1
|
Alonaizan R, Purnama U, Malandraki-Miller S, Gunadasa-Rohling M, Lewis A, Smart N, Carr C. MicroRNA-210 Enhances Cell Survival and Paracrine Potential for Cardiac Cell Therapy While Targeting Mitophagy. J Funct Biomater 2025; 16:147. [PMID: 40278255 PMCID: PMC12028018 DOI: 10.3390/jfb16040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The therapeutic potential of presumed cardiac progenitor cells (CPCs) in heart regeneration has garnered significant interest, yet clinical trials have revealed limited efficacy due to challenges in cell survival, retention, and expansion. Priming CPCs to survive the hostile hypoxic environment may be key to enhancing their regenerative capacity. We demonstrate that microRNA-210 (miR-210), known for its role in hypoxic adaptation, significantly improves CPC survival by inhibiting apoptosis through the downregulation of Casp8ap2, a ~40% reduction in caspase activity, and a ~90% decrease in DNA fragmentation. Contrary to the expected induction of Bnip3-dependent mitophagy by hypoxia, miR-210 did not upregulate Bnip3, indicating a distinct anti-apoptotic mechanism. Instead, miR-210 reduced markers of mitophagy and increased mitochondrial biogenesis and oxidative metabolism, suggesting a role in metabolic reprogramming. Furthermore, miR-210 enhanced the secretion of paracrine growth factors from CPCs, with a ~1.6-fold increase in the release of stem cell factor and of insulin growth factor 1, which promoted in vitro endothelial cell proliferation and cardiomyocyte survival. These findings elucidate the multifaceted role of miR-210 in CPC biology and its potential to enhance cell-based therapies for myocardial repair by promoting cell survival, metabolic adaptation, and paracrine signalling.
Collapse
Affiliation(s)
- Rita Alonaizan
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
- King Faisal Specialist Hospital & Research Centre, Riyadh 12713, Saudi Arabia
| | - Ujang Purnama
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | | | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Andrew Lewis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Carolyn Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| |
Collapse
|
2
|
Amin A, Mohajerian A, Ghalehnoo SR, Mohamadinia M, Ahadi S, Sohbatzadeh T, Pazoki M, Hasanvand A, Faghihkhorasani F, Habibi Z. Potential Player of Platelet in the Pathogenesis of Cardiotoxicity: Molecular Insight and Future Perspective. Cardiovasc Toxicol 2024; 24:1381-1394. [PMID: 39397196 DOI: 10.1007/s12012-024-09924-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Cancer patients may encounter the onset of cardiovascular disease due to tumor advancement or chemotherapy, commonly known as "cardiotoxicity." In this respect, the conventional chemotherapy treatment protocol involves a mixture of different medications. These medications can be detrimental to cardiac tissue, consequently exposing the patient to the possibility of irreversible cardiac injury. The enhancement of oxidative stress and inflammation is an important mechanism of chemotherapeutic agents for developing cardiotoxicity. Regarding their dual pro- and anti-inflammatory functions, platelets can significantly influence the progression or suppression of cardiotoxicity. Therefore, the expression of platelet activatory markers can serve as valuable prognostic indicators for cardiotoxicity. The primary objective of this study is to examine the significance of platelets in cardiotoxicity and explore potential strategies that could effectively target malignant cells while minimizing their cytotoxic impact, such as cardiotoxicity and thrombosis.
Collapse
Affiliation(s)
- Arash Amin
- Department of Cardiology, School of Medicine, Shahid Madani Hospital, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Ahmad Mohajerian
- Department of Emergency Medicine, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Rashki Ghalehnoo
- Department of Cardiology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Mehdi Mohamadinia
- Department of Dental Prosthesis, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shana Ahadi
- School of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tooba Sohbatzadeh
- Student Research Committee, School of Medicine, Alborz University of Medical Science, Alborz, Iran
| | - Mahboubeh Pazoki
- Department of Cardiology, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Hasanvand
- Department of General Surgery, Lorestan University of Medical Science, Khorramabad, Iran
| | | | - Zeinab Habibi
- Lorestan University of Medical Science, Lorestan, Iran.
| |
Collapse
|
3
|
Skliutė G, Staponkutė G, Skliutas E, Malinauskas M, Navakauskienė R. Molecular changes in endometrium origin stromal cells during initiation of cardiomyogenic differentiation induced with Decitabine, Angiotensin II and TGF- β1. Sci Rep 2024; 14:16966. [PMID: 39043870 PMCID: PMC11266582 DOI: 10.1038/s41598-024-68108-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024] Open
Abstract
Stem cells' differentiation toward cardiac lineage is a complex process dependent on various alterations in molecular basis and regulation pathways. The aim of the study is to show that endometrium-derived stromal cells - menstrual, endometrial and endometriotic, could be an attractive source for examination of the mechanisms underlying cardiomyogenesis. After treatment with Decitabine, Angiotensin II and TGF-β1, cells demonstrated morphological dedifferentiation into early cardiomyocyte-like cells and expressed CD36, CD106, CD172a typically used to sort for human pluripotent stem cell-derived cardiomyocytes. RT-qPCR revealed changed cells' genetic profiles, as majority of cardiac lineage differentiation related genes and cardiac ion channels (calcium, sodium, potassium) coding genes were upregulated after 6 and 13 days of exposure. Additionally, analysis of expression of various signaling proteins (FOXO1, PDGFB, TGFBR1, mTOR, VEGFA, WNT4, Notch1) coding genes showed differences between cell cultures as they seem to employ distinct signaling pathways through differentiation initiation. Early stages of differentiation had biggest impact on cardiomyogenesis related proteins (Nkx-2.5, EZH2, FOXO3a, H3K9Ac) levels, as we noticed after conducting Western blot and as expected, early cardiac transcription factor Nkx-2.5 was highly expressed and localized in nucleus of differentiating cells. These findings led us to assess endometrium origin stromal cells' potential to differentiate towards cardiomyogenic lineage and better understand the regulation of complex differentiation processes in ex vivo model systems.
Collapse
Affiliation(s)
- Giedrė Skliutė
- Department of Molecular Cell Biology, Life Sciences Center, Institute of Biochemistry, Vilnius University, Saulėtekio Av. 7, 10257, Vilnius, Lithuania.
| | - Giedrė Staponkutė
- Department of Molecular Cell Biology, Life Sciences Center, Institute of Biochemistry, Vilnius University, Saulėtekio Av. 7, 10257, Vilnius, Lithuania
| | - Edvinas Skliutas
- Faculty of Physics, Laser Research Center, Vilnius University, Saulėtekio Av. 10, 10223, Vilnius, Lithuania
| | - Mangirdas Malinauskas
- Faculty of Physics, Laser Research Center, Vilnius University, Saulėtekio Av. 10, 10223, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Life Sciences Center, Institute of Biochemistry, Vilnius University, Saulėtekio Av. 7, 10257, Vilnius, Lithuania.
| |
Collapse
|
4
|
Frangogiannis NG. TGF-β as a therapeutic target in the infarcted and failing heart: cellular mechanisms, challenges, and opportunities. Expert Opin Ther Targets 2024; 28:45-56. [PMID: 38329809 DOI: 10.1080/14728222.2024.2316735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Myocardial fibrosis accompanies most cardiac conditions and can be reparative or maladaptive. Transforming Growth Factor (TGF)-β is a potent fibrogenic mediator, involved in repair, remodeling, and fibrosis of the injured heart. AREAS COVERED This review manuscript discusses the role of TGF-β in heart failure focusing on cellular mechanisms and therapeutic implications. TGF-β is activated in infarcted, remodeling and failing hearts. In addition to its fibrogenic actions, TGF-β has a broad range of effects on cardiomyocytes, immune, and vascular cells that may have both protective and detrimental consequences. TGF-β-mediated effects on macrophages promote anti-inflammatory transition, whereas actions on fibroblasts mediate reparative scar formation and effects on pericytes are involved in maturation of infarct neovessels. On the other hand, TGF-β actions on cardiomyocytes promote adverse remodeling, and prolonged activation of TGF-β signaling in fibroblasts stimulates progression of fibrosis and heart failure. EXPERT OPINION Understanding of the cell-specific actions of TGF-β is necessary to design therapeutic strategies in patients with myocardial disease. Moreover, to implement therapeutic interventions in the heterogeneous population of heart failure patients, mechanism-driven classification of both HFrEF and HFpEF patients is needed. Heart failure patients with prolonged or overactive fibrogenic TGF-β responses may benefit from cautious TGF-β inhibition.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
5
|
Bachamanda Somesh D, Klose K, Maring JA, Kunkel D, Jürchott K, Protze SI, Klein O, Nebrich G, Becker M, Krüger U, Nazari-Shafti TZ, Falk V, Kurtz A, Gossen M, Stamm C. Cardiomyocyte precursors generated by direct reprogramming and molecular beacon selection attenuate ventricular remodeling after experimental myocardial infarction. Stem Cell Res Ther 2023; 14:296. [PMID: 37840130 PMCID: PMC10577947 DOI: 10.1186/s13287-023-03519-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 09/25/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Direct cardiac reprogramming is currently being investigated for the generation of cells with a true cardiomyocyte (CM) phenotype. Based on the original approach of cardiac transcription factor-induced reprogramming of fibroblasts into CM-like cells, various modifications of that strategy have been developed. However, they uniformly suffer from poor reprogramming efficacy and a lack of translational tools for target cell expansion and purification. Therefore, our group has developed a unique approach to generate proliferative cells with a pre-CM phenotype that can be expanded in vitro to yield substantial cell doses. METHODS Cardiac fibroblasts were reprogrammed toward CM fate using lentiviral transduction of cardiac transcriptions factors (GATA4, MEF2C, TBX5, and MYOCD). The resulting cellular phenotype was analyzed by RNA sequencing and immunocytology. Live target cells were purified based on intracellular CM marker expression using molecular beacon technology and fluorescence-activated cell sorting. CM commitment was assessed using 5-azacytidine-based differentiation assays and the therapeutic effect was evaluated in a mouse model of acute myocardial infarction using echocardiography and histology. The cellular secretome was analyzed using mass spectrometry. RESULTS We found that proliferative CM precursor-like cells were part of the phenotype spectrum arising during direct reprogramming of fibroblasts toward CMs. These induced CM precursors (iCMPs) expressed CPC- and CM-specific proteins and were selectable via hairpin-shaped oligonucleotide hybridization probes targeting Myh6/7-mRNA-expressing cells. After purification, iCMPs were capable of extensive expansion, with preserved phenotype when under ascorbic acid supplementation, and gave rise to CM-like cells with organized sarcomeres in differentiation assays. When transplanted into infarcted mouse hearts, iCMPs prevented CM loss, attenuated fibrotic scarring, and preserved ventricular function, which can in part be attributed to their substantial secretion of factors with documented beneficial effect on cardiac repair. CONCLUSIONS Fibroblast reprogramming combined with molecular beacon-based cell selection yields an iCMP-like cell population with cardioprotective potential. Further studies are needed to elucidate mechanism-of-action and translational potential.
Collapse
Affiliation(s)
- Dipthi Bachamanda Somesh
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| | - Kristin Klose
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| | - Janita A Maring
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, 13353, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Désirée Kunkel
- Cytometry Core Facility, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Karsten Jürchott
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute for Medical Immunology, 13353, Berlin, Germany
| | - Stephanie I Protze
- University Health Network, McEwen Stem Cell Institute, Toronto, ON, M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Oliver Klein
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Imaging Mass Spectrometry Core Unit, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Grit Nebrich
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Imaging Mass Spectrometry Core Unit, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Matthias Becker
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Ulrike Krüger
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute for Medical Immunology, 13353, Berlin, Germany
| | - Timo Z Nazari-Shafti
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, 10785, Berlin, Germany
| | - Volkmar Falk
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, 10785, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | - Andreas Kurtz
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, 13353, Berlin, Germany
| | - Christof Stamm
- BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies, 13353, Berlin, Germany.
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- German Centre for Cardiovascular Research, Partner Site Berlin, 10785, Berlin, Germany.
| |
Collapse
|
6
|
Sun B, Wang L, Guo W, Chen S, Ma Y, Wang D. New treatment methods for myocardial infarction. Front Cardiovasc Med 2023; 10:1251669. [PMID: 37840964 PMCID: PMC10569499 DOI: 10.3389/fcvm.2023.1251669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/31/2023] [Indexed: 10/17/2023] Open
Abstract
For a long time, cardiovascular clinicians have focused their research on coronary atherosclerotic cardiovascular disease and acute myocardial infarction due to their high morbidity, high mortality, high disability rate, and limited treatment options. Despite the continuous optimization of the therapeutic methods and pharmacological therapies for myocardial ischemia-reperfusion, the incidence rate of heart failure continues to increase year by year. This situation is speculated to be caused by the current therapies, such as reperfusion therapy after ischemic injury, drugs, rehabilitation, and other traditional treatments, that do not directly target the infarcted myocardium. Consequently, these therapies cannot fundamentally solve the problems of myocardial pathological remodeling and the reduction of cardiac function after myocardial infarction, allowing for the progression of heart failure after myocardial infarction. Coupled with the decline in mortality caused by acute myocardial infarction in recent years, this combination leads to an increase in the incidence of heart failure. As a new promising therapy rising at the beginning of the twenty-first century, cardiac regenerative medicine provides a new choice and hope for the recovery of cardiac function and the prevention and treatment of heart failure after myocardial infarction. In the past two decades, regeneration engineering researchers have explored and summarized the elements, such as cells, scaffolds, and cytokines, required for myocardial regeneration from all aspects and various levels day and night, paving the way for our later scholars to carry out relevant research and also putting forward the current problems and directions for us. Here, we describe the advantages and challenges of cardiac tissue engineering, a contemporary innovative therapy after myocardial infarction, to provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Bingbing Sun
- Department of Critical Care Medicine, The Air Force Characteristic Medical Center, Air Force Medical University, Beijing, China
| | - Long Wang
- Department of General Internal Medicine, Beijing Dawanglu Emergency Hospital, Beijing, China
| | - Wenmin Guo
- Department of Critical Care Medicine, The Air Force Characteristic Medical Center, Air Force Medical University, Beijing, China
| | - Shixuan Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yujie Ma
- Department of Critical Care Medicine, The Air Force Characteristic Medical Center, Air Force Medical University, Beijing, China
| | - Dongwei Wang
- Department of Cardiac Rehabilitation, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Okamura A, Yoshioka Y, Saito Y, Ochiya T. Can Extracellular Vesicles as Drug Delivery Systems Be a Game Changer in Cardiac Disease? Pharm Res 2022; 40:889-908. [PMID: 36577860 PMCID: PMC10126064 DOI: 10.1007/s11095-022-03463-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022]
Abstract
Cardiac diseases such as myocardial infarction and heart failure have been the leading cause of death worldwide for more than 20 years, and new treatments continue to be investigated. Heart transplantation, a curative treatment for severe cardiac dysfunction, is available to only a small number of patients due to the rarity of donors and high costs. Cardiac regenerative medicine using embryonic stem cells and induced pluripotent stem cells is expected to be a new alternative to heart transplantation, but it has problems such as induction of immune response, tumor formation, and low survival rate of transplanted cells. On the other hand, there has been a focus on cell-free therapy using extracellular vesicles (EVs) due to their high biocompatibility and target specificity. Exosomes, one type of EV, play a role in the molecular transport system in vivo and can be considered a drug delivery system (DDS) innate to all living things. Exosomes contain nucleic acids and proteins, which are transported from secretory cells to recipient cells. Molecules in exosomes are encapsulated in a lipid bilayer, which allows them to exist stably in body fluids without being affected by nuclease degradation enzymes. Therefore, the therapeutic use of exosomes as DDSs has been widely explored and is being used in clinical trials and other clinical settings. This review summarizes the current topics of EVs as DDSs in cardiac disease.
Collapse
Affiliation(s)
- Akihiko Okamura
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.,Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
8
|
The Vascular Niche for Adult Cardiac Progenitor Cells. Antioxidants (Basel) 2022; 11:antiox11050882. [PMID: 35624750 PMCID: PMC9137669 DOI: 10.3390/antiox11050882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
Research on cardiac progenitor cell populations has generated expectations about their potential for cardiac regeneration capacity after acute myocardial infarction and during physiological aging; however, the endogenous capacity of the adult mammalian heart is limited. The modest efficacy of exogenous cell-based treatments can guide the development of new approaches that, alone or in combination, can be applied to boost clinical efficacy. The identification and manipulation of the adult stem cell environment, termed niche, will be critical for providing new evidence on adult stem cell populations and improving stem-cell-based therapies. Here, we review and discuss the state of our understanding of the interaction of adult cardiac progenitor cells with other cardiac cell populations, with a focus on the description of the B-CPC progenitor population (Bmi1+ cardiac progenitor cell), which is a strong candidate progenitor for all main cardiac cell lineages, both in the steady state and after cardiac damage. The set of all interactions should be able to define the vascular cardiac stem cell niche, which is associated with low oxidative stress domains in vasculature, and whose manipulation would offer new hope in the cardiac regeneration field.
Collapse
|
9
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
10
|
MicroRNAs and exosomes: Cardiac stem cells in heart diseases. Pathol Res Pract 2021; 229:153701. [PMID: 34872024 DOI: 10.1016/j.prp.2021.153701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/09/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022]
Abstract
Treating cardiovascular diseases with cardiac stem cells (CSCs) is a valid treatment among various stem cell-based therapies. With supplying the physiological need for cardiovascular cells as their main function, under pathological circumstances, CSCs can also reproduce the myocardial cells. Although studies have identified many of CSCs' functions, our knowledge of molecular pathways that regulate these functions is not complete enough. Either physiological or pathological studies have shown, stem cells proliferation and differentiation could be regulated by microRNAs (miRNAs). How miRNAs regulate CSC behavior is an interesting area of research that can help us study and control the function of these cells in vitro; an achievement that may be beneficial for patients with cardiovascular diseases. The secretome of stem and progenitor cells has been studied and it has been determined that exosomes are the main source of their secretion which are very small vesicles at the nanoscale and originate from endosomes, which are secreted into the extracellular space and act as key signaling organelles in intercellular communication. Mesenchymal stem cells, cardiac-derived progenitor cells, embryonic stem cells, induced pluripotent stem cells (iPSCs), and iPSC-derived cardiomyocytes release exosomes that have been shown to have cardioprotective, immunomodulatory, and reparative effects. Herein, we summarize the regulation roles of miRNAs and exosomes in cardiac stem cells.
Collapse
|
11
|
Kasai-Brunswick TH, Carvalho AB, Campos de Carvalho AC. Stem cell therapies in cardiac diseases: Current status and future possibilities. World J Stem Cells 2021; 13:1231-1247. [PMID: 34630860 PMCID: PMC8474720 DOI: 10.4252/wjsc.v13.i9.1231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases represent the world's leading cause of death. In this heterogeneous group of diseases, ischemic cardiomyopathies are the most devastating and prevalent, estimated to cause 17.9 million deaths per year. Despite all biomedical efforts, there are no effective treatments that can replace the myocytes lost during an ischemic event or progression of the disease to heart failure. In this context, cell therapy is an emerging therapeutic alternative to treat cardiovascular diseases by cell administration, aimed at cardiac regeneration and repair. In this review, we will cover more than 30 years of cell therapy in cardiology, presenting the main milestones and drawbacks in the field and signaling future challenges and perspectives. The outcomes of cardiac cell therapies are discussed in three distinct aspects: The search for remuscularization by replacement of lost cells by exogenous adult cells, the endogenous stem cell era, which pursued the isolation of a progenitor with the ability to induce heart repair, and the utilization of pluripotent stem cells as a rich and reliable source of cardiomyocytes. Acellular therapies using cell derivatives, such as microvesicles and exosomes, are presented as a promising cell-free therapeutic alternative.
Collapse
Affiliation(s)
- Tais Hanae Kasai-Brunswick
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Adriana Bastos Carvalho
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Antonio Carlos Campos de Carvalho
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil.
| |
Collapse
|
12
|
Abstract
Cardiovascular diseases top the list of fatal illnesses worldwide. Cardiac tissues is known to be one of te least proliferative in the human body, with very limited regenraive capacity. Stem cell therapy has shown great potential for treatment of cardiovascular diseases in the experimental setting, but success in human trials has been limited. Applications of stem cell therapy for cardiovascular regeneration necessitate understamding of the complex and unique structure of the heart unit, and the embryologic development of the heart muscles and vessels. This chapter aims to provide an insight into cardiac progenitor cells and their potential applications in regenerative medicine. It also provides an overview of the embryological development of cardiac tissue, and the major findings on the development of cardiac stem cells, their characterization, and differentiation, and their regenerative potential. It concludes with clinical applications in treating cardiac disease using different approaches, and concludes with areas for future research.
Collapse
|
13
|
Pakzad KK, Tan JJ, Anderson S, Board M, Clarke K, Carr CA. Metabolic maturation of differentiating cardiosphere-derived cells. Stem Cell Res 2021; 54:102422. [PMID: 34118565 PMCID: PMC8271094 DOI: 10.1016/j.scr.2021.102422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Collagen IV promotes proliferation of cardiosphere-derived cells. Fibronectin supports differentiation of cardiosphere-derived cells. Oxidative metabolism increases as cardiac progenitors mature. Stimulating fatty acid oxidation promotes cardiac progenitor cell maturation. Cardiosphere-derived cells (CDCs) can be expanded in vitro and induced to differentiate along the cardiac lineage. To recapitulate the phenotype of an adult cardiomyocyte, differentiating progenitors need to upregulate mitochondrial glucose and fatty acid oxidation. Here we cultured and differentiated CDCs using protocols aimed to maintain stemness or to promote differentiation, including triggering fatty acid oxidation using an agonist of peroxisome proliferator-activated receptor alpha (PPARα). Metabolic changes were characterised in undifferentiated CDCs and during differentiation towards a cardiac phenotype. CDCs from rat atria were expanded on fibronectin or collagen IV via cardiosphere formation. Differentiation was assessed using flow cytometry and qPCR and substrate metabolism was quantified using radiolabelled substrates. Collagen IV promoted proliferation of CDCs whereas fibronectin primed cells for differentiation towards a cardiac phenotype. In both populations, treatment with 5-Azacytidine induced a switch towards oxidative metabolism, as shown by changes in gene expression, decreased glycolytic flux and increased oxidation of glucose and palmitate. Addition of a PPARα agonist during differentiation increased both glucose and fatty acid oxidation and expression of cardiac genes. We conclude that oxidative metabolism and cell differentiation act in partnership with increases in one driving an increase in the other.
Collapse
Affiliation(s)
| | - Jun Jie Tan
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK; Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | | | - Mary Board
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK
| | - Kieran Clarke
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK.
| |
Collapse
|
14
|
Extracellular Vesicle-Based Therapeutics for Heart Repair. NANOMATERIALS 2021; 11:nano11030570. [PMID: 33668836 PMCID: PMC7996323 DOI: 10.3390/nano11030570] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) are constituted by a group of heterogeneous membrane vesicles secreted by most cell types that play a crucial role in cell–cell communication. In recent years, EVs have been postulated as a relevant novel therapeutic option for cardiovascular diseases, including myocardial infarction (MI), partially outperforming cell therapy. EVs may present several desirable features, such as no tumorigenicity, low immunogenic potential, high stability, and fine cardiac reparative efficacy. Furthermore, the natural origin of EVs makes them exceptional vehicles for drug delivery. EVs may overcome many of the limitations associated with current drug delivery systems (DDS), as they can travel long distances in body fluids, cross biological barriers, and deliver their cargo to recipient cells, among others. Here, we provide an overview of the most recent discoveries regarding the therapeutic potential of EVs for addressing cardiac damage after MI. In addition, we review the use of bioengineered EVs for targeted cardiac delivery and present some recent advances for exploiting EVs as DDS. Finally, we also discuss some of the most crucial aspects that should be addressed before a widespread translation to the clinical arena.
Collapse
|
15
|
Pawan KC, Mickey S, Rubia S, Yi H, Ge Z. Preseeding of Mesenchymal Stem Cells Increases Integration of an iPSC-Derived CM Sheet into a Cardiac Matrix. ACS Biomater Sci Eng 2020; 6:6808-6818. [PMID: 33320624 PMCID: PMC9841440 DOI: 10.1021/acsbiomaterials.0c00788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell sheet technology has demonstrated great promise in delivering a large amount of therapeutic cells for tissue repair, including in the myocardium. However, the lack of host integration remains one of the key challenges in using cell sheets for cardiac repair. Paracrine factors secreted by mesenchymal stem cells (MSCs) have been reported to facilitate tissue repair and regeneration in a variety of ways. It has been demonstrated that paracrine factors from MSCs could enhance scaffold recellularization and vascularization. In this study, we used an in vitro cardiac matrix mimic platform to examine the effects of hMSCs preseeding on the interactions between cell sheets and cardiac matrix. The fabricated human induced pluripotent stem cells-derived cardiomyocyte sheets were attached to a decellularized porcine myocardium slice with or without preseeding of hMSCs. The hMSCs preseeding significantly enhanced the interactions between cardiomyocyte sheets and cardiac matrix in terms of cell migration distance, cell distribution, and mature vascular and cardiomyocyte marker expressions in the matrix. Growth factor and matrix metalloproteinases array analysis suggested that hMSCs- induced vascularization and MMPs regulation are the two possible mechanisms that lead to the improved CMs and cardiac matrix interactions. Further examination of these two mechanisms will enable the development of new approaches to facilitate transplanted cells for tissue repair.
Collapse
Affiliation(s)
- KC Pawan
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Shah Mickey
- Department of Biomedical Engineering and Department of Integrated Bioscience, The University of Akron, Akron, Ohio 44325, United States
| | - Shaik Rubia
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Hong Yi
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Zhang Ge
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
16
|
Xing W, Li T, Wang Y, Qiang Y, Ai C, Tang H. MiR-33a-5p targets NOMO1 to modulate human cardiomyocyte progenitor cells proliferation and differentiation and apoptosis. J Recept Signal Transduct Res 2020; 41:476-487. [PMID: 33054489 DOI: 10.1080/10799893.2020.1825492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE MicroRNA (miRNA) is known to be involved in the pathological process of congenital heart disease (CHD), and nodal modulator1 (NOMO1) is a critical determinant of heart formation. The present study aims to discover the effect of miR-33a-5p and NOMO1 on CHD. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect expressions of miR-33a-5p mimic or inhibitor and overexpressed NOMO1 plasmid orNOMO1 knockdown. Human cardiomyocyte progenitor cells (hCMPCs) proliferation was measured by cell counting kit-8 (CCK-8) at 24, 48 and 72 h. Flow cytometry was applied to investigate hCMPCs cell cycle progression and apoptosis. Expressions of cell apoptotic proteins Bax, Cleaved(C) caspase-3 and Bcl-2, and expressions of cardiomyocyte differentiation markers GATA4, troponin T (cTnT) and myocyte enhancer factor2C (MEF2C) in hCMPCs were identified by qRT-PCR and western blot. Target genes and potential binding sites of NOMO1 and miR-33a-5p were predicted with Targetscan 7.2, and was confirmed through dual-luciferase reporter assay. RESULTS Up-regulation of miR-33a-5p inhibited hCMPCs proliferation, cell cycle G0/S transition but promoted hCMPCs apoptosis, which was partially mitigated by overexpressed NOMO1. NOMO1 was the target gene of miR-33a-5p. Expressions of Bax and C caspase-3 were enhanced but expressions of Bcl-2, GATA4, cTnT and MEF2C were reduced by up-regulation of miR-33a-5p, which was partially mitigated by overexpressed NOMO1. CONCLUSION Up-regulation of miR-33a-5p inhibited hCMPCs proliferation, cell cycle G0/S transition and differentiation into cardiomyocytes but promoted apoptosis via targeting NOMO1.
Collapse
Affiliation(s)
- Wang Xing
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Tiangang Li
- Department of Ultrasonography, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Yixuan Wang
- Department of Ultrasonography, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Yi Qiang
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Chencheng Ai
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Hanbo Tang
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| |
Collapse
|
17
|
Markmee R, Aungsuchawan S, Tancharoen W, Narakornsak S, Pothacharoen P. Differentiation of cardiomyocyte-like cells from human amniotic fluid mesenchymal stem cells by combined induction with human platelet lysate and 5-azacytidine. Heliyon 2020; 6:e04844. [PMID: 32995593 PMCID: PMC7502343 DOI: 10.1016/j.heliyon.2020.e04844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/01/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
Human amniotic fluid mesenchymal stem cells (hAF-MSCs) have been shown to be effective in the treatment of many diseases. Platelet lysate (PL) contains multiple growth and differentiation factors; therefore, it can be used as a differentiation inducer. In this study, we attempted to evaluate the efficiency of human platelet lysate (hPL) on cell viability and the effects on cardiomyogenic differentiation of hAF-MSCs. When treating the cells with hPL, the result showed an increase in cell viability. Expressions of cardiomyogenic specific genes, including GATA4, cTnT, Cx43 and Nkx2.5, were higher in the combined treatment groups of 5-azacytidine (5-aza) and hPL than the expressions of cardiomyogenic specific genes in the control group and in the 5-aza treatment group. In terms of the results of immunofluorescence and immunoenzymatic staining, the highest expressions of cardiomyogenic specific proteins were revealed in combined treatment groups. It can be summarized that hPL may be an effective supporting cardiomyogenic supplementary factor for cardiomyogenic differentiation in hAF-MSCs.
Collapse
Affiliation(s)
- Runchana Markmee
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinda Aungsuchawan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Waleephan Tancharoen
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Suteera Narakornsak
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Peraphan Pothacharoen
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
18
|
Rallapalli S, Guhathakurta S, Korrapati PS. Isolation, growth kinetics, and immunophenotypic characterization of adult human cardiac progenitor cells. J Cell Physiol 2020; 236:1840-1853. [PMID: 33242343 DOI: 10.1002/jcp.29965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 11/10/2022]
Abstract
The discovery of cardiac progenitor cells (CPCs) has raised expectations for the development of cell-based therapy of the heart. Although cell therapy is emerging as a novel treatment for heart failure, several issues still exist concerning an unambiguous definition of the phenotype of CPC types. There is a need to define and validate the methods for the generation of quality CPC populations used in cell therapy applications. Considering the critical roles of cardiac cell progenitors in cellular therapy, we speculate that long term culture might modulate the immunophenotypes of CPCs. Hence, a strategy to validate the isolation and cell culture expansion of cardiac cell populations was devised. Isolation of three subpopulations of human CPCs was done from a single tissue sample using explant, enzymatic isolation, and c-kit+ immunomagnetic sorting methods. The study assessed the effects of ex vivo expansion on proliferation, immunophenotypes, and differentiation of CPCs. Additionally, we report that an explant culture can take over 2 months to achieve similar cell yields, and cell sorting requires a much larger starting population to match this expansion time frame. In comparison, an enzymatic method is expected to yield equivalent quantities of CPCs in 2-3 weeks, notably at a significantly lower cost, which may intensify their use in therapeutic approaches. We determined that ex vivo expansion caused changes in cellular characteristics, and hence propose validated molecular signatures should be established to evaluate the impact of ex vivo expansion for a safe cell therapy product.
Collapse
Affiliation(s)
- Suneel Rallapalli
- Biological Material Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India
| | | | - Purna S Korrapati
- Biological Material Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India
| |
Collapse
|
19
|
Buskermolen AB, Ristori T, Mostert D, van Turnhout MC, Shishvan SS, Loerakker S, Kurniawan NA, Deshpande VS, Bouten CV. Cellular Contact Guidance Emerges from Gap Avoidance. CELL REPORTS. PHYSICAL SCIENCE 2020; 1:100055. [PMID: 32685934 PMCID: PMC7357833 DOI: 10.1016/j.xcrp.2020.100055] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/28/2020] [Accepted: 03/20/2020] [Indexed: 05/17/2023]
Abstract
In the presence of anisotropic biochemical or topographical patterns, cells tend to align in the direction of these cues-a widely reported phenomenon known as "contact guidance." To investigate the origins of contact guidance, here, we created substrates micropatterned with parallel lines of fibronectin with dimensions spanning multiple orders of magnitude. Quantitative morphometric analysis of our experimental data reveals two regimes of contact guidance governed by the length scale of the cues that cannot be explained by enforced alignment of focal adhesions. Adopting computational simulations of cell remodeling on inhomogeneous substrates based on a statistical mechanics framework for living cells, we show that contact guidance emerges from anisotropic cell shape fluctuation and "gap avoidance," i.e., the energetic penalty of cell adhesions on non-adhesive gaps. Our findings therefore point to general biophysical mechanisms underlying cellular contact guidance, without the necessity of invoking specific molecular pathways.
Collapse
Affiliation(s)
- Antonetta B.C. Buskermolen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Dylan Mostert
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mark C. van Turnhout
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Siamak S. Shishvan
- Department of Structural Engineering, University of Tabriz, Tabriz, Iran
- Department of Mechanical Engineering, University of Cambridge, Cambridge, UK
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Corresponding author
| | - Vikram S. Deshpande
- Department of Mechanical Engineering, University of Cambridge, Cambridge, UK
| | - Carlijn V.C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Corresponding author
| |
Collapse
|
20
|
Jia Y, Chang Y, Sun P, Li H, Guo Z. Inhibition of profibrotic signalling enhances the 5-azacytidine-induced reprogramming of fibroblasts into cardiomyocytes. Int J Biochem Cell Biol 2020; 122:105733. [DOI: 10.1016/j.biocel.2020.105733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/22/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
|
21
|
Abstract
Experimental models of cardiac disease play a key role in understanding the pathophysiology of the disease and developing new therapies. The features of the experimental models should reflect the clinical phenotype, which can have a wide spectrum of underlying mechanisms. We review characteristics of commonly used experimental models of cardiac physiology and pathophysiology in all translational steps including in vitro, small animal, and large animal models. Understanding their characteristics and relevance to clinical disease is the key for successful translation to effective therapies.
Collapse
|
22
|
Dokanehiifard S, Soltani BM, Ghiasi P, Baharvand H, Reza Ganjali M, Hosseinkhani S. hsa-miR-766-5p as a new regulator of mitochondrial apoptosis pathway for discriminating of cell death from cardiac differentiation. Gene 2020; 736:144448. [DOI: 10.1016/j.gene.2020.144448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/26/2020] [Accepted: 02/04/2020] [Indexed: 12/19/2022]
|
23
|
Bagheri-Hosseinabadi Z, Seyedi F, Mollaei HR, Moshrefi M, Seifalian A. Combination of 5-azaytidine and hanging drop culture convert fat cell into cardiac cell. Biotechnol Appl Biochem 2020; 68:92-101. [PMID: 32028539 DOI: 10.1002/bab.1897] [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: 10/16/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
One of the promising approaches for the treatment of cardiac disease is stem cell therapy. In this study, we compared the cardiomyogenic differentiation rate, from human adipose-derived stem cells (hADSCs) in a three-dimensional (3D) hanging drop (HD) spheroid culture system, versus a two-dimensional (2D) culture condition at different concentrations of 5-azacytidine (5-Aza). 5-Azaytidine (5-Aza) is a pyrimidine nucleoside analogue of cytidine that initiates cell differentiation programs through DNA demethylation. The hADSCs were isolated and cultured both in 2D and 3D HD conditions, with either 10 or 50 μM concentrations of 5-Aza. Then DNA content, gene expression, and protein content were analyzed. 3D HD culture resulted in a higher percentage of cells in G0/G1 and S phase in the cell division cycle, whereas 2D culture led to a greater percentage of cells in the G2/M phase. A significantly higher gene expression rate of HAND1, HAND2, cTnI, Cx43, βMHC, GATA4, NKX2.5, and MLC2V was observed in HD treated with 50 μM 5-Aza. This was confirmed by immunocytochemistry. These findings suggest that 50 μM concentration of 5-Aza can induce hADSCs to differentiate into cardiomyocytes. The differentiation rate was significantly higher when accompanied by the 3D HD culture system. This work provides a new culture system for cell differentiation for cardiovascular tissue engineering.
Collapse
Affiliation(s)
- Zahra Bagheri-Hosseinabadi
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Seyedi
- Department of Anatomy, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hamid Reza Mollaei
- Department of Medical Microbiology, Afzalipour Medical Faculty, Kerman University of Medical Science, Kerman, Iran
| | - Mojgan Moshrefi
- Medical Nanotechnology & Tissue Engineering Research Centre, Yazd Reproductive Science Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd.), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
24
|
Gorabi AM, Bianconi V, Pirro M, Banach M, Sahebkar A. Regulation of cardiac stem cells by microRNAs: State-of-the-art. Biomed Pharmacother 2019; 120:109447. [PMID: 31580971 DOI: 10.1016/j.biopha.2019.109447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/27/2022] Open
Abstract
Stem cells have a therapeutic potential in various medical conditions. In cases without sufficient response to conventional drug treatments, stem cells represent a next generation therapeutic strategy in cardiovascular diseases. Cardiac stem cells (CSCs), among a wide variety of stem cell sources, have been identified as a valid option for stem cell-based therapy in cardiovascular diseases. CSCs mainly act as a cell source to supply the physiological need for cardiovascular cells. However, they have been demonstrated to reproduce the myocardial cells under pathological settings. Despite their roles and functions have somewhat been clarified, molecular pathways underlying the regulatory mechanisms of CSCs are still not fully elucidated. Several studies have recently shown that different microRNAs (miRNAs) play a substantial role in regulating and controlling both the physiological and pathological proliferation and differentiation of stem cells. MiRNAs are small non-coding RNA molecules that regulate gene expression and may undergo aberrant expression levels during pathological conditions. Understanding the way through which miRNAs regulate CSC behavior may open up new horizons in modulating these cells in vitro to devise sophisticated approaches for treating patients with cardiovascular diseases. In this review article, we tried to discuss available evidence about the role of miRNAs in regulating CSCs.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
25
|
Li Calzi S, Cook T, Della Rocca DG, Zhang J, Shenoy V, Yan Y, Espejo A, Rathinasabapathy A, Jacobsen MH, Salazar T, Sandusky GE, Shaw LC, March K, Raizada MK, Pepine CJ, Katovich MJ, Grant MB. Complementary Embryonic and Adult Cell Populations Enhance Myocardial Repair in Rat Myocardial Injury Model. Stem Cells Int 2019; 2019:3945850. [PMID: 31781239 PMCID: PMC6875168 DOI: 10.1155/2019/3945850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/09/2019] [Accepted: 10/01/2019] [Indexed: 11/18/2022] Open
Abstract
We compared the functional outcome of Isl-1+ cardiac progenitors, CD90+ bone marrow-derived progenitor cells, and the combination of the two in a rat myocardial infarction (MI) model. Isl-1+ cells were isolated from embryonic day 12.5 (E12.5) rat hearts and expanded in vitro. Thy-1+/CD90+ cells were isolated from the bone marrow of adult Sprague-Dawley rats by immunomagnetic cell sorting. Six-week-old female Sprague-Dawley rats underwent permanent left anterior descending (LAD) coronary artery ligation and received intramyocardial injection of either saline, Isl-1+ cells, CD90+ cells, or a combination of Isl-1+ and CD90+ cells, at the time of infarction. Cells were delivered transepicardially to the peri-infarct zone. Left ventricular function was assessed by transthoracic echocardiography at 1- and 4-week post-MI and by Millar catheterization (-dP/dt and +dP/dt) at 4-week post-MI. Fluorescence in situ hybridization (Isl-1+cells) and monochrystalline iron oxide nanoparticles labeling (MION; CD90+ cells) were performed to assess biodistribution of transplanted cells. Only the combination of cells demonstrated a significant improvement of cardiac function as assessed by anterior wall contractility, dP/dt (max), and dP/dt (min), compared to Isl-1+ or CD90+ cell monotherapies. In the combination cell group, viable cells were detected at week 4 when anterior wall motion was completely restored. In conclusion, the combination of Isl-1+ cardiac progenitors and adult bone marrow-derived CD90+ cells shows prolonged and robust myocardial tissue repair and provides support for the use of complementary cell populations to enhance myocardial repair.
Collapse
Affiliation(s)
- Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0001, USA
| | - Todd Cook
- Department of Medicine, IUPUI, Indianapolis, IN 46202, USA
| | | | - Juan Zhang
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32611, USA
| | - Vinayak Shenoy
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611, USA
| | - Yuanqing Yan
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Espejo
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32611, USA
| | | | - Max H. Jacobsen
- Pathology and Laboratory Med., IUPUI, Indianapolis, IN 46202, USA
| | - Tatiana Salazar
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0001, USA
| | | | - Lynn C. Shaw
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0001, USA
| | - Keith March
- Department of Medicine, IUPUI, Indianapolis, IN 46202, USA
| | - Mohan K. Raizada
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611, USA
| | - Carl J. Pepine
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Michael J. Katovich
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32611, USA
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0001, USA
| |
Collapse
|
26
|
André E, De Pauw A, Verdoy R, Brusa D, Bouzin C, Timmermans A, Bertrand L, Balligand JL. Changes of Metabolic Phenotype of Cardiac Progenitor Cells During Differentiation: Neutral Effect of Stimulation of AMP-Activated Protein Kinase. Stem Cells Dev 2019; 28:1498-1513. [PMID: 31530214 DOI: 10.1089/scd.2019.0129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac progenitor cells (CPCs) in the adult mammalian heart, as well as exogenous CPCs injected at the border zone of infarcted tissue, display very low differentiation rate into cardiac myocytes and marginal repair capacity in the injured heart. Emerging evidence supports an obligatory metabolic shift from glycolysis to oxidative phosphorylation (OXPHOS) during stem cells differentiation, suggesting that pharmacological modulation of metabolism may improve CPC differentiation and, potentially, healing properties. In this study, we investigated the metabolic transition underlying CPC differentiation toward cardiac myocytes. In addition, we tested whether activators of adenosine monophosphate-activated protein kinase (AMPK), known to promote mitochondrial biogenesis in other cell types would also improve CPC differentiation. Stem cell antigen 1 (Sca1+) CPCs were isolated from adult mouse hearts and their phenotype compared with more mature neonatal rat cardiac myocytes (NRCMs). Under normoxia, glucose consumption and lactate release were significantly higher in CPCs than in NRCMs. Both parameters were increased in hypoxia together with increased abundance of Glut1 (glucose transporter), of the monocarboxylic transporter Mct4 (lactate efflux mediator) and of Pfkfb3 (key regulator of glycolytic rate). CPC proliferation was critically dependent on glucose and glutamine availability in the media. Oxygen consumption analysis indicates that, compared with NRCMs, CPCs exhibited lower basal and maximal respirations with lower Tomm20 protein expression and mitochondrial DNA content. This CPC metabolic phenotype profoundly changed upon in vitro differentiation, with a decrease of glucose consumption and lactate release together with increased abundance of Tnnt2, Pgc-1α, Tomm20, and mitochondrial DNA content. Proliferative CPCs express both alpha1 and -2 catalytic subunits of AMPK that is activated by A769662. However, A769662 or resveratrol (an activator of Pgc-1α and AMPK) did not promote either mitochondrial biogenesis or CPC maturation during their differentiation. We conclude that although CPC differentiation is accompanied with an increase of mitochondrial oxidative metabolism, this is not potentiated by activation of AMPK in these cells.
Collapse
Affiliation(s)
- Emilie André
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain) and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Aurélia De Pauw
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain) and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Roxane Verdoy
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain) and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Davide Brusa
- Flow Cytometry Platform, Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- I2P Imaging Platform, Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Aurélie Timmermans
- Pole of Cardiovascular Research (CARD), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain) and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Luc Bertrand
- Pole of Cardiovascular Research (CARD), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain) and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain) and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
27
|
Gasiūnienė M, Petkus G, Matuzevičius D, Navakauskas D, Navakauskienė R. Angiotensin II and TGF- β1 Induce Alterations in Human Amniotic Fluid-Derived Mesenchymal Stem Cells Leading to Cardiomyogenic Differentiation Initiation. Int J Stem Cells 2019; 12:251-264. [PMID: 31023001 PMCID: PMC6657950 DOI: 10.15283/ijsc18126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Background and Objectives Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) may be a valuable source for cardiovascular tissue engineering and cell therapy. The aim of this study is to verify angiotensin II and transforming growth factor-beta 1 (TGF-β1) as potential cardiomyogenic differentiation inducers of AF-MSCs. Methods and Results AF-MSCs were obtained from amniocentesis samples from second-trimester pregnant women, isolated and characterized by the expression of cell surface markers (CD44, CD90, CD105 positive; CD34 negative) and pluripotency genes (OCT4, SOX2, NANOG, REX1). Cardiomyogenic differentiation was induced using different concentrations of angiotensin II and TGF-β1. Successful initiation of differentiation was confirmed by alterations in cell morphology, upregulation of cardiac genes-markers NKX2-5, TBX5, GATA4, MYH6, TNNT2, DES and main cardiac ion channels genes (sodium, calcium, potassium) as determined by RT-qPCR. Western blot and immunofluorescence analysis revealed the increased expression of Connexin43, the main component of gap junctions, and Nkx2.5, the early cardiac transcription factor. Induced AF-MSCs switched their phenotype towards more energetic and started utilizing oxidative phosphorylation more than glycolysis for energy production as assessed using Agilent Seahorse XF analyzer. The immune analysis of chromatin-modifying enzymes DNMT1, HDAC1/2 and Polycomb repressive complex 1 and 2 (PRC1/2) proteins BMI1, EZH2 and SUZ12 as well as of modified histones H3 and H4 indicated global chromatin remodeling during the induced differentiation. Conclusions Angiotensin II and TGF-β1 are efficient cardiomyogenic inducers of human AF-MSCs; they initiate alterations at the gene and protein expression, metabolic and epigenetic levels in stem cells leading towards cardiomyocyte- like phenotype formation.
Collapse
Affiliation(s)
- Monika Gasiūnienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gintautas Petkus
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dalius Matuzevičius
- Electronic Systems Department, Electronics Faculty, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Dalius Navakauskas
- Electronic Systems Department, Electronics Faculty, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
28
|
Malandraki-Miller S, Lopez CA, Alonaizan R, Purnama U, Perbellini F, Pakzad K, Carr CA. Metabolic flux analyses to assess the differentiation of adult cardiac progenitors after fatty acid supplementation. Stem Cell Res 2019; 38:101458. [PMID: 31102832 PMCID: PMC6618003 DOI: 10.1016/j.scr.2019.101458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction is the most prevalent of cardiovascular diseases and pharmacological interventions do not lead to restoration of the lost cardiomyocytes. Despite extensive stem cell therapy studies, clinical trials using cardiac progenitor cells have shown moderate results. Furthermore, differentiation of endogenous progenitors to mature cardiomyocytes is rarely reported. A metabolic switch from glucose to fatty acid oxidation occurs during cardiac development and cardiomyocyte maturation, however in vitro differentiation protocols do not consider the lack of fatty acids in cell culture media. The aim of this study was to assess the effect of this metabolic switch on control and differentiated adult cardiac progenitors, by fatty acid supplementation. Addition of oleic acid stimulated the peroxisome proliferator-activated receptor alpha pathway and led to maturation of the cardiac progenitors, both before and after transforming growth factor-beta 1 differentiation. Addition of oleic acid following differentiation increased expression of myosin heavy chain 7 and connexin 43. Also, total glycolytic metabolism increased, as did mitochondrial membrane potential and glucose and fatty acid transporter expression. This work provides new insights into the importance of fatty acids, and of peroxisome proliferator-activated receptor alpha, in cardiac progenitor differentiation. Harnessing the oxidative metabolic switch induced maturation of differentiated endogenous stem cells. (200 words).
Collapse
Affiliation(s)
- Sophia Malandraki-Miller
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Colleen A Lopez
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Rita Alonaizan
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Ujang Purnama
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Filippo Perbellini
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK.
| | - Kathy Pakzad
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Carolyn A Carr
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| |
Collapse
|
29
|
Cardiomyocyte Progenitor Cells as a Functional Gene Delivery Vehicle for Long-Term Biological Pacing. Molecules 2019; 24:molecules24010181. [PMID: 30621310 PMCID: PMC6337610 DOI: 10.3390/molecules24010181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 01/16/2023] Open
Abstract
Sustained pacemaker function is a challenge in biological pacemaker engineering. Human cardiomyocyte progenitor cells (CMPCs) have exhibited extended survival in the heart after transplantation. We studied whether lentivirally transduced CMPCs that express the pacemaker current If (encoded by HCN4) can be used as functional gene delivery vehicle in biological pacing. Human CMPCs were isolated from fetal hearts using magnetic beads coated with Sca-1 antibody, cultured in nondifferentiating conditions, and transduced with a green fluorescent protein (GFP)- or HCN4-GFP-expressing lentivirus. A patch-clamp analysis showed a large hyperpolarization-activated, time-dependent inward current (−20 pA/pF at −140 mV, n = 14) with properties typical of If in HCN4-GFP-expressing CMPCs. Gap-junctional coupling between CMPCs and neonatal rat ventricular myocytes (NRVMs) was demonstrated by efficient dye transfer and changes in spontaneous beating activity. In organ explant cultures, the number of preparations showing spontaneous beating activity increased from 6.3% in CMPC/GFP-injected preparations to 68.2% in CMPC/HCN4-GFP-injected preparations (P < 0.05). Furthermore, in CMPC/HCN4-GFP-injected preparations, isoproterenol induced a significant reduction in cycle lengths from 648 ± 169 to 392 ± 71 ms (P < 0.05). In sum, CMPCs expressing HCN4-GFP functionally couple to NRVMs and induce physiologically controlled pacemaker activity and may therefore provide an attractive delivery platform for sustained pacemaker function.
Collapse
|
30
|
Scalise M, Marino F, Cianflone E, Mancuso T, Marotta P, Aquila I, Torella M, Nadal-Ginard B, Torella D. Heterogeneity of Adult Cardiac Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:141-178. [PMID: 31487023 DOI: 10.1007/978-3-030-24108-7_8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cardiac biology and heart regeneration have been intensively investigated and debated in the last 15 years. Nowadays, the well-established and old dogma that the adult heart lacks of any myocyte-regenerative capacity has been firmly overturned by the evidence of cardiomyocyte renewal throughout the mammalian life as part of normal organ cell homeostasis, which is increased in response to injury. Concurrently, reproducible evidences from independent laboratories have convincingly shown that the adult heart possesses a pool of multipotent cardiac stem/progenitor cells (CSCs or CPCs) capable of sustaining cardiomyocyte and vascular tissue refreshment after injury. CSC transplantation in animal models displays an effective regenerative potential and may be helpful to treat chronic heart failure (CHF), obviating at the poor/modest results using non-cardiac cells in clinical trials. Nevertheless, the degree/significance of cardiomyocyte turnover in the adult heart, which is insufficient to regenerate extensive damage from ischemic and non-ischemic origin, remains strongly disputed. Concurrently, different methodologies used to detect CSCs in situ have created the paradox of the adult heart harboring more than seven different cardiac progenitor populations. The latter was likely secondary to the intrinsic heterogeneity of any regenerative cell agent in an adult tissue but also to the confusion created by the heterogeneity of the cell population identified by a single cell marker used to detect the CSCs in situ. On the other hand, some recent studies using genetic fate mapping strategies claimed that CSCs are an irrelevant endogenous source of new cardiomyocytes in the adult. On the basis of these contradictory findings, here we critically reviewed the available data on adult CSC biology and their role in myocardial cell homeostasis and repair.
Collapse
Affiliation(s)
- Mariangela Scalise
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Fabiola Marino
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Teresa Mancuso
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pina Marotta
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Surgery, University of Campania "L.Vanvitelli", Naples, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
31
|
Maring JA, Lodder K, Mol E, Verhage V, Wiesmeijer KC, Dingenouts CKE, Moerkamp AT, Deddens JC, Vader P, Smits AM, Sluijter JPG, Goumans MJ. Cardiac Progenitor Cell-Derived Extracellular Vesicles Reduce Infarct Size and Associate with Increased Cardiovascular Cell Proliferation. J Cardiovasc Transl Res 2018; 12:5-17. [PMID: 30456736 PMCID: PMC6394631 DOI: 10.1007/s12265-018-9842-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022]
Abstract
Cell transplantation studies have shown that injection of progenitor cells can improve cardiac function after myocardial infarction (MI). Transplantation of human cardiac progenitor cells (hCPCs) results in an increased ejection fraction, but survival and integration are low. Therefore, paracrine factors including extracellular vesicles (EVs) are likely to contribute to the beneficial effects. We investigated the contribution of EVs by transplanting hCPCs with reduced EV secretion. Interestingly, these hCPCs were unable to reduce infarct size post-MI. Moreover, injection of hCPC-EVs did significantly reduce infarct size. Analysis of EV uptake showed cardiomyocytes and endothelial cells primarily positive and a higher Ki67 expression in these cell types. Yes-associated protein (YAP), a proliferation marker associated with Ki67, was also increased in the entire infarcted area. In summary, our data suggest that EV secretion is the driving force behind the short-term beneficial effect of hCPC transplantation on cardiac recovery after MI.
Collapse
Affiliation(s)
- Janita A Maring
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kirsten Lodder
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Emma Mol
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Vera Verhage
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Karien C Wiesmeijer
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Calinda K E Dingenouts
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Asja T Moerkamp
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Janine C Deddens
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Pieter Vader
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.,Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anke M Smits
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Joost P G Sluijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marie-José Goumans
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands.
| |
Collapse
|
32
|
Bollini S, Smits AM, Balbi C, Lazzarini E, Ameri P. Triggering Endogenous Cardiac Repair and Regeneration via Extracellular Vesicle-Mediated Communication. Front Physiol 2018; 9:1497. [PMID: 30405446 PMCID: PMC6206049 DOI: 10.3389/fphys.2018.01497] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
A variety of paracrine signals create networks within the myocardium and mediate intercellular communications. Indeed, paracrine stimulation of the endogenous regenerative program of the heart, mainly based on resident cardiac progenitor cell (CPC) activation together with cardiomyocyte proliferation, has become increasingly relevant for future cardiac medicine. In the last years, it has been shown that extracellular vesicles (EV), including exosomes (Ex), are powerful conveyors of relevant biological effects. EV have been proposed not only as promising therapeutic tool for triggering cardiac regeneration and improving repair, but also as means of better understanding the physiological and pathological relationships between specific cardiac components, including cardiomyocytes and fibroblasts. Actually, EV from different kinds of exogenous stem cells have been shown to mediate beneficial effects on the injured myocardium. Moreover, endogenous cells, like CPC can instruct cardiovascular cell types, including cardiomyocytes, while cardiac stromal cells, especially fibroblasts, secrete EV that modulate relevant aspects of cardiomyocyte biology, such as hypertrophy and electrophysiological properties. Finally, cardiomyocytes too may release EV influencing the function of other cardiac cell types. Therefore, EV-based crosstalk is thought to be important in both physiology and pathology, being involved in the responses of the heart to noxious stimuli. In this review we will discuss the role of EV in both regulating cardiac homeostasis and driving heart regeneration. In particular, we will address their role in: (i) providing cardio-protection and enhancing cardiac repair mechanisms; (ii) CPC biology; and (iii) influencing adult cardiomyocyte behavior.
Collapse
Affiliation(s)
- Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Anke M Smits
- Laboratory of Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Carolina Balbi
- Laboratory of Molecular and Cellular Cardiology, CardioCentro Ticino, Lugano, Switzerland
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine, University of Genova, Genoa, Italy.,Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
33
|
Malandraki-Miller S, Lopez CA, Al-Siddiqi H, Carr CA. Changing Metabolism in Differentiating Cardiac Progenitor Cells-Can Stem Cells Become Metabolically Flexible Cardiomyocytes? Front Cardiovasc Med 2018; 5:119. [PMID: 30283788 PMCID: PMC6157401 DOI: 10.3389/fcvm.2018.00119] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
The heart is a metabolic omnivore and the adult heart selects the substrate best suited for each circumstance, with fatty acid oxidation preferred in order to fulfill the high energy demand of the contracting myocardium. The fetal heart exists in an hypoxic environment and obtains the bulk of its energy via glycolysis. After birth, the "fetal switch" to oxidative metabolism of glucose and fatty acids has been linked to the loss of the regenerative phenotype. Various stem cell types have been used in differentiation studies, but most are cultured in high glucose media. This does not change in the majority of cardiac differentiation protocols. Despite the fact that metabolic state affects marker expression and cellular function and activity, the substrate composition is currently being overlooked. In this review we discuss changes in cardiac metabolism during development, the various protocols used to differentiate progenitor cells to cardiomyocytes, what is known about stem cell metabolism and how consideration of metabolism can contribute toward maturation of stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
| | | | | | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
34
|
Specific Cell (Re-)Programming: Approaches and Perspectives. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:71-115. [PMID: 29071403 DOI: 10.1007/10_2017_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Many disorders are manifested by dysfunction of key cell types or their disturbed integration in complex organs. Thereby, adult organ systems often bear restricted self-renewal potential and are incapable of achieving functional regeneration. This underlies the need for novel strategies in the field of cell (re-)programming-based regenerative medicine as well as for drug development in vitro. The regenerative field has been hampered by restricted availability of adult stem cells and the potentially hazardous features of pluripotent embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Moreover, ethical concerns and legal restrictions regarding the generation and use of ESCs still exist. The establishment of direct reprogramming protocols for various therapeutically valuable somatic cell types has overcome some of these limitations. Meanwhile, new perspectives for safe and efficient generation of different specified somatic cell types have emerged from numerous approaches relying on exogenous expression of lineage-specific transcription factors, coding and noncoding RNAs, and chemical compounds.It should be of highest priority to develop protocols for the production of mature and physiologically functional cells with properties ideally matching those of their endogenous counterparts. Their availability can bring together basic research, drug screening, safety testing, and ultimately clinical trials. Here, we highlight the remarkable successes in cellular (re-)programming, which have greatly advanced the field of regenerative medicine in recent years. In particular, we review recent progress on the generation of cardiomyocyte subtypes, with a focus on cardiac pacemaker cells. Graphical Abstract.
Collapse
|
35
|
Pagano F, Picchio V, Angelini F, Iaccarino A, Peruzzi M, Cavarretta E, Biondi-Zoccai G, Sciarretta S, De Falco E, Chimenti I, Frati G. The Biological Mechanisms of Action of Cardiac Progenitor Cell Therapy. Curr Cardiol Rep 2018; 20:84. [PMID: 30105430 DOI: 10.1007/s11886-018-1031-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Cell therapy for cardiovascular diseases is regarded as a rapidly growing field within regenerative medicine. Different cellular populations enriched for cardiac progenitor cells (CPCs), or derivate a-cellular products, are currently under preclinical and clinical evaluation. Here, we have reviewed the described mechanisms whereby resident post-natal CPCs, isolated in different ways, act as a therapeutic product on the damaged myocardium. RECENT FINDINGS Several biological mechanisms of action have been described which can explain the multiple therapeutic effects of CPC treatment observed on cardiac function and remodelling. These mechanisms span from direct cardiovascular differentiation, through induction of resident progenitor proliferation, to paracrine effects on cardiac and non-cardiac cells mediated by exosomes and non-coding RNAs. All the reported mechanisms of action support an integrated view including cardiomyogenesis, cardioprotection, and anti-fibrotic effects. Moreover, future developments of CPC therapy approaches may support cell-free strategies, exploiting effective pleiotropic cell-derived products, such as exosomes.
Collapse
Affiliation(s)
- Francesca Pagano
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
| | - Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
| | - Francesco Angelini
- Medical Oncology Unit, San Filippo Neri Hospital, Via Giovanni Martinotti, 20, 00135, Rome, Italy
- Experimental and Clinical Pharmacology Unit, CRO-National Cancer Institute, Via Franco Gallini 2, 33081, Aviano (PN), Italy
| | - Alessandra Iaccarino
- Department of Thoracic Surgery, "La Sapienza" University of Rome, viale Regina Margherita 324, 00161, Rome, Italy
| | - Mariangela Peruzzi
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
| | - Elena Cavarretta
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
- Department of AngioCardioNeurology, IRCCS Neuromed Institute, Via Atinense 18, 86077, Pozzilli (IS), Italy
| | - Sebastiano Sciarretta
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
- Department of AngioCardioNeurology, IRCCS Neuromed Institute, Via Atinense 18, 86077, Pozzilli (IS), Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy.
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnologies, "La Sapienza" University of Rome, Corso della Repubblica 79, 04100, Latina, Italy
- Department of AngioCardioNeurology, IRCCS Neuromed Institute, Via Atinense 18, 86077, Pozzilli (IS), Italy
| |
Collapse
|
36
|
Quantitative Secretomics Reveals Extrinsic Signals Involved in Human Pluripotent Stem Cell Cardiomyogenesis. Proteomics 2018; 18:e1800102. [DOI: 10.1002/pmic.201800102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/17/2018] [Indexed: 12/22/2022]
|
37
|
van den Akker F, Vrijsen KR, Deddens JC, Buikema JW, Mokry M, van Laake LW, Doevendans PA, Sluijter JPG. Suppression of T cells by mesenchymal and cardiac progenitor cells is partly mediated via extracellular vesicles. Heliyon 2018; 4:e00642. [PMID: 30003150 PMCID: PMC6040605 DOI: 10.1016/j.heliyon.2018.e00642] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/11/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Adverse remodeling after myocardial infarction (MI) is strongly influenced by T cells. Stem cell therapy after MI, using mesenchymal stem cells (MSC) or cardiomyocyte progenitor cells (CMPC), improved cardiac function, despite low cell retention and limited differentiation. As MSC secrete many factors affecting T cell proliferation and function, we hypothesized the immune response could be affected as one of the targets of stem cell therapy. Therefore, we studied the immunosuppressive properties of human BM-MSC and CMPC and their extracellular vesicles (EVs) in co-culture with activated T cells. Proliferation of T cells, measured by carboxyfluorescein succinimidyl ester dilution, was significantly reduced in the presence of BM-MSC and CMPC. The inflammatory cytokine panel of the T cells in co-culture, measured by Luminex assay, changed, with strong downregulation of IFN-gamma and TNF-alpha. The effect on proliferation was observed in both direct cell contact and transwell co-culture systems. Transfer of conditioned medium to unrelated T cells abrogated proliferation in these cells. EVs isolated from the conditioned medium of BM-MSC and CMPC prevented T cell proliferation in a dose-dependent fashion. Progenitor cells presence induces up- and downregulation of multiple previously unreported pathways in T cells. In conclusion, both BM-MSC and CMPC have a strong capacity for in vitro immunosuppression. This effect is mediated by paracrine factors, such as extracellular vesicles. Besides proliferation, many additional pathways are influenced by both BM-MSC and CMPC.
Collapse
Affiliation(s)
- F van den Akker
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands
| | - K R Vrijsen
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands
| | - J C Deddens
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands
| | - J W Buikema
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands
| | - M Mokry
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, The Netherlands
| | - L W van Laake
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands
| | - P A Doevendans
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands.,ICIN - Netherlands Heart Institute, Utrecht, The Netherlands
| | - J P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, The Netherlands.,ICIN - Netherlands Heart Institute, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
38
|
Middleton RC, Rogers RG, De Couto G, Tseliou E, Luther K, Holewinski R, Soetkamp D, Van Eyk JE, Antes TJ, Marbán E. Newt cells secrete extracellular vesicles with therapeutic bioactivity in mammalian cardiomyocytes. J Extracell Vesicles 2018; 7:1456888. [PMID: 29696078 PMCID: PMC5912190 DOI: 10.1080/20013078.2018.1456888] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 03/17/2018] [Indexed: 01/18/2023] Open
Abstract
Newts can regenerate amputated limbs and cardiac tissue, unlike mammals which lack broad regenerative capacity. Several signaling pathways involved in cell proliferation, differentiation and survival during newt tissue regeneration have been elucidated, however the factors that coordinate signaling between cells, as well as the conservation of these factors in other animals, are not well defined. Here we report that media conditioned by newt limb explant cells (A1 cells) protect mammalian cardiomyocytes from oxidative stress-induced apoptosis. The cytoprotective effect of A1-conditioned media was negated by exposing A1 cells to GW4869, which suppresses the generation of extracellular vesicles (EVs). A1-EVs are similar in diameter (~100–150 nm), structure, and share several membrane surface and cargo proteins with mammalian exosomes. However, isolated A1-EVs contain significantly higher levels of both RNA and protein per particle than mammalian EVs. Additionally, numerous cargo RNAs and proteins are unique to A1-EVs. Of particular note, A1-EVs contain numerous mRNAs encoding nuclear receptors, membrane ligands, as well as transcription factors. Mammalian cardiomyocytes treated with A1-EVs showed increased expression of genes in the PI3K/AKT pathway, a pivotal player in survival signaling. We conclude that newt cells secrete EVs with diverse, distinctive RNA and protein contents. Despite ~300 million years of evolutionary divergence between newts and mammals, newt EVs confer cytoprotective effects on mammalian cardiomyocytes.
Collapse
Affiliation(s)
- Ryan C Middleton
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Russell G Rogers
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Geoffrey De Couto
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eleni Tseliou
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kristin Luther
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ronald Holewinski
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel Soetkamp
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Travis J Antes
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
39
|
Muncie JM, Weaver VM. The Physical and Biochemical Properties of the Extracellular Matrix Regulate Cell Fate. Curr Top Dev Biol 2018; 130:1-37. [PMID: 29853174 DOI: 10.1016/bs.ctdb.2018.02.002] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The extracellular matrix is a complex network of hydrated macromolecular proteins and sugars that, in concert with bound soluble factors, comprise the acellular stromal microenvironment of tissues. Rather than merely providing structural information to cells, the extracellular matrix plays an instructive role in development and is critical for the maintenance of tissue homeostasis. In this chapter, we review the composition of the extracellular matrix and summarize data illustrating its importance in embryogenesis, tissue-specific development, and stem cell differentiation. We discuss how the biophysical and biochemical properties of the extracellular matrix ligate specific transmembrane receptors to activate intracellular signaling that alter cell shape and cytoskeletal dynamics to modulate cell growth and viability, and direct cell migration and cell fate. We present examples describing how the extracellular matrix functions as a highly complex physical and chemical entity that regulates tissue organization and cell behavior through a dynamic and reciprocal dialogue with the cellular constituents of the tissue. We suggest that the extracellular matrix not only transmits cellular and tissue-level force to shape development and tune cellular activities that are key for coordinated tissue behavior, but that it is itself remodeled such that it temporally evolves to maintain the integrated function of the tissue. Accordingly, we argue that perturbations in extracellular matrix composition and structure compromise key developmental events and tissue homeostasis, and promote disease.
Collapse
Affiliation(s)
- Jonathon M Muncie
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA, United States; Graduate Program in Bioengineering, University of California San Francisco and University of California Berkeley, San Francisco, CA, United States
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
40
|
Zhang J, Guo F, Wu H, Wei J, Xian M, Fan F, Tang S, Zhao Y, Zhang Y, Li D, Yang H. Yixin-Shu facilitated cardiac-like differentiation of mesenchymal stem cells in vitro. RSC Adv 2018; 8:10032-10039. [PMID: 35540854 PMCID: PMC9078824 DOI: 10.1039/c7ra13326j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/20/2018] [Indexed: 11/21/2022] Open
Abstract
Yixin-Shu capsules (YXS) are a Chinese Materia Medica standardized product used for heart disease and their effectiveness has been demonstrated through both clinical and experimental research. However, the mechanism involved has remained unclear. The effect of YXS on the cardiac-like differentiation of mesenchymal stem cells was investigated in this study. The intestinal absorption liquid of YXS was prepared using an in vitro intestinal absorption method and 62 compounds have been identified. A compound-target-function network constructed by a network pharmacology-based approach indicated that these compounds had an effect on cell differentiation. The effects of YXS on cardiac-like differentiation of mesenchymal stem cells was verified by detecting cardiac-specific protein expression such as α-actinin, cardiac troponin-I and desmin through real time-PCR, western blotting and immunofluorescence staining. A network pharmacology analysis indicated that the facilitation of YXS on the cardiac-like differentiation may be through the TGF-β signaling pathway, Wnt signaling pathway and MAPK signaling pathway. The observed improvements on cardiac differentiation may be due to the novel molecular mechanism for YXS that could also benefit developments in cardiac tissue engineering.
Collapse
Affiliation(s)
- Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Hongwei Wu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Junying Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Minghua Xian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Fangfang Fan
- College of Ethnic Medicine, Chengdu University of TCM Chengdu 610072 China
| | - Shihuan Tang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Ye Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Yi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Defeng Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences Beijing 100700 China +86-10-64013996 +86-10-64032656
| |
Collapse
|
41
|
Nummi A, Nieminen T, Pätilä T, Lampinen M, Lehtinen ML, Kivistö S, Holmström M, Wilkman E, Teittinen K, Laine M, Sinisalo J, Kupari M, Kankuri E, Juvonen T, Vento A, Suojaranta R, Harjula A. Epicardial delivery of autologous atrial appendage micrografts during coronary artery bypass surgery-safety and feasibility study. Pilot Feasibility Stud 2017; 3:74. [PMID: 29276625 PMCID: PMC5738681 DOI: 10.1186/s40814-017-0217-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/01/2017] [Indexed: 12/14/2022] Open
Abstract
Background The atrial appendages are a tissue reservoir for cardiac stem cells. During on-pump coronary artery bypass graft (CABG) surgery, part of the right atrial appendage can be excised upon insertion of the right atrial cannula of the heart-lung machine. In the operating room, the removed tissue can be easily cut into micrografts for transplantation. This trial aims to assess the safety and feasibility of epicardial transplantation of atrial appendage micrografts in patients undergoing CABG surgery. Methods/design Autologous cardiac micrografts are made from leftover right atrial appendage during CABG of 6 patients. Atrial appendage is mechanically processed to micrografts consisting of atrial appendage-derived cells (AADCs) and their extracellular matrix (ECM). The micrografts are epicardially transplanted in a fibrin gel and covered with a tissue-engineered ECM sheet. Parameters including echocardiography—reflecting cardiac insufficiency—are studied pre- and post-operatively as well as at 3 and 6 months of the follow-up. Cardiac functional magnetic resonance imaging is performed preoperatively and at 6-month follow-up. The primary outcome measures are patient safety in terms of hemodynamic and cardiac function over time and feasibility of therapy administration in a clinical setting. Secondary outcome measures are left ventricular wall thickness, change in the amount of myocardial scar tissue, changes in left ventricular ejection fraction, plasma concentrations of N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels, New York Heart Association class, days in hospital, and changes in the quality of life. Twenty patients undergoing routine CAGB surgery will be recruited to serve as a control group. Discussion This study aims to address the surgical feasibility and patient safety of epicardially delivered atrial appendage micrografts during CABG surgery. Delivery of autologous micrografts and AADCs has potential applications for cell and cell-based gene therapies. Trial registration ClinicalTrials.gov Identifier: NCT02672163. Date of registration: 02.02.2016
Collapse
Affiliation(s)
- Annu Nummi
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomo Nieminen
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Internal Medicine, South Karelia Central Hospital, Lappeenranta, Finland
| | - Tommi Pätilä
- Pediatric Cardiac Surgery, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Milla Lampinen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Miia L Lehtinen
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sari Kivistö
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Miia Holmström
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Erika Wilkman
- Department of Anesthesiology and Intensive Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kari Teittinen
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Laine
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Markku Kupari
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tatu Juvonen
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Vento
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Raili Suojaranta
- Department of Anesthesiology and Intensive Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ari Harjula
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | |
Collapse
|
42
|
Myocardial Regeneration via Progenitor Cell-Derived Exosomes. Stem Cells Int 2017; 2017:7849851. [PMID: 29333167 PMCID: PMC5733225 DOI: 10.1155/2017/7849851] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/11/2017] [Indexed: 12/16/2022] Open
Abstract
In the past 20 years, a variety of cell products has been evaluated in terms of their capacity to treat patients with acute myocardial infarction and chronic heart failure. Despite initial enthusiasm, therapeutic efficacy has overall been disappointing, and clinical application is costly and complex. Recently, a subset of small extracellular vesicles (EVs), commonly referred to as "exosomes," was shown to confer cardioprotective and regenerative signals at a magnitude similar to that of their donor cells. The conceptual advantage is that they may be produced in industrial quantities and stored at the point-of-care for off-the-shelf application, ideally without eliciting a relevant recipient immune response or other adverse effects associated with viable cells. The body of evidence on beneficial exosome-mediated effects in animal models of heart diseases is rapidly growing. However, there is significant heterogeneity in terms of exosome source cells, isolation process, therapeutic dosage, and delivery mode. This review summarizes the current state of research on exosomes as experimental therapy of heart diseases and seeks to identify roadblocks that need to be overcome prior to clinical application.
Collapse
|
43
|
Moerkamp AT, Leung HW, Bax NAM, Holst S, Lodder K, Berends T, Dingenouts CKE, Choo A, Smits AM, Goumans MJ. Glycosylated Cell Surface Markers for the Isolation of Human Cardiac Progenitors. Stem Cells Dev 2017; 26:1552-1565. [PMID: 28891400 DOI: 10.1089/scd.2017.0048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of stem cell therapy after cardiac injury is to replace damaged cardiac tissue. Human cardiac progenitor cells (CPCs) represent an interesting cell population for clinical strategies to treat cardiac disease and human CPC-specific antibodies would aid in the clinical implementation of cardiac progenitor-based cell therapy. However, the field of CPC biology suffers from the lack of human CPC-specific markers. Therefore, we raised a panel of monoclonal antibodies (mAb) against CPCs. Of this panel of antibodies, we show that mAb C1096 recognizes a progenitor-like population in the fetal and adult human heart and partially colocalize with reported CPC populations in vitro. Furthermore, mAb C1096 can be used to isolate a multipotent progenitor population from human heart tissue. Interestingly, the two lead candidates, mAb C1096 and mAb C19, recognize glycosylated residues on PECAM1 (platelet and endothelial cell adhesion molecule 1) and GRP78, respectively, and de-N-glycosylation significantly abolishes their binding. Thereby, this report describes new clinically applicable antibodies against human CPCs, and for the first time demonstrates the importance of glycosylated residues as CPCs specific markers.
Collapse
Affiliation(s)
- Asja T Moerkamp
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands
| | - Hau Wan Leung
- 2 Bioprocessing Technology Institute , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Noortje A M Bax
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands .,3 Department of Biomedical Engineering, Eindhoven University of Technology , Eindhoven, the Netherlands
| | - Stephanie Holst
- 4 Center for Proteomics and Metabolomics, Leiden University Medical Center , Leiden, the Netherlands
| | - Kirsten Lodder
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands
| | - Thijs Berends
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands
| | - Calinda K E Dingenouts
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands
| | - Andre Choo
- 2 Bioprocessing Technology Institute , Agency for Science, Technology and Research (A*STAR), Singapore, Singapore .,5 Department of Bioengineering, National University of Singapore , Singapore, Singapore
| | - Anke M Smits
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands
| | - Marie-José Goumans
- 1 Department of Molecular Cell Biology, Leiden University Medical Center , Leiden, the Netherlands
| |
Collapse
|
44
|
Smit NW, Cócera Ortega L, Végh AMD, Meijborg VMF, Smits AM, Klerk M, Tijsen AJM, Tan HL, Goumans MJHT, Boink GJJ, Coronel R. Human Cardiomyocyte Progenitor Cells in Co-culture with Rat Cardiomyocytes Form a Pro-arrhythmic Substrate: Evidence for Two Different Arrhythmogenic Mechanisms. Front Physiol 2017; 8:797. [PMID: 29075204 PMCID: PMC5644204 DOI: 10.3389/fphys.2017.00797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/28/2017] [Indexed: 01/05/2023] Open
Abstract
Background: Cardiomyocyte progenitor cells (CMPCs) are a promising cell source for regenerative cell therapy to improve cardiac function after myocardial infarction. However, it is unknown whether undifferentiated CMPCs have arrhythmogenic risks. We investigate whether undifferentiated, regionally applied, human fetal CMPCs form a pro-arrhythmic substrate in co-culture with neonatal rat ventricular myocytes (NRVMs). Method: Unipolar extracellular electrograms, derived from micro-electrode arrays (8 × 8 electrodes) containing monolayers of NRVMs (control), or co-cultures of NRVMs and locally seeded CMPCs were used to determine conduction velocity and the incidence of tachy-arrhythmias. Micro-electrodes were used to record action potentials. Conditioned medium (Cme) of CMPCs was used to distinguish between coupling or paracrine effects. Results: Co-cultures demonstrated conduction slowing (5.6 ± 0.3 cm/s, n = 50) compared to control monolayers (13.4 ± 0.4 cm/s, n = 26) and monolayers subjected to Cme (13.7 ± 0.6 cm/s, n = 11, all p < 0.001). Furthermore, co-cultures had a more depolarized resting membrane than control monolayers (−47.3 ± 17.4 vs. −64.8 ± 7.7 mV, p < 0.001) and monolayers subjected to Cme (−64.4 ± 8.1 mV, p < 0.001). Upstroke velocity was significantly decreased in co-cultures and action potential duration was prolonged. The CMPC region was characterized by local ST-elevation in the recorded electrograms. The spontaneous rhythm was faster and tachy-arrhythmias occurred more often in co-cultured monolayers than in control monolayers (42.0 vs. 5.4%, p < 0.001). Conclusion: CMPCs form a pro-arrhythmic substrate when co-cultured with neonatal cardiomyocytes. Electrical coupling between both cell types leads to current flow between a, slowly conducting, depolarized and the normal region leading to local ST-elevations and the occurrence of tachy-arrhythmias originating from the non-depolarized zone.
Collapse
Affiliation(s)
- Nicoline W Smit
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| | - Lucia Cócera Ortega
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Anna M D Végh
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Veronique M F Meijborg
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| | - Anke M Smits
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Mischa Klerk
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Anke J M Tijsen
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hanno L Tan
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Marie-José H T Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Gerard J J Boink
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Ruben Coronel
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,IHU Liryc, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, Bordeaux, France
| |
Collapse
|
45
|
(Re-)programming of subtype specific cardiomyocytes. Adv Drug Deliv Rev 2017; 120:142-167. [PMID: 28916499 DOI: 10.1016/j.addr.2017.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/29/2017] [Accepted: 09/07/2017] [Indexed: 01/10/2023]
Abstract
Adult cardiomyocytes (CMs) possess a highly restricted intrinsic regenerative potential - a major barrier to the effective treatment of a range of chronic degenerative cardiac disorders characterized by cellular loss and/or irreversible dysfunction and which underlies the majority of deaths in developed countries. Both stem cell programming and direct cell reprogramming hold promise as novel, potentially curative approaches to address this therapeutic challenge. The advent of induced pluripotent stem cells (iPSCs) has introduced a second pluripotent stem cell source besides embryonic stem cells (ESCs), enabling even autologous cardiomyocyte production. In addition, the recent achievement of directly reprogramming somatic cells into cardiomyocytes is likely to become of great importance. In either case, different clinical scenarios will require the generation of highly pure, specific cardiac cellular-subtypes. In this review, we discuss these themes as related to the cardiovascular stem cell and programming field, including a focus on the emergent topic of pacemaker cell generation for the development of biological pacemakers and in vitro drug testing.
Collapse
|
46
|
Cardiac Progenitor Cells and the Interplay with Their Microenvironment. Stem Cells Int 2017; 2017:7471582. [PMID: 29075298 PMCID: PMC5623801 DOI: 10.1155/2017/7471582] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The microenvironment plays a crucial role in the behavior of stem and progenitor cells. In the heart, cardiac progenitor cells (CPCs) reside in specific niches, characterized by key components that are altered in response to a myocardial infarction. To date, there is a lack of knowledge on these niches and on the CPC interplay with the niche components. Insight into these complex interactions and into the influence of microenvironmental factors on CPCs can be used to promote the regenerative potential of these cells. In this review, we discuss cardiac resident progenitor cells and their regenerative potential and provide an overview of the interactions of CPCs with the key elements of their niche. We focus on the interaction between CPCs and supporting cells, extracellular matrix, mechanical stimuli, and soluble factors. Finally, we describe novel approaches to modulate the CPC niche that can represent the next step in recreating an optimal CPC microenvironment and thereby improve their regeneration capacity.
Collapse
|
47
|
Castilho M, Feyen D, Flandes-Iparraguirre M, Hochleitner G, Groll J, Doevendans PAF, Vermonden T, Ito K, Sluijter JPG, Malda J. Melt Electrospinning Writing of Poly-Hydroxymethylglycolide-co-ε-Caprolactone-Based Scaffolds for Cardiac Tissue Engineering. Adv Healthc Mater 2017; 6:10.1002/adhm.201700311. [PMID: 28699224 PMCID: PMC7116102 DOI: 10.1002/adhm.201700311] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/24/2017] [Indexed: 12/31/2022]
Abstract
Current limitations in cardiac tissue engineering revolve around the inability to fully recapitulate the structural organization and mechanical environment of native cardiac tissue. This study aims at developing organized ultrafine fiber scaffolds with improved biocompatibility and architecture in comparison to the traditional fiber scaffolds obtained by solution electrospinning. This is achieved by combining the additive manufacturing of a hydroxyl-functionalized polyester, (poly(hydroxymethylglycolide-co-ε-caprolactone) (pHMGCL), with melt electrospinning writing (MEW). The use of pHMGCL with MEW vastly improves the cellular response to the mechanical anisotropy. Cardiac progenitor cells (CPCs) are able to align more efficiently along the preferential direction of the melt electrospun pHMGCL fiber scaffolds in comparison to electrospun poly(ε-caprolactone)-based scaffolds. Overall, this study describes for the first time that highly ordered microfiber (4.0-7.0 µm) scaffolds based on pHMGCL can be reproducibly generated with MEW and that these scaffolds can support and guide the growth of CPCs and thereby potentially enhance their therapeutic potential.
Collapse
Affiliation(s)
- Miguel Castilho
- Department of Orthopaedics, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, P. O. Box 513, Eindhoven, MB 5600, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, Utrecht, CT 3584, The Netherlands
| | - Dries Feyen
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, Utrecht, CT 3584, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
| | - María Flandes-Iparraguirre
- Department of Orthopaedics, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, Utrecht, CT 3584, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
| | - Gernot Hochleitner
- Department of Functional Materials in Medicine and Dentistry, and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, Würzburg 97070, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, Würzburg 97070, Germany
| | - Pieter A. F. Doevendans
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P. O. Box 80082, Utrecht, TB 3508, The Netherlands
| | - Keita Ito
- Department of Orthopaedics, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, P. O. Box 513, Eindhoven, MB 5600, The Netherlands
| | - Joost P. G. Sluijter
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, Utrecht, CT 3584, The Netherlands
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, P.O. Box 85500, Utrecht, GA 3508, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, Utrecht, CT 3584, The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CM, The Netherlands
| |
Collapse
|
48
|
Castaldi A, Dodia RM, Orogo AM, Zambrano CM, Najor RH, Gustafsson ÅB, Heller Brown J, Purcell NH. Decline in cellular function of aged mouse c-kit + cardiac progenitor cells. J Physiol 2017; 595:6249-6262. [PMID: 28737214 PMCID: PMC5621489 DOI: 10.1113/jp274775] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS While autologous stem cell-based therapies are currently being tested on elderly patients, there are limited data on the function of aged stem cells and in particular c-kit+ cardiac progenitor cells (CPCs). We isolated c-kit+ cells from young (3 months) and aged (24 months) C57BL/6 mice to compare their biological properties. Aged CPCs have increased senescence, decreased stemness and reduced capacity to proliferate or to differentiate following dexamethasone (Dex) treatment in vitro, as evidenced by lack of cardiac lineage gene upregulation. Aged CPCs fail to activate mitochondrial biogenesis and increase proteins involved in mitochondrial oxidative phosphorylation in response to Dex. Aged CPCs fail to upregulate paracrine factors that are potentially important for proliferation, survival and angiogenesis in response to Dex. The results highlight marked differences between young and aged CPCs, which may impact future design of autologous stem cell-based therapies. ABSTRACT Therapeutic use of c-kit+ cardiac progenitor cells (CPCs) is being evaluated for regenerative therapy in older patients with ischaemic heart failure. Our understanding of the biology of these CPCs has, however, largely come from studies of young cells and animal models. In the present study we examined characteristics of CPCs isolated from young (3 months) and aged (24 months) mice that could underlie the diverse outcomes reported for CPC-based therapeutics. We observed morphological differences and altered senescence indicated by increased senescence-associated markers β-galactosidase and p16 mRNA in aged CPCs. The aged CPCs also proliferated more slowly than their young counterparts and expressed lower levels of the stemness marker LIN28. We subsequently treated the cells with dexamethasone (Dex), routinely used to induce commitment in CPCs, for 7 days and analysed expression of cardiac lineage marker genes. While MEF2C, GATA4, GATA6 and PECAM mRNAs were significantly upregulated in response to Dex treatment in young CPCs, their expression was not increased in aged CPCs. Interestingly, Dex treatment of aged CPCs also failed to increase mitochondrial biogenesis and expression of the mitochondrial proteins Complex III and IV, consistent with a defect in mitochondria complex assembly in the aged CPCs. Dex-treated aged CPCs also had impaired ability to upregulate expression of paracrine factor genes and the conditioned media from these cells had reduced ability to induce angiogenesis in vitro. These findings could impact the design of future CPC-based therapeutic approaches for the treatment of older patients suffering from cardiac injury.
Collapse
Affiliation(s)
- Alessandra Castaldi
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Ramsinh Mansinh Dodia
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.,California State University, Channel Islands, Camarillo, CA, USA
| | - Amabel M Orogo
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Cristina M Zambrano
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Rita H Najor
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Joan Heller Brown
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Nicole H Purcell
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
49
|
Adult cardiac stem cells are multipotent and robustly myogenic: c-kit expression is necessary but not sufficient for their identification. Cell Death Differ 2017; 24:2101-2116. [PMID: 28800128 PMCID: PMC5686347 DOI: 10.1038/cdd.2017.130] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/14/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022] Open
Abstract
Multipotent adult resident cardiac stem cells (CSCs) were first identified by the expression of c-kit, the stem cell factor receptor. However, in the adult myocardium c-kit alone cannot distinguish CSCs from other c-kit-expressing (c-kitpos) cells. The adult heart indeed contains a heterogeneous mixture of c-kitpos cells, mainly composed of mast and endothelial/progenitor cells. This heterogeneity of cardiac c-kitpos cells has generated confusion and controversy about the existence and role of CSCs in the adult heart. Here, to unravel CSC identity within the heterogeneous c-kit-expressing cardiac cell population, c-kitpos cardiac cells were separated through CD45-positive or -negative sorting followed by c-kitpos sorting. The blood/endothelial lineage-committed (Lineagepos) CD45posc-kitpos cardiac cells were compared to CD45neg(Lineageneg/Linneg) c-kitpos cardiac cells for stemness and myogenic properties in vitro and in vivo. The majority (~90%) of the resident c-kitpos cardiac cells are blood/endothelial lineage-committed CD45posCD31posc-kitpos cells. In contrast, the LinnegCD45negc-kitpos cardiac cell cohort, which represents ⩽10% of the total c-kitpos cells, contain all the cardiac cells with the properties of adult multipotent CSCs. These characteristics are absent from the c-kitneg and the blood/endothelial lineage-committed c-kitpos cardiac cells. Single Linnegc-kitpos cell-derived clones, which represent only 1-2% of total c-kitpos myocardial cells, when stimulated with TGF-β/Wnt molecules, acquire full transcriptome and protein expression, sarcomere organisation, spontaneous contraction and electrophysiological properties of differentiated cardiomyocytes (CMs). Genetically tagged cloned progeny of one Linnegc-kitpos cell when injected into the infarcted myocardium, results in significant regeneration of new CMs, arterioles and capillaries, derived from the injected cells. The CSC's myogenic regenerative capacity is dependent on commitment to the CM lineage through activation of the SMAD2 pathway. Such regeneration was not apparent when blood/endothelial lineage-committed c-kitpos cardiac cells were injected. Thus, among the cardiac c-kitpos cell cohort only a very small fraction has the phenotype and the differentiation/regenerative potential characteristics of true multipotent CSCs.
Collapse
|
50
|
Du Pré BC, Demkes EJ, Feyen DAM, Dierickx P, Crnko S, Kok BJM, Sluijter JPG, Doevendans PA, Vos MA, Van Veen TAB, Van Laake LW. SCA1 + Cells from the Heart Possess a Molecular Circadian Clock and Display Circadian Oscillations in Cellular Functions. Stem Cell Reports 2017; 9:762-769. [PMID: 28803917 PMCID: PMC5599230 DOI: 10.1016/j.stemcr.2017.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 11/16/2022] Open
Abstract
Stem cell antigen 1-positive (SCA1+) cells (SPCs) have been investigated in cell-based cardiac repair and pharmacological research, although improved cardiac function after injection has been variable and the mode of action remains unclear. Circadian (24-hr) rhythms are biorhythms regulated by molecular clocks that play an important role in (patho)physiology. Here, we describe (1) the presence of a molecular circadian clock in SPCs and (2) circadian rhythmicity in SPC function. We isolated SPCs from human fetal heart and found that these cells possess a molecular clock based on typical oscillations in core clock components BMAL1 and CRY1. Functional analyses revealed that circadian rhythmicity also governs SPC proliferation, stress tolerance, and growth factor release, with large differences between peaks and troughs. We conclude that SPCs contain a circadian molecular clock that controls crucial cellular functions. Taking circadian rhythms into account may improve reproducibility and outcome of research and therapies using SPCs.
SCA1+ cells are a cell source used in pharmacology studies and cardiac repair SCA1+ cells possess a molecular circadian (24-hr) clock Proliferation, stress tolerance, and paracrine secretion follow a circadian pattern Taking rhythmicity into account may improve studies using SCA1+ cells
Collapse
Affiliation(s)
- Bastiaan C Du Pré
- Department of Medical Physiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Evelyne J Demkes
- Department of Medical Physiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Dries A M Feyen
- Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Pieterjan Dierickx
- Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Hubrecht Institute-KNAW, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Sandra Crnko
- Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Bart J M Kok
- Department of Medical Physiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Marc A Vos
- Department of Medical Physiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Toon A B Van Veen
- Department of Medical Physiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Linda W Van Laake
- Department of Cardiology, Division of Heart and Lungs, and Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|