1
|
Schiffer M, Wagner K, Carls E, Nicke J, Hesse M, Fratila RM, Hildebrand S, Eberbeck D, Mohr T, Mohammadi MM, de la Fuente JM, Fleischmann BK, Roell W. Nanoparticle-assisted targeting of heart lesions with cardiac myofibroblasts: Combined gene and cell therapy. Theranostics 2025; 15:4287-4307. [PMID: 40225585 PMCID: PMC11984384 DOI: 10.7150/thno.103816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/10/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: The cardiac scar is an area rich in collagen. It is populated by myofibroblasts and lacks Connexin 43 expressing cardiomyocytes. Myocardial infarctions have so far proven little amenable to gene- and cell-based therapeutic interventions. Our aim was to establish an experimental approach with translational potential for effective cell-based gene therapy of the cardiac scar. Methods: We have developed a targeting strategy for myocardial infarctions by grafting ex vivo lentivirus-transduced and magnetic nanoparticle-loaded embryonic cardiac myofibroblasts into mouse hearts with magnetic steering. Results: Our approach yielded highly efficient targeting and cell grafting into the cardiac scar. Engraftment rates of myofibroblasts proved very high (30% of injected cells) due to cell proliferation and a low apoptosis rate. We also demonstrate that grafting lentivirus-transduced Connexin 43 overexpressing myofibroblasts into the lesion resulted in increased Connexin 43 protein content and strong protection against ventricular arrhythmias in vivo, as their incidence was reduced by ~ 50% at 2- and 8 weeks after myocardial infarction. Conclusion: The combination of ex vivo gene and in vivo cell therapy, along with magnetic steering of cardiac myofibroblasts, enables, efficient targeting of the cardiac scar and can even modulate its functional properties.
Collapse
Affiliation(s)
- Miriam Schiffer
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Cardiac Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kevin Wagner
- Department of Cardiac Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Esther Carls
- Department of Cardiac Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Julia Nicke
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Michael Hesse
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Raluca M. Fratila
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), Campus Río Ebro, Edificio CIRCE, despacho 00.070 C/ Mariano Esquillor, s/n - 50018 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 Madrid, Spain
| | - Staffan Hildebrand
- Institute of Pharmacology and Toxicology, Medical Faculty, University of Bonn, Biomedizinisches Zentrum, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Dietmar Eberbeck
- Physikalisch-Technische Bundesanstalt (PTB), Abbestraße 2-12, 10587 Berlin, Germany
| | - Timo Mohr
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Cardiac Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Mona Malek Mohammadi
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jesus Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), Campus Río Ebro, Edificio CIRCE, despacho 00.070 C/ Mariano Esquillor, s/n - 50018 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029 Madrid, Spain
| | - Bernd K. Fleischmann
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Wilhelm Roell
- Department of Cardiac Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
2
|
Castillo-Casas JM, Caño-Carrillo S, Sánchez-Fernández C, Franco D, Lozano-Velasco E. Comparative Analysis of Heart Regeneration: Searching for the Key to Heal the Heart-Part II: Molecular Mechanisms of Cardiac Regeneration. J Cardiovasc Dev Dis 2023; 10:357. [PMID: 37754786 PMCID: PMC10531542 DOI: 10.3390/jcdd10090357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, among which ischemic heart disease is the most representative. Myocardial infarction results from occlusion of a coronary artery, which leads to an insufficient blood supply to the myocardium. As it is well known, the massive loss of cardiomyocytes cannot be solved due the limited regenerative ability of the adult mammalian hearts. In contrast, some lower vertebrate species can regenerate the heart after an injury; their study has disclosed some of the involved cell types, molecular mechanisms and signaling pathways during the regenerative process. In this 'two parts' review, we discuss the current state-of-the-art of the main response to achieve heart regeneration, where several processes are involved and essential for cardiac regeneration.
Collapse
Affiliation(s)
- Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Cristina Sánchez-Fernández
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| |
Collapse
|
3
|
Barnes AM, Holmstoen TB, Bonham AJ, Rowland TJ. Differentiating Human Pluripotent Stem Cells to Cardiomyocytes Using Purified Extracellular Matrix Proteins. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120720. [PMID: 36550926 PMCID: PMC9774171 DOI: 10.3390/bioengineering9120720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) can be differentiated into cardiomyocytes (hESC-CMs and iPSC-CMs, respectively), which hold great promise for cardiac regenerative medicine and disease modeling efforts. However, the most widely employed differentiation protocols require undefined substrates that are derived from xenogeneic (animal) products, contaminating resultant hESC- and iPSC-CM cultures with xenogeneic proteins and limiting their clinical applicability. Additionally, typical hESC- and iPSC-CM protocols produce CMs that are significantly contaminated by non-CMs and that are immature, requiring lengthy maturation procedures. In this review, we will summarize recent studies that have investigated the ability of purified extracellular matrix (ECM) proteins to support hESC- and iPSC-CM differentiation, with a focus on commercially available ECM proteins and coatings to make such protocols widely available to researchers. The most promising of the substrates reviewed here include laminin-521 with laminin-221 together or Synthemax (a synthetic vitronectin-based peptide coating), which both resulted in highly pure CM cultures. Future efforts are needed to determine whether combinations of specific purified ECM proteins or derived peptides could further improve CM maturation and culture times, and significantly improve hESC- and iPSC-CM differentiation protocols.
Collapse
Affiliation(s)
- Ashlynn M. Barnes
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Tessa B. Holmstoen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Andrew J. Bonham
- Department of Chemistry & Biochemistry, Metropolitan State University of Denver, Denver, CO 80217, USA
| | - Teisha J. Rowland
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
- Correspondence:
| |
Collapse
|
4
|
Kinases of the Focal Adhesion Complex Contribute to Cardiomyocyte Specification. Int J Mol Sci 2021; 22:ijms221910430. [PMID: 34638793 PMCID: PMC8508671 DOI: 10.3390/ijms221910430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/03/2021] [Accepted: 09/22/2021] [Indexed: 11/17/2022] Open
Abstract
Differentiation of pluripotent stem cells to cardiomyocytes is influenced by culture conditions including the extracellular matrices or similar synthetic scaffolds on which they are grown. However, the molecular mechanisms that link the scaffold with differentiation outcomes are not fully known. Here, we determined by immunofluorescence staining and mass spectrometry approaches that extracellular matrix (ECM) engagement by mouse pluripotent stem cells activates critical components of canonical wingless/integrated (Wnt) signaling pathways via kinases of the focal adhesion to drive cardiomyogenesis. These kinases were found to be differentially activated depending on type of ECM engaged. These outcomes begin to explain how varied ECM composition of in vivo tissues with development and in vitro model systems gives rise to different mature cell types, having broad practical applicability for the design of engineered tissues.
Collapse
|
5
|
Schussler O, Chachques JC, Alifano M, Lecarpentier Y. Key Roles of RGD-Recognizing Integrins During Cardiac Development, on Cardiac Cells, and After Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:179-203. [PMID: 34342855 DOI: 10.1007/s12265-021-10154-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Cardiac cells interact with the extracellular matrix (ECM) proteins through integrin mechanoreceptors that control many cellular events such as cell survival, apoptosis, differentiation, migration, and proliferation. Integrins play a crucial role in cardiac development as well as in cardiac fibrosis and hypertrophy. Integrins recognize oligopeptides present on ECM proteins and are involved in three main types of interaction, namely with collagen, laminin, and the oligopeptide RGD (Arg-Gly-Asp) present on vitronectin and fibronectin proteins. To date, the specific role of integrins recognizing the RGD has not been addressed. In this review, we examine their role during cardiac development, their role on cardiac cells, and their upregulation during pathological processes such as heart fibrosis and hypertrophy. We also examine their role in regenerative and angiogenic processes after myocardial infarction (MI) in the peri-infarct area. Specific targeting of these integrins may be a way of controlling some of these pathological events and thereby improving medical outcomes.
Collapse
Affiliation(s)
- Olivier Schussler
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.
| | - Juan C Chachques
- Department of Cardiac Surgery Pompidou Hospital, Laboratory of Biosurgical Research, Carpentier Foundation, University Paris Descartes, 75015, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.,INSERM U1138 Team "Cancer, Immune Control, and Escape", Cordeliers Research Center, University of Paris, Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
6
|
Wysoczynski M, Bolli R. A realistic appraisal of the use of embryonic stem cell-based therapies for cardiac repair. Eur Heart J 2021; 41:2397-2404. [PMID: 31778154 DOI: 10.1093/eurheartj/ehz787] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/06/2019] [Accepted: 10/25/2019] [Indexed: 12/22/2022] Open
Abstract
Despite the well-documented capacity of embryonic stem cells (ESCs) to differentiate into cardiomyocytes, transplantation of ESCs or ESC-derived cells is plagued by several formidable problems, including graft rejection, arrhythmias, and potential risk of teratomas. Life-long immunosuppression is a disease in itself. Transplantation of human ESC-derived cells in primates causes life-threatening arrhythmias, and the doses used to show efficacy are not clinically relevant. In contemporary clinical research, the margin of tolerance for such catastrophic effects as malignancies is zero, and although the probability of tumours can be reduced by ESC differentiation, it is unlikely to be completely eliminated, particularly when billions of cells are injected. Although ESCs and ESC-derived cells were touted as capable of long-term regeneration, these cells disappear rapidly after transplantation and there is no evidence of long-term engraftment, let alone regeneration. There is, however, mounting evidence that they act via paracrine mechanisms-just like adult cells. To date, no controlled clinical trial of ESC-derived cells in cardiovascular disease has been conducted or even initiated. In contrast, adult cells have been used in thousands of patients with heart disease, with no significant adverse effects and with results that were sufficiently encouraging to warrant Phase II and III trials. Furthermore, induced pluripotent stem cells offer pluripotency similar to ESCs without the need for lifelong immunosuppression. After two decades, the promise that ESC-derived cells would regenerate dead myocardium has not been fulfilled. The most reasonable interpretation of current data is that ESC-based therapies are not likely to have clinical application for heart disease.
Collapse
Affiliation(s)
- Marcin Wysoczynski
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| | - Roberto Bolli
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
7
|
Cell surface markers for immunophenotyping human pluripotent stem cell-derived cardiomyocytes. Pflugers Arch 2021; 473:1023-1039. [PMID: 33928456 DOI: 10.1007/s00424-021-02549-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 02/08/2023]
Abstract
Human pluripotent stem cells (hPSC) self-renew and represent a potentially unlimited source for the production of cardiomyocytes (CMs) suitable for studies of human cardiac development, drug discovery, cardiotoxicity testing, and disease modelling and for cell-based therapies. However, most cardiac differentiation protocols yield mixed cultures of atrial-, ventricular-, and pacemaker-like cells at various stages of development, as well as non-CMs. The proportions and maturation states of these cell types result from disparities among differentiation protocols and time of cultivation, as well as hPSC reprogramming inconsistencies and genetic background variations. The reproducible use of hPSC-CMs for research and therapy is therefore limited by issues of cell population heterogeneity and functional states of maturation. A validated method that overcomes issues of cell heterogeneity is immunophenotyping coupled with live cell sorting, an approach that relies on accessible surface markers restricted to the desired cell type(s). Here we review current progress in unravelling heterogeneity in hPSC-cardiac cultures and in the identification of surface markers suitable for defining cardiac identity, subtype specificity, and maturation states.
Collapse
|
8
|
Cruz-Samperio R, Jordan M, Perriman A. Cell augmentation strategies for cardiac stem cell therapies. Stem Cells Transl Med 2021; 10:855-866. [PMID: 33660953 PMCID: PMC8133336 DOI: 10.1002/sctm.20-0489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) has been the primary cause of death in developed countries, resulting in a major psychological and financial burden for society. Current treatments for acute MI are directed toward rapid restoration of perfusion to limit damage to the myocardium, rather than promoting tissue regeneration and subsequent contractile function recovery. Regenerative cell therapies (CTs), in particular those using multipotent stem cells (SCs), are in the spotlight for treatment post‐MI. Unfortunately, the efficacy of CTs is somewhat limited by their poor long‐term viability, homing, and engraftment to the myocardium. In response, a range of novel SC‐based technologies are in development to provide additional cellular modalities, bringing CTs a step closer to the clinic. In this review, the current landscape of emerging CTs and their augmentation strategies for the treatment post‐MI are discussed. In doing so, we highlight recent advances in cell membrane reengineering via genetic modifications, recombinant protein immobilization, and the utilization of soft biomimetic scaffold interfaces.
Collapse
Affiliation(s)
| | - Millie Jordan
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Adam Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
9
|
Leitolis A, Robert AW, Pereira IT, Correa A, Stimamiglio MA. Cardiomyogenesis Modeling Using Pluripotent Stem Cells: The Role of Microenvironmental Signaling. Front Cell Dev Biol 2019; 7:164. [PMID: 31448277 PMCID: PMC6695570 DOI: 10.3389/fcell.2019.00164] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
Pluripotent stem cells (PSC) can be used as a model to study cardiomyogenic differentiation. In vitro modeling can reproduce cardiac development through modulation of some key signaling pathways. Therefore, many studies make use of this strategy to better understand cardiomyogenesis complexity and to determine possible ways to modulate cell fate. However, challenges remain regarding efficiency of differentiation protocols, cardiomyocyte (CM) maturation and therapeutic applications. Considering that the extracellular milieu is crucial for cellular behavior control, cardiac niche studies, such as those identifying secreted molecules from adult or neonatal tissues, allow the identification of extracellular factors that may contribute to CM differentiation and maturation. This review will focus on cardiomyogenesis modeling using PSC and the elements involved in cardiac microenvironmental signaling (the secretome - extracellular vesicles, extracellular matrix and soluble factors) that may contribute to CM specification and maturation.
Collapse
Affiliation(s)
- Amanda Leitolis
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Anny W Robert
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Isabela T Pereira
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Alejandro Correa
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| | - Marco A Stimamiglio
- Stem Cell Basic Biology Laboratory, Carlos Chagas Institute, FIOCRUZ-PR, Curitiba, Brazil
| |
Collapse
|
10
|
Zhang C, Wang W, He W, Xi N, Wang Y, Liu L. Dynamic Model for Characterizing Contractile Behaviors and Mechanical Properties of a Cardiomyocyte. Biophys J 2018; 114:188-200. [PMID: 29320686 PMCID: PMC5773758 DOI: 10.1016/j.bpj.2017.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 01/27/2023] Open
Abstract
Studies on the contractile dynamics of heart cells have attracted broad attention for the development of both heart disease therapies and cardiomyocyte-actuated micro-robotics. In this study, a linear dynamic model of a single cardiomyocyte cell was proposed at the subcellular scale to characterize the contractile behaviors of heart cells, with system parameters representing the mechanical properties of the subcellular components of living cardiomyocytes. The system parameters of the dynamic model were identified with the cellular beating pattern measured by a scanning ion conductance microscope. The experiments were implemented with cardiomyocytes in one control group and two experimental groups with the drugs cytochalasin-D or nocodazole, to identify the system parameters of the model based on scanning ion conductance microscope measurements, measurement of the cellular Young's modulus with atomic force microscopy indentation, measurement of cellular contraction forces using the micro-pillar technique, and immunofluorescence staining and imaging of the cytoskeleton. The proposed mathematical model was both indirectly and qualitatively verified by the variation in cytoskeleton, beating amplitude, and contractility of cardiomyocytes among the control and the experimental groups, as well as directly and quantitatively validated by the simulation and the significant consistency of 90.5% in the comparison between the ratios of the Young's modulus and the equivalent comprehensive cellular elasticities of cells in the experimental groups to those in the control group. Apart from mechanical properties (mass, elasticity, and viscosity) of subcellular structures, other properties of cardiomyocytes have also been studied, such as the properties of the relative action potential pattern and cellular beating frequency. This work has potential implications for research on cytobiology, drug screening, mechanisms of the heart, and cardiomyocyte-based bio-syncretic robotics.
Collapse
Affiliation(s)
- Chuang Zhang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wenxue Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China.
| | - Wenhui He
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ning Xi
- Emerging Technologies Institute, Department of Industrial and Manufacturing Systems Engineering, University of Hong Kong Pokfulam, Pokfulam, Hong Kong
| | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China.
| |
Collapse
|
11
|
Li Y, Tian S, Lei I, Liu L, Ma P, Wang Z. Transplantation of multipotent Isl1+ cardiac progenitor cells preserves infarcted heart function in mice. Am J Transl Res 2017; 9:1530-1542. [PMID: 28386378 PMCID: PMC5376043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/18/2017] [Indexed: 06/07/2023]
Abstract
Cell-based cardiac therapy is a promising therapeutic strategy to restore heart function after myocardial infarction (MI). However, the cell type selection and ensuing effects remain controversial. Here, we intramyocardially injected Isl1+ cardiac progenitor cells (CPCs) derived from EGFP/luciferase double-tagged mouse embryonic stem (dt-mES) cells with vehicle (fibrin gel) or phosphate-buffered saline (PBS) into the infarcted area in nude mice to assess the contribution of CPCs to the recovery of cardiac function post-MI. Our results showed that Isl1+ CPCs differentiated normally into three cardiac lineages (cardiomyocytes (CMs), endothelial cells and smooth muscle cells) both on cell culture plates and in fibrin gel. Cell retention was significantly increased when the transplanted cells were injected with vehicle. Importantly, 28 days after injection, CPCs were observed to differentiate into CMs within the infarcted area. Moreover, numerous CD31+ endothelial cells derived from endogenous revascularization and differentiation of the injected CPCs were detected. SMMHC-, Ki67- and CX-43-positive cells were identified in the injected CPC population, further demonstrating the proliferation, differentiation and integration of the transplanted CPCs in host cells. Furthermore, animal hearts injected with CPCs showed increased angiogenesis, decreased infarct size, and improved heart function. In conclusion, our studies showed that Isl1+ CPCs, when combined with a suitable vehicle, can produce notable therapeutic effects in the infarcted heart, suggesting that CPCs might be an ideal cell source for cardiac therapy.
Collapse
Affiliation(s)
- Yunpeng Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical UniversityXian 710032, Shaanxi, China
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Shuo Tian
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
- Faculty of Health Sciences, University of MacauMacau SAR, China
| | - Liu Liu
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Peter Ma
- Department of Biologic and Materials Sciences, Biomedical Engineering, Macromolecular Science and Engineering Center, and Materials Science and Engineering, The University of MichiganAnn Arbor 48109, MI, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| |
Collapse
|
12
|
Zhang W, Kong C, Tong M, Chooi W, Huang N, Li R, Chan B. Maturation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) in 3D collagen matrix: Effects of niche cell supplementation and mechanical stimulation. Acta Biomater 2017; 49:204-217. [PMID: 27890729 DOI: 10.1016/j.actbio.2016.11.058] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 11/18/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes derived from human embryonic stem cells (hESC-CMs) are regarded as a promising source for regenerative medicine, drug testing and disease modeling. Nevertheless, cardiomyocytes are immature in terms of their contractile structure, metabolism and electrophysiological properties. Here, we fabricate cardiac muscle strips by encapsulating hESC-CMs in collagen-based biomaterials. Supplementation of niche cells at 3% to the number of hESC-CMs enhance the maturation of the hESC-CMs in 3D tissue matrix. The benefits of adding mesenchymal stem cells (MSCs) are comparable to that of adding fibroblasts. These two cell types demonstrate similar effects in promoting the compaction and cell spreading, as well as expression of maturation markers at both gene and protein levels. Mechanical loading, particularly cyclic stretch, produces engineered cardiac tissues with higher maturity in terms of twitch force, elastic modulus, sarcomere length and molecular signature, when comparing to static stretch or non-stretched controls. The current study demonstrates that the application of niche cells and mechanical stretch both stimulate the maturation of hESC-CMs in 3D architecture. Our results therefore suggest that this 3D model can be used for in vitro cardiac maturation study. STATEMENT OF SIGNIFICANCE Cardiomyocytes derived from human embryonic stem cells (hESC-CMs) are regarded as being a promising source of cells for regenerative medicine, drug testing and disease modeling. Nevertheless, cardiomyocytes are immature in terms of their contractile structure, metabolism and electrophysiological properties. In the current study, we have fabricated cardiac muscle strips by encapsulating hESC-CMs in collagen-based biomaterials and demonstrated that supplementation of mesenchymal niche cells as well as provision of mechanical loading particularly stretching have significantly promoted the maturation of the cardiomyocytes and hence improved the mechanical functional characteristics of the tissue strips. Specifically, with 3% niche cells including both fibroblasts and mesenchymal stem cells, a more mature hESC-CMs derived cardiac strip was resulted, in terms of compaction and spreading of cells, and upregulation of molecular signature in both gene and protein expression of maturation. Mechanical loading, particularly cyclic stretch, produces engineered cardiac tissues with higher maturity in terms of molecular signature markers and functional parameters including twitch force, elastic modulus and sarcomere length, when comparing with static stretch or non-stretched controls. The current study demonstrates that the application of niche cells and mechanical stretch both stimulate the maturation of hESC-CMs in 3D architecture, resulting in more mature cardiac strips. Our results contribute to bioengineering of functional heart tissue strips for drug screening and disease modeling.
Collapse
|
13
|
Abstract
Soluble morphogen gradients have long been studied in the context of heart specification and patterning. However, recent data have begun to challenge the notion that long-standing in vivo observations are driven solely by these gradients alone. Evidence from multiple biological models, from stem cells to ex vivo biophysical assays, now supports a role for mechanical forces in not only modulating cell behavior but also inducing it de novo in a process termed mechanotransduction. Structural proteins that connect the cell to its niche, for example, integrins and cadherins, and that couple to other growth factor receptors, either directly or indirectly, seem to mediate these changes, although specific mechanistic details are still being elucidated. In this review, we summarize how the wingless (Wnt), transforming growth factor-β, and bone morphogenetic protein signaling pathways affect cardiomyogenesis and then highlight the interplay between each pathway and mechanical forces. In addition, we will outline the role of integrins and cadherins during cardiac development. For each, we will describe how the interplay could change multiple processes during cardiomyogenesis, including the specification of undifferentiated cells, the establishment of heart patterns to accomplish tube and chamber formation, or the maturation of myocytes in the fully formed heart.
Collapse
Affiliation(s)
- Cassandra L Happe
- From the Department of Bioengineering, University of California, San Diego, La Jolla; and Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Adam J Engler
- From the Department of Bioengineering, University of California, San Diego, La Jolla; and Sanford Consortium for Regenerative Medicine, La Jolla, CA.
| |
Collapse
|
14
|
Xu Y, Fu M, Li Z, Fan Z, Li X, Liu Y, Anderson PM, Xie X, Liu Z, Guan J. A prosurvival and proangiogenic stem cell delivery system to promote ischemic limb regeneration. Acta Biomater 2016; 31:99-113. [PMID: 26689466 DOI: 10.1016/j.actbio.2015.12.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/17/2015] [Accepted: 12/11/2015] [Indexed: 12/20/2022]
Abstract
Stem cell therapy is one of the most promising strategies to restore blood perfusion and promote muscle regeneration in ischemic limbs. Yet its therapeutic efficacy remains low owing to the inferior cell survival under the low oxygen and nutrient environment of the injured limbs. To increase therapeutic efficacy, high rates of both short- and long-term cell survival are essential, which current approaches do not support. In this work, we hypothesized that a high rate of short-term cell survival can be achieved by introducing a prosurvival environment into the stem cell delivery system to enhance cell survival before vascularization is established; and that a high rate of long-term cell survival can be attained by building a proangiogenic environment in the system to quickly vascularize the limbs. The system was based on a biodegradable and thermosensitive poly(N-Isopropylacrylamide)-based hydrogel, a prosurvival and proangiogenic growth factor bFGF, and bone marrow-derived mesenchymal stem cells (MSCs). bFGF can be continuously released from the system for 4weeks. The released bFGF significantly improved MSC survival and paracrine effects under low nutrient and oxygen conditions (0% FBS and 1% O2) in vitro. The prosurvival effect of the bFGF on MSCs was resulted from activating cell Kruppel-like factor 4 (KLF4) pathway. When transplanted into the ischemic limbs, the system dramatically improved MSC survival. Some of the engrafted cells were differentiated into skeletal muscle and endothelial cells, respectively. The system also promoted the proliferation of host cells. After only 2weeks of implantation, tissue blood perfusion was completely recovered; and after 4weeks, the muscle fiber diameter was restored similarly to that of the normal limbs. These pronounced results demonstrate that the developed stem cell delivery system has a potential for ischemic limb regeneration. STATEMENT OF SIGNIFICANCE Stem cell therapy is a promising strategy to restore blood perfusion and promote muscle regeneration in ischemic limbs. Yet its therapeutic efficacy remains low owing to the inferior cell survival under the ischemic environment of the injured limbs. To increase therapeutic efficacy, high rate of cell survival is essential, which current approaches do not support. In this work, we tested the hypothesis that a stem cell delivery system that can continuously release a prosurvival and proangiogenic growth factor will promote high rates of cell survival in the ischemic limbs. The prosurvival effect could augment cell survival before vascularization is established, while the proangiogenic effect could stimulate quick angiogenesis to achieve long-term cell survival. Meanwhile, the differentiation of stem cells into endothelial and myogenic lineages, and cell paracrine effects will enhance vascularization and muscle regeneration.
Collapse
Affiliation(s)
- Yanyi Xu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Minghuan Fu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Department of Gerontology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Zhihong Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Division of General Surgery, Shanghai Pudong New District Zhoupu Hospital, Shanghai 201200, China
| | - Zhaobo Fan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaofei Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Ying Liu
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Peter M Anderson
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaoyun Xie
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Zhenguo Liu
- Davis Heart and Lung Research Institute, The Ohio State University, OH 43210, United States
| | - Jianjun Guan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Tongji Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
15
|
Ja KPMM, Miao Q, Zhen Tee NG, Lim SY, Nandihalli M, Ramachandra CJA, Mehta A, Shim W. iPSC-derived human cardiac progenitor cells improve ventricular remodelling via angiogenesis and interstitial networking of infarcted myocardium. J Cell Mol Med 2015; 20:323-32. [PMID: 26612359 PMCID: PMC4727567 DOI: 10.1111/jcmm.12725] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/29/2015] [Indexed: 12/27/2022] Open
Abstract
We investigate the effects of myocardial transplantation of human induced pluripotent stem cell (iPSC)‐derived progenitors and cardiomyocytes into acutely infarcted myocardium in severe combined immune deficiency mice. A total of 2 × 105 progenitors, cardiomyocytes or cell‐free saline were injected into peri‐infarcted anterior free wall. Sham‐operated animals received no injection. Myocardial function was assessed at 2‐week and 4‐week post‐infarction by using echocardiography and pressure‐volume catheterization. Early myocardial remodelling was observed at 2‐week with echocardiography derived stroke volume (SV) in saline (20.45 ± 7.36 μl, P < 0.05) and cardiomyocyte (19.52 ± 3.97 μl, P < 0.05) groups, but not in progenitor group (25.65 ± 3.61 μl), significantly deteriorated as compared to sham control group (28.41 ± 4.41 μl). Consistently, pressure–volume haemodynamic measurements showed worsening chamber dilation in saline (EDV: 23.24 ± 5.01 μl, P < 0.05; ESV: 17.08 ± 5.82 μl, P < 0.05) and cardiomyocyte (EDV: 26.45 ± 5.69 μl, P < 0.05; ESV: 18.03 ± 6.58 μl, P < 0.05) groups by 4‐week post‐infarction as compared to control (EDV: 15.26 ± 2.96 μl; ESV: 8.41 ± 2.94 μl). In contrast, cardiac progenitors (EDV: 20.09 ± 7.76 μl; ESV: 13.98 ± 6.74 μl) persistently protected chamber geometry against negative cardiac remodelling. Similarly, as compared to sham control (54.64 ± 11.37%), LV ejection fraction was preserved in progenitor group from 2‐(38.68 ± 7.34%) to 4‐week (39.56 ± 13.26%) while cardiomyocyte (36.52 ± 11.39%, P < 0.05) and saline (35.34 ± 11.86%, P < 0.05) groups deteriorated early at 2‐week. Improvements of myocardial function in the progenitor group corresponded to increased vascularization (16.12 ± 1.49/mm2 to 25.48 ± 2.08/mm2 myocardial tissue, P < 0.05) and coincided with augmented networking of cardiac telocytes in the interstitial space of infarcted zone.
Collapse
Affiliation(s)
- K P Myu Mia Ja
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Qingfeng Miao
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore.,Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nicole Gui Zhen Tee
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Sze Yun Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Manasi Nandihalli
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | | | - Ashish Mehta
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore.,DUKE-NUS Graduate Medical School
| | - Winston Shim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore.,DUKE-NUS Graduate Medical School
| |
Collapse
|
16
|
Ye J, Gaur M, Zhang Y, Sievers RE, Woods BJ, Aurigui J, Bernstein HS, Yeghiazarians Y. Treatment with hESC-Derived Myocardial Precursors Improves Cardiac Function after a Myocardial Infarction. PLoS One 2015; 10:e0131123. [PMID: 26230835 PMCID: PMC4521814 DOI: 10.1371/journal.pone.0131123] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/27/2015] [Indexed: 01/05/2023] Open
Abstract
Background We previously reported the generation of a reporter line of human embryonic stem cells (hESCs) with enhanced green fluorescent protein (eGFP) expression driven by the α-myosin heavy chain (αMHC) promoter. The GFP+/αMHC+ cells derived from this cell line behave as multipotent, human myocardial precursors (hMPs) in vitro. In this study, we evaluated the therapeutic effects of GFP+/αMHC+ cells isolated from the reporter line in a mouse model of myocardial infarction (MI). Methods MI was generated in immunodeficient mice. hMPs were injected into murine infarcted hearts under ultrasound guidance at 3 days post-MI. Human fetal skin fibroblasts (hFFs) were injected as control. Cardiac function was evaluated by echocardiography. Infarct size, angiogenesis, apoptosis, cell fate, and teratoma formation were analyzed by immunohistochemical staining. Results Compared with control, hMPs resulted in improvement of cardiac function post-MI with smaller infarct size, induced endogenous angiogenesis, and reduced apoptosis of host cardiomyocytes at the peri-infarct zone at 28 days post-MI. Conclusion Intramyocardial injection of hMPs improved cardiac function post-MI. The engraftment rate of these cells in the myocardium post-MI was low, suggesting that the majority of effect occurs via paracrine mechanisms.
Collapse
Affiliation(s)
- Jianqin Ye
- Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Meenakshi Gaur
- Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Yan Zhang
- Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Richard E. Sievers
- Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Brandon J. Woods
- Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Julian Aurigui
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Harold S. Bernstein
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, 94143, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Yerem Yeghiazarians
- Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, 94143, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, 94143, United States of America
- * E-mail:
| |
Collapse
|
17
|
Thimm TN, Squirrell JM, Liu Y, Eliceiri KW, Ogle BM. Endogenous Optical Signals Reveal Changes of Elastin and Collagen Organization During Differentiation of Mouse Embryonic Stem Cells. Tissue Eng Part C Methods 2015; 21:995-1004. [PMID: 25923353 DOI: 10.1089/ten.tec.2014.0699] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Components of the extracellular matrix (ECM) have recently been shown to influence stem cell specification. However, it has been challenging to assess the spatial and temporal dynamics of stem cell-ECM interactions because most methodologies utilized to date require sample destruction or fixation. We examined the efficacy of utilizing the endogenous optical signals of two important ECM proteins, elastin (Eln), through autofluorescence, and type I collagen (ColI), through second harmonic generation (SHG), during mouse embryonic stem cell differentiation. After finding favorable overlap between antibody labeling and the endogenous fluorescent signal of Eln, we used this endogenous signal to map temporal changes in Eln and ColI during murine embryoid body differentiation and found that Eln increases until day 9 and then decreases slightly by day 12, while Col1 steadily increases over the 12-day period. Furthermore, we combined endogenous fluorescence imaging and SHG with antibody labeling of cardiomyocytes to examine the spatial relationship between Eln and ColI accumulation and cardiomyocyte differentiation. Eln was ubiquitously present, with enrichment in regions with cardiomyocyte differentiation, while there was an inverse correlation between ColI and cardiomyocyte differentiation. This work provides an important first step for utilizing endogenous optical signals, which can be visualized in living cells, to understand the relationship between the ECM and cardiomyocyte development and sets the stage for future studies of stem cell-ECM interactions and dynamics relevant to stem cells as well as other cell and tissue types.
Collapse
Affiliation(s)
- Terra N Thimm
- 1 Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison , Madison, Wisconsin
| | - Jayne M Squirrell
- 1 Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison , Madison, Wisconsin
| | - Yuming Liu
- 1 Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison , Madison, Wisconsin
| | - Kevin W Eliceiri
- 1 Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison , Madison, Wisconsin.,2 Morgridge Institute for Research, University of Wisconsin-Madison , Madison, Wisconsin
| | - Brenda M Ogle
- 3 Department of Biomedical Engineering, University of Minnesota-Twin Cities , Minneapolis, Minnesota
| |
Collapse
|
18
|
Rodriguez ML, Graham BT, Pabon LM, Han SJ, Murry CE, Sniadecki NJ. Measuring the contractile forces of human induced pluripotent stem cell-derived cardiomyocytes with arrays of microposts. J Biomech Eng 2015; 136:051005. [PMID: 24615475 DOI: 10.1115/1.4027145] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/10/2014] [Indexed: 12/31/2022]
Abstract
Human stem cell-derived cardiomyocytes hold promise for heart repair, disease modeling, drug screening, and for studies of developmental biology. All of these applications can be improved by assessing the contractility of cardiomyocytes at the single cell level. We have developed an in vitro platform for assessing the contractile performance of stem cell-derived cardiomyocytes that is compatible with other common endpoints such as microscopy and molecular biology. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were seeded onto elastomeric micropost arrays in order to characterize the contractile force, velocity, and power produced by these cells. We assessed contractile function by tracking the deflection of microposts beneath an individual hiPSC-CM with optical microscopy. Immunofluorescent staining of these cells was employed to assess their spread area, nucleation, and sarcomeric structure on the microposts. Following seeding of hiPSC-CMs onto microposts coated with fibronectin, laminin, and collagen IV, we found that hiPSC-CMs on laminin coatings demonstrated higher attachment, spread area, and contractile velocity than those seeded on fibronectin or collagen IV coatings. Under optimized conditions, hiPSC-CMs spread to an area of approximately 420 μm2, generated systolic forces of approximately 15 nN/cell, showed contraction and relaxation rates of 1.74 μm/s and 1.46 μm/s, respectively, and had a peak contraction power of 29 fW. Thus, elastomeric micropost arrays can be used to study the contractile strength and kinetics of hiPSC-CMs. This system should facilitate studies of hiPSC-CM maturation, disease modeling, and drug screens as well as fundamental studies of human cardiac contraction.
Collapse
|
19
|
Prowse AB, Timmins NE, Yau TM, Li RK, Weisel RD, Keller G, Zandstra PW. Transforming the Promise of Pluripotent Stem Cell-Derived Cardiomyocytes to a Therapy: Challenges and Solutions for Clinical Trials. Can J Cardiol 2014; 30:1335-49. [DOI: 10.1016/j.cjca.2014.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/06/2014] [Accepted: 08/11/2014] [Indexed: 01/08/2023] Open
|
20
|
Young JL, Kretchmer K, Ondeck MG, Zambon AC, Engler AJ. Mechanosensitive kinases regulate stiffness-induced cardiomyocyte maturation. Sci Rep 2014; 4:6425. [PMID: 25236849 PMCID: PMC4168277 DOI: 10.1038/srep06425] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/28/2014] [Indexed: 01/26/2023] Open
Abstract
Cells secrete and assemble extracellular matrix throughout development, giving rise to time-dependent, tissue-specific stiffness. Mimicking myocardial matrix stiffening, i.e. ~10-fold increase over 1 week, with a hydrogel system enhances myofibrillar organization of embryonic cardiomyocytes compared to static hydrogels, and thus we sought to identify specific mechanosensitive proteins involved. Expression and/or phosphorylation state of 309 unique protein kinases were examined in embryonic cardiomyocytes plated on either dynamically stiffening or static mature myocardial stiffness hydrogels. Gene ontology analysis of these kinases identified cardiogenic pathways that exhibited time-dependent up-regulation on dynamic versus static matrices, including PI3K/AKT and p38 MAPK, while GSK3β, a known antagonist of cardiomyocyte maturation, was down-regulated. Additionally, inhibiting GSK3β on static matrices improved spontaneous contraction and myofibril organization, while inhibiting agonist AKT on dynamic matrices reduced myofibril organization and spontaneous contraction, confirming its role in mechanically-driven maturation. Together, these data indicate that mechanically-driven maturation is at least partially achieved via active mechanosensing at focal adhesions, affecting expression and phosphorylation of a variety of protein kinases important to cardiomyogenesis.
Collapse
Affiliation(s)
- Jennifer L. Young
- Department of Bioengineering, University of California, San Diego, CA 92093
| | - Kyle Kretchmer
- Department of Bioengineering, University of California, San Diego, CA 92093
| | - Matthew G. Ondeck
- Department of Material Science Program, University of California, San Diego, CA 92093
| | | | - Adam J. Engler
- Department of Bioengineering, University of California, San Diego, CA 92093
- Department of Material Science Program, University of California, San Diego, CA 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| |
Collapse
|
21
|
Lee TJ, Kang S, Jeong GJ, Yoon JK, Bhang SH, Oh J, Kim BS. Incorporation of gold-coated microspheres into embryoid body of human embryonic stem cells for cardiomyogenic differentiation. Tissue Eng Part A 2014; 21:374-81. [PMID: 25065511 DOI: 10.1089/ten.tea.2014.0015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human embryonic stem cells (hESCs) are a useful cell source for cardiac regeneration by stem cell therapy. In this study, we show that incorporation of gold-coated microspheres into hESC-derived embryoid bodies (EBs) enhances the cardiomyogenic differentiation process of pluripotent embryonic stem cells. A polycaprolactone (PCL) microsphere surface was coated with gold. Either gold-coated PCL microspheres (AuMS) or PCL microspheres (MS) were incorporated into hESC-derived EBs. AuMS and MS were not cytotoxic. AuMS promoted the expression of genes for mesodermal and cardiac mesodermal lineage cells, both of which are intermediates in the process of cardiac differentiation of hESCs on day 4 and the expression of cardiomyogenic differentiation markers on day 14 compared to MS. AuMS also enhanced gene expression of cardiac-specific extracellular matrices. Incorporation of gold-coated MS into hESC-derived EBs may provide a new platform for inducing cardiomyogenic differentiation of pluripotent embryonic stem cells.
Collapse
Affiliation(s)
- Tae-Jin Lee
- 1 Engineering Research Institute, Seoul National University , Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Lee TJ, Park S, Bhang SH, Yoon JK, Jo I, Jeong GJ, Hong BH, Kim BS. Graphene enhances the cardiomyogenic differentiation of human embryonic stem cells. Biochem Biophys Res Commun 2014; 452:174-80. [PMID: 25152405 DOI: 10.1016/j.bbrc.2014.08.062] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/15/2014] [Indexed: 12/11/2022]
Abstract
Graphene has drawn attention as a substrate for stem cell culture and has been reported to stimulate the differentiation of multipotent adult stem cells. Here, we report that graphene enhances the cardiomyogenic differentiation of human embryonic stem cells (hESCs) at least in part, due to nanoroughness of graphene. Large-area graphene on glass coverslips was prepared via the chemical vapor deposition method. The coating of the graphene with vitronectin (VN) was required to ensure high viability of the hESCs cultured on the graphene. hESCs were cultured on either VN-coated glass (glass group) or VN-coated graphene (graphene group) for 21 days. The cells were also cultured on glass coated with Matrigel (Matrigel group), which is a substrate used in conventional, directed cardiomyogenic differentiation systems. The culture of hESCs on graphene promoted the expression of genes involved in the stepwise differentiation into mesodermal and endodermal lineage cells and subsequently cardiomyogenic differentiation compared with the culture on glass or Matrigel. In addition, the culture on graphene enhanced the gene expression of cardiac-specific extracellular matrices. Culture on graphene may provide a new platform for the development of stem cell therapies for ischemic heart diseases by enhancing the cardiomyogenic differentiation of hESCs.
Collapse
Affiliation(s)
- Tae-Jin Lee
- Engineering Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Subeom Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Insu Jo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Gun-Jae Jeong
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Byung Hee Hong
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
| | - Byung-Soo Kim
- Engineering Research Institute, Seoul National University, Seoul, Republic of Korea; School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea; Institute of Bioengineering, Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Luo J, Weaver MS, Cao B, Dennis JE, Van Biber B, Laflamme MA, Allen MD. Cobalt protoporphyrin pretreatment protects human embryonic stem cell-derived cardiomyocytes from hypoxia/reoxygenation injury in vitro and increases graft size and vascularization in vivo. Stem Cells Transl Med 2014; 3:734-44. [PMID: 24736402 DOI: 10.5966/sctm.2013-0189] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) can regenerate infarcted myocardium. However, when implanted into acutely infarcted hearts, few cells survive the first week postimplant. To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). When hESC-CMs were challenged with an in vitro hypoxia/reoxygenation injury, mimicking cell transplantation into an ischemic site, survival was significantly greater among cells pretreated with CoPP versus phosphate-buffered saline (PBS)-pretreated controls. Compared with PBS-pretreated cells, CoPP-pretreated hESC-CM preparations exhibited higher levels of HO-1 expression, Akt phosphorylation, and vascular endothelial growth factor production, with reduced apoptosis, and a 30% decrease in intracellular reactive oxygen species. For in vivo translation, 1 × 10(7) hESC-CMs were pretreated ex vivo with CoPP or PBS and then injected intramyocardially into rat hearts immediately following acute infarction (permanent coronary ligation). At 1 week, hESC-CM content, assessed by quantitative polymerase chain reaction for human Alu sequences, was 17-fold higher in hearts receiving CoPP- than PBS-pretreated cells. On histomorphometry, cardiomyocyte graft size was 2.6-fold larger in hearts receiving CoPP- than PBS-pretreated cells, occupying up to 12% of the ventricular area. Vascular density of host-perfused human-derived capillaries was significantly greater in grafts composed of CoPP- than PBS-pretreated cells. Taken together, these experiments demonstrate that ex vivo pretreatment of hESC-CMs with a single dose of CoPP before intramyocardial implantation more than doubled resulting graft size and improved early graft vascularization in acutely infarcted hearts. These findings open the door for delivery of these, or other, stem cells during acute interventional therapy following myocardial infarction or ischemia.
Collapse
Affiliation(s)
- Jun Luo
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew S Weaver
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Baohong Cao
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - James E Dennis
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Benjamin Van Biber
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Michael A Laflamme
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| | - Margaret D Allen
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA; Departments of Pathology and Surgery, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
24
|
Abstract
Integrins are heterodimeric, transmembrane receptors that are expressed in all cells, including those in the heart. They participate in multiple critical cellular processes including adhesion, extracellular matrix organization, signaling, survival, and proliferation. Particularly relevant for a contracting muscle cell, integrins are mechanotransducers, translating mechanical to biochemical information. Although it is likely that cardiovascular clinicians and scientists have the highest recognition of integrins in the cardiovascular system from drugs used to inhibit platelet aggregation, the focus of this article will be on the role of integrins specifically in the cardiac myocyte. After a general introduction to integrin biology, the article will discuss important work on integrin signaling, mechanotransduction, and lessons learned about integrin function from a range of model organisms. Then we will detail work on integrin-related proteins in the myocyte, how integrins may interact with ion channels and mediate viral uptake into cells, and also play a role in stem cell biology. Finally, we will discuss directions for future study.
Collapse
Affiliation(s)
- Sharon Israeli-Rosenberg
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Ana Maria Manso
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Hideshi Okada
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Robert S Ross
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
25
|
Shiba Y, Filice D, Fernandes S, Minami E, Dupras SK, Biber BV, Trinh P, Hirota Y, Gold JD, Viswanathan M, Laflamme MA. Electrical Integration of Human Embryonic Stem Cell-Derived Cardiomyocytes in a Guinea Pig Chronic Infarct Model. J Cardiovasc Pharmacol Ther 2014; 19:368-381. [PMID: 24516260 DOI: 10.1177/1074248413520344] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were recently shown to be capable of electromechanical integration following direct injection into intact or recently injured guinea pig hearts, and hESC-CM transplantation in recently injured hearts correlated with improvements in contractile function and a reduction in the incidence of arrhythmias. The present study was aimed at determining the ability of hESC-CMs to integrate and modulate electrical stability following transplantation in a chronic model of cardiac injury. METHODS AND RESULTS At 28 days following cardiac cryoinjury, guinea pigs underwent intracardiac injection of hESC-CMs, noncardiac hESC derivatives (non-CMs), or vehicle. Histology confirmed partial remuscularization of the infarct zone in hESC-CM recipients while non-CM recipients showed heterogeneous xenografts. The 3 experimental groups showed no significant difference in the left ventricular dimensions or fractional shortening by echocardiography or in the incidence of spontaneous arrhythmias by telemetric monitoring. Although recipients of hESC-CMs and vehicle showed a similar incidence of arrhythmias induced by programmed electrical stimulation at 4 weeks posttransplantation, non-CM recipients proved to be highly inducible, with a ∼3-fold greater incidence of induced arrhythmias. In parallel studies, we investigated the ability of hESC-CMs to couple with host myocardium in chronically injured hearts by the intravital imaging of hESC-CM grafts that stably expressed a fluorescent reporter of graft activation, the genetically encoded calcium sensor GCaMP3. In this work, we found that only ∼38% (5 of 13) of recipients of GCaMP3+ hESC-CMs showed fluorescent transients that were coupled to the host electrocardiogram. CONCLUSIONS Human embryonic stem cell-derived cardiomyocytes engraft in chronically injured hearts without increasing the incidence of arrhythmias, but their electromechanical integration is more limited than previously reported following their transplantation in a subacute injury model. Moreover, non-CM grafts may promote arrhythmias under certain conditions, a finding that underscores the need for input preparations of high cardiac purity.
Collapse
Affiliation(s)
- Yuji Shiba
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Dominic Filice
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA Department of Bioengineering, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Sarah Fernandes
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA Gilead Sciences, Fremont, CA, USA
| | - Elina Minami
- Department of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Sarah K Dupras
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Benjamin Van Biber
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Peter Trinh
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Yusuke Hirota
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Joseph D Gold
- Geron Corporation, Menlo Park, CA, USA Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Mohan Viswanathan
- Department of Medicine/Cardiology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Michael A Laflamme
- Department of Pathology, Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Park J, Park S, Ryu S, Bhang SH, Kim J, Yoon JK, Park YH, Cho SP, Lee S, Hong BH, Kim BS. Graphene-regulated cardiomyogenic differentiation process of mesenchymal stem cells by enhancing the expression of extracellular matrix proteins and cell signaling molecules. Adv Healthc Mater 2014; 3:176-81. [PMID: 23949999 DOI: 10.1002/adhm.201300177] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/28/2013] [Indexed: 12/26/2022]
Abstract
The potential of graphene as a mesenchymal stem cell (MSC) culture substrate to promote cardiomyogenic differentiation is demonstrated. Graphene exhibits no sign of cytotoxicity for stem cell culture. MSCs are committed toward cardiomyogenic lineage by simply culturing them on graphene. This may be attributed, at least partially, to the regulation of expression levels of extracellular matrix and signaling molecules.
Collapse
Affiliation(s)
- Jooyeon Park
- School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Doppler SA, Deutsch MA, Lange R, Krane M. Cardiac regeneration: current therapies-future concepts. J Thorac Dis 2013; 5:683-97. [PMID: 24255783 DOI: 10.3978/j.issn.2072-1439.2013.08.71] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) continues to be one of the main causes of death in the western world. A high burden of disease and the high costs for the healthcare systems claim for novel therapeutic strategies besides current conventional medical care. One decade ago first clinical trials addressed stem cell based therapies as a potential alternative therapeutic strategy for myocardial regeneration and repair. Besides bone marrow derived stem cells (BMCs), adult stem cells from adipose or cardiac tissue have been used in current clinical studies with inconsistent results. Although outcomes in terms of safety and feasibility are generally encouraging, functional improvements were mostly disappointingly low and have failed to reach expectations. In the future, new concepts for myocardial regeneration, especially concerning recovery of cardiomyocyte loss, have to be developed. Transplantation of novel stem or progenitor cell populations with "true" regenerative potential, direct reprogramming of scar tissue into functional myocardium, tissue engineering or stimulation of endogenous cardiac repair by pharmacological agents are conceivable. This review summarizes current evidence of stem cell based regenerative therapies and discusses future strategies to improve functional outcomes.
Collapse
Affiliation(s)
- Stefanie A Doppler
- Department of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München (TUM), Munich Heart Alliance, Munich, Germany
| | | | | | | |
Collapse
|
28
|
Don CW, Murry CE. Improving survival and efficacy of pluripotent stem cell-derived cardiac grafts. J Cell Mol Med 2013; 17:1355-62. [PMID: 24118766 PMCID: PMC4049630 DOI: 10.1111/jcmm.12147] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/28/2013] [Indexed: 01/02/2023] Open
Abstract
Human embryonic stem cells (hESCs) can be differentiated into structurally and electrically functional myocardial tissue and have the potential to regenerate large regions of infarcted myocardium. One of the key challenges that needs to be addressed towards full-scale clinical application of hESCs is enhancing survival of the transplanted cells within ischaemic or scarred, avascular host tissue. Shortly after transplantation, most hESCs are lost as a result of multiple mechanical, cellular and host factors, and a large proportion of the remaining cells undergo apoptosis or necrosis shortly thereafter, as a result of loss of adhesion-related signals, ischaemia, inflammation or immunological rejection. Blocking the apoptotic signalling pathways of the cells, using pro-survival cocktails, conditioning hESCs prior to transplant, promoting angiogenesis, immunosuppressing the host and using of bioengineered matrices are among the emerging techniques that have been shown to optimize cell survival. This review presents an overview of the current strategies for optimizing cell and host tissue to improve the survival and efficacy of cardiac cells derived from pluripotent stem cells.
Collapse
Affiliation(s)
- Creighton W Don
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
29
|
Sreejit P, Verma RS. Natural ECM as biomaterial for scaffold based cardiac regeneration using adult bone marrow derived stem cells. Stem Cell Rev Rep 2013; 9:158-71. [PMID: 23319217 DOI: 10.1007/s12015-013-9427-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cellular therapy using stem cells for cardiac diseases has recently gained much interest in the scientific community due to its potential in regenerating damaged and even dead tissue and thereby restoring the organ function. Stem cells from various sources and origin are being currently used for regeneration studies directly or along with differentiation inducing agents. Long term survival and minimal side effects can be attained by using autologous cells and reduced use of inducing agents. Cardiomyogenic differentiation of adult derived stem cells has been previously reported using various inducing agents but the use of a potentially harmful DNA demethylating agent 5-azacytidine (5-azaC) has been found to be critical in almost all studies. Alternate inducing factors and conditions/stimulant like physical condition including electrical stimulation, chemical inducers and biological agents have been attempted by numerous groups to induce cardiac differentiation. Biomaterials were initially used as artificial scaffold in in vitro studies and later as a delivery vehicle. Natural ECM is the ideal biological scaffold since it contains all the components of the tissue from which it was derived except for the living cells. Constructive remodeling can be performed using such natural ECM scaffolds and stem cells since, the cells can be delivered to the site of infraction and once delivered the cells adhere and are not "lost". Due to the niche like conditions of ECM, stem cells tend to differentiate into tissue specific cells and attain several characteristics similar to that of functional cells even in absence of any directed differentiation using external inducers. The development of niche mimicking biomaterials and hybrid biomaterial can further advance directed differentiation without specific induction. The mechanical and electrical integration of these materials to the functional tissue is a problem to be addressed. The search for the perfect extracellular matrix for therapeutic applications including engineering cardiac tissue structures for post ischemic cardiac tissue regeneration continues.
Collapse
Affiliation(s)
- P Sreejit
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, TN, India
| | | |
Collapse
|
30
|
du Pré BC, Doevendans PA, van Laake LW. Stem cells for cardiac repair: an introduction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2013; 10:186-97. [PMID: 23888179 PMCID: PMC3708059 DOI: 10.3969/j.issn.1671-5411.2013.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 02/16/2013] [Accepted: 04/22/2013] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease is a major cause of morbidity and mortality throughout the world. Most cardiovascular diseases, such as ischemic heart disease and cardiomyopathy, are associated with loss of functional cardiomyocytes. Unfortunately, the heart has a limited regenerative capacity and is not able to replace these cardiomyocytes once lost. In recent years, stem cells have been put forward as a potential source for cardiac regeneration. Pre-clinical studies that use stem cell-derived cardiac cells show promising results. The mechanisms, though, are not well understood, results have been variable, sometimes transient in the long term, and often without a mechanistic explanation. There are still several major hurdles to be taken. Stem cell-derived cardiac cells should resemble original cardiac cell types and be able to integrate in the damaged heart. Integration requires administration of stem cell-derived cardiac cells at the right time using the right mode of delivery. Once delivered, transplanted cells need vascularization, electrophysiological coupling with the injured heart, and prevention of immunological rejection. Finally, stem cell therapy needs to be safe, reproducible, and affordable. In this review, we will give an introduction to the principles of stem cell based cardiac repair.
Collapse
Affiliation(s)
- Bastiaan C du Pré
- Departments of Cardiology and Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, P.O. box 85500, 3508 GA Utrecht, the Netherlands
| | | | | |
Collapse
|
31
|
Enhanced cardiomyogenic lineage differentiation of adult bone-marrow-derived stem cells grown on cardiogel. Cell Tissue Res 2013; 353:443-56. [PMID: 23771778 DOI: 10.1007/s00441-013-1661-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/06/2013] [Indexed: 02/03/2023]
Abstract
The extracellular matrix (ECM) and its components are known to promote growth and cellular differentiation in vitro. Cardiogel, a three-dimensional extracellular matrix derived from cardiac fibroblasts, is evaluated for its cardiomyogenic-differentiation-inducing potential on bone-marrow-derived stem cells (BMSC). BMSC from adult mice were grown on cardiogel and induced to differentiate into specific lineages that were validated by morphological, phenotypic and molecular assays. The data revealed that the cardiogel enhanced cardiomyogenic and adipogenic differentiation and relegated osteogenic differentiation following specific induction. More importantly, increased cardiomyogenic differentiation was also observed following BMSC growth on cardiogel without specific chemical (5-azacytidine) induction. This is the first report of an attempt to use cardiogel as a biomaterial on which to achieve cardiomyogenic differentiation of BMSC without chemical induction. Our study suggests that cardiogel is an efficient extracellular matrix that enhances the cardiomyogenic differentiation of BMSC and that it can therefore be used as a scaffold for cardiac tissue regeneration.
Collapse
|
32
|
Maher KO, Xu C. Marching towards regenerative cardiac therapy with human pluripotent stem cells. DISCOVERY MEDICINE 2013; 15:349-356. [PMID: 23819949 PMCID: PMC4144195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Damage in cardiac tissues from ischemia or other pathological conditions leads to heart failure; and cell loss or dysfunction in pacemaker tissues due to congenital heart defects, aging, and acquired diseases can cause severe arrhythmias. The promise of successful therapies with stem cells to treat these conditions has remained elusive to the scientific community. However, recent advances in this field have opened new opportunities for regenerative cardiac therapy. Transplantation of cardiomyocytes derived from human pluripotent stem cells has the potential to alleviate heart disease. Since the initial derivation of human embryonic stem cells, significant progress has been made in the generation and characterization of enriched cardiomyocytes and the demonstration of the ability of these cardiomyocytes to survive, integrate, and function in animal models. The scope of therapeutic potential from pluripotent stem cell-derived cardiomyocytes has been further expanded with the invention of induced pluripotent stem cells, which can be induced to generate functional cardiomyocytes for regenerative cardiac therapy in a patient specific manner. The reprogramming technology has also inspired the recent discovery of direct conversion of fibroblasts into cardiomyocyte-like cells, which may allow endogenous cardiac repair. Regenerative cardiac therapy with human pluripotent stem cells is now moving closer to clinic testing.
Collapse
Affiliation(s)
- Kevin O Maher
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
33
|
Moyes KW, Sip CG, Obenza W, Yang E, Horst C, Welikson RE, Hauschka SD, Folch A, Laflamme MA. Human embryonic stem cell-derived cardiomyocytes migrate in response to gradients of fibronectin and Wnt5a. Stem Cells Dev 2013; 22:2315-25. [PMID: 23517131 DOI: 10.1089/scd.2012.0586] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
An improved understanding of the factors that regulate the migration of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) would provide new insights into human heart development and suggest novel strategies to improve their electromechanical integration after intracardiac transplantation. Since nothing has been reported as to the factors controlling hESC-CM migration, we hypothesized that hESC-CMs would migrate in response to the extracellular matrix and soluble signaling molecules previously implicated in heart morphogenesis. To test this, we screened candidate factors by transwell assay for effects on hESC-CM motility, followed by validation via live-cell imaging and/or gap-closure assays. Fibronectin (FN) elicited a haptotactic response from hESC-CMs, with cells seeded on a steep FN gradient showing nearly a fivefold greater migratory activity than cells on uniform FN. Studies with neutralizing antibodies indicated that adhesion and migration on FN are mediated by integrins α-5 and α-V. Next, we screened 10 soluble candidate factors by transwell assay and found that the noncanonical Wnt, Wnt5a, elicited an approximately twofold increase in migration over controls. This effect was confirmed using the gap-closure assay, in which Wnt5a-treated hESC-CMs showed approximately twofold greater closure than untreated cells. Studies with microfluidic-generated Wnt5a gradients showed that this factor was chemoattractive as well as chemokinetic, and Wnt5a-mediated responses were inhibited by the Frizzled-1/2 receptor antagonist, UM206. In summary, hESC-CMs show robust promigratory responses to FN and Wnt5a, findings that have implications on both cardiac development and cell-based therapies.
Collapse
Affiliation(s)
- Kara White Moyes
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Moon SH, Kang SW, Park SJ, Bae D, Kim SJ, Lee HA, Kim KS, Hong KS, Kim JS, Do JT, Byun KH, Chung HM. The use of aggregates of purified cardiomyocytes derived from human ESCs for functional engraftment after myocardial infarction. Biomaterials 2013; 34:4013-4026. [DOI: 10.1016/j.biomaterials.2013.02.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 02/10/2013] [Indexed: 11/15/2022]
|
35
|
Didié M, Christalla P, Rubart M, Muppala V, Döker S, Unsöld B, El-Armouche A, Rau T, Eschenhagen T, Schwoerer AP, Ehmke H, Schumacher U, Fuchs S, Lange C, Becker A, Tao W, Scherschel JA, Soonpaa MH, Yang T, Lin Q, Zenke M, Han DW, Schöler HR, Rudolph C, Steinemann D, Schlegelberger B, Kattman S, Witty A, Keller G, Field LJ, Zimmermann WH. Parthenogenetic stem cells for tissue-engineered heart repair. J Clin Invest 2013; 123:1285-98. [PMID: 23434590 DOI: 10.1172/jci66854] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 01/03/2013] [Indexed: 01/14/2023] Open
Abstract
Uniparental parthenotes are considered an unwanted byproduct of in vitro fertilization. In utero parthenote development is severely compromised by defective organogenesis and in particular by defective cardiogenesis. Although developmentally compromised, apparently pluripotent stem cells can be derived from parthenogenetic blastocysts. Here we hypothesized that nonembryonic parthenogenetic stem cells (PSCs) can be directed toward the cardiac lineage and applied to tissue-engineered heart repair. We first confirmed similar fundamental properties in murine PSCs and embryonic stem cells (ESCs), despite notable differences in genetic (allelic variability) and epigenetic (differential imprinting) characteristics. Haploidentity of major histocompatibility complexes (MHCs) in PSCs is particularly attractive for allogeneic cell-based therapies. Accordingly, we confirmed acceptance of PSCs in MHC-matched allotransplantation. Cardiomyocyte derivation from PSCs and ESCs was equally effective. The use of cardiomyocyte-restricted GFP enabled cell sorting and documentation of advanced structural and functional maturation in vitro and in vivo. This included seamless electrical integration of PSC-derived cardiomyocytes into recipient myocardium. Finally, we enriched cardiomyocytes to facilitate engineering of force-generating myocardium and demonstrated the utility of this technique in enhancing regional myocardial function after myocardial infarction. Collectively, our data demonstrate pluripotency, with unrestricted cardiogenicity in PSCs, and introduce this unique cell type as an attractive source for tissue-engineered heart repair.
Collapse
Affiliation(s)
- Michael Didié
- Institute of Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Human Embryonic Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
37
|
Sheng X, Reppel M, Nguemo F, Mohammad FI, Kuzmenkin A, Hescheler J, Pfannkuche K. Human pluripotent stem cell-derived cardiomyocytes: response to TTX and lidocain reveals strong cell to cell variability. PLoS One 2012; 7:e45963. [PMID: 23029342 PMCID: PMC3459939 DOI: 10.1371/journal.pone.0045963] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 08/28/2012] [Indexed: 02/07/2023] Open
Abstract
Stem cell derived cardiomyocytes generated either from human embryonic stem cells (hESC-CMs) or human induced pluripotent stem cells (hiPSC-CMs) hold great promise for the investigation of early developmental processes in human cardiomyogenesis and future cell replacement strategies. We have analyzed electrophysiological properties of hESC-CMs (HES2) and hiPSC-CMs, derived from reprogrammed adult foreskin fibroblasts that have previously been found to be highly similar in terms of gene expression. In contrast to the similarity found in the expression profile we found substantial differences in action potentials (APs) and sodium currents at late stage (day 60) of in vitro differentiation with higher sodium currents in hiPSC-CMs. Sensitivity to lidocain was considerably reduced in hESC-CMs as compared to hiPSC-CMs, and the effect could not be explained by differences in beating frequency. In contrast, sensitivity to tetrodotoxin (TTX) was higher in hESC-CMs suggesting different contributions of TTX-sensitive and TTX-resistant sodium channels to AP generation. These data point to physiological differences that are not necessarily detected by genomics. We conclude that novel pharmacological screening-assays using hiPSC-CMs need to be applied with some caution.
Collapse
Affiliation(s)
- Xiaowu Sheng
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
- Department of Physiology and German-Chinese Stem Cell Center, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | - Filomain Nguemo
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Farooq Ibrahem Mohammad
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
- Biotechnology Research Center, Al Nahrain University, Baghdad, Iraq
| | - Alexey Kuzmenkin
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Kurt Pfannkuche
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
- Clinic and Polyclinic for Paedriatric Cardiology, University of Cologne, Cologne, Germany
| |
Collapse
|
38
|
Blazeski A, Zhu R, Hunter DW, Weinberg SH, Boheler KR, Zambidis ET, Tung L. Electrophysiological and contractile function of cardiomyocytes derived from human embryonic stem cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:178-95. [PMID: 22958937 DOI: 10.1016/j.pbiomolbio.2012.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/23/2022]
Abstract
Human embryonic stem cells have emerged as the prototypical source from which cardiomyocytes can be derived for use in drug discovery and cell therapy. However, such applications require that these cardiomyocytes (hESC-CMs) faithfully recapitulate the physiology of adult cells, especially in relation to their electrophysiological and contractile function. We review what is known about the electrophysiology of hESC-CMs in terms of beating rate, action potential characteristics, ionic currents, and cellular coupling as well as their contractility in terms of calcium cycling and contraction. We also discuss the heterogeneity in cellular phenotypes that arises from variability in cardiac differentiation, maturation, and culture conditions, and summarize present strategies that have been implemented to reduce this heterogeneity. Finally, we present original electrophysiological data from optical maps of hESC-CM clusters.
Collapse
Affiliation(s)
- Adriana Blazeski
- Department of Biomedical Engineering, The Johns Hopkins University, 720 Rutland Ave., Baltimore, MD 21205, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Synergistic effects of hypoxia and extracellular matrix cues in cardiomyogenesis. Biomaterials 2012; 33:6313-9. [PMID: 22717366 DOI: 10.1016/j.biomaterials.2012.05.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 05/27/2012] [Indexed: 11/23/2022]
Abstract
Limited characterization of how the stem cell niche evolves has hindered our ability to mimic the physiological environment. In this paper, we hypothesized that hypoxia-induced extracellular matrix (ECM) cues may facilitate cardiomyogenesis. We evaluated the expression of four ECM proteins - fibronectin, collagen I, collagen IV, and laminin - over a period of 20 days in H1 and H9 human embryonic stem cell-derived embryoid bodies (EBs) under hypoxic (5% oxygen) and normoxic (21% oxygen) conditions. Hypoxic EBs exhibited increased collagen I, collagen IV and fibronectin expression relative to normoxic EBs between days 9-13, which coincided with increased expression of mesoderm genes. The effect of ECM cues was confirmed by plating day 9 EBs on collagen IV, gelatin, and fibronectin-rich substrates for 11 days. Hypoxia/gelatin cultures synergistically increased the cardiomyocyte yield by 1.7 and 5.5 fold relative to normoxia/gelatin and normoxia/collagen IV cultures, respectively. Current differentiation protocols may underestimate the contribution of hypoxia and ECM cues that evolve during EB maturation.
Collapse
|
40
|
Lui KO, Bu L, Li RA, Chan CW. Pluripotent stem cell-based heart regeneration: From the developmental and immunological perspectives. ACTA ACUST UNITED AC 2012; 96:98-108. [DOI: 10.1002/bdrc.21004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
41
|
Vunjak-Novakovic G, Lui KO, Tandon N, Chien KR. Bioengineering heart muscle: a paradigm for regenerative medicine. Annu Rev Biomed Eng 2012; 13:245-67. [PMID: 21568715 DOI: 10.1146/annurev-bioeng-071910-124701] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The idea of extending the lifetime of our organs is as old as humankind, fueled by major advances in organ transplantation, novel drugs, and medical devices. However, true regeneration of human tissue has become increasingly plausible only in recent years. The human heart has always been a focus of such efforts, given its notorious inability to repair itself following injury or disease. We discuss here the emerging bioengineering approaches to regeneration of heart muscle as a paradigm for regenerative medicine. Our focus is on biologically inspired strategies for heart regeneration, knowledge gained thus far about how to make a "perfect" heart graft, and the challenges that remain to be addressed for tissue-engineered heart regeneration to become a clinical reality. We emphasize the need for interdisciplinary research and training, as recent progress in the field is largely being made at the interfaces between cardiology, stem cell science, and bioengineering.
Collapse
|
42
|
Forte G, Pietronave S, Nardone G, Zamperone A, Magnani E, Pagliari S, Pagliari F, Giacinti C, Nicoletti C, Musaró A, Rinaldi M, Ribezzo M, Comoglio C, Traversa E, Okano T, Minieri M, Prat M, Di Nardo P. Human cardiac progenitor cell grafts as unrestricted source of supernumerary cardiac cells in healthy murine hearts. Stem Cells 2012; 29:2051-61. [PMID: 22009661 DOI: 10.1002/stem.763] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Human heart harbors a population of resident progenitor cells that can be isolated by stem cell antigen-1 antibody and expanded in culture. These cells can differentiate into cardiomyocytes in vitro and contribute to cardiac regeneration in vivo. However, when directly injected as single cell suspension, less than 1%-5% survive and differentiate. Among the major causes of this failure are the distressing protocols used to culture in vitro and implant progenitor cells into damaged hearts. Human cardiac progenitors obtained from the auricles of patients were cultured as scaffoldless engineered tissues fabricated using temperature-responsive surfaces. In the engineered tissue, progenitor cells established proper three-dimensional intercellular relationships and were embedded in self-produced extracellular matrix preserving their phenotype and multipotency in the absence of significant apoptosis. After engineered tissues were leant on visceral pericardium, a number of cells migrated into the murine myocardium and in the vascular walls, where they integrated in the respective textures. The study demonstrates the suitability of such an approach to deliver stem cells to the myocardium. Interestingly, the successful delivery of cells in murine healthy hearts suggests that myocardium displays a continued cell cupidity that is strictly regulated by the limited release of progenitor cells by the adopted source. When an unregulated cell source is added to the system, cells are delivered to the myocardium. The exploitation of this novel concept may pave the way to the setup of new protocols in cardiac cell therapy.
Collapse
Affiliation(s)
- Giancarlo Forte
- Laboratorio di Cardiologia Molecolare e Cellulare, Dipartimento di Medicina Interna, Università di Roma Tor Vergata, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Jung JP, Squirrell JM, Lyons GE, Eliceiri KW, Ogle BM. Imaging cardiac extracellular matrices: a blueprint for regeneration. Trends Biotechnol 2011; 30:233-40. [PMID: 22209562 DOI: 10.1016/j.tibtech.2011.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 11/19/2022]
Abstract
Once damaged, cardiac tissue does not readily repair and is therefore a primary target of regenerative therapies. One regenerative approach is the development of scaffolds that functionally mimic the cardiac extracellular matrix (ECM) to deliver stem cells or cardiac precursor populations to the heart. Technological advances in micro/nanotechnology, stem cell biology, biomaterials and tissue decellularization have propelled this promising approach forward. Surprisingly, technological advances in optical imaging methods have not been fully utilized in the field of cardiac regeneration. Here, we describe and provide examples to demonstrate how advanced imaging techniques could revolutionize how ECM-mimicking cardiac tissues are informed and evaluated.
Collapse
Affiliation(s)
- Jangwook P Jung
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
44
|
Fernandes S, Kuklok S, McGonigle J, Reinecke H, Murry CE. Synthetic matrices to serve as niches for muscle cell transplantation. Cells Tissues Organs 2011; 195:48-59. [PMID: 22005610 DOI: 10.1159/000331414] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Poor cell retention and limited cell survival after grafting are major limitations of cell therapy. Recent studies showed that the use of matrices as vehicles at the time of cell injection can significantly improve cell engraftment by providing an appropriate structure and physical support for the injected cells. Properly designed matrices can also promote the organization of the cells into a functioning cardiac-like tissue and enhance integration between the host and the engrafted tissue. Furthermore, the use of an injectable biomaterial provides an opportunity to release in situ bioactive molecules that can further enhance the beneficial effects of cell transplantation. In this article we review a large variety of biologically derived synthetic and hybrid materials that have been tested as matrices for cardiac repair. We summarize the optimal parameters required for an ideal matrix including biocompatibility, injectability, degradation rate, and mechanical properties. Using an in vivo subcutaneous grafting model, we also provide novel data involving a side-by-side comparison of six synthetic matrices derived from maltodextrin. By systematically varying polymer molecular weight, cross-link density, and availability of cell adhesion motifs, a synthetic matrix was identified that supported skeletal myotube formation similar to Matrigel™. Our results emphasize not only the need to have a range of tunable matrices for cardiac cell therapy but also the importance of further characterizing the physical properties required for an ideal injectable matrix.
Collapse
Affiliation(s)
- Sarah Fernandes
- Center for Cardiovascular Biology, University of Washington, Seattle, USA
| | | | | | | | | |
Collapse
|
45
|
Hudson JE, Brooke G, Blair C, Wolvetang E, Cooper-White JJ. Development of Myocardial Constructs Using Modulus-Matched Acrylated Polypropylene Glycol Triol Substrate and Different Nonmyocyte Cell Populations. Tissue Eng Part A 2011; 17:2279-89. [DOI: 10.1089/ten.tea.2010.0743] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- James E. Hudson
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Gary Brooke
- School of Medicine, The University of Queensland, Brisbane, Australia
| | - Chris Blair
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Ernst Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Justin John Cooper-White
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, Australia
| |
Collapse
|
46
|
Stem cell integrins: Implications for ex-vivo culture and cellular therapies. Stem Cell Res 2011; 6:1-12. [DOI: 10.1016/j.scr.2010.09.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 09/28/2010] [Accepted: 09/28/2010] [Indexed: 12/15/2022] Open
|
47
|
Kruegel J, Miosge N. Basement membrane components are key players in specialized extracellular matrices. Cell Mol Life Sci 2010; 67:2879-95. [PMID: 20428923 PMCID: PMC2921489 DOI: 10.1007/s00018-010-0367-x] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 03/17/2010] [Accepted: 03/24/2010] [Indexed: 01/11/2023]
Abstract
More than three decades ago, basement membranes (BMs) were described as membrane-like structures capable of isolating a cell from and connecting a cell to its environment. Since this time, it has been revealed that BMs are specialized extracellular matrices (sECMs) with unique components that support important functions including differentiation, proliferation, migration, and chemotaxis of cells during development. The composition of these sECM is as unique as the tissues to which they are localized, opening the possibility that such matrices can fulfill distinct functions. Changes in BM composition play significant roles in facilitating the development of various diseases. Furthermore, tissues have to provide sECM for their stem cells during development and for their adult life. Here, we briefly review the latest research on these unique sECM and their components with a special emphasis on embryonic and adult stem cells and their niches.
Collapse
Affiliation(s)
- Jenny Kruegel
- Tissue Regeneration Work Group, Department of Prosthodontics, Georg August University, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Nicolai Miosge
- Tissue Regeneration Work Group, Department of Prosthodontics, Georg August University, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| |
Collapse
|