101
|
Tang J, Cui X, Caranasos TG, Hensley MT, Vandergriff AC, Hartanto Y, Shen D, Zhang H, Zhang J, Cheng K. Heart Repair Using Nanogel-Encapsulated Human Cardiac Stem Cells in Mice and Pigs with Myocardial Infarction. ACS NANO 2017; 11:9738-9749. [PMID: 28929735 PMCID: PMC5656981 DOI: 10.1021/acsnano.7b01008] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/01/2017] [Indexed: 05/20/2023]
Abstract
Stem cell transplantation is currently implemented clinically but is limited by low retention and engraftment of transplanted cells and the adverse effects of inflammation and immunoreaction when allogeneic or xenogeneic cells are used. Here, we demonstrate the safety and efficacy of encapsulating human cardiac stem cells (hCSCs) in thermosensitive poly(N-isopropylacrylamine-co-acrylic acid) or P(NIPAM-AA) nanogel in mouse and pig models of myocardial infarction (MI). Unlike xenogeneic hCSCs injected in saline, injection of nanogel-encapsulated hCSCs does not elicit systemic inflammation or local T cell infiltrations in immunocompetent mice. In mice and pigs with acute MI, injection of encapsulated hCSCs preserves cardiac function and reduces scar sizes, whereas injection of hCSCs in saline has an adverse effect on heart healing. In conclusion, thermosensitive nanogels can be used as a stem cell carrier: the porous and convoluted inner structure allows nutrient, oxygen, and secretion diffusion but can prevent the stem cells from being attacked by immune cells.
Collapse
Affiliation(s)
- Junnan Tang
- Department
of Cardiology, The First Affiliated Hospital
of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department
of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Department
of Biomedical Engineering, University of
North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Xiaolin Cui
- School
of Chemical Engineering, The University
of Adelaide, Adelaide, SA 5005, Australia
| | - Thomas G. Caranasos
- Division
of Cardiothoracic Surgery, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - M. Taylor Hensley
- Department
of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Department
of Biomedical Engineering, University of
North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Adam C. Vandergriff
- Department
of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Department
of Biomedical Engineering, University of
North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Yusak Hartanto
- School
of Chemical Engineering, The University
of Adelaide, Adelaide, SA 5005, Australia
| | - Deliang Shen
- Department
of Cardiology, The First Affiliated Hospital
of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hu Zhang
- School
of Chemical Engineering, The University
of Adelaide, Adelaide, SA 5005, Australia
| | - Jinying Zhang
- Department
of Cardiology, The First Affiliated Hospital
of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ke Cheng
- Department
of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Department
of Biomedical Engineering, University of
North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
- Pharmacoengineering
and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
102
|
Chen Z, Zhu W, Bender I, Gong W, Kwak IY, Yellamilli A, Hodges TJ, Nemoto N, Zhang J, Garry DJ, van Berlo JH. Pathologic Stimulus Determines Lineage Commitment of Cardiac C-kit + Cells. Circulation 2017; 136:2359-2372. [PMID: 29021323 DOI: 10.1161/circulationaha.117.030137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 09/20/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although cardiac c-kit+ cells are being tested in clinical trials, the circumstances that determine lineage differentiation of c-kit+ cells in vivo are unknown. Recent findings suggest that endogenous cardiac c-kit+ cells rarely contribute cardiomyocytes to the adult heart. We assessed whether various pathological stimuli differentially affect the eventual cell fates of c-kit+ cells. METHODS We used single-cell sequencing and genetic lineage tracing of c-kit+ cells to determine whether various pathological stimuli would result in different fates of c-kit+ cells. RESULTS Single-cell sequencing of cardiac CD45-c-kit+ cells showed innate heterogeneity, indicative of the existence of vascular and mesenchymal c-kit+ cells in normal hearts. Cardiac pressure overload resulted in a modest increase in c-kit-derived cardiomyocytes, with significant increases in the numbers of endothelial cells and fibroblasts. Doxorubicin-induced acute cardiotoxicity did not increase c-kit-derived endothelial cell fates but instead induced cardiomyocyte differentiation. Mechanistically, doxorubicin-induced DNA damage in c-kit+ cells resulted in expression of p53. Inhibition of p53 blocked cardiomyocyte differentiation in response to doxorubicin, whereas stabilization of p53 was sufficient to increase c-kit-derived cardiomyocyte differentiation. CONCLUSIONS These results demonstrate that different pathological stimuli induce different cell fates of c-kit+ cells in vivo. Although the overall rate of cardiomyocyte formation from c-kit+ cells is still below clinically relevant levels, we show that p53 is central to the ability of c-kit+ cells to adopt cardiomyocyte fates, which could lead to the development of strategies to preferentially generate cardiomyocytes from c-kit+ cells.
Collapse
Affiliation(s)
- Zhongming Chen
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.)
| | - Wuqiang Zhu
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Department of Biomedical Engineering, University of Alabama at Birmingham (W.Z., J.Z.)
| | - Ingrid Bender
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.)
| | - Wuming Gong
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.).,Stem Cell Institute, University of Minnesota, Minneapolis (W.G., D.J.G., J.H.v.B.)
| | - Il-Youp Kwak
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.)
| | - Amritha Yellamilli
- Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.).,Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis (A.Y., J.H.v.B.)
| | - Thomas J Hodges
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.)
| | - Natsumi Nemoto
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.)
| | - Jianyi Zhang
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Department of Biomedical Engineering, University of Alabama at Birmingham (W.Z., J.Z.)
| | - Daniel J Garry
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.).,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.).,Stem Cell Institute, University of Minnesota, Minneapolis (W.G., D.J.G., J.H.v.B.)
| | - Jop H van Berlo
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis (Z.C., W.Z., I.B., W.G., I-Y.K., T.J.H., N.N., J.Z., D.J.G., J.H.v.B.) .,Lillehei Heart Institute, University of Minnesota, Minneapolis (Z.C., I.B., W.G., I-Y.K., A.Y., N.N., D.J.G., J.H.v.B.).,Stem Cell Institute, University of Minnesota, Minneapolis (W.G., D.J.G., J.H.v.B.).,Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis (A.Y., J.H.v.B.)
| |
Collapse
|
103
|
Ciulla MM. Epistemology of Natural Strategies for Cardiac Tissue Repair. Front Cardiovasc Med 2017; 4:61. [PMID: 29038784 PMCID: PMC5630689 DOI: 10.3389/fcvm.2017.00061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/15/2017] [Indexed: 12/01/2022] Open
Affiliation(s)
- Michele M Ciulla
- Laboratory of Clinical Informatics and Cardiovascular Imaging, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Cardiovascular Diseases Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
104
|
Santoso MR, Yang PC. Molecular Imaging of Stem Cells and Exosomes for Myocardial Regeneration. CURRENT CARDIOVASCULAR IMAGING REPORTS 2017. [DOI: 10.1007/s12410-017-9433-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
105
|
Cardiac Progenitor Cells and the Interplay with Their Microenvironment. Stem Cells Int 2017; 2017:7471582. [PMID: 29075298 PMCID: PMC5623801 DOI: 10.1155/2017/7471582] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The microenvironment plays a crucial role in the behavior of stem and progenitor cells. In the heart, cardiac progenitor cells (CPCs) reside in specific niches, characterized by key components that are altered in response to a myocardial infarction. To date, there is a lack of knowledge on these niches and on the CPC interplay with the niche components. Insight into these complex interactions and into the influence of microenvironmental factors on CPCs can be used to promote the regenerative potential of these cells. In this review, we discuss cardiac resident progenitor cells and their regenerative potential and provide an overview of the interactions of CPCs with the key elements of their niche. We focus on the interaction between CPCs and supporting cells, extracellular matrix, mechanical stimuli, and soluble factors. Finally, we describe novel approaches to modulate the CPC niche that can represent the next step in recreating an optimal CPC microenvironment and thereby improve their regeneration capacity.
Collapse
|
106
|
Weinberger F, Mannhardt I, Eschenhagen T. Engineering Cardiac Muscle Tissue: A Maturating Field of Research. Circ Res 2017; 120:1487-1500. [PMID: 28450366 DOI: 10.1161/circresaha.117.310738] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Twenty years after the initial description of a tissue engineered construct, 3-dimensional human cardiac tissues of different kinds are now generated routinely in many laboratories. Advances in stem cell biology and engineering allow for the generation of constructs that come close to recapitulating the complex structure of heart muscle and might, therefore, be amenable to industrial (eg, drug screening) and clinical (eg, cardiac repair) applications. Whether the more physiological structure of 3-dimensional constructs provides a relevant advantage over standard 2-dimensional cell culture has yet to be shown in head-to-head-comparisons. The present article gives an overview on current strategies of cardiac tissue engineering with a focus on different hydrogel methods and discusses perspectives and challenges for necessary steps toward the real-life application of cardiac tissue engineering for disease modeling, drug development, and cardiac repair.
Collapse
Affiliation(s)
- Florian Weinberger
- From the Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Ingra Mannhardt
- From the Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Thomas Eschenhagen
- From the Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Germany; and DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany.
| |
Collapse
|
107
|
Spontaneous activation of a MAVS-dependent antiviral signaling pathway determines high basal interferon-β expression in cardiac myocytes. J Mol Cell Cardiol 2017; 111:102-113. [PMID: 28822807 DOI: 10.1016/j.yjmcc.2017.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 07/31/2017] [Accepted: 08/14/2017] [Indexed: 01/09/2023]
Abstract
Viral myocarditis is a leading cause of sudden death in young adults as the limited turnover of cardiac myocytes renders the heart particularly vulnerable to viral damage. Viruses induce an antiviral type I interferon (IFN-α/β) response in essentially all cell types, providing an immediate innate protection. Cardiac myocytes express high basal levels of IFN-β to help pre-arm them against viral infections, however the mechanism underlying this expression remains unclear. Using primary cultures of murine cardiac and skeletal muscle cells, we demonstrate here that the mitochondrial antiviral signaling (MAVS) pathway is spontaneously activated in unstimulated cardiac myocytes but not cardiac fibroblasts or skeletal muscle cells. Results suggest that MAVS association with the mitochondrial-associated ER membranes (MAM) is a determinant of high basal IFN-β expression, and demonstrate that MAVS is essential for spontaneous high basal expression of IFN-β in cardiac myocytes and the heart. Together, results provide the first mechanism for spontaneous high expression of the antiviral cytokine IFN-β in a poorly replenished and essential cell type.
Collapse
|
108
|
Adult cardiac stem cells are multipotent and robustly myogenic: c-kit expression is necessary but not sufficient for their identification. Cell Death Differ 2017; 24:2101-2116. [PMID: 28800128 PMCID: PMC5686347 DOI: 10.1038/cdd.2017.130] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/14/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022] Open
Abstract
Multipotent adult resident cardiac stem cells (CSCs) were first identified by the expression of c-kit, the stem cell factor receptor. However, in the adult myocardium c-kit alone cannot distinguish CSCs from other c-kit-expressing (c-kitpos) cells. The adult heart indeed contains a heterogeneous mixture of c-kitpos cells, mainly composed of mast and endothelial/progenitor cells. This heterogeneity of cardiac c-kitpos cells has generated confusion and controversy about the existence and role of CSCs in the adult heart. Here, to unravel CSC identity within the heterogeneous c-kit-expressing cardiac cell population, c-kitpos cardiac cells were separated through CD45-positive or -negative sorting followed by c-kitpos sorting. The blood/endothelial lineage-committed (Lineagepos) CD45posc-kitpos cardiac cells were compared to CD45neg(Lineageneg/Linneg) c-kitpos cardiac cells for stemness and myogenic properties in vitro and in vivo. The majority (~90%) of the resident c-kitpos cardiac cells are blood/endothelial lineage-committed CD45posCD31posc-kitpos cells. In contrast, the LinnegCD45negc-kitpos cardiac cell cohort, which represents ⩽10% of the total c-kitpos cells, contain all the cardiac cells with the properties of adult multipotent CSCs. These characteristics are absent from the c-kitneg and the blood/endothelial lineage-committed c-kitpos cardiac cells. Single Linnegc-kitpos cell-derived clones, which represent only 1-2% of total c-kitpos myocardial cells, when stimulated with TGF-β/Wnt molecules, acquire full transcriptome and protein expression, sarcomere organisation, spontaneous contraction and electrophysiological properties of differentiated cardiomyocytes (CMs). Genetically tagged cloned progeny of one Linnegc-kitpos cell when injected into the infarcted myocardium, results in significant regeneration of new CMs, arterioles and capillaries, derived from the injected cells. The CSC's myogenic regenerative capacity is dependent on commitment to the CM lineage through activation of the SMAD2 pathway. Such regeneration was not apparent when blood/endothelial lineage-committed c-kitpos cardiac cells were injected. Thus, among the cardiac c-kitpos cell cohort only a very small fraction has the phenotype and the differentiation/regenerative potential characteristics of true multipotent CSCs.
Collapse
|
109
|
Abstract
The stem cells keep us young by endogenously repairing us upon need. They do so bysmartly one step forward towards differentiation while another step backward to nurturethe undifferentiated stem cells. They are building blocks for every organ witha differential rate of repair of worn out tissues. Since stem cells can be cultured with a normal karyo type, they could be the ideal source for heart repair after myocardial infarction. As opposed to lower vertebrates and neonatal mice, cardiac regeneration in adult mammalian heart seems to be difficult to assess with a solid evidence of cytokinesis. It becomes more difficult to quantify the level of regeneration after myocardial infarction injury against a background of a large invasion of proliferating inflammatory cells. The question to beraised is how the renewal of a piece of myocardium follows the time line of picking upcell types in series: cardiomyocytes, endothelial cells, smooth muscle cells, fibroblast, pacemaker cells, conducting and Purkinje cells to bring the orchestration of rhythmically contracting and relaxing heart. This review focuses on where we are onthe status of heart repair and cardiac regeneration.
Collapse
Affiliation(s)
- H R Ahmad
- Dr. HR Ahmad, MD PhD Bochum, FCPS. Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Satwat Hashmi
- Dr. Satwat Hashmi, MBBS MS PhD. Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
110
|
Cahill TJ, Choudhury RP, Riley PR. Heart regeneration and repair after myocardial infarction: translational opportunities for novel therapeutics. Nat Rev Drug Discov 2017; 16:699-717. [DOI: 10.1038/nrd.2017.106] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
111
|
Pathophysiology and therapeutic potential of cardiac fibrosis. Inflamm Regen 2017; 37:13. [PMID: 29259712 PMCID: PMC5725925 DOI: 10.1186/s41232-017-0046-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/28/2017] [Indexed: 12/24/2022] Open
Abstract
Inflammatory and fibrotic responses to myocardial damage are essential for cardiac repair; however, these responses often result in extensive fibrotic remodeling with impaired systolic function. Recent reports have suggested that such acute phase responses provide a favorable environment for endogenous cardiac regeneration, which is mainly driven by the division of pre-existing cardiomyocytes (CMs). Existing CMs in mammals can re-acquire proliferative activity after substantial cardiac damage, and elements other than CMs in the physiological and/or pathological environment, such as hypoxia, angiogenesis, and the polarity of infiltrating macrophages, have been reported to regulate replication. Cardiac fibroblasts comprise the largest cell population in terms of cell number in the myocardium, and they play crucial roles in the proliferation and protection of CMs. The in vivo direct reprogramming of functional CMs has been investigated in cardiac regeneration. Currently, growth factors, transcription factors, microRNAs, and small molecules promoting the regeneration and protection of these CMs have also been actively researched. Here, we summarize and discuss current studies on the relationship between cardiac inflammation and fibrosis, and cardiac regeneration and protection, which would be useful for the development of therapeutic strategies to treat and prevent advanced heart failure.
Collapse
|
112
|
The use and abuse of Cre/Lox recombination to identify adult cardiomyocyte renewal rate and origin. Pharmacol Res 2017; 127:116-128. [PMID: 28655642 DOI: 10.1016/j.phrs.2017.06.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/19/2017] [Accepted: 06/21/2017] [Indexed: 11/20/2022]
Abstract
The adult mammalian heart, including the human, is unable to regenerate segmental losses after myocardial infarction. This evidence has been widely and repeatedly used up-to-today to suggest that the myocardium, contrary to most adult tissues, lacks an endogenous stem cell population or more specifically a bona-fide cardiomyocyte-generating progenitor cell of biological significance. In the last 15 years, however, the field has slowly evolved from the dogma that no new cardiomyocytes were produced from shortly after birth to the present consensus that new cardiomyocytes are formed throughout lifespan. This endogenous regenerative potential increases after various forms of injury. Nevertheless, the degree/significance and more importantly the origin of adult new cardiomyocytes remains strongly disputed. Evidence from independent laboratories has shown that the adult myocardium harbours bona-fide tissue-specific cardiac stem cells (CSCs). Their transplantation and in situ activation have demonstrated the CSCs regenerative potential and have been used to develop regeneration protocols which in pre-clinical tests have shown to be effective in the prevention and treatment of heart failure. Recent reports purportedly tracking the c-kit+CSC's fate using Cre/lox recombination in the mouse have challenged the existence and regenerative potential of the CSCs and have raised scepticism about their role in myocardial homeostasis and regeneration. The validity of these reports, however, is controversial because they failed to show that the experimental approach used is capable to both identify and tract the fate of the CSCs. Despite these serious shortcomings, in contraposition to the CSCs, these publications have proposed the proliferation of existing adult fully-matured cardiomyocytes as the relevant mechanism to explain cardiomyocyte renewal in the adult. This review critically ponders the available evidence showing that the adult mammalian heart possesses a definable myocyte-generating progenitor cell of biological significance. This endogenous regenerative potential is expected to provide the bases for novel approaches of myocardial repair in the near future.
Collapse
|
113
|
Cai CL, Molkentin JD. The Elusive Progenitor Cell in Cardiac Regeneration: Slip Slidin' Away. Circ Res 2017; 120:400-406. [PMID: 28104772 DOI: 10.1161/circresaha.116.309710] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/31/2022]
Abstract
The adult human heart is unable to regenerate after various forms of injury, suggesting that this organ lacks a biologically meaningful endogenous stem cell pool. However, injecting the infarcted area of the adult mammalian heart with exogenously prepared progenitor cells of various types has been reported to create new myocardium by the direct conversion of these progenitor cells into cardiomyocytes. These reports remain controversial because follow-up studies from independent laboratories failed to observe such an effect. Also, the exact nature of various putative myocyte-producing progenitor cells remains elusive and undefined across laboratories. By comparison, the field has gradually worked toward a consensus viewpoint that proposes that the adult mammalian myocardium can undergo a low level of new cardiomyocyte renewal of ≈1% per year, which is primarily because of proliferation of existing cardiomyocytes but not from the differentiation of putative progenitor cells. This review will weigh the emerging evidence, suggesting that the adult mammalian heart lacks a definable myocyte-generating progenitor cell of biological significance.
Collapse
Affiliation(s)
- Chen-Leng Cai
- From the Department of Developmental and Regenerative Biology, and Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.-L.C.); and Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center and Howard Hughes Medical Institute, OH (J.D.M.).
| | - Jeffery D Molkentin
- From the Department of Developmental and Regenerative Biology, and Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.-L.C.); and Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center and Howard Hughes Medical Institute, OH (J.D.M.).
| |
Collapse
|
114
|
Miyawaki A, Obana M, Mitsuhara Y, Orimoto A, Nakayasu Y, Yamashita T, Fukada SI, Maeda M, Nakayama H, Fujio Y. Adult murine cardiomyocytes exhibit regenerative activity with cell cycle reentry through STAT3 in the healing process of myocarditis. Sci Rep 2017; 7:1407. [PMID: 28469272 PMCID: PMC5431117 DOI: 10.1038/s41598-017-01426-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/29/2017] [Indexed: 11/09/2022] Open
Abstract
Mammalian cardiomyocytes substantially lose proliferative capacity immediately after birth, limiting adult heart regeneration after injury. However, clinical myocarditis appears to be self-limiting with tissue-reparative properties. Here, we investigated the molecular mechanisms underlying the recovery from myocarditis with regard to cardiomyocyte proliferation using an experimental autoimmune myocarditis (EAM) model. Three weeks after EAM induction (EAM3w), cardiac tissue displayed infiltration of inflammatory cells with cardiomyocyte apoptosis. However, by EAM5w, the myocardial damage was remarkably attenuated, associated with an increase in cardiomyocytes that were positively stained with cell cycle markers at EAM3w. Cardiomyocyte fate mapping study revealed that the proliferating cardiomyocytes primarily derived from pre-existing cardiomyocytes. Signal transducer and activator of transcription 3 (STAT3) was robustly activated in cardiomyocytes during inflammation, accompanied by induction of interleukin-6 family cytokines. Cardiomyocyte-specific ablation of STAT3 gene suppressed the frequency of cycling cardiomyocytes in the recovery period without influencing inflammatory status, resulting in impaired tissue repair and cardiac dysfunction. Finally, microarray analysis revealed that the expression of regeneration-related genes, metallothioneins and clusterin, in cardiomyocytes was decreased by STAT3 gene deletion. These data show that adult mammalian cardiomyocytes restore regenerative capacity with cell cycle reentry through STAT3 as the heart recovers from myocarditis-induced cardiac damage.
Collapse
Affiliation(s)
- Akimitsu Miyawaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Mitsuhara
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Aya Orimoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Nakayasu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomomi Yamashita
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makiko Maeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
115
|
Wollert KC, Meyer GP, Müller-Ehmsen J, Tschöpe C, Bonarjee V, Larsen AI, May AE, Empen K, Chorianopoulos E, Tebbe U, Waltenberger J, Mahrholdt H, Ritter B, Pirr J, Fischer D, Korf-Klingebiel M, Arseniev L, Heuft HG, Brinchmann JE, Messinger D, Hertenstein B, Ganser A, Katus HA, Felix SB, Gawaz MP, Dickstein K, Schultheiss HP, Ladage D, Greulich S, Bauersachs J. Intracoronary autologous bone marrow cell transfer after myocardial infarction: the BOOST-2 randomised placebo-controlled clinical trial. Eur Heart J 2017; 38:2936-2943. [DOI: 10.1093/eurheartj/ehx188] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/23/2017] [Indexed: 01/21/2023] Open
|
116
|
Abstract
Dramatic evolution in medical and catheter interventions and complex surgeries to treat children with congenital heart disease (CHD) has led to a growing number of patients with a multitude of long-term complications associated with morbidity and mortality. Heart failure in patients with hypoplastic left heart syndrome predicated by functional single ventricle lesions is associated with an increase in CHD prevalence and remains a significant challenge. Pathophysiological mechanisms contributing to the progression of CHD, including single ventricle lesions and dilated cardiomyopathy, and adult heart disease may inevitably differ. Although therapeutic options for advanced cardiac failure are restricted to heart transplantation or mechanical circulatory support, there is a strong impetus to develop novel therapeutic strategies. As lower vertebrates, such as the newt and zebrafish, have a remarkable ability to replace lost cardiac tissue, this intrinsic self-repair machinery at the early postnatal stage in mice was confirmed by partial ventricular resection. Although the underlying mechanistic insights might differ among the species, mammalian heart regeneration occurs even in humans, with the highest degree occurring in early childhood and gradually declining with age in adulthood, suggesting the advantage of stem cell therapy to ameliorate ventricular dysfunction in patients with CHD. Although effective clinical translation by a variety of stem cells in adult heart disease remains inconclusive with respect to the improvement of cardiac function, case reports and clinical trials based on stem cell therapies in patients with CHD may be invaluable for the next stage of therapeutic development. Dissecting the differential mechanisms underlying progressive ventricular dysfunction in children and adults may lead us to identify a novel regenerative therapy. Future regenerative technologies to treat patients with CHD are exciting prospects for heart regeneration in general practice.
Collapse
Affiliation(s)
- Hidemasa Oh
- From the Department of Regenerative Medicine, Center for Innovative Clinical Medicine, Okayama University Hospital, Japan
| |
Collapse
|
117
|
Hypoxia modulates cell migration and proliferation in placenta-derived mesenchymal stem cells. J Thorac Cardiovasc Surg 2017; 154:543-552.e3. [PMID: 28526501 DOI: 10.1016/j.jtcvs.2017.03.141] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 02/28/2017] [Accepted: 03/13/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVES For more than a decade, stem cells isolated from different tissues have been evaluated in cell therapy. Among them, the human bone marrow-derived mesenchymal stem cells (hBM-MSCs) were investigated extensively in the treatment of myocardial infarction. Recently, the human placenta-derived mesenchymal stem cells (hPD-MSCs), which are readily available from a biological waste, appear to be a viable alternative to hBM-MSCs. METHODS C-X-C chemokine receptor type 4 (CXCR4) gene expression and localization were detected and validated in hPD-MSCs and hBM-MSCs via polymerase chain reaction and immunofluorescence. Subsequently, cell culture conditions for CXCR4 expression were optimized in stromal-derived factor-1 alpha (SDF1-α), glucose, and cobalt chloride (CoCl2) by the use of cell viability, proliferation, and migration assays. To elucidate the cell signaling pathway, protein expression of CXCR4, hypoxia-inducible factor-1α, interleukin-6, Akt, and extracellular signal-regulated kinase were analyzed by Western blot. CXCR4-positive cells were sorted and analyzed by florescence-activated cell sorting. RESULTS CXCR4 was expressed on both hPD-MSCs and hBM-MSCs at the basal level. HPD-MSCs were shown to have a greater sensitivity to SDF-1α-dependent cell migration compared with hBM-MSCs. In addition, CXCR4 expression was significantly greater in both hPD-MSCs and hBM-MSCs with SDF-1α or CoCl2-induced hypoxia treatment. However, CXCR4+ hPD-MSCs population increased by 10-fold in CoCl2-induced hypoxia. In contrast, only a 2-fold increase was observed in the CXCR4+ hBM-MSCs population in similar conditions. After CoCl2-induced hypoxia, the CXCR4/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase signaling pathway was activated prominently in hPD-MSCs, whereas in hBM-MSCs, the CXCR4/phosphatidylinositol 3-kinase/Akt pathway was triggered. CONCLUSIONS Our current results suggest that hPD-MSCs could represent a viable and effective alternative to hBM-MSCs for translational studies in cardiocellular repair.
Collapse
|
118
|
Li Y, Tian S, Lei I, Liu L, Ma P, Wang Z. Transplantation of multipotent Isl1+ cardiac progenitor cells preserves infarcted heart function in mice. Am J Transl Res 2017; 9:1530-1542. [PMID: 28386378 PMCID: PMC5376043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/18/2017] [Indexed: 06/07/2023]
Abstract
Cell-based cardiac therapy is a promising therapeutic strategy to restore heart function after myocardial infarction (MI). However, the cell type selection and ensuing effects remain controversial. Here, we intramyocardially injected Isl1+ cardiac progenitor cells (CPCs) derived from EGFP/luciferase double-tagged mouse embryonic stem (dt-mES) cells with vehicle (fibrin gel) or phosphate-buffered saline (PBS) into the infarcted area in nude mice to assess the contribution of CPCs to the recovery of cardiac function post-MI. Our results showed that Isl1+ CPCs differentiated normally into three cardiac lineages (cardiomyocytes (CMs), endothelial cells and smooth muscle cells) both on cell culture plates and in fibrin gel. Cell retention was significantly increased when the transplanted cells were injected with vehicle. Importantly, 28 days after injection, CPCs were observed to differentiate into CMs within the infarcted area. Moreover, numerous CD31+ endothelial cells derived from endogenous revascularization and differentiation of the injected CPCs were detected. SMMHC-, Ki67- and CX-43-positive cells were identified in the injected CPC population, further demonstrating the proliferation, differentiation and integration of the transplanted CPCs in host cells. Furthermore, animal hearts injected with CPCs showed increased angiogenesis, decreased infarct size, and improved heart function. In conclusion, our studies showed that Isl1+ CPCs, when combined with a suitable vehicle, can produce notable therapeutic effects in the infarcted heart, suggesting that CPCs might be an ideal cell source for cardiac therapy.
Collapse
Affiliation(s)
- Yunpeng Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical UniversityXian 710032, Shaanxi, China
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Shuo Tian
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
- Faculty of Health Sciences, University of MacauMacau SAR, China
| | - Liu Liu
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| | - Peter Ma
- Department of Biologic and Materials Sciences, Biomedical Engineering, Macromolecular Science and Engineering Center, and Materials Science and Engineering, The University of MichiganAnn Arbor 48109, MI, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of MichiganAnn Arbor 48109, MI, USA
| |
Collapse
|
119
|
Ludlow AT, Gratidão L, Ludlow LW, Spangenburg EE, Roth SM. Acute exercise activates p38 MAPK and increases the expression of telomere-protective genes in cardiac muscle. Exp Physiol 2017; 102:397-410. [PMID: 28166612 DOI: 10.1113/ep086189] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/20/2017] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? A positive association between telomere length and exercise training has been shown in cardiac tissue of mice. It is currently unknown how each bout of exercise influences telomere-length-regulating proteins. We sought to determine how a bout of exercise altered the expression of telomere-length-regulating genes and a related signalling pathway in cardiac tissue of mice. What is the main finding and its importance? Acute exercise altered the expression of telomere-length-regulating genes in cardiac tissue and might be related to altered mitogen-activated protein kinase signalling. These findings are important in understanding how exercise provides a cardioprotective phenotype with ageing. Age is the greatest risk factor for cardiovascular disease. Telomere length is shorter in the hearts of aged mice compared with young mice, and short telomere length has been associated with an increased risk of cardiovascular disease. One year of voluntary wheel-running exercise attenuates the age-associated loss of telomere length and results in altered gene expression of telomere-length-maintaining and genome-stabilizing proteins in heart tissue of mice. Understanding the early adaptive response of the heart to an endurance exercise bout is paramount to understanding the impact of endurance exercise on heart tissue and cells. To this end, we studied mice before (BL), immediately after (TP1) and 1 h after a treadmill running bout (TP2). We measured the changes in expression of telomere-related genes (shelterin components), DNA-damage-sensing (p53 and Chk2) and DNA-repair genes (Ku70 and Ku80) and mitogen-activated protein kinase (MAPK) signalling. The TP1 animals had increased TRF1 and TRF2 protein and mRNA levels, greater expression of DNA-repair and -response genes (Chk2 and Ku80) and greater protein content of phosphorylated p38 MAPK compared with both BL and TP2 animals. These data provide insights into how physiological stressors remodel the heart tissue and how an early adaptive response mediated by exercise may be maintaining telomere length and/or stabilizing the heart genome through the upregulation of telomere-protective genes.
Collapse
Affiliation(s)
- Andrew T Ludlow
- Department of Kinesiology, School of Public Health, University of Maryland at College Park, College Park, MD, USA.,Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Laila Gratidão
- Department of Kinesiology, School of Public Health, University of Maryland at College Park, College Park, MD, USA.,Kinesiology Graduate Program, Catholic University of Brasilia, Brasilia, Brazil
| | - Lindsay W Ludlow
- Department of Kinesiology, School of Public Health, University of Maryland at College Park, College Park, MD, USA.,Department of Applied Physiology, Southern Methodist University, Dallas, TX, USA
| | - Espen E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Stephen M Roth
- Department of Kinesiology, School of Public Health, University of Maryland at College Park, College Park, MD, USA
| |
Collapse
|
120
|
Ellison-Hughes GM, Madeddu P. Exploring pericyte and cardiac stem cell secretome unveils new tactics for drug discovery. Pharmacol Ther 2017; 171:1-12. [PMID: 27916652 PMCID: PMC5636619 DOI: 10.1016/j.pharmthera.2016.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ischaemic diseases remain a major cause of morbidity and mortality despite continuous advancements in medical and interventional treatments. Moreover, available drugs reduce symptoms associated with tissue ischaemia, without providing a definitive repair. Cardiovascular regenerative medicine is an expanding field of research that aims to improve the treatment of ischaemic disorders through restorative methods, such as gene therapy, stem cell therapy, and tissue engineering. Stem cell transplantation has salutary effects through direct and indirect actions, the latter being attributable to growth factors and cytokines released by stem cells and influencing the endogenous mechanisms of repair. Autologous stem cell therapies offer less scope for intellectual property coverage and have limited scalability. On the other hand, off-the-shelf cell products and derivatives from the stem cell secretome have a greater potential for large-scale distribution, thus enticing commercial investors and reciprocally producing more significant medical and social benefits. This review focuses on the paracrine properties of cardiac stem cells and pericytes, two stem cell populations that are increasingly attracting the attention of regenerative medicine operators. It is likely that new cardiovascular drugs are introduced in the next future by applying different approaches based on the refinement of the stem cell secretome.
Collapse
Affiliation(s)
- Georgina M Ellison-Hughes
- Centre of Human & Aerospace Physiological Sciences, Centre for Stem Cells and Regenerative Medicine, Faculty of Medicine & Life Sciences, Guy's Campus, King's College London, London SE1 1UL, United Kingdom
| | - Paolo Madeddu
- Chair Experimental Cardiovascular Medicine, Bristol Heart Institute, School of Clinical Sciences University of Bristol Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, United Kingdom.
| |
Collapse
|
121
|
Mesenchymal stromal cell therapy to promote cardiac tissue regeneration and repair. Curr Opin Organ Transplant 2017; 22:86-96. [DOI: 10.1097/mot.0000000000000379] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
122
|
Blice-Baum AC, Zambon AC, Kaushik G, Viswanathan MC, Engler AJ, Bodmer R, Cammarato A. Modest overexpression of FOXO maintains cardiac proteostasis and ameliorates age-associated functional decline. Aging Cell 2017; 16:93-103. [PMID: 28090761 PMCID: PMC5242305 DOI: 10.1111/acel.12543] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2016] [Indexed: 11/27/2022] Open
Abstract
Heart performance declines with age. Impaired protein quality control (PQC), due to reduced ubiquitin‐proteasome system (UPS) activity, autophagic function, and/or chaperone‐mediated protein refolding, contributes to cardiac deterioration. The transcription factor FOXO participates in regulating genes involved in PQC, senescence, and numerous other processes. Here, a comprehensive approach, involving molecular genetics, novel assays to probe insect cardiac physiology, and bioinformatics, was utilized to investigate the influence of heart‐restricted manipulation of dFOXO expression in the rapidly aging Drosophila melanogaster model. Modest dFOXO overexpression was cardioprotective, ameliorating nonpathological functional decline with age. This was accompanied by increased expression of genes associated predominantly with the UPS, relative to other PQC components, which was validated by a significant decrease in ubiquitinated proteins. RNAi knockdown of UPS candidates accordingly compromised myocardial physiology in young flies. Conversely, excessive dFOXO overexpression or suppression proved detrimental to heart function and/or organismal development. This study highlights D. melanogaster as a model of cardiac aging and FOXO as a tightly regulated mediator of proteostasis and heart performance over time.
Collapse
Affiliation(s)
- Anna C. Blice-Baum
- Division of Cardiology; Department of Medicine; Johns Hopkins University; Baltimore MD 21205 USA
| | - Alexander C. Zambon
- Department of Biopharmaceutical Sciences; Keck Graduate Institute; Claremont CA 91711 USA
- Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program; La Jolla CA 92037 USA
| | - Gaurav Kaushik
- Department of Bioengineering; University of California, San Diego; La Jolla CA 92093 USA
| | - Meera C. Viswanathan
- Division of Cardiology; Department of Medicine; Johns Hopkins University; Baltimore MD 21205 USA
| | - Adam J. Engler
- Department of Bioengineering; University of California, San Diego; La Jolla CA 92093 USA
| | - Rolf Bodmer
- Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program; La Jolla CA 92037 USA
| | - Anthony Cammarato
- Division of Cardiology; Department of Medicine; Johns Hopkins University; Baltimore MD 21205 USA
| |
Collapse
|
123
|
Abstract
The hearts of lower vertebrates such as fish and salamanders display scarless regeneration following injury, although this feature is lost in adult mammals. The remarkable capacity of the neonatal mammalian heart to regenerate suggests that the underlying machinery required for the regenerative process is evolutionarily retained. Recent studies highlight the epicardial covering of the heart as an important source of the signalling factors required for the repair process. The developing epicardium is also a major source of cardiac fibroblasts, smooth muscle, endothelial cells and stem cells. Here, we examine animal models that are capable of scarless regeneration, the role of the epicardium as a source of cells, signalling mechanisms implicated in the regenerative process and how these mechanisms influence cardiomyocyte proliferation. We also discuss recent advances in cardiac stem cell research and potential therapeutic targets arising from these studies.
Collapse
Affiliation(s)
| | - Nadia Rosenthal
- National Heart and Lung Institute, Imperial College London, London, UK Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia The Jackson Laboratory, Bar Harbor, ME, USA
| |
Collapse
|
124
|
Dulce RA, Kulandavelu S, Schulman IH, Fritsch J, Hare JM. Nitric Oxide Regulation of Cardiovascular Physiology and Pathophysiology. Nitric Oxide 2017. [DOI: 10.1016/b978-0-12-804273-1.00024-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
125
|
|
126
|
Duffey OJ, Smart N. Approaches to augment vascularisation and regeneration of the adult heart via the reactivated epicardium. Glob Cardiol Sci Pract 2016; 2016:e201628. [PMID: 28979901 PMCID: PMC5624183 DOI: 10.21542/gcsp.2016.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 12/15/2016] [Indexed: 11/05/2022] Open
Abstract
Survival rates following myocardial infarction have increased in recent years but current treatments for post-infarction recovery are inadequate and cannot induce regeneration of damaged hearts. Regenerative medicine could provide disease-reversing treatments by harnessing modern concepts in cell and developmental biology. A recently-established paradigm in regenerative medicine is that regeneration of a tissue can be achieved by reactivation of the coordinated developmental processes that originally formed the tissue. In the heart, the epicardium has emerged as an important regulator of cardiac development and reactivation of epicardial developmental processes may provide a means to enable cardiac regeneration. Indeed, in adult mouse hearts, treatment with thymosin β4 and other drug-like molecules reactivates the epicardium and improves outcomes after myocardial infarction by inducing regenerative paracrine signalling, neovascularisation and de novo cardiomyocyte production. However, there are considerable limitations to current methods of epicardial reactivation that prevent direct translation into clinical practice. Here, we describe the rationale for targeting the epicardium and the successes and limitations of this approach. We consider how several recent advances in epicardial biology could be used to overcome these limitations. These advances include insight into epicardial signalling and heterogeneity, epicardial modulation of inflammation and epicardial remodelling of extracellular matrix.
Collapse
Affiliation(s)
- Owen J. Duffey
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
127
|
Affiliation(s)
- Richard T Lee
- From Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, and the Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); and Whitaker Cardiovascular Institute, Boston University, Boston, MA (K.W.).
| | - Kenneth Walsh
- From Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, and the Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (R.T.L.); and Whitaker Cardiovascular Institute, Boston University, Boston, MA (K.W.).
| |
Collapse
|
128
|
Cantero Peral S, Bernstein D, Nelson TJ. Regenerative medicine - From stem cell biology to clinical trials for pediatric heart failure. PROGRESS IN PEDIATRIC CARDIOLOGY 2016. [DOI: 10.1016/j.ppedcard.2016.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
129
|
Stone GW. Periprocedural Myocardial Infarction. JACC Cardiovasc Interv 2016; 9:2229-2231. [DOI: 10.1016/j.jcin.2016.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 09/09/2016] [Accepted: 09/10/2016] [Indexed: 11/16/2022]
|
130
|
Depletion of Tip60 from In Vivo Cardiomyocytes Increases Myocyte Density, Followed by Cardiac Dysfunction, Myocyte Fallout and Lethality. PLoS One 2016; 11:e0164855. [PMID: 27768769 PMCID: PMC5074524 DOI: 10.1371/journal.pone.0164855] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/15/2016] [Indexed: 12/20/2022] Open
Abstract
Tat-interactive protein 60 (Tip60), encoded by the Kat5 gene, is a member of the MYST family of acetyltransferases. Cancer biology studies have shown that Tip60 induces the DNA damage response, apoptosis, and cell-cycle inhibition. Although Tip60 is expressed in the myocardium, its role in cardiomyocytes (CMs) is unclear. Earlier studies here showed that application of cardiac stress to globally targeted Kat5+/—haploinsufficient mice resulted in inhibition of apoptosis and activation of the CM cell-cycle, despite only modest reduction of Tip60 protein levels. It was therefore of interest to ascertain the effects of specifically and substantially depleting Tip60 from CMs using Kat5LoxP/-;Myh6-Cre mice in the absence of stress. We report initial findings using this model, in which the effects of specifically depleting Tip60 protein from ventricular CMs, beginning at early neonatal stages, were assessed in 2–12 week-old mice. Although 5’-bromodeoxyuridine immunostaining indicated that CM proliferation was not altered at any of these stages, CM density was increased in 2 week-old ventricles, which persisted in 4 week-old hearts when TUNEL staining revealed inhibition of apoptosis. By week 4, levels of connexin-43 were depleted, and its patterning was dysmorphic, concomitant with an increase in cardiac hypertrophy marker expression and interstitial fibrosis. This was followed by systolic dysfunction at 8 weeks, after which extensive apoptosis and CM fallout occurred, followed by lethality as mice approached 12 weeks of age. In summary, chronic depletion of Tip60 from the ventricular myocardium beginning at early stages of neonatal heart development causes CM death after 8 weeks; hence, Tip60 protein has a crucial function in the heart.
Collapse
|
131
|
Macrophages and regeneration: Lessons from the heart. Semin Cell Dev Biol 2016; 58:26-33. [DOI: 10.1016/j.semcdb.2016.04.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/18/2016] [Accepted: 04/17/2016] [Indexed: 12/24/2022]
|
132
|
Plaisance I, Perruchoud S, Fernandez-Tenorio M, Gonzales C, Ounzain S, Ruchat P, Nemir M, Niggli E, Pedrazzini T. Cardiomyocyte Lineage Specification in Adult Human Cardiac Precursor Cells Via Modulation of Enhancer-Associated Long Noncoding RNA Expression. JACC Basic Transl Sci 2016; 1:472-493. [PMID: 29707678 PMCID: PMC5916868 DOI: 10.1016/j.jacbts.2016.06.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human CPCs produce predominantly smooth muscle cells. CPCs can be redirected to the cardiomyocyte fate by transient activation followed by inhibition of NOTCH signaling. Inhibition of NOTCH signaling during differentiation represses MIR-143/145 expression and blocks smooth muscle differentiation. Expression of the microRNAs is under control of CARMEN, a long noncoding RNA associated with an enhancer located in the MIR-143/145 locus and target of NOTCH signaling. The CARMEN/MIR-145/143 locus represents a promising therapeutic target to favor production of cardiomyocytes in cell replacement therapies.
The mechanisms controlling differentiation in adult cardiac precursor cells (CPCs) are still largely unknown. In this study, CPCs isolated from the human heart were found to produce predominantly smooth muscle cells but could be redirected to the cardiomyocyte fate by transient activation followed by inhibition of NOTCH signaling. NOTCH inhibition repressed MIR-143/145 expression, and blocked smooth muscle differentiation. Expression of the microRNAs is under control of CARMEN, a long noncoding RNA associated with an enhancer located in the MIR-143/145 locus and target of NOTCH signaling. The CARMEN/MIR-145/143 axis represents, therefore, a promising target to favor production of cardiomyocytes in cell replacement therapies.
Collapse
Affiliation(s)
- Isabelle Plaisance
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Stéphanie Perruchoud
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | | | - Christine Gonzales
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Samir Ounzain
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Patrick Ruchat
- Department of Cardiovascular Surgery, University of Lausanne Medical School, Lausanne, Switzerland
| | - Mohamed Nemir
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Ernst Niggli
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Thierry Pedrazzini
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
- Reprint requests and correspondence: Dr. Thierry Pedrazzini, Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, CH-1011 Lausanne, Switzerland.
| |
Collapse
|
133
|
Aguado T, Gutiérrez FJ, Aix E, Schneider RP, Giovinazzo G, Blasco MA, Flores I. Telomere Length Defines the Cardiomyocyte Differentiation Potency of Mouse Induced Pluripotent Stem Cells. Stem Cells 2016; 35:362-373. [PMID: 27612935 DOI: 10.1002/stem.2497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 12/21/2022]
Abstract
Induced pluripotent stem cells (iPSCs) can be differentiated in vitro and in vivo to all cardiovascular lineages and are therefore a promising cell source for cardiac regenerative therapy. However, iPSC lines do not all differentiate into cardiomyocytes (CMs) with the same efficiency. Here, we show that telomerase-competent iPSCs with relatively long telomeres and high expression of the shelterin-complex protein TRF1 (iPSChighT ) differentiate sooner and more efficiently into CMs than those with relatively short telomeres and low TRF1 expression (iPSClowT ). Ascorbic acid, an enhancer of cardiomyocyte differentiation, further increases the cardiomyocyte yield from iPSChighT but does not rescue the cardiomyogenic potential of iPSClowT . Interestingly, although iPSCslowT differentiate very poorly to the mesoderm and endoderm lineages, they differentiate very efficiently to the ectoderm lineage, indicating that cell fate can be determined by in vitro selection of iPSCs with different telomere content. Our findings highlight the importance of selecting iPSCs with ample telomere reserves in order to generate high numbers of CMs in a fast, reliable, and efficient way. Stem Cells 2017;35:362-373.
Collapse
Affiliation(s)
- Tania Aguado
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Francisco J Gutiérrez
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Esther Aix
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Ralph P Schneider
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Giovanna Giovinazzo
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - María A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ignacio Flores
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| |
Collapse
|
134
|
A reproducible protocol for neonatal ischemic injury and cardiac regeneration in neonatal mice. Basic Res Cardiol 2016; 111:64. [PMID: 27665606 PMCID: PMC5035663 DOI: 10.1007/s00395-016-0580-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/09/2016] [Indexed: 01/12/2023]
Abstract
Cardiac regeneration is one of the prime visions in cardiovascular research. The mouse neonatal apical resection and left anterior descending artery (LAD) ligation model introduced novel in vivo mammalian assays to study cardiac regeneration. However, recent reports and editorials discussed and critically questioned the value and technical reproducibility of the mouse neonatal myocardial infarction approach, making it paramount to develop and use a reproducible model system. We established a mouse neonatal myocardial infarction model by visually confirmed ligation of the LAD using microsurgery. TdT-mediated dUTP nick-end labeling (TUNEL) proved reproducible massive myocardial infarctions in a defined region of the apex and anterior wall of neonatal and 7-day-old mice. Whereas hearts ligated on postnatal day 7 displayed chronic injury, cardiac samples ligated immediately after birth always showed complete structural regeneration after long-term follow-up. Cardiac regeneration was observed in all mouse stains (C57BL/6J, ICR, and mixed background C57BL/6JxSv129) tested so far. We present a detailed in vivo protocol to study complex mechanisms of complete cardiac repair following ischemic cardiac damage. Neonatal LAD ligation surgery is feasible, and results in reproducible myocardial infarctions 24 h after ligation, and no structural myocardial defects are detectable following long-term follow-up. We encourage the cardiovascular community to use our protocol and teaching video to answer key scientific questions in the field of cardiac regeneration.
Collapse
|
135
|
Hamid T, Xu Y, Ismahil MA, Li Q, Jones SP, Bhatnagar A, Bolli R, Prabhu SD. TNF receptor signaling inhibits cardiomyogenic differentiation of cardiac stem cells and promotes a neuroadrenergic-like fate. Am J Physiol Heart Circ Physiol 2016; 311:H1189-H1201. [PMID: 27591224 DOI: 10.1152/ajpheart.00904.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 08/25/2016] [Indexed: 01/23/2023]
Abstract
Despite expansion of resident cardiac stem cells (CSCs; c-kit+Lin-) after myocardial infarction, endogenous repair processes are insufficient to prevent adverse cardiac remodeling and heart failure (HF). This suggests that the microenvironment in post-ischemic and failing hearts compromises CSC regenerative potential. Inflammatory cytokines, such as tumor necrosis factor-α (TNF), are increased after infarction and in HF; whether they modulate CSC function is unknown. As the effects of TNF are specific to its two receptors (TNFRs), we tested the hypothesis that TNF differentially modulates CSC function in a TNFR-specific manner. CSCs were isolated from wild-type (WT), TNFR1-/-, and TNFR2-/- adult mouse hearts, expanded and evaluated for cell competence and differentiation in vitro in the absence and presence of TNF. Our results indicate that TNF signaling in murine CSCs is constitutively related primarily to TNFR1, with TNFR2 inducible after stress. TNFR1 signaling modestly diminished CSC proliferation, but, along with TNFR2, augmented CSC resistance to oxidant stress. Deficiency of either TNFR1 or TNFR2 did not impact CSC telomerase activity. Importantly, TNF, primarily via TNFR1, inhibited cardiomyogenic commitment during CSC differentiation, and instead promoted smooth muscle and endothelial fates. Moreover, TNF, via both TNFR1 and TNFR2, channeled an alternate CSC neuroadrenergic-like fate (capable of catecholamine synthesis) during differentiation. Our results suggest that elevated TNF in the heart restrains cardiomyocyte differentiation of resident CSCs and may enhance adrenergic activation, both effects that would reduce the effectiveness of endogenous cardiac repair and the response to exogenous stem cell therapy, while promoting adverse cardiac remodeling.
Collapse
Affiliation(s)
- Tariq Hamid
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and
| | - Yuanyuan Xu
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and
| | - Mohamed Ameen Ismahil
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and
| | - Qianhong Li
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Steven P Jones
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Aruni Bhatnagar
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Roberto Bolli
- Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Sumanth D Prabhu
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham and Birmingham Veterans Affairs Medical Center, Birmingham, Alabama; and
| |
Collapse
|
136
|
Directed Differentiation of Zebrafish Pluripotent Embryonic Cells to Functional Cardiomyocytes. Stem Cell Reports 2016; 7:370-382. [PMID: 27569061 PMCID: PMC5032289 DOI: 10.1016/j.stemcr.2016.07.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 12/24/2022] Open
Abstract
A cardiomyocyte differentiation in vitro system from zebrafish embryos remains to be established. Here, we have determined pluripotency window of zebrafish embryos by analyzing their gene-expression patterns of pluripotency factors together with markers of three germ layers, and have found that zebrafish undergoes a very narrow period of pluripotency maintenance from zygotic genome activation to a brief moment after oblong stage. Based on the pluripotency and a combination of appropriate conditions, we established a rapid and efficient method for cardiomyocyte generation in vitro from primary embryonic cells. The induced cardiomyocytes differentiated into functional and specific cardiomyocyte subtypes. Notably, these in vitro generated cardiomyocytes exhibited typical contractile kinetics and electrophysiological features. The system provides a new paradigm of cardiomyocyte differentiation from primary embryonic cells in zebrafish. The technology provides a new platform for the study of heart development and regeneration, in addition to drug discovery, disease modeling, and assessment of cardiotoxic agents. Zebrafish embryos may start to exit from pluripotency shortly after the oblong stage Beating cell clusters are efficiently generated from zebrafish blastomeres Beating cell clusters contain specific cardiomyocyte subtypes Induced cardiomyocytes possess normal electrophysiological features
Collapse
|
137
|
De Pauw A, Massion P, Sekkali B, Andre E, Dubroca C, Kmecova J, Bouzin C, Friart A, Sibille C, Gilon P, De Mulder D, Esfahani H, Strapart A, Martherus R, Payen V, Sonveaux P, Brouckaert P, Janssens S, Balligand JL. Paracrine nitric oxide induces expression of cardiac sarcomeric proteins in adult progenitor cells through soluble guanylyl cyclase/cyclic-guanosine monophosphate and Wnt/β-catenin inhibition. Cardiovasc Res 2016; 112:478-90. [PMID: 27520736 DOI: 10.1093/cvr/cvw196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/29/2016] [Indexed: 01/05/2023] Open
Abstract
AIM Cardiac progenitor cells (CPC) from adult hearts can differentiate to several cell types composing the myocardium but the underlying molecular pathways are poorly characterized. We examined the role of paracrine nitric oxide (NO) in the specification of CPC to the cardiac lineage, particularly through its inhibition of the canonical Wnt/β-catenin pathway, a critical step preceding cardiac differentiation. METHODS AND RESULTS Sca1 + CPC from adult mouse hearts were isolated by magnetic-activated cell sorting and clonally expanded. Pharmacologic NO donors increased their expression of cardiac myocyte-specific sarcomeric proteins in a concentration and time-dependent manner. The optimal time window for NO efficacy coincided with up-regulation of CPC expression of Gucy1a3 (coding the alpha1 subunit of guanylyl cyclase). The effect of paracrine NO was reproduced in vitro upon co-culture of CPC with cardiac myocytes expressing a transgenic NOS3 (endothelial nitric oxide synthase) and in vivo upon injection of CPC in infarcted hearts from cardiac-specific NOS3 transgenic mice. In mono- and co-cultures, this effect was abrogated upon inhibition of soluble guanylyl cyclase or nitric oxide synthase, and was lost in CPC genetically deficient in Gucy1a3. Mechanistically, NO inhibits the constitutive activity of the canonical Wnt/β-catenin in CPC and in cell reporter assays in a guanylyl cyclase-dependent fashion. This was paralleled with decreased expression of β-catenin and down-regulation of Wnt target genes in CPC and abrogated in CPC with a stabilized, non-inhibitable β-catenin. CONCLUSIONS Exogenous or paracrine sources of NO promote the specification towards the myocyte lineage and expression of cardiac sarcomeric proteins of adult CPC. This is contingent upon the expression and activity of the alpha1 subunit of guanylyl cyclase in CPC that is necessary for NO-mediated inhibition of the canonical Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Aurelia De Pauw
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Paul Massion
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Belaid Sekkali
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Emilie Andre
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Caroline Dubroca
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Jana Kmecova
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Caroline Bouzin
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Ann Friart
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Catherine Sibille
- Department of Human Genetics, Cliniques Saint-Luc, Université Catholique de Louvain, 10 avenue Hippocrate, 1200 Brussels, Belgium
| | - Patrick Gilon
- Pole of Endocrinology, Diabetes and Nutrition (EDIN), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, B1.55.06, 55 avenue Hippocrate, 1200 Brussels, Belgium
| | - Delphine De Mulder
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Hrag Esfahani
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Adrien Strapart
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Ruben Martherus
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Valéry Payen
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Universiteit Gent, Technologiepark 927, 9052 Gent, Belgium
| | - Stefan Janssens
- Department of Cardiology, Katholieke Universiteit Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Departement de Medecine Interne et Cliniques Saint-Luc, Université Catholique de Louvain, B1.53.09, 52 Ave. Mounier, 1200 Brussels, Belgium
| |
Collapse
|
138
|
Foglia MJ, Poss KD. Building and re-building the heart by cardiomyocyte proliferation. Development 2016; 143:729-40. [PMID: 26932668 DOI: 10.1242/dev.132910] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adult human heart does not regenerate significant amounts of lost tissue after injury. Rather than making new, functional muscle, human hearts are prone to scarring and hypertrophy, which can often lead to fatal arrhythmias and heart failure. The most-cited basis of this ineffective cardiac regeneration in mammals is the low proliferative capacity of adult cardiomyocytes. However, mammalian cardiomyocytes can avidly proliferate during fetal and neonatal development, and both adult zebrafish and neonatal mice can regenerate cardiac muscle after injury, suggesting that latent regenerative potential exists. Dissecting the cellular and molecular mechanisms that promote cardiomyocyte proliferation throughout life, deciphering why proliferative capacity normally dissipates in adult mammals, and deriving means to boost this capacity are primary goals in cardiovascular research. Here, we review our current understanding of how cardiomyocyte proliferation is regulated during heart development and regeneration.
Collapse
Affiliation(s)
- Matthew J Foglia
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
139
|
Hatzistergos KE, Saur D, Seidler B, Balkan W, Breton M, Valasaki K, Takeuchi LM, Landin AM, Khan A, Hare JM. Stimulatory Effects of Mesenchymal Stem Cells on cKit+ Cardiac Stem Cells Are Mediated by SDF1/CXCR4 and SCF/cKit Signaling Pathways. Circ Res 2016; 119:921-30. [PMID: 27481956 DOI: 10.1161/circresaha.116.309281] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 07/29/2016] [Indexed: 01/13/2023]
Abstract
RATIONALE Culture-expanded cells originating from cardiac tissue that express the cell surface receptor cKit are undergoing clinical testing as a cell source for heart failure and congenital heart disease. Although accumulating data support that mesenchymal stem cells (MSCs) enhance the efficacy of cardiac cKit(+) cells (CSCs), the underlying mechanism for this synergistic effect remains incompletely understood. OBJECTIVE To test the hypothesis that MSCs stimulate endogenous CSCs to proliferate, migrate, and differentiate via the SDF1/CXCR4 and stem cell factor/cKit pathways. METHODS AND RESULTS Using genetic lineage-tracing approaches, we show that in the postnatal murine heart, cKit(+) cells proliferate, migrate, and form cardiomyocytes, but not endothelial cells. CSCs exhibit marked chemotactic and proliferative responses when cocultured with MSCs but not with cardiac stromal cells. Antagonism of the CXCR4 pathway with AMD3100 (an SDF1/CXCR4 antagonist) inhibited MSC-induced CSC chemotaxis but stimulated CSC cardiomyogenesis (P<0.0001). Furthermore, MSCs enhanced CSC proliferation via the stem cell factor/cKit and SDF1/CXCR4 pathways (P<0.0001). CONCLUSIONS Together these findings show that MSCs exhibit profound, yet differential, effects on CSC migration, proliferation, and differentiation and suggest a mechanism underlying the improved cardiac regeneration associated with combination therapy using CSCs and MSCs. These findings have important therapeutic implications for cell-based therapy strategies that use mixtures of CSCs and MSCs.
Collapse
Affiliation(s)
- Konstantinos E Hatzistergos
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Dieter Saur
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Barbara Seidler
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Wayne Balkan
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Matthew Breton
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Krystalenia Valasaki
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Lauro M Takeuchi
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Ana Marie Landin
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Aisha Khan
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.)
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, FL (K.E.H., W.B., M.B., K.V., L.M.T., A.M.L., A.K., J.M.H.); Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Germany (D.S., B.S.); and German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany (D.S., B.S.).
| |
Collapse
|
140
|
Valiente-Alandi I, Albo-Castellanos C, Herrero D, Sanchez I, Bernad A. Bmi1 (+) cardiac progenitor cells contribute to myocardial repair following acute injury. Stem Cell Res Ther 2016; 7:100. [PMID: 27472922 PMCID: PMC4967328 DOI: 10.1186/s13287-016-0355-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/25/2016] [Accepted: 06/28/2016] [Indexed: 01/19/2023] Open
Abstract
Background The inability of the adult mammalian heart to replace cells lost after severe cardiac injury compromises organ function. Although the heart is one of the least regenerative organs in the body, evidence accumulated in recent decades indicates a certain degree of renewal after injury. We have evaluated the role of cardiac Bmi1+ progenitor cells (Bmi1-CPC) following acute myocardial infarction (AMI). Methods Bmi1Cre/+;Rosa26YFP/+ (Bmi1-YFP) mice were used for lineage tracing strategy. After tamoxifen (TM) induction, yellow fluorescent protein (YFP) is expressed under the control of Rosa26 regulatory sequences in Bmi1+ cells. YFP+ cells were tracked following myocardial infarction. Additionally, whole transcriptome analysis of isolated YFP+ cells was performed in unchallenged hearts and after myocardial infarction. Results Deep-sequencing analysis of Bmi1-CPC from unchallenged hearts suggests that this population expresses high levels of pluripotency markers. Conversely, transcriptome evaluation of Bmi1-CPC following AMI shows a rich representation of genes related to cell proliferation, movement, and cell cycle. Lineage-tracing studies after cardiac infarction show that the progeny of Bmi1-expressing cells contribute to de novo cardiomyocytes (CM) (13.8 ± 5 % new YFP+ CM compared to 4.7 ± 0.9 % in age-paired non-infarcted hearts). However, apical resection of TM-induced day 1 Bmi1-YFP pups indicated a very minor contribution of Bmi1-derived cells to de novo CM. Conclusions Cardiac Bmi1 progenitor cells respond to cardiac injury, contributing to the generation of de novo CM in the adult mouse heart. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0355-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iñigo Valiente-Alandi
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.,Current address: The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carmen Albo-Castellanos
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.,Current address: Vivebiotech, San Sebastian, Spain
| | - Diego Herrero
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Iria Sanchez
- Unidad de Medicina Comparada, Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Antonio Bernad
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
141
|
The myocardial regenerative potential of three-dimensional engineered cardiac tissues composed of multiple human iPS cell-derived cardiovascular cell lineages. Sci Rep 2016; 6:29933. [PMID: 27435115 PMCID: PMC4951692 DOI: 10.1038/srep29933] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/24/2016] [Indexed: 12/27/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are a robust source for cardiac regenerative therapy due to their potential to support autologous and allogeneic transplant paradigms. The in vitro generation of three-dimensional myocardial tissue constructs using biomaterials as an implantable hiPSC-derived myocardium provides a path to realize sustainable myocardial regeneration. We generated engineered cardiac tissues (ECTs) from three cellular compositions of cardiomyocytes (CMs), endothelial cells (ECs), and vascular mural cells (MCs) differentiated from hiPSCs. We then determined the impact of cell composition on ECT structural and functional properties. In vitro force measurement showed that CM+EC+MC ECTs possessed preferential electromechanical properties versus ECTs without vascular cells indicating that incorporation of vascular cells augmented tissue maturation and function. The inclusion of MCs facilitated more mature CM sarcomeric structure, preferential alignment, and activated multiple tissue maturation pathways. The CM+EC+MC ECTs implanted onto infarcted, immune tolerant rat hearts engrafted, displayed both host and graft-derived vasculature, and ameliorated myocardial dysfunction. Thus, a composition of CMs and multiple vascular lineages derived from hiPSCs and incorporated into ECTs promotes functional maturation and demonstrates myocardial replacement and perfusion relevant for clinical translation.
Collapse
|
142
|
Ottaviani L, De Windt LJ, da Costa Martins PA. Exosomes: scytales in the damaged heart. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:222. [PMID: 27384882 DOI: 10.21037/atm.2016.05.17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Lara Ottaviani
- 1 Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands ; 2 Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Leon J De Windt
- 1 Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands ; 2 Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Paula A da Costa Martins
- 1 Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands ; 2 Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
143
|
Tepp K, Timohhina N, Puurand M, Klepinin A, Chekulayev V, Shevchuk I, Kaambre T. Bioenergetics of the aging heart and skeletal muscles: Modern concepts and controversies. Ageing Res Rev 2016; 28:1-14. [PMID: 27063513 DOI: 10.1016/j.arr.2016.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/03/2023]
Abstract
Age-related alterations in the bioenergetics of the heart and oxidative skeletal muscle tissues are of crucial influence on their performance. Until now the prevailing concept of aging was the mitochondrial theory, the increased production of reactive oxygen species, mediated by deficiency in the activity of respiratory chain complexes. However, studies with mitochondria in situ have presented results which, to some extent, disagree with previous ones, indicating that the mitochondrial theory of aging may be overestimated. The studies reporting age-related decline in mitochondrial function were performed using mainly isolated mitochondria. Measurements on this level are not able to take into account the system level properties. The relevant information can be obtained only from appropriate studies using cells or tissue fibers. The functional interactions between the components of Intracellular Energetic Unit (ICEU) regulate the energy production and consumption in oxidative muscle cells. The alterations of these interactions in ICEU should be studied in order to find a more effective protocol to decelerate the age-related changes taking place in the energy metabolism. In this article, an overview is given of the present theories and controversies of causes of age-related alterations in bioenergetics. Also, branches of study, which need more emphasis, are indicated.
Collapse
Affiliation(s)
- Kersti Tepp
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia.
| | - Natalja Timohhina
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Marju Puurand
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Aleksandr Klepinin
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Vladimir Chekulayev
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Tuuli Kaambre
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia; Faculty of Science, Tallinn University, Narva mnt. 25, 10120, Estonia
| |
Collapse
|
144
|
Tahara N, Brush M, Kawakami Y. Cell migration during heart regeneration in zebrafish. Dev Dyn 2016; 245:774-87. [PMID: 27085002 PMCID: PMC5839122 DOI: 10.1002/dvdy.24411] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/17/2016] [Accepted: 04/12/2016] [Indexed: 12/27/2022] Open
Abstract
Zebrafish possess the remarkable ability to regenerate injured hearts as adults, which contrasts the very limited ability in mammals. Although very limited, mammalian hearts do in fact have measurable levels of cardiomyocyte regeneration. Therefore, elucidating mechanisms of zebrafish heart regeneration would provide information of naturally occurring regeneration to potentially apply to mammalian studies, in addition to addressing this biologically interesting phenomenon in itself. Studies over the past 13 years have identified processes and mechanisms of heart regeneration in zebrafish. After heart injury, pre-existing cardiomyocytes dedifferentiate, enter the cell cycle, and repair the injured myocardium. This process requires interaction with epicardial cells, endocardial cells, and vascular endothelial cells. Epicardial cells envelope the heart, while endocardial cells make up the inner lining of the heart. They provide paracrine signals to cardiomyocytes to regenerate the injured myocardium, which is vascularized during heart regeneration. In addition, accumulating results suggest that local migration of these major cardiac cell types have roles in heart regeneration. In this review, we summarize the characteristics of various heart injury methods used in the research community and regeneration of the major cardiac cell types. Then, we discuss local migration of these cardiac cell types and immune cells during heart regeneration. Developmental Dynamics 245:774-787, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Naoyuki Tahara
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Michael Brush
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
145
|
Hatzistergos KE, Hare JM. Murine Models Demonstrate Distinct Vasculogenic and Cardiomyogenic cKit+ Lineages in the Heart. Circ Res 2016; 118:382-7. [PMID: 26846638 DOI: 10.1161/circresaha.115.308061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
After 2 recent genetic studies in mice addressing the developmental origins and regenerative activity of cardiac cKit+ cells, 2 additional reports by Sultana et al and Liu et al provide further information on the expression of cKit in the embryonic and adult hearts. Here, we synthesize the findings from the 4 distinct cKit models to gain insights into the biology of this important cell type.
Collapse
Affiliation(s)
- Konstantinos E Hatzistergos
- From the Interdisciplinary Stem Cell Institute (K.E.H.), and Department of Medicine, Division of Cardiology and Department of Molecular and Cellular Pharmacology (J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (K.E.H.), and Department of Medicine, Division of Cardiology and Department of Molecular and Cellular Pharmacology (J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL.
| |
Collapse
|
146
|
Affiliation(s)
- Ian Y Chen
- From Stanford Cardiovascular Institute (I.Y.C., J.C.W.), Division of Cardiovascular Medicine, Department of Medicine (I.Y.C., J.C.W.), and Department of Radiology (J.C.W.), Stanford University School of Medicine, CA
| | - Joseph C Wu
- From Stanford Cardiovascular Institute (I.Y.C., J.C.W.), Division of Cardiovascular Medicine, Department of Medicine (I.Y.C., J.C.W.), and Department of Radiology (J.C.W.), Stanford University School of Medicine, CA.
| |
Collapse
|
147
|
Miyawaki A, Mitsuhara Y, Orimoto A, Nakayasu Y, Tsunoda SI, Obana M, Maeda M, Nakayama H, Yoshioka Y, Tsutsumi Y, Fujio Y. Moesin is activated in cardiomyocytes in experimental autoimmune myocarditis and mediates cytoskeletal reorganization with protrusion formation. Am J Physiol Heart Circ Physiol 2016; 311:H476-86. [PMID: 27342875 DOI: 10.1152/ajpheart.00180.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/16/2016] [Indexed: 12/19/2022]
Abstract
Acute myocarditis is a self-limiting disease. Most patients with myocarditis recover without cardiac dysfunction in spite of limited capacity of myocardial regeneration. Therefore, to address intrinsic reparative machinery of inflamed hearts, we investigated the cellular dynamics of cardiomyocytes in response to inflammation using experimental autoimmune myocarditis (EAM) model. EAM was induced by immunization of BALB/c mice with α-myosin heavy chain peptides twice. The inflammatory reaction was evoked with myocardial damage with the peak at 3 wk after the first immunization (EAM3w). Morphological and functional restoration started from EAM3w, when active protrusion formation, a critical process of myocardial healing, was observed in cardiomyocytes. Shotgun proteomics revealed that cytoskeletal proteins were preferentially increased in cardiomyocytes at EAM3w, compared with preimmunized (EAM0w) hearts, and that moesin was the most remarkably upregulated among them. Immunoblot analyses demonstrated that the expression of both total and phosphorylated moesin was upregulated in isolated cardiomyocytes from EAM3w hearts. Immunofluorescence staining showed that moesin was localized at cardiomyocyte protrusions at EAM3w. Adenoviral vectors expressing wild-type, constitutively active and inactive form of moesin (wtMoesin, caMoesin, and iaMoesin, respectively) were transfected in neonatal rat cardiomyocytes. The overexpression of wtMoesin and caMoesin resulted in protrusion formation, while not iaMoesin. Finally, we found that cardiomyocyte protrusions were accompanied by cell-cell contact formation. The expression of moesin was upregulated in cardiomyocytes under inflammation, inducing protrusion formation in a phosphorylation-dependent fashion. Moesin signal could be a novel therapeutic target that stimulates myocardial repair by promoting contact formation of cardiomyocytes.
Collapse
Affiliation(s)
- Akimitsu Miyawaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Yusuke Mitsuhara
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Aya Orimoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Yusuke Nakayasu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Shin-Ichi Tsunoda
- Laboratory of Biopharmaceutical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Saitoasagi, Ibaraki, Osaka, Japan; and
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Makiko Maeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Yasuo Yoshioka
- Department of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Yasuo Tsutsumi
- Department of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-oka, Suita, Osaka, Japan;
| |
Collapse
|
148
|
van Berlo JH, Molkentin JD. Most of the Dust Has Settled: cKit+ Progenitor Cells Are an Irrelevant Source of Cardiac Myocytes In Vivo. Circ Res 2016; 118:17-9. [PMID: 26837741 DOI: 10.1161/circresaha.115.307934] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jop H van Berlo
- From the Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis (J.H.v.B.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, OH (J.D.M.); and Howard Hughes Medical Institute, Cincinnati, OH (J.D.M.).
| | - Jeffery D Molkentin
- From the Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis (J.H.v.B.); Department of Pediatrics, Cincinnati Children's Hospital Medical Center, OH (J.D.M.); and Howard Hughes Medical Institute, Cincinnati, OH (J.D.M.).
| |
Collapse
|
149
|
Mahmoudi M, Tachibana A, Goldstone AB, Woo YJ, Chakraborty P, Lee KR, Foote CS, Piecewicz S, Barrozo JC, Wakeel A, Rice BW, Bell III CB, Yang PC. Novel MRI Contrast Agent from Magnetotactic Bacteria Enables In Vivo Tracking of iPSC-derived Cardiomyocytes. Sci Rep 2016; 6:26960. [PMID: 27264636 PMCID: PMC4893600 DOI: 10.1038/srep26960] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 05/09/2016] [Indexed: 11/17/2022] Open
Abstract
Therapeutic delivery of human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMs) represents a novel clinical approach to regenerate the injured myocardium. However, methods for robust and accurate in vivo monitoring of the iCMs are still lacking. Although superparamagnetic iron oxide nanoparticles (SPIOs) are recognized as a promising tool for in vivo tracking of stem cells using magnetic resonance imaging (MRI), their signal persists in the heart even weeks after the disappearance of the injected cells. This limitation highlights the inability of SPIOs to distinguish stem cell viability. In order to overcome this shortcoming, we demonstrate the use of a living contrast agent, magneto-endosymbionts (MEs) derived from magnetotactic bacteria for the labeling of iCMs. The ME-labeled iCMs were injected into the infarcted area of murine heart and probed by MRI and bioluminescence imaging (BLI). Our findings demonstrate that the MEs are robust and effective biological contrast agents to track iCMs in an in vivo murine model. We show that the MEs clear within one week of cell death whereas the SPIOs remain over 2 weeks after cell death. These findings will accelerate the clinical translation of in vivo MRI monitoring of transplanted stem cell at high spatial resolution and sensitivity.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Atsushi Tachibana
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Andrew B. Goldstone
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Y. Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | - Kayla R. Lee
- Bell Biosystems Inc., San Francisco, CA 94107, USA
| | | | | | | | - Abdul Wakeel
- Bell Biosystems Inc., San Francisco, CA 94107, USA
| | | | | | - Phillip C. Yang
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
150
|
Ebert AD, Diecke S, Chen IY, Wu JC. Reprogramming and transdifferentiation for cardiovascular development and regenerative medicine: where do we stand? EMBO Mol Med 2016; 7:1090-103. [PMID: 26183451 PMCID: PMC4568945 DOI: 10.15252/emmm.201504395] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Heart disease remains a leading cause of mortality and a major worldwide healthcare burden. Recent advances in stem cell biology have made it feasible to derive large quantities of cardiomyocytes for disease modeling, drug development, and regenerative medicine. The discoveries of reprogramming and transdifferentiation as novel biological processes have significantly contributed to this paradigm. This review surveys the means by which reprogramming and transdifferentiation can be employed to generate induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and induced cardiomyocytes (iCMs). The application of these patient-specific cardiomyocytes for both in vitro disease modeling and in vivo therapies for various cardiovascular diseases will also be discussed. We propose that, with additional refinement, human disease-specific cardiomyocytes will allow us to significantly advance the understanding of cardiovascular disease mechanisms and accelerate the development of novel therapeutic options.
Collapse
Affiliation(s)
- Antje D Ebert
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sebastian Diecke
- Max Delbrück Center, Berlin, Germany Berlin Institute of Health, Berlin, Germany
| | - Ian Y Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|