1
|
Kesavan K, Panchakshari S, Abdelwahab H, Rabelo ESG, Chaudhary KR. Endothelial characteristics of cardiac stem cell antigen-1 expressing cells and their relevance to right ventricular adaptation. Can J Physiol Pharmacol 2025; 103:98-110. [PMID: 39841976 DOI: 10.1139/cjpp-2024-0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
A growing body of evidence suggest that the stem cell antigen-1 expressing (Sca-1+) cells in the heart may be the cardiac endothelial stem/progenitor cells. Their endothelial cell (EC) functions, and their role in right ventricle (RV) physiology and pathophysiology of right heart failure (RHF) remains poorly defined. This study investigated EC characteristics of rat cardiac Sca-1+ cells, assessed spatial distribution and studied changes in Sca-1+ cells during RV remodelling in monocrotaline (MCT) model of pulmonary hypertension and RV remodeling. First, flow-cytometry analysis of adult male and female Sprague Dawley (SD) and Fischer CDF rat heart cells was performed, and we observed that the majority of Sca-1+ cells also expressed CD31, an EC marker. Furthermore, Sca-1+ cells showed acetylated low-density lipoprotein (ac-LDL) uptake and lectin binding similar to CD31+ cells from the same heart. The Sca-1+ cells also demonstrated network formation when plated on Matrigel. In the MCT treated rats, we observed increase in RV hypertrophy that correlated with the reduction in the abundance of Sca-1+CD31+ cells in the RV. Together, the cardiac Sca-1+ cells in the heart are endothelial stem/progenitor-like cells. These cells have higher abundance in the RV and may play a role in RV adaptation.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Female
- Rats, Sprague-Dawley
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Heart Ventricles/cytology
- Endothelial Cells/metabolism
- Ventricular Remodeling
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Rats, Inbred F344
- Antigens, Ly/metabolism
- Adaptation, Physiological
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/physiopathology
- Monocrotaline
- Stem Cells/metabolism
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
Collapse
Affiliation(s)
- Kirishani Kesavan
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Sheethal Panchakshari
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Haya Abdelwahab
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | | | - Ketul R Chaudhary
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Dalhousie Cardiac Research Excellence Wave, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
2
|
Fiorino E, Rossin D, Vanni R, Aubry M, Giachino C, Rastaldo R. Recent Insights into Endogenous Mammalian Cardiac Regeneration Post-Myocardial Infarction. Int J Mol Sci 2024; 25:11747. [PMID: 39519298 PMCID: PMC11546116 DOI: 10.3390/ijms252111747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial infarction (MI) is a critical global health issue and a leading cause of heart failure. Indeed, while neonatal mammals can regenerate cardiac tissue mainly through cardiomyocyte proliferation, this ability is lost shortly after birth, resulting in the adult heart's inability to regenerate after injury effectively. In adult mammals, the adverse cardiac remodelling, which compensates for the loss of cardiac cells, impairs cardiac function due to the non-contractile nature of fibrotic tissue. Moreover, the neovascularisation after MI is inadequate to restore blood flow to the infarcted myocardium. This review aims to synthesise the most recent insights into the molecular and cellular players involved in endogenous myocardial and vascular regeneration, facilitating the identification of mechanisms that could be targeted to trigger cardiac regeneration, reduce fibrosis, and improve functional recovery post-MI. Reprogramming adult cardiomyocytes to regain their proliferative potential, along with the modulation of target cells responsible for neovascularisation, represents promising therapeutic strategies. An updated overview of endogenous mechanisms that regulate both myocardial and coronary vasculature regeneration-including stem and progenitor cells, growth factors, cell cycle regulators, and key signalling pathways-could help identify new critical intervention points for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy; (E.F.); (D.R.); (R.V.); (M.A.); (C.G.)
| |
Collapse
|
3
|
Du L, Wang X, Guo Y, Tao T, Wu H, Xu X, Zhang B, Chen T, Xu Q, Guo X. Altered lipid metabolism promoting cardiac fibrosis is mediated by CD34 + cell-derived FABP4 + fibroblasts. Exp Mol Med 2024; 56:1869-1886. [PMID: 39198543 PMCID: PMC11372182 DOI: 10.1038/s12276-024-01309-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 09/01/2024] Open
Abstract
Hyperlipidemia and hypertension might play a role in cardiac fibrosis, in which a heterogeneous population of fibroblasts seems important. However, it is unknown whether CD34+ progenitor cells are involved in the pathogenesis of heart fibrosis. This study aimed to explore the mechanism of CD34+ cell differentiation in cardiac fibrosis during hyperlipidemia. Through the analysis of transcriptomes from 50,870 single cells extracted from mouse hearts and 76,851 single cells from human hearts, we have effectively demonstrated the evolving cellular landscape throughout cardiac fibrosis. Disturbances in lipid metabolism can accelerate the development of fibrosis. Through the integration of bone marrow transplantation models and lineage tracing, our study showed that hyperlipidemia can expedite the differentiation of non-bone marrow-derived CD34+ cells into fibroblasts, particularly FABP4+ fibroblasts, in response to angiotensin II. Interestingly, the partial depletion of CD34+ cells led to a notable reduction in triglycerides in the heart, mitigated fibrosis, and improved cardiac function. Furthermore, immunostaining of human heart tissue revealed colocalization of CD34+ cells and fibroblasts. Mechanistically, our investigation of single-cell RNA sequencing data through pseudotime analysis combined with in vitro cellular studies revealed the crucial role of the PPARγ/Akt/Gsk3β pathway in orchestrating the differentiation of CD34+ cells into FABP4+ fibroblasts. Through our study, we generated valuable insights into the cellular landscape of CD34+ cell-derived cells in the hypertrophic heart with hyperlipidemia, indicating that the differentiation of non-bone marrow-derived CD34+ cells into FABP4+ fibroblasts during this process accelerates lipid accumulation and promotes heart failure via the PPARγ/Akt/Gsk3β pathway.
Collapse
Affiliation(s)
- Luping Du
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Tao
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Wu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Health care, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Tan FH, Bronner ME. Regenerative loss in the animal kingdom as viewed from the mouse digit tip and heart. Dev Biol 2024; 507:44-63. [PMID: 38145727 PMCID: PMC10922877 DOI: 10.1016/j.ydbio.2023.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
The myriad regenerative abilities across the animal kingdom have fascinated us for centuries. Recent advances in developmental, molecular, and cellular biology have allowed us to unearth a surprising diversity of mechanisms through which these processes occur. Developing an all-encompassing theory of animal regeneration has thus proved a complex endeavor. In this chapter, we frame the evolution and loss of animal regeneration within the broad developmental constraints that may physiologically inhibit regenerative ability across animal phylogeny. We then examine the mouse as a model of regeneration loss, specifically the experimental systems of the digit tip and heart. We discuss the digit tip and heart as a positionally-limited system of regeneration and a temporally-limited system of regeneration, respectively. We delve into the physiological processes involved in both forms of regeneration, and how each phase of the healing and regenerative process may be affected by various molecular signals, systemic changes, or microenvironmental cues. Lastly, we also discuss the various approaches and interventions used to induce or improve the regenerative response in both contexts, and the implications they have for our understanding regenerative ability more broadly.
Collapse
Affiliation(s)
- Fayth Hui Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
5
|
Shen Y, Kim IM, Tang Y. Re: Single-cell transcriptome analysis for characterizing primary Sca-1 positive, non-endothelial cardiac cells. J Mol Cell Cardiol 2023; 182:28-29. [PMID: 37454412 PMCID: PMC11987645 DOI: 10.1016/j.yjmcc.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/06/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Affiliation(s)
- Yan Shen
- Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Il-Man Kim
- Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Yaoliang Tang
- Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
6
|
Du L, Sun X, Gong H, Wang T, Jiang L, Huang C, Xu X, Li Z, Xu H, Ma L, Li W, Chen T, Xu Q. Single cell and lineage tracing studies reveal the impact of CD34 + cells on myocardial fibrosis during heart failure. Stem Cell Res Ther 2023; 14:33. [PMID: 36805782 PMCID: PMC9942332 DOI: 10.1186/s13287-023-03256-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND CD34+ cells have been used to treat the patients with heart failure, but the outcome is variable. It is of great significance to scrutinize the fate and the mechanism of CD34+ cell differentiation in vivo during heart failure and explore its intervention strategy. METHODS We performed single-cell RNA sequencing (scRNA-seq) of the total non-cardiomyocytes and enriched Cd34-tdTomato+ lineage cells in the murine (male Cd34-CreERT2; Rosa26-tdTomato mice) pressure overload model (transverse aortic constriction, TAC), and total non-cardiomyocytes from human adult hearts. Then, in order to determine the origin of CD34+ cell that plays a role in myocardial fibrosis, bone marrow transplantation model was performed. Furthermore, to further clarify the role of CD34 + cells in myocardial remodeling in response to TAC injury, we generated Cd34-CreERT2; Rosa26-eGFP-DTA (Cre/DTA) mice. RESULTS By analyzing the transcriptomes of 59,505 single cells from the mouse heart and 22,537 single cells from the human heart, we illustrated the dynamics of cell landscape during the progression of heart hypertrophy, including CD34+ cells, fibroblasts, endothelial and immune cells. By combining genetic lineage tracing and bone marrow transplantation models, we demonstrated that non-bone-marrow-derived CD34+ cells give rise to fibroblasts and endothelial cells, while bone-marrow-derived CD34+ cell turned into immune cells only in response to pressure overload. Interestingly, partial depletion of CD34+ cells alleviated the severity of myocardial fibrosis with a significant improvement of cardiac function in Cd34-CreERT2; Rosa26-eGFP-DTA model. Similar changes of non-cardiomyocyte composition and cellular heterogeneity of heart failure were also observed in human patient with heart failure. Furthermore, immunostaining showed a double labeling of CD34 and fibroblast markers in human heart tissue. Mechanistically, our single-cell pseudotime analysis of scRNA-seq data and in vitro cell culture study revealed that Wnt-β-catenin and TGFβ1/Smad pathways are critical in regulating CD34+ cell differentiation toward fibroblasts. CONCLUSIONS Our study provides a cellular landscape of CD34+ cell-derived cells in the hypertrophy heart of human and animal models, indicating that non-bone-marrow-derived CD34+ cells differentiating into fibroblasts largely account for cardiac fibrosis. These findings may provide novel insights for the pathogenesis of cardiac fibrosis and have further potential therapeutic implications for the heart failure.
Collapse
Affiliation(s)
- Luping Du
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Xiaotong Sun
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Hui Gong
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Ting Wang
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Liujun Jiang
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Chengchen Huang
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Xiaodong Xu
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Zhoubin Li
- grid.13402.340000 0004 1759 700XDepartment of Lung Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Hongfei Xu
- grid.13402.340000 0004 1759 700XDepartment of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Liang Ma
- grid.13402.340000 0004 1759 700XDepartment of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Weidong Li
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Ting Chen
- Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China.
| | - Qingbo Xu
- Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
7
|
Wang J, An M, Haubner BJ, Penninger JM. Cardiac regeneration: Options for repairing the injured heart. Front Cardiovasc Med 2023; 9:981982. [PMID: 36712238 PMCID: PMC9877631 DOI: 10.3389/fcvm.2022.981982] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
Cardiac regeneration is one of the grand challenges in repairing injured human hearts. Numerous studies of signaling pathways and metabolism on cardiac development and disease pave the way for endogenous cardiomyocyte regeneration. New drug delivery approaches, high-throughput screening, as well as novel therapeutic compounds combined with gene editing will facilitate the development of potential cell-free therapeutics. In parallel, progress has been made in the field of cell-based therapies. Transplantation of human pluripotent stem cell (hPSC)-derived cardiomyocytes (hPSC-CMs) can partially rescue the myocardial defects caused by cardiomyocyte loss in large animals. In this review, we summarize current cell-based and cell-free regenerative therapies, discuss the importance of cardiomyocyte maturation in cardiac regenerative medicine, and envision new ways of regeneration for the injured heart.
Collapse
Affiliation(s)
- Jun Wang
- Department of Medical Genetics, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Meilin An
- Department of Medical Genetics, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Bernhard Johannes Haubner
- Department of Internal Medicine III (Cardiology and Angiology), Innsbruck Medical University, Innsbruck, Austria
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Josef M. Penninger
- Department of Medical Genetics, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC – Vienna BioCenter, Vienna, Austria
| |
Collapse
|
8
|
Role of smooth muscle progenitor cells in vascular mechanical injury and repair. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
9
|
Salerno N, Salerno L, Marino F, Scalise M, Chiefalo A, Panuccio G, De Angelis A, Cianflone E, Urbanek K, Torella D. Myocardial regeneration protocols towards the routine clinical scenario: An unseemly path from bench to bedside. EClinicalMedicine 2022; 50:101530. [PMID: 35799845 PMCID: PMC9253597 DOI: 10.1016/j.eclinm.2022.101530] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Heart failure secondary to cardiomyocyte loss and/or dysfunction is the number one killer worldwide. The field of myocardial regeneration with its far-reaching primary goal of cardiac remuscularization and its hard-to-accomplish translation from bench to bedside, has been filled with ups and downs, steps forward and steps backward, controversies galore and, unfortunately, scientific scandals. Despite the present morass in which cardiac remuscularization is stuck in, the search for clinically effective regenerative approaches remains keenly active. Starting with a concise overview of the still highly debated regenerative capacity of the adult mammalian heart, we focus on the main interventions, that have reached or are close to clinical use, critically discussing key findings, successes, and failures. Finally, some promising and innovative approaches for myocardial repair/regeneration still at the pre-clinical stage are discussed to offer a holistic view on the future of myocardial repair/regeneration for the prevention/management of heart failure in the clinical scenario. FUNDING This research was funded by Grants from the Ministry of University and Research PRIN2015 2015ZTT5KB_004; PRIN2017NKB2N4_005; PON-AIM - 1829805-2.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80125, Naples, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
- Corresponding author.
| |
Collapse
|
10
|
Garbern JC, Lee RT. Heart regeneration: 20 years of progress and renewed optimism. Dev Cell 2022; 57:424-439. [PMID: 35231426 PMCID: PMC8896288 DOI: 10.1016/j.devcel.2022.01.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide, and thus there remains great interest in regenerative approaches to treat heart failure. In the past 20 years, the field of heart regeneration has entered a renaissance period with remarkable progress in the understanding of endogenous heart regeneration, stem cell differentiation for exogenous cell therapy, and cell-delivery methods. In this review, we highlight how this new understanding can lead to viable strategies for human therapy. For the near term, drugs, electrical and mechanical devices, and heart transplantation will remain mainstays of cardiac therapies, but eventually regenerative therapies based on fundamental regenerative biology may offer more permanent solutions for patients with heart failure.
Collapse
Affiliation(s)
- Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, USA,Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, USA,Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115, USA,Corresponding author and lead contact: Richard T. Lee, Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave, Cambridge, MA 02138, Phone: 617-496-5394, Fax: 617-496-8351,
| |
Collapse
|
11
|
Jatho A, Zieseniss A, Brechtel-Curth K, Guo J, Böker KO, Salinas G, Wenger RH, Katschinski DM. The HIFα-Stabilizing Drug Roxadustat Increases the Number of Renal Epo-Producing Sca-1 + Cells. Cells 2022; 11:cells11040753. [PMID: 35203399 PMCID: PMC8869801 DOI: 10.3390/cells11040753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Inhibition of the prolyl-4-hydroxylase domain (PHD) enzymes, leading to the stabilization of hypoxia-inducible factor (HIF) α as well as to the stimulation of erythropoietin (Epo) synthesis, is the functional mechanism of the new anti-anemia drug roxadustat. Little is known about the effects of roxadustat on the Epo-producing cell pool. To gain further insights into the function of PHD inhibitors, we characterized the abundance of mesenchymal stem cell (MSC)-like cells after roxadustat treatment of mice. The number of Sca-1+ mesenchymal cells following roxadustat treatment increased exclusively in the kidneys. Isolated Sca-1+ cells demonstrated typical features of MSC-like cells, including adherence to tissue culture plates, trilineage differentiation potential, and expression of MSC markers. Kidney-derived Sca-1+ MSC-like cells were cultured for up to 21 days. Within the first few days in culture, cells stabilized HIF-1α and HIF-2α and temporarily increased Epo production upon incubation in hypoxia. In summary, we have identified a Sca-1+ MSC-like cell population that is involved in renal Epo production and might contribute to the strong anti-anemic effect of the PHD inhibitor roxadustat.
Collapse
Affiliation(s)
- Aline Jatho
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University Göttingen, 37073 Goettingen, Germany; (A.Z.); (K.B.-C.); (J.G.)
- Correspondence: (A.J.); (D.M.K.)
| | - Anke Zieseniss
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University Göttingen, 37073 Goettingen, Germany; (A.Z.); (K.B.-C.); (J.G.)
| | - Katja Brechtel-Curth
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University Göttingen, 37073 Goettingen, Germany; (A.Z.); (K.B.-C.); (J.G.)
| | - Jia Guo
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University Göttingen, 37073 Goettingen, Germany; (A.Z.); (K.B.-C.); (J.G.)
| | - Kai Oliver Böker
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen, Georg-August-University Göttingen, 37075 Goettingen, Germany;
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Center Göttingen, Georg-August-University Göttingen, 37073 Goettingen, Germany;
| | - Roland H. Wenger
- National Centre of Competence in Research “Kidney.CH”, 8057 Zurich, Switzerland;
- Institute of Physiology, University of Zürich, 8057 Zurich, Switzerland
| | - Dörthe M. Katschinski
- Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University Göttingen, 37073 Goettingen, Germany; (A.Z.); (K.B.-C.); (J.G.)
- Correspondence: (A.J.); (D.M.K.)
| |
Collapse
|
12
|
Cardiac regeneration following myocardial infarction: the need for regeneration and a review of cardiac stromal cell populations used for transplantation. Biochem Soc Trans 2022; 50:269-281. [PMID: 35129611 PMCID: PMC9042388 DOI: 10.1042/bst20210231] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/02/2022] [Accepted: 01/06/2022] [Indexed: 02/07/2023]
Abstract
Myocardial infarction is a leading cause of death globally due to the inability of the adult human heart to regenerate after injury. Cell therapy using cardiac-derived progenitor populations emerged about two decades ago with the aim of replacing cells lost after ischaemic injury. Despite early promise from rodent studies, administration of these populations has not translated to the clinic. We will discuss the need for cardiac regeneration and review the debate surrounding how cardiac progenitor populations exert a therapeutic effect following transplantation into the heart, including their ability to form de novo cardiomyocytes and the release of paracrine factors. We will also discuss limitations hindering the cell therapy field, which include the challenges of performing cell-based clinical trials and the low retention of administered cells, and how future research may overcome them.
Collapse
|
13
|
Wang Y, Chen J, Cowan DB, Wang DZ. Non-coding RNAs in cardiac regeneration: Mechanism of action and therapeutic potential. Semin Cell Dev Biol 2021; 118:150-162. [PMID: 34284952 PMCID: PMC8434979 DOI: 10.1016/j.semcdb.2021.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/19/2022]
Abstract
In the past two decades, thousands of non-coding RNAs (ncRNAs) have been discovered, annotated, and characterized in nearly every tissue under both physiological and pathological conditions. Here, we will focus on the role of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) in ischemic heart disease (IHD), which remains the leading cause of morbidity and mortality in humans-resulting in 8.9 million deaths annually. Cardiomyocyte (CM) proliferation, differentiation, and survival in addition to neovascularization of injured tissues and the prevention of fibrosis are commonly regarded as critically important for the recovery of the heart following myocardial infarction (MI). An abundance of evidence has been accumulated to show ncRNAs participate in cardiac recovery after MI. Because miRNAs are important regulators of cardiac regeneration, the therapeutic potential of at least five of these molecules has been assessed in large animal models of human IHD. In particular, miRNA-based interventions based on miR-132 and miR-92a inhibition in related diseases have displayed favorable outcomes that have provided the impetus for miRNA-based clinical trials for IHD. At the same time, the functional roles of lncRNAs and circRNAs in cardiac regeneration are also being explored. In the present review, we will summarize the latest ncRNA studies aimed at reversing damage to the ischemic heart and discuss the therapeutic potential of targeting miRNAs, lncRNAs, and circRNAs to stimulate cardiac regeneration.
Collapse
Affiliation(s)
- Yi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jinghai Chen
- Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University, 268 Kaixuan Road, Hangzhou, China
| | - Douglas B Cowan
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Kasai-Brunswick TH, Carvalho AB, Campos de Carvalho AC. Stem cell therapies in cardiac diseases: Current status and future possibilities. World J Stem Cells 2021; 13:1231-1247. [PMID: 34630860 PMCID: PMC8474720 DOI: 10.4252/wjsc.v13.i9.1231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases represent the world's leading cause of death. In this heterogeneous group of diseases, ischemic cardiomyopathies are the most devastating and prevalent, estimated to cause 17.9 million deaths per year. Despite all biomedical efforts, there are no effective treatments that can replace the myocytes lost during an ischemic event or progression of the disease to heart failure. In this context, cell therapy is an emerging therapeutic alternative to treat cardiovascular diseases by cell administration, aimed at cardiac regeneration and repair. In this review, we will cover more than 30 years of cell therapy in cardiology, presenting the main milestones and drawbacks in the field and signaling future challenges and perspectives. The outcomes of cardiac cell therapies are discussed in three distinct aspects: The search for remuscularization by replacement of lost cells by exogenous adult cells, the endogenous stem cell era, which pursued the isolation of a progenitor with the ability to induce heart repair, and the utilization of pluripotent stem cells as a rich and reliable source of cardiomyocytes. Acellular therapies using cell derivatives, such as microvesicles and exosomes, are presented as a promising cell-free therapeutic alternative.
Collapse
Affiliation(s)
- Tais Hanae Kasai-Brunswick
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Adriana Bastos Carvalho
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Antonio Carlos Campos de Carvalho
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil.
| |
Collapse
|
15
|
Zhu D, Cheng K. Cardiac Cell Therapy for Heart Repair: Should the Cells Be Left Out? Cells 2021; 10:641. [PMID: 33805763 PMCID: PMC7999733 DOI: 10.3390/cells10030641] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is still the leading cause of death worldwide. Coronary artery occlusion, or myocardial infarction (MI) causes massive loss of cardiomyocytes. The ischemia area is eventually replaced by a fibrotic scar. From the mechanical dysfunctions of the scar in electronic transduction, contraction and compliance, pathological cardiac dilation and heart failure develops. Once end-stage heart failure occurs, the only option is to perform heart transplantation. The sequential changes are termed cardiac remodeling, and are due to the lack of endogenous regenerative actions in the adult human heart. Regenerative medicine and biomedical engineering strategies have been pursued to repair the damaged heart and to restore normal cardiac function. Such strategies include both cellular and acellular products, in combination with biomaterials. In addition, substantial progress has been made to elucidate the molecular and cellular mechanisms underlying heart repair and regeneration. In this review, we summarize and discuss current therapeutic approaches for cardiac repair and provide a perspective on novel strategies that holding potential opportunities for future research and clinical translation.
Collapse
Affiliation(s)
- Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA;
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
16
|
Deng J, Ni Z, Gu W, Chen Q, Nowak WN, Chen T, Issa Bhaloo S, Zhang Z, Hu Y, Zhou B, Zhang L, Xu Q. Single-cell gene profiling and lineage tracing analyses revealed novel mechanisms of endothelial repair by progenitors. Cell Mol Life Sci 2020; 77:5299-5320. [PMID: 32166394 PMCID: PMC11104897 DOI: 10.1007/s00018-020-03480-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/18/2020] [Accepted: 02/07/2020] [Indexed: 12/20/2022]
Abstract
Stem/progenitor cells (SPCs) have been implicated to participate in vascular repair. However, the exact role of SPCs in endothelial repair of large vessels still remains controversial. This study aimed to delineate the cellular heterogeneity and possible functional role of endogenous vascular SPCs in large vessels. Using single-cell RNA-sequencing (scRNA-seq) and genetic lineage tracing mouse models, we uncovered the cellular heterogeneity of SPCs, i.e., c-Kit+ cells in the mouse aorta, and found that endogenous c-Kit+ cells acquire endothelial cell fate in the aorta under both physiological and pathological conditions. While c-Kit+ cells contribute to aortic endothelial turnover in the atheroprone regions during homeostasis, recipient c-Kit+ cells of nonbone marrow source replace both luminal and microvessel endothelial cells in transplant arteriosclerosis. Single-cell pseudotime analysis of scRNA-seq data and in vitro cell experiments suggest that vascular SPCs display endothelial differentiation potential and undergo metabolic reprogramming during cell differentiation, in which AKT/mTOR-dependent glycolysis is critical for endothelial gene expression. These findings demonstrate a critical role for c-Kit lineage cells in aortic endothelial turnover and replacement, and may provide insights into therapeutic strategies for vascular diseases.
Collapse
Affiliation(s)
- Jiacheng Deng
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Zhichao Ni
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Wenduo Gu
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Qishan Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Witold Norbert Nowak
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Shirin Issa Bhaloo
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Zhongyi Zhang
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Yanhua Hu
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai, 200031, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
- School of Cardiovascular Medicine and Science, BHF Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
17
|
Li N, Rignault-Clerc S, Bielmann C, Bon-Mathier AC, Déglise T, Carboni A, Ducrest M, Rosenblatt-Velin N. Increasing heart vascularisation after myocardial infarction using brain natriuretic peptide stimulation of endothelial and WT1 + epicardial cells. eLife 2020; 9:61050. [PMID: 33245046 PMCID: PMC7695454 DOI: 10.7554/elife.61050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Brain natriuretic peptide (BNP) treatment increases heart function and decreases heart dilation after myocardial infarction (MI). Here, we investigated whether part of the cardioprotective effect of BNP in infarcted hearts related to improved neovascularisation. Infarcted mice were treated with saline or BNP for 10 days. BNP treatment increased vascularisation and the number of endothelial cells in all areas of infarcted hearts. Endothelial cell lineage tracing showed that BNP directly stimulated the proliferation of resident endothelial cells via NPR-A binding and p38 MAP kinase activation. BNP also stimulated the proliferation of WT1+ epicardium-derived cells but only in the hypoxic area of infarcted hearts. Our results demonstrated that these immature cells have a natural capacity to differentiate into endothelial cells in infarcted hearts. BNP treatment increased their proliferation but not their differentiation capacity. We identified new roles for BNP that hold potential for new therapeutic strategies to improve recovery and clinical outcome after MI.
Collapse
Affiliation(s)
- Na Li
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Stephanie Rignault-Clerc
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Christelle Bielmann
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Anne-Charlotte Bon-Mathier
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Tamara Déglise
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Alexia Carboni
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Mégane Ducrest
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Nathalie Rosenblatt-Velin
- Division of Angiology, Heart and Vessel Department, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Christensen R, Gunnarsson AP, Jensen UB. The role of stem cell antigen-1/Lymphocyte antigen 6A-2/6E-1 knock out in murine epidermis. Stem Cell Res 2020; 49:102047. [PMID: 33157392 DOI: 10.1016/j.scr.2020.102047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 11/27/2022] Open
Abstract
Stem Cell Antigen-1 (SCA-1) is a central positive marker for isolating stem cells in several tissues in the mouse. However, for the epidermis, this appears to be the opposite since lack of SCA-1 has been shown to identify keratinocyte populations with progenitor characteristics. This study investigates the effect of SCA-1 knockout in murine keratinocytes. We compared Sca-1EGFP/EGFP knockout and wildtype mice with respect to the three-dimensional morphology of the epidermis, performed functional assays, and generated gene expression profiles on FACS sorted cells. There were no morphological abnormalities on skin, fur, or hair follicles in transgenic knockout mice compared to wild type mice. SCA-1 knockout keratinocytes showed significantly reduced colony-forming efficiency, colony size and proliferation rate in vitro, however, SCA-1 knockout did not alter wound healing efficiency or keratinocyte proliferation rate in vivo. Moreover, gene expression profiling shows that the effect from knockout of SCA-1 in keratinocytes is dissimilar from what has been observed in other tissues. Additionally, tumor assay indicated that SCA-1 knockout decreases the number of formed papillomas. The results indicate a more complex role for SCA-1, which might differ between epidermal keratinocytes during homeostasis and activated conditions.
Collapse
Affiliation(s)
- Rikke Christensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| | - Anders Patrik Gunnarsson
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark.
| |
Collapse
|
19
|
Parekh KR, Nawroth J, Pai A, Busch SM, Senger CN, Ryan AL. Stem cells and lung regeneration. Am J Physiol Cell Physiol 2020; 319:C675-C693. [PMID: 32783658 PMCID: PMC7654650 DOI: 10.1152/ajpcell.00036.2020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022]
Abstract
The ability to replace defective cells in an airway with cells that can engraft, integrate, and restore a functional epithelium could potentially cure a number of lung diseases. Progress toward the development of strategies to regenerate the adult lung by either in vivo or ex vivo targeting of endogenous stem cells or pluripotent stem cell derivatives is limited by our fundamental lack of understanding of the mechanisms controlling human lung development, the precise identity and function of human lung stem and progenitor cell types, and the genetic and epigenetic control of human lung fate. In this review, we intend to discuss the known stem/progenitor cell populations, their relative differences between rodents and humans, their roles in chronic lung disease, and their therapeutic prospects. Additionally, we highlight the recent breakthroughs that have increased our understanding of these cell types. These advancements include novel lineage-traced animal models and single-cell RNA sequencing of human airway cells, which have provided critical information on the stem cell subtypes, transition states, identifying cell markers, and intricate pathways that commit a stem cell to differentiate or to maintain plasticity. As our capacity to model the human lung evolves, so will our understanding of lung regeneration and our ability to target endogenous stem cells as a therapeutic approach for lung disease.
Collapse
Affiliation(s)
- Kalpaj R Parekh
- Department Surgery, Division of Cardiothoracic Surgery, University of Iowa, Iowa City, Iowa
| | - Janna Nawroth
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, California
| | - Albert Pai
- Department Surgery, Division of Cardiothoracic Surgery, University of Iowa, Iowa City, Iowa
| | - Shana M Busch
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, California
| | - Christiana N Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, California
| | - Amy L Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, California
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
20
|
Constantinou C, Miranda AMA, Chaves P, Bellahcene M, Massaia A, Cheng K, Samari S, Rothery SM, Chandler AM, Schwarz RP, Harding SE, Punjabi P, Schneider MD, Noseda M. Human pluripotent stem cell-derived cardiomyocytes as a target platform for paracrine protection by cardiac mesenchymal stromal cells. Sci Rep 2020; 10:13016. [PMID: 32747668 PMCID: PMC7400574 DOI: 10.1038/s41598-020-69495-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic heart disease remains the foremost cause of death globally, with survivors at risk for subsequent heart failure. Paradoxically, cell therapies to offset cardiomyocyte loss after ischemic injury improve long-term cardiac function despite a lack of durable engraftment. An evolving consensus, inferred preponderantly from non-human models, is that transplanted cells benefit the heart via early paracrine signals. Here, we tested the impact of paracrine signals on human cardiomyocytes, using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as the target of mouse and human cardiac mesenchymal stromal cells (cMSC) with progenitor-like features. In co-culture and conditioned medium studies, cMSCs markedly inhibited human cardiomyocyte death. Little or no protection was conferred by mouse tail tip or human skin fibroblasts. Consistent with the results of transcriptomic profiling, functional analyses showed that the cMSC secretome suppressed apoptosis and preserved cardiac mitochondrial transmembrane potential. Protection was independent of exosomes under the conditions tested. In mice, injecting cMSC-conditioned media into the infarct border zone reduced apoptotic cardiomyocytes > 70% locally. Thus, hPSC-CMs provide an auspicious, relevant human platform to investigate extracellular signals for cardiac muscle survival, substantiating human cardioprotection by cMSCs, and suggesting the cMSC secretome or its components as potential cell-free therapeutic products.
Collapse
Affiliation(s)
- Chrystalla Constantinou
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Antonio M A Miranda
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Patricia Chaves
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Mohamed Bellahcene
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Andrea Massaia
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Kevin Cheng
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Sara Samari
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Stephen M Rothery
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Anita M Chandler
- Kardia Therapeutics, Houston, TX, 77030, USA
- Department of Bioengineering, BioScience Research Collaborative, Rice University, Houston, TX, 77005, USA
| | - Richard P Schwarz
- Kardia Therapeutics, Houston, TX, 77030, USA
- CV Ventures, LLC, Blue Bell, PA, 19422, USA
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
| | - Prakash Punjabi
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
| | - Michael D Schneider
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK.
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK.
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, W12 0NN, UK.
- British Heart Foundation Centre for Research Excellence and Centre for Regenerative Medicine, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
21
|
He L, Nguyen NB, Ardehali R, Zhou B. Heart Regeneration by Endogenous Stem Cells and Cardiomyocyte Proliferation: Controversy, Fallacy, and Progress. Circulation 2020; 142:275-291. [PMID: 32687441 DOI: 10.1161/circulationaha.119.045566] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ischemic heart disease is the leading cause of death worldwide. Myocardial infarction results in an irreversible loss of cardiomyocytes with subsequent adverse remodeling and heart failure. Identifying new sources for cardiomyocytes and promoting their formation represents a goal of cardiac biology and regenerative medicine. Within the past decade, many types of putative cardiac stem cells (CSCs) have been reported to regenerate the injured myocardium by differentiating into new cardiomyocytes. Some of these CSCs have been translated from bench to bed with reported therapeutic effectiveness. However, recent basic research studies on stem cell tracing have begun to question their fundamental biology and mechanisms of action, raising serious concerns over the myogenic potential of CSCs. We review the history of different types of CSCs within the past decade and provide an update of recent cell tracing studies that have challenged the origin and existence of CSCs. In addition to the potential role of CSCs in heart regeneration, proliferation of preexisting cardiomyocytes has recently gained more attention. This review will also evaluate the methodologic and technical aspects of past and current studies on CSCs and cardiomyocyte proliferation, with emphasis on technical strengths, advantages, and potential limitations of research approaches. While our understanding of cardiomyocyte generation and regeneration continues to evolve, it is important to address the shortcomings and inaccuracies in this field. This is best achieved by embracing technological advancements and improved methods to label single cardiomyocytes/progenitors and accurately investigate their developmental potential and fate/lineage commitment.
Collapse
Affiliation(s)
- Lingjuan He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China (L.H., B.Z.)
| | - Ngoc B Nguyen
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine (N.B.N., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (N.B.N., R.A.), University of California, Los Angeles
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine (N.B.N., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (N.B.N., R.A.), University of California, Los Angeles
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China (L.H., B.Z.).,School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.).,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China (B.Z.).,Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.)
| |
Collapse
|
22
|
Hao C, Lu Z, Zhao Y, Chen Z, Shen C, Ma G, Chen L. Overexpression of GATA4 enhances the antiapoptotic effect of exosomes secreted from cardiac colony-forming unit fibroblasts via miRNA221-mediated targeting of the PTEN/PI3K/AKT signaling pathway. Stem Cell Res Ther 2020; 11:251. [PMID: 32586406 PMCID: PMC7318537 DOI: 10.1186/s13287-020-01759-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/25/2020] [Accepted: 06/04/2020] [Indexed: 01/04/2023] Open
Abstract
Background GATA4 is an early cardiac-specific transcription factor, and endogenous GATA4-positive cells play a critical role in cardioprotection after myocardial injury. As functional paracrine units of therapeutic cells, exosomes can partially reproduce the reparative properties of their parental cells. Here, we investigated the cardioprotective capabilities of exosomes derived from cardiac colony-forming unit fibroblasts (cCFU-Fs) overexpressing GATA4 (cCFU-FsGATA4) and the underlying mechanism through which these exosomes use microRNA (miRNA) delivery to regulate target proteins in myocardial infarction (MI). Methods Exosomes were harvested from cCFU-Fs by ultracentrifugation. miRNA arrays were performed to determine differential miRNA expression between exosomes derived from cCFU-FsGATA4 (GATA4-Exo) and control cCFU-Fs (NC-Exo). A dual-luciferase reporter assay confirmed that miR221 directly targets the 3′ untranslated region (UTR) of the phosphatase and tensin homolog on chromosome ten (PTEN) gene. Cardiac function and myocardial infarct size were evaluated by echocardiography and Masson trichrome staining, respectively. Results Compared with NC-Exo, GATA4-Exo increased the survival and reduced the apoptosis of H9c2 cells. Direct intramyocardial transplantation of GATA4-Exo at the border of the ischemic region following ligation of the left anterior descending (LAD) coronary artery significantly restored cardiac contractile function and reduced infarct size. Microarray analysis revealed significantly increased miR221 expression in GATA4-Exo. qPCR confirmed higher miR221 levels in H9c2 cells treated with GATA4-Exo than in those treated with NC-Exo. miR221 mimic-transfected H9c2 cells demonstrated a significantly higher survival rate following exposure to hypoxic conditions than those transfected with miR221 inhibitor. A dual-luciferase reporter gene assay confirmed the PTEN gene as a target of miR221. Western blot analysis showed that H9c2 cells treated with GATA4-Exo exhibited lower PTEN protein expression and higher p-Akt expression. Conclusion GATA4 overexpression enhances the protective effect of cCFU-F-derived exosomes on myocardial ischemic injury. In terms of the mechanism, it is at least partly due to the miR221 transferred by GATA4-Exo, which inhibits PTEN expression, activates the phosphatidylinositol 3 kinase (PI3K)/AKT signaling pathway, and subsequently alleviates apoptosis of myocardial cells (CMs).
Collapse
Affiliation(s)
- Chunshu Hao
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.,Medical School of Southeast University, Nanjing, China
| | - Zhengri Lu
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.,Medical School of Southeast University, Nanjing, China
| | - Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.,Medical School of Southeast University, Nanjing, China
| | - Zhong Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, China.
| |
Collapse
|
23
|
Soliman H, Rossi FMV. Cardiac fibroblast diversity in health and disease. Matrix Biol 2020; 91-92:75-91. [PMID: 32446910 DOI: 10.1016/j.matbio.2020.05.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
The cardiac stroma plays essential roles in health and following cardiac damage. The major player of the stroma with respect to extracellular matrix deposition, maintenance and remodeling is the poorly defined fibroblast. It has long been recognized that there is considerable variability to the fibroblast phenotype. With the advent of new, high throughput analytical methods our understanding and appreciation of this heterogeneity has grown dramatically. This review aims to explore the diversity of cardiac fibroblasts and highlights new insights into the diverse nature of these cells and their progenitors as revealed by single cell sequencing and fate mapping studies. We propose that at least in part the observed heterogeneity is related to the existence of a differentiation cascade within stromal cells. Beyond in-organ heterogeneity, we also discuss how the stromal response to damage differs between non-regenerating organs such as the heart and regenerating organs such as skeletal muscle. In exploring possible causes for these differences, we outline that although fibrogenic cells from different organs overlap in many properties, they still possess organ-specific transcriptional signatures and differentiation biases that make them functionally distinct.
Collapse
Affiliation(s)
- Hesham Soliman
- Biomedical Research Centre, University of British Columbia, Vancouver, Canada; School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada; Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Fabio M V Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, Canada; School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada.
| |
Collapse
|
24
|
Maghin E, Garbati P, Quarto R, Piccoli M, Bollini S. Young at Heart: Combining Strategies to Rejuvenate Endogenous Mechanisms of Cardiac Repair. Front Bioeng Biotechnol 2020; 8:447. [PMID: 32478060 PMCID: PMC7237726 DOI: 10.3389/fbioe.2020.00447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
True cardiac regeneration of the injured heart has been broadly described in lower vertebrates by active replacement of lost cardiomyocytes to functionally and structurally restore the myocardial tissue. On the contrary, following severe injury (i.e., myocardial infarction) the adult mammalian heart is endowed with an impaired reparative response by means of meager wound healing program and detrimental remodeling, which can lead over time to cardiomyopathy and heart failure. Lately, a growing body of basic, translational and clinical studies have supported the therapeutic use of stem cells to provide myocardial regeneration, with the working hypothesis that stem cells delivered to the cardiac tissue could result into new cardiovascular cells to replenish the lost ones. Nevertheless, multiple independent evidences have demonstrated that injected stem cells are more likely to modulate the cardiac tissue via beneficial paracrine effects, which can enhance cardiac repair and reinstate the embryonic program and cell cycle activity of endogenous cardiac stromal cells and resident cardiomyocytes. Therefore, increasing interest has been addressed to the therapeutic profiling of the stem cell-derived secretome (namely the total of cell-secreted soluble factors), with specific attention to cell-released extracellular vesicles, including exosomes, carrying cardioprotective and regenerative RNA molecules. In addition, the use of cardiac decellularized extracellular matrix has been recently suggested as promising biomaterial to develop novel therapeutic strategies for myocardial repair, as either source of molecular cues for regeneration, biological scaffold for cardiac tissue engineering or biomaterial platform for the functional release of factors. In this review, we will specifically address the translational relevance of these two approaches with ad hoc interest in their feasibility to rejuvenate endogenous mechanisms of cardiac repair up to functional regeneration.
Collapse
Affiliation(s)
- Edoardo Maghin
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.,Department of Women's and Children Health, University of Padova, Padua, Italy
| | - Patrizia Garbati
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Rodolfo Quarto
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy.,UOC Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Martina Piccoli
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
25
|
Balbi C, Costa A, Barile L, Bollini S. Message in a Bottle: Upgrading Cardiac Repair into Rejuvenation. Cells 2020; 9:cells9030724. [PMID: 32183455 PMCID: PMC7140681 DOI: 10.3390/cells9030724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Ischaemic cardiac disease is associated with a loss of cardiomyocytes and an intrinsic lack of myocardial renewal. Recent work has shown that the heart retains limited cardiomyocyte proliferation, which remains inefficient when facing pathological conditions. While broadly active in the neonatal mammalian heart, this mechanism becomes quiescent soon after birth, suggesting loss of regenerative potential with maturation into adulthood. A key question is whether this temporary regenerative window can be enhanced via appropriate stimulation and further extended. Recently the search for novel therapeutic approaches for heart disease has centred on stem cell biology. The “paracrine effect” has been proposed as a promising strategy to boost endogenous reparative and regenerative mechanisms from within the cardiac tissue by exploiting the modulatory potential of soluble stem cell-secreted factors. As such, growing interest has been specifically addressed towards stem/progenitor cell-secreted extracellular vesicles (EVs), which can be easily isolated in vitro from cell-conditioned medium. This review will provide a comprehensive overview of the current paradigm on cardiac repair and regeneration, with a specific focus on the role and mechanism(s) of paracrine action of EVs from cardiac stromal progenitors as compared to exogenous stem cells in order to discuss the optimal choice for future therapy. In addition, the challenges to overcoming translational EV biology from bench to bedside for future cardiac regenerative medicine will be discussed.
Collapse
Affiliation(s)
- Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland;
| | - Ambra Costa
- Regenerative Medicine Laboratory, Dept. of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Correspondence: (L.B.); (S.B.)
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Dept. of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
- Correspondence: (L.B.); (S.B.)
| |
Collapse
|
26
|
Chen K, Huang Y, Singh R, Wang ZZ. Arrhythmogenic risks of stem cell replacement therapy for cardiovascular diseases. J Cell Physiol 2020; 235:6257-6267. [PMID: 31994198 DOI: 10.1002/jcp.29554] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Ischemic heart disease and congestive heart failure are major contributors to high morbidity and mortality. Approximately 1.5 million cases of myocardial infarction occur annually in the United States; the yearly incidence rate is approximately 600 cases per 100,000 people. Although significant progress to improve the survival rate has been made by medications and implantable medical devices, damaged cardiomyocytes are unable to be recovered by current treatment strategies. After almost two decades of research, stem cell therapy has become a very promising approach to generate new cardiomyocytes and enhance the function of the heart. Along with clinical trials with stem cells conducted in cardiac regeneration, concerns regarding safety and potential risks have emerged. One of the contentious issues is the electrical dysfunctions of cardiomyocytes and cardiac arrhythmia after stem cell therapy. In this review, we focus on the cell sources currently used for stem cell therapy and discuss related arrhythmogenic risk.
Collapse
Affiliation(s)
- Kang Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Huang
- Department of Medicine, University of Maryland Medical Center Midtown Campus, Baltimore, Maryland
| | - Radhika Singh
- Center for Biotechnology Education, Johns Hopkins University, Baltimore, Maryland
| | - Zack Z Wang
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
27
|
Halver J, Wenzel K, Sendker J, Carrillo García C, Erdelmeier CAJ, Willems E, Mercola M, Symma N, Könemann S, Koch E, Hensel A, Schade D. Crataegus Extract WS®1442 Stimulates Cardiomyogenesis and Angiogenesis From Stem Cells: A Possible New Pharmacology for Hawthorn? Front Pharmacol 2019; 10:1357. [PMID: 31849643 PMCID: PMC6902660 DOI: 10.3389/fphar.2019.01357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022] Open
Abstract
Extracts from the leaves and flowers of Crataegus spp. (i.e., hawthorn species) have been traditionally used with documented preclinical and clinical activities in cardiovascular medicine. Based on reported positive effects on heart muscle after ischemic injury and the overall cardioprotective profile, the present study addressed potential contributions of Crataegus extracts to cardiopoietic differentiation from stem cells. The quantified Crataegus extract WS®1442 stimulated cardiomyogenesis from murine and human embryonic stem cells (ESCs). Mechanistically, this effect was found to be induced by promoting differentiation of cardiovascular progenitor cell populations but not by proliferation. Bioassay-guided fractionation, phytochemical and analytical profiling suggested high-molecular weight ingredients as the active principle with at least part of the activity due to oligomeric procyanidines (OPCs) with a degree of polymerization between 3 and 6 (DP3–6). Transcriptome profiling in mESCs suggested two main, plausible mechanisms: These were early, stress-associated cellular events along with the modulation of distinct developmental pathways, including the upregulation of brain-derived neurotrophic factor (BDNF) and retinoic acid as well as the inhibition of transforming growth factor β/bone morphogenetic protein (TGFβ/BMP) and fibroblast growth factor (FGF) signaling. In addition, WS®1442 stimulated angiogenesis ex vivo in Sca-1+ progenitor cells from adult mice hearts. These in vitro data provide evidence for a differentiation promoting activity of WS®1442 on distinct cardiovascular stem/progenitor cells that could be valuable for therapeutic heart regeneration after myocardial infarction. However, the in vivo relevance of this new pharmacological activity of Crataegus spp. remains to be investigated and active ingredients from bioactive fractions will have to be further characterized.
Collapse
Affiliation(s)
- Jonas Halver
- Department of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Kristin Wenzel
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,Partner site Greifswald, DZHK, German Centre for Cardiovascular Research, Greifswald, Germany
| | - Jandirk Sendker
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Carmen Carrillo García
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | - Erik Willems
- Muscle Development and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, United States
| | - Mark Mercola
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, United States
| | - Nico Symma
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Stephanie Könemann
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,Partner site Greifswald, DZHK, German Centre for Cardiovascular Research, Greifswald, Germany
| | - Egon Koch
- Preclinical Research, Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Dennis Schade
- Department of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany.,Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Christian-Albrechts-University of Kiel, Kiel, Germany.,Partner site Kiel, DZHK, German Centre for Cardiovascular Research, Kiel, Germany
| |
Collapse
|
28
|
Tassone NM, Li B, Patel MS, Devine MY, Firmiss PR, Gould AD, Kochan KS, Stubbee RA, Bowen DK, Dettman RW, Gong EM. Stem cell antigen/Ly6a protects against bladder fibrosis in mice. Am J Physiol Renal Physiol 2019; 317:F1503-F1512. [PMID: 31532245 DOI: 10.1152/ajprenal.00160.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have defined a population of stem cell antigen (Sca)-1+/CD34+/lin- mesenchymal stem cells in the mouse urinary bladder. These cells are reduced after partial bladder outlet obstruction (PO). To test the role of Sca-1 expressed by these cells, we analyzed bladders from Sca-1 knockout (KO) mice in both uninjured male mice and male mice subjected to PO. We found that loss of Sca-1 alone had little effect on bladder development or function but reduced the total number of mesenchymal stem cells by 30%. After PO, bladders from Sca-1-null KO male mice were larger, with more collagen and less muscle, than obstructed wild-type mice. Steady-state levels of caldesmon were significantly reduced and levels of fibroblast-specific protein 1 were significantly increased in Sca-1 KO mice compared with wild-type mice after PO. In investigating the effects of PO on cell proliferation, we found that loss of Sca-1 changed the timing of cell division in CD34+/lin-, collagen-producing, and smooth muscle cells. PO in combination with loss of Sca-1 drastically reduced the ability of CD34+/lin- cells to form colonies in vitro. Our findings therefore support the hypothesis that Sca-1 protects the bladder from fibrotic remodeling after obstruction, in part by influencing the proliferation of cells responding to the injury.
Collapse
Affiliation(s)
- Nicholas M Tassone
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Belinda Li
- Department of Urology, Loyola University Health System, Maywood, Illinois
| | - Mehul S Patel
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Megan Y Devine
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Paula R Firmiss
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Andrew D Gould
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Kirsten S Kochan
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Reid A Stubbee
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Diana K Bowen
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Robert W Dettman
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Edward M Gong
- Pediatric Urology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
29
|
Li Y, Lv Z, He L, Huang X, Zhang S, Zhao H, Pu W, Li Y, Yu W, Zhang L, Liu X, Liu K, Tang J, Tian X, Wang QD, Lui KO, Zhou B. Genetic Tracing Identifies Early Segregation of the Cardiomyocyte and Nonmyocyte Lineages. Circ Res 2019; 125:343-355. [PMID: 31185811 DOI: 10.1161/circresaha.119.315280] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE The developing heart is composed of cardiomyocytes and noncardiomyocytes since the early stage. It is generally believed that noncardiomyocytes including the cardiac progenitors contribute to new cardiomyocytes of the looping heart. However, it remains unclear what the cellular dynamics of nonmyocyte to cardiomyocyte conversion are and when the lineage segregation occurs during development. It also remains unknown whether nonmyocyte to cardiomyocyte conversion contributes to neonatal heart regeneration. OBJECTIVE We quantify the lineage conversion of noncardiomyocytes to cardiomyocytes in the embryonic and neonatal hearts and determine when the 2 cell lineages segregate during heart development. Moreover, we directly test if nonmyocyte to cardiomyocyte conversion contributes to neonatal heart regeneration. METHODS AND RESULTS We generated a dual genetic lineage tracing strategy in which cardiomyocytes and noncardiomyocytes of the developing heart could be simultaneously labeled by 2 orthogonal recombination systems. Genetic fate mapping showed that nonmyocyte to cardiomyocyte conversion peaks at E8.0 (embryonic day) to E8.5 and gradually declines at E9.5 and E10.5. Noncardiomyocytes do not generate any cardiomyocyte at and beyond E11.5 to E12.5. In the neonatal heart, noncardiomyocytes also do not contribute to any new cardiomyocyte in homeostasis or after injury. CONCLUSIONS Noncardiomyocytes contribute to new cardiomyocytes of the developing heart at early embryonic stage before E11.5. The noncardiomyocyte and cardiomyocyte lineage segregation occurs between E10.5 and E11.5, which is maintained afterward even during neonatal heart regeneration.
Collapse
Affiliation(s)
- Yan Li
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Zan Lv
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Lingjuan He
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Xiuzhen Huang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Shaohua Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Huan Zhao
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Wenjuan Pu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Yi Li
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Wei Yu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Libo Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Xiuxiu Liu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Kuo Liu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.).,School of Life Science and Technology, ShanghaiTech University, China (K.L., B.Z.)
| | - Juan Tang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.)
| | - Xueying Tian
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China (X.T., B.Z.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Early Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.-D.W.)
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China (K.O.L.)
| | - Bin Zhou
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (Yan Li, Z.L., L.H., X.H., S.Z., H.Z., W.P., Yi Li, W.Y., L.Z., X.L., K.L., J.T., B.Z.).,School of Life Science and Technology, ShanghaiTech University, China (K.L., B.Z.).,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China (X.T., B.Z.).,The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (B.Z.).,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China (B.Z.)
| |
Collapse
|
30
|
Oxygen as a key regulator of cardiomyocyte proliferation: New results about cell culture conditions! BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118460. [PMID: 30885672 DOI: 10.1016/j.bbamcr.2019.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/21/2019] [Accepted: 03/13/2019] [Indexed: 01/16/2023]
Abstract
The goal of the new therapeutically strategies aimed to treat cardiovascular diseases (CVDs) is to enhance the natural ability of the heart to regenerate. This represents a great challenge for the coming years as all the mechanisms underlying the replacement of dying cells by functional cells of the same type are not completely elucidated. Among these, stimulating cardiomyocyte proliferation seems to be crucial for the restoration of normal cardiac function after CVDs. In this review, we summarized the recent advances about the modulation of cardiomyocyte proliferation in physiological (during ageing) and pathological conditions. We highlighted the role of oxygen and we presented new results demonstrating that performing neonatal cardiomyocyte cell cultures in "normoxic" oxygen conditions (i.e. 3% oxygen) increases their proliferation rate, when compared to "hyperoxic" conventional conditions (i.e. 20% oxygen). Thus, oxygen concentration seems to be a key factor in the control of cardiomyocyte proliferation.
Collapse
|
31
|
Affiliation(s)
- Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA, and the Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|