1
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [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: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Bak ST, Harvald EB, Ellman DG, Mathiesen SB, Chen T, Fang S, Andersen KS, Fenger CD, Burton M, Thomassen M, Andersen DC. Ploidy-stratified single cardiomyocyte transcriptomics map Zinc Finger E-Box Binding Homeobox 1 to underly cardiomyocyte proliferation before birth. Basic Res Cardiol 2023; 118:8. [PMID: 36862248 PMCID: PMC9981540 DOI: 10.1007/s00395-023-00979-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/31/2022] [Accepted: 01/21/2023] [Indexed: 03/03/2023]
Abstract
Whereas cardiomyocytes (CMs) in the fetal heart divide, postnatal CMs fail to undergo karyokinesis and/or cytokinesis and therefore become polyploid or binucleated, a key process in terminal CM differentiation. This switch from a diploid proliferative CM to a terminally differentiated polyploid CM remains an enigma and seems an obstacle for heart regeneration. Here, we set out to identify the transcriptional landscape of CMs around birth using single cell RNA sequencing (scRNA-seq) to predict transcription factors (TFs) involved in CM proliferation and terminal differentiation. To this end, we established an approach combining fluorescence activated cell sorting (FACS) with scRNA-seq of fixed CMs from developing (E16.5, P1, and P5) mouse hearts, and generated high-resolution single-cell transcriptomic maps of in vivo diploid and tetraploid CMs, increasing the CM resolution. We identified TF-networks regulating the G2/M phases of developing CMs around birth. ZEB1 (Zinc Finger E-Box Binding Homeobox 1), a hereto unknown TF in CM cell cycling, was found to regulate the highest number of cell cycle genes in cycling CMs at E16.5 but was downregulated around birth. CM ZEB1-knockdown reduced proliferation of E16.5 CMs, while ZEB1 overexpression at P0 after birth resulted in CM endoreplication. These data thus provide a ploidy stratified transcriptomic map of developing CMs and bring new insight to CM proliferation and endoreplication identifying ZEB1 as a key player in these processes.
Collapse
Affiliation(s)
- Sara Thornby Bak
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Eva Bang Harvald
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Ditte Gry Ellman
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Sabrina Bech Mathiesen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Ting Chen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Shu Fang
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Kristian Skriver Andersen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | | | - Mark Burton
- Clinical Institute, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Mads Thomassen
- Clinical Institute, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Ditte Caroline Andersen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark.
- Clinical Institute, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
3
|
Vadakke‐Madathil S, Chaudhry HW. Concepts of Cell Therapy and Myocardial Regeneration. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
4
|
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
|
5
|
Beliën H, Evens L, Hendrikx M, Bito V, Bronckaers A. Combining stem cells in myocardial infarction: The road to superior repair? Med Res Rev 2021; 42:343-373. [PMID: 34114238 DOI: 10.1002/med.21839] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/04/2021] [Accepted: 05/29/2021] [Indexed: 12/25/2022]
Abstract
Myocardial infarction irreversibly destroys millions of cardiomyocytes in the ventricle, making it the leading cause of heart failure worldwide. Over the past two decades, many progenitor and stem cell types were proposed as the ideal candidate to regenerate the heart after injury. The potential of stem cell therapy has been investigated thoroughly in animal and human studies, aiming at cardiac repair by true tissue replacement, by immune modulation, or by the secretion of paracrine factors that stimulate endogenous repair processes. Despite some successful results in animal models, the outcome from clinical trials remains overall disappointing, largely due to the limited stem cell survival and retention after transplantation. Extensive interest was developed regarding the combinational use of stem cells and various priming strategies to improve the efficacy of regenerative cell therapy. In this review, we provide a critical discussion of the different stem cell types investigated in preclinical and clinical studies in the field of cardiac repair. Moreover, we give an update on the potential of stem cell combinations as well as preconditioning and explore the future promises of these novel regenerative strategies.
Collapse
Affiliation(s)
- Hanne Beliën
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Lize Evens
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Virginie Bito
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| |
Collapse
|
6
|
Portillo Esquivel LE, Zhang B. Application of Cell, Tissue, and Biomaterial Delivery in Cardiac Regenerative Therapy. ACS Biomater Sci Eng 2021; 7:1000-1021. [PMID: 33591735 DOI: 10.1021/acsbiomaterials.0c01805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death around the world, being responsible for 31.8% of all deaths in 2017 (Roth, G. A. et al. The Lancet 2018, 392, 1736-1788). The leading cause of CVD is ischemic heart disease (IHD), which caused 8.1 million deaths in 2013 (Benjamin, E. J. et al. Circulation 2017, 135, e146-e603). IHD occurs when coronary arteries in the heart are narrowed or blocked, preventing the flow of oxygen and blood into the cardiac muscle, which could provoke acute myocardial infarction (AMI) and ultimately lead to heart failure and death. Cardiac regenerative therapy aims to repair and refunctionalize damaged heart tissue through the application of (1) intramyocardial cell delivery, (2) epicardial cardiac patch, and (3) acellular biomaterials. In this review, we aim to examine these current approaches and challenges in the cardiac regenerative therapy field.
Collapse
Affiliation(s)
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontaria L8S 4L8, Canada
| |
Collapse
|
7
|
Haller C, Friedberg MK, Laflamme MA. The role of regenerative therapy in the treatment of right ventricular failure: a literature review. Stem Cell Res Ther 2020; 11:502. [PMID: 33239066 PMCID: PMC7687832 DOI: 10.1186/s13287-020-02022-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/09/2020] [Indexed: 01/13/2023] Open
Abstract
Right ventricular (RV) failure is a commonly encountered problem in patients with congenital heart disease but can also be a consequence of left ventricular disease, primary pulmonary hypertension, or RV-specific cardiomyopathies. Improved survival of the aforementioned pathologies has led to increasing numbers of patients suffering from RV dysfunction, making it a key contributor to morbidity and mortality in this population. Currently available therapies for heart failure were developed for the left ventricle (LV), and there is clear evidence that LV-specific strategies are insufficient or inadequate for the RV. New therapeutic strategies are needed to address this growing clinical problem, and stem cells show significant promise. However, to properly evaluate the prospects of a potential stem cell-based therapy for RV failure, one needs to understand the unique pathophysiology of RV dysfunction and carefully consider available data from animal models and human clinical trials. In this review, we provide a comprehensive overview of the molecular mechanisms involved in RV failure such as hypertrophy, fibrosis, inflammation, changes in energy metabolism, calcium handling, decreasing RV contractility, and apoptosis. We also summarize the available preclinical and clinical experience with RV-specific stem cell therapies, covering the broad spectrum of stem cell sources used to date. We describe two different scientific rationales for stem cell transplantation, one of which seeks to add contractile units to the failing myocardium, while the other aims to augment endogenous repair mechanisms and/or attenuate harmful remodeling. We emphasize the limitations and challenges of regenerative strategies, but also highlight the characteristics of the failing RV myocardium that make it a promising target for stem cell therapy.
Collapse
Affiliation(s)
- Christoph Haller
- Division of Cardiovascular Surgery, The Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,McEwen Stem Cell Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Canada
| | - Mark K Friedberg
- Division of Cardiology, The Labatt Family Heart Centre, The Hospital for Sick Children, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, Peter Munk Cardiac Centre, University Health Network, Toronto, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada. .,McEwen Stem Cell Institute, Toronto Medical Discovery Tower, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.
| |
Collapse
|
8
|
Mount S, Kanda P, Parent S, Khan S, Michie C, Davila L, Chan V, Davies RA, Haddad H, Courtman D, Stewart DJ, Davis DR. Physiologic expansion of human heart-derived cells enhances therapeutic repair of injured myocardium. Stem Cell Res Ther 2019; 10:316. [PMID: 31685023 PMCID: PMC6829847 DOI: 10.1186/s13287-019-1418-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/03/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
Background Serum-free xenogen-free defined media and continuous controlled physiological cell culture conditions have been developed for stem cell therapeutics, but the effect of these conditions on the relative potency of the cell product is unknown. As such, we conducted a head-to-head comparison of cell culture conditions on human heart explant-derived cells using established in vitro measures of cell potency and in vivo functional repair. Methods Heart explant-derived cells cultured from human atrial or ventricular biopsies within a serum-free xenogen-free media and a continuous physiological culture environment were compared to cells cultured under traditional (high serum) cell culture conditions in a standard clean room facility. Results Transitioning from traditional high serum cell culture conditions to serum-free xenogen-free conditions had no effect on cell culture yields but provided a smaller, more homogenous, cell product with only minor antigenic changes. Culture within continuous physiologic conditions markedly boosted cell proliferation while increasing the expression of stem cell-related antigens and ability of cells to stimulate angiogenesis. Intramyocardial injection of physiologic cultured cells into immunodeficient mice 1 week after coronary ligation translated into improved cardiac function and reduced scar burden which was attributable to increased production of pro-healing cytokines, extracellular vesicles, and microRNAs. Conclusions Continuous physiological cell culture increased cell growth, paracrine output, and treatment outcomes to provide the greatest functional benefit after experimental myocardial infarction.
Collapse
Affiliation(s)
- Seth Mount
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada
| | - Pushpinder Kanda
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada
| | - Sandrine Parent
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada
| | - Saad Khan
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada
| | - Connor Michie
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada
| | - Liliana Davila
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada
| | - Vincent Chan
- University of Ottawa Heart Institute, Division of Cardiac Surgery, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Ross A Davies
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada
| | | | - David Courtman
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada
| | - Duncan J Stewart
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H8M5, Canada.,Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, K1H8L6, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, H3214 40 Ruskin Ave, Ottawa, ON, K1Y4W7, Canada. .,Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, K1H8L6, Canada.
| |
Collapse
|
9
|
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
|
10
|
Xu J, Lian W, Li L, Huang Z. Generation of induced cardiac progenitor cells via somatic reprogramming. Oncotarget 2018; 8:29442-29457. [PMID: 28199972 PMCID: PMC5438743 DOI: 10.18632/oncotarget.15272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/24/2017] [Indexed: 12/15/2022] Open
Abstract
It has been demonstrated that cardiac progenitor cells (CPCs) represent a more effective cell-based therapy for treatment of myocardial infarction. Unfortunately, their therapeutic application is limited by low yield of cell harvesting, declining quality and quantity during the ageing process, and the need for highly invasive heart biopsy. Therefore, there is an emerging interest in generating CPC-like stem cells from somatic cells via somatic reprogramming. This novel approach would provide an unlimited source of stem cells with cardiac differentiation potential. Here we would firstly discuss the different types of CPC and their importance in stem cell therapy for treatment of myocardial infarction; secondly, the necessity of generating induced CPC from somatic cells via somatic reprogramming; and finally the current progress of somatic reprogramming in cardiac cells, especially induced CPC generation.
Collapse
Affiliation(s)
- Jianyong Xu
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| | - Wei Lian
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| | - Lingyun Li
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| | - Zhong Huang
- Institute of Biological Therapy, Shenzhen University, Shenzhen, China.,Department of Pathogen Biology and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Shenzhen City Shenzhen University Immunodiagnostic Technology Platform, Shenzhen, China
| |
Collapse
|
11
|
Kasai-Brunswick TH, Costa ARD, Barbosa RAQ, Farjun B, Mesquita FCP, Silva dos Santos D, Ramos IP, Suhett G, Brasil GV, Cunha STD, Brito JOR, Passipieri JDA, Carvalho AB, Campos de Carvalho AC. Cardiosphere-derived cells do not improve cardiac function in rats with cardiac failure. Stem Cell Res Ther 2017; 8:36. [PMID: 28202059 PMCID: PMC5312520 DOI: 10.1186/s13287-017-0481-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Heart failure represents an important public health issue due to its high costs and growing incidence worldwide. Evidence showing the regenerative potential of postmitotic heart tissue has suggested the existence of endogenous cardiac stem cells in adult hearts. Cardiosphere-derived cells (CDC) constitute a candidate pool of such cardiac stem cells. Previous studies using acute myocardial infarction (MI) models in rodents demonstrated an improvement in cardiac function after cell therapy with CDC. We evaluated the therapeutic potential of CDC 60 days after MI in a rat model. METHODS CDC were obtained from human discarded myocardial tissue and rat hearts by enzymatic digestion with collagenase II. At 10-15 days after isolation, small, round, phase-bright cells (PBCs) appeared on top of the adherent fibroblast-like cells. The PBCs were collected and placed on a nonadherent plate for 2 days, where they formed cardiospheres which were then transferred to adherent plates, giving rise to CDC. These CDC were characterized by flow cytometry. Wistar rats were submitted to MI through permanent occlusion of the anterior descending coronary artery. After 60 days, they were immunosuppressed with cyclosporine A during 10 days. On the third day, infarcted animals were treated with 5 × 105 human CDC (hCDC) or placebo through intramyocardial injection guided by echocardiogram. Another group of animals was treated with rat CDC (rCDC) without immunosuppression. hCDC and rCDC were stably transduced with a viral construct expressing luciferase under control of a constitutive promoter. CDC were then used in a bioluminescence assay. Functional parameters were evaluated by echocardiogram 90 and 120 days after MI and by Langendorff at 120 days. RESULTS CDC had a predominantly mesenchymal phenotype. Cell tracking by bioluminescence demonstrated over 85% decrease in signal at 5-7 days after cell therapy. Cardiac function evaluation by echocardiography showed no differences in ejection fraction, end-diastolic volume, or end-systolic volume between groups receiving human cells, rat cells, or placebo. Hemodynamic analyses and infarct area quantification confirmed that there was no improvement in cardiac remodeling after cell therapy with CDC. CONCLUSION Our study challenges the effectiveness of CDC in post-ischemic heart failure.
Collapse
Affiliation(s)
- Taís Hanae Kasai-Brunswick
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
- 0000 0004 0481 7106grid.414444.5National Institute of Cardiology, Rua das Laranjeiras, n°374—Laranjeiras, CEP:22240-006 Rio de Janeiro, RJ Brazil
- 0000 0001 2294 473Xgrid.8536.8National Center for Structural Biology and Bioimaging—CENABIO, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, buiding M, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Andréa Rodrigues da Costa
- 0000 0004 0481 7106grid.414444.5National Institute of Cardiology, Rua das Laranjeiras, n°374—Laranjeiras, CEP:22240-006 Rio de Janeiro, RJ Brazil
| | - Raiana Andrade Quintanilha Barbosa
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
- 0000 0004 0481 7106grid.414444.5National Institute of Cardiology, Rua das Laranjeiras, n°374—Laranjeiras, CEP:22240-006 Rio de Janeiro, RJ Brazil
| | - Bruna Farjun
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Fernanda Cristina Paccola Mesquita
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Danúbia Silva dos Santos
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Isalira Peroba Ramos
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
- 0000 0001 2294 473Xgrid.8536.8National Center for Structural Biology and Bioimaging—CENABIO, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, buiding M, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Grazielle Suhett
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Guilherme Visconde Brasil
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Sandro Torrentes da Cunha
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - José Oscar R. Brito
- 0000 0004 0481 7106grid.414444.5National Institute of Cardiology, Rua das Laranjeiras, n°374—Laranjeiras, CEP:22240-006 Rio de Janeiro, RJ Brazil
| | - Juliana do Amaral Passipieri
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
- 0000 0004 0481 7106grid.414444.5National Institute of Cardiology, Rua das Laranjeiras, n°374—Laranjeiras, CEP:22240-006 Rio de Janeiro, RJ Brazil
| | - Adriana Bastos Carvalho
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
- National Institute of Science and Technology for Regenerative Medicine, Av. Carlos Chagas Filho, n°373, CEP:21941-902 Rio de Janeiro, RJ Brazil
| | - Antonio Carlos Campos de Carvalho
- 0000 0001 2294 473Xgrid.8536.8Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, room G2-053, CEP:21941-902 Rio de Janeiro, RJ Brazil
- 0000 0004 0481 7106grid.414444.5National Institute of Cardiology, Rua das Laranjeiras, n°374—Laranjeiras, CEP:22240-006 Rio de Janeiro, RJ Brazil
- 0000 0001 2294 473Xgrid.8536.8National Center for Structural Biology and Bioimaging—CENABIO, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, n°373, buiding M, CEP:21941-902 Rio de Janeiro, RJ Brazil
- National Institute of Science and Technology for Regenerative Medicine, Av. Carlos Chagas Filho, n°373, CEP:21941-902 Rio de Janeiro, RJ Brazil
| |
Collapse
|
12
|
Hao M, Wang R, Wang W. Cell Therapies in Cardiomyopathy: Current Status of Clinical Trials. Anal Cell Pathol (Amst) 2017; 2017:9404057. [PMID: 28194324 PMCID: PMC5282433 DOI: 10.1155/2017/9404057] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/06/2016] [Accepted: 12/08/2016] [Indexed: 12/28/2022] Open
Abstract
Because the human heart has limited potential for regeneration, the loss of cardiomyocytes during cardiac myopathy and ischaemic injury can result in heart failure and death. Stem cell therapy has emerged as a promising strategy for the treatment of dead myocardium, directly or indirectly, and seems to offer functional benefits to patients. The ideal candidate donor cell for myocardial reconstitution is a stem-like cell that can be easily obtained, has a robust proliferation capacity and a low risk of tumour formation and immune rejection, differentiates into functionally normal cardiomyocytes, and is suitable for minimally invasive clinical transplantation. The ultimate goal of cardiac repair is to regenerate functionally viable myocardium after myocardial infarction (MI) to prevent or heal heart failure. This review provides a comprehensive overview of treatment with stem-like cells in preclinical and clinical studies to assess the feasibility and efficacy of this novel therapeutic strategy in ischaemic cardiomyopathy.
Collapse
Affiliation(s)
- Ming Hao
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai 200233, China
- Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA 95014, USA
| | - Richard Wang
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai 200233, China
- Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA 95014, USA
| | - Wen Wang
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai 200233, China
- Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA 95014, USA
| |
Collapse
|
13
|
Le TYL, Thavapalachandran S, Kizana E, Chong JJ. New Developments in Cardiac Regeneration. Heart Lung Circ 2016; 26:316-322. [PMID: 27916592 DOI: 10.1016/j.hlc.2016.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 01/13/2023]
Abstract
Numerous pharmacological and device therapies have improved adverse cardiac remodelling and mortality in heart failure. However, none are able to regenerate damaged cardiac tissue. Stem cell based therapies using multipotent (adult) stem cells and pluripotent stem cells are new approaches that could potentially achieve the elusive goal of true cardiac regeneration. Over the past two decades, various stem cell based approaches have been shown to improve left ventricular function in pre-clinical animal models. Promising results rapidly led to clinical trials, initially using bone marrow-derived mononuclear cells, then mesenchymal stromal cell populations and, more recently, progenitor cells from the adult heart itself. These have been shown to be safe and have advanced our understanding of potential suitable recipients, cell delivery routes, and possible mechanisms of action. However, efficacy in these trials has been inconsistent. Human pluripotent stem cells (hPSCs) are another potential source of stem cells for cardiac regeneration. They could theoretically provide an unlimited source of cardiomyocytes or cardiac progenitors. Pre-clinical studies in both small and large animal models have shown robust engraftment and improvements in cardiac function. The first clinical trial using hPSC-derived cardiac derivatives has now commenced and others are imminent. In this brief review article, we summarise recent developments in stem cell therapies aimed at cardiac regeneration, including discussion of types of cell and non-cell-based strategies being explored.
Collapse
Affiliation(s)
- Thi Yen Loan Le
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Sujitha Thavapalachandran
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - James Jh Chong
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
14
|
Andersen DC, Jensen CH, Skovrind I, Johnsen RH, Traustadottir GA, Aagaard KS, Ganesalingam S, Sheikh SP. Neonatal epicardial-derived progenitors aquire myogenic traits in skeletal muscle, but not cardiac muscle. Int J Cardiol 2016; 222:448-456. [PMID: 27505332 DOI: 10.1016/j.ijcard.2016.07.165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 07/27/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND/OBJECTIVES Epicardium-derived progenitor cells (EPDCs) differentiate into all heart cell types in the embryonic heart, yet their differentiation into cardiomyocytes in the adult heart is limited and poorly described. This may be due to EPDCs lacking myogenic potential or the inert adult heart missing regenerative signals essential for directed differentiation of EPDCs. Herein, we aimed to evaluate the myogenic potential of neonatal EPDCs in adult and neonatal mouse myocardium, as well as in skeletal muscle. The two latter tissues have an intrinsic capability to develop and regenerate, in contrast to the adult heart. METHODS Highly purified mouse EPDCs were transplanted into damaged neonatal and adult myocardium as well as regenerating skeletal muscle. Co-cultures with skeletal myoblasts were used to distinguish fusion independent myogenic conversion. RESULTS No donor EPDC-derived cardiomyocytes were observed in hearts. In contrast, a remarkable contribution of EPDCs to skeletal muscle myofiber formation was evident in vivo. Furthermore, co-cultures of EPDCs with myoblasts showed that EPDCs became part of multinucleated fibers and appeared to acquire myogenic traits independent of a fusion event. Fluorescence activated cell sorting of EPDCs co-cultured with and without myoblasts and subsequent qRT-PCR of 64 transcripts established that the myogenic phenotype conversion was accomplished through induction of a transcriptional myogenic program. CONCLUSION These results suggest that EPDCs may be more myogenic than previously anticipated. But, the heart may lack factors for induction of myogenesis of EPDCs, a scenario that should be taken into consideration when aiming for repair of damaged myocardium by stem cell transplantation.
Collapse
Affiliation(s)
- Ditte C Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark.
| | - Charlotte H Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Ida Skovrind
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Rikke Helin Johnsen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Clinical Institute, University of Southern Denmark, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Gunnhildur Asta Traustadottir
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Katrine S Aagaard
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Suganya Ganesalingam
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| | - Søren P Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark; Cardiovascular and Renal Research, University of Southern Denmark, Winsloewparken 21(3rd), 5000 Odense, Denmark; The Danish Regenerative Center (danishcrm.com), Odense University Hospital, Sdr. Boulevard 29, 5000 Odense, Denmark
| |
Collapse
|
15
|
Dergilev KV, Tsokolaeva ZI, Rubina KA, Sysoeva VY, Makarevich PI, Boldyreva MA, Beloglazova IB, Zubkova ES, Sharonov GV, Akchurin RS, Parfyonova YV. Isolation and characterization of cardiac progenitor cells from myocardial right atrial appendage tissue. ACTA ACUST UNITED AC 2016. [DOI: 10.1134/s1990519x16050035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Cardiac atrial appendage stem cells engraft and differentiate into cardiomyocytes in vivo: A new tool for cardiac repair after MI. Int J Cardiol 2015; 201:10-9. [DOI: 10.1016/j.ijcard.2015.07.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/16/2015] [Accepted: 07/26/2015] [Indexed: 12/22/2022]
|
17
|
Fanton Y, Robic B, Rummens JL, Daniëls A, Windmolders S, Willems L, Jamaer L, Dubois J, Bijnens E, Heuts N, Notelaers K, Paesen R, Ameloot M, Mees U, Bito V, Declercq J, Hensen K, Koninckx R, Hendrikx M. Possibilities and limitations for co-transplantation of cardiac atrial appendage stem cells and mesenchymal stem cells for myocardial repair. Int J Cardiol 2015; 203:1155-6. [PMID: 26549562 DOI: 10.1016/j.ijcard.2015.10.141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/18/2015] [Indexed: 11/19/2022]
Affiliation(s)
- Yanick Fanton
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
| | - Boris Robic
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| | - Jean-Luc Rummens
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Annick Daniëls
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium
| | - Severina Windmolders
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Leen Willems
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Luc Jamaer
- Department of Cardiac Anesthesia, Jessa Hospital, Hasselt, Belgium
| | - Jasperina Dubois
- Department of Cardiac Anesthesia, Jessa Hospital, Hasselt, Belgium
| | - Eric Bijnens
- MRI Unit-Department of Radiology, Jessa Hospital, Hasselt, Belgium
| | - Nic Heuts
- MRI Unit-Department of Radiology, Jessa Hospital, Hasselt, Belgium
| | - Kristof Notelaers
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Rik Paesen
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Marcel Ameloot
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Urbain Mees
- Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| | - Virginie Bito
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Jeroen Declercq
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Karen Hensen
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Remco Koninckx
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| |
Collapse
|
18
|
Kshitiz, Afzal J, Kim DH, Levchenko A. Concise review: Mechanotransduction via p190RhoGAP regulates a switch between cardiomyogenic and endothelial lineages in adult cardiac progenitors. Stem Cells 2015; 32:1999-2007. [PMID: 24710857 DOI: 10.1002/stem.1700] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/18/2014] [Indexed: 01/01/2023]
Abstract
Mechanical cues can have pleiotropic influence on stem cell shape, proliferation, differentiation, and morphogenesis, and are increasingly realized to play an instructive role in regeneration and maintenance of tissue structure and functions. To explore the putative effects of mechanical cues in regeneration of the cardiac tissue, we investigated therapeutically important cardiosphere-derived cells (CDCs), a heterogeneous patient- or animal-specific cell population containing c-Kit(+) multipotent stem cells. We showed that mechanical cues can instruct c-Kit(+) cell differentiation along two lineages with corresponding morphogenic changes, while also serving to amplify the initial c-Kit(+) subpopulation. In particular, mechanical cues mimicking the structure of myocardial extracellular matrix specify cardiomyogenic fate, while cues mimicking myocardium rigidity specify endothelial fates. Furthermore, we found that these cues dynamically regulate the same molecular species, p190RhoGAP, which then acts through both RhoA-dependent and independent mechanisms. Thus, differential regulation of p190RhoGAP molecule by either mechanical inputs or genetic manipulation can determine lineage type specification. Since human CDCs are already in phase II clinical trials, the potential therapeutic use of mechanical or genetic manipulation of the cell fate could enhance effectiveness of these progenitor cells in cardiac repair, and shed new light on differentiation mechanisms in cardiac and other tissues.
Collapse
Affiliation(s)
- Kshitiz
- Department of Bioengineering, Institute of Stem Cells and Regenerative Medicine and Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA; Institute of Stem Cells and Regenerative Medicine and Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| | | | | | | |
Collapse
|
19
|
Ghazizadeh Z, Vahdat S, Fattahi F, Fonoudi H, Omrani G, Gholampour M, Aghdami N. Isolation and characterization of cardiogenic, stem-like cardiac precursors from heart samples of patients with congenital heart disease. Life Sci 2015; 137:105-15. [PMID: 26165749 DOI: 10.1016/j.lfs.2015.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 06/04/2015] [Accepted: 07/02/2015] [Indexed: 01/14/2023]
Abstract
AIMS Regenerative therapies based on resident human cardiac progenitor cells (hCPCs) are a promising alternative to medical treatments for patients with myocardial infarction. However, hCPCs are rare in human heart and finding efficient source and proper surface marker for isolation of these cells would make them a good candidate for therapy. MAIN METHODS We have isolated 5.34∗10(6)±2.04∗10(5)/g viable cells from 35 heart tissue samples of 23 patients with congenital heart disease obtained during their heart surgery along with 6 samples from 3 normal subjects during cardiac biopsy. KEY FINDINGS According to FACS analysis, younger ages, atrial specimen and disease with increased pulmonary vascular resistance were associated with higher percentage of c-kit(+) (CD117) hCPCs. Analysis for other stemness markers revealed increased CD133(+) cells in the hearts of patients with congenital heart disease. By using both immune-labeling and PCR, we demonstrated that these cells express key cardiac lineage and endothelial transcription factors and structural proteins during in vitro differentiation and do express stemness transcription factors in undifferentiated state. Another novel datum of potentially relevant interest is their ability in promoting greater myocardial regeneration and better survival in rat model of myocardial infarction following transplantation. SIGNIFICANCE Our results could provide evidence for conditions associated with enriched hCPCs in patients with congenital heart disease. Moreover, we showed presence of a significant number of CD133 expressing cardiogenic stem-like cardiac precursors in the heart of patients with congenital heart disease, which could be isolated and stored for future regenerative therapies in these patients.
Collapse
Affiliation(s)
- Zaniar Ghazizadeh
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadaf Vahdat
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faranak Fattahi
- Department of Molecular Systems Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hananeh Fonoudi
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Gholamreza Omrani
- Department of Cardiac Surgery, Rajaei Cardiovascular Medical Research Center, Tehran, Iran
| | - Maziar Gholampour
- Department of Cardiac Surgery, Rajaei Cardiovascular Medical Research Center, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Biomedicine at the Cell Science Research, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
20
|
A simplified protocol for the isolation and culture of cardiomyocytes and progenitor cells from neonatal mouse ventricles. Eur J Cell Biol 2015; 94:444-52. [PMID: 26153430 DOI: 10.1016/j.ejcb.2015.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 01/04/2023] Open
Abstract
The neonatal heart is a very useful tool for the study of biochemical pathways and properties of cardiomyocytes and as it has the potential to regenerate for a brief period of time from birth; it is also useful to study cardiac regeneration. However, as the heart matures, this proficiency for regeneration is reduced. This regenerative potential may be influenced by the microenvironment of the heart in the early stages of postnatal development and therefore, cell cultures derived at this stage may contain functional cardiomyocytes and progenitor cells. The aim of this study was to identify key steps in the isolation and culture of such early stage-neonatal mouse hearts to allow maximum migration of cardiomyocytes from the explant and their maintenance as functional, long term cultures. Explant cultures of mouse ventricles preserved 3-dimensional structure and generated migrating layers of cardiomyocytes that expressed alpha sarcomeric actin which could be further sub-cultured by enzymatic dissociation. Western blotting demonstrated expression of c-KIT, GATA4, alpha sarcomeric actin and connexin43 proteins after 20 days of explant culture. ACTA1, GATA4, and CX43 continued to express in five weeks old explant cultures while the c-KIT protein was expressed up to two passages during sub-culture. Real time PCR and SQRT PCR also demonstrated gene expression of cardiomyocyte markers in long term cultures. Migrating cells from the explants assembled into contracting spheroids after subculture and expressed the c-KIT protein. Progenitor markers CD44, CD90, and extracellular proteins, periostin and vimentin demonstrated the preservation of cellular heterogeneity in such cultures. Supplementation with Hydrocortisone maintained a cardioprotective environment and reduced the non-myocyte population. This is an optimized and efficient method for the generation of neonatal heart cultures that is not labor intensive and does not require supplementation with cytokines.
Collapse
|
21
|
Kim JT, Chung HJ, Seo JY, Yang YI, Choi MY, Kim HI, Yang TH, Lee WJ, Youn YC, Kim HJ, Kim YM, Lee H, Jang YS, Lee SJ. A fibrin-supported myocardial organ culture for isolation of cardiac stem cells via the recapitulation of cardiac homeostasis. Biomaterials 2015; 48:66-83. [DOI: 10.1016/j.biomaterials.2015.01.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/11/2015] [Accepted: 01/20/2015] [Indexed: 12/22/2022]
|
22
|
Ge Z, Lal S, Le TYL, Dos Remedios C, Chong JJH. Cardiac stem cells: translation to human studies. Biophys Rev 2014; 7:127-139. [PMID: 28509972 DOI: 10.1007/s12551-014-0148-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 02/08/2023] Open
Abstract
The discovery of multiple classes of cardiac progenitor cells in the adult mammalian heart has generated hope for their use as a therapeutic in heart failure. However, successful results from animal models have not always yielded similar findings in human studies. Recent Phase I/II trials of c-Kit (SCIPIO) and cardiosphere-based (CADUCEUS) cardiac progenitor cells have demonstrated safety and some therapeutic efficacy. Gaps remain in our understanding of the origins, function and relationships between the different progenitor cell families, many of which are heterogeneous populations with overlapping definitions. Another challenge lies in the limitations of small animal models in replicating the human heart. Cryopreserved human cardiac tissue provides a readily available source of cardiac progenitor cells and may help address these questions. We review important findings and relative unknowns of the main classes of cardiac progenitor cells, highlighting differences between animal and human studies.
Collapse
Affiliation(s)
- Zijun Ge
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Sean Lal
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Thi Y L Le
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia.,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145
| | | | - James J H Chong
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW, Australia. .,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145.
| |
Collapse
|
23
|
Sepúlveda DE, Cabeza Meckert P, Locatelli P, Olea FD, Pérez NG, Pinilla OA, Díaz RG, Crottogini A, Laguens RP. Activated macrophages as a feeder layer for growth of resident cardiac progenitor cells. Cytotechnology 2014; 68:665-74. [PMID: 25432330 DOI: 10.1007/s10616-014-9814-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 11/03/2014] [Indexed: 01/05/2023] Open
Abstract
The adult heart contains a population of cardiac progenitor cells (CPCs). Growing and collecting an adequate number of CPCs demands complex culture media containing growth factors. Since activated macrophages secrete many growth factors, we investigated if activated isolated heart cells seeded on a feeder layer of activated peritoneal macrophages (PM) could result in CPCs and if these, in turn, could exert cardioprotection in rats with myocardial infarction (MI). Heart cells of inbred Wistar rats were isolated by collagenase digestion and cultured on PM obtained 72 h after intraperitoneal injection of 12 ml thioglycollate. Cells (1 × 10(6)) exhibiting CPC phenotype (immunohistochemistry) were injected in the periphery of rat MI 10 min after coronary artery occlusion. Control rats received vehicle. Three weeks later, left ventricular (LV) function (echocardiogram) was assessed, animals were euthanized and the hearts removed for histological studies. Five to six days after seeding heart cells on PM, spherical clusters composed of small bright and spherical cells expressing mostly c-Kit and Sca-1 antigens were apparent. After explant, those clusters developed cobblestone-like monolayers that expressed smooth muscle actin and sarcomeric actin and were successfully transferred for more than ten passages. When injected in the MI periphery, many of them survived at 21 days after coronary ligature, improved LV ejection fraction and decreased scar size as compared with control rats. CPC-derived cells with cardiocyte and smooth muscle phenotypes can be successfully grown on a feeder layer of activated syngeneic PM. These cells decreased scar size and improved heart function in rats with MI.
Collapse
Affiliation(s)
- Diana E Sepúlveda
- Department of Pathology, Universidad Favaloro, Solís 453, Buenos Aires, Argentina
| | | | - Paola Locatelli
- Department of Physiology, Universidad Favaloro, Solís 453, Buenos Aires, Argentina
| | - Fernanda D Olea
- Department of Physiology, Universidad Favaloro, Solís 453, Buenos Aires, Argentina
| | - Néstor G Pérez
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, 60 y 120, La Plata, Argentina
| | - Oscar A Pinilla
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, 60 y 120, La Plata, Argentina
| | - Romina G Díaz
- Facultad de Ciencias Médicas, Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, 60 y 120, La Plata, Argentina
| | - Alberto Crottogini
- Department of Physiology, Universidad Favaloro, Solís 453, Buenos Aires, Argentina
| | - Rubén P Laguens
- Department of Pathology, Universidad Favaloro, Solís 453, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Cho GS, Fernandez L, Kwon C. Regenerative medicine for the heart: perspectives on stem-cell therapy. Antioxid Redox Signal 2014; 21:2018-31. [PMID: 25133793 PMCID: PMC4208610 DOI: 10.1089/ars.2014.6063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Despite decades of progress in cardiovascular biology and medicine, heart disease remains the leading cause of death, and there is no cure for the failing heart. Since heart failure is mostly caused by loss or dysfunction of cardiomyocytes (CMs), replacing dead or damaged CMs with new CMs might be an ideal way to reverse the disease. However, the adult heart is composed mainly of terminally differentiated CMs that have no significant self-regeneration capacity. RECENT ADVANCES Stem cells have tremendous regenerative potential and, thus, current cardiac regenerative research has focused on developing stem cell sources to repair damaged myocardium. CRITICAL ISSUES In this review, we examine the potential sources of cells that could be used for heart therapies, including embryonic stem cells and induced pluripotent stem cells, as well as alternative methods for activating the endogenous regenerative mechanisms of the heart via transdifferentiation and cell reprogramming. We also discuss the current state of knowledge of cell purification, delivery, and retention. FUTURE DIRECTIONS Efforts are underway to improve the current stem cell strategies and methodologies, which will accelerate the development of innovative stem-cell therapies for heart regeneration.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Johns Hopkins University , Baltimore, Maryland
| | | | | |
Collapse
|
25
|
Chong JJ, Forte E, Harvey RP. Developmental origins and lineage descendants of endogenous adult cardiac progenitor cells. Stem Cell Res 2014; 13:592-614. [DOI: 10.1016/j.scr.2014.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/24/2014] [Accepted: 09/26/2014] [Indexed: 12/30/2022] Open
|
26
|
Zhang H, Wang H, Li N, Duan CE, Yang YJ. Cardiac progenitor/stem cells on myocardial infarction or ischemic heart disease: what we have known from current research. Heart Fail Rev 2014; 19:247-58. [PMID: 23381197 DOI: 10.1007/s10741-013-9372-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell therapy has become a promising method for many diseases, including ischemic heart disease and heart failure. Several kinds of stem cells have been studied for heart diseases. Of them, bone marrow stem cells (BMSCs), which have been used in many clinical trials, are the most understood one. But the effect of BMSCs is mediated by paracrine factors instead of direct turning into cardiomyocytes. On the other hand, a lot of evidences have shown that resident cardiac stem cells could turn into cardiomyocytes directly in vivo. Currently, seven kinds of resident cardiac stem cells have been discovered. However, their mechanisms, development origins, and relationships have yet to be fully understood. Moreover, two Phase I clinical trials have been performed recently. They show promising results. In this review, we will summarize the current research on these cardiac stem cells and the methods to enhance their effects in clinical applications.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Translational Cardiovascular Medicine, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | | | | | | | | |
Collapse
|
27
|
Abstract
Various stem cell types have been tested for regenerating damaged myocardium after myocardial infarction. However, the results of clinical trials have not been consistent, with only some of the trials reporting small improvements in cardiac function. It seems that engraftment and survival of injected cells is limited and transplanted stem cells either do not differentiate into cardiac cells or differentiate into only limited number of cardiac cells. The exact mechanism(s) of cardiac functional improvement by cell therapy are unclear, but paracrine effect may play a central role. The resident cardiac progenitor cells identified within the adult myocardium have distinct advantages over other stem cell types for cardiac cell therapy, as they are likely precommitted to the cardiovascular fate. However, isolating and expanding these cells from cardiac biopsies is a challenge. More recently, direct reprogramming of fibroblasts into cardiomyocytes has given new hope for myocardial regeneration. Here we will review different stem cells used in cardiac cell therapy with a focus on the native cardiac progenitor cells and briefly outline future directions of cardiac cell therapy.
Collapse
|
28
|
Maxeiner H, Mufti S, Krehbiehl N, Dülfer F, Helmig S, Schneider J, Böning A, Matejec R, Weigand MA, Schlüter KD, Wenzel S. Interleukin-6 contributes to the paracrine effects of cardiospheres cultured from human, murine and rat hearts. J Cell Physiol 2014; 229:1681-9. [PMID: 24623002 DOI: 10.1002/jcp.24613] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 03/11/2014] [Indexed: 12/12/2022]
Abstract
Cardiosphere-derived cells (CDCs) were cultured from human, murine, and rat hearts. Diluted supernatant (conditioned-medium) of the cultures improved the contractile behavior of isolated rat cardiomyocytes (CMCs). This effect is mediated by the paracrine release of cytokines. The present study tested the hypothesis, that the cardiovascular state of the donor's heart influences this effect on CMCs and tries to identify the responsible factors. CDCs were cultured from human tissue samples of cardiac surgery and from murine and rat hearts. The supernatants of cultured CDCs from hypertensive humans and rats showed a higher improvement of the contractile behavior of CMCs compared to CDCs of normotensive origin. Subsequently, the cytokine profile of the supernatants was analyzed. Among the cytokines elevated in supernatants originating from hypertensive humans or rats was Interleukin-6. CDCs were also generated from Interleukin-6(-/-) -mice and their wildtype littermates. The supernatant of the cultured Interleukin-6(-/-) -CDCs had no effect on the contractile behavior, whereas the supernatant of the Interleukin-6(+/+) -CDCs showed a positive effect. To confirm the hypothesis that Interleukin-6 contributes to the paracrine effects, CMCs were incubated with Interleukin-6. It improved the contractile function in a concentration dependent way. Finally, the effect of the supernatant of cultured CDCs derived from a hypertensive human sample could be abolished by simultaneous incubation with a specific Interleukin-6 antibody. CDCs release cytokines that improve the contractile behavior of CMCs. This effect is more intense in CDCs from hypertensive donors. Interleukin-6 is involved in this phenomenon.
Collapse
Affiliation(s)
- Hagen Maxeiner
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Giessen and Marburg, Campus Giessen, Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Matar AA, Chong JJ. Stem cell therapy for cardiac dysfunction. SPRINGERPLUS 2014; 3:440. [PMID: 25191634 PMCID: PMC4153875 DOI: 10.1186/2193-1801-3-440] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/11/2014] [Indexed: 01/05/2023]
Abstract
Following significant injury, the heart undergoes induced compensation and gradually deteriorates towards impending heart failure. Current therapy slows but does not halt the resultant adverse remodeling. Stem cell therapy, however, has the potential to regenerate or repair infarcted heart tissue and therefore is a promising therapeutic strategy undergoing intensive investigation. Due to the wide range of stem cells investigated, it is difficult to navigate this field. This review aims to summarize the main types of stem cells (both of cardiac and extra-cardiac origin) that possess promising therapeutic potential. Particular focus is placed on clinical trials supporting this therapeutic strategy.
Collapse
Affiliation(s)
- Amer A Matar
- Sydney Medical School, University of Sydney, Sydney, NSW Australia
| | - James Jh Chong
- Sydney Medical School, University of Sydney, Sydney, NSW Australia ; Department of Cardiology, Westmead Hospital, Sydney, NSW Australia ; Centre for Heart Research, Westmead Millennium Institute, Sydney, NSW Australia
| |
Collapse
|
30
|
Wang H, Chen H, Feng B, Wang X, He X, Hu R, Yin M, Wang W, Fu W, Xu Z. Isolation and characterization of a Sca-1+/CD31- progenitor cell lineage derived from mouse heart tissue. BMC Biotechnol 2014; 14:75. [PMID: 25106452 PMCID: PMC4133720 DOI: 10.1186/1472-6750-14-75] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 08/04/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Myocardial infarction remains the leading cause of mortality in developed countries despite recent advances in its prevention and treatment. Regenerative therapies based on resident cardiac progenitor cells (CPCs) are a promising alternative to conventional treatments. However, CPCs resident in the heart are quite rare. It is unclear how these CPCs can be isolated and cultured efficiently and what the effects of long-term culture in vitro are on their 'stemness' and differentiation potential, but this is critical knowledge for CPCs' clinical application. RESULTS Here, we isolated stem cell antigen-1 positive cells from postnatal mouse heart by magnetic active cell sorting using an iron-labeled anti-mouse Sca-1 antibody, and cultured them long-term in vitro. We tested stemness marker expression and the proliferation ability of long-term cultured Sca-1+ cells at early, middle and late passages. Furthermore, we determined the differentiation potential of these three passages into cardiac cell lineages (cardiomyocytes, smooth muscle and endothelial cells) after induction in vitro. The expression of myocardial, smooth muscle and endothelial cell-specific genes and surface markers were analyzed by RT-PCR and IF staining. We also investigated the oncogenicity of the three passages by subcutaneously injecting cells in nude mice. Overall, heart-derived Sca-1+ cells showed CPC characteristics: long-term propagation ability in vitro, non-tumorigenic in vivo, persistent expression of stemness and cardiac-specific markers, and multipotent differentiation into cardiac cell lineages. CONCLUSIONS Our research may bring new insights to myocardium regeneration, for which even a small number of biopsy-derived CPCs could be enriched and propagated long term in vitro to obtain sufficient seed cells for cell injection or cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China.
| | | |
Collapse
|
31
|
Eskildsen TV, Schneider M, Sandberg MB, Skov V, Brønnum H, Thomassen M, Kruse TA, Andersen DC, Sheikh SP. The microRNA-132/212 family fine-tunes multiple targets in Angiotensin II signalling in cardiac fibroblasts. J Renin Angiotensin Aldosterone Syst 2014; 16:1288-97. [PMID: 25031299 DOI: 10.1177/1470320314539367] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION MicroRNAs (miRNAs) are emerging as key regulators of cardiovascular development and disease; however, the cardiac miRNA target molecules are not well understood. We and others have described the Angiotensin II (AngII)-induced miR-132/212 family as novel regulators of cardiovascular function including regulation of cardiac hypertrophy, heart failure and blood pressure possibly through AT1R signalling. However, the miR-132/212 targets in the heart remain unknown. MATERIALS AND METHODS To understand the role of these miRNAs in cardiac signalling networks, we undertook comprehensive in silico and in vitro experiments to identify miR-132/212 molecular targets in primary rat cardiac fibroblasts. RESULTS MiR-132/212 overexpression increased fibroblast cell size and mRNA arrays detected several hundred genes that were differentially expressed, including a wide panel of receptors, signalling molecules and transcription factors. Subsequent comprehensive in silico analysis identified 24 target genes, of which 22 genes were qPCR validated. We identified seven genes involved in AngII signalling pathways. CONCLUSION We here report novel insight of an extensive network of molecular pathways that fine-tuned by miR-132/212, suggesting a role for this miRNA family as master signalling switches in cardiac fibroblasts. Our data underscore the potential for miRNA tools to manipulate a large array of molecules and thereby control biological function.
Collapse
Affiliation(s)
- Tilde V Eskildsen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Denmark Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| | - Mikael Schneider
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| | - Maria B Sandberg
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| | - Vibe Skov
- Department of Clinical Genetics, Odense University Hospital, Denmark
| | - Hasse Brønnum
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Denmark Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Denmark
| | - Torben A Kruse
- Department of Clinical Genetics, Odense University Hospital, Denmark
| | - Ditte C Andersen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Denmark Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| | - Søren P Sheikh
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Denmark Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| |
Collapse
|
32
|
Sommer P. Can stem cells really regenerate the human heart? Use your noggin, dickkopf! Lessons from developmental biology. Cardiovasc J Afr 2014; 24:189-93. [PMID: 24217168 PMCID: PMC3748454 DOI: 10.5830/cvja-2013-045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/07/2013] [Indexed: 11/06/2022] Open
Abstract
The human heart is the first organ to develop and its development is fairly well characterised. In theory, the heart has the capacity to regenerate, as its cardiomyocytes may be capable of cell division and the adult heart contains a cardiac stem cell niche, presumably capable of differentiating into cardiomyocytes and other cardiac-associated cell types. However, as with most other organs, these mechanisms are not activated upon serious injury. Several experimental options to induce regeneration of the damaged heart tissue are available: activate the endogenous cardiomyocytes to divide, coax the endogenous population of stem cells to divide and differentiate, or add exogenous cell-based therapy to replace the lost cardiac tissue. This review is a summary of the recent research into all these avenues, discussing the reasons for the limited successes of clinical trials using stem cells after cardiac injury and explaining new advances in basic science. It concludes with a reiteration that chances of successful regeneration would be improved by understanding and implementing the basics of heart development and stem cell biology.
Collapse
Affiliation(s)
- Paula Sommer
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
33
|
Abstract
Human heart failure (HF) is one of the leading causes of morbidity and mortality worldwide. Currently, heart transplantation and implantation of mechanical devices represent the only available treatments for advanced HF. Two alternative strategies have emerged to treat patients with HF. One approach relies on transplantation of exogenous stem cells (SCs) of non-cardiac or cardiac origin to induce cardiac regeneration and improve ventricular function. Another complementary strategy relies on stimulation of the endogenous regenerative capacity of uninjured cardiac progenitor cells to rebuild cardiac muscle and restore ventricular function. Various SC types and delivery strategies have been examined in the experimental and clinical settings; however, neither the ideal cell type nor the cell delivery method for cardiac cell therapy has yet emerged. Although the use of bone marrow (BM)-derived cells, most frequently exploited in clinical trials, appears to be safe, the results are controversial. Two recent randomized trials have failed to document any beneficial effects of intracardiac delivery of autologous BM mononuclear cells on cardiac function of patients with HF. The remarkable discovery that various populations of cardiac progenitor cells (CPCs) are present in the adult human heart and that it possesses limited regeneration capacity has opened a new era in cardiac repair. Importantly, unlike BM-derived SCs, autologous CPCs from myocardial biopsies cultured and subsequently delivered by coronary injection to patients have given positive results. Although these data are promising, a better understanding of how to control proliferation and differentiation of CPCs, to enhance their recruitment and survival, is required before CPCs become clinically applicable therapeutics.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ, 08904, USA
| | | |
Collapse
|
34
|
Sluijter JPG, Condorelli G, Davidson SM, Engel FB, Ferdinandy P, Hausenloy DJ, Lecour S, Madonna R, Ovize M, Ruiz-Meana M, Schulz R, Van Laake LW. Novel therapeutic strategies for cardioprotection. Pharmacol Ther 2014; 144:60-70. [PMID: 24837132 DOI: 10.1016/j.pharmthera.2014.05.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/12/2022]
Abstract
The morbidity and mortality from ischemic heart disease (IHD) remain significant worldwide. The treatment for acute myocardial infarction has improved over the past decades, including early reperfusion of occluded coronary arteries. Although it is essential to re-open the artery as soon as possible, paradoxically this leads to additional myocardial injury, called acute ischemia-reperfusion injury (IRI), for which currently no effective therapy is available. Therefore, novel therapeutic strategies are required to protect the heart from acute IRI in order to reduce myocardial infarction size, preserve cardiac function and improve clinical outcomes in patients with IHD. In this review article, we will first outline the pathophysiology of acute IRI and review promising therapeutic strategies for cardioprotection. These include novel aspects of mitochondrial function, epigenetics, circadian clocks, the immune system, microvesicles, growth factors, stem cell therapy and gene therapy. We discuss the therapeutic potential of these novel cardioprotective strategies in terms of pharmacological targeting and clinical application.
Collapse
Affiliation(s)
- Joost P G Sluijter
- Department of Cardiology, University Medical Center Utrecht, The Netherlands; ICIN, Netherlands Heart Institute, Utrecht, The Netherlands
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Derek J Hausenloy
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, South Africa
| | - Rosalinda Madonna
- Department of Neurosciences and Imaging, Institute of Cardiology, University of Chieti, Chieti, Italy
| | - Michel Ovize
- Service d'Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, France; Inserm U1060-CarMeN, CIC de Lyon, Université Claude Bernard Lyon, Lyon, France
| | - Marisol Ruiz-Meana
- Laboratori Cardiologia, Vall d'Hebron Institut de Recerca, Universitat Autonoma de Barcelona, Spain
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig Universität, Gießen, Germany
| | - Linda W Van Laake
- Department of Cardiology, University Medical Center Utrecht, The Netherlands.
| | | |
Collapse
|
35
|
Xiong JW, Chang NN. Recent advances in heart regeneration. ACTA ACUST UNITED AC 2014; 99:160-9. [PMID: 24078494 DOI: 10.1002/bdrc.21039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/25/2022]
Abstract
Although cardiac stem cells (CSCs) and tissue engineering are very promising for cardiac regenerative medicine, studies with model organisms for heart regeneration will provide alternative therapeutic targets and opportunities. Here, we present a review on heart regeneration, with a particular focus on the most recent work in mouse and zebrafish. We attempt to summarize the recent progresses and bottlenecks of CSCs and tissue engineering for heart regeneration; and emphasize what we have learned from mouse and zebrafish regenerative models on discovering crucial genetic and epigenetic factors for stimulating heart regeneration; and speculate the potential application of these regenerative factors for heart failure. A brief perspective highlights several important and promising research directions in this exciting field.
Collapse
Affiliation(s)
- Jing-Wei Xiong
- are from Institute of Molecular Medicine, Peking University, Beijing, 100871, China and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | | |
Collapse
|
36
|
Ellison GM, Smith AJ, Waring CD, Henning BJ, Burdina AO, Polydorou J, Vicinanza C, Lewis FC, Nadal-Ginard B, Torella D. Adult Cardiac Stem Cells: Identity, Location and Potential. ADULT STEM CELLS 2014. [DOI: 10.1007/978-1-4614-9569-7_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Pavlova SV, Perovskii PP, Chepeleva EV, Malakhova AA, Dement’eva EV, Pokushalov EA, Sukhikh GT, Zakiyan SM. Characteristics of Cardiac Cell Cultures Derived from Human Myocardial Explants. Bull Exp Biol Med 2013; 156:127-35. [DOI: 10.1007/s10517-013-2295-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
38
|
Abstract
The heart consists of many types of cells, including cardiomyocytes, vascular cells, neural cells, and cardiac fibroblasts. Adult cardiomyocytes are terminally differentiated cells, and loss of cardiomyocytes as a result of heart damage is irreversible. To regenerate damaged hearts and restore cardiac function, understanding the cellular and molecular basis of heart development is of considerable importance. Although it is well known that heart function is tightly regulated by cell-cell interactions, their roles in heart development are not clear. Recent studies, including ours, identified important roles of cell-cell interactions in heart development and function. The balance between neural chemoattractants and chemorepellents secreted from cardiomyocytes determines cardiac nervous development. Nerve growth factor is a potent chemoattractant synthesized by cardiomyocytes, whereas Sema3a is a neural chemorepellent expressed specifically in the subendocardium. Disruption of this molecular balance induces disorganized cardiac innervation and may lead to sudden cardiac death due to lethal arrhythmias. Cardiac fibroblasts, of which there are large populations in the heart, secrete high levels of specific extracellular matrix and growth factors. Embryonic cardiac fibroblast-specific secreted factors collaboratively promote mitotic activity of embryonic cardiomyocytes and expansion of ventricular chambers during cardiogenesis. More recently, utilizing knowledge of the regulatory mechanisms of heart development, we found that cardiac fibroblasts can be directly reprogrammed into cardiomyocyte-like cells in vitro and in vivo by gene transfer of cardiac-specific transcription factors. Understanding the mechanisms of heart development and cardiac reprogramming technology may provide new therapeutic approaches for heart disease in the future.
Collapse
|
39
|
Cell shape and cardiosphere differentiation: a revelation by proteomic profiling. Biochem Res Int 2013; 2013:730874. [PMID: 24073335 PMCID: PMC3773893 DOI: 10.1155/2013/730874] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/08/2013] [Indexed: 01/16/2023] Open
Abstract
Stem cells (embryonic stem cells, somatic stem cells such as neural stem cells, and cardiac stem cells) and cancer cells are known to aggregate and form spheroid structures. This behavior is common in undifferentiated cells and may be necessary for adapting to certain conditions such as low-oxygen levels or to maintain undifferentiated status in microenvironments including stem cell niches. In order to decipher the meaning of this spheroid structure, we established a cardiosphere clone (CSC-21E) derived from the rat heart which can switch its morphology between spheroid and nonspheroid. Two forms, floating cardiospheres and dish-attached flat cells, could be switched reversibly by changing the cell culture condition. We performed differential proteome analysis studies and obtained protein profiles distinct between spherical forms and flat cells. From protein profiling analysis, we found upregulation of glycolytic enzymes in spheroids with some stress proteins switched in expression levels between these two forms. Evidence has been accumulating that certain chaperone/stress proteins are upregulated in concert with cellular changes including proliferation and differentiation. We would like to discuss the possible mechanism of how these aggregates affect cell differentiation and/or other cellular functions.
Collapse
|
40
|
Brønnum H, Eskildsen T, Andersen DC, Schneider M, Sheikh SP. IL-1β suppresses TGF-β-mediated myofibroblast differentiation in cardiac fibroblasts. Growth Factors 2013; 31:81-9. [PMID: 23734837 DOI: 10.3109/08977194.2013.787994] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiac fibrosis is a maladaptive response of the injured myocardium and is mediated through a complex interplay between molecular triggers and cellular responses. Interleukin (IL)-1β is a key inflammatory inducer in cardiac disease and promotes cell invasion and cardiomyocyte injury, but little is known of its impact on fibrosis. A major cornerstone of fibrosis is the differentiation of cardiac fibroblasts (CFs) into myofibroblasts (myoFbs), which is highly promoted by Transforming Growth Factor (TGF)-β. Therefore, we asked how IL-1β functionally modulated CF-to-myoFb differentiation. Using a differentiation model of ventricular fibroblasts, we found that IL-1β instigated substantial anti-fibrogenic effects. In specific, IL-1β reduced proliferation, matrix activity, cell motility and α-smooth muscle actin expression, which are all hallmarks of myoFb differentiation. These findings suggest that IL-1β, besides from its acknowledged adverse role in the inflammatory response, can also exert beneficial effects in cardiac fibrosis by actively suppressing differentiation of CFs into fibrogenic myoFbs.
Collapse
Affiliation(s)
- Hasse Brønnum
- Laboratory for Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital and Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | | | | | | | | |
Collapse
|
41
|
Angiotensin II regulates microRNA-132/-212 in hypertensive rats and humans. Int J Mol Sci 2013; 14:11190-207. [PMID: 23712358 PMCID: PMC3709727 DOI: 10.3390/ijms140611190] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/25/2013] [Accepted: 05/15/2013] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs), a group of small non-coding RNAs that fine tune translation of multiple target mRNAs, are emerging as key regulators in cardiovascular development and disease. MiRNAs are involved in cardiac hypertrophy, heart failure and remodeling following cardiac infarction; however, miRNAs involved in hypertension have not been thoroughly investigated. We have recently reported that specific miRNAs play an integral role in Angiotensin II receptor (AT1R) signaling, especially after activation of the Gαq signaling pathway. Since AT1R blockers are widely used to treat hypertension, we undertook a detailed analysis of potential miRNAs involved in Angiotensin II (AngII) mediated hypertension in rats and hypertensive patients, using miRNA microarray and qPCR analysis. The miR-132 and miR-212 are highly increased in the heart, aortic wall and kidney of rats with hypertension (159 ± 12 mm Hg) and cardiac hypertrophy following chronic AngII infusion. In addition, activation of the endothelin receptor, another Gαq coupled receptor, also increased miR-132 and miR-212. We sought to extend these observations using human samples by reasoning that AT1R blockers may decrease miR-132 and miR-212. We analyzed tissue samples of mammary artery obtained from surplus arterial tissue after coronary bypass operations. Indeed, we found a decrease in expression levels of miR-132 and miR-212 in human arteries from bypass-operated patients treated with AT1R blockers, whereas treatment with β-blockers had no effect. Taken together, these data suggest that miR-132 and miR-212 are involved in AngII induced hypertension, providing a new perspective in hypertensive disease mechanisms.
Collapse
|
42
|
Human cardiospheres as a source of multipotent stem and progenitor cells. Stem Cells Int 2013; 2013:916837. [PMID: 23766771 PMCID: PMC3666231 DOI: 10.1155/2013/916837] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/19/2013] [Indexed: 12/20/2022] Open
Abstract
Cardiospheres (CSs) are self-assembling multicellular clusters from the cellular outgrowth from cardiac explants cultured in nonadhesive substrates. They contain a core of primitive, proliferating cells, and an outer layer of mesenchymal/stromal cells and differentiating cells that express cardiomyocyte proteins and connexin 43. Because CSs contain both primitive cells and committed progenitors for the three major cell types present in the heart, that is, cardiomyocytes, endothelial cells, and smooth muscle cells, and because they are derived from percutaneous endomyocardial biopsies, they represent an attractive cell source for cardiac regeneration. In preclinical studies, CS-derived cells (CDCs) delivered to infarcted hearts resulted in improved cardiac function. CDCs have been tested safely in an initial phase-1 clinical trial in patients after myocardial infarction. Whether or not CDCs are superior to purified populations, for example, c-kit(+) cardiac stem cells, or to gene therapy approaches for cardiac regeneration remains to be evaluated.
Collapse
|
43
|
Horse serum reduces expression of membrane-bound and soluble isoforms of the preadipocyte marker Delta-like 1 homolog (Dlk1), but is inefficient for adipogenic differentiation of mouse preadipocytes. Acta Histochem 2013; 115:401-6. [PMID: 22975115 DOI: 10.1016/j.acthis.2012.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/18/2012] [Accepted: 08/19/2012] [Indexed: 11/24/2022]
Abstract
Downregulation of the preadipocyte marker Delta-like 1 homologue (Dlk1), an inhibitor of adipogenesis, has been suggested to be a prerequisite for adipogenic differentiation to occur, and low Dlk1 levels are often used to verify adipogenesis. Mouse preadipocytic cell lines such as 3T3-L1, as well as primary derived preadipocytes, are important models to study adipogenic differentiation and obesity. However, in vitro adipogenic differentiation of primary derived preadipocytes remains incomplete, and identification of factors that will improve the adipogenic differentiation process is thus of high value. In this study we show that horse serum fails to improve adipogenic differentiation of mouse preadipocytes (both 3T3-L1 cells and primary derived mouse preadipocytes) as otherwise reported for bone marrow derived adipogenic precursors. Unexpectedly, while Dlk1 levels were indeed decreased using horse serum, this did not correlate with a high degree of adipogenic differentiation. In conclusion, our novel results thus reveal that horse serum clearly is insufficient for adipogenic differentiation of mouse preadipocytes and that low levels of Dlk1 alone are a poor marker of mouse in vitro adipogenesis. We would also like to emphasize that it is very important for the field of cellular differentiation that researchers thoroughly investigate the effect of individual reagents in their protocols. Such data will increase understanding of the limitations and possibilities of individual systems.
Collapse
|
44
|
Left atrial appendages from adult hearts contain a reservoir of diverse cardiac progenitor cells. PLoS One 2013; 8:e59228. [PMID: 23555001 PMCID: PMC3595246 DOI: 10.1371/journal.pone.0059228] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 02/13/2013] [Indexed: 11/19/2022] Open
Abstract
AIMS There is strong evidence supporting the claim that endogenous cardiac progenitor cells (CPCs) are key players in cardiac regeneration, but the anatomic source and phenotype of the master cardiac progenitors remains uncertain. Our aim was to investigate the different cardiac stem cell populations in the left atrial appendage (LAA) and their fates. METHODS AND RESULTS We investigated the CPC content and profile of adult murine LAAs using immunohistochemistry and flow cytometry. We demonstrate that the LAA contains a large number of CPCs relative to other areas of the heart, representing over 20% of the total cell number. We grew two distinct CPC populations from the LAA by varying the degree of proteolysis. These differed by their histological location, surface marker profiles and growth dynamics. Specifically, CD45(pos) cells grew with milder proteolysis, while CD45(neg) cells grew mainly with more intense proteolysis. Both cell types could be induced to differentiate into cells with cardiomyocyte markers and organelles, albeit by different protocols. Many CD45(pos) cells expressed CD45 initially and rapidly lost its expression while differentiating. CONCLUSIONS Our results demonstrate that the left atrial appendage plays a role as a reservoir of multiple types of progenitor cells in murine adult hearts. Two different types of CPCs were isolated, differing in their epicardial-myocardial localization. Considering studies demonstrating layer-specific origins of different cardiac progenitor cells, our findings may shed light on possible pathways to study and utilize the diversity of endogenous progenitor cells in the adult heart.
Collapse
|
45
|
Reprogramming toward Heart Regeneration: Stem Cells and Beyond. Cell Stem Cell 2013; 12:275-84. [DOI: 10.1016/j.stem.2013.02.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/11/2013] [Accepted: 02/19/2013] [Indexed: 12/28/2022]
|
46
|
miR-21 promotes fibrogenic epithelial-to-mesenchymal transition of epicardial mesothelial cells involving Programmed Cell Death 4 and Sprouty-1. PLoS One 2013; 8:e56280. [PMID: 23441172 PMCID: PMC3575372 DOI: 10.1371/journal.pone.0056280] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 01/11/2013] [Indexed: 12/21/2022] Open
Abstract
The lining of the adult heart contains epicardial mesothelial cells (EMCs) that have the potential to undergo fibrogenic Epithelial-to-Mesenchymal Transition (EMT) during cardiac injury. EMT of EMCs has therefore been suggested to contribute to the heterogeneous fibroblast pool that mediates cardiac fibrosis. However, the molecular basis of this process is poorly understood. Recently, microRNAs (miRNAs) have been shown to regulate a number of sub-cellular events in cardiac disease. Hence, we hypothesized that miRNAs regulate fibrogenic EMT in the adult heart. Indeed pro-fibrogenic stimuli, especially TGF-β, promoted EMT progression in EMC cultures, which resulted in differential expression of numerous miRNAs, especially the pleiotropic miR-21. Accordingly, ectopic expression of miR-21 substantially promoted the fibroblast-like phenotype arising from fibrogenic EMT, whereas an antagonist that targeted miR-21 blocked this effect, as assessed on the E-cadherin/α-smooth muscle actin balance, cell viability, matrix activity, and cell motility, thus making miR-21 a relevant target of EMC-derived fibrosis. Several mRNA targets of miR-21 was differentially regulated during fibrogenic EMT of EMCs and miR-21-dependent targeting of Programmed Cell Death 4 (PDCD4) and Sprouty Homolog 1 (SPRY1) significantly contributed to the development of a fibroblastoid phenotype. However, PDCD4- and SPRY1-targeting was not entirely ascribable to all phenotypic effects from miR-21, underscoring the pleiotropic biological role of miR-21 and the increasing number of recognized miR-21 targets.
Collapse
|
47
|
Kawaguchi N, Nakanishi T. Cardiomyocyte regeneration. Cells 2013; 2:67-82. [PMID: 24709645 PMCID: PMC3972659 DOI: 10.3390/cells2010067] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 12/25/2012] [Accepted: 01/05/2013] [Indexed: 01/14/2023] Open
Abstract
The heart was initially believed to be a terminally differentiated organ; once the cardiomyocytes died, no recovery could be made to replace the dead cells. However, around a decade ago, the concept of cardiac stem cells (CSCs) in adult hearts was proposed. CSCs differentiate into cardiomyocytes, keeping the heart functioning. Studies have proved the existence of stem cells in the heart. These somatic stem cells have been studied for use in cardiac regeneration. Moreover, recently, induced pluripotent stem cells (iPSCs) were invented, and methodologies have now been developed to induce stable cardiomyocyte differentiation and purification of mature cardiomyocytes. A reprogramming method has also been applied to direct reprogramming using cardiac fibroblasts into cardiomyocytes. Here, we address cardiomyocyte differentiation of CSCs and iPSCs. Furthermore, we describe the potential of CSCs in regenerative biology and regenerative medicine.
Collapse
Affiliation(s)
- Nanako Kawaguchi
- Department of Pediatric Cardiology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Toshio Nakanishi
- Department of Pediatric Cardiology, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
48
|
Generation of human secondary cardiospheres as a potent cell processing strategy for cell-based cardiac repair. Biomaterials 2013; 34:651-61. [DOI: 10.1016/j.biomaterials.2012.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 10/04/2012] [Indexed: 12/12/2022]
|
49
|
Abstract
Cellular cardiomyoplasty is a cell therapy using stem cells or progenitor cells for myocardial regeneration to improve cardiac function and mitigate heart failure. Since we first published cellular cardiomyoplasty in 1989, this procedure became the innovative method to treat damaged myocardium other than heart transplantation. A significant improvement in cardiac function, metabolism, and perfusion is generally observed in experimental and clinical studies, but the improvement is mild and incomplete. Although safety, feasibility, and efficacy have been well documented for the procedure, the beneficial mechanisms remain unclear and optimization of the procedure requires further study. This chapter briefly reviews the stem cells used for cellular cardiomyoplasty and their clinical outcomes with possible improvements in future studies.
Collapse
Affiliation(s)
- Elizabeth K Lamb
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | | | | |
Collapse
|
50
|
High density sphere culture of adult cardiac cells increases the levels of cardiac and progenitor markers and shows signs of vasculogenesis. BIOMED RESEARCH INTERNATIONAL 2012; 2013:696837. [PMID: 23484142 PMCID: PMC3591148 DOI: 10.1155/2013/696837] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/05/2012] [Indexed: 11/17/2022]
Abstract
3D environment and high cell density play an important role in restoring and supporting the phenotypes of cells represented in cardiac tissues. The aim of this study was therefore to investigate the suitability of high density sphere (HDS) cultures for studies of cardiomyocyte-, endothelial-, and stem-cell biology. Primary adult cardiac cells from nine human biopsies were cultured using different media for up to 9 weeks. The possibilities to favor a certain cell phenotype and induce production of extra cellular matrix (ECM) were studied by histology, immunohistochemistry, and quantitative real-time PCR. Defined media gave significant increase in both cardiac- and progenitor-specific markers and also an intraluminal position of endothelial cells over time. Cardiac media showed indication of differentiation and maturity of HDS considering the ECM production and activities within NOTCH regulation but no additional cardiac differentiation. Endothelial media gave no positive effects on endothelial phenotype but increased proliferation without fibroblast overgrowth. In addition, indications for early vasculogenesis were found. It was also possible to affect the Wnt signaling in HDS by addition of a glycogen synthase kinase 3 (GSK3) inhibitor. In conclusion, these findings show the suitability of HDS as in vitro model for studies of cardiomyocyte-, endothelial-, and stem-cell biology.
Collapse
|