1
|
Bornhorst D, Hejjaji AV, Steuter L, Woodhead NM, Maier P, Gentile A, Alhajkadour A, Santis Larrain O, Weber M, Kikhi K, Guenther S, Huisken J, Tamplin OJ, Stainier DYR, Gunawan F. The heart is a resident tissue for hematopoietic stem and progenitor cells in zebrafish. Nat Commun 2024; 15:7589. [PMID: 39217144 PMCID: PMC11366026 DOI: 10.1038/s41467-024-51920-7] [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: 12/01/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The contribution of endocardial cells (EdCs) to the hematopoietic lineages has been strongly debated. Here, we provide evidence that in zebrafish, the endocardium gives rise to and maintains a stable population of hematopoietic cells. Using single-cell sequencing, we identify an endocardial subpopulation expressing enriched levels of hematopoietic-promoting genes. High-resolution microscopy and photoconversion tracing experiments uncover hematopoietic cells, mainly hematopoietic stem and progenitor cells (HSPCs)/megakaryocyte-erythroid precursors (MEPs), derived from EdCs as well as the dorsal aorta stably attached to the endocardium. Emergence of HSPCs/MEPs in hearts cultured ex vivo without external hematopoietic sources, as well as longitudinal imaging of the beating heart using light sheet microscopy, support endocardial contribution to hematopoiesis. Maintenance of these hematopoietic cells depends on the adhesion factors Integrin α4 and Vcam1 but is at least partly independent of cardiac trabeculation or shear stress. Finally, blocking primitive erythropoiesis increases cardiac-residing hematopoietic cells, suggesting that the endocardium is a hematopoietic reservoir. Altogether, these studies uncover the endocardium as a resident tissue for HSPCs/MEPs and a de novo source of hematopoietic cells.
Collapse
Affiliation(s)
- Dorothee Bornhorst
- Institute of Cell Biology, Faculty of Medicine, University of Münster, Münster, 48149, Germany
- 'Cells-in-Motion' Interfaculty Center, University of Münster, Münster, 48149, Germany
| | - Amulya V Hejjaji
- Institute of Cell Biology, Faculty of Medicine, University of Münster, Münster, 48149, Germany
- 'Cells-in-Motion' Interfaculty Center, University of Münster, Münster, 48149, Germany
| | - Lena Steuter
- Institute of Cell Biology, Faculty of Medicine, University of Münster, Münster, 48149, Germany
- 'Cells-in-Motion' Interfaculty Center, University of Münster, Münster, 48149, Germany
| | - Nicole M Woodhead
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Paul Maier
- Multiscale Biology, Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, 37077, Germany
| | - Alessandra Gentile
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research (MPI-HLR), Bad Nauheim, 61231, Germany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alice Alhajkadour
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Octavia Santis Larrain
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Michael Weber
- Multiscale Biology, Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, 37077, Germany
| | - Khrievono Kikhi
- Flow Cytometry and Cell Sorting Core Facility, MPI-HLR, Bad Nauheim, 61231, Germany
| | - Stefan Guenther
- Deep Sequencing Platform, MPI-HLR, Bad Nauheim, 61231, Germany
| | - Jan Huisken
- Multiscale Biology, Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, 37077, Germany
| | - Owen J Tamplin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research (MPI-HLR), Bad Nauheim, 61231, Germany
| | - Felix Gunawan
- Institute of Cell Biology, Faculty of Medicine, University of Münster, Münster, 48149, Germany.
- 'Cells-in-Motion' Interfaculty Center, University of Münster, Münster, 48149, Germany.
| |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 0] [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
|
3
|
The adventitia in arterial development, remodeling, and hypertension. Biochem Pharmacol 2022; 205:115259. [PMID: 36150432 DOI: 10.1016/j.bcp.2022.115259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022]
Abstract
The adventitia receives input signals from the vessel wall, the immune system, perivascular nerves and from surrounding tissues to generate effector responses that regulate structural and mechanical properties of blood vessels. It is a complex and dynamic tissue that orchestrates multiple functions for vascular development, homeostasis, repair, and disease. The purpose of this review is to highlight recent advances in our understanding of the origins, phenotypes, and functions of adventitial and perivascular cells with particular emphasis on hypertensive vascular remodeling.
Collapse
|
4
|
Yao J, Ma F, Zhang L, Zhu C, Jumabay M, Yao Z, Wang L, Cai X, Zhang D, Qiao X, Shivkumar K, Pellegrini M, Yao Y, Wu X, Boström KI. Single-Cell RNA-Seq Identifies Dynamic Cardiac Transition Program from Adipose Derived Cells Induced by Leukemia Inhibitory Factor. Stem Cells 2022; 40:932-948. [PMID: 35896368 PMCID: PMC9585902 DOI: 10.1093/stmcls/sxac048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022]
Abstract
Adipose-derived cells (ADCs) from white adipose tissue (WAT) are promising stem cell candidates because of their large regenerative reserves and the potential for cardiac regeneration. However, given the heterogeneity of ADC and its unsolved mechanisms of cardiac acquisition, ADC-cardiac transition efficiency remains low. In this study, we explored the heterogeneity of ADCs and the cellular kinetics of 39,432 single-cell transcriptomes along the leukemia inhibitory factor (LIF) induced ADC-cardiac transition. We identified distinct ADC subpopulations that reacted differentially to LIF when entering the cardiomyogenic program, further demonstrating that ADC-myogenesis is time-dependent and initiates from transient changes in nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. At later stages, pseudotime analysis of ADCs navigated a trajectory with two branches corresponding to activated myofibroblast or cardiomyocyte-like cells. Our findings offer a high-resolution dissection of ADC heterogeneity and cell fate during ADC-cardiac transition, thus providing new insights into potential cardiac stem cells.
Collapse
Affiliation(s)
- Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Feiyang Ma
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.,Chongqing International Institute for Immunology, Chongqing 401338, China
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Ching Zhu
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Medet Jumabay
- Division of Allergy, Immunology Center for Immunity, Infection, and Inflammation Pediatrics, Dept of Medicine, University of California, San Diego, San Diego, CA
| | - Zehao Yao
- Peking Union Medical College, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Lumin Wang
- Institute of Precision Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Daoqin Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Xiaojing Qiao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | | | - Matteo Pellegrini
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.,Dept of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
5
|
Pogontke C, Guadix JA, Sánchez-Tévar AM, Muñoz-Chápuli R, Ruiz-Villalba A, Pérez-Pomares JM. Dynamic Epicardial Contribution to Cardiac Interstitial c-Kit and Sca1 Cellular Fractions. Front Cell Dev Biol 2022; 10:864765. [PMID: 35706902 PMCID: PMC9189417 DOI: 10.3389/fcell.2022.864765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 12/02/2022] Open
Abstract
Background: The cardiac interstitial cellular fraction is composed of multiple cell types. Some of these cells are known to express some well-known stem cell markers such as c-Kit and Sca1, but they are no longer accepted to be true cardiac stem cells. Although their existence in the cardiac interstitium has not been disputed, their dynamic throughout development, specific embryonic origin, and potential heterogeneity remain unknown. In this study, we hypothesized that both c-KitPOS and Sca1POS cardiac interstitial cell (CIC) subpopulations are related to the Wilms’ tumor 1 (Wt1) epicardial lineage. Methods: In this study, we have used genetic cell lineage tracing methods, immunohistochemistry, and FACS techniques to characterize cardiac c-KitPOS and Sca1POS cells. Results: Our data show that approximately 50% of cardiac c-KitPOS cells are derived from the Wt1-lineage at E15.5. This subpopulation decreased along with embryonic development, disappearing from P7 onwards. We found that a large proportion of cardiac c-KitPOS cells express specific markers strongly suggesting they are blood-borne cells. On the contrary, the percentage of Sca1POS cells within the Wt1-lineage increases postnatally. In accordance with these findings, 90% of adult epicardial-derived endothelial cells and 60% of mEFSK4POS cardiac fibroblasts expressed Sca1. Conclusion: Our study revealed a minor contribution of the Wt1-epicardial lineage to c-KitPOS CIC from embryonic stages to adulthood. Remarkably, a major part of the adult epicardial-derived cell fraction is enriched in Sca1, suggesting that this subpopulation of CICs is heterogeneous from their embryonic origin. The study of this heterogeneity can be instrumental to the development of diagnostic and prognostic tests for the evaluation of cardiac homeostasis and cardiac interstitium response to pathologic stimuli.
Collapse
Affiliation(s)
- C. Pogontke
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - J. A. Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - A. M. Sánchez-Tévar
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - R. Muñoz-Chápuli
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
| | - A. Ruiz-Villalba
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- *Correspondence: A. Ruiz-Villalba, ; J. M. Pérez-Pomares,
| | - J. M. Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto Malagueño de Biomedicina (IBIMA)-Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
- *Correspondence: A. Ruiz-Villalba, ; J. M. Pérez-Pomares,
| |
Collapse
|
6
|
The Vascular Niche for Adult Cardiac Progenitor Cells. Antioxidants (Basel) 2022; 11:antiox11050882. [PMID: 35624750 PMCID: PMC9137669 DOI: 10.3390/antiox11050882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
Research on cardiac progenitor cell populations has generated expectations about their potential for cardiac regeneration capacity after acute myocardial infarction and during physiological aging; however, the endogenous capacity of the adult mammalian heart is limited. The modest efficacy of exogenous cell-based treatments can guide the development of new approaches that, alone or in combination, can be applied to boost clinical efficacy. The identification and manipulation of the adult stem cell environment, termed niche, will be critical for providing new evidence on adult stem cell populations and improving stem-cell-based therapies. Here, we review and discuss the state of our understanding of the interaction of adult cardiac progenitor cells with other cardiac cell populations, with a focus on the description of the B-CPC progenitor population (Bmi1+ cardiac progenitor cell), which is a strong candidate progenitor for all main cardiac cell lineages, both in the steady state and after cardiac damage. The set of all interactions should be able to define the vascular cardiac stem cell niche, which is associated with low oxidative stress domains in vasculature, and whose manipulation would offer new hope in the cardiac regeneration field.
Collapse
|
7
|
In Vivo Methods to Monitor Cardiomyocyte Proliferation. J Cardiovasc Dev Dis 2022; 9:jcdd9030073. [PMID: 35323621 PMCID: PMC8950582 DOI: 10.3390/jcdd9030073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/07/2022] Open
Abstract
Adult mammalian cardiomyocytes demonstrate scarce cycling and even lower proliferation rates in response to injury. Signals that enhance cardiomyocyte proliferation after injury will be groundbreaking, address unmet clinical needs, and represent new strategies to treat cardiovascular diseases. In vivo methods to monitor cardiomyocyte proliferation are critical to addressing this challenge. Fortunately, advances in transgenic approaches provide sophisticated techniques to quantify cardiomyocyte cycling and proliferation.
Collapse
|
8
|
Prat-Vidal C, Crisóstomo V, Moscoso I, Báez-Díaz C, Blanco-Blázquez V, Gómez-Mauricio G, Albericio G, Aguilar S, Fernández-Santos ME, Fernández-Avilés F, Sánchez-Margallo FM, Bayes-Genis A, Bernad A. Intracoronary Delivery of Porcine Cardiac Progenitor Cells Overexpressing IGF-1 and HGF in a Pig Model of Sub-Acute Myocardial Infarction. Cells 2021; 10:cells10102571. [PMID: 34685551 PMCID: PMC8534140 DOI: 10.3390/cells10102571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human cardiac progenitor cells (hCPC) are considered a good candidate in cell therapy for ischemic heart disease, demonstrating capacity to improve functional recovery after myocardial infarction (MI), both in small and large preclinical animal models. However, improvements are required in terms of cell engraftment and efficacy. Based on previously published reports, insulin-growth factor 1 (IGF-1) and hepatocyte growth factor (HGF) have demonstrated substantial cardioprotective, repair and regeneration activities, so they are good candidates to be evaluated in large animal model of MI. We have validated porcine cardiac progenitor cells (pCPC) and lentiviral vectors to overexpress IGF-1 (co-expressing eGFP) and HGF (co-expressing mCherry). pCPC were transduced and IGF1-eGFPpos and HGF-mCherrypos populations were purified by cell sorting and further expanded. Overexpression of IGF-1 has a limited impact on pCPC expression profile, whereas results indicated that pCPC-HGF-mCherry cultures could be counter selecting high expresser cells. In addition, pCPC-IGF1-eGFP showed a higher cardiogenic response, evaluated in co-cultures with decellularized extracellular matrix, compared with native pCPC or pCPC-HGF-mCherry. In vivo intracoronary co-administration of pCPC-IGF1-eGFP and pCPC-HFG-mCherry (1:1; 40 × 106/animal), one week after the induction of an MI model in swine, revealed no significant improvement in cardiac function.
Collapse
Affiliation(s)
- Cristina Prat-Vidal
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain; (C.P.-V.); (A.B.-G.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08908 L’Hospitalet de Llobregat, Spain
| | - Verónica Crisóstomo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Isabel Moscoso
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Claudia Báez-Díaz
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Virginia Blanco-Blázquez
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | | | - Guillermo Albericio
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
| | - Susana Aguilar
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
| | - María-Eugenia Fernández-Santos
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Laboratorio Investigación Traslacional en Cardiología (LITC), Unidad de Producción Celular-GMP (UPC-GMP), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), TERCEL, 28007 Madrid, Spain
| | - Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Laboratorio Investigación Traslacional en Cardiología (LITC), Unidad de Producción Celular-GMP (UPC-GMP), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), TERCEL, 28007 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Francisco M. Sánchez-Margallo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Jesús Usón Minimally Invasive Surgery Center, 10071 Cáceres, Spain;
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain; (C.P.-V.); (A.B.-G.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.C.); (I.M.); (C.B.-D.); (V.B.-B.); (M.-E.F.-S.); (F.F.-A.); (F.M.S.-M.)
- Cardiology Service, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, National Center for Biotechnology, 28049 Madrid, Spain; (G.A.); (S.A.)
- Correspondence: ; Tel.: +34-915-855-424
| |
Collapse
|
9
|
Abstract
Endocardial cells are specialized endothelial cells that, during embryogenesis, form a lining on the inside of the developing heart, which is maintained throughout life. Endocardial cells are an essential source for several lineages of the cardiovascular system including coronary endothelium, endocardial cushion mesenchyme, cardiomyocytes, mural cells, fibroblasts, liver vasculature, adipocytes, and hematopoietic cells. Alterations in the differentiation programs that give rise to these lineages has detrimental effects, including premature lethality or significant structural malformations present at birth. Here, we will review the literature pertaining to the contribution of endocardial cells to valvular, and nonvalvular lineages and highlight critical pathways required for these processes. The lineage differentiation potential of embryonic, and possibly adult, endocardial cells has therapeutic potential in the regeneration of damaged cardiac tissue or treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Bailey Dye
- Biomedical Sciences Graduate Program at The Ohio State University, Columbus, Ohio 43210, USA.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.,Division of Pediatric Cardiology, Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Joy Lincoln
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.,Division of Pediatric Cardiology, Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin 53226, USA
| |
Collapse
|
10
|
Balaji Ragunathrao VA, Anwar M, Akhter MZ, Chavez A, Mao DY, Natarajan V, Lakshmikanthan S, Chrzanowska-Wodnicka M, Dudek AZ, Claesson-Welsh L, Kitajewski JK, Wary KK, Malik AB, Mehta D. Sphingosine-1-Phosphate Receptor 1 Activity Promotes Tumor Growth by Amplifying VEGF-VEGFR2 Angiogenic Signaling. Cell Rep 2020; 29:3472-3487.e4. [PMID: 31825830 PMCID: PMC6927555 DOI: 10.1016/j.celrep.2019.11.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/06/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelial growth factor-A (VEGF-A)-VEGFR2 pathway drives tumor vascularization by activating proangiogenic signaling in endothelial cells (ECs). Here, we show that EC-sphingosine-1-phosphate receptor 1 (S1PR1) amplifies VEGFR2-mediated angiogenic signaling to enhance tumor growth. We show that cancer cells induce S1PR1 activity in ECs, and thereby, conditional deletion of S1PR1 in ECs (EC-S1pr1−/− mice) impairs tumor vascularization and growth. Mechanistically, we show that S1PR1 engages the heterotrimeric G-protein Gi, which amplifies VEGF-VEGFR2 signaling due to an increase in the activity of the tyrosine kinase c-Abl1. c-Abl1, by phosphorylating VEGFR2 at tyrosine-951, prolongs VEGFR2 retention on the plasmalemma to sustain Rac1 activity and EC migration. Thus, S1PR1 or VEGFR2 antagonists, alone or in combination, reverse the tumor growth in control mice to the level seen in EC-S1pr1−/− mice. Our findings suggest that blocking S1PR1 activity in ECs has the potential to suppress tumor growth by preventing amplification of VEGF-VEGFR2 signaling. Vijay Avin et al. demonstrate an essential role of endothelial cell (EC)-S1PR1 signaling in amplifying VEGFR2-mediated tumor growth. S1PR1 by Gi and c-Abl1 phosphorylates VEGFR2 at Y951, which retains VEGFR2 at EC plasmalemma, thus enabling EC migration, tumor angiogenesis, and growth.
Collapse
Affiliation(s)
- Vijay Avin Balaji Ragunathrao
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Mumtaz Anwar
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Md Zahid Akhter
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Alejandra Chavez
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - De Yu Mao
- Department of Physiology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA; Department of Medicine, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | - Arkadiusz Z Dudek
- Department of Medicine, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Jan K Kitajewski
- Department of Physiology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kishore K Wary
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Asrar B Malik
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Dolly Mehta
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
11
|
Ma J, Sanchez-Duffhues G, Goumans MJ, ten Dijke P. TGF-β-Induced Endothelial to Mesenchymal Transition in Disease and Tissue Engineering. Front Cell Dev Biol 2020; 8:260. [PMID: 32373613 PMCID: PMC7187792 DOI: 10.3389/fcell.2020.00260] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
Endothelial to mesenchymal transition (EndMT) is a complex biological process that gives rise to cells with multipotent potential. EndMT is essential for the formation of the cardiovascular system during embryonic development. Emerging results link EndMT to the postnatal onset and progression of fibrotic diseases and cancer. Moreover, recent reports have emphasized the potential for EndMT in tissue engineering and regenerative applications by regulating the differentiation status of cells. Transforming growth factor β (TGF-β) engages in many important physiological processes and is a potent inducer of EndMT. In this review, we first summarize the mechanisms of the TGF-β signaling pathway as it relates to EndMT. Thereafter, we discuss the pivotal role of TGF-β-induced EndMT in the development of cardiovascular diseases, fibrosis, and cancer, as well as the potential application of TGF-β-induced EndMT in tissue engineering.
Collapse
Affiliation(s)
- Jin Ma
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | | | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Peter ten Dijke
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
12
|
Yellamilli A, Ren Y, McElmurry RT, Lambert JP, Gross P, Mohsin S, Houser SR, Elrod JW, Tolar J, Garry DJ, van Berlo JH. Abcg2-expressing side population cells contribute to cardiomyocyte renewal through fusion. FASEB J 2020; 34:5642-5657. [PMID: 32100368 DOI: 10.1096/fj.201902105r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
The adult mammalian heart has a limited regenerative capacity. Therefore, identification of endogenous cells and mechanisms that contribute to cardiac regeneration is essential for the development of targeted therapies. The side population (SP) phenotype has been used to enrich for stem cells throughout the body; however, SP cells isolated from the heart have been studied exclusively in cell culture or after transplantation, limiting our understanding of their function in vivo. We generated a new Abcg2-driven lineage-tracing mouse model with efficient labeling of SP cells. Labeled SP cells give rise to terminally differentiated cells in bone marrow and intestines. In the heart, labeled SP cells give rise to lineage-traced cardiomyocytes under homeostatic conditions with an increase in this contribution following cardiac injury. Instead of differentiating into cardiomyocytes like proposed cardiac progenitor cells, cardiac SP cells fuse with preexisting cardiomyocytes to stimulate cardiomyocyte cell cycle reentry. Our study is the first to show that fusion between cardiomyocytes and non-cardiomyocytes, identified by the SP phenotype, contribute to endogenous cardiac regeneration by triggering cardiomyocyte cell cycle reentry in the adult mammalian heart.
Collapse
Affiliation(s)
- Amritha Yellamilli
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Yi Ren
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ron T McElmurry
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jonathan P Lambert
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Polina Gross
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Sadia Mohsin
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jakub Tolar
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jop H van Berlo
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.,Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
13
|
Wang L, Yu P, Zhou B, Song J, Li Z, Zhang M, Guo G, Wang Y, Chen X, Han L, Hu S. Single-cell reconstruction of the adult human heart during heart failure and recovery reveals the cellular landscape underlying cardiac function. Nat Cell Biol 2020; 22:108-119. [PMID: 31915373 DOI: 10.1038/s41556-019-0446-7] [Citation(s) in RCA: 232] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 11/28/2019] [Indexed: 01/04/2023]
Abstract
Owing to the prevalence and high mortality rates of cardiac diseases, a more detailed characterization of the human heart is necessary; however, this has been largely impeded by the cellular diversity of cardiac tissue and limited access to samples. Here, we show transcriptome profiling of 21,422 single cells-including cardiomyocytes (CMs) and non-CMs (NCMs)-from normal, failed and partially recovered (left ventricular assist device treatment) adult human hearts. Comparative analysis of atrial and ventricular cells revealed pronounced inter- and intracompartmental CM heterogeneity as well as compartment-specific utilization of NCM cell types as major cell-communication hubs. Systematic analysis of cellular compositions and cell-cell interaction networks showed that CM contractility and metabolism are the most prominent aspects that are correlated with changes in heart function. We also uncovered active engagement of NCMs in regulating the behaviour of CMs, exemplified by ACKR1+-endothelial cells, injection of which preserved cardiac function after injury. Beyond serving as a rich resource, our study provides insights into cell-type-targeted intervention of heart diseases.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Peng Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingzhi Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangran Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Mekala SR, Wörsdörfer P, Bauer J, Stoll O, Wagner N, Reeh L, Loew K, Eckner G, Kwok CK, Wischmeyer E, Dickinson ME, Schulze H, Stegner D, Benndorf RA, Edenhofer F, Pfeiffer V, Kuerten S, Frantz S, Ergün S. Generation of Cardiomyocytes From Vascular Adventitia-Resident Stem Cells. Circ Res 2019; 123:686-699. [PMID: 30355234 DOI: 10.1161/circresaha.117.312526] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE Regeneration of lost cardiomyocytes is a fundamental unresolved problem leading to heart failure. Despite several strategies developed from intensive studies performed in the past decades, endogenous regeneration of heart tissue is still limited and presents a big challenge that needs to be overcome to serve as a successful therapeutic option for myocardial infarction. OBJECTIVE One of the essential prerequisites for cardiac regeneration is the identification of endogenous cardiomyocyte progenitors and their niche that can be targeted by new therapeutic approaches. In this context, we hypothesized that the vascular wall, which was shown to harbor different types of stem and progenitor cells, might serve as a source for cardiac progenitors. METHODS AND RESULTS We describe generation of spontaneously beating mouse aortic wall-derived cardiomyocytes without any genetic manipulation. Using aortic wall-derived cells (AoCs) of WT (wild type), αMHC (α-myosin heavy chain), and Flk1 (fetal liver kinase 1)-reporter mice and magnetic bead-associated cell sorting sorting of Flk1+ AoCs from GFP (green fluorescent protein) mice, we identified Flk1+CD (cluster of differentiation) 34+Sca-1 (stem cell antigen-1)-CD44- AoCs as the population that gives rise to aortic wall-derived cardiomyocytes. This AoC subpopulation delivered also endothelial cells and macrophages with a particular accumulation within the aortic wall-derived cardiomyocyte containing colonies. In vivo, cardiomyocyte differentiation capacity was studied by implantation of fluorescently labeled AoCs into chick embryonic heart. These cells acquired cardiomyocyte-like phenotype as shown by αSRA (α-sarcomeric actinin) expression. Furthermore, coronary adventitial Flk1+ and CD34+ cells proliferated, migrated into the myocardium after mouse myocardial infarction, and expressed Isl-1+ (insulin gene enhancer protein-1) indicative of cardiovascular progenitor potential. CONCLUSIONS Our data suggest Flk1+CD34+ vascular adventitia-resident stem cells, including those of coronary adventitia, as a novel endogenous source for generating cardiomyocytes. This process is essentially supported by endothelial cells and macrophages. In summary, the therapeutic manipulation of coronary adventitia-resident cardiac stem and their supportive cells may open new avenues for promoting cardiac regeneration and repair after myocardial infarction and for preventing heart failure.
Collapse
Affiliation(s)
- Subba Rao Mekala
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Philipp Wörsdörfer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Jochen Bauer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Olga Stoll
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Nicole Wagner
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Laurens Reeh
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Kornelia Loew
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Georg Eckner
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Chee Keong Kwok
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Erhard Wischmeyer
- Institute of Physiology (E.W.).,University of Würzburg, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, Center of Mental Health (E.W.)
| | - Mary Eleanor Dickinson
- University Hospital of Wuerzburg, Germany; Baylor College of Medicine, Houston, TX (M.E.D.)
| | | | | | - Ralf A Benndorf
- Department of Clinical Pharmacy and Pharmacotherapy, University of Halle-Wittenberg, Germany (R.A.B.)
| | - Frank Edenhofer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Verena Pfeiffer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Stefanie Kuerten
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Stefan Frantz
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.).,Department of Internal Medicine I, ZIM (Zentrum für Innere Medizin) (S.F.)
| | - Süleyman Ergün
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| |
Collapse
|
15
|
Abstract
Endocardial cells are specialized endothelial cells that form the innermost layer of the heart wall. By virtue of genetic lineage-tracing technology, many of the unexpected roles of endocardium during murine heart development, diseases, and regeneration have been identified recently. In addition to heart valves developed from the well-known endothelial to mesenchymal transition, recent fate-mapping studies using mouse models reveal that multiple cardiac cell lineages are also originated from the endocardium. This review focuses on a variety of different cell types that are recently reported to be endocardium derived during murine heart development, diseases, and regeneration. These multiple cell fates underpin the unprecedented roles of endocardial progenitors in function, pathological progression, and regeneration of the heart. Because emerging studies suggest that developmental mechanisms can be redeployed and recapitulated in promoting heart disease development and also cardiac repair and regeneration, understanding the mechanistic regulation of endocardial plasticity and modulation of their cell fate conversion may uncover new therapeutic potential in facilitating heart regeneration.
Collapse
Affiliation(s)
- Hui Zhang
- From the The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.Z., B.Z.); School of Life Science and Technology, ShanghaiTech University, China (H.Z., B.Z.); 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.); and Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).
| | - Kathy O Lui
- From the The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.Z., B.Z.); School of Life Science and Technology, ShanghaiTech University, China (H.Z., B.Z.); 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.); and Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).
| | - Bin Zhou
- From the The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.Z., B.Z.); School of Life Science and Technology, ShanghaiTech University, China (H.Z., B.Z.); 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.); and Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).
| |
Collapse
|
16
|
Herrero D, Cañón S, Pelacho B, Salvador-Bernáldez M, Aguilar S, Pogontke C, Carmona RM, Salvador JM, Perez-Pomares JM, Klein OD, Prósper F, Jimenez-Borreguero LJ, Bernad A. Bmi1-Progenitor Cell Ablation Impairs the Angiogenic Response to Myocardial Infarction. Arterioscler Thromb Vasc Biol 2019; 38:2160-2173. [PMID: 29930004 DOI: 10.1161/atvbaha.118.310778] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective- Cardiac progenitor cells reside in the heart in adulthood, although their physiological relevance remains unknown. Here, we demonstrate that after myocardial infarction, adult Bmi1+ (B lymphoma Mo-MLV insertion region 1 homolog [PCGF4]) cardiac cells are a key progenitor-like population in cardiac neovascularization during ventricular remodeling. Approach and Results- These cells, which have a strong in vivo differentiation bias, are a mixture of endothelial- and mesenchymal-related cells with in vitro spontaneous endothelial cell differentiation capacity. Genetic lineage tracing analysis showed that heart-resident Bmi1+ progenitor cells proliferate after acute myocardial infarction and differentiate to generate de novo cardiac vasculature. In a mouse model of induced myocardial infarction, genetic ablation of these cells substantially deteriorated both heart angiogenesis and the ejection fraction, resulting in an ischemic-dilated cardiac phenotype. Conclusions- These findings imply that endothelial-related Bmi1+ progenitor cells are necessary for injury-induced neovascularization in adult mouse heart and highlight these cells as a suitable therapeutic target for preventing dysfunctional left ventricular remodeling after injury.
Collapse
Affiliation(s)
- Diego Herrero
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| | - Susana Cañón
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| | - Beatriz Pelacho
- Center for Applied Medical Research (CIMA) Regenerative Medicine Area, University of Navarra, Pamplona, Spain (B.P., F.P.).,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain (B.P., F.P.)
| | - María Salvador-Bernáldez
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| | - Susana Aguilar
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| | - Cristina Pogontke
- Department of Animal Biology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga (IBIMA) and BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía), Universidad de Málaga, Spain (C.P., J.M.P.-P.)
| | - Rosa María Carmona
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| | - Jesús María Salvador
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| | - Jose María Perez-Pomares
- Department of Animal Biology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga (IBIMA) and BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía), Universidad de Málaga, Spain (C.P., J.M.P.-P.)
| | - Ophir David Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco (O.D.K.)
| | - Felipe Prósper
- Center for Applied Medical Research (CIMA) Regenerative Medicine Area, University of Navarra, Pamplona, Spain (B.P., F.P.).,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain (B.P., F.P.)
| | - Luis Jesús Jimenez-Borreguero
- Cardiovascular Development and Repair Department, National Cardiovascular Research Center (CNIC) and Hospital de La Princesa, Madrid, Spain (L.J.J.-B.)
| | - Antonio Bernad
- From the Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), Madrid, Spain (D.H., S.C., M.S.-B., S.A., R.M.C., J.M.S., A.B.)
| |
Collapse
|
17
|
Torán JL, López JA, Gomes-Alves P, Aguilar S, Torroja C, Trevisan-Herraz M, Moscoso I, Sebastião MJ, Serra M, Brito C, Cruz FM, Sepúlveda JC, Abad JL, Galán-Arriola C, Ibanez B, Martínez F, Fernández ME, Fernández-Aviles F, Palacios I, R-Borlado L, Vázquez J, Alves PM, Bernad A. Definition of a cell surface signature for human cardiac progenitor cells after comprehensive comparative transcriptomic and proteomic characterization. Sci Rep 2019; 9:4647. [PMID: 30874584 PMCID: PMC6420620 DOI: 10.1038/s41598-019-39571-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 01/22/2019] [Indexed: 12/24/2022] Open
Abstract
Adult cardiac progenitor/stem cells (CPC/CSC) are multipotent resident populations involved in cardiac homeostasis and heart repair. Assisted by complementary RNAseq analysis, we defined the fraction of the CPC proteome associable with specific functions by comparison with human bone marrow mesenchymal stem cells (MSC), the reference population for cell therapy, and human dermal fibroblasts (HDF), as a distant reference. Label-free proteomic analysis identified 526 proteins expressed differentially in CPC. iTRAQ analysis confirmed differential expression of a substantial proportion of those proteins in CPC relative to MSC, and systems biology analysis defined a clear overrepresentation of several categories related to enhanced angiogenic potential. The CPC plasma membrane compartment comprised 1,595 proteins, including a minimal signature of 167 proteins preferentially or exclusively expressed by CPC. CDH5 (VE-cadherin), OX2G (OX-2 membrane glycoprotein; CD200), GPR4 (G protein-coupled receptor 4), CACNG7 (calcium voltage-gated channel auxiliary subunit gamma 7) and F11R (F11 receptor; junctional adhesion molecule A; JAM-A; CD321) were selected for validation. Their differential expression was confirmed both in expanded CPC batches and in early stages of isolation, particularly when compared against cardiac fibroblasts. Among them, GPR4 demonstrated the highest discrimination capacity between all cell lineages analyzed.
Collapse
Affiliation(s)
- José Luis Torán
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Antonio López
- Laboratory of Cardiovascular Proteomics, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Patricia Gomes-Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Susana Aguilar
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Carlos Torroja
- Bioinformatics Unit, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Marco Trevisan-Herraz
- Laboratory of Cardiovascular Proteomics, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Isabel Moscoso
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.,CIMUS, Avda Barcelona s/n, Santiago de Compostela, 15782A, Coruña, Spain
| | - Maria João Sebastião
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Francisco Miguel Cruz
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Juan Carlos Sepúlveda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - José Luis Abad
- Coretherapix S.L. U. Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Fernando Martínez
- Bioinformatics Unit, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - María Eugenia Fernández
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Dr Esquerdo, 46, 28007, Madrid, Spain
| | - Francisco Fernández-Aviles
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Dr Esquerdo, 46, 28007, Madrid, Spain
| | - Itziar Palacios
- Coretherapix S.L. U. Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Luis R-Borlado
- Coretherapix S.L. U. Santiago Grisolia 2, 28769, Tres Cantos, Madrid, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - Antonio Bernad
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
18
|
Herrero D, Cañón S, Albericio G, Carmona RM, Aguilar S, Mañes S, Bernad A. Age-related oxidative stress confines damage-responsive Bmi1 + cells to perivascular regions in the murine adult heart. Redox Biol 2019; 22:101156. [PMID: 30851670 PMCID: PMC6407305 DOI: 10.1016/j.redox.2019.101156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/23/2019] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Adult progenitor cells reside in specialized microenvironments which maintain their undifferentiated cell state and trigger regenerative responses following injury. Although these environments are well described in several tissues, the cellular components that comprise the cardiac environment where progenitor cells are located remain unknown. Here we use Bmi1CreERT and Bmi1GFP mice as genetic tools to trace cardiac damage-responsive cells throughout the mouse lifespan. In adolescent mice, Bmi1+ damage-responsive cells are broadly distributed throughout the myocardium. In adult mice, however, Bmi1+ cells are confined predominately in perivascular areas with low levels of reactive oxygen species (ROS) and their number decline in an age-dependent manner. In vitro co-culture experiments with endothelial cells supported a regulatory role of the endothelium in damage-responsive cell behavior. Accordingly, in vivo genetic decrease of ROS levels in adult heart disengaged Bmi1+ cells from the cardiovascular network, recapitulating an adolescent-like Bmi1 expression profile. Thus, we identify cardiac perivascular regions as low-stress microenvironments that favor the maintenance of adult damage-responsive cells. Bmi1+ cardiac damage-responsive cells are sheltered in areas with low ROS levels. Aging-related oxidative damage confines cardiac Bmi1+ cells to perivascular regions. Microvasculature-derived signals regulate adult Bmi1+ damage-responsive cell behavior. Genetic ROS levels manipulation modifies the percentage and identity of Bmi1+ cells.
Collapse
Affiliation(s)
- Diego Herrero
- Cardiac Stem Cells Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Susana Cañón
- Cardiac Stem Cells Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Guillermo Albericio
- Cardiac Stem Cells Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Rosa María Carmona
- Cardiac Stem Cells Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Susana Aguilar
- Cardiac Stem Cells Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Santos Mañes
- Signaling Networks in Inflammation and Cancer Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain
| | - Antonio Bernad
- Cardiac Stem Cells Group, Department of Immunology and Oncology, National Center for Biotechnology (CNB-CSIC), 28049, Madrid, Spain.
| |
Collapse
|
19
|
Doppler SA, Deutsch MA, Serpooshan V, Li G, Dzilic E, Lange R, Krane M, Wu SM. Mammalian Heart Regeneration: The Race to the Finish Line. Circ Res 2019; 120:630-632. [PMID: 28209796 DOI: 10.1161/circresaha.116.310051] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Stefanie A Doppler
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Marcus-Andre Deutsch
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Vahid Serpooshan
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Guang Li
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Elda Dzilic
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Rüdiger Lange
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.)
| | - Markus Krane
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.).
| | - Sean M Wu
- From the Division of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Germany (S.A.D., M.-A.D., E.D., R.L., M.K.); Stanford Cardiovascular Institute (V.S., G.L., E.D., S.M.W.), Institute for Stem Cell and Regenerative Medicine (S.M.W.), and Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA; DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (M.-A.D., M.K.).
| |
Collapse
|
20
|
Robich MP, Ryzhov S, Sawyer DB. Successful rebuilding after disaster, even in the heart, starts with infrastructure. J Thorac Dis 2019; 10:S4165-S4167. [PMID: 30631583 DOI: 10.21037/jtd.2018.10.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Michael P Robich
- Maine Medical Center, Cardiovascular Institute, Portland, ME, USA.,Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Sergey Ryzhov
- Maine Medical Center, Cardiovascular Institute, Portland, ME, USA
| | - Douglas B Sawyer
- Maine Medical Center, Cardiovascular Institute, Portland, ME, USA.,Maine Medical Center Research Institute, Scarborough, ME, USA
| |
Collapse
|
21
|
Pogontke C, Guadix JA, Ruiz-Villalba A, Pérez-Pomares JM. Development of the Myocardial Interstitium. Anat Rec (Hoboken) 2018; 302:58-68. [PMID: 30288955 DOI: 10.1002/ar.23915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/26/2018] [Accepted: 05/11/2018] [Indexed: 12/24/2022]
Abstract
The space between cardiac myocytes is commonly referred-to as the cardiac interstitium (CI). The CI is a unique, complex and dynamic microenvironment in which multiple cell types, extracellular matrix molecules, and instructive signals interact to crucially support heart homeostasis and promote cardiac responses to normal and pathologic stimuli. Despite the biomedical and clinical relevance of the CI, its detailed cellular structure remains to be elucidated. In this review, we will dissect the organization of the cardiac interstitium by following its changing cellular and molecular composition from embryonic developmental stages to adulthood, providing a systematic analysis of the biological components of the CI. The main goal of this review is to contribute to our understanding of the CI roles in health and disease. Anat Rec, 302:58-68, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cristina Pogontke
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, 29080, Málaga, Spain.,BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), Severo Ochoa n°25, 29590 Campanillas (Málaga), Spain
| | - Juan A Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, 29080, Málaga, Spain.,BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), Severo Ochoa n°25, 29590 Campanillas (Málaga), Spain
| | - Adrián Ruiz-Villalba
- Stem Cell Therapy Area, Foundation for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - José M Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Instituto Malagueño de Biomedicina (IBIMA), Campus de Teatinos s/n, 29080, Málaga, Spain.,BIONAND, Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía, Universidad de Málaga), Severo Ochoa n°25, 29590 Campanillas (Málaga), Spain
| |
Collapse
|
22
|
Identification of a multipotent Twist2-expressing cell population in the adult heart. Proc Natl Acad Sci U S A 2018; 115:E8430-E8439. [PMID: 30127033 DOI: 10.1073/pnas.1800526115] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Twist transcription factors function as ancestral regulators of mesodermal cell fates in organisms ranging from Drosophila to mammals. Through lineage tracing of Twist2 (Tw2)-expressing cells with tamoxifen-inducible Tw2-CreERT2 and tdTomato (tdTO) reporter mice, we discovered a unique cell population that progressively contributes to cardiomyocytes (CMs), endothelial cells, and fibroblasts in the adult heart. Clonal analysis confirmed the ability of Tw2-derived tdTO+ (Tw2-tdTO+) cells to form CMs in vitro. Within the adult heart, Tw2-tdTO+ CMs accounted for ∼13% of total CMs, the majority of which resulted from fusion of Tw2-tdTO+ cells with existing CMs. Tw2-tdTO+ cells also contribute to cardiac remodeling after injury. We conclude that Tw2-tdTO+ cells participate in lifelong maintenance of cardiac function, at least in part through de novo formation of CMs and fusion with preexisting CMs, as well as in the genesis of other cellular components of the adult heart.
Collapse
|
23
|
Man S, Sanchez Duffhues G, Ten Dijke P, Baker D. The therapeutic potential of targeting the endothelial-to-mesenchymal transition. Angiogenesis 2018; 22:3-13. [PMID: 30076548 PMCID: PMC6510911 DOI: 10.1007/s10456-018-9639-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022]
Abstract
Endothelial cells (ECs) have been found to be capable of acquiring a mesenchymal phenotype through a process known as endothelial-to-mesenchymal transition (EndMT). First seen in the developing embryo, EndMT can be triggered postnatally under certain pathological conditions. During this process, ECs dedifferentiate into mesenchymal stem-like cells (MSCs) and subsequently give rise to cell types belonging to the mesoderm lineage. As EndMT contributes to a multitude of diseases, pharmacological modulation of the signaling pathways underlying EndMT may prove to be effective as a therapeutic treatment. Additionally, EndMT in ECs could also be exploited to acquire multipotent MSCs, which can be readily re-differentiated into various distinct cell types. In this review, we will consider current models of EndMT, how manipulation of this process might improve treatment of clinically important pathologies and how it could be harnessed to advance regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Shirley Man
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Gonzalo Sanchez Duffhues
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands.
| | - David Baker
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
24
|
Abstract
Death of adult cardiac myocytes and supportive tissues resulting from cardiovascular diseases such as myocardial infarction is the proximal driver of pathological ventricular remodeling that often culminates in heart failure. Unfortunately, no currently available therapeutic barring heart transplantation can directly replenish myocytes lost from the injured heart. For decades, the field has struggled to define the intrinsic capacity and cellular sources for endogenous myocyte turnover in pursuing more innovative therapeutic strategies aimed at regenerating the injured heart. Although controversy persists to this day as to the best therapeutic regenerative strategy to use, a growing consensus has been reached that the very limited capacity for new myocyte formation in the adult mammalian heart is because of proliferation of existing cardiac myocytes but not because of the activity of an endogenous progenitor cell source of some sort. Hence, future therapeutic approaches should take into consideration the fundamental biology of myocyte renewal in designing strategies to potentially replenish these cells in the injured heart.
Collapse
Affiliation(s)
| | - Jeffery D Molkentin
- From the Department of Pediatrics (R.J.V., J.D.M.)
- Howard Hughes Medical Institute (J.D.M.)
| | - Steven R Houser
- Cincinnati Children's Hospital Medical Center, OH; and the Lewis Katz School of Medicine, Cardiovascular Research Center, Temple University, Philadelphia, PA (S.R.H.)
| |
Collapse
|
25
|
Tang J, Zhang H, He L, Huang X, Li Y, Pu W, Yu W, Zhang L, Cai D, Lui KO, Zhou B. Genetic Fate Mapping Defines the Vascular Potential of Endocardial Cells in the Adult Heart. Circ Res 2018; 122:984-993. [DOI: 10.1161/circresaha.117.312354] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Juan Tang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Hui Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Lingjuan He
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Xiuzhen Huang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Yan Li
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Wenjuan Pu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Wei Yu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Libo Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Dongqing Cai
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Kathy O. Lui
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Bin Zhou
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| |
Collapse
|
26
|
See K, Tan WLW, Lim EH, Tiang Z, Lee LT, Li PYQ, Luu TDA, Ackers-Johnson M, Foo RS. Single cardiomyocyte nuclear transcriptomes reveal a lincRNA-regulated de-differentiation and cell cycle stress-response in vivo. Nat Commun 2017; 8:225. [PMID: 28790305 PMCID: PMC5548780 DOI: 10.1038/s41467-017-00319-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/21/2017] [Indexed: 12/14/2022] Open
Abstract
Cardiac regeneration may revolutionize treatment for heart failure but endogenous progenitor-derived cardiomyocytes in the adult mammalian heart are few and pre-existing adult cardiomyocytes divide only at very low rates. Although candidate genes that control cardiomyocyte cell cycle re-entry have been implicated, expression heterogeneity in the cardiomyocyte stress-response has never been explored. Here, we show by single nuclear RNA-sequencing of cardiomyocytes from both mouse and human failing, and non-failing adult hearts that sub-populations of cardiomyocytes upregulate cell cycle activators and inhibitors consequent to the stress-response in vivo. We characterize these subgroups by weighted gene co-expression network analysis and discover long intergenic non-coding RNAs (lincRNA) as key nodal regulators. KD of nodal lincRNAs affects expression levels of genes related to dedifferentiation and cell cycle, within the same gene regulatory network. Our study reveals that sub-populations of adult cardiomyocytes may have a unique endogenous potential for cardiac regeneration in vivo. Adult mammalian cardiomyocytes are predominantly binucleated and unable to divide. Using single nuclear RNA-sequencing of cardiomyocytes from mouse and human failing and non-failing adult hearts, See et al. show that some cardiomyocytes respond to stress by dedifferentiation and cell cycle re-entry regulated by lncRNAs.
Collapse
Affiliation(s)
- Kelvin See
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Wilson L W Tan
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.,Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore
| | - Eng How Lim
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.,Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore
| | - Zenia Tiang
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore.,Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore
| | - Li Ting Lee
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore
| | - Peter Y Q Li
- Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore
| | - Tuan D A Luu
- Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore
| | - Matthew Ackers-Johnson
- Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore
| | - Roger S Foo
- Genome Institute of Singapore, 60 Biopolis Street, Singapore, 138672, Singapore. .,Cardiovascular Research Institute, National University Health System, Centre for Translational Medicine, 14 Medical Drive, Singapore, 117599, Singapore.
| |
Collapse
|
27
|
Abstract
The stem cells keep us young by endogenously repairing us upon need. They do so bysmartly one step forward towards differentiation while another step backward to nurturethe undifferentiated stem cells. They are building blocks for every organ witha differential rate of repair of worn out tissues. Since stem cells can be cultured with a normal karyo type, they could be the ideal source for heart repair after myocardial infarction. As opposed to lower vertebrates and neonatal mice, cardiac regeneration in adult mammalian heart seems to be difficult to assess with a solid evidence of cytokinesis. It becomes more difficult to quantify the level of regeneration after myocardial infarction injury against a background of a large invasion of proliferating inflammatory cells. The question to beraised is how the renewal of a piece of myocardium follows the time line of picking upcell types in series: cardiomyocytes, endothelial cells, smooth muscle cells, fibroblast, pacemaker cells, conducting and Purkinje cells to bring the orchestration of rhythmically contracting and relaxing heart. This review focuses on where we are onthe status of heart repair and cardiac regeneration.
Collapse
Affiliation(s)
- H R Ahmad
- Dr. HR Ahmad, MD PhD Bochum, FCPS. Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Satwat Hashmi
- Dr. Satwat Hashmi, MBBS MS PhD. Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
28
|
|
29
|
The vascular adventitia: An endogenous, omnipresent source of stem cells in the body. Pharmacol Ther 2017; 171:13-29. [DOI: 10.1016/j.pharmthera.2016.07.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/22/2022]
|
30
|
Abstract
The hearts of lower vertebrates such as fish and salamanders display scarless regeneration following injury, although this feature is lost in adult mammals. The remarkable capacity of the neonatal mammalian heart to regenerate suggests that the underlying machinery required for the regenerative process is evolutionarily retained. Recent studies highlight the epicardial covering of the heart as an important source of the signalling factors required for the repair process. The developing epicardium is also a major source of cardiac fibroblasts, smooth muscle, endothelial cells and stem cells. Here, we examine animal models that are capable of scarless regeneration, the role of the epicardium as a source of cells, signalling mechanisms implicated in the regenerative process and how these mechanisms influence cardiomyocyte proliferation. We also discuss recent advances in cardiac stem cell research and potential therapeutic targets arising from these studies.
Collapse
Affiliation(s)
| | - Nadia Rosenthal
- National Heart and Lung Institute, Imperial College London, London, UK Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia The Jackson Laboratory, Bar Harbor, ME, USA
| |
Collapse
|
31
|
Brunner R, Lai HL, Deliu Z, Melman E, Geenen DL, Wang QT. Asxl2 -/- Mice Exhibit De Novo Cardiomyocyte Production during Adulthood. J Dev Biol 2016; 4:jdb4040032. [PMID: 29615595 PMCID: PMC5831801 DOI: 10.3390/jdb4040032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022] Open
Abstract
Heart attacks affect more than seven million people worldwide each year. A heart attack, or myocardial infarction, may result in the death of a billion cardiomyocytes within hours. The adult mammalian heart does not have an effective mechanism to replace lost cardiomyocytes. Instead, lost muscle is replaced with scar tissue, which decreases blood pumping ability and leads to heart failure over time. Here, we report that the loss of the chromatin factor ASXL2 results in spontaneous proliferation and cardiogenic differentiation of a subset of interstitial non-cardiomyocytes. The adult Asxl2-/- heart displays spontaneous overgrowth without cardiomyocyte hypertrophy. Thymidine analog labeling and Ki67 staining of 12-week-old hearts revealed 3- and 5-fold increases of proliferation rate for vimentin⁺ non-cardiomyocytes in Asxl2-/- over age- and sex-matched wildtype controls, respectively. Approximately 10% of proliferating non-cardiomyocytes in the Asxl2-/- heart express the cardiogenic marker NKX2-5, a frequency that is ~7-fold higher than that observed in the wildtype. EdU lineage tracing experiments showed that ~6% of pulsed-labeled non-cardiomyocytes in Asxl2-/- hearts differentiate into mature cardiomyocytes after a four-week chase, a phenomenon not observed for similarly pulse-chased wildtype controls. Taken together, these data indicate de novo cardiomyocyte production in the Asxl2-/- heart due to activation of a population of proliferative cardiogenic non-cardiomyocytes. Our study suggests the existence of an epigenetic barrier to cardiogenicity in the adult heart and raises the intriguing possibility of unlocking regenerative potential via transient modulation of epigenetic activity.
Collapse
Affiliation(s)
- Rachel Brunner
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | - Hsiao-Lei Lai
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
- PTM Biolabs Inc., Chicago, IL 60612, USA.
| | - Zane Deliu
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| | - Elan Melman
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
- The School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Champaign, IL 61801, USA.
| | - David L Geenen
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA.
- Physician Assistant Studies, Grand Valley State University, Grand Rapids, MI 49503, USA.
| | - Q Tian Wang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
- Congressionally Directed Medical Research Programs, Frederick, MD 21702, USA.
| |
Collapse
|
32
|
Cencioni C, Atlante S, Savoia M, Martelli F, Farsetti A, Capogrossi MC, Zeiher AM, Gaetano C, Spallotta F. The double life of cardiac mesenchymal cells: Epimetabolic sensors and therapeutic assets for heart regeneration. Pharmacol Ther 2016; 171:43-55. [PMID: 27742569 DOI: 10.1016/j.pharmthera.2016.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organ-specific mesenchymal cells naturally reside in the stroma, where they are exposed to some environmental variables affecting their biology and functions. Risk factors such as diabetes or aging influence their adaptive response. In these cases, permanent epigenetic modifications may be introduced in the cells with important consequences on their local homeostatic activity and therapeutic potential. Numerous results suggest that mesenchymal cells, virtually present in every organ, may contribute to tissue regeneration mostly by paracrine mechanisms. Intriguingly, the heart is emerging as a source of different cells, including pericytes, cardiac progenitors, and cardiac fibroblasts. According to phenotypic, functional, and molecular criteria, these should be classified as mesenchymal cells. Not surprisingly, in recent years, the attention on these cells as therapeutic tools has grown exponentially, although only very preliminary data have been obtained in clinical trials to date. In this review, we summarized the state of the art about the phenotypic features, functions, regenerative properties, and clinical applicability of mesenchymal cells, with a particular focus on those of cardiac origin.
Collapse
Affiliation(s)
- Chiara Cencioni
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Sandra Atlante
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Matteo Savoia
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Universitá Cattolica, Institute of Medical Pathology, 00138 Rome, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan 20097, Italy.
| | - Antonella Farsetti
- Consiglio Nazionale delle Ricerche, Istituto di Biologia Cellulare e Neurobiologia, Roma, Italy; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Maurizio C Capogrossi
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, Roma, Italy.
| | - Andreas M Zeiher
- Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany; Internal Medicine Clinic III, Department of Cardiology, Goethe University, Frankfurt am Main 60596, Germany.
| |
Collapse
|
33
|
Valiente-Alandi I, Albo-Castellanos C, Herrero D, Sanchez I, Bernad A. Bmi1 (+) cardiac progenitor cells contribute to myocardial repair following acute injury. Stem Cell Res Ther 2016; 7:100. [PMID: 27472922 PMCID: PMC4967328 DOI: 10.1186/s13287-016-0355-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/25/2016] [Accepted: 06/28/2016] [Indexed: 01/19/2023] Open
Abstract
Background The inability of the adult mammalian heart to replace cells lost after severe cardiac injury compromises organ function. Although the heart is one of the least regenerative organs in the body, evidence accumulated in recent decades indicates a certain degree of renewal after injury. We have evaluated the role of cardiac Bmi1+ progenitor cells (Bmi1-CPC) following acute myocardial infarction (AMI). Methods Bmi1Cre/+;Rosa26YFP/+ (Bmi1-YFP) mice were used for lineage tracing strategy. After tamoxifen (TM) induction, yellow fluorescent protein (YFP) is expressed under the control of Rosa26 regulatory sequences in Bmi1+ cells. YFP+ cells were tracked following myocardial infarction. Additionally, whole transcriptome analysis of isolated YFP+ cells was performed in unchallenged hearts and after myocardial infarction. Results Deep-sequencing analysis of Bmi1-CPC from unchallenged hearts suggests that this population expresses high levels of pluripotency markers. Conversely, transcriptome evaluation of Bmi1-CPC following AMI shows a rich representation of genes related to cell proliferation, movement, and cell cycle. Lineage-tracing studies after cardiac infarction show that the progeny of Bmi1-expressing cells contribute to de novo cardiomyocytes (CM) (13.8 ± 5 % new YFP+ CM compared to 4.7 ± 0.9 % in age-paired non-infarcted hearts). However, apical resection of TM-induced day 1 Bmi1-YFP pups indicated a very minor contribution of Bmi1-derived cells to de novo CM. Conclusions Cardiac Bmi1 progenitor cells respond to cardiac injury, contributing to the generation of de novo CM in the adult mouse heart. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0355-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iñigo Valiente-Alandi
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.,Current address: The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carmen Albo-Castellanos
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.,Current address: Vivebiotech, San Sebastian, Spain
| | - Diego Herrero
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - Iria Sanchez
- Unidad de Medicina Comparada, Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain
| | - Antonio Bernad
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
34
|
Hatzistergos KE, Hare JM. Murine Models Demonstrate Distinct Vasculogenic and Cardiomyogenic cKit+ Lineages in the Heart. Circ Res 2016; 118:382-7. [PMID: 26846638 DOI: 10.1161/circresaha.115.308061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
After 2 recent genetic studies in mice addressing the developmental origins and regenerative activity of cardiac cKit+ cells, 2 additional reports by Sultana et al and Liu et al provide further information on the expression of cKit in the embryonic and adult hearts. Here, we synthesize the findings from the 4 distinct cKit models to gain insights into the biology of this important cell type.
Collapse
Affiliation(s)
- Konstantinos E Hatzistergos
- From the Interdisciplinary Stem Cell Institute (K.E.H.), and Department of Medicine, Division of Cardiology and Department of Molecular and Cellular Pharmacology (J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (K.E.H.), and Department of Medicine, Division of Cardiology and Department of Molecular and Cellular Pharmacology (J.M.H.), Leonard M. Miller School of Medicine, University of Miami, FL.
| |
Collapse
|
35
|
Medici D. Endothelial-Mesenchymal Transition in Regenerative Medicine. Stem Cells Int 2016; 2016:6962801. [PMID: 27143978 PMCID: PMC4838799 DOI: 10.1155/2016/6962801] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/12/2016] [Accepted: 03/22/2016] [Indexed: 12/29/2022] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is a fundamental cellular mechanism that regulates embryonic development and diseases such as cancer and fibrosis. Recent developments in biomedical research have shown remarkable potential to harness the EndMT process for tissue engineering and regeneration. As an alternative to traditional or artificial stem cell therapies, EndMT may represent a safe method for engineering new tissues to treat degenerative diseases by mimicking a process that occurs in nature. This review discusses the signaling mechanisms and therapeutic inhibitors of EndMT, as well as the role of EndMT in development, disease, acquiring stem cell properties and generating connective tissues, and its potential as a novel mechanism for tissue regeneration.
Collapse
Affiliation(s)
- Damian Medici
- Department of Orthopaedics, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
- Division of Hematology/Oncology, Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
- Center for Regenerative Medicine, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
- Cardiovascular Research Center, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
36
|
Nakano A, Nakano H, Smith KA, Palpant NJ. The developmental origins and lineage contributions of endocardial endothelium. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1937-47. [PMID: 26828773 DOI: 10.1016/j.bbamcr.2016.01.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/21/2015] [Accepted: 01/28/2016] [Indexed: 10/22/2022]
Abstract
Endocardial development involves a complex orchestration of cell fate decisions that coordinate with endoderm formation and other mesodermal cell lineages. Historically, investigations into the contribution of endocardium in the developing embryo was constrained to the heart where these cells give rise to the inner lining of the myocardium and are a major contributor to valve formation. In recent years, studies have continued to elucidate the complexities of endocardial fate commitment revealing a much broader scope of lineage potential from developing endocardium. These studies cover a wide range of species and model systems and show direct contribution or fate potential of endocardium giving rise to cardiac vasculature, blood, fibroblast, and cardiomyocyte lineages. This review focuses on the marked expansion of knowledge in the area of endocardial fate potential. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Atsushi Nakano
- Department of Molecular Cell and Developmental Biology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Haruko Nakano
- Department of Molecular Cell and Developmental Biology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Kelly A Smith
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
37
|
Wu J, Montaniel KRC, Saleh MA, Xiao L, Chen W, Owens GK, Humphrey JD, Majesky MW, Paik DT, Hatzopoulos AK, Madhur MS, Harrison DG. Origin of Matrix-Producing Cells That Contribute to Aortic Fibrosis in Hypertension. Hypertension 2015; 67:461-8. [PMID: 26693821 DOI: 10.1161/hypertensionaha.115.06123] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/08/2015] [Indexed: 11/16/2022]
Abstract
Various hypertensive stimuli lead to exuberant adventitial collagen deposition in large arteries, exacerbating blood pressure elevation and end-organ damage. Collagen production is generally attributed to resident fibroblasts; however, other cells, including resident and bone marrow-derived stem cell antigen positive (Sca-1(+)) cells and endothelial and vascular smooth muscle cells, can produce collagen and contribute to vascular stiffening. Using flow cytometry and immunofluorescence, we found that adventitial Sca-1(+) progenitor cells begin to produce collagen and acquire a fibroblast-like phenotype in hypertension. We also found that bone marrow-derived cells represent more than half of the matrix-producing cells in hypertension, and that one-third of these are Sca-1(+). Cell sorting and lineage-tracing studies showed that cells of endothelial origin contribute to no more than one fourth of adventitial collagen I(+) cells, whereas those of vascular smooth muscle lineage do not contribute. Our findings indicate that Sca-1(+) progenitor cells and bone marrow-derived infiltrating fibrocytes are major sources of arterial fibrosis in hypertension. Endothelial to mesenchymal transition likely also contributes, albeit to a lesser extent and pre-existing resident fibroblasts represent a minority of aortic collagen-producing cells in hypertension. This study shows that vascular stiffening represents a complex process involving recruitment and transformation of multiple cells types that ultimately elaborate adventitial extracellular matrix.
Collapse
Affiliation(s)
- Jing Wu
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Kim Ramil C Montaniel
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Mohamed A Saleh
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Liang Xiao
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Wei Chen
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Gary K Owens
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Jay D Humphrey
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Mark W Majesky
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - David T Paik
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Antonis K Hatzopoulos
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - Meena S Madhur
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.)
| | - David G Harrison
- From the Division of Clinical Pharmacology, Department of Medicine (J.W., K.R.C.M., M.A.S., L.X., W.C., M.S.M., D.G.H.), Department of Molecular Physiology and Biophysics (K.R.C.M., M.S.M., D.G.H.), Division of Cardiovascular Medicine, Department of Medicine (D.T.P., A.K.H.), and Department of Cell and Developmental Biology (D.T.P., A.K.H.), School of Medicine, Vanderbilt University, Nashville, TN; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt (M.A.S.); Robert M. Berne Cardiovascular Research Center, Department of Physiology, University of Virginia, Charlottesville (G.K.O.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.); Seattle Children's Research Institute, WA (M.W.M.).
| |
Collapse
|
38
|
Sultana N, Zhang L, Yan J, Chen J, Cai W, Razzaque S, Jeong D, Sheng W, Bu L, Xu M, Huang GY, Hajjar RJ, Zhou B, Moon A, Cai CL. Resident c-kit(+) cells in the heart are not cardiac stem cells. Nat Commun 2015; 6:8701. [PMID: 26515110 PMCID: PMC4846318 DOI: 10.1038/ncomms9701] [Citation(s) in RCA: 227] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022] Open
Abstract
Identifying a bona fide population of cardiac stem cells (CSCs) is a critical step for developing cell-based therapies for heart failure patients. Previously, cardiac c-kit+ cells were reported to be CSCs with a potential to become myocardial, endothelial and smooth muscle cells in vitro and after cardiac injury. Here we provide further insights into the nature of cardiac c-kit+ cells. By targeting the c-kit locus with multiple reporter genes in mice, we find that c-kit expression rarely co-localizes with the expression of the cardiac progenitor and myogenic marker Nkx2.5, or that of the myocardial marker, cardiac troponin T (cTnT). Instead, c-kit predominantly labels a cardiac endothelial cell population in developing and adult hearts. After acute cardiac injury, c-kit+ cells retain their endothelial identity and do not become myogenic progenitors or cardiomyocytes. Thus, our work strongly suggests that c-kit+ cells in the murine heart are endothelial cells and not CSCs. The issue whether the cell surface protein c-kit identifies resident cardiac stem cells (CSC) is controversial. By using novel reporter mouse models, Sultana et al. show that c-kit+ cells represent a subpopulation of endothelial cells in the developing and adult heart and do not exhibit CSC traits in health or disease.
Collapse
Affiliation(s)
- Nishat Sultana
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Lu Zhang
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jianyun Yan
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jiqiu Chen
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Weibin Cai
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Shegufta Razzaque
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Dongtak Jeong
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Wei Sheng
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Lei Bu
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York 10016, USA
| | - Mingjiang Xu
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Guo-Ying Huang
- Cardiovascular Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Roger J Hajjar
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York 10461, USA
| | - Anne Moon
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania 17822, USA
| | - Chen-Leng Cai
- Department of Developmental and Regenerative Biology, The Black Family Stem Cell Institute, and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
39
|
Valiente-Alandi I, Albo-Castellanos C, Herrero D, Arza E, Garcia-Gomez M, Segovia JC, Capecchi M, Bernad A. Cardiac Bmi1(+) cells contribute to myocardial renewal in the murine adult heart. Stem Cell Res Ther 2015; 6:205. [PMID: 26503423 PMCID: PMC4620653 DOI: 10.1186/s13287-015-0196-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/15/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
Introduction The mammalian adult heart maintains a continuous, low cardiomyocyte turnover rate throughout life. Although many cardiac stem cell populations have been studied, the natural source for homeostatic repair has not yet been defined. The Polycomb protein BMI1 is the most representative marker of mouse adult stem cell systems. We have evaluated the relevance and role of cardiac Bmi1+ cells in cardiac physiological homeostasis. Methods Bmi1CreER/+;Rosa26YFP/+ (Bmi1-YFP) mice were used for lineage tracing strategy. After tamoxifen (TM) induction, yellow fluorescent protein (YFP) is expressed under the control of Rosa26 regulatory sequences in Bmi1+ cells. These cells and their progeny were tracked by FACS, immunofluorescence and RT-qPCR techniques from 5 days to 1 year. Results FACS analysis of non-cardiomyocyte compartment from TM-induced Bmi1-YFP mice showed a Bmi1+-expressing cardiac progenitor cell (Bmi1-CPC: B-CPC) population, SCA-1 antigen-positive (95.9 ± 0.4 %) that expresses some stemness-associated genes. B-CPC were also able to differentiate in vitro to the three main cardiac lineages. Pulse-chase analysis showed that B-CPC remained quite stable for extended periods (up to 1 year), which suggests that this Bmi1+ population contains cardiac progenitors with substantial self-maintenance potential. Specific immunostaining of Bmi1-YFP hearts serial sections 5 days post-TM induction indicated broad distribution of B-CPC, which were detected in variably sized clusters, although no YFP+ cardiomyocytes (CM) were detected at this time. Between 2 to 12 months after TM induction, YFP+ CM were clearly identified (3 ± 0.6 % to 6.7 ± 1.3 %) by immunohistochemistry of serial sections and by flow cytometry of total freshly isolated CM. B-CPC also contributed to endothelial and smooth muscle (SM) lineages in vivo. Conclusions High Bmi1 expression identifies a non-cardiomyocyte resident cardiac population (B-CPC) that contributes to the main lineages of the heart in vitro and in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0196-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iñigo Valiente-Alandi
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Carmen Albo-Castellanos
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Vivebiotech, San Sebastian, Spain.
| | - Diego Herrero
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| | - Elvira Arza
- Microscopy Unit, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
| | - Maria Garcia-Gomez
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - José C Segovia
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - Mario Capecchi
- Howard Hughes Medical Institute University of Utah, Salt Lake City, UT, USA.
| | - Antonio Bernad
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
40
|
Abstract
The degree to which cKit-expressing progenitors generate cardiomyocytes in the heart is controversial. Genetic fate-mapping studies suggest minimal contribution; however, whether or not minimal contribution reflects minimal cardiomyogenic capacity is unclear because the embryonic origin and role in cardiogenesis of these progenitors remain elusive. Using high-resolution genetic fate-mapping approaches with cKit(CreERT2/+) and Wnt1::Flpe mouse lines, we show that cKit delineates cardiac neural crest progenitors (CNC(kit)). CNC(kit) possess full cardiomyogenic capacity and contribute to all CNC derivatives, including cardiac conduction system cells. Furthermore, by modeling cardiogenesis in cKit(CreERT2)-induced pluripotent stem cells, we show that, paradoxically, the cardiogenic fate of CNC(kit) is regulated by bone morphogenetic protein antagonism, a signaling pathway activated transiently during establishment of the cardiac crescent, and extinguished from the heart before CNC invasion. Together, these findings elucidate the origin of cKit(+) cardiac progenitors and suggest that a nonpermissive cardiac milieu, rather than minimal cardiomyogenic capacity, controls the degree of CNC(kit) contribution to myocardium.
Collapse
|
41
|
Paik DT, Rai M, Ryzhov S, Sanders LN, Aisagbonhi O, Funke MJ, Feoktistov I, Hatzopoulos AK. Wnt10b Gain-of-Function Improves Cardiac Repair by Arteriole Formation and Attenuation of Fibrosis. Circ Res 2015; 117:804-16. [PMID: 26338900 DOI: 10.1161/circresaha.115.306886] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/03/2015] [Indexed: 01/10/2023]
Abstract
RATIONALE Myocardial infarction causes irreversible tissue damage, leading to heart failure. We recently discovered that canonical Wnt signaling and the Wnt10b ligand are strongly induced in mouse hearts after infarction. Wnt10b regulates cell fate in various organs, but its role in the heart is unknown. OBJECTIVE To investigate the effect of Wnt10b gain-of-function on cardiac repair mechanisms and to assess its potential to improve ventricular function after injury. METHODS AND RESULTS Histological and molecular analyses showed that Wnt10b is expressed in cardiomyocytes and localized in the intercalated discs of mouse and human hearts. After coronary artery ligation or cryoinjury in mice, Wnt10b is strongly and transiently induced in peri-infarct cardiomyocytes during granulation tissue formation. To determine the effect of Wnt10b on neovascularization and fibrosis, we generated a mouse line to increase endogenous Wnt10b levels in cardiomyocytes. We found that gain of Wnt10b function orchestrated a recovery phenotype characterized by robust neovascularization of the injury zone, less myofibroblasts, reduced scar size, and improved ventricular function compared with wild-type mice. Wnt10b stimulated expression of vascular endothelial growth factor receptor 2 in endothelial cells and angiopoietin-1 in vascular smooth muscle cells through nuclear factor-κB activation. These effects coordinated endothelial growth and smooth muscle cell recruitment, promoting robust formation of large, coronary-like blood vessels. CONCLUSION Wnt10b gain-of-function coordinates arterial formation and attenuates fibrosis in cardiac tissue after injury. Because generation of mature blood vessels is necessary for efficient perfusion, our findings could lead to novel strategies to optimize the inherent repair capacity of the heart and prevent the onset of heart failure.
Collapse
Affiliation(s)
- David T Paik
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Meena Rai
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Sergey Ryzhov
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Lehanna N Sanders
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Omonigho Aisagbonhi
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Mitchell J Funke
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Igor Feoktistov
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.)
| | - Antonis K Hatzopoulos
- From the Division of Cardiovascular Medicine, Department of Medicine (D.T.P., M.R., S.R., L.N.S., O.A., M.J.F., I.F., A.K.H.), Department of Cell and Developmental Biology (D.T.P., M.R., L.N.S., O.A., A.K.H.), and Department of Pharmacology, Vanderbilt University, Nashville, TN (I.F.); Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (S.R.); Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston (O.A.); and Golden Rule Medical, Cincinnati, OH (M.J.F.).
| |
Collapse
|
42
|
Goss GM, Chaudhari N, Hare JM, Nwojo R, Seidler B, Saur D, Goldstein BJ. Differentiation potential of individual olfactory c-Kit+ progenitors determined via multicolor lineage tracing. Dev Neurobiol 2015; 76:241-51. [PMID: 26016700 DOI: 10.1002/dneu.22310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/14/2015] [Accepted: 05/22/2015] [Indexed: 11/10/2022]
Abstract
Olfactory tissue undergoes lifelong renewal, due to the presence of basal neural stem cells. Multiple categories of globose basal stem cells have been identified, expressing markers such as Lgr5, Ascl1, GBC-2, and c-Kit. The differentiation potential of individual globose cells has remained unclear. Here, we utilized Cre/loxP lineage tracing with a multicolor reporter system to define c-Kit+ cell contributions at clonal resolution. We determined that reporter expression permitted identification of c-Kit derived progeny with fine cellular detail, and that clones were found to be comprised by neurons only, microvillar cells only, microvillar cells and neurons, or gland/duct cells. Quantification of reporter-labeled cells indicated that c-Kit+ cells behave as transit amplifying or immediate precursors, although we also found evidence for longer-term c-Kit+ cell contributions. Our results from the application of multicolor fate mapping delineate the clonal contributions of c-Kit+ cells to olfactory epithelial renewal, and provide novel insight into tissue maintenance of an adult neuroepithelium.
Collapse
Affiliation(s)
- Garrett M Goss
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Nirupa Chaudhari
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Raphael Nwojo
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Barbara Seidler
- Department of Internal Medicine, Technical University of Munich, München, Germany
| | - Dieter Saur
- Department of Internal Medicine, Technical University of Munich, München, Germany
| | - Bradley J Goldstein
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
43
|
Keith MCL, Bolli R. "String theory" of c-kit(pos) cardiac cells: a new paradigm regarding the nature of these cells that may reconcile apparently discrepant results. Circ Res 2015; 116:1216-30. [PMID: 25814683 DOI: 10.1161/circresaha.116.305557] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although numerous preclinical investigations have consistently demonstrated salubrious effects of c-kit(pos) cardiac cells administered after myocardial infarction, the mechanism of action remains highly controversial. We and others have found little or no evidence that these cells differentiate into mature functional cardiomyocytes, suggesting paracrine effects. In this review, we propose a new paradigm predicated on a comprehensive analysis of the literature, including studies of cardiac development; we have (facetiously) dubbed this conceptual construct "string theory" of c-kit(pos) cardiac cells because it reconciles multifarious and sometimes apparently discrepant results. There is strong evidence that, during development, the c-kit receptor is expressed in different pools of cardiac progenitors (some capable of robust cardiomyogenesis and others with little or no contribution to myocytes). Accordingly, c-kit positivity, in itself, does not define the embryonic origins, lineage capabilities, or differentiation capacities of specific cardiac progenitors. C-kit(pos) cells derived from the first heart field exhibit cardiomyogenic potential during development, but these cells are likely depleted shortly before or after birth. The residual c-kit(pos) cells found in the adult heart are probably of proepicardial origin, possess a mesenchymal phenotype (resembling bone marrow mesenchymal stem/stromal cells), and are capable of contributing significantly only to nonmyocytic lineages (fibroblasts, smooth muscle cells, and endothelial cells). If these 2 populations (first heart field and proepicardium) express different levels of c-kit, the cardiomyogenic potential of first heart field progenitors might be reconciled with recent results of c-kit(pos) cell lineage tracing studies. The concept that c-kit expression in the adult heart identifies epicardium-derived, noncardiomyogenic precursors with a mesenchymal phenotype helps to explain the beneficial effects of c-kit(pos) cell administration to ischemically damaged hearts despite the observed paucity of cardiomyogenic differentiation of these cells.
Collapse
Affiliation(s)
- Matthew C L Keith
- From the Division of Cardiovascular Medicine, Department of Cardiology, University of Louisville, KY
| | - Roberto Bolli
- From the Division of Cardiovascular Medicine, Department of Cardiology, University of Louisville, KY.
| |
Collapse
|
44
|
Optogenetic intervention to the vascular endothelium. Vascul Pharmacol 2015; 74:122-129. [PMID: 26015375 DOI: 10.1016/j.vph.2015.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 05/08/2015] [Accepted: 05/16/2015] [Indexed: 11/21/2022]
Abstract
Endothelium lining the interior of cardiovascular system and most visceral organs plays an important role in vascular function. Its dysfunction occurs in some of the most challenging diseases. An important function of the endothelium is to release vasoactive substances that act on the smooth muscle to change vascular tones. Substance secretion from endocrine cells relies on membrane potentials and firing activity, while it is unclear whether the membrane potential regulates substance release from the ECs. Understanding of this requires selective intervention to membrane potentials of the endothelial cells in situ. Here we show a novel intervention to endothelial cells using the optogenetic approach. A strain of transgenic mice was developed with the Cre-loxP recombination system. These transgenic mice expressed channelrhodopsin (ChR) in endothelial cells driven by the vascular endothelial cadherin or cdh5 promoter. Linked in a tandem with YFP, the ChR expression was detected by YFP fluorescence in various endothelium-lining tissues and organs. The YFP fluorescence was observed in the lumen of blood vessels and pericardium, but not in tissues beneath the endothelium lining. Optostimulation of dissociated endothelial cells evoked inward currents and depolarization. In the isolated and perfused heart, surprisingly, optostimulation of endothelial cells produced fast, robust, reproducible and long-lasting vasoconstriction that was not blocked by either ET-1A or TXA2 receptor antagonist. Similar optical vasoconstriction was found in the isolated and perfused kidney. These results indicate that the optogenetics is an effective intervention to vascular endothelium where optostimulation produces vasoconstriction.
Collapse
|
45
|
Leite CF, Almeida TR, Lopes CS, Dias da Silva VJ. Multipotent stem cells of the heart-do they have therapeutic promise? Front Physiol 2015; 6:123. [PMID: 26005421 PMCID: PMC4424849 DOI: 10.3389/fphys.2015.00123] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/06/2015] [Indexed: 01/26/2023] Open
Abstract
The last decade has brought a comprehensive change in our view of cardiac remodeling processes under both physiological and pathological conditions, and cardiac stem cells have become important new players in the general mainframe of cardiac homeostasis. Different types of cardiac stem cells show different capacities for differentiation into the three major cardiac lineages: myocytes, endothelial cells and smooth muscle cells. Physiologically, cardiac stem cells contribute to cardiac homeostasis through continual cellular turnover. Pathologically, these cells exhibit a high level of proliferative activity in an apparent attempt to repair acute cardiac injury, indicating that these cells possess (albeit limited) regenerative potential. In addition to cardiac stem cells, mesenchymal stem cells represent another multipotent cell population in the heart; these cells are located in regions near pericytes and exhibit regenerative, angiogenic, antiapoptotic, and immunosuppressive properties. The discovery of these resident cardiac stem cells was followed by a number of experimental studies in animal models of cardiomyopathies, in which cardiac stem cells were tested as a therapeutic option to overcome the limited transdifferentiating potential of hematopoietic or mesenchymal stem cells derived from bone marrow. The promising results of these studies prompted clinical studies of the role of these cells, which have demonstrated the safety and practicability of cellular therapies for the treatment of heart disease. However, questions remain regarding this new therapeutic approach. Thus, the aim of the present review was to discuss the multitude of different cardiac stem cells that have been identified, their possible functional roles in the cardiac regenerative process, and their potential therapeutic uses in treating cardiac diseases.
Collapse
Affiliation(s)
- Camila F Leite
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Thalles R Almeida
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Carolina S Lopes
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Valdo J Dias da Silva
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| |
Collapse
|
46
|
An emerging consensus on cardiac regeneration. Nat Med 2015; 20:1386-93. [PMID: 25473919 DOI: 10.1038/nm.3764] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 10/31/2014] [Indexed: 12/22/2022]
Abstract
Cardiac regeneration is a rapidly evolving and controversial field of research. The identification some 12 years ago of progenitor cells that reside within the heart spurred enthusiasm for cell-based regenerative therapies. However, recent evidence has called into question both the presence of a biologically important stem cell population in the heart and the ability of exogenously derived cells to promote regeneration through direct formation of new cardiomyocytes. Here, we discuss recent developments that suggest an emerging consensus on the ability of different cell types to regenerate the adult mammalian heart.
Collapse
|
47
|
Raynaud CM, Ahmad FS, Allouba M, Abou-Saleh H, Lui KO, Yacoub M. Reprogramming for cardiac regeneration. Glob Cardiol Sci Pract 2014; 2014:309-29. [PMID: 25763379 PMCID: PMC4352683 DOI: 10.5339/gcsp.2014.44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/18/2014] [Indexed: 01/10/2023] Open
Abstract
Treatment of cardiovascular diseases remains challenging considering the limited regeneration capacity of the heart muscle. Developments of reprogramming strategies to create in vitro and in vivo cardiomyocytes have been the focus point of a considerable amount of research in the past decades. The choice of cells to employ, the state-of-the-art methods for different reprogramming strategies, and their promises and future challenges before clinical entry, are all discussed here.
Collapse
Affiliation(s)
| | | | - Mona Allouba
- Aswan Heart Center, Magdi Yacoub Foundation, Aswan, Egypt
| | - Haissam Abou-Saleh
- Qatar Cardiovascular Research Center, Qatar Foundation-Education City, Doha, Qatar
| | - Kathy O Lui
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, USA
| | | |
Collapse
|