101
|
Liu T, Wu H, Wu S, Wang C. Single-Cell Sequencing Technologies for Cardiac Stem Cell Studies. Stem Cells Dev 2017; 26:1540-1551. [PMID: 28859577 DOI: 10.1089/scd.2017.0050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Today with the rapid advancements in stem cell studies and the promising potential of using stem cells in clinical therapy, there is an increasing demand for in-depth comprehensive analysis on individual cell transcriptome and epigenome, as they play critical roles in a number of cell functions such as cell differentiation, growth, and reprogramming. The development of single-cell sequencing technologies has helped in revealing some exciting new perspectives in stem cells and regenerative medicine research. Among the various potential applications, single-cell analysis for cardiac stem cells (CSCs) holds tremendous promises in understanding the mechanisms of heart development and regeneration, which might light up the path toward cell therapy for cardiovascular diseases. This review briefly highlights the recent progresses in single-cell sequencing analysis technologies and their applications in CSC research.
Collapse
Affiliation(s)
- Tiantian Liu
- 1 Center for Genomics & Department of Basic Sciences, School of Medicine, Loma Linda University , Loma Linda, California
| | - Hongjin Wu
- 1 Center for Genomics & Department of Basic Sciences, School of Medicine, Loma Linda University , Loma Linda, California.,2 Cancer Research Institute, Hangzhou Cancer Hospital , Hangzhou, Zhejiang Province, P.R. China
| | - Shixiu Wu
- 2 Cancer Research Institute, Hangzhou Cancer Hospital , Hangzhou, Zhejiang Province, P.R. China
| | - Charles Wang
- 1 Center for Genomics & Department of Basic Sciences, School of Medicine, Loma Linda University , Loma Linda, California
| |
Collapse
|
102
|
Milstone ZJ, Lawson G, Trivedi CM. Histone deacetylase 1 and 2 are essential for murine neural crest proliferation, pharyngeal arch development, and craniofacial morphogenesis. Dev Dyn 2017; 246:1015-1026. [PMID: 28791750 DOI: 10.1002/dvdy.24563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Craniofacial anomalies involve defective pharyngeal arch development and neural crest function. Copy number variation at 1p35, containing histone deacetylase 1 (Hdac1), or 6q21-22, containing Hdac2, are implicated in patients with craniofacial defects, suggesting an important role in guiding neural crest development. However, the roles of Hdac1 and Hdac2 within neural crest cells remain unknown. RESULTS The neural crest and its derivatives express both Hdac1 and Hdac2 during early murine development. Ablation of Hdac1 and Hdac2 within murine neural crest progenitor cells cause severe hemorrhage, atrophic pharyngeal arches, defective head morphogenesis, and complete embryonic lethality. Embryos lacking Hdac1 and Hdac2 in the neural crest exhibit decreased proliferation and increased apoptosis in both the neural tube and the first pharyngeal arch. Mechanistically, loss of Hdac1 and Hdac2 upregulates cyclin-dependent kinase inhibitors Cdkn1a, Cdkn1b, Cdkn1c, Cdkn2b, Cdkn2c, and Tp53 within the first pharyngeal arch. CONCLUSIONS Our results show that Hdac1 and Hdac2 function redundantly within the neural crest to regulate proliferation and the development of the pharyngeal arches by means of repression of cyclin-dependent kinase inhibitors. Developmental Dynamics 246:1015-1026, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zachary J Milstone
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Grace Lawson
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Chinmay M Trivedi
- Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.,Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
103
|
The epicardium as a source of multipotent adult cardiac progenitor cells: Their origin, role and fate. Pharmacol Res 2017; 127:129-140. [PMID: 28751220 DOI: 10.1016/j.phrs.2017.07.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/12/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
Since the regenerative capacity of the adult mammalian heart is limited, cardiac injury leads to the formation of scar tissue and thereby increases the risk of developing compensatory heart failure. Stem cell therapy is a promising therapeutic approach but is facing problems with engraftment and clinical feasibility. Targeting an endogenous stem cell population could circumvent these limitations. The epicardium, a membranous layer covering the outside of the myocardium, is an accessible cell population which plays a key role in the developing heart. Epicardial cells undergo epithelial to mesenchymal transition (EMT), thus providing epicardial derived cells (EPDCs) that migrate into the myocardium and cooperate in myocardial vascularisation and compaction. In the adult heart, injury activates the epicardium, and an embryonic-like response is observed which includes EMT and differentiation of the EPDCs into cardiac cell types. Furthermore, paracrine communication between the epicardium and myocardium improves the regenerative response. The significant role of the epicardium has been shown in both the developing and the regenerating heart. Interestingly, the epicardial contribution to cardiac repair can be improved in several ways. In this review, an overview of the epicardial origin and fate will be given and potential therapeutic approaches will be discussed.
Collapse
|
104
|
Hasin T, Iakobishvili Z, Weisz G. Associated Risk of Malignancy in Patients with Cardiovascular Disease: Evidence and Possible Mechanism. Am J Med 2017; 130:780-785. [PMID: 28344133 DOI: 10.1016/j.amjmed.2017.02.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease and malignancy are leading causes of morbidity and mortality. Increased risk of malignancy was identified in patients with cardiovascular disease, including patients with heart failure, heart failure after myocardial infarction, patients undergoing cardiac intervention, and patients after a thrombotic event. Common risk factors and biological pathways can explain this association and are explored in this review. Further research is needed to establish the causes of malignancy in this population and direct possible intervention.
Collapse
Affiliation(s)
- Tal Hasin
- Department of Cardiology, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Zaza Iakobishvili
- Department of Cardiology, Rabin Medical Center, Petach Tiqwa, Israel
| | - Giora Weisz
- Department of Cardiology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
105
|
[Genetics of congenital heart diseases]. Presse Med 2017; 46:612-619. [PMID: 28583745 DOI: 10.1016/j.lpm.2017.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/01/2017] [Accepted: 05/11/2017] [Indexed: 01/24/2023] Open
Abstract
Developmental genetics of congenital heart diseases has evolved from analysis of serial slices in embryos towards molecular genetics of cardiac morphogenesis with a dynamic view of cardiac development. Genetics of congenital heart diseases has also changed from formal genetic analysis of familial recurrences or population-based analysis to screening for mutations in candidates genes identified in animal models. Close cooperation between molecular embryologists, pathologists involved in heart development and pediatric cardiologists is crucial for further increase of knowledge in the field of cardiac morphogenesis and genetics of cardiac defects. The genetic model for congenital heart disease has to be revised to favor a polygenic origin rather than a monogenic one. The main mechanism is altered genic dosage that can account for heart diseases in chromosomal anomalies as well as in point mutations in syndromic and isolated congenital heart diseases. The use of big data grouping information from cardiac development, interactions between genes and proteins, epigenetic factors such as chromatin remodeling or DNA methylation is the current source for improving our knowledge in the field and to give clues for future therapies.
Collapse
|
106
|
Ocorr K, Zambon A, Nudell Y, Pineda S, Diop S, Tang M, Akasaka T, Taylor E. Age-dependent electrical and morphological remodeling of the Drosophila heart caused by hERG/seizure mutations. PLoS Genet 2017; 13:e1006786. [PMID: 28542428 PMCID: PMC5459509 DOI: 10.1371/journal.pgen.1006786] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 06/05/2017] [Accepted: 04/30/2017] [Indexed: 12/03/2022] Open
Abstract
Understanding the cellular-molecular substrates of heart disease is key to the development of cardiac specific therapies and to the prevention of off-target effects by non-cardiac targeted drugs. One of the primary targets for therapeutic intervention has been the human ether a go-go (hERG) K+ channel that, together with the KCNQ channel, controls the rate and efficiency of repolarization in human myocardial cells. Neither of these channels plays a major role in adult mouse heart function; however, we show here that the hERG homolog seizure (sei), along with KCNQ, both contribute significantly to adult heart function as they do in humans. In Drosophila, mutations in or cardiac knockdown of sei channels cause arrhythmias that become progressively more severe with age. Intracellular recordings of semi-intact heart preparations revealed that these perturbations also cause electrical remodeling that is reminiscent of the early afterdepolarizations seen in human myocardial cells defective in these channels. In contrast to KCNQ, however, mutations in sei also cause extensive structural remodeling of the myofibrillar organization, which suggests that hERG channel function has a novel link to sarcomeric and myofibrillar integrity. We conclude that deficiency of ion channels with similar electrical functions in cardiomyocytes can lead to different types or extents of electrical and/or structural remodeling impacting cardiac output. We have used the fruit fly cardiac model to show that seizure, the fly homolog of the human ether a go-go K+ channel hERG, is functional in the fly heart. This channel plays a major role in cardiac repolarization in humans but not in adult rodent hearts. Loss of channel function in the fly causes bradycardia, electrical arrhythmia and altered myofibrillar structure. Gene expression analysis indicates that Wnt signaling is affected and we show a genetic interaction between sei and pygopus, a Wnt pathway component, on heart function.
Collapse
Affiliation(s)
- Karen Ocorr
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
- * E-mail:
| | - Alexander Zambon
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Yoav Nudell
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Santiago Pineda
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Soda Diop
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Min Tang
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Takeshi Akasaka
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Erika Taylor
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, United States of America
| |
Collapse
|
107
|
Desjardins CA, Naya FJ. Antagonistic regulation of cell-cycle and differentiation gene programs in neonatal cardiomyocytes by homologous MEF2 transcription factors. J Biol Chem 2017; 292:10613-10629. [PMID: 28473466 DOI: 10.1074/jbc.m117.776153] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/03/2017] [Indexed: 12/30/2022] Open
Abstract
Cardiomyocytes acquire their primary specialized function (contraction) before exiting the cell cycle. In this regard, proliferation and differentiation must be precisely coordinated for proper cardiac morphogenesis. Here, we have investigated the complex transcriptional mechanisms employed by cardiomyocytes to coordinate antagonistic cell-cycle and differentiation gene programs through the molecular dissection of the core cardiac transcription factor, MEF2. Knockdown of individual MEF2 proteins, MEF2A, -C, and -D, in primary neonatal cardiomyocytes resulted in radically distinct and opposite effects on cellular homeostasis and gene regulation. MEF2A and MEF2D were absolutely required for cardiomyocyte survival, whereas MEF2C, despite its major role in cardiac morphogenesis and direct reprogramming, was dispensable for this process. Inhibition of MEF2A or -D also resulted in the activation of cell-cycle genes and down-regulation of markers of terminal differentiation. In striking contrast, the regulation of cell-cycle and differentiation gene programs by MEF2C was antagonistic to that of MEF2A and -D. Computational analysis of regulatory regions from MEF2 isoform-dependent gene sets identified the Notch and Hedgehog signaling pathways as key determinants in coordinating MEF2 isoform-specific control of antagonistic gene programs. These results reveal that mammalian MEF2 family members have distinct transcriptional functions in cardiomyocytes and suggest that these differences are critical for proper development and maturation of the heart. Analysis of MEF2 isoform-specific function in neonatal cardiomyocytes has yielded insight into an unexpected transcriptional regulatory mechanism by which these specialized cells utilize homologous members of a core cardiac transcription factor to coordinate cell-cycle and differentiation gene programs.
Collapse
Affiliation(s)
- Cody A Desjardins
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Francisco J Naya
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
108
|
Galdos FX, Guo Y, Paige SL, VanDusen NJ, Wu SM, Pu WT. Cardiac Regeneration: Lessons From Development. Circ Res 2017; 120:941-959. [PMID: 28302741 DOI: 10.1161/circresaha.116.309040] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 02/06/2023]
Abstract
Palliative surgery for congenital heart disease has allowed patients with previously lethal heart malformations to survive and, in most cases, to thrive. However, these procedures often place pressure and volume loads on the heart, and over time, these chronic loads can cause heart failure. Current therapeutic options for initial surgery and chronic heart failure that results from failed palliation are limited, in part, by the mammalian heart's low inherent capacity to form new cardiomyocytes. Surmounting the heart regeneration barrier would transform the treatment of congenital, as well as acquired, heart disease and likewise would enable development of personalized, in vitro cardiac disease models. Although these remain distant goals, studies of heart development are illuminating the path forward and suggest unique opportunities for heart regeneration, particularly in fetal and neonatal periods. Here, we review major lessons from heart development that inform current and future studies directed at enhancing cardiac regeneration.
Collapse
Affiliation(s)
- Francisco X Galdos
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Yuxuan Guo
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Sharon L Paige
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Nathan J VanDusen
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Sean M Wu
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| | - William T Pu
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| |
Collapse
|
109
|
Reduced dosage of β-catenin provides significant rescue of cardiac outflow tract anomalies in a Tbx1 conditional null mouse model of 22q11.2 deletion syndrome. PLoS Genet 2017; 13:e1006687. [PMID: 28346476 PMCID: PMC5386301 DOI: 10.1371/journal.pgen.1006687] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/10/2017] [Accepted: 03/13/2017] [Indexed: 11/19/2022] Open
Abstract
The 22q11.2 deletion syndrome (22q11.2DS; velo-cardio-facial syndrome; DiGeorge syndrome) is a congenital anomaly disorder in which haploinsufficiency of TBX1, encoding a T-box transcription factor, is the major candidate for cardiac outflow tract (OFT) malformations. Inactivation of Tbx1 in the anterior heart field (AHF) mesoderm in the mouse results in premature expression of pro-differentiation genes and a persistent truncus arteriosus (PTA) in which septation does not form between the aorta and pulmonary trunk. Canonical Wnt/β-catenin has major roles in cardiac OFT development that may act upstream of Tbx1. Consistent with an antagonistic relationship, we found the opposite gene expression changes occurred in the AHF in β-catenin loss of function embryos compared to Tbx1 loss of function embryos, providing an opportunity to test for genetic rescue. When both alleles of Tbx1 and one allele of β-catenin were inactivated in the Mef2c-AHF-Cre domain, 61% of them (n = 34) showed partial or complete rescue of the PTA defect. Upregulated genes that were oppositely changed in expression in individual mutant embryos were normalized in significantly rescued embryos. Further, β-catenin was increased in expression when Tbx1 was inactivated, suggesting that there may be a negative feedback loop between canonical Wnt and Tbx1 in the AHF to allow the formation of the OFT. We suggest that alteration of this balance may contribute to variable expressivity in 22q11.2DS.
Collapse
|
110
|
Li YJ, Yang YQ. An update on the molecular diagnosis of congenital heart disease: focus on loss-of-function mutations. Expert Rev Mol Diagn 2017; 17:393-401. [PMID: 28274167 DOI: 10.1080/14737159.2017.1300062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yan-Jie Li
- Department of Cardiology, Cardiovascular Research Laboratory, and Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi-Qing Yang
- Department of Cardiology, Cardiovascular Research Laboratory, and Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
111
|
Zamir L, Singh R, Nathan E, Patrick R, Yifa O, Yahalom-Ronen Y, Arraf AA, Schultheiss TM, Suo S, Han JDJ, Peng G, Jing N, Wang Y, Palpant N, Tam PP, Harvey RP, Tzahor E. Nkx2.5 marks angioblasts that contribute to hemogenic endothelium of the endocardium and dorsal aorta. eLife 2017; 6:20994. [PMID: 28271994 PMCID: PMC5400512 DOI: 10.7554/elife.20994] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 03/06/2017] [Indexed: 01/10/2023] Open
Abstract
Novel regenerative therapies may stem from deeper understanding of the mechanisms governing cardiovascular lineage diversification. Using enhancer mapping and live imaging in avian embryos, and genetic lineage tracing in mice, we investigated the spatio-temporal dynamics of cardiovascular progenitor populations. We show that expression of the cardiac transcription factor Nkx2.5 marks a mesodermal population outside of the cardiac crescent in the extraembryonic and lateral plate mesoderm, with characteristics of hemogenic angioblasts. Extra-cardiac Nkx2.5 lineage progenitors migrate into the embryo and contribute to clusters of CD41+/CD45+ and RUNX1+ cells in the endocardium, the aorta-gonad-mesonephros region of the dorsal aorta and liver. We also demonstrated that ectopic expression of Nkx2.5 in chick embryos activates the hemoangiogenic gene expression program. Taken together, we identified a hemogenic angioblast cell lineage characterized by transient Nkx2.5 expression that contributes to hemogenic endothelium and endocardium, suggesting a novel role for Nkx2.5 in hemoangiogenic lineage specification and diversification. DOI:http://dx.doi.org/10.7554/eLife.20994.001 As an animal embryo develops, it establishes a circulatory system that includes the heart, vessels and blood. Vessels and blood initially form in the yolk sac, a membrane that surrounds the embryo. These yolk sac vessels act as a rudimentary circulatory system, connecting to the heart and blood vessels within the embryo itself. In older embryos, cells in the inner layer of the largest blood vessel (known as the dorsal aorta) generate blood stem cells that give rise to the different types of blood cells. A gene called Nkx2.5 encodes a protein that controls the activity of a number of complex genetic programs and has been long studied as a key player in the development of the heart. Nkx2.5 is essential for forming normal heart muscle cells and for shaping the primitive heart and its surrounding vessels into a working organ. Interfering with the normal activity of the Nkx2.5 gene results in severe defects in blood vessels and the heart. However, many details are missing on the role played by Nkx2.5 in specifying the different cellular components of the circulatory system and heart. Zamir et al. genetically engineered chick and mouse embryos to produce fluorescent markers that could be used to trace the cells that become part of blood vessels and heart. The experiments found that some of the cells that form the blood and vessels in the yolk sac originate from within the membranes surrounding the embryo, outside of the areas previously reported to give rise to the heart. The Nkx2.5 gene is active in these cells for only a short period of time as they migrate toward the heart and dorsal aorta, where they give rise to blood stem cells These findings suggest that Nkx2.5 plays an important role in triggering developmental processes that eventually give rise to blood vessels and blood cells. The next step following on from this work will be to find out what genes the protein encoded by Nkx2.5 regulates to drive these processes. Mapping the genes that control the early origins of blood and blood-forming vessels will help biologists understand this complex and vital tissue system, and develop new treatments for patients with conditions that affect their circulatory system. In the future, this knowledge may also help to engineer synthetic blood and blood products for use in trauma and genetic diseases. DOI:http://dx.doi.org/10.7554/eLife.20994.002
Collapse
Affiliation(s)
- Lyad Zamir
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Reena Singh
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Elisha Nathan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Oren Yifa
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yfat Yahalom-Ronen
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alaa A Arraf
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shengbao Suo
- Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing-Dong Jackie Han
- Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guangdun Peng
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, The University of Washington, Seattle, United States
| | - Nathan Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Patrick Pl Tam
- School of Medical Sciences, Sydney Medical School, The University of Sydney, Westmead, Australia.,Embryology Unit, Children's Medical Research Institute, Westmead, Australia
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Sydney, Australia.,St. Vincent's Clinical School, School of Biological and Biomolecular Sciences, University of New South Wales, Kensington, Australia
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
112
|
Wu B, Wang Y, Xiao F, Butcher JT, Yutzey KE, Zhou B. Developmental Mechanisms of Aortic Valve Malformation and Disease. Annu Rev Physiol 2017; 79:21-41. [DOI: 10.1146/annurev-physiol-022516-034001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Bingruo Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
| | - Yidong Wang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
| | - Feng Xiao
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029 China
| | - Jonathan T. Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853;
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio 45229;
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York 10461;
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029 China
| |
Collapse
|
113
|
Isl2b regulates anterior second heart field development in zebrafish. Sci Rep 2017; 7:41043. [PMID: 28106108 PMCID: PMC5247716 DOI: 10.1038/srep41043] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
After initial formation, the heart tube grows by addition of second heart field progenitor cells to its poles. The transcription factor Isl1 is expressed in the entire second heart field in mouse, and Isl1-deficient mouse embryos show defects in arterial and venous pole development. The expression of Isl1 is conserved in zebrafish cardiac progenitors; however, Isl1 is required for cardiomyocyte differentiation only at the venous pole. Here we show that Isl1 homologues are expressed in specific patterns in the developing zebrafish heart and play distinct roles during cardiac morphogenesis. In zebrafish, isl2a mutants show defects in cardiac looping, whereas isl2b is required for arterial pole development. Moreover, Isl2b controls the expression of key cardiac transcription factors including mef2ca, mef2cb, hand2 and tbx20. The specific roles of individual Islet family members in the development of distinct regions of the zebrafish heart renders this system particularly well-suited for dissecting Islet-dependent gene regulatory networks controlling the behavior and function of second heart field progenitors in distinct steps of cardiac development.
Collapse
|
114
|
Bloomekatz J, Singh R, Prall OW, Dunn AC, Vaughan M, Loo CS, Harvey RP, Yelon D. Platelet-derived growth factor (PDGF) signaling directs cardiomyocyte movement toward the midline during heart tube assembly. eLife 2017; 6:21172. [PMID: 28098558 PMCID: PMC5298878 DOI: 10.7554/elife.21172] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/17/2017] [Indexed: 01/23/2023] Open
Abstract
Communication between neighboring tissues plays a central role in guiding organ morphogenesis. During heart tube assembly, interactions with the adjacent endoderm control the medial movement of cardiomyocytes, a process referred to as cardiac fusion. However, the molecular underpinnings of this endodermal-myocardial relationship remain unclear. Here, we show an essential role for platelet-derived growth factor receptor alpha (Pdgfra) in directing cardiac fusion. Mutation of pdgfra disrupts heart tube assembly in both zebrafish and mouse. Timelapse analysis of individual cardiomyocyte trajectories reveals misdirected cells in zebrafish pdgfra mutants, suggesting that PDGF signaling steers cardiomyocytes toward the midline during cardiac fusion. Intriguingly, the ligand pdgfaa is expressed in the endoderm medial to the pdgfra-expressing myocardial precursors. Ectopic expression of pdgfaa interferes with cardiac fusion, consistent with an instructive role for PDGF signaling. Together, these data uncover a novel mechanism through which endodermal-myocardial communication can guide the cell movements that initiate cardiac morphogenesis. DOI:http://dx.doi.org/10.7554/eLife.21172.001 In the growing embryo, the heart initially develops in the form of a simple tube. Its outer layer is made up of muscular cells, called myocardial cells, that pump blood through the tube. Before the heart tube develops, two groups of myocardial cells exist – one on each side of the embryo. To assemble the heart, these two populations of cells must move as a group to the middle of the embryo, where they meet and merge through a process called cardiac fusion. This movement of myocardial cells toward the middle of the embryo depends upon interactions with a neighboring tissue called the endoderm. How the endoderm directs the movement of the myocardial cells was not well understood. The PDGF signaling pathway guides the movement of several different types of cells in the body, but it had not been previously linked to the early stages of heart tube assembly. In this pathway, a molecule called platelet-derived growth factor (PDGF) binds to PDGF receptors that sit on the surface of cells. Using microscopy and genetic analysis to study zebrafish and mouse embryos, Bloomekatz et al. now show that embryos that carry mutations in a gene that encodes a PDGF receptor suffer from defects in heart tube assembly. Further examination of the mutant zebrafish embryos revealed that the myocardial cells were not properly directed toward the middle of the embryo. In fact, many of these cells appeared to move away from the midline. Bloomekatz et al. also observed that, in normal embryos, the endoderm cells that lie adjacent to the myocardial cells produce PDGF. Therefore, it appears that PDGF produced by the endoderm could interact with PDGF receptors on the myocardial cells to direct these cells toward the middle of the embryo. The next step will be to figure out how this signaling influences the machinery inside the myocardial cells that controls their movement. Ultimately, this knowledge could lead to new ways to identify and treat congenital heart diseases. DOI:http://dx.doi.org/10.7554/eLife.21172.002
Collapse
Affiliation(s)
- Joshua Bloomekatz
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Reena Singh
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, Australia
| | - Owen Wj Prall
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ariel C Dunn
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Megan Vaughan
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Chin-San Loo
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, Australia
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, United States
| |
Collapse
|
115
|
Isolation of an ES-Derived Cardiovascular Multipotent Cell Population Based on VE-Cadherin Promoter Activity. Stem Cells Int 2016; 2016:8305624. [PMID: 28101109 PMCID: PMC5215608 DOI: 10.1155/2016/8305624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/16/2016] [Indexed: 12/26/2022] Open
Abstract
Embryonic Stem (ES) or induced Pluripotent Stem (iPS) cells are important sources for cardiomyocyte generation, targeted for regenerative therapies. Several in vitro protocols are currently utilized for their differentiation, but the value of cell-based approaches remains unclear. Here, we characterized a cardiovascular progenitor population derived during ES differentiation, after selection based on VE-cadherin promoter (Pvec) activity. ESCs were genetically modified with an episomal vector, allowing the expression of puromycin resistance gene, under Pvec activity. Puromycin-surviving cells displayed cardiac and endothelial progenitor cells characteristics. Expansion and self-renewal of this cardiac and endothelial dual-progenitor population (CEDP) were achieved by Wnt/β-catenin pathway activation. CEDPs express early cardiac developmental stage-specific markers but not markers of differentiated cardiomyocytes. Similarly, CEDPs express endothelial markers. However, CEDPs can undergo differentiation predominantly to cTnT+ (~47%) and VE-cadherin+ (~28%) cells. Transplantation of CEDPs in the left heart ventricle of adult rats showed that CEDPs-derived cells survive and differentiate in vivo for at least 14 days after transplantation. A novel, dual-progenitor population was isolated during ESCs differentiation, based on Pvec activity. This lineage can self-renew, permitting its maintenance as a source of cardiovascular progenitor cells and constitutes a useful source for regenerative approaches.
Collapse
|
116
|
Pacheco-Leyva I, Matias AC, Oliveira DV, Santos JMA, Nascimento R, Guerreiro E, Michell AC, van De Vrugt AM, Machado-Oliveira G, Ferreira G, Domian I, Bragança J. CITED2 Cooperates with ISL1 and Promotes Cardiac Differentiation of Mouse Embryonic Stem Cells. Stem Cell Reports 2016; 7:1037-1049. [PMID: 27818139 PMCID: PMC5161512 DOI: 10.1016/j.stemcr.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/07/2023] Open
Abstract
The transcriptional regulator CITED2 is essential for heart development. Here, we investigated the role of CITED2 in the specification of cardiac cell fate from mouse embryonic stem cells (ESC). The overexpression of CITED2 in undifferentiated ESC was sufficient to promote cardiac cell emergence upon differentiation. Conversely, the depletion of Cited2 at the onset of differentiation resulted in a decline of ESC ability to generate cardiac cells. Moreover, loss of Cited2 expression impairs the expression of early mesoderm markers and cardiogenic transcription factors (Isl1, Gata4, Tbx5). The cardiogenic defects in Cited2-depleted cells were rescued by treatment with recombinant CITED2 protein. We showed that Cited2 expression is enriched in cardiac progenitors either derived from ESC or mouse embryonic hearts. Finally, we demonstrated that CITED2 and ISL1 proteins interact physically and cooperate to promote ESC differentiation toward cardiomyocytes. Collectively, our results show that Cited2 plays a pivotal role in cardiac commitment of ESC.
Collapse
Affiliation(s)
- Ivette Pacheco-Leyva
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Ana Catarina Matias
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Daniel V Oliveira
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - João M A Santos
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Rita Nascimento
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Eduarda Guerreiro
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Anna C Michell
- Division of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Annebel M van De Vrugt
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Gisela Machado-Oliveira
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal
| | - Guilherme Ferreira
- DSM Biotechnology Center, Alexander Fleminglaan 1, 2613 AX Delft, the Netherlands
| | - Ibrahim Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Charles River Plaza/CPZN 3200, 185 Cambridge Street, Boston, MA 02114-2790, USA; Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA
| | - José Bragança
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal; Centre for Biomedical Research - CBMR, University of Algarve, Campus of Gambelas, Building 8, Room 2.22, 8005-139 Faro, Portugal; ABC - Algarve Biomedical Centre, 8005-139 Faro, Portugal.
| |
Collapse
|
117
|
Wang X, Chen D, Chen K, Jubran A, Ramirez A, Astrof S. Endothelium in the pharyngeal arches 3, 4 and 6 is derived from the second heart field. Dev Biol 2016; 421:108-117. [PMID: 27955943 DOI: 10.1016/j.ydbio.2016.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/18/2016] [Accepted: 12/03/2016] [Indexed: 12/22/2022]
Abstract
Oxygenated blood from the heart is directed into the systemic circulation through the aortic arch arteries (AAAs). The AAAs arise by remodeling of three symmetrical pairs of pharyngeal arch arteries (PAAs), which connect the heart with the paired dorsal aortae at mid-gestation. Aberrant PAA formation results in defects frequently observed in patients with lethal congenital heart disease. How the PAAs form in mammals is not understood. The work presented in this manuscript shows that the second heart field (SHF) is the major source of progenitors giving rise to the endothelium of the pharyngeal arches 3 - 6, while the endothelium in the pharyngeal arches 1 and 2 is derived from a different source. During the formation of the PAAs 3 - 6, endothelial progenitors in the SHF extend cellular processes toward the pharyngeal endoderm, migrate from the SHF and assemble into a uniform vascular plexus. This plexus then undergoes remodeling, whereby plexus endothelial cells coalesce into a large PAA in each pharyngeal arch. Taken together, our studies establish a platform for investigating cellular and molecular mechanisms regulating PAA formation and alterations that lead to disease.
Collapse
Affiliation(s)
- Xia Wang
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | - Dongying Chen
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kelley Chen
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Jefferson Medical College of Thomas Jefferson University, Clinical & Translational Research Track, USA
| | - Ali Jubran
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; The Master's of Science Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - AnnJosette Ramirez
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sophie Astrof
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
118
|
Albanese I, Yu B, Al-Kindi H, Barratt B, Ott L, Al-Refai M, de Varennes B, Shum-Tim D, Cerruti M, Gourgas O, Rhéaume E, Tardif JC, Schwertani A. Role of Noncanonical Wnt Signaling Pathway in Human Aortic Valve Calcification. Arterioscler Thromb Vasc Biol 2016; 37:543-552. [PMID: 27932350 DOI: 10.1161/atvbaha.116.308394] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 11/28/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The mechanisms underlying the pathogenesis of aortic valve calcification remain unclear. With accumulating evidence demonstrating that valve calcification recapitulates bone development, the crucial roles of noncanonical Wnt ligands WNT5a, WNT5b, and WNT11 in osteogenesis make them critical targets in the study of aortic valve calcification. APPROACH AND RESULTS Using immunohistochemistry, real-time qPCR, Western blotting, and tissue culture, we examined the tissue distribution of WNT5a, WNT5b, and WNT11 in noncalcified and calcified aortic valves and their effects on human aortic valve interstitial cells (HAVICs). Only focal strong immunostaining for WNT5a was seen in and around areas of calcification. Abundant immunostaining for WNT5b and WNT11 was seen in inflammatory cells, fibrosis, and activated myofibroblasts in areas of calcified foci. There was significant correlation between WNT5b and WNT11 overall staining and presence of calcification, lipid score, fibrosis, and microvessels (P<0.05). Real-time qPCR and Western blotting revealed abundant expression of both Wnts in stenotic aortic valves, particularly in bicuspid valves. Incubation of HAVICs from noncalcified valves with the 3 noncanonical Wnts significantly increased cell apoptosis and calcification (P<0.05). Treatment of HAVICs with the mitogen-activated protein kinase-38β and GSK3β inhibitors significantly reduced their mineralization (P<0.01). Raman spectroscopy identified the inorganic phosphate deposits as hydroxyapatite and showed a significant increase in hydroxyapatite deposition in HAVICs in response to WNT5a and WNT11 (P<0.05). Similar crystallinity was seen in the deposits found in HAVICs treated with Wnts and in calcified human aortic valves. CONCLUSIONS These findings suggest a potential role for noncanonical Wnt signaling in the pathogenesis of aortic valve calcification.
Collapse
Affiliation(s)
- Isabella Albanese
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Bin Yu
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Hamood Al-Kindi
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Bianca Barratt
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Leah Ott
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Mohammad Al-Refai
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Benoit de Varennes
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Dominique Shum-Tim
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Marta Cerruti
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Ophélie Gourgas
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Eric Rhéaume
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Jean-Claude Tardif
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.)
| | - Adel Schwertani
- From the Division of Cardiology and Division of Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada (I.A., B.Y., H.A.-K., B.B., L.O., M.A.-R., B.d.V., D.S.-T., A.S.); Department of Material Engineering, McGill University, Montreal, Quebec, Canada (M.C., O.G.); and Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada (E.R., J.C.T.).
| |
Collapse
|
119
|
Wilson NK, Gould RA, Gallo MacFarlane E, Consortium ML. Pathophysiology of aortic aneurysm: insights from human genetics and mouse models. Pharmacogenomics 2016; 17:2071-2080. [PMID: 27922338 DOI: 10.2217/pgs-2016-0127] [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] [Indexed: 12/13/2022] Open
Abstract
Aneurysms are local dilations of an artery that predispose the vessel to sudden rupture. They are often asymptomatic and undiagnosed, resulting in a high mortality rate. The predisposition to develop thoracic aortic aneurysms is often genetically inherited and associated with syndromes affecting connective tissue homeostasis. This review discusses how elucidation of the genetic causes of syndromic forms of thoracic aortic aneurysm has helped identify pathways that contribute to disease progression, including those activated by TGF-β, angiotensin II and Notch ligands. We also discuss how pharmacological manipulation of these signaling pathways has provided further insight into the mechanism of disease and identified compounds with therapeutic potential in these and related disorders.
Collapse
Affiliation(s)
- Nicole K Wilson
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Russell A Gould
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | | |
Collapse
|
120
|
Sharma S, Mishra R, Bigham GE, Wehman B, Khan MM, Xu H, Saha P, Goo YA, Datla SR, Chen L, Tulapurkar ME, Taylor BS, Yang P, Karathanasis S, Goodlett DR, Kaushal S. A Deep Proteome Analysis Identifies the Complete Secretome as the Functional Unit of Human Cardiac Progenitor Cells. Circ Res 2016; 120:816-834. [PMID: 27908912 DOI: 10.1161/circresaha.116.309782] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/28/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Cardiac progenitor cells are an attractive cell type for tissue regeneration, but their mechanism for myocardial remodeling is still unclear. OBJECTIVE This investigation determines how chronological age influences the phenotypic characteristics and the secretome of human cardiac progenitor cells (CPCs), and their potential to recover injured myocardium. METHODS AND RESULTS Adult (aCPCs) and neonatal (nCPCs) cells were derived from patients aged >40 years or <1 month, respectively, and their functional potential was determined in a rodent myocardial infarction model. A more robust in vitro proliferative capacity of nCPCs, compared with aCPCs, correlated with significantly greater myocardial recovery mediated by nCPCs in vivo. Strikingly, a single injection of nCPC-derived total conditioned media was significantly more effective than nCPCs, aCPC-derived TCM, or nCPC-derived exosomes in recovering cardiac function, stimulating neovascularization, and promoting myocardial remodeling. High-resolution accurate mass spectrometry with reverse phase liquid chromatography fractionation and mass spectrometry was used to identify proteins in the secretome of aCPCs and nCPCs, and the literature-based networking software identified specific pathways affected by the secretome of CPCs in the setting of myocardial infarction. Examining the TCM, we quantified changes in the expression pattern of 804 proteins in nCPC-derived TCM and 513 proteins in aCPC-derived TCM. The literature-based proteomic network analysis identified that 46 and 6 canonical signaling pathways were significantly targeted by nCPC-derived TCM and aCPC-derived TCM, respectively. One leading candidate pathway is heat-shock factor-1, potentially affecting 8 identified pathways for nCPC-derived TCM but none for aCPC-derived TCM. To validate this prediction, we demonstrated that the modulation of heat-shock factor-1 by knockdown in nCPCs or overexpression in aCPCs significantly altered the quality of their secretome. CONCLUSIONS A deep proteomic analysis revealed both detailed and global mechanisms underlying the chronological age-based differences in the ability of CPCs to promote myocardial recovery via the components of their secretome.
Collapse
Affiliation(s)
- Sudhish Sharma
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Rachana Mishra
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Grace E Bigham
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Brody Wehman
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Mohd M Khan
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Huichun Xu
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Progyaparamita Saha
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Young Ah Goo
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Srinivasa Raju Datla
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Ling Chen
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Mohan E Tulapurkar
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Bradley S Taylor
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Peixin Yang
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Sotirios Karathanasis
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - David R Goodlett
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.)
| | - Sunjay Kaushal
- From the Division of Cardiac Surgery, School of Medicine (S.S., R.M., G.E.B., B.W., P.S., S.R.D., B.S.T., S.K.), Department of Pharmaceutical Sciences, School of Pharmacy (M.M.K., Y.A.G., D.R.G.), Division of Endocrinology, Diabetes and Nutrition, Department of Medicine (H.X.), Department of Physiology and Medicine, School of Medicine (L.C.), Department of OB/GYN & Reproductive Science, Department of Biochemistry and Molecular Biology, School of Medicine (P.Y.), and Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine (M.E.T.), University of Maryland, Baltimore; and Cardiovascular and Metabolic Diseases, Innovative Medicines Biotech Unit MedImmune, Inc., Gaithersburg, MD (S.K.).
| |
Collapse
|
121
|
Shaikh Qureshi WM, Miao L, Shieh D, Li J, Lu Y, Hu S, Barroso M, Mazurkiewicz J, Wu M. Imaging Cleared Embryonic and Postnatal Hearts at Single-cell Resolution. J Vis Exp 2016. [PMID: 27768060 DOI: 10.3791/54303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Single clonal tracing and analysis at the whole-heart level can determine cardiac progenitor cell behavior and differentiation during cardiac development, and allow for the study of the cellular and molecular basis of normal and abnormal cardiac morphogenesis. Recent emerging technologies of retrospective single clonal analyses make the study of cardiac morphogenesis at single cell resolution feasible. However, tissue opacity and light scattering of the heart as imaging depth is increased hinder whole-heart imaging at single cell resolution. To overcome these obstacles, a whole-embryo clearing system that can render the heart highly transparent for both illumination and detection must be developed. Fortunately, in the last several years, many methodologies for whole-organism clearing systems such as CLARITY, Scale, SeeDB, ClearT, 3DISCO, CUBIC, DBE, BABB and PACT have been reported. This lab is interested in the cellular and molecular mechanisms of cardiac morphogenesis. Recently, we established single cell lineage tracing via the ROSA26-CreERT2; ROSA26-Confetti system to sparsely label cells during cardiac development. We adapted several whole embryo-clearing methodologies including Scale and CUBIC (clear, unobstructed brain imaging cocktails and computational analysis) to clear the embryo in combination with whole mount staining to image single clones inside the heart. The heart was successfully imaged at single cell resolution. We found that Scale can clear the embryonic heart, but cannot effectively clear the postnatal heart, while CUBIC can clear the postnatal heart, but damages the embryonic heart by dissolving the tissue. The methods described here will permit the study of gene function at a single clone resolution during cardiac morphogenesis, which, in turn, can reveal the cellular and molecular basis of congenital heart defects.
Collapse
Affiliation(s)
| | - Lianjie Miao
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - David Shieh
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Jingjing Li
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Yangyang Lu
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Saiyang Hu
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College
| | - Joseph Mazurkiewicz
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Mingfu Wu
- Department of Molecular and Cellular Physiology, Albany Medical College;
| |
Collapse
|
122
|
Calderon D, Bardot E, Dubois N. Probing early heart development to instruct stem cell differentiation strategies. Dev Dyn 2016; 245:1130-1144. [PMID: 27580352 DOI: 10.1002/dvdy.24441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/20/2016] [Accepted: 08/20/2016] [Indexed: 12/19/2022] Open
Abstract
Scientists have studied organs and their development for centuries and, along that path, described models and mechanisms explaining the developmental principles of organogenesis. In particular, with respect to the heart, new fundamental discoveries are reported continuously that keep changing the way we think about early cardiac development. These discoveries are driven by the need to answer long-standing questions regarding the origin of the earliest cells specified to the cardiac lineage, the differentiation potential of distinct cardiac progenitor cells, and, very importantly, the molecular mechanisms underlying these specification events. As evidenced by numerous examples, the wealth of developmental knowledge collected over the years has had an invaluable impact on establishing efficient strategies to generate cardiovascular cell types ex vivo, from either pluripotent stem cells or via direct reprogramming approaches. The ability to generate functional cardiovascular cells in an efficient and reliable manner will contribute to therapeutic strategies aimed at alleviating the increasing burden of cardiovascular disease and morbidity. Here we will discuss the recent discoveries in the field of cardiac progenitor biology and their translation to the pluripotent stem cell model to illustrate how developmental concepts have instructed regenerative model systems in the past and promise to do so in the future. Developmental Dynamics 245:1130-1144, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Damelys Calderon
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Evan Bardot
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Nicole Dubois
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| |
Collapse
|
123
|
Nelson DO, Lalit PA, Biermann M, Markandeya YS, Capes DL, Addesso L, Patel G, Han T, John MC, Powers PA, Downs KM, Kamp TJ, Lyons GE. Irx4 Marks a Multipotent, Ventricular-Specific Progenitor Cell. Stem Cells 2016; 34:2875-2888. [PMID: 27570947 DOI: 10.1002/stem.2486] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/18/2016] [Indexed: 12/11/2022]
Abstract
While much progress has been made in the resolution of the cellular hierarchy underlying cardiogenesis, our understanding of chamber-specific myocardium differentiation remains incomplete. To better understand ventricular myocardium differentiation, we targeted the ventricle-specific gene, Irx4, in mouse embryonic stem cells to generate a reporter cell line. Using an antibiotic-selection approach, we purified Irx4+ cells in vitro from differentiating embryoid bodies. The isolated Irx4+ cells proved to be highly proliferative and presented Cxcr4, Pdgfr-alpha, Flk1, and Flt1 on the cell surface. Single Irx4+ ventricular progenitor cells (VPCs) exhibited cardiovascular potency, generating endothelial cells, smooth muscle cells, and ventricular myocytes in vitro. The ventricular specificity of the Irx4+ population was further demonstrated in vivo as VPCs injected into the cardiac crescent subsequently produced Mlc2v+ myocytes that exclusively contributed to the nascent ventricle at E9.5. These findings support the existence of a newly identified ventricular myocardial progenitor. This is the first report of a multipotent cardiac progenitor that contributes progeny specific to the ventricular myocardium. Stem Cells 2016;34:2875-2888.
Collapse
Affiliation(s)
- Daryl O Nelson
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Pratik A Lalit
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mitch Biermann
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yogananda S Markandeya
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Deborah L Capes
- Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Luke Addesso
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Gina Patel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Tianxiao Han
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Manorama C John
- University of Wisconsin Biotechnology Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Patricia A Powers
- University of Wisconsin Biotechnology Center, University of Wisconsin, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Karen M Downs
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Timothy J Kamp
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gary E Lyons
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
124
|
Santos R, Kawauchi S, Jacobs RE, Lopez-Burks ME, Choi H, Wikenheiser J, Hallgrimsson B, Jamniczky HA, Fraser SE, Lander AD, Calof AL. Conditional Creation and Rescue of Nipbl-Deficiency in Mice Reveals Multiple Determinants of Risk for Congenital Heart Defects. PLoS Biol 2016; 14:e2000197. [PMID: 27606604 PMCID: PMC5016002 DOI: 10.1371/journal.pbio.2000197] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/05/2016] [Indexed: 12/16/2022] Open
Abstract
Elucidating the causes of congenital heart defects is made difficult by the complex morphogenesis of the mammalian heart, which takes place early in development, involves contributions from multiple germ layers, and is controlled by many genes. Here, we use a conditional/invertible genetic strategy to identify the cell lineage(s) responsible for the development of heart defects in a Nipbl-deficient mouse model of Cornelia de Lange Syndrome, in which global yet subtle transcriptional dysregulation leads to development of atrial septal defects (ASDs) at high frequency. Using an approach that allows for recombinase-mediated creation or rescue of Nipbl deficiency in different lineages, we uncover complex interactions between the cardiac mesoderm, endoderm, and the rest of the embryo, whereby the risk conferred by genetic abnormality in any one lineage is modified, in a surprisingly non-additive way, by the status of others. We argue that these results are best understood in the context of a model in which the risk of heart defects is associated with the adequacy of early progenitor cell populations relative to the sizes of the structures they must eventually form.
Collapse
Affiliation(s)
- Rosaysela Santos
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America.,Center for Complex Biological Systems, University of California, Irvine, California, United States of America
| | - Shimako Kawauchi
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America.,Center for Complex Biological Systems, University of California, Irvine, California, United States of America
| | - Russell E Jacobs
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Martha E Lopez-Burks
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America.,Center for Complex Biological Systems, University of California, Irvine, California, United States of America
| | - Hojae Choi
- Center for Complex Biological Systems, University of California, Irvine, California, United States of America
| | - Jamie Wikenheiser
- Department of Anatomy and Neurobiology, University of California, Irvine, California, United States of America
| | - Benedikt Hallgrimsson
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Heather A Jamniczky
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Scott E Fraser
- Departments of Biology and Bioengineering, University of Southern California, Los Angeles, California, United States of America
| | - Arthur D Lander
- Center for Complex Biological Systems, University of California, Irvine, California, United States of America.,Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Anne L Calof
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America.,Center for Complex Biological Systems, University of California, Irvine, California, United States of America.,Department of Anatomy and Neurobiology, University of California, Irvine, California, United States of America
| |
Collapse
|
125
|
Santini MP, Forte E, Harvey RP, Kovacic JC. Developmental origin and lineage plasticity of endogenous cardiac stem cells. Development 2016; 143:1242-58. [PMID: 27095490 DOI: 10.1242/dev.111591] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past two decades, several populations of cardiac stem cells have been described in the adult mammalian heart. For the most part, however, their lineage origins and in vivo functions remain largely unexplored. This Review summarizes what is known about different populations of embryonic and adult cardiac stem cells, including KIT(+), PDGFRα(+), ISL1(+)and SCA1(+)cells, side population cells, cardiospheres and epicardial cells. We discuss their developmental origins and defining characteristics, and consider their possible contribution to heart organogenesis and regeneration. We also summarize the origin and plasticity of cardiac fibroblasts and circulating endothelial progenitor cells, and consider what role these cells have in contributing to cardiac repair.
Collapse
Affiliation(s)
- Maria Paola Santini
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Elvira Forte
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington 2052, Australia
| | - Jason C Kovacic
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
126
|
Touma M, Kang X, Zhao Y, Cass AA, Gao F, Biniwale R, Coppola G, Xiao X, Reemtsen B, Wang Y. Decoding the Long Noncoding RNA During Cardiac Maturation: A Roadmap for Functional Discovery. ACTA ACUST UNITED AC 2016; 9:395-407. [PMID: 27591185 DOI: 10.1161/circgenetics.115.001363] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 08/08/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND Cardiac maturation during perinatal transition of heart is critical for functional adaptation to hemodynamic load and nutrient environment. Perturbation in this process has major implications in congenital heart defects. Transcriptome programming during perinatal stages is an important information but incomplete in current literature, particularly, the expression profiles of the long noncoding RNAs (lncRNAs) are not fully elucidated. METHODS AND RESULTS From comprehensive analysis of transcriptomes derived from neonatal mouse heart left and right ventricles, a total of 45 167 unique transcripts were identified, including 21 916 known and 2033 novel lncRNAs. Among these lncRNAs, 196 exhibited significant dynamic regulation along maturation process. By implementing parallel weighted gene co-expression network analysis of mRNA and lncRNA data sets, several lncRNA modules coordinately expressed in a developmental manner similar to protein coding genes, while few lncRNAs revealed chamber-specific patterns. Out of 2262 lncRNAs located within 50 kb of protein coding genes, 5% significantly correlate with the expression of their neighboring genes. The impact of Ppp1r1b-lncRNA on the corresponding partner gene Tcap was validated in cultured myoblasts. This concordant regulation was also conserved in human infantile hearts. Furthermore, the Ppp1r1b-lncRNA/Tcap expression ratio was identified as a molecular signature that differentiated congenital heart defect phenotypes. CONCLUSIONS The study provides the first high-resolution landscape on neonatal cardiac lncRNAs and reveals their potential interaction with mRNA transcriptome during cardiac maturation. Ppp1r1b-lncRNA was identified as a regulator of Tcap expression, with dynamic interaction in postnatal cardiac development and congenital heart defects.
Collapse
Affiliation(s)
- Marlin Touma
- The Children's Discovery and Innovation Institute (CDI), Department of Pediatrics, University of California, Los Angeles, CA.,Cardiovascular Research Laboratory, University of California, Los Angeles, CA
| | - Xuedong Kang
- The Children's Discovery and Innovation Institute (CDI), Department of Pediatrics, University of California, Los Angeles, CA
| | - Yan Zhao
- The Children's Discovery and Innovation Institute (CDI), Department of Pediatrics, University of California, Los Angeles, CA
| | - Ashley A Cass
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Fuying Gao
- Department of Neurology and Psychiatry, University of California, Los Angeles, CA
| | - Reshma Biniwale
- Department of Cardiothoracic Surgery, University of California, Los Angeles, CA
| | - Giovanni Coppola
- Department of Neurology and Psychiatry, University of California, Los Angeles, CA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Brian Reemtsen
- Department of Cardiothoracic Surgery, University of California, Los Angeles, CA
| | - Yibin Wang
- Cardiovascular Research Laboratory, University of California, Los Angeles, CA.,Department of Anesthesiology, Physiology and Medicine, University of California, Los Angeles, CA
| |
Collapse
|
127
|
Abstract
Recent data have paved the way to mechanistic studies into the role of Tbx1 during development. Tbx1 is haploinsufficient and is involved in an important genetic disorder. The gene encodes a T-box transcription factor that is expressed from approximately E7.5 in mouse embryos and continues to be expressed in a highly dynamic manner. It is neither a strong transcriptional activator nor a strong repressor, but it regulates a large number of genes through epigenetic modifications. Here, we review recent literature concerning mechanisms of gene regulation by Tbx1 and its role in mammalian development, with a special focus on the cardiac, vascular, and central nervous systems.
Collapse
|
128
|
Fair JV, Voronova A, Bosiljcic N, Rajgara R, Blais A, Skerjanc IS. BRG1 interacts with GLI2 and binds Mef2c gene in a hedgehog signalling dependent manner during in vitro cardiomyogenesis. BMC DEVELOPMENTAL BIOLOGY 2016; 16:27. [PMID: 27484899 PMCID: PMC4970297 DOI: 10.1186/s12861-016-0127-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/28/2016] [Indexed: 12/22/2022]
Abstract
Background The Hedgehog (HH) signalling pathway regulates cardiomyogenesis in vivo and in differentiating P19 embryonal carcinoma (EC) cells, a mouse embryonic stem (mES) cell model. To further assess the transcriptional role of HH signalling during cardiomyogenesis in stem cells, we studied the effects of overexpressing GLI2, a primary transducer of the HH signalling pathway, in mES cells. Results Stable GLI2 overexpression resulted in an enhancement of cardiac progenitor-enriched genes, Mef2c, Nkx2-5, and Tbx5 during mES cell differentiation. In contrast, pharmacological blockade of the HH pathway in mES cells resulted in lower expression of these genes. Mass spectrometric analysis identified the chromatin remodelling factor BRG1 as a protein which co-immunoprecipitates with GLI2 in differentiating mES cells. We then determined that BRG1 is recruited to a GLI2-specific Mef2c gene element in a HH signalling-dependent manner during cardiomyogenesis in P19 EC cells, a mES cell model. Conclusions Thus, we propose a mechanism where HH/GLI2 regulates the expression of Mef2c by recruiting BRG1 to the Mef2c gene, most probably via chromatin remodelling, to ultimately regulate in vitro cardiomyogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0127-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joel Vincent Fair
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada
| | - Anastassia Voronova
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada
| | - Neven Bosiljcic
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada
| | - Rashida Rajgara
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada
| | - Alexandre Blais
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada. .,Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada.
| | - Ilona Sylvia Skerjanc
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada.
| |
Collapse
|
129
|
Cardiac stem cell transplantation with 2,3,5,4′-tetrahydroxystilbehe-2-O-β-d-glucoside improves cardiac function in rat myocardial infarction model. Life Sci 2016; 158:37-45. [DOI: 10.1016/j.lfs.2016.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/06/2016] [Accepted: 06/09/2016] [Indexed: 01/01/2023]
|
130
|
Jahangiri L, Sharpe M, Novikov N, González-Rosa JM, Borikova A, Nevis K, Paffett-Lugassy N, Zhao L, Adams M, Guner-Ataman B, Burns CE, Burns CG. The AP-1 transcription factor component Fosl2 potentiates the rate of myocardial differentiation from the zebrafish second heart field. Development 2016; 143:113-22. [PMID: 26732840 DOI: 10.1242/dev.126136] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vertebrate heart forms through successive phases of cardiomyocyte differentiation. Initially, cardiomyocytes derived from first heart field (FHF) progenitors assemble the linear heart tube. Thereafter, second heart field (SHF) progenitors differentiate into cardiomyocytes that are accreted to the poles of the heart tube over a well-defined developmental window. Although heart tube elongation deficiencies lead to life-threatening congenital heart defects, the variables controlling the initiation, rate and duration of myocardial accretion remain obscure. Here, we demonstrate that the AP-1 transcription factor, Fos-like antigen 2 (Fosl2), potentiates the rate of myocardial accretion from the zebrafish SHF. fosl2 mutants initiate accretion appropriately, but cardiomyocyte production is sluggish, resulting in a ventricular deficit coupled with an accumulation of SHF progenitors. Surprisingly, mutant embryos eventually correct the myocardial deficit by extending the accretion window. Overexpression of Fosl2 also compromises production of SHF-derived ventricular cardiomyocytes, a phenotype that is consistent with precocious depletion of the progenitor pool. Our data implicate Fosl2 in promoting the progenitor to cardiomyocyte transition and uncover the existence of regulatory mechanisms to ensure appropriate SHF-mediated cardiomyocyte contribution irrespective of embryonic stage.
Collapse
Affiliation(s)
- Leila Jahangiri
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Michka Sharpe
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Natasha Novikov
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Asya Borikova
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen Nevis
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Noelle Paffett-Lugassy
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Long Zhao
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Meghan Adams
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Burcu Guner-Ataman
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Caroline E Burns
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - C Geoffrey Burns
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
131
|
Haack T, Abdelilah-Seyfried S. The force within: endocardial development, mechanotransduction and signalling during cardiac morphogenesis. Development 2016; 143:373-86. [PMID: 26839341 DOI: 10.1242/dev.131425] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endocardial cells are cardiac endothelial cells that line the interior of the heart tube. Historically, their contribution to cardiac development has mainly been considered from a morphological perspective. However, recent studies have begun to define novel instructive roles of the endocardium, as a sensor and signal transducer of biophysical forces induced by blood flow, and as an angiocrine signalling centre that is involved in myocardial cellular morphogenesis, regeneration and reprogramming. In this Review, we discuss how the endocardium develops, how endocardial-myocardial interactions influence the developing embryonic heart, and how the dysregulation of blood flow-responsive endocardial signalling can result in pathophysiological changes.
Collapse
Affiliation(s)
- Timm Haack
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, Potsdam D-14476, Germany
| |
Collapse
|
132
|
Fulcoli FG, Franzese M, Liu X, Zhang Z, Angelini C, Baldini A. Rebalancing gene haploinsufficiency in vivo by targeting chromatin. Nat Commun 2016; 7:11688. [PMID: 27256596 PMCID: PMC4895808 DOI: 10.1038/ncomms11688] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/19/2016] [Indexed: 11/28/2022] Open
Abstract
Congenital heart disease (CHD) affects eight out of 1,000 live births and is a major social and health-care burden. A common genetic cause of CHD is the 22q11.2 deletion, which is the basis of the homonymous deletion syndrome (22q11.2DS), also known as DiGeorge syndrome. Most of its clinical spectrum is caused by haploinsufficiency of Tbx1, a gene encoding a T-box transcription factor. Here we show that Tbx1 positively regulates monomethylation of histone 3 lysine 4 (H3K4me1) through interaction with and recruitment of histone methyltransferases. Treatment of cells with tranylcypromine (TCP), an inhibitor of histone demethylases, rebalances the loss of H3K4me1 and rescues the expression of approximately one-third of the genes dysregulated by Tbx1 suppression. In Tbx1 mouse mutants, TCP treatment ameliorates substantially the cardiovascular phenotype. These data suggest that epigenetic drugs may represent a potential therapeutic strategy for rescue of gene haploinsufficiency phenotypes, including structural defects. Deficit in transcription factor Tbx1 causes heart defects in humans and mice. Here the authors show that Tbx1 regulates gene expression by recruiting histone methyltransferases that affect chromatin marks, and that a drug inhibiting histone demethylation ameliorates the cardiovascular phenotype in Tbx1 haploinsufficient or hypomorphic mice.
Collapse
Affiliation(s)
- Filomena Gabriella Fulcoli
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, Naples 80131, Italy
| | - Monica Franzese
- Istituto per le Applicazioni del Calcolo, CNR, Naples, Italy
| | - Xiangyang Liu
- Shanghai Pediatric Congenital Heart Institute, Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Institute, Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | | | - Antonio Baldini
- CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, Naples 80131, Italy.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| |
Collapse
|
133
|
Hara Y, Wake K, Inoue K, Kuroda N, Sato A, Inamatsu M, Tateno C, Sato T. Development of the Heart Endocardium at an Early Stage of Chick Embryos Evaluated at Light- and Electron-Microscopic Levels. Anat Rec (Hoboken) 2016; 299:1080-9. [DOI: 10.1002/ar.23372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 03/14/2016] [Indexed: 01/20/2023]
Affiliation(s)
- Yaiko Hara
- Department of Anatomy, Tissue and Cell Biology; Tsurumi University School of Dental Medicine; Yokohama Japan
| | - Kenjiro Wake
- Department of Anatomy, Tissue and Cell Biology; Tsurumi University School of Dental Medicine; Yokohama Japan
- Liver Research Unit; Minophagen Pharmaceutical Co., Ltd.; Tokyo Japan
| | - Kouji Inoue
- Research Center of Electron Microscopy, School of Dental Medicine; Tsurumi University; Yokohama Japan
| | - Noriyuki Kuroda
- Department of Anatomy, Tissue and Cell Biology; Tsurumi University School of Dental Medicine; Yokohama Japan
| | - Akie Sato
- Department of Anatomy, Tissue and Cell Biology; Tsurumi University School of Dental Medicine; Yokohama Japan
| | | | | | - Tetsuji Sato
- Department of Anatomy, Tissue and Cell Biology; Tsurumi University School of Dental Medicine; Yokohama Japan
| |
Collapse
|
134
|
Higashiyama H, Hirasawa T, Oisi Y, Sugahara F, Hyodo S, Kanai Y, Kuratani S. On the vagal cardiac nerves, with special reference to the early evolution of the head-trunk interface. J Morphol 2016; 277:1146-58. [PMID: 27216138 DOI: 10.1002/jmor.20563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 04/28/2016] [Accepted: 05/02/2016] [Indexed: 12/11/2022]
Abstract
The vagus nerve, or the tenth cranial nerve, innervates the heart in addition to other visceral organs, including the posterior visceral arches. In amniotes, the anterior and posterior cardiac branches arise from the branchial and intestinal portions of the vagus nerve to innervate the arterial and venous poles of the heart, respectively. The evolution of this innervation pattern has yet to be elucidated, due mainly to the lack of morphological data on the vagus in basal vertebrates. To investigate this topic, we observed the vagus nerves of the lamprey (Lethenteron japonicum), elephant shark (Callorhinchus milii), and mouse (Mus musculus), focusing on the embryonic patterns of the vagal branches in the venous pole. In the lamprey, no vagus branch was found in the venous pole throughout development, whereas the arterial pole was innervated by a branch from the branchial portion. In contrast, the vagus innervated the arterial and venous poles in the mouse and elephant shark. Based on the morphological patterns of these branches, the venous vagal branches of the mouse and elephant shark appear to belong to the intestinal part of the vagus, implying that the cardiac nerve pattern is conserved among crown gnathostomes. Furthermore, we found a topographical shift of the structures adjacent to the venous pole (i.e., the hypoglossal nerve and pronephros) between the extant gnathostomes and lamprey. Phylogenetically, the lamprey morphology is likely to be the ancestral condition for vertebrates, suggesting that the evolution of the venous branch occurred early in the gnathostome lineage, in parallel with the remodeling of the head-trunk interfacial domain during the acquisition of the neck. J. Morphol. 277:1146-1158, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hiroki Higashiyama
- Department of Biology, Graduate School of Science, Kobe University, Kobe, 657-8501, Japan.,Evolutionary Morphology Laboratory, RIKEN, Kobe, 650-0047, Japan.,Laboratory of Veterinary Anatomy, the University of Tokyo, Tokyo, 113-8657, Japan
| | - Tatsuya Hirasawa
- Evolutionary Morphology Laboratory, RIKEN, Kobe, 650-0047, Japan
| | - Yasuhiro Oisi
- Evolutionary Morphology Laboratory, RIKEN, Kobe, 650-0047, Japan.,Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
| | - Fumiaki Sugahara
- Evolutionary Morphology Laboratory, RIKEN, Kobe, 650-0047, Japan.,Division of Biology, Hyogo College of Medicine, Nishinomiya, Hyogo, 663-8501, Japan
| | - Susumu Hyodo
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, the University of Tokyo, Chiba, 277-8564, Japan
| | - Yoshiakira Kanai
- Laboratory of Veterinary Anatomy, the University of Tokyo, Tokyo, 113-8657, Japan
| | - Shigeru Kuratani
- Evolutionary Morphology Laboratory, RIKEN, Kobe, 650-0047, Japan
| |
Collapse
|
135
|
Functions of miRNAs during Mammalian Heart Development. Int J Mol Sci 2016; 17:ijms17050789. [PMID: 27213371 PMCID: PMC4881605 DOI: 10.3390/ijms17050789] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/26/2016] [Accepted: 05/13/2016] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs (miRNAs) play essential roles during mammalian heart development and have emerged as attractive therapeutic targets for cardiovascular diseases. The mammalian embryonic heart is mainly derived from four major cell types during development. These include cardiomyocytes, endocardial cells, epicardial cells, and neural crest cells. Recent data have identified various miRNAs as critical regulators of the proper differentiation, proliferation, and survival of these cell types. In this review, we briefly introduce the contemporary understanding of mammalian cardiac development. We also focus on recent developments in the field of cardiac miRNAs and their functions during the development of different cell types.
Collapse
|
136
|
Arora H, Boulberdaa M, Qureshi R, Bitirim V, Gasser A, Messaddeq N, Dolle P, Nebigil CG. Prokineticin receptor-1 signaling promotes Epicardial to Mesenchymal Transition during heart development. Sci Rep 2016; 6:25541. [PMID: 27150455 PMCID: PMC4858698 DOI: 10.1038/srep25541] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/18/2016] [Indexed: 01/20/2023] Open
Abstract
The epicardium plays an essential role in coronary artery formation and myocardial development. However, signals controlling the developing epicardium and epicardial-mesenchymal transition (EMT) in the normal and diseased adult heart are studied less rigorously. Here we investigated the role of angiogenic hormone, prokineticin-2 and its receptor PKR1 in the epicardium of developing and adult heart. Genetic ablation of PKR1 in epicardium leads to partial embryonic and postnatal lethality with abnormal heart development. Cardiac developmental defects are manifested in the adult stage as ischemic cardiomyopathy with systolic dysfunction. We discovered that PKR1 regulates epicardial-mesenchymal transition (EMT) for epicardial-derived progenitor cell (EPDC), formation. This event affects at least three consequential steps during heart development: (i) EPDC and cardiomyocyte proliferation involved in thickening of an outer compact ventricular chamber wall, (ii) rhythmicity, (iii) formation of coronary circulation. In isolated embryonic EPDCs, overexpression or activation of PKR1 alters cell morphology and EMT markers via activating Akt signaling. Lack of PKR1 signal in epicardium leads to defective heart development and underlies the origin of congenital heart disease in adult mice. Our mice provide genetic models for congenital dysfunction of the heart and should facilitate studies of both pathogenesis and therapy of cardiac disorders in humans.
Collapse
Affiliation(s)
- Himanshu Arora
- CNRS, Université de Strasbourg, UMR7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Mounia Boulberdaa
- CNRS, Université de Strasbourg, UMR7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Rehana Qureshi
- CNRS, Université de Strasbourg, UMR7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Verda Bitirim
- CNRS, Université de Strasbourg, UMR7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Adeline Gasser
- CNRS, Université de Strasbourg, UMR7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Nadia Messaddeq
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS, UMR 7104 and INSERM Unité 964, Université de Strasbourg, Illkirch-Strasbourg, France
| | - Pascal Dolle
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS, UMR 7104 and INSERM Unité 964, Université de Strasbourg, Illkirch-Strasbourg, France
| | - Canan G. Nebigil
- CNRS, Université de Strasbourg, UMR7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
137
|
Paksa A, Bandemer J, Hoeckendorf B, Razin N, Tarbashevich K, Minina S, Meyen D, Biundo A, Leidel SA, Peyrieras N, Gov NS, Keller PJ, Raz E. Repulsive cues combined with physical barriers and cell-cell adhesion determine progenitor cell positioning during organogenesis. Nat Commun 2016; 7:11288. [PMID: 27088892 PMCID: PMC4837475 DOI: 10.1038/ncomms11288] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/09/2016] [Indexed: 01/15/2023] Open
Abstract
The precise positioning of organ progenitor cells constitutes an essential, yet poorly understood step during organogenesis. Using primordial germ cells that participate in gonad formation, we present the developmental mechanisms maintaining a motile progenitor cell population at the site where the organ develops. Employing high-resolution live-cell microscopy, we find that repulsive cues coupled with physical barriers confine the cells to the correct bilateral positions. This analysis revealed that cell polarity changes on interaction with the physical barrier and that the establishment of compact clusters involves increased cell–cell interaction time. Using particle-based simulations, we demonstrate the role of reflecting barriers, from which cells turn away on contact, and the importance of proper cell–cell adhesion level for maintaining the tight cell clusters and their correct positioning at the target region. The combination of these developmental and cellular mechanisms prevents organ fusion, controls organ positioning and is thus critical for its proper function. The precise positioning of organ progenitor cells is essential for organ development and function. Here the authors use live imaging and mathematical modelling to show that the confinement of a motile progenitor cell population results from coupled physical barriers and cell-cell interactions.
Collapse
Affiliation(s)
- Azadeh Paksa
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| | - Jan Bandemer
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| | | | - Nitzan Razin
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Sofia Minina
- Germ Cell Development, Max-Planck Institute of Biophysical Chemistry, Am Fassberg 11, 37070 Göttingen, Germany
| | - Dana Meyen
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| | - Antonio Biundo
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, Von-Esmarch-Strasse 54, 48149 Muenster, Germany
| | - Sebastian A Leidel
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, Von-Esmarch-Strasse 54, 48149 Muenster, Germany
| | - Nadine Peyrieras
- USR3695 BioEmergences, CNRS, Université Paris-Saclay, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Nir S Gov
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Erez Raz
- Institute for Cell Biology, ZMBE, Von-Esmarch-Street 56, 48149 Muenster, Germany
| |
Collapse
|
138
|
Wittig JG, Münsterberg A. The Early Stages of Heart Development: Insights from Chicken Embryos. J Cardiovasc Dev Dis 2016; 3:jcdd3020012. [PMID: 29367563 PMCID: PMC5715676 DOI: 10.3390/jcdd3020012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 01/01/2023] Open
Abstract
The heart is the first functioning organ in the developing embryo and a detailed understanding of the molecular and cellular mechanisms involved in its formation provides insights into congenital malformations affecting its function and therefore the survival of the organism. Because many developmental mechanisms are highly conserved, it is possible to extrapolate from observations made in invertebrate and vertebrate model organisms to humans. This review will highlight the contributions made through studying heart development in avian embryos, particularly the chicken. The major advantage of chick embryos is their accessibility for surgical manipulation and functional interference approaches, both gain- and loss-of-function. In addition to experiments performed in ovo, the dissection of tissues for ex vivo culture, genomic, or biochemical approaches is straightforward. Furthermore, embryos can be cultured for time-lapse imaging, which enables tracking of fluorescently labeled cells and detailed analysis of tissue morphogenesis. Owing to these features, investigations in chick embryos have led to important discoveries, often complementing genetic studies in mice and zebrafish. As well as including some historical aspects, we cover here some of the crucial advances made in understanding early heart development using the chicken model.
Collapse
Affiliation(s)
- Johannes G Wittig
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| |
Collapse
|
139
|
Lambers E, Kume T. Navigating the labyrinth of cardiac regeneration. Dev Dyn 2016; 245:751-61. [PMID: 26890576 DOI: 10.1002/dvdy.24397] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/26/2016] [Accepted: 02/10/2016] [Indexed: 12/20/2022] Open
Abstract
Heart disease is the number one cause of morbidity and mortality in the world and is a major health and economic burden, costing the United States Health Care System more than $200 billion annually. A major cause of heart disease is the massive loss or dysfunction of cardiomyocytes caused by myocardial infarctions and hypertension. Due to the limited regenerative capacity of the heart, much research has focused on better understanding the process of differentiation toward cardiomyocytes. This review will highlight what is currently known about cardiac cell specification during mammalian development, areas of controversy, cellular sources of cardiomyocytes, and current and potential uses of stem cell derived cardiomyocytes for cardiac therapies. Developmental Dynamics 245:751-761, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Erin Lambers
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
140
|
Hox Genes in Cardiovascular Development and Diseases. J Dev Biol 2016; 4:jdb4020014. [PMID: 29615581 PMCID: PMC5831787 DOI: 10.3390/jdb4020014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/16/2016] [Accepted: 03/23/2016] [Indexed: 11/23/2022] Open
Abstract
Congenital heart defects (CHD) are the leading cause of death in the first year of life. Over the past 20 years, much effort has been focused on unraveling the genetic bases of CHD. In particular, studies in human genetics coupled with those of model organisms have provided valuable insights into the gene regulatory networks underlying CHD pathogenesis. Hox genes encode transcription factors that are required for the patterning of the anterior–posterior axis in the embryo. In this review, we focus on the emerging role of anteriorly expressed Hox genes (Hoxa1, Hoxb1, and Hoxa3) in cardiac development, specifically their contribution to patterning of cardiac progenitor cells and formation of the great arteries. Recent evidence regarding the cooperative regulation of heart development by Hox proteins with members of the TALE-class of homeodomain proteins such as Pbx and Meis transcription factors is also discussed. These findings are highly relevant to human pathologies as they pinpoint new genes that increase susceptibility to cardiac anomalies and provide novel mechanistic insights into CHD.
Collapse
|
141
|
Endothelin-1 supports clonal derivation and expansion of cardiovascular progenitors derived from human embryonic stem cells. Nat Commun 2016; 7:10774. [PMID: 26952167 PMCID: PMC4786749 DOI: 10.1038/ncomms10774] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 01/19/2016] [Indexed: 01/10/2023] Open
Abstract
Coronary arteriogenesis is a central step in cardiogenesis, requiring coordinated generation and integration of endothelial cell and vascular smooth muscle cells. At present, it is unclear whether the cell fate programme of cardiac progenitors to generate complex muscular or vascular structures is entirely cell autonomous. Here we demonstrate the intrinsic ability of vascular progenitors to develop and self-organize into cardiac tissues by clonally isolating and expanding second heart field cardiovascular progenitors using WNT3A and endothelin-1 (EDN1) human recombinant proteins. Progenitor clones undergo long-term expansion and differentiate primarily into endothelial and smooth muscle cell lineages in vitro, and contribute extensively to coronary-like vessels in vivo, forming a functional human–mouse chimeric circulatory system. Our study identifies EDN1 as a key factor towards the generation and clonal derivation of ISL1+ vascular intermediates, and demonstrates the intrinsic cell-autonomous nature of these progenitors to differentiate and self-organize into functional vasculatures in vivo. Understanding coronary vessels development provides basis for regenerative strategies. Here, Soh et al. identify endothelin-1 as a key molecule driving long-term expansion of ISL1+ bipotent vascular progenitors derived from human embryonic stem cells, and show that these cells can regenerate coronary vessels in mice.
Collapse
|
142
|
Spatial regulation of cell cohesion by Wnt5a during second heart field progenitor deployment. Dev Biol 2016; 412:18-31. [PMID: 26916252 DOI: 10.1016/j.ydbio.2016.02.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 01/11/2023]
Abstract
Wnt5a, a non-canonical Wnt ligand critical for outflow tract (OFT) morphogenesis, is expressed specifically in second heart field (SHF) progenitors in the caudal splanchnic mesoderm (SpM) near the inflow tract (IFT). Using a conditional Wnt5a gain of function (GOF) allele and Islet1-Cre, we broadly over-expressed Wnt5a throughout the SHF lineage, including the entire SpM between the IFT and OFT. Wnt5a over-expression in Wnt5a null mutants can rescue the cell polarity and actin polymerization defects as well as severe SpM shortening, but fails to rescue OFT shortening. Moreover, Wnt5a over-expression in wild-type background is able to cause OFT shortening. We find that Wnt5a over-expression does not perturb SHF cell proliferation, apoptosis or differentiation, but affects the deployment of SHF cells by causing them to accumulate into a large bulge at the rostral SpM and fail to enter the OFT. Our immunostaining analyses suggest an inverse correlation between cell cohesion and Wnt5a level in the wild-type SpM. Ectopic Wnt5a expression in the rostral SpM of Wn5a-GOF mutants diminishes the upregulation of adherens junction; whereas loss of Wnt5a in Wnt5a null mutants causes premature increase in adherens junction level in the caudal SpM. Over-expression of mouse Wnt5a in Xenopus animal cap cells also reduces C-cadherin distribution on the plasma membrane without affecting its overall protein level, suggesting that Wnt5a may play an evolutionarily conserved role in controlling the cell surface level of cadherin to modulate cell cohesion during tissue morphogenesis. Collectively, our data indicate that restricted expression of Wnt5a in the caudal SpM is essential for normal OFT morphogenesis, and uncover a novel function of spatially regulated cell cohesion by Wnt5a in driving the deployment of SHF cells from the SpM into the OFT.
Collapse
|
143
|
Orlova VV, Chuva de Sousa Lopes S, Valdimarsdottir G. BMP-SMAD signaling: From pluripotent stem cells to cardiovascular commitment. Cytokine Growth Factor Rev 2016; 27:55-63. [DOI: 10.1016/j.cytogfr.2015.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
|
144
|
Fuchs C, Gawlas S, Heher P, Nikouli S, Paar H, Ivankovic M, Schultheis M, Klammer J, Gottschamel T, Capetanaki Y, Weitzer G. Desmin enters the nucleus of cardiac stem cells and modulates Nkx2.5 expression by participating in transcription factor complexes that interact with the nkx2.5 gene. Biol Open 2016; 5:140-53. [PMID: 26787680 PMCID: PMC4823984 DOI: 10.1242/bio.014993] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/13/2015] [Indexed: 12/30/2022] Open
Abstract
The transcription factor Nkx2.5 and the intermediate filament protein desmin are simultaneously expressed in cardiac progenitor cells during commitment of primitive mesoderm to the cardiomyogenic lineage. Up-regulation of Nkx2.5 expression by desmin suggests that desmin may contribute to cardiogenic commitment and myocardial differentiation by directly influencing the transcription of the nkx2.5 gene in cardiac progenitor cells. Here, we demonstrate that desmin activates transcription of nkx2.5 reporter genes, rescues nkx2.5 haploinsufficiency in cardiac progenitor cells, and is responsible for the proper expression of Nkx2.5 in adult cardiac side population stem cells. These effects are consistent with the temporary presence of desmin in the nuclei of differentiating cardiac progenitor cells and its physical interaction with transcription factor complexes bound to the enhancer and promoter elements of the nkx2.5 gene. These findings introduce desmin as a newly discovered and unexpected player in the regulatory network guiding cardiomyogenesis in cardiac stem cells.
Collapse
Affiliation(s)
- Christiane Fuchs
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Sonja Gawlas
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Philipp Heher
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 115 27, Greece
| | - Hannah Paar
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Mario Ivankovic
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Martina Schultheis
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Julia Klammer
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Teresa Gottschamel
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 115 27, Greece
| | - Georg Weitzer
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| |
Collapse
|
145
|
Liu J, Laksman Z, Backx PH. The electrophysiological development of cardiomyocytes. Adv Drug Deliv Rev 2016; 96:253-73. [PMID: 26788696 DOI: 10.1016/j.addr.2015.12.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/23/2015] [Accepted: 12/31/2015] [Indexed: 02/07/2023]
Abstract
The generation of human cardiomyocytes (CMs) from human pluripotent stem cells (hPSCs) has become an important resource for modeling human cardiac disease and for drug screening, and also holds significant potential for cardiac regeneration. Many challenges remain to be overcome however, before innovation in this field can translate into a change in the morbidity and mortality associated with heart disease. Of particular importance for the future application of this technology is an improved understanding of the electrophysiologic characteristics of CMs, so that better protocols can be developed and optimized for generating hPSC-CMs. Many different cell culture protocols are currently utilized to generate CMs from hPSCs and all appear to yield relatively “developmentally” immature CMs with highly heterogeneous electrical properties. These hPSC-CMs are characterized by spontaneous beating at highly variable rates with a broad range of depolarization-repolarization patterns, suggestive of mixed populations containing atrial, ventricular and nodal cells. Many recent studies have attempted to introduce approaches to promote maturation and to create cells with specific functional properties. In this review, we summarize the studies in which the electrical properties of CMs derived from stem cells have been examined. In order to place this information in a useful context, we also review the electrical properties of CMs as they transition from the developing embryo to the adult human heart. The signal pathways involved in the regulation of ion channel expression during development are also briefly considered.
Collapse
|
146
|
Dorn T, Goedel A, Lam JT, Haas J, Tian Q, Herrmann F, Bundschu K, Dobreva G, Schiemann M, Dirschinger R, Guo Y, Kühl SJ, Sinnecker D, Lipp P, Laugwitz KL, Kühl M, Moretti A. Direct nkx2-5 transcriptional repression of isl1 controls cardiomyocyte subtype identity. Stem Cells 2016; 33:1113-29. [PMID: 25524439 PMCID: PMC6750130 DOI: 10.1002/stem.1923] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/29/2014] [Accepted: 11/08/2014] [Indexed: 12/31/2022]
Abstract
During cardiogenesis, most myocytes arise from cardiac progenitors expressing the transcription factors Isl1 and Nkx2-5. Here, we show that a direct repression of Isl1 by Nkx2-5 is necessary for proper development of the ventricular myocardial lineage. Overexpression of Nkx2-5 in mouse embryonic stem cells (ESCs) delayed specification of cardiac progenitors and inhibited expression of Isl1 and its downstream targets in Isl1(+) precursors. Embryos deficient for Nkx2-5 in the Isl1(+) lineage failed to downregulate Isl1 protein in cardiomyocytes of the heart tube. We demonstrated that Nkx2-5 directly binds to an Isl1 enhancer and represses Isl1 transcriptional activity. Furthermore, we showed that overexpression of Isl1 does not prevent cardiac differentiation of ESCs and in Xenopus laevis embryos. Instead, it leads to enhanced specification of cardiac progenitors, earlier cardiac differentiation, and increased cardiomyocyte number. Functional and molecular characterization of Isl1-overexpressing cardiomyocytes revealed higher beating frequencies in both ESC-derived contracting areas and Xenopus Isl1-gain-of-function hearts, which associated with upregulation of nodal-specific genes and downregulation of transcripts of working myocardium. Immunocytochemistry of cardiomyocyte lineage-specific markers demonstrated a reduction of ventricular cells and an increase of cells expressing the pacemaker channel Hcn4. Finally, optical action potential imaging of single cardiomyocytes combined with pharmacological approaches proved that Isl1 overexpression in ESCs resulted in normally electrophysiologically functional cells, highly enriched in the nodal subtype at the expense of the ventricular lineage. Our findings provide an Isl1/Nkx2-5-mediated mechanism that coordinately regulates the specification of cardiac progenitors toward the different myocardial lineages and ensures proper acquisition of myocyte subtype identity.
Collapse
Affiliation(s)
- Tatjana Dorn
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
|
148
|
|
149
|
Ahmad SM, Bhattacharyya P, Jeffries N, Gisselbrecht SS, Michelson AM. Two Forkhead transcription factors regulate cardiac progenitor specification by controlling the expression of receptors of the fibroblast growth factor and Wnt signaling pathways. Development 2015; 143:306-17. [PMID: 26657774 PMCID: PMC4725337 DOI: 10.1242/dev.122952] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 11/26/2015] [Indexed: 11/20/2022]
Abstract
Cardiogenesis involves the coordinated regulation of multiple biological processes by a finite set of transcription factors (TFs). Here, we show that the Forkhead TFs Checkpoint suppressor homologue (CHES-1-like) and Jumeau (Jumu), which govern cardiac progenitor cell divisions by regulating Polo kinase activity, play an additional, mutually redundant role in specifying the cardiac mesoderm (CM) as eliminating the functions of both Forkhead genes in the same Drosophila embryo results in defective hearts with missing hemisegments. This process is mediated by the Forkhead TFs regulating the fibroblast growth factor receptor Heartless (Htl) and the Wnt receptor Frizzled (Fz): CHES-1-like and jumu exhibit synergistic genetic interactions with htl and fz in CM specification, thereby implying that they function through the same genetic pathways, and transcriptionally activate the expression of both receptor-encoding genes. Furthermore, ectopic overexpression of either htl or fz in the mesoderm partially rescues the defective CM specification phenotype in embryos lacking both Forkhead genes. Together, these data emphasize the functional redundancy that leads to robustness in the cardiac progenitor specification process, and illustrate the pleiotropic functions of Forkhead TFs in different aspects of cardiogenesis. Summary: Checkpoint suppressor homologue and Jumeau, which are known to govern cardiac progenitor cell divisions, play additional, mutually redundant roles in specifying cardiac mesoderm in Drosophila.
Collapse
Affiliation(s)
- Shaad M Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pritha Bhattacharyya
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neal Jeffries
- Office of Biostatistics Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen S Gisselbrecht
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alan M Michelson
- Laboratory of Developmental Systems Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
150
|
Abstract
The developmental mechanisms that control head muscle formation are distinct from those that operate in the trunk. Head and neck muscles derive from various mesoderm populations in the embryo and are regulated by distinct transcription factors and signaling molecules. Throughout the last decade, developmental, and lineage studies in vertebrates and invertebrates have revealed the peculiar nature of the pharyngeal mesoderm that forms certain head muscles and parts of the heart. Studies in chordates, the ancestors of vertebrates, revealed an evolutionarily conserved cardiopharyngeal field that progressively facilitates the development of both heart and craniofacial structures during vertebrate evolution. This ancient regulatory circuitry preceded and facilitated the emergence of myogenic cell types and hierarchies that exist in vertebrates. This chapter summarizes studies related to the origins, signaling circuits, genetics, and evolution of the head musculature, highlighting its heterogeneous characteristics in all these aspects, with a special focus on the FGF-ERK pathway. Additionally, we address the processes of head muscle regeneration, and the development of stem cell-based therapies for treatment of muscle disorders.
Collapse
Affiliation(s)
- Inbal Michailovici
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Eigler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|