1
|
Zhang J, Zhang S, Yang Y, Liu L. Transplantation of umbilical cord blood-derived mesenchymal stem cells as therapy for adriamycin induced-cardiomyopathy. Bioengineered 2022; 13:9564-9574. [PMID: 35387551 PMCID: PMC9161987 DOI: 10.1080/21655979.2022.2061145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) have been reported to possess cardioprotective effects in diseases. However, its effects on cardiomyopathy remain unclear. This study aimed to the therapeutic effects of UCBMSC transplantation on adriamycin (ADR)-induced cardiomyopathy. UCBMSCs isolated from human UCB were identified by detecting surface markers (CD29, CD90, CD34, and CD45) using flow cytometry. The effect of UCBMSCs on left ventricular end-diastolic dimension (LVEDD), left ventricular systolic end-diastolic diameter (LVESD), left ventricular ejection fraction (LVEF), and left ventricular fraction shortening (LVFS) were determined by echocardiography. Histological changes were observed by HE and Masson staining. The serum levels of collagen-I (Col-I), brain natriuretic peptide (BNP), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), creatine kinase (CK), CK-MB, interleukin (IL)-6, IL-10, and tumor necrosis factor alpha (TNF-α) were measured by corresponding kits. The protein levels of IL-6, IL-10, and TNF-α were measured by Western blotting. The isolated UCBMSCs manifested the positive expression of CD29 and CD90, and the negative expression of CD34 and CD45. UCBMSC transplantation significantly reduced LVEDD and LVESD, and increased LVEF and LVFS in ADR-induced cardiomyopathy model rats. Cardiac injury and high collagen deposition in model rats were alleviated by UCBMSC treatment. Moreover, UCBMSCs decreased the serum levels of Col-I, BNP, AST, LDH, CK, CK-MB, IL-6, IL-10, and TNF-α in model rats. Overall, UCBMSCs exert the therapeutic effects on ADR-induced cardiomyopathy through recovering the myocadiac function and alleviating the inflammatory response.
Collapse
Affiliation(s)
- Jingyue Zhang
- Department of Hematologic Oncology, Dalian Municipal Women and Children's Medical Center, Dalian, Liaoning, China
| | - Shiheng Zhang
- Department of Hematologic Oncology, Dalian Municipal Women and Children's Medical Center, Dalian, Liaoning, China
| | - Yueming Yang
- Department of Hematologic Oncology, Dalian Municipal Women and Children's Medical Center, Dalian, Liaoning, China
| | - Ling Liu
- Department of Hematologic Oncology, Dalian Municipal Women and Children's Medical Center, Dalian, Liaoning, China
| |
Collapse
|
2
|
Tajabadi M, Goran Orimi H, Ramzgouyan MR, Nemati A, Deravi N, Beheshtizadeh N, Azami M. Regenerative strategies for the consequences of myocardial infarction: Chronological indication and upcoming visions. Biomed Pharmacother 2021; 146:112584. [PMID: 34968921 DOI: 10.1016/j.biopha.2021.112584] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Heart muscle injury and an elevated troponin level signify myocardial infarction (MI), which may result in defective and uncoordinated segments, reduced cardiac output, and ultimately, death. Physicians apply thrombolytic therapy, coronary artery bypass graft (CABG) surgery, or percutaneous coronary intervention (PCI) to recanalize and restore blood flow to the coronary arteries, albeit they were not convincingly able to solve the heart problems. Thus, researchers aim to introduce novel substitutional therapies for regenerating and functionalizing damaged cardiac tissue based on engineering concepts. Cell-based engineering approaches, utilizing biomaterials, gene, drug, growth factor delivery systems, and tissue engineering are the most leading studies in the field of heart regeneration. Also, understanding the primary cause of MI and thus selecting the most efficient treatment method can be enhanced by preparing microdevices so-called heart-on-a-chip. In this regard, microfluidic approaches can be used as diagnostic platforms or drug screening in cardiac disease treatment. Additionally, bioprinting technique with whole organ 3D printing of human heart with major vessels, cardiomyocytes and endothelial cells can be an ideal goal for cardiac tissue engineering and remarkable achievement in near future. Consequently, this review discusses the different aspects, advancements, and challenges of the mentioned methods with presenting the advantages and disadvantages, chronological indications, and application prospects of various novel therapeutic approaches.
Collapse
Affiliation(s)
- Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran
| | - Hanif Goran Orimi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Roya Ramzgouyan
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alireza Nemati
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
3
|
Chang T, Liu C, Lu K, Wu Y, Xu M, Yu Q, Shen Z, Jiang T, Zhang Y. Biomaterials based cardiac patches for the treatment of myocardial infarction. JOURNAL OF MATERIALS SCIENCE & TECHNOLOGY 2021; 94:77-89. [DOI: 10.1016/j.jmst.2021.03.062] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Wharton's Jelly Mesenchymal Stromal Cells and Derived Extracellular Vesicles as Post-Myocardial Infarction Therapeutic Toolkit: An Experienced View. Pharmaceutics 2021; 13:pharmaceutics13091336. [PMID: 34575412 PMCID: PMC8471243 DOI: 10.3390/pharmaceutics13091336] [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: 07/09/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
Outstanding progress has been achieved in developing therapeutic options for reasonably alleviating symptoms and prolonging the lifespan of patients suffering from myocardial infarction (MI). Current treatments, however, only partially address the functional recovery of post-infarcted myocardium, which is in fact the major goal for effective primary care. In this context, we largely investigated novel cell and TE tissue engineering therapeutic approaches for cardiac repair, particularly using multipotent mesenchymal stromal cells (MSC) and natural extracellular matrices, from pre-clinical studies to clinical application. A further step in this field is offered by MSC-derived extracellular vesicles (EV), which are naturally released nanosized lipid bilayer-delimited particles with a key role in cell-to-cell communication. Herein, in this review, we further describe and discuss the rationale, outcomes and challenges of our evidence-based therapy approaches using Wharton's jelly MSC and derived EV in post-MI management.
Collapse
|
5
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|
6
|
Shi L, Ren J, Li J, Wang D, Wang Y, Qin T, Li X, Zhang G, Li C, Wang Y. Extracellular vesicles derived from umbilical cord mesenchymal stromal cells alleviate pulmonary fibrosis by means of transforming growth factor-β signaling inhibition. Stem Cell Res Ther 2021; 12:230. [PMID: 33845892 PMCID: PMC8041243 DOI: 10.1186/s13287-021-02296-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF), the end point of interstitial lung diseases, is characterized by myofibroblast over differentiation and excessive extracellular matrix accumulation, leading to progressive organ dysfunction and usually a terminal outcome. Studies have shown that umbilical cord-derived mesenchymal stromal cells (uMSCs) could alleviate PF; however, the underlying mechanism remains to be elucidated. METHODS The therapeutic effects of uMSC-derived extracellular vesicles (uMSC-EVs) on PF were evaluated using bleomycin (BLM)-induced mouse models. Then, the role and mechanism of uMSC-EVs in inhibiting myofibroblast differentiation were investigated in vivo and in vitro. RESULTS Treatment with uMSC-EVs alleviated the PF and enhanced the proliferation of alveolar epithelial cells in BLM-induced mice, thus improved the life quality, including the survival rate, body weight, fibrosis degree, and myofibroblast over differentiation of lung tissue. Moreover, these effects of uMSC-EVs on PF are likely achieved by inhibiting the transforming growth factor-β (TGF-β) signaling pathway, evidenced by decreased expression levels of TGF-β2 and TGF-βR2. Using mimics of uMSC-EV-specific miRNAs, we found that miR-21 and miR-23, which are highly enriched in uMSC-EVs, played a critical role in inhibiting TGF-β2 and TGF-βR2, respectively. CONCLUSION The effects of uMSCs on PF alleviation are likely achieved via EVs, which reveals a new role of uMSC-EV-derived miRNAs, opening a novel strategy for PF treatment in the clinical setting.
Collapse
Affiliation(s)
- Liyan Shi
- China-Japan Union Hospital of Jilin University, 126 Xiantai St., Changchun, 130033, Jilin, China
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China
| | - Jiping Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China
| | - Dongxu Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China
| | - Yusu Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China
| | - Tao Qin
- School of Ecology and Environment, Northwestern Polytechnical University, 1 Dongxiang Rd, Xi'an, 710129, Shaanxi, China
| | - Xiuying Li
- China-Japan Union Hospital of Jilin University, 126 Xiantai St., Changchun, 130033, Jilin, China
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China.
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), 4899 Juye St., Changchun, 130112, Jilin, China.
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, Jilin, China.
| | - Yimin Wang
- China-Japan Union Hospital of Jilin University, 126 Xiantai St., Changchun, 130033, Jilin, China.
| |
Collapse
|
7
|
Monguió-Tortajada M, Prat-Vidal C, Moron-Font M, Clos-Sansalvador M, Calle A, Gastelurrutia P, Cserkoova A, Morancho A, Ramírez MÁ, Rosell A, Bayes-Genis A, Gálvez-Montón C, Borràs FE, Roura S. Local administration of porcine immunomodulatory, chemotactic and angiogenic extracellular vesicles using engineered cardiac scaffolds for myocardial infarction. Bioact Mater 2021; 6:3314-3327. [PMID: 33778207 PMCID: PMC7973387 DOI: 10.1016/j.bioactmat.2021.02.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The administration of extracellular vesicles (EV) from mesenchymal stromal cells (MSC) is a promising cell-free nanotherapy for tissue repair after myocardial infarction (MI). However, the optimal EV delivery strategy remains undetermined. Here, we designed a novel MSC-EV delivery, using 3D scaffolds engineered from decellularised cardiac tissue as a cell-free product for cardiac repair. EV from porcine cardiac adipose tissue-derived MSC (cATMSC) were purified by size exclusion chromatography (SEC), functionally analysed and loaded to scaffolds. cATMSC-EV markedly reduced polyclonal proliferation and pro-inflammatory cytokines production (IFNγ, TNFα, IL12p40) of allogeneic PBMC. Moreover, cATMSC-EV recruited outgrowth endothelial cells (OEC) and allogeneic MSC, and promoted angiogenesis. Fluorescently labelled cATMSC-EV were mixed with peptide hydrogel, and were successfully retained in decellularised scaffolds. Then, cATMSC-EV-embedded pericardial scaffolds were administered in vivo over the ischemic myocardium in a pig model of MI. Six days from implantation, the engineered scaffold efficiently integrated into the post-infarcted myocardium. cATMSC-EV were detected within the construct and MI core, and promoted an increase in vascular density and reduction in macrophage and T cell infiltration within the damaged myocardium. The confined administration of multifunctional MSC-EV within an engineered pericardial scaffold ensures local EV dosage and release, and generates a vascularised bioactive niche for cell recruitment, engraftment and modulation of short-term post-ischemic inflammation.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Prat-Vidal
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Miriam Moron-Font
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Marta Clos-Sansalvador
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Alexandra Calle
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Paloma Gastelurrutia
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Adriana Cserkoova
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Anna Morancho
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), UAB, Barcelona, Spain
| | - Miguel Ángel Ramírez
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute (VHIR), UAB, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, UAB, Barcelona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc E Borràs
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain.,Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Barcelona, 08500, Spain
| |
Collapse
|
8
|
Montazeri L, Kowsari-Esfahan R, Pahlavan S, Sobat M, Rabbani S, Ansari H, Varzideh F, Barekat M, Rajabi S, Navaee F, Bonakdar S, Renaud P, Braun T, Baharvand H. Oxygen-rich Environment Ameliorates Cell Therapy Outcomes of Cardiac Progenitor Cells for Myocardial Infarction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111836. [PMID: 33579474 DOI: 10.1016/j.msec.2020.111836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 11/20/2022]
Abstract
To some extent, cell therapy for myocardial infarction (MI) has supported the idea of cardiac repair; however, further optimizations are inevitable. Combined approaches that comprise suitable cell sources and supporting molecules considerably improved its effect. Here, we devised a strategy of simultaneous transplantation of human cardiac progenitor cells (CPCs) and an optimized oxygen generating microparticles (MPs) embedded in fibrin hydrogel, which was injected into a left anterior descending artery (LAD) ligating-based rat model of acute myocardial infarction (AMI). Functional parameters of the heart, particularly left ventricular systolic function, markedly improved and reached pre-AMI levels. This functional restoration was well correlated with substantially lower fibrotic tissue formation and greater vascular density in the infarct area. Our novel approach promoted CPCs retention and differentiation into cardiovascular lineages. We propose this novel co-transplantation strategy for more efficient cell therapy of AMI which may function by providing an oxygen-rich microenvironment, and thus regulate cell survival and differentiation.
Collapse
Affiliation(s)
- Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Reza Kowsari-Esfahan
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Motahareh Sobat
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Shahram Rabbani
- Tehran Heart Center, Medical Sciences University of Tehran, Tehran, Iran
| | - Hassan Ansari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fahimeh Varzideh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Barekat
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sarah Rajabi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Navaee
- Microsystems Laboratory, École Polytechnique Fédérale de Lausanne, EPFL-STIIMT- LMIS4, Station 17, Lausanne, 1015, Switzerland
| | | | - Philippe Renaud
- Microsystems Laboratory, École Polytechnique Fédérale de Lausanne, EPFL-STIIMT- LMIS4, Station 17, Lausanne, 1015, Switzerland
| | - Thomas Braun
- Max-Planck Institute for Heart and Lung Research, Department of Cardiac Development and Remodeling, Bad Nauheim, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
9
|
Mei X, Cheng K. Recent Development in Therapeutic Cardiac Patches. Front Cardiovasc Med 2020; 7:610364. [PMID: 33330673 PMCID: PMC7728668 DOI: 10.3389/fcvm.2020.610364] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/03/2020] [Indexed: 01/03/2023] Open
Abstract
For the past decades, heart diseases remain the leading cause of death worldwide. In the adult mammalian heart, damaged cardiomyocytes will be replaced by non-contractile fibrotic scar tissues due to the poor regenerative ability of heart, causing heart failure subsequently. The development of tissue engineering has launched a new medical innovation for heart regeneration. As one of the most outstanding technology, cardiac patches hold the potential to restore cardiac function clinically. Consisted of two components: therapeutic ingredients and substrate scaffolds, the fabrication of cardiac patches requires both advanced bioactive molecules and biomaterials. In this review, we will present the most state-of-the-art cardiac patches and analysis their compositional details. The therapeutic ingredients will be discussed from cell sources to bioactive molecules. In the meanwhile, the recent advances to obtain scaffold biomaterials will be highlighted, including synthetic and natural materials. Also, we have focused on the challenges and potential strategies to fabricate clinically applicable cardiac patches.
Collapse
Affiliation(s)
- Xuan Mei
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
10
|
Chen C, Lou Y, Li XY, Lv ZT, Zhang LQ, Mao W. Mapping current research and identifying hotspots on mesenchymal stem cells in cardiovascular disease. Stem Cell Res Ther 2020; 11:498. [PMID: 33239082 PMCID: PMC7687818 DOI: 10.1186/s13287-020-02009-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have important research value and broad application prospects in the cardiovascular disease. This study provides information on the latest progress, evolutionary path, frontier research hotspots, and future research developmental trends in this field. METHODS A knowledge map was generated by CiteSpace and VOSviewer analysis software based on data obtained from the literature on MSCs in the cardiovascular field. RESULTS The USA and China ranked at the top in terms of the percentage of articles, accounting for 34.306% and 28.550%, respectively. The institution with the highest number of research publications in this field was the University of Miami, followed by the Chinese Academy of Medical Sciences and Harvard University. The research institution with the highest ACI value was Harvard University, followed by the Mayo Clinic and the University of Cincinnati. The top three subjects in terms of the number of published articles were cell biology, cardiovascular system cardiology, and research experimental medicine. The journal with the most publications in this field was Circulation Research, followed by Scientific Reports and Biomaterials. The direction of research on MSCs in the cardiovascular system was divided into four parts: (1) tissue engineering, scaffolds, and extracellular matrix research; (2) cell transplantation, differentiation, proliferation, and signal transduction pathway research; (3) assessment of the efficacy of stem cells from different sources and administration methods in the treatment of acute myocardial infarction, myocardial hypertrophy, and heart failure; and (4) exosomes and extracellular vesicles research. Tissue research is the hotspot and frontier in this field. CONCLUSION MSC research has presented a gradual upward trend in the cardiovascular field. Multidisciplinary intersection is a characteristic of this field. Engineering and materials disciplines are particularly valued and have received attention from researchers. The progress in multidisciplinary research will provide motivation and technical support for the development of this field.
Collapse
Affiliation(s)
- Chan Chen
- Hangzhou Xiaoshan district Hospital of TCM, Jiangnan Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 311201, Zhejiang, China. .,Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Yang Lou
- The first Affiliated Hospital Zhejiang Chinese Medical University, Hangzhou, 311006, Zhejiang, China
| | - Xin-Yi Li
- The first Affiliated Hospital Zhejiang Chinese Medical University, Hangzhou, 311006, Zhejiang, China
| | - Zheng-Tian Lv
- The first Affiliated Hospital Zhejiang Chinese Medical University, Hangzhou, 311006, Zhejiang, China
| | - Lu-Qiu Zhang
- The first Affiliated Hospital Zhejiang Chinese Medical University, Hangzhou, 311006, Zhejiang, China
| | - Wei Mao
- The first Affiliated Hospital Zhejiang Chinese Medical University, Hangzhou, 311006, Zhejiang, China.
| |
Collapse
|
11
|
Prat-Vidal C, Rodríguez-Gómez L, Aylagas M, Nieto-Nicolau N, Gastelurrutia P, Agustí E, Gálvez-Montón C, Jorba I, Teis A, Monguió-Tortajada M, Roura S, Vives J, Torrents-Zapata S, Coca MI, Reales L, Cámara-Rosell ML, Cediel G, Coll R, Farré R, Navajas D, Vilarrodona A, García-López J, Muñoz-Guijosa C, Querol S, Bayes-Genis A. First-in-human PeriCord cardiac bioimplant: Scalability and GMP manufacturing of an allogeneic engineered tissue graft. EBioMedicine 2020; 54:102729. [PMID: 32304998 PMCID: PMC7163319 DOI: 10.1016/j.ebiom.2020.102729] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/24/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Small cardiac tissue engineering constructs show promise for limiting post-infarct sequelae in animal models. This study sought to scale-up a 2-cm2 preclinical construct into a human-size advanced therapy medicinal product (ATMP; PeriCord), and to test it in a first-in-human implantation. Methods The PeriCord is a clinical-size (12–16 cm2) decellularised pericardial matrix colonised with human viable Wharton's jelly-derived mesenchymal stromal cells (WJ-MSCs). WJ-MSCs expanded following good manufacturing practices (GMP) met safety and quality standards regarding the number of cumulative population doublings, genomic stability, and sterility. Human decellularised pericardial scaffolds were tested for DNA content, matrix stiffness, pore size, and absence of microbiological growth. Findings PeriCord implantation was surgically performed on a large non-revascularisable scar in the inferior wall of a 63-year-old male patient. Coronary artery bypass grafting was concomitantly performed in the non-infarcted area. At implantation, the 16-cm2 pericardial scaffold contained 12·5 × 106 viable WJ-MSCs (85·4% cell viability; <0·51 endotoxin units (EU)/mL). Intraoperative PeriCord delivery was expeditious, and secured with surgical glue. The post-operative course showed non-adverse reaction to the PeriCord, without requiring host immunosuppression. The three-month clinical follow-up was uneventful, and three-month cardiac magnetic resonance imaging showed ~9% reduction in scar mass in the treated area. Interpretation This preliminary report describes the development of a scalable clinical-size allogeneic PeriCord cardiac bioimplant, and its first-in-human implantation. Funding La Marató de TV3 Foundation, Government of Catalonia, Catalan Society of Cardiology, “La Caixa” Banking Foundation, Spanish Ministry of Science, Innovation and Universities, Institute of Health Carlos III, and the European Regional Development Fund.
Collapse
Affiliation(s)
- Cristina Prat-Vidal
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Luciano Rodríguez-Gómez
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Miriam Aylagas
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Autonomous University of Barcelona, Barcelona, Spain
| | | | - Paloma Gastelurrutia
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Elba Agustí
- Barcelona Tissue Bank (BTB), Banc de Sang i Teixits (BST), Barcelona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignasi Jorba
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, CIBERES, University of Barcelona, Barcelona, Spain
| | - Albert Teis
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Marta Monguió-Tortajada
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
| | - Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquim Vives
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain
| | - Silvia Torrents-Zapata
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - María Isabel Coca
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Laura Reales
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - María Luisa Cámara-Rosell
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Germán Cediel
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Ruth Coll
- Research and Education. Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Ramon Farré
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona-IDIBAPS-CIBERES, Barcelona, Spain
| | - Daniel Navajas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, CIBERES, University of Barcelona, Barcelona, Spain
| | - Anna Vilarrodona
- Barcelona Tissue Bank (BTB), Banc de Sang i Teixits (BST), Barcelona, Spain
| | - Joan García-López
- Research and Education. Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Christian Muñoz-Guijosa
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Sergi Querol
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Autonomous University of Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain.
| |
Collapse
|
12
|
Eissa M, Elarabany N, Hyder A. In vitro efficacy of liver microenvironment in bone marrow mesenchymal stem cell differentiation. In Vitro Cell Dev Biol Anim 2020; 56:341-348. [PMID: 32270392 DOI: 10.1007/s11626-020-00436-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/08/2020] [Indexed: 12/31/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) represent an interesting alternative to liver or hepatocyte transplantation to treat liver injuries. Many studies have reported that MSCs can treat several diseases, including liver damage, just by injection into the bloodstream, without evidence of differentiation. The improvements were attributed to the organotrophic factors, low immunogenicity, immunomodulatory, and anti-inflammatory effects of MSCs, rather than their differentiation. The aim of the present study was to answer the question of whether the presence of BM-MSCs in the hepatic microenvironment will lead to their differentiation to functional hepatocyte-like cells. The hepatic microenvironment was mimicked in vitro by culture for 21 d with liver extract. The resulted cells expressed marker genes of the hepatic lineage including AFP, CK18, and Hnf4a. Functionally, they were able to detoxify ammonia into urea, to store glycogen as observed by PAS staining, and to synthesize glucose from pyruvate/lactate mixture. Phenotypically, the expression of MSC surface markers CD90 and CD105 decreased by differentiation. This evidenced differentiation into hepatocyte-like cells was accompanied by a downregulation of the stem cell marker genes sox2 and Nanog and the cell cycle regulatory genes ANAPC2, CDC2, Cyclin A1, and ABL1. The present results suggest a clear differentiation of BM-MSCs into functional hepatocyte-like cells by the extracted liver microenvironment. This differentiation is confirmed by a decrease in the stemness and mitotic activities. Tracking transplanted BM-MSCs and proving their in vivo differentiation remains to be elucidated.
Collapse
Affiliation(s)
- Manar Eissa
- Faculty of Science, Damietta University, New Damietta, 34517, Egypt
| | - Naglaa Elarabany
- Faculty of Science, Damietta University, New Damietta, 34517, Egypt
| | - Ayman Hyder
- Faculty of Science, Damietta University, New Damietta, 34517, Egypt.
| |
Collapse
|
13
|
Huang K, Ozpinar EW, Su T, Tang J, Shen D, Qiao L, Hu S, Li Z, Liang H, Mathews K, Scharf V, Freytes DO, Cheng K. An off-the-shelf artificial cardiac patch improves cardiac repair after myocardial infarction in rats and pigs. Sci Transl Med 2020; 12:eaat9683. [PMID: 32269164 PMCID: PMC7293901 DOI: 10.1126/scitranslmed.aat9683] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 02/26/2019] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
Cell therapy has been a promising strategy for cardiac repair after injury or infarction; however, low retention and engraftment of transplanted cells limit potential therapeutic efficacy. Seeding scaffold material with cells to create cardiac patches that are transplanted onto the surface of the heart can overcome these limitations. However, because patches need to be freshly prepared to maintain cell viability, long-term storage is not feasible and limits clinical applicability. Here, we developed an off-the-shelf therapeutic cardiac patch composed of a decellularized porcine myocardial extracellular matrix scaffold and synthetic cardiac stromal cells (synCSCs) generated by encapsulating secreted factors from isolated human cardiac stromal cells. This fully acellular artificial cardiac patch (artCP) maintained its potency after long-term cryopreservation. In a rat model of acute myocardial infarction, transplantation of the artCP supported cardiac recovery by reducing scarring, promoting angiomyogenesis, and boosting cardiac function. The safety and efficacy of the artCP were further confirmed in a porcine model of myocardial infarction. The artCP is a clinically feasible, easy-to-store, and cell-free alternative to myocardial repair using cell-based cardiac patches.
Collapse
Affiliation(s)
- Ke Huang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| | - Emily W Ozpinar
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Teng Su
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Deliang Shen
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Li Qiao
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Hongxia Liang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Kyle Mathews
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Valery Scharf
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Donald O Freytes
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA.
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
14
|
Anil Kumar S, Alonzo M, Allen SC, Abelseth L, Thakur V, Akimoto J, Ito Y, Willerth SM, Suggs L, Chattopadhyay M, Joddar B. A Visible Light-Cross-Linkable, Fibrin-Gelatin-Based Bioprinted Construct with Human Cardiomyocytes and Fibroblasts. ACS Biomater Sci Eng 2019; 5:4551-4563. [PMID: 32258387 PMCID: PMC7117097 DOI: 10.1021/acsbiomaterials.9b00505] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In this study, fibrin was added to a photo-polymerizable gelatin-based bioink mixture to fabricate cardiac cell-laden constructs seeded with human induced pluripotent stem cell-derived cardiomyocytes (iPS-CM) or CM cell lines with cardiac fibroblasts (CF). The extensive use of platelet-rich fibrin, its capacity to offer patient specificity, and the similarity in composition to surgical glue prompted us to include fibrin in the existing bioink composition. The cell-laden bioprinted constructs were cross-linked to retain a herringbone pattern via a two-step procedure including the visible light cross-linking of furfuryl-gelatin followed by the chemical cross-linking of fibrinogen via thrombin and calcium chloride. The printed constructs revealed an extremely porous, networked structure that afforded long-term in vitro stability. Cardiomyocytes printed within the sheet structure showed excellent viability, proliferation, and expression of the troponin I cardiac marker. We extended the utility of this fibrin-gelatin bioink toward coculturing and coupling of CM and cardiac fibroblasts (CF), the interaction of which is extremely important for maintenance of normal physiology of the cardiac wall in vivo. This enhanced "cardiac construct" can be used for drug cytotoxicity screening or unraveling triggers for heart diseases in vitro.
Collapse
Affiliation(s)
- Shweta Anil Kumar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, M201 Metallurgy Building, United States
| | - Matthew Alonzo
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, M201 Metallurgy Building, United States
| | - Shane C. Allen
- Department of Biomedical Engineering, The University of Texas at Austin, 110 Inner Campus Drive, Austin, Texas 78712, United States
| | - Laila Abelseth
- Department of Mechanical Engineering, University of Victoria, Engineering Office Wing, Room 548, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
- Biomedical Engineering Program, University of Victoria, Engineering Office Wing, Room 548, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
| | - Vikram Thakur
- Department of Molecular and Translational Medicine, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, Texas 79905, United States
| | - Jun Akimoto
- Nano Medical Engineering Laboratory, RIKEN Custer for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Custer for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, Engineering Office Wing, Room 548, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
- Biomedical Engineering Program, University of Victoria, Engineering Office Wing, Room 548, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Engineering Office Wing, Room 548, 3800 Finnerty Road, Victoria, British Columbia V8P 5C2, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, 818 West 10th Avenue, Vancouver, British Columbia V5Z 1M9, Canada
| | - Laura Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, 110 Inner Campus Drive, Austin, Texas 78712, United States
| | - Munmun Chattopadhyay
- Department of Molecular and Translational Medicine, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, Texas 79905, United States
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, M201 Metallurgy Building, United States
- Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas 79968, United States
- Nano Medical Engineering Laboratory, RIKEN Custer for Pioneering Research, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
15
|
Saberianpour S, Heidarzadeh M, Geranmayeh MH, Hosseinkhani H, Rahbarghazi R, Nouri M. Tissue engineering strategies for the induction of angiogenesis using biomaterials. J Biol Eng 2018; 12:36. [PMID: 30603044 PMCID: PMC6307144 DOI: 10.1186/s13036-018-0133-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is touted as a fundamental procedure in the regeneration and restoration of different tissues. The induction of de novo blood vessels seems to be vital to yield a successful cell transplantation rate loaded on various scaffolds. Scaffolds are natural or artificial substances that are considered as one of the means for delivering, aligning, maintaining cell connection in a favor of angiogenesis. In addition to the potential role of distinct scaffold type on vascularization, the application of some strategies such as genetic manipulation, and conjugation of pro-angiogenic factors could intensify angiogenesis potential. In the current review, we focused on the status of numerous scaffolds applicable in the field of vascular biology. Also, different strategies and priming approaches useful for the induction of pro-angiogenic signaling pathways were highlighted.
Collapse
Affiliation(s)
- Shirin Saberianpour
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 2Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Heidarzadeh
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
| | - Mohammad Hossein Geranmayeh
- 3Neuroscience Research Center, Imam Reza Medical Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 5Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- 2Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 5Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Tang J, Wang J, Huang K, Ye Y, Su T, Qiao L, Hensley MT, Caranasos TG, Zhang J, Gu Z, Cheng K. Cardiac cell-integrated microneedle patch for treating myocardial infarction. SCIENCE ADVANCES 2018; 4:eaat9365. [PMID: 30498778 PMCID: PMC6261659 DOI: 10.1126/sciadv.aat9365] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 10/26/2018] [Indexed: 05/17/2023]
Abstract
We engineered a microneedle patch integrated with cardiac stromal cells (MN-CSCs) for therapeutic heart regeneration after acute myocardial infarction (MI). To perform cell-based heart regeneration, cells are currently delivered to the heart via direct muscle injection, intravascular infusion, or transplantation of epicardial patches. The first two approaches suffer from poor cell retention, while epicardial patches integrate slowly with host myocardium. Here, we used polymeric MNs to create "channels" between host myocardium and therapeutic CSCs. These channels allow regenerative factors secreted by CSCs to be released into the injured myocardium to promote heart repair. In the rat MI model study, the application of the MN-CSC patch effectively augmented cardiac functions and enhanced angiomyogenesis. In the porcine MI model study, MN-CSC patch application was nontoxic and resulted in cardiac function protection. The MN system represents an innovative approach delivering therapeutic cells for heart regeneration.
Collapse
Affiliation(s)
- Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Jinqiang Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Teng Su
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Li Qiao
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
- Department of Cardiology, The Second Affiliated Hospital of Hebei Medicial University, Shijiazhuang, Hebei 050000, China
| | - Michael Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
| | - Thomas George Caranasos
- Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, USA
- Corresponding author. (K.C.); (Z.G.)
| | - Ke Cheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27607, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Corresponding author. (K.C.); (Z.G.)
| |
Collapse
|
17
|
Yong KW, Choi JR, Mohammadi M, Mitha AP, Sanati-Nezhad A, Sen A. Mesenchymal Stem Cell Therapy for Ischemic Tissues. Stem Cells Int 2018; 2018:8179075. [PMID: 30402112 PMCID: PMC6196793 DOI: 10.1155/2018/8179075] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/01/2018] [Accepted: 08/29/2018] [Indexed: 12/13/2022] Open
Abstract
Ischemic diseases such as myocardial infarction, ischemic stroke, and critical limb ischemia are immense public health challenges. Current pharmacotherapy and surgical approaches are insufficient to completely heal ischemic diseases and are associated with a considerable risk of adverse effects. Alternatively, human mesenchymal stem cells (hMSCs) have been shown to exhibit immunomodulation, angiogenesis, and paracrine secretion of bioactive factors that can attenuate inflammation and promote tissue regeneration, making them a promising cell source for ischemic disease therapy. This review summarizes the pathogenesis of ischemic diseases, discusses the potential therapeutic effects and mechanisms of hMSCs for these diseases, and provides an overview of challenges of using hMSCs clinically for treating ischemic diseases.
Collapse
Affiliation(s)
- Kar Wey Yong
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Jane Ru Choi
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, V6T 1Z4, Canada
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Mehdi Mohammadi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Alim P. Mitha
- Department of Clinical Neurosciences, Foothills Medical Centre, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- Center of Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
- Center of Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
18
|
Monguió-Tortajada M, Roura S, Gálvez-Montón C, Franquesa M, Bayes-Genis A, Borràs FE. Mesenchymal Stem Cells Induce Expression of CD73 in Human Monocytes In Vitro and in a Swine Model of Myocardial Infarction In Vivo. Front Immunol 2017; 8:1577. [PMID: 29209319 PMCID: PMC5701925 DOI: 10.3389/fimmu.2017.01577] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/02/2017] [Indexed: 11/30/2022] Open
Abstract
The ectoenzymes CD39 and CD73 regulate the purinergic signaling through the hydrolysis of adenosine triphosphate (ATP)/ADP to AMP and to adenosine (Ado), respectively. This shifts the pro-inflammatory milieu induced by extracellular ATP to the anti-inflammatory regulation by Ado. Mesenchymal stem cells (MSCs) have potent immunomodulatory capabilities, including monocyte modulation toward an anti-inflammatory phenotype aiding tissue repair. In vitro, we observed that human cardiac adipose tissue-derived MSCs (cATMSCs) and umbilical cord MSCs similarly polarize monocytes toward a regulatory M2 phenotype, which maintained the expression of CD39 and induced expression of CD73 in a cell contact dependent fashion, correlating with increased functional activity. In addition, the local treatment with porcine cATMSCs using an engineered bioactive graft promoted the in vivo CD73 expression on host monocytes in a swine model of myocardial infarction. Our results suggest the upregulation of ectonucleotidases on MSC-conditioned monocytes as an effective mechanism to amplify the long-lasting immunomodulatory and healing effects of MSCs delivery.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.,Center of Regenerative Medicine in Barcelona, Barcelona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Marcella Franquesa
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.,Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Francesc E Borràs
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| |
Collapse
|
19
|
Roura S, Gálvez-Montón C, Mirabel C, Vives J, Bayes-Genis A. Mesenchymal stem cells for cardiac repair: are the actors ready for the clinical scenario? Stem Cell Res Ther 2017; 8:238. [PMID: 29078809 PMCID: PMC5658929 DOI: 10.1186/s13287-017-0695-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
For years, sufficient progress has been made in treating heart failure following myocardial infarction; however, the social and economic burdens and the costs to world health systems remain high. Moreover, treatment advances have not resolved the underlying problem of functional heart tissue loss. In this field of research, for years we have actively explored innovative biotherapies for cardiac repair. Here, we present a general, critical overview of our experience in using mesenchymal stem cells, derived from cardiac adipose tissue and umbilical cord blood, in a variety of cell therapy and tissue engineering approaches. We also include the latest advances and future challenges, including good manufacturing practice and regulatory issues. Finally, we evaluate whether recent approaches hold potential for reliable translation to clinical trials.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,Center of Regenerative Medicine in Barcelona, Barcelona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,ICREC (Heart Failure and Cardiac Regeneration) Research Programme, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Carretera de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Clémentine Mirabel
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain. .,Cardiology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain. .,Heart Institute, Hospital Universitari Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
20
|
Cao W, Chang YF, Zhao AC, Chen BD, Liu F, Ma YT, Ma X. Synergistic cardioprotective effects of rAAV9-CyclinA2 combined with fibrin glue in rats after myocardial infarction. J Mol Histol 2017. [PMID: 28643114 DOI: 10.1007/s10735-017-9725-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The present study aimed to investigate the protective effects of rAAV9-CyclinA2 combined with fibrin glue (FG) in vivo in rats after myocardial infarction (MI). Ninety male Sprague-Dawley rats were randomized into 6 groups (15 in each group): sham, MI, rAAV9-green fluorescent protein (GFP) + MI, rAAV9-CyclinA2 + MI, FG + MI, and rAAV9-CyclinA2 + FG + MI. Packed virus (5 × 1011vg/ml) in 150 µl of normal saline or FG was injected into the infarcted myocardium at five locations in rAAV9-GFP + MI, rAAV9-CyclinA2 + MI, and rAAV9-CyclinA2 + FG + MI groups. The sham, MI, and FG + MI groups were injected with an equal volume of normal saline or FG at the same sites. Five weeks after injection, echocardiography was performed to evaluate the left ventricular function. The expressions of CyclinA2, proliferating cell nuclear antigen (PCNA), and phospho-histone-H3 (H3P), vascular density, and infarct area were assessed by Western blot, immunohistochemistry, immunofluorescence, and Masson staining. As a result, the combination of rAAV9-CyclinA2 and FG increased ejection fraction and fractional shortening compared with FG or rAAV9-CyclinA2 alone. The expression level of CyclinA2 was significantly higher in the rAAV9-CyclinA2 + FG + MI group compared with the rAAV9-CyclinA2 + MI and FG + MI groups (70.1 ± 1.86% vs. 14.74 ± 2.02%, P < 0.01; or vs. 50.13 ± 3.80%; P < 0.01). A higher expression level of PCNA and H3P was found in the rAAV9-CyclinA2 + FG + MI group compared with other groups. Comparing with other experiment groups, collagen deposition and the infarct size significantly decreased in rAAV9-CyclinA2 + Fibrin + MI group. The vascular density was much higher in the rAAV9-CyclinA2 + FG + MI group compared with the rAAV9-CyclinA2 + MI group. We concluded that fibrin glue combined with rAAV9-CyclinA2 was found to be effective in cardiac remodeling and improving myocardial protection.
Collapse
Affiliation(s)
- Wen Cao
- Department of Cardiology, Second People's Hospital of Wuxi, 68 Zhongshan Road, Wuxi, 214000, China
| | - Ya-Fei Chang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Road, Urumqi, 830054, China
| | - Ai-Chao Zhao
- Department of Critical Care Medicine, People's Hospital of Dezhou, 1751 Xihu Road, Dezhou, 253000, China
| | - Bang-Dang Chen
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Road, Urumqi, 830054, China
| | - Fen Liu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Road, Urumqi, 830054, China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Road, Urumqi, 830054, China.
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Road, Urumqi, 830054, China.
| |
Collapse
|
21
|
Clayton ZE, Yuen GS, Sadeghipour S, Hywood JD, Wong JW, Huang NF, Ng MK, Cooke JP, Patel S. A comparison of the pro-angiogenic potential of human induced pluripotent stem cell derived endothelial cells and induced endothelial cells in a murine model of peripheral arterial disease. Int J Cardiol 2017; 234:81-89. [DOI: 10.1016/j.ijcard.2017.01.125] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 12/28/2016] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
|
22
|
Cho HM, Kim PH, Chang HK, Shen YM, Bonsra K, Kang BJ, Yum SY, Kim JH, Lee SY, Choi MC, Kim HH, Jang G, Cho JY. Targeted Genome Engineering to Control VEGF Expression in Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells: Potential Implications for the Treatment of Myocardial Infarction. Stem Cells Transl Med 2017; 6:1040-1051. [PMID: 28186692 PMCID: PMC5442764 DOI: 10.1002/sctm.16-0114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/11/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022] Open
Abstract
Human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs) exhibit potency for the regeneration of infarcted hearts. Vascular endothelial growth factor (VEGF) is capable of inducing angiogenesis and can boost stem cell‐based therapeutic effects. However, high levels of VEGF can cause abnormal blood vessel growth and hemangiomas. Thus, a controllable system to induce therapeutic levels of VEGF is required for cell therapy. We generated an inducible VEGF‐secreting stem cell (VEGF/hUCB‐MSC) that controls the expression of VEGF and tested the therapeutic efficacy in rat myocardial infarction (MI) model to apply functional stem cells to MI. To introduce the inducible VEGF gene cassette into a safe harbor site of the hUCB‐MSC chromosome, the transcription activator‐like effector nucleases system was used. After confirming the integration of the cassette into the locus, VEGF secretion in physiological concentration from VEGF/hUCB‐MSCs after doxycycline (Dox) induction was proved in conditioned media. VEGF secretion was detected in mice implanted with VEGF/hUCB‐MSCs grown via a cell sheet system. Vessel formation was induced in mice transplanted with Matrigel containing VEGF/hUCB‐MSCs treated with Dox. Moreover, seeding of the VEGF/hUCB‐MSCs onto the cardiac patch significantly improved the left ventricle ejection fraction and fractional shortening in a rat MI model upon VEGF induction. Induced VEGF/hUCB‐MSC patches significantly decreased the MI size and fibrosis and increased muscle thickness, suggesting improved survival of cardiomyocytes and protection from MI damage. These results suggest that our inducible VEGF‐secreting stem cell system is an effective therapeutic approach for the treatment of MI. Stem Cells Translational Medicine2017;6:1040–1051
Collapse
Affiliation(s)
- Hyun-Min Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Pyung-Hwan Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyun-Kyung Chang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yi-Ming Shen
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kwaku Bonsra
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Byung-Jae Kang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Soo-Young Yum
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Joo-Hyun Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - So-Yeong Lee
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Min-Cheol Choi
- Department of Veterinary Radiology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, College of Medicine, Yonsei University, Seoul, South Korea
| | - Goo Jang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
23
|
Monguió-Tortajada M, Roura S, Gálvez-Montón C, Pujal JM, Aran G, Sanjurjo L, Franquesa M, Sarrias MR, Bayes-Genis A, Borràs FE. Nanosized UCMSC-derived extracellular vesicles but not conditioned medium exclusively inhibit the inflammatory response of stimulated T cells: implications for nanomedicine. Theranostics 2017; 7:270-284. [PMID: 28042333 PMCID: PMC5197063 DOI: 10.7150/thno.16154] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/18/2016] [Indexed: 12/20/2022] Open
Abstract
Undesired immune responses have drastically hampered outcomes after allogeneic organ transplantation and cell therapy, and also lead to inflammatory diseases and autoimmunity. Umbilical cord mesenchymal stem cells (UCMSCs) have powerful regenerative and immunomodulatory potential, and their secreted extracellular vesicles (EVs) are envisaged as a promising natural source of nanoparticles to increase outcomes in organ transplantation and control inflammatory diseases. However, poor EV preparations containing highly-abundant soluble proteins may mask genuine vesicular-associated functions and provide misleading data. Here, we used Size-Exclusion Chromatography (SEC) to successfully isolate EVs from UCMSCs-conditioned medium. These vesicles were defined as positive for CD9, CD63, CD73 and CD90, and their size and morphology characterized by NTA and cryo-EM. Their immunomodulatory potential was determined in polyclonal T cell proliferation assays, analysis of cytokine profiles and in the skewing of monocyte polarization. In sharp contrast to the non-EV containing fractions, to the complete conditioned medium and to ultracentrifuged pellet, SEC-purified EVs from UCMSCs inhibited T cell proliferation, resembling the effect of parental UCMSCs. Moreover, while SEC-EVs did not induce cytokine response, the non-EV fractions, conditioned medium and ultracentrifuged pellet promoted the secretion of pro-inflammatory cytokines by polyclonally stimulated T cells and supported Th17 polarization. In contrast, EVs did not induce monocyte polarization, but the non-EV fraction induced CD163 and CD206 expression and TNF-α production in monocytes. These findings increase the growing evidence confirming that EVs are an active component of MSC's paracrine immunosuppressive function and affirm their potential for therapeutics in nanomedicine. In addition, our results highlight the importance of well-purified and defined preparations of MSC-derived EVs to achieve the immunosuppressive effect.
Collapse
|
24
|
Shadrin IY, Khodabukus A, Bursac N. Striated muscle function, regeneration, and repair. Cell Mol Life Sci 2016; 73:4175-4202. [PMID: 27271751 PMCID: PMC5056123 DOI: 10.1007/s00018-016-2285-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022]
Abstract
As the only striated muscle tissues in the body, skeletal and cardiac muscle share numerous structural and functional characteristics, while exhibiting vastly different size and regenerative potential. Healthy skeletal muscle harbors a robust regenerative response that becomes inadequate after large muscle loss or in degenerative pathologies and aging. In contrast, the mammalian heart loses its regenerative capacity shortly after birth, leaving it susceptible to permanent damage by acute injury or chronic disease. In this review, we compare and contrast the physiology and regenerative potential of native skeletal and cardiac muscles, mechanisms underlying striated muscle dysfunction, and bioengineering strategies to treat muscle disorders. We focus on different sources for cellular therapy, biomaterials to augment the endogenous regenerative response, and progress in engineering and application of mature striated muscle tissues in vitro and in vivo. Finally, we discuss the challenges and perspectives in translating muscle bioengineering strategies to clinical practice.
Collapse
Affiliation(s)
- I Y Shadrin
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - A Khodabukus
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - N Bursac
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA.
| |
Collapse
|
25
|
Llucià‐Valldeperas A, Soler‐Botija C, Gálvez‐Montón C, Roura S, Prat‐Vidal C, Perea‐Gil I, Sanchez B, Bragos R, Vunjak‐Novakovic G, Bayes‐Genis A. Electromechanical Conditioning of Adult Progenitor Cells Improves Recovery of Cardiac Function After Myocardial Infarction. Stem Cells Transl Med 2016; 6:970-981. [PMID: 28297585 PMCID: PMC5442794 DOI: 10.5966/sctm.2016-0079] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/29/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiac cells are subjected to mechanical and electrical forces, which regulate gene expression and cellular function. Therefore, in vitro electromechanical stimuli could benefit further integration of therapeutic cells into the myocardium. Our goals were (a) to study the viability of a tissue-engineered construct with cardiac adipose tissue-derived progenitor cells (cardiac ATDPCs) and (b) to examine the effect of electromechanically stimulated cardiac ATDPCs within a myocardial infarction (MI) model in mice for the first time. Cardiac ATDPCs were electromechanically stimulated at 2-millisecond pulses of 50 mV/cm at 1 Hz and 10% stretching during 7 days. The cells were harvested, labeled, embedded in a fibrin hydrogel, and implanted over the infarcted area of the murine heart. A total of 39 animals were randomly distributed and sacrificed at 21 days: groups of grafts without cells and with stimulated or nonstimulated cells. Echocardiography and gene and protein analyses were also carried out. Physiologically stimulated ATDPCs showed increased expression of cardiac transcription factors, structural genes, and calcium handling genes. At 21 days after implantation, cardiac function (measured as left ventricle ejection fraction between presacrifice and post-MI) increased up to 12% in stimulated grafts relative to nontreated animals. Vascularization and integration with the host blood supply of grafts with stimulated cells resulted in increased vessel density in the infarct border region. Trained cells within the implanted fibrin patch expressed main cardiac markers and migrated into the underlying ischemic myocardium. To conclude, synchronous electromechanical cell conditioning before delivery may be a preferred alternative when considering strategies for heart repair after myocardial infarction. Stem Cells Translational Medicine 2017;6:970-981.
Collapse
Affiliation(s)
- Aida Llucià‐Valldeperas
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Carolina Soler‐Botija
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Carolina Gálvez‐Montón
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Santiago Roura
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
- Center of Regenerative Medicine in Barcelona, Barcelona, Spain
| | - Cristina Prat‐Vidal
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Isaac Perea‐Gil
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Benjamin Sanchez
- Electronic and Biomedical Instrumentation Group, Departament d’Enginyeria Electrònica, Universitat Politècnica de Catalunya, Barcelona, Spain
- Department of Neurology, Division of Neuromuscular Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ramon Bragos
- Electronic and Biomedical Instrumentation Group, Departament d’Enginyeria Electrònica, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Gordana Vunjak‐Novakovic
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- Department of Medicine, Columbia University, New York, New York, USA
| | - Antoni Bayes‐Genis
- Heart Failure and Cardiac Regeneration Research Programme, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
- Cardiology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
26
|
Stoppel WL, Gao AE, Greaney AM, Partlow BP, Bretherton RC, Kaplan DL, Black LD. Elastic, silk-cardiac extracellular matrix hydrogels exhibit time-dependent stiffening that modulates cardiac fibroblast response. J Biomed Mater Res A 2016; 104:3058-3072. [PMID: 27480328 DOI: 10.1002/jbm.a.35850] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/15/2022]
Abstract
Heart failure is the leading cause of death in the United States and rapidly becoming the leading cause of death worldwide. While pharmacological treatments can reduce progression to heart failure following myocardial infarction, there still exists a need for new therapies that promote better healing postinjury for a more functional cardiac repair and methods to understand how the changes to tissue mechanical properties influence cell phenotype and function following injury. To address this need, we have optimized a silk-based hydrogel platform containing cardiac tissue-derived extracellular matrix (cECM). These silk-cECM hydrogels have tunable mechanical properties, as well as rate-controllable hydrogel stiffening over time. In vitro, silk-cECM scaffolds led to enhanced cardiac fibroblast (CF) cell growth and viability with culture time. cECM incorporation improved expression of integrin an focal adhesion proteins, suggesting that CFs were able to interact with the cECM in the hydrogel. Subcutaneous injection of silk hydrogels in rats demonstrated that addition of the cECM led to endogenous cell infiltration and promoted endothelial cell ingrowth after 4 weeks in vivo. This naturally derived silk fibroin platform is applicable to the development of more physiologically relevant constructs that replicate healthy and diseased tissue in vitro and has the potential to be used as an injectable therapeutic for cardiac repair. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 3058-3072, 2016.
Collapse
Affiliation(s)
- Whitney L Stoppel
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155
| | - Albert E Gao
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155
| | - Allison M Greaney
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155
| | - Benjamin P Partlow
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155
| | - Ross C Bretherton
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, 02155. .,Cellular, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, 02111.
| |
Collapse
|
27
|
Roura S, Pujal JM, Gálvez-Montón C, Bayes-Genis A. Quality and exploitation of umbilical cord blood for cell therapy: Are we beyond our capabilities? Dev Dyn 2016; 245:710-7. [PMID: 27043849 DOI: 10.1002/dvdy.24408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/03/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023] Open
Abstract
There is increasing interest in identifying novel stem cell sources for application in emerging cell therapies. In this context, umbilical cord blood (UCB) shows great promise in multiple clinical settings. The number of UCB banks has therefore increased worldwide, with the objective of preserving potentially life-saving cells that are usually discarded after birth. After a rather long and costly processing procedure, the resultant UCB-derived cell products are cryopreserved until transplantation to patients. However, in many cases, only a small proportion of administered cells engraft successfully. Thus, can we do any better regarding current UCB-based therapeutic approaches? Here we discuss concerns about the use of UCB that are not critically pondered by researchers, clinicians, and banking services, including wasting samples with small volumes and the need for more reliable quality and functional controls to ensure the biological activity of stem cells and subsequent engraftment and treatment efficacy. Finally, we appeal for collaborative agreements between research institutions and UCB banks in order to redirect currently discarded small-volume UCB units for basic and clinical research purposes. Developmental Dynamics 245:710-717, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Santiago Roura
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
- Center of Regenerative Medicine in Barcelona, c/ Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Josep Maria Pujal
- Cell Processing Laboratory, Edifici Giroemprèn, Pic de Peguera 11, Parc Científic i Tecnològic Universitat de Girona, 17003, Girona, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
| | - Antoni Bayes-Genis
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
- Cardiology Service, Germans Trias i Pujol University Hospital, Crta.Can Ruti-Camí Escoles s/n, 08916, Badalona, Spain
- Department of Medicine, Crta. Can Ruti-Camí Escoles s/n, Universitat Autònoma de Barcelona, 08916, Badalona, Spain
| |
Collapse
|
28
|
Bayes-Genis A, Gastelurrutia P, Cámara ML, Teis A, Lupón J, Llibre C, Zamora E, Alomar X, Ruyra X, Roura S, Revilla A, San Román JA, Gálvez-Montón C. First-in-man Safety and Efficacy of the Adipose Graft Transposition Procedure (AGTP) in Patients With a Myocardial Scar. EBioMedicine 2016; 7:248-54. [PMID: 27322478 PMCID: PMC4909363 DOI: 10.1016/j.ebiom.2016.03.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The present study evaluates the safety and efficacy of the Adipose Graft Transposition Procedure (AGTP) as a biological regenerative innovation for patients with a chronic myocardial scar. METHODS This prospective, randomized single-center controlled study included 10 patients with established chronic transmural myocardial scars. Candidates for myocardial revascularization were randomly allocated into two treatment groups. In the control arm (n=5), the revascularizable area was treated with CABG and the non-revascularizable area was left untouched. Patients in the AGTP-treated arm (n=5) were treated with CABG and the non-revascularizable area was covered by a biological adipose graft. The primary endpoint was the appearance of adverse effects derived from the procedure including hospital admissions and death, and 24-hour Holter monitoring arrhythmias at baseline, 1week, and 3 and 12months. Secondary endpoints of efficacy were assessed by cardiac MRI. FINDINGS No differences in safety were observed between groups in terms of clinical or arrhythmic events. On follow-up MRI testing, participants in the AGTP-treated arm showed a borderline smaller left ventricular end systolic volume (LVESV; p=0.09) and necrosis ratio (p=0.06) at 3months but not at 12months. The AGTP-treated patient with the largest necrotic area and most dilated chambers experienced a noted improvement in necrotic mass size (-10.8%), and ventricular volumes (LVEDV: -55.2mL and LVESV: -37.8mL at one year follow-up) after inferior AGTP. INTERPRETATION Our results indicate that AGTP is safe and may be efficacious in selected patients. Further studies are needed to assess its clinical value. (ClinicalTrials.org NCT01473433, AdiFlap Trial).
Collapse
Affiliation(s)
- Antoni Bayes-Genis
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain.
| | - Paloma Gastelurrutia
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| | - Maria-Luisa Cámara
- Cardiac Surgery Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Albert Teis
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain; Clínica Creu Blanca, Barcelona, Spain
| | - Josep Lupón
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cinta Llibre
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Elisabet Zamora
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | | | - Xavier Ruyra
- Cardiac Surgery Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain; Center of Regenerative Medicine in Barcelona, Barcelona, Spain
| | - Ana Revilla
- ICICORELAB, Clinic University Hospital, Valladolid, Spain
| | | | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
29
|
Roura S, Gálvez-Montón C, Bayes-Genis A. Fibrin, the preferred scaffold for cell transplantation after myocardial infarction? An old molecule with a new life. J Tissue Eng Regen Med 2016; 11:2304-2313. [PMID: 27061269 DOI: 10.1002/term.2129] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/13/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022]
Abstract
Fibrin is a topical haemostat, sealant and tissue glue, which consists of concentrated fibrinogen and thrombin. It has broad medical and research uses. Recently, several studies have shown that engineered patches comprising mixtures of biological or synthetic materials and progenitor cells showed therapeutic promise for regenerating damaged tissues. In that context, fibrin maintains cell adherence at the site of injury, where cells are required for tissue repair, and offers a nurturing environment that protects implanted cells without interfering with their expected benefit. Here we review the past, present and future uses of fibrin, with a focus on its use as a scaffold material for cardiac repair. Fibrin patches filled with regenerative cells can be placed over the scarring myocardium; this methodology avoids many of the drawbacks of conventional cell-infusion systems. Advantages of using fibrin also include extraction from the patient's blood, an easy readjustment and implantation procedure, increase in viability and early proliferation of delivered cells, and benefits even with the patch alone. In line with this, we discuss the numerous preclinical studies that have used fibrin-cell patches, the practical issues inherent in their generation, and the necessary process of scaling-up from animal models to patients. In the light of the data presented, fibrin stands out as a valuable biomaterial for delivering cells to damaged tissue and for promoting beneficial effects. However, before the fibrin scaffold can be translated from bench to bedside, many issues must be explored further, including suboptimal survival and limited migration of the implanted cells to underlying ischaemic myocardium. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Santiago Roura
- Heart Failure and Cardiac Regeneration (ICREC) Research Programme, Germans Trias i Pujol Health Science Research Institute, Badalona, Barcelona, Spain.,Center of Regenerative Medicine in Barcelona, Barcelona, Spain
| | - Carolina Gálvez-Montón
- Heart Failure and Cardiac Regeneration (ICREC) Research Programme, Germans Trias i Pujol Health Science Research Institute, Badalona, Barcelona, Spain
| | - Antoni Bayes-Genis
- Heart Failure and Cardiac Regeneration (ICREC) Research Programme, Germans Trias i Pujol Health Science Research Institute, Badalona, Barcelona, Spain.,Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Spain
| |
Collapse
|
30
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
31
|
Perea-Gil I, Prat-Vidal C, Bayes-Genis A. In vivo experience with natural scaffolds for myocardial infarction: the times they are a-changin'. Stem Cell Res Ther 2015; 6:248. [PMID: 26670389 PMCID: PMC4681026 DOI: 10.1186/s13287-015-0237-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Treating a myocardial infarction (MI), the most frequent cause of death worldwide, remains one of the most exciting medical challenges in the 21st century. Cardiac tissue engineering, a novel emerging treatment, involves the use of therapeutic cells supported by a scaffold for regenerating the infarcted area. It is essential to select the appropriate scaffold material; the ideal one should provide a suitable cellular microenvironment, mimic the native myocardium, and allow mechanical and electrical coupling with host tissues. Among available scaffold materials, natural scaffolds are preferable for achieving these purposes because they possess myocardial extracellular matrix properties and structures. Here, we review several natural scaffolds for applications in MI management, with a focus on pre-clinical studies and clinical trials performed to date. We also evaluate scaffolds combined with different cell types and proteins for their ability to promote improved heart function, contractility and neovascularization, and attenuate adverse ventricular remodeling. Although further refinement is necessary in the coming years, promising results indicate that natural scaffolds may be a valuable translational therapeutic option with clinical impact in MI repair.
Collapse
Affiliation(s)
- Isaac Perea-Gil
- ICREC (Heart Failure and Cardiac Regeneration) Research Lab, Health Sciences Research Institute Germans Trias i Pujol (IGTP). Cardiology Service, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Barcelona, Spain
| | - Cristina Prat-Vidal
- ICREC (Heart Failure and Cardiac Regeneration) Research Lab, Health Sciences Research Institute Germans Trias i Pujol (IGTP). Cardiology Service, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Barcelona, Spain.
| | - Antoni Bayes-Genis
- ICREC (Heart Failure and Cardiac Regeneration) Research Lab, Health Sciences Research Institute Germans Trias i Pujol (IGTP). Cardiology Service, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Barcelona, Spain.,Department of Medicine, Autonomous University of Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
32
|
Gong X, Wang P, Wu Q, Wang S, Yu L, Wang G. Human umbilical cord blood derived mesenchymal stem cells improve cardiac function in cTnT(R141W) transgenic mouse of dilated cardiomyopathy. Eur J Cell Biol 2015; 95:57-67. [PMID: 26655348 DOI: 10.1016/j.ejcb.2015.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/15/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Cell transplantation is a promising strategy in regenerative medicine. Beneficial effects of bone marrow mesenchymal stem cells (BM-MSCs) on heart disease have been widely reported. However, the MSCs in these studies have been mainly derived from autologous animals, and data on MSCs from human umbilical cord blood (UCB-MSCs) are still scarce. We investigated whether intramyocardial xenogeneic administration of UCB-MSCs is beneficial for preserving heart function in a cTnT(R141W) transgenic mouse of dilated cardiomyopathy (DCM). Cultured UCB-MSCs, which were identified by there morphology, differentiation and cell surface markers, were transplanted into cTnT(R141W) transgenic mice to examine apoptosis, fibrosis, vasculogenesis and the associated Akt pathway. Moreover, we measured the expression levels of VEGF and IGF-1, which are growth factors required for differentiation into cardiomyocytes, and are also involved in cardiac regeneration and improving heart function. One month after transplantation, MSCs significantly decreased chamber dilation and contractile dysfunction in the cTnT(R141W) mice. MSCs transplanted hearts showed a significant decrease in cardiac apoptosis and its regulation by the Akt pathway. Cardiac fibrosis and cytoplasmic vacuolisation were significantly attenuated in the MSCs group. Importantly, the levels of VEGF and IGF-1 were increased in the MSCs transplanted hearts. In vitro, the MSC-conditioned medium displayed anti-apoptotic activity in h9c2 cardiomyocytes subjected to hypoxia. These results further confirm the paracrine effects of MSCs. In conclusion, UCB-MSCs preserve cardiac function after intramyocardial transplantation in a DCM mouse, and this effect may be associated with reductions in cellular apoptosis, inflammation, hypertrophy and myocardial fibrosis; in addition to; up-regulation of Akt, VEGF and IGF-1; and enhanced angiogenesis.
Collapse
Affiliation(s)
- Xuhe Gong
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Pengbo Wang
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qingqing Wu
- Departments of Obstetrics and Gynaecology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Sijia Wang
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Litian Yu
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Guogan Wang
- Emergency and Critical Center, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Departments of Obstetrics and Gynaecology, Fuxing Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|