardium was observed. Left ventricular wall thickening associated with reduced dilatation index was promoted by transplantation of the autologous cells. According to the results, multipotent stromal cell transplantation prevents adverse remodeling and stimulates left ventricular reverse remodeling.
Collapse
Affiliation(s)
- Timur Fatkhudinov
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- 2Scientific Research Institute of Human Morphology, Russian Academy of Medical Sciences, 3 Tsurupa Street, Moscow 117418, Russia
- 3Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 1 Ostrovitianov Street, Moscow 117997, Russia
- *Timur Fatkhudinov:
| | - Galina Bolshakova
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
| | - Irina Arutyunyan
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- 2Scientific Research Institute of Human Morphology, Russian Academy of Medical Sciences, 3 Tsurupa Street, Moscow 117418, Russia
| | - Andrey Elchaninov
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- 2Scientific Research Institute of Human Morphology, Russian Academy of Medical Sciences, 3 Tsurupa Street, Moscow 117418, Russia
- 3Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 1 Ostrovitianov Street, Moscow 117997, Russia
| | - Andrey Makarov
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- 2Scientific Research Institute of Human Morphology, Russian Academy of Medical Sciences, 3 Tsurupa Street, Moscow 117418, Russia
| | - Evgeniya Kananykhina
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
- 2Scientific Research Institute of Human Morphology, Russian Academy of Medical Sciences, 3 Tsurupa Street, Moscow 117418, Russia
| | - Oksana Khokhlova
- 4Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, Pushchino 142290, Russia
| | - Arkady Murashev
- 4Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, Pushchino 142290, Russia
| | - Valeria Glinkina
- 3Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 1 Ostrovitianov Street, Moscow 117997, Russia
| | - Dmitry Goldshtein
- 5Research Centre of Medical Genetics of the Russian Academy of Medical Sciences, 1 Moskvorechie Street, Moscow 115478, Russia
| | - Gennady Sukhikh
- 1Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of the Russian Federation, 4 Oparina Street, Moscow 117997, Russia
| |
Collapse
|
41
|
Hodonsky C, Mundada L, Wang S, Witt R, Raff G, Kaushal S, Si MS. Effects of scaffold material used in cardiovascular surgery on mesenchymal stem cells and cardiac progenitor cells. Ann Thorac Surg 2014; 99:605-11. [PMID: 25497071 DOI: 10.1016/j.athoracsur.2014.08.071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 08/04/2014] [Accepted: 08/15/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polytetrafluoroethylene (PTFE) and porcine small intestinal submucosa (pSIS) are patch materials used in congenital heart surgery. Porcine SIS is an extracellular-matrix scaffold that may interact with stem or progenitor cells. To evaluate this, we determined the in vitro effects of pSIS and PTFE on human bone marrow mesenchymal stromal cells (MSCs) and cardiac progenitor cells (CPCs) in 3 areas; cell proliferation, angiogenic growth-factor production, and differentiation. METHODS Human MSCs and CPCs were seeded onto pSIS and PTFE patches. Cell-seeded patches were cultured and then assessed for cell viability and proliferation and supernatant vascular endothelial growth factor A (VEGFA) levels. Cell proliferation was quantified by MTT assay (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide). Quantitative real-time polymerase chain reaction was performed on cell-seeded scaffolds to determine relative changes in gene expression related to angiogenesis and cardiogenesis. RESULTS The MSCs and CPCs were able to attach and proliferate on pSIS and PTFE. The proliferation rate of each cell type was similar on pSIS. Total RNA isolation was only possible from the cell-seeded pSIS patches. The MSC VEGFA production was increased by pSIS. Porcine SIS promoted an angiogenic gene profile in MSCs and an early cardiogenic profile in CPCs. CONCLUSIONS Both PTFE and pSIS allow for varying degrees of cell proliferation. Porcine SIS elicits different phenotypical responses in MSCs as compared with CPCs, which indicates that pSIS may be a bioactive scaffold that modulates stem cell activation and proliferation. These findings highlight the differences in scaffold material strategies and suggest potential advantages of bioactive approaches.
Collapse
Affiliation(s)
- Chani Hodonsky
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan
| | - Lakshmi Mundada
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan
| | - Shuyun Wang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan
| | - Russell Witt
- Department of Surgery, University of California at Davis Medical Center, Sacramento, California
| | - Gary Raff
- Department of Surgery, University of California at Davis Medical Center, Sacramento, California
| | - Sunjay Kaushal
- Department of Surgery, University Maryland, Baltimore, Maryland
| | - Ming-Sing Si
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
42
|
Pham TLB, Nguyen TT, Van Bui A, Nguyen MT, Van Pham P. Fetal heart extract facilitates the differentiation of human umbilical cord blood-derived mesenchymal stem cells into heart muscle precursor cells. Cytotechnology 2014; 68:645-58. [PMID: 25377264 DOI: 10.1007/s10616-014-9812-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/27/2014] [Indexed: 02/07/2023] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) are a promising stem cell source with the potential to modulate the immune system as well as the capacity to differentiate into osteoblasts, chondrocytes, and adipocytes. In previous publications, UCB-MSCs have been successfully differentiated into cardiomyocytes. This study aimed to improve the efficacy of differentiation of UCB-MSCs into cardiomyocytes by combining 5-azacytidine (Aza) with mouse fetal heart extract (HE) in the induction medium. UCB-MSCs were isolated from umbilical cord blood according to a published protocol. Murine fetal hearts were used to produce fetal HE using a rapid freeze-thaw procedure. MSCs at the 3rd to 5th passage were differentiated into cardiomyocytes in two kinds of induction medium: complete culture medium plus Aza (Aza group) and complete culture medium plus Aza and fetal HE (Aza + HE group). The results showed that the cells in both kinds of induction medium exhibited the phenotype of cardiomyocytes. At the transcriptional level, the cells expressed a number of cardiac muscle-specific genes such as Nkx2.5, Gata 4, Mef2c, HCN2, hBNP, α-Ca, cTnT, Desmin, and β-MHC on day 27 in the Aza group and on day 18 in the Aza + HE group. At the translational level, sarcomic α-actin was expressed on day 27 in the Aza group and day 18 in the Aza + HE group. Although they expressed specific genes and proteins of cardiac muscle cells, the induced cells in both groups did not contract and beat spontaneously. These properties are similar to properties of heart muscle precursor cells in vivo. These results demonstrated that the fetal HE facilitates the differentiation process of human UCB-MSCs into heart muscle precursor cells.
Collapse
Affiliation(s)
- Truc Le-Buu Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Tam Thanh Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Anh Van Bui
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - My Thu Nguyen
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
43
|
Di Felice V, Barone R, Nardone G, Forte G. Cardiac tissue engineering: a reflection after a decade of hurry. Front Physiol 2014; 5:365. [PMID: 25295012 PMCID: PMC4171986 DOI: 10.3389/fphys.2014.00365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Valentina Di Felice
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy
| | - Rosario Barone
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo Palermo, Italy ; Department of Stress Biology, Epigenetics and Biomarkers, Euro-Mediterranean Institute of Science and Technology (IEMEST) Palermo, Italy
| | - Giorgia Nardone
- Integrated Center for Cell Therapy and Regenerative Medicine (ICCT), International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic
| | - Giancarlo Forte
- Integrated Center for Cell Therapy and Regenerative Medicine (ICCT), International Clinical Research Center, St. Anne's University Hospital Brno, Czech Republic
| |
Collapse
|
44
|
Li Z, Zhang Z, Zhao L, Li H, Wang S, Shen Y. Bone marrow mesenchymal stem cells with Nogo-66 receptor gene silencing for repair of spinal cord injury. Neural Regen Res 2014; 9:806-14. [PMID: 25206893 PMCID: PMC4146260 DOI: 10.4103/1673-5374.131595] [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] [Accepted: 02/08/2014] [Indexed: 12/13/2022] Open
Abstract
We hypothesized that RNA interference to silence Nogo-66 receptor gene expression in bone marrow mesenchymal stem cells before transplantation might further improve neurological function in rats with spinal cord transection injury. After 2 weeks, the number of neurons and BrdU-positive cells in the Nogo-66 receptor gene silencing group was higher than in the bone marrow mesenchymal stem cell group, and significantly greater compared with the model group. After 4 weeks, behavioral performance was significantly enhanced in the model group. After 8 weeks, the number of horseradish peroxidase-labeled nerve fibers was higher in the Nogo-66 receptor gene silencing group than in the bone marrow mesenchymal stem cell group, and significantly higher than in the model group. The newly formed nerve fibers and myelinated nerve fibers were detectable in the central transverse plane section in the bone marrow mesenchymal stem cell group and in the Nogo-66 receptor gene silencing group.
Collapse
Affiliation(s)
- Zhiyuan Li
- Department of Joint Orthopedics, Hebei Provincial Xingtai People's Hospital, Xingtai, Hebei Province, China
| | - Zhanxiu Zhang
- Department of Joint Orthopedics, Hebei Provincial Xingtai People's Hospital, Xingtai, Hebei Province, China
| | - Lili Zhao
- Department of Joint Orthopedics, Hebei Provincial Xingtai People's Hospital, Xingtai, Hebei Province, China
| | - Hui Li
- Department of Joint Orthopedics, Hebei Provincial Xingtai People's Hospital, Xingtai, Hebei Province, China
| | - Suxia Wang
- Department of Joint Orthopedics, Hebei Provincial Xingtai People's Hospital, Xingtai, Hebei Province, China
| | - Yong Shen
- Department of Spinal Orthopedics, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
45
|
Zhang Q, Wang H, Yang YJ, Dong QT, Wang TJ, Qian HY, Li N, Wang XM, Jin C. Atorvastatin treatment improves the effects of mesenchymal stem cell transplantation on acute myocardial infarction: the role of the RhoA/ROCK/ERK pathway. Int J Cardiol 2014; 176:670-9. [PMID: 25139321 DOI: 10.1016/j.ijcard.2014.07.071] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 07/03/2014] [Accepted: 07/23/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Statins protect mesenchymal stem cells (MSCs) against the harsh microenvironment and improve the efficacy of MSC transplantation after acute myocardial infarction (AMI); however, the mechanism remains uncertain. Furthermore, the transdifferentiation potential of MSCs in the post-infarct heart remains highly controversial. The RhoA/Rho-associated coiled-coil-forming kinase (ROCK) pathway participates in many aspects of the damaged heart after AMI and related to the "pleiotropic" effects of statins. This study aimed to explore whether atorvastatin (ATV) facilitates the survival and therapeutic efficacy of MSCs via the inhibition of RhoA/ROCK pathway and subsequently its downstream molecular extracellular regulated protein kinase (ERK1/2), and to investigate the transdifferentiation potential of MSCs in vivo. METHODS AND RESULTS Female rats received myocardial injections of male rat MSCs 30 min after AMI. Four weeks after AMI, ATV combined with MSC treatment resulted in improved cardiac function and reduced infarct area. ATV facilitated the MSC survival, as revealed by the increased expression of Y chromosomal genes and the increased number of Y chromosome-positive cells; however, no transdifferentiation markers were observed. ATV inhibited the production of inflammatory cytokines both in vitro and vivo, accompanied by suppression of ROCK and ERK activities. Geranylgeranyl pyrophosphate (GGPP) abrogated the effects of ATV in the H9c2 cells under hypoxia/serum deprivation (H/SD), while the ROCK inhibitor fasudil mimicked the benefits of ATV after AMI. CONCLUSIONS ATV improves the post-infarct microenvironment via RhoA/ROCK/ERK inhibition and thus facilitates the survival and efficacy of implanted MSCs. Transdifferentiation may be not responsible for the cardiac benefits that follow MSC transplantation.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Hong Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China.
| | - Qiu-Ting Dong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Tian-Jie Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Hai-Yan Qian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Na Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Xi-Mei Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, PR China
| |
Collapse
|
46
|
Altmann P, Mildner M, Haider T, Traxler D, Beer L, Ristl R, Golabi B, Gabriel C, Leutmezer F, Ankersmit HJ. Secretomes of apoptotic mononuclear cells ameliorate neurological damage in rats with focal ischemia. F1000Res 2014; 3:131. [PMID: 25383184 PMCID: PMC4215751 DOI: 10.12688/f1000research.4219.2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2014] [Indexed: 12/15/2022] Open
Abstract
The pursuit of targeting multiple pathways in the ischemic cascade of cerebral stroke is a promising treatment option. We examined the regenerative potential of conditioned medium derived from rat and human apoptotic mononuclear cells (MNC), rMNC
apo sec and hMNC
apo sec, in experimental stroke. We performed middle cerebral artery occlusion on Wistar rats and administered apoptotic MNC-secretomes intraperitoneally in two experimental settings. Ischemic lesion volumes were determined 48 hours after cerebral ischemia. Neurological evaluations were performed after 6, 24 and 48 hours. Immunoblots were conducted to analyze neuroprotective signal-transduction in human primary glia cells and neurons. Neuronal sprouting assays were performed and neurotrophic factors in both hMNC
apo sec and rat plasma were quantified using ELISA. Administration of rat as well as human apoptotic MNC-secretomes significantly reduced ischemic lesion volumes by 36% and 37%, respectively. Neurological examinations revealed improvement after stroke in both treatment groups. Co-incubation of human astrocytes, Schwann cells and neurons with hMNC
apo sec resulted in activation of several signaling cascades associated with the regulation of cytoprotective gene products and enhanced neuronal sprouting
in vitro. Analysis of neurotrophic factors in hMNC
apo sec and rat plasma revealed high levels of brain derived neurotrophic factor (BDNF). Our data indicate that apoptotic MNC-secretomes elicit neuroprotective effects on rats that have undergone ischemic stroke.
Collapse
Affiliation(s)
- Patrick Altmann
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria ; Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, 1090, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| | - Thomas Haider
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria ; Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, 1090, Austria
| | - Denise Traxler
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria ; Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, 1090, Austria
| | - Lucian Beer
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria
| | - Robin Ristl
- Section for Medical Statistics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, 1090, Austria
| | - Bahar Golabi
- Department of Dermatology, Medical University of Vienna, Vienna, 1090, Austria
| | - Christian Gabriel
- Red Cross Transfusion Service for Upper Austria, Linz, 4017, Austria
| | - Fritz Leutmezer
- Department of Neurology, Medical University of Vienna, Vienna, 1090, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, 1090, Austria ; Christian Doppler Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, Vienna, 1090, Austria
| |
Collapse
|
47
|
Injection of mesenchymal stromal cells into a mechanically stimulated in vitro model of cardiac fibrosis has paracrine effects on resident fibroblasts. Cytotherapy 2014; 16:906-14. [PMID: 24713331 DOI: 10.1016/j.jcyt.2014.01.416] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 12/18/2013] [Accepted: 01/31/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND AIMS Myocardial infarction results in the formation of scar tissue populated by myofibroblasts, a phenotype characterized by increased contractility and matrix deposition. Mesenchymal stromal cells (MSC) delivered to the myocardium can attenuate scar growth and restore cardiac function, though the mechanism is unclear. METHODS This study describes a simple yet robust three-dimensional (3D) in vitro co-culture model to examine the paracrine effects of implanted MSC on resident myofibroblasts in a controlled biochemical and mechanical environment. The fibrosis model consisted of fibroblasts embedded in a 3D collagen gel cultured under defined oxygen tensions and exposed to either cyclic strain or interstitial fluid flow. MSC were injected into this model, and the effect on fibroblast phenotype was evaluated 48 h after cell injection. RESULTS Analysis of gene and protein expression of the fibroblasts indicated that injection of MSC attenuated the myofibroblast transition in response to reduced oxygen and mechanical stress. Assessment of vascular endothelial growth factor and insulin-like growth factor-1 levels demonstrated that their release by fibroblasts was markedly upregulated in hypoxic conditions but attenuated by strain or fluid flow. In fibroblast-MSC co-cultures, vascular endothelial growth factor levels were increased by hypoxia but not affected by mechanical stimuli, whereas insulin-like growth factor-1 levels were generally low and not affected by experimental conditions. CONCLUSIONS This study demonstrates how a 3D in vitro model of the cardiac scar can be used to examine paracrine effects of MSC on the phenotype of resident fibroblasts and therefore illuminates the role of injected progenitor cells on the progression of cardiac fibrosis.
Collapse
|
48
|
Kota DJ, DiCarlo B, Hetz RA, Smith P, Cox CS, Olson SD. Differential MSC activation leads to distinct mononuclear leukocyte binding mechanisms. Sci Rep 2014; 4:4565. [PMID: 24691433 PMCID: PMC3972508 DOI: 10.1038/srep04565] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/13/2014] [Indexed: 02/06/2023] Open
Abstract
Advances in the field of Multipotent Mesenchymal Stromal cell (MSC) biology have demonstrated that MSCs can improve disease outcome when ‘activated' to exert immunomodulatory effects. However, the precise mechanisms modulating MSC-immune cells interactions remain largely elusive. In here, we activated MSC based on a recent polarization paradigm, in which MSCs can be polarized towards a pro- or anti-inflammatory phenotype depending on the Toll-like receptor stimulated, to dissect the mechanisms through which MSCs physically interact with and modulate leukocytes in this context. Our data show that MSCs activated through the Toll-like receptor (TLR) 4 pathway increased VCAM-1 and ICAM-1 dependent binding of leukocytes. On the other hand, TLR3 stimulation strongly increases leukocytes affinity to MSC comparatively, through the formation of cable-like hyaluronic acid structures. In addition, TLR4 activation elicited secretion of pro-inflammatory mediators by MSCs, whereas TLR3-activated MSCs displayed a milder pro-inflammatory phenotype, similar to inactivated MSCs. However, the differently activated MSCs maintained their ability to suppress leukocyte activation at similar levels in our in vitro model, and this immunomodulatory property was shown here to be partially mediated by prostaglandin. These results reinforce the concept that alternate activation profiles control MSC responses and may impact the therapeutic use of MSCs.
Collapse
Affiliation(s)
- Daniel J Kota
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin St., MSB 5.233, Houston, TX, USA 77030
| | - Bryan DiCarlo
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin St., MSB 5.233, Houston, TX, USA 77030
| | - Robert A Hetz
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin St., MSB 5.233, Houston, TX, USA 77030
| | - Philippa Smith
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin St., MSB 5.233, Houston, TX, USA 77030
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin St., MSB 5.233, Houston, TX, USA 77030
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Medical School at Houston, 6431 Fannin St., MSB 5.233, Houston, TX, USA 77030
| |
Collapse
|
49
|
Mesenchymal stem cells for cardiac therapy: practical challenges and potential mechanisms. Stem Cell Rev Rep 2014; 9:254-65. [PMID: 22577007 DOI: 10.1007/s12015-012-9375-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell based treatments for myocardial infarction have demonstrated efficacy in the laboratory and in phase I clinical trials, but the understanding of such therapies remains incomplete. Mesenchymal stem cells (MSCs) are classically defined as maintaining the ability to generate mesenchyme-derived cell types, namely adipocytes, chondrocytes and osteocytes. Recent evidence suggests these cells may in fact harbor much greater potency than originally realized, as several groups have found that MSCs can form cardiac lineage cells in vitro. Additionally, experimental coculture of MSCs with cardiomyocytes appears to improve contractile function of the latter. Bolstered by such findings, several clinical trials have begun to test MSC transplantation for improving post-infarct cardiac function in human patients. The results of these trials have been mixed, underscoring the need to develop a deeper understanding of the underlying stem cell biology. To help synthesize the breadth of studies on the topic, this paper discusses current challenges in the field of MSC cellular therapies for cardiac repair, including methods of cell delivery and the identification of molecular markers that accurately specify the therapeutically relevant mesenchymal cell types. The various possible mechanisms of MSC mediated cardiac improvement, including somatic reprogramming, transdifferentiation, paracrine signaling, and direct electrophysiological coupling are also reviewed. Finally, we consider the traditional cell culture microenvironment, and the promise of cardiac tissue engineering to provide biomimetic in vitro model systems to more faithfully investigate MSC biology, helping to safely and effectively translate exciting discoveries in the laboratory to meaningful therapies in the clinic.
Collapse
|
50
|
Martinez EC, Vu DT, Wang J, Lilyanna S, Ling LH, Gan SU, Tan AL, Phan TT, Lee CN, Kofidis T. Grafts enriched with subamnion-cord-lining mesenchymal stem cell angiogenic spheroids induce post-ischemic myocardial revascularization and preserve cardiac function in failing rat hearts. Stem Cells Dev 2013; 22:3087-99. [PMID: 23869939 DOI: 10.1089/scd.2013.0119] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A crucial question in post-ischemic cell therapy refers to the ideal method of cell delivery to the heart. We hypothesized that epicardial implantation of subamnion-cord-lining mesenchymal stem cells (CL-MSC) angiogenic spheroids embedded within fibrin grafts (SASG) facilitates donor cell survival and enhances cardiac function in failing rat hearts. Furthermore, we compared the efficacy of this approach applied through two delivery methods. Spheroids made of 1.5×10(4) human CL-MSC coated with 2×10(3) human umbilical vein endothelial cells were self-assembled in hanging drops. SASG were constructed by embedding 150 spheroids in fibrin matrix. Except for untreated rats (MI, n=8), grafts were implanted 2 weeks after myocardial infarction upon confirmation of ensued heart failure through thoracotomy: SASG (n=8) and fibrin graft (FG, n=8); or video-assisted thoracoscopic surgery (VATS): SASG-VATS (n=8) and FG-VATS (n=7). In vivo CL-MSC survival was comparable between both SASG-treated groups throughout the study. SASG and SASG-VATS animals had decreased left ventricular end-diastolic pressure relative to untreated animals, and increased fractional shortening compared to MI and FG controls, 4 weeks after treatment. A 14.1% and 6.2% enhancement in ejection fraction from week 2 to 6 after injury was observed in SASG/SASG-VATS, paralleled by improvement in cardiac output. Treated hearts had smaller scar size, and more blood vessels than MI, while donor CL-MSC contributed to arteriogenesis within the graft and infarct areas. Taken together, our data suggest that SASG treatment has the potential to restore failing hearts by preserving cardiac function and inducing myocardial revascularization, while attenuating cardiac fibrosis. Furthermore, we introduce a method for minimally invasive in situ graft assembly.
Collapse
Affiliation(s)
- Eliana C Martinez
- 1 Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|