1
|
Kaur N, Gare SR, Ruiz-Velasco A, Miller JM, Abouleisa RR, Ou Q, Shen J, Soran H, Mohamed TM, Liu W. FGF21/FGFR1-β-KL cascade in cardiomyocytes modulates angiogenesis and inflammation under metabolic stress. Heliyon 2023; 9:e14952. [PMID: 37123894 PMCID: PMC10133673 DOI: 10.1016/j.heliyon.2023.e14952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
Diabetes is a metabolic disorder with an increased risk of developing heart failure. Inflammation and damaged vasculature are the cardinal features of diabetes-induced cardiac damage. Moreover, systemic metabolic stress triggers discordant intercellular communication, thus culminating in cardiac dysfunction. Fibroblast growth factor 21 (FGF21) is a pleiotropic hormone transducing cellular signals via fibroblast growth factor receptor 1 (FGFR1) and its co-receptor beta-klotho (β-KL). This study first demonstrated a decreased expression or activity of FGFR1 and β-KL in both human and mouse diabetic hearts. Reinforcing cardiac FGFR1 and β-KL expression can alleviate pro-inflammatory response and endothelial dysfunction upon diabetic stress. Using proteomics, novel cardiomyocyte-derived anti-inflammatory and proangiogenic factors regulated by FGFR1-β-KL signaling were identified. Although not exhaustive, this study provides a unique insight into the protective topology of the cardiac FGFR1-β-KL signaling-mediated intercellular reactions in the heart in response to metabolic stress.
Collapse
Affiliation(s)
- Namrita Kaur
- University of Manchester, Oxford Road, M13 9PT, Manchester, UK
| | | | | | - Jessica M. Miller
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St., Louisville, KY, 40202, USA
| | - Riham R.E. Abouleisa
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St., Louisville, KY, 40202, USA
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St., Louisville, KY, 40202, USA
| | - Jiahan Shen
- University of Manchester, Oxford Road, M13 9PT, Manchester, UK
| | - Handrean Soran
- University of Manchester, Oxford Road, M13 9PT, Manchester, UK
| | - Tamer M.A. Mohamed
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St., Louisville, KY, 40202, USA
| | - Wei Liu
- University of Manchester, Oxford Road, M13 9PT, Manchester, UK
| |
Collapse
|
2
|
Kaur N, Gare SR, Shen J, Raja R, Fonseka O, Liu W. Multi-organ FGF21-FGFR1 signaling in metabolic health and disease. Front Cardiovasc Med 2022; 9:962561. [PMID: 35983184 PMCID: PMC9378980 DOI: 10.3389/fcvm.2022.962561] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolic syndrome is a chronic systemic disease that is particularly manifested by obesity, diabetes, and hypertension, affecting multiple organs. The increasing prevalence of metabolic syndrome poses a threat to public health due to its complications, such as liver dysfunction and cardiovascular disease. Impaired adipose tissue plasticity is another factor contributing to metabolic syndrome. Emerging evidence demonstrates that fibroblast growth factors (FGFs) are critical players in organ crosstalk via binding to specific FGF receptors (FGFRs) and their co-receptors. FGFRs activation modulates intracellular responses in various cell types under metabolic stress. FGF21, in particular is considered as the key regulator for mediating systemic metabolic effects by binding to receptors FGFR1, FGFR3, and FGFR4. The complex of FGFR1 and beta Klotho (β-KL) facilitates endocrine and paracrine communication networks that physiologically regulate global metabolism. This review will discuss FGF21-mediated FGFR1/β-KL signaling pathways in the liver, adipose, and cardiovascular systems, as well as how this signaling is involved in the interplay of these organs during the metabolic syndrome. Furthermore, the clinical implications and therapeutic strategies for preventing metabolic syndrome and its complications by targeting FGFR1/β-KL are also discussed.
Collapse
Affiliation(s)
| | | | - Jiahan Shen
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Rida Raja
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | - Oveena Fonseka
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| | | |
Collapse
|
3
|
Zhang J, Gregorich ZR, Tao R, Kim GC, Lalit PA, Carvalho JL, Markandeya Y, Mosher DF, Palecek SP, Kamp TJ. Cardiac differentiation of human pluripotent stem cells using defined extracellular matrix proteins reveals essential role of fibronectin. eLife 2022; 11:e69028. [PMID: 35758861 PMCID: PMC9236614 DOI: 10.7554/elife.69028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/05/2022] [Indexed: 11/13/2022] Open
Abstract
Research and therapeutic applications using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) require robust differentiation strategies. Efforts to improve hPSC-CM differentiation have largely overlooked the role of extracellular matrix (ECM). The present study investigates the ability of defined ECM proteins to promote hPSC cardiac differentiation. Fibronectin (FN), laminin-111, and laminin-521 enabled hPSCs to attach and expand. However, only addition of FN promoted cardiac differentiation in response to growth factors Activin A, BMP4, and bFGF in contrast to the inhibition produced by laminin-111 or laminin-521. hPSCs in culture produced endogenous FN which accumulated in the ECM to a critical level necessary for effective cardiac differentiation. Inducible shRNA knockdown of FN prevented Brachyury+ mesoderm formation and subsequent hPSC-CM generation. Antibodies blocking FN binding integrins α4β1 or αVβ1, but not α5β1, inhibited cardiac differentiation. Furthermore, inhibition of integrin-linked kinase led to a decrease in phosphorylated AKT, which was associated with increased apoptosis and inhibition of cardiac differentiation. These results provide new insights into defined matrices for culture of hPSCs that enable production of FN-enriched ECM which is essential for mesoderm formation and efficient cardiac differentiation.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
| | - Zachery R Gregorich
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Ran Tao
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Gina C Kim
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Pratik A Lalit
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Juliana L Carvalho
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Department of Genomic Sciences and Biotechnology, University of BrasíliaBrasíliaBrazil
| | - Yogananda Markandeya
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Deane F Mosher
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Morgridge Institute for ResearchMadisonUnited States
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Sean P Palecek
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
- Department of Chemical and Biological Engineering, College of Engineering, University of WisconsinMadisonUnited States
| | - Timothy J Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| |
Collapse
|
4
|
Valussi M, Besser J, Wystub-Lis K, Zukunft S, Richter M, Kubin T, Boettger T, Braun T. Repression of Osmr and Fgfr1 by miR-1/133a prevents cardiomyocyte dedifferentiation and cell cycle entry in the adult heart. SCIENCE ADVANCES 2021; 7:eabi6648. [PMID: 34644107 PMCID: PMC8514096 DOI: 10.1126/sciadv.abi6648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Dedifferentiation of cardiomyocytes is part of the survival program in the remodeling myocardium and may be essential for enabling cardiomyocyte proliferation. In addition to transcriptional processes, non-coding RNAs play important functions for the control of cell cycle regulation in cardiomyocytes and cardiac regeneration. Here, we demonstrate that suppression of FGFR1 and OSMR by miR-1/133a is instrumental to prevent cardiomyocyte dedifferentiation and cell cycle entry in the adult heart. Concomitant inactivation of both miR-1/133a clusters in adult cardiomyocytes activates expression of cell cycle regulators, induces a switch from fatty acid to glycolytic metabolism, and changes expression of extracellular matrix genes. Inhibition of FGFR and OSMR pathways prevents most effects of miR-1/133a inactivation. Short-term miR-1/133a depletion protects cardiomyocytes against ischemia, while extended loss of miR-1/133a causes heart failure. Our results demonstrate a crucial role of miR-1/133a–mediated suppression of Osmr and Ffgfr1 in maintaining the postmitotic differentiated state of cardiomyocytes.
Collapse
Affiliation(s)
- Melissa Valussi
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
| | - Johannes Besser
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
| | - Katharina Wystub-Lis
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, D-60590 Frankfurt am Main, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, D-61231 Bad Nauheim, Germany
| | - Thomas Kubin
- Department of Cardiac Surgery, Kerckhoff Heart Center, Benekestrasse 2-8, D-61231 Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
- Corresponding author. (T.Bo.); (T.Br.)
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, D-61231 Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Corresponding author. (T.Bo.); (T.Br.)
| |
Collapse
|
5
|
Zhang Z, Li S, Wang K, Zhao Z, Zhang H, Li S, Jiang X. Whole-genome sequencing identifies novel candidate pathogenic variants associated with left ventricular non-compaction in a three-generation family. Clin Transl Med 2021; 11:e501. [PMID: 34459129 PMCID: PMC8351521 DOI: 10.1002/ctm2.501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zhe Zhang
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, P. R. China
| | - Shiying Li
- Shenzhen Byoryn Technology Co., Ltd, Shenzhen, Guangdong, P. R. China
| | - Kun Wang
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, P. R. China
| | - Zicheng Zhao
- Shenzhen Byoryn Technology Co., Ltd, Shenzhen, Guangdong, P. R. China
| | - Heng Zhang
- Department of Ultrasonography, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, P. R. China
| | - Shuaicheng Li
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, Guangdong, P. R. China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, P. R. China
| |
Collapse
|
6
|
Santos ARMP, Jang Y, Son I, Kim J, Park Y. Recapitulating Cardiac Structure and Function In Vitro from Simple to Complex Engineering. MICROMACHINES 2021; 12:mi12040386. [PMID: 33916254 PMCID: PMC8067203 DOI: 10.3390/mi12040386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Cardiac tissue engineering aims to generate in vivo-like functional tissue for the study of cardiac development, homeostasis, and regeneration. Since the heart is composed of various types of cells and extracellular matrix with a specific microenvironment, the fabrication of cardiac tissue in vitro requires integrating technologies of cardiac cells, biomaterials, fabrication, and computational modeling to model the complexity of heart tissue. Here, we review the recent progress of engineering techniques from simple to complex for fabricating matured cardiac tissue in vitro. Advancements in cardiomyocytes, extracellular matrix, geometry, and computational modeling will be discussed based on a technology perspective and their use for preparation of functional cardiac tissue. Since the heart is a very complex system at multiscale levels, an understanding of each technique and their interactions would be highly beneficial to the development of a fully functional heart in cardiac tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Jongseong Kim
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| | - Yongdoo Park
- Correspondence: (J.K.); (Y.P.); Tel.: +82-10-8858-7260 (J.K.); +82-10-4260-6460 (Y.P.)
| |
Collapse
|
7
|
Cooper SL, Sandhu H, Hussain A, Mee C, Maddock H. Involvement of mitogen activated kinase kinase 7 intracellular signalling pathway in Sunitinib-induced cardiotoxicity. Toxicology 2018; 394:72-83. [DOI: 10.1016/j.tox.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 12/30/2022]
|
8
|
Prathipati P, Nandi SS, Mishra PK. Stem Cell-Derived Exosomes, Autophagy, Extracellular Matrix Turnover, and miRNAs in Cardiac Regeneration during Stem Cell Therapy. Stem Cell Rev Rep 2017; 13:79-91. [PMID: 27807762 DOI: 10.1007/s12015-016-9696-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell therapy (SCT) raises the hope for cardiac regeneration in ischemic hearts. However, underlying molecular mechanisms for repair of dead myocardium by SCT in the ischemic heart is poorly understood. Growing evidences suggest that cardiac matrix stiffness and differential expressions of miRNAs play a crucial role in stem cell survival and differentiation. However, their roles on transplanted stem cells, for myocardial repair of the ischemic heart, remain unclear. Transplanted stem cells may act in an autocrine and/or paracrine manner to regenerate the dead myocardium. Paracrine mediators such as stem cell-derived exosomes are emerging as a novel therapeutic strategy to overcome some of the limitations of SCT. These exosomes carry microRNAs (miRNAs) that may regulate stem cell differentiation into a specific lineage. MicroRNAs may also contribute to stiffness of surrounding matrix by regulating extracellular matrix (ECM) turnover. The survival of transplanted stem cell depends on its autophagic process that maintains cellular homeostasis. Therefore, exosomes, miRNAs, extracellular matrix turnover, and autophagy may have an integral role in improving the efficacy of SCT. This review elaborates the specific roles of these regulatory components on cardiac regeneration in the ischemic heart during SCT.
Collapse
Affiliation(s)
- Priyanka Prathipati
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shyam Sundar Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Paras Kumar Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
9
|
Differential proteomic analysis of respiratory samples from patients suffering from influenza. Virusdisease 2016; 27:226-233. [PMID: 28466033 DOI: 10.1007/s13337-016-0332-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/20/2016] [Indexed: 02/03/2023] Open
Abstract
The exact molecular pathways involved in the pathogenesis of influenza are yet unclear. In the present study we investigated the upper respiratory proteome in influenza patients. 200 nasal and throat swab samples were collected from patients suffering from acute respiratory illness. These samples were confirmed for influenza pandemic A/H1N1/2009 and influenza type B using qRT-PCR. 10 similar swabs were collected from healthy individuals and were used as controls. Proteins were extracted from the cell pellets and were subjected to 2-D gel electrophoresis. The differentially expressed proteins were identified using MALDI-TOF. Identified proteins were classified into different functional groups based on functions reported in the databases. 25 % of these proteins were involved in cytoskeletal formation, whereas 14 % were involved in signal transduction. Proteins involved in anti-viral responses, Ca-signaling, transport, and tumor suppression constituted 10 % each, where as 5 % of proteins each belong to Nicotinic acetylcholine receptor, Protein Synthesis and anti-bacterial proteins. 10 % of the proteins have not been described previously. This is the first report on respiratory proteome profile in Influenza patients. The study emphasizes the role of such profiling studies using multiple platforms for bio-marker discoveries, especially non-invasive diagnostic marker in Influenza and other infectious diseases.
Collapse
|
10
|
Integrating Transcriptomic and Proteomic Data Using Predictive Regulatory Network Models of Host Response to Pathogens. PLoS Comput Biol 2016; 12:e1005013. [PMID: 27403523 PMCID: PMC4942116 DOI: 10.1371/journal.pcbi.1005013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 06/06/2016] [Indexed: 12/17/2022] Open
Abstract
Mammalian host response to pathogenic infections is controlled by a complex regulatory network connecting regulatory proteins such as transcription factors and signaling proteins to target genes. An important challenge in infectious disease research is to understand molecular similarities and differences in mammalian host response to diverse sets of pathogens. Recently, systems biology studies have produced rich collections of omic profiles measuring host response to infectious agents such as influenza viruses at multiple levels. To gain a comprehensive understanding of the regulatory network driving host response to multiple infectious agents, we integrated host transcriptomes and proteomes using a network-based approach. Our approach combines expression-based regulatory network inference, structured-sparsity based regression, and network information flow to infer putative physical regulatory programs for expression modules. We applied our approach to identify regulatory networks, modules and subnetworks that drive host response to multiple influenza infections. The inferred regulatory network and modules are significantly enriched for known pathways of immune response and implicate apoptosis, splicing, and interferon signaling processes in the differential response of viral infections of different pathogenicities. We used the learned network to prioritize regulators and study virus and time-point specific networks. RNAi-based knockdown of predicted regulators had significant impact on viral replication and include several previously unknown regulators. Taken together, our integrated analysis identified novel module level patterns that capture strain and pathogenicity-specific patterns of expression and helped identify important regulators of host response to influenza infection. An important challenge in infectious disease research is to understand how the human immune system responds to different types of pathogenic infections. An important component of mounting proper response is the transcriptional regulatory network that specifies the context-specific gene expression program in the host cell. However, our understanding of this regulatory network and how it drives context-specific transcriptional programs is incomplete. To address this gap, we performed a network-based analysis of host response to influenza viruses that integrated high-throughput mRNA- and protein measurements and protein-protein interaction networks to identify virus and pathogenicity-specific modules and their upstream physical regulatory programs. We inferred regulatory networks for human cell line and mouse host systems, which recapitulated several known regulators and pathways of the immune response and viral life cycle. We used the networks to study time point and strain-specific subnetworks and to prioritize important regulators of host response. We predicted several novel regulators, both at the mRNA and protein levels, and experimentally verified their role in the virus life cycle based on their ability to significantly impact viral replication.
Collapse
|
11
|
Krejci E, Pesevski Z, Nanka O, Sedmera D. Physiological role of FGF signaling in growth and remodeling of developing cardiovascular system. Physiol Res 2016; 65:425-35. [PMID: 27070743 DOI: 10.33549/physiolres.933216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling plays an important role during embryonic induction and patterning, as well as in modulating proliferative and hypertrophic growth in fetal and adult organs. Hemodynamically induced stretching is a powerful physiological stimulus for embryonic myocyte proliferation. The aim of this study was to assess the effect of FGF2 signaling on growth and vascularization of chick embryonic ventricular wall and its involvement in transmission of mechanical stretch-induced signaling to myocyte growth in vivo. Myocyte proliferation was significantly higher at the 48 h sampling interval in pressure-overloaded hearts. Neither Western blotting, nor immunohistochemistry performed on serial paraffin sections revealed any changes in the amount of myocardial FGF2 at that time point. ELISA showed a significant increase of FGF2 in the serum. Increased amount of FGF2 mRNA in the heart was confirmed by real time PCR. Blocking of FGF signaling by SU5402 led to decreased myocyte proliferation, hemorrhages in the areas of developing vasculature in epicardium and digit tips. FGF2 synthesis is increased in embryonic ventricular cardiomyocytes in response to increased stretch due to pressure overload. Inhibition of FGF signaling impacts also vasculogenesis, pointing to partial functional redundancy in paracrine control of cell proliferation in the developing heart.
Collapse
Affiliation(s)
- E Krejci
- Institute of Anatomy, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | |
Collapse
|
12
|
Cardiotrophin-1 promotes cardiomyocyte differentiation from mouse induced pluripotent stem cells via JAK2/STAT3/Pim-1 signaling pathway. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2016; 12:591-9. [PMID: 26788034 PMCID: PMC4712363 DOI: 10.11909/j.issn.1671-5411.2015.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND The induced pluripotent stem cell (iPSC) has shown great potential in cellular therapy of myocardial infarction (MI), while its application is hampered by the low efficiency of cardiomyocyte differentiation. The present study was designed to investigate the effects of cardiotrophin-1 (CT-1) on cardiomyocyte differentiation from mouse induced pluripotent stem cells (miPSCs) and the underlying mechanisms involved. METHODS The optimal treatment condition for cardiomyocyte differentiation from miPSCs was established with ideal concentration (10 ng/mL) and duration (from day 3 to day 14) of CT-1 administration. Up-regulated expression of cardiac specific genes that accounted for embryonic cardiogenesis was observed by quantitative RT-PCR. Elevated amount of α-myosin heavy chain (α-MHC) and cardiac troponin I (cTn I) positive cells were detected by immunofluorescence staining and flow cytometry analysis in CT-1 group. RESULTS Transmission electron microscopic analysis revealed that cells treated with CT-1 showed better organized sacromeric structure and more mitochondria, which are morphological characteristic of matured cardiomyocytes. Western blot demonstrated that CT-1 promotes cardiomyocyte differentiation from miPSCs partly via JAK2/STAT3/Pim-1 pathway as compared with control group. CONCLUSIONS These findings suggested that CT-1 could enhance the cardiomyocyte differentiation as well as the maturation of mouse induced pluripotent stem cell derived cardiomyocytes by regulating JAK2/STAT3/Pim-1signaling pathway.
Collapse
|
13
|
A Functional Role of Fibroblast Growth Factor Receptor 1 (FGFR1) in the Suppression of Influenza A Virus Replication. PLoS One 2015; 10:e0124651. [PMID: 25909503 PMCID: PMC4409105 DOI: 10.1371/journal.pone.0124651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/17/2015] [Indexed: 12/28/2022] Open
Abstract
Influenza A virus causes annual epidemics and occasional pandemics in humans. Here, we investigated four members of the fibroblast growth factor receptor (FGFR) family; FGFR1 to 4, and examined their expression patterns in human lung epithelial cells A549 with influenza A virus infection. We identified a functional role of FGFR1 in influenza A/Puerto Rico/8/1934 (PR8) and A/Anhui/01/2005 (H5N1) virus replication. Our results showed that FGFR1 silencing by siRNA interference promoted influenza A/PR8 and H5N1 virus replication in A549 cells, while lentivirus-mediated exogenous FGFR1 expression significantly suppressed influenza A virus replication; however, FGFR4 did not have the same effects. Moreover, FGFR1 phosphorylation levels were downregulated in A549 cells by influenza A virus infection, while the repression of FGFR1 kinase using PD173074, a potent and selective FGFR1 inhibitor, could enhance virus replication. Furthermore, we found that FGFR1 inhibits influenza virus internalization, but not binding, during viral entry. These results suggested that FGFR1 specifically antagonizes influenza A virus replication, probably by blocking viral entry.
Collapse
|
14
|
Meganathan K, Sotiriadou I, Natarajan K, Hescheler J, Sachinidis A. Signaling molecules, transcription growth factors and other regulators revealed from in-vivo and in-vitro models for the regulation of cardiac development. Int J Cardiol 2015; 183:117-28. [PMID: 25662074 DOI: 10.1016/j.ijcard.2015.01.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/19/2014] [Accepted: 01/25/2015] [Indexed: 02/08/2023]
Abstract
Several in-vivo heart developmental models have been applied to decipher the cardiac developmental patterning encompassing early, dorsal, cardiac and visceral mesoderm as well as various transcription factors such as Gata, Hand, Tin, Dpp, Pnr. The expression of cardiac specific transcription factors, such as Gata4, Tbx5, Tbx20, Tbx2, Tbx3, Mef2c, Hey1 and Hand1 are of fundamental significance for the in-vivo cardiac development. Not only the transcription factors, but also the signaling molecules involved in cardiac development were conserved among various species. Enrichment of the bone morphogenic proteins (BMPs) in the anterior lateral plate mesoderm is essential for the initiation of myocardial differentiation and the cardiac developmental process. Moreover, the expression of a number of cardiac transcription factors and structural genes initiate cardiac differentiation in the medial mesoderm. Other signaling molecules such as TGF-beta, IGF-1/2 and the fibroblast growth factor (FGF) play a significant role in cardiac repair/regeneration, ventricular heart development and specification of early cardiac mesoderm, respectively. The role of the Wnt signaling in cardiac development is still controversial discussed, as in-vitro results differ dramatically in relation to the animal models. Embryonic stem cells (ESC) were utilized as an important in-vitro model for the elucidation of the cardiac developmental processes since they can be easily manipulated by numerous signaling molecules, growth factors, small molecules and genetic manipulation. Finally, in the present review the dynamic role of the long noncoding RNA and miRNAs in the regulation of cardiac development are summarized and discussed.
Collapse
Affiliation(s)
- Kesavan Meganathan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Isaia Sotiriadou
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Karthick Natarajan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Jürgen Hescheler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Germany.
| |
Collapse
|
15
|
The heart: mostly postmitotic or mostly premitotic? Myocyte cell cycle, senescence, and quiescence. Can J Cardiol 2014; 30:1270-8. [PMID: 25442430 DOI: 10.1016/j.cjca.2014.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/21/2014] [Accepted: 08/21/2014] [Indexed: 11/21/2022] Open
Abstract
The concept of myocyte division and myocyte-mediated regeneration has re-emerged in the past 5 years through development of sophisticated transgenic mice and carbon-dating of cells. Although recently, a couple of studies have been conducted as an attempt to intervene in myocyte division, the efficiency in adult animals remains discouragingly low. Re-enforcing myocyte division is a vision that has been desired for decades, leading to years of experience in myocyte resistance to proproliferative stimuli. Previous attempts have indeed provided a platform for basic knowledge on molecular players and signalling in myocytes. However, natural biological processes such as hypertrophy and binucleation provide layers of complexity in interpretation of previous and current findings. A major hurdle in mediating myocyte division is a lack of insight in the myocyte cell cycle. To date, no knowledge is gained on myoycte cell cycle progression and/or duration. This review will include an overview of previous and current literature on myocyte cell cycle and division. Furthermore, the limitations of current approaches and basic questions that might be essential in understanding myocardial resistance to division will be discussed.
Collapse
|
16
|
Sullivan KE, Black LD. The role of cardiac fibroblasts in extracellular matrix-mediated signaling during normal and pathological cardiac development. J Biomech Eng 2014; 135:71001. [PMID: 23720014 DOI: 10.1115/1.4024349] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 04/30/2013] [Indexed: 01/18/2023]
Abstract
The extracellular matrix is no longer considered a static support structure for cells but a dynamic signaling network with the power to influence cell, tissue, and whole organ physiology. In the myocardium, cardiac fibroblasts are the primary cell type responsible for the synthesis, deposition, and degradation of matrix proteins, and they therefore play a critical role in the development and maintenance of functional heart tissue. This review will summarize the extensive research conducted in vivo and in vitro, demonstrating the influence of both physical and chemical stimuli on cardiac fibroblasts and how these interactions impact both the extracellular matrix and, by extension, cardiomyocytes. This work is of considerable significance, given that cardiovascular diseases are marked by extensive remodeling of the extracellular matrix, which ultimately impairs the functional capacity of the heart. We seek to summarize the unique role of cardiac fibroblasts in normal cardiac development and the most prevalent cardiac pathologies, including congenital heart defects, hypertension, hypertrophy, and the remodeled heart following myocardial infarction. We will conclude by identifying existing holes in the research that, if answered, have the potential to dramatically improve current therapeutic strategies for the repair and regeneration of damaged myocardium via mechanotransductive signaling.
Collapse
|
17
|
Samsa LA, Yang B, Liu J. Embryonic cardiac chamber maturation: Trabeculation, conduction, and cardiomyocyte proliferation. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2013; 163C:157-68. [PMID: 23720419 PMCID: PMC3723796 DOI: 10.1002/ajmg.c.31366] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Congenital heart diseases are some of the most common human birth defects. Though some congenital heart defects can be surgically corrected, treatment options for other congenital heart diseases are very limited. In many congenital heart diseases, genetic defects lead to impaired embryonic heart development or growth. One of the key development processes in cardiac development is chamber maturation, and alterations in this maturation process can manifest as a variety of congenital defects including non-compaction, systolic dysfunction, diastolic dysfunction, and arrhythmia. During development, to meet the increasing metabolic demands of the developing embryo, the myocardial wall undergoes extensive remodeling characterized by the formation of muscular luminal protrusions called cardiac trabeculae, increased cardiomyocyte mass, and development of the ventricular conduction system. Though the basic morphological and cytological changes involved in early heart development are clear, much remains unknown about the complex biomolecular mechanisms governing chamber maturation. In this review, we highlight evidence suggesting that a wide variety of basic signaling pathways and biomechanical forces are involved in cardiac wall maturation.
Collapse
Affiliation(s)
- Leigh Ann Samsa
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Betsy Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
18
|
Sakurai T, Tsuchida M, Lampe PD, Murakami M. Cardiomyocyte FGF signaling is required for Cx43 phosphorylation and cardiac gap junction maintenance. Exp Cell Res 2013; 319:2152-65. [PMID: 23742896 DOI: 10.1016/j.yexcr.2013.05.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/22/2013] [Accepted: 05/25/2013] [Indexed: 02/06/2023]
Abstract
Cardiac remodeling resulting from impairment of myocardial integrity leads to heart failure, through still incompletely understood mechanisms. The fibroblast growth factor (FGF) system has been implicated in tissue maintenance, but its role in the adult heart is not well defined. We hypothesized that the FGF system plays a role in the maintenance of cardiac homeostasis, and the impairment of cardiomyocyte FGF signaling leads to pathological cardiac remodeling. We showed that FGF signaling is required for connexin 43 (Cx43) localization at cell-cell contacts in isolated cardiomyocytes and COS7 cells. Lack of FGF signaling led to decreased Cx43 phosphorylation at serines 325/328/330 (S325/328/330), sites known to be important for assembly of gap junctions. Cx43 instability induced by FGF inhibition was restored by the Cx43 S325/328/330 phospho-mimetic mutant, suggesting FGF-dependent phosphorylation of these sites. Consistent with these in vitro findings, cardiomyocyte-specific inhibition of FGF signaling in adult mice demonstrated mislocalization of Cx43 at intercalated discs, whereas localization of N-cadherin and desmoplakin was not affected. This led to premature death resulting from impaired cardiac remodeling. We conclude that cardiomyocyte FGF signaling is essential for cardiomyocyte homeostasis through phosphorylation of Cx43 at S325/328/330 residues which are important for the maintenance of gap junction.
Collapse
Affiliation(s)
- Takashi Sakurai
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA.
| | | | | | | |
Collapse
|
19
|
El-Mounayri O, Mihic A, Shikatani EA, Gagliardi M, Steinbach SK, Dubois N, DaCosta R, Li RK, Keller G, Husain M. Serum-free differentiation of functional human coronary-like vascular smooth muscle cells from embryonic stem cells. Cardiovasc Res 2012; 98:125-35. [DOI: 10.1093/cvr/cvs357] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
20
|
Schlueter J, Brand T. Epicardial progenitor cells in cardiac development and regeneration. J Cardiovasc Transl Res 2012; 5:641-53. [PMID: 22653801 DOI: 10.1007/s12265-012-9377-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 05/15/2012] [Indexed: 01/25/2023]
Abstract
The epicardium forms an epithelial layer on the surface of the heart. It is derived from a cluster of mesothelial cells, which is termed the proepicardium. The proepicardium gives rise not only to the epicardium but also to epicardium-derived cells. These cells populate the myocardial wall and differentiate into smooth muscle cells, fibroblast, and possibly endothelial cells. In this review, the formation of the proepicardium is discussed. Marker genes, suitable to identify these cells in the embryo and in the adult, are introduced. Recent evidence suggests that the PE is made up of distinct cell populations. These cell lineages can be distinguished on the basis of marker gene expression and differ in their differentiation potential. The role of the epicardium as a resource for cardiac stem cells and its importance in cardiac regeneration is also discussed.
Collapse
Affiliation(s)
- Jan Schlueter
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Hill End Road, Harefield, Middlesex, UK
| | | |
Collapse
|
21
|
Abstract
Abstract
The embryonic heart initially consists of only two cell layers, the endocardium and the myocardium. The epicardium, which forms an epithelial layer on the surface of the heart, is derived from a cluster of mesothelial cells developing at the base of the venous inflow tract of the early embryonic heart. This cell cluster is termed the proepicardium and gives rise not only to the epicardium but also to epicardium-derived cells. These cells populate the myocardial wall and differentiate into smooth muscle cells and fibroblasts, while the contribution to the vascular endothelial lineage is uncertain. In this review we will discuss the signaling molecules involved in recruiting mesodermal cells to undergo proepicardium formation and guide these cells to the myocardial surface. Marker genes which are suitable to follow these cells during proepicardium formation and cell migration will be introduced. We will address whether the proepicardium consists of a homogenous cell population or whether different cell lineages are present. Finally the role of the epicardium as a source for cardiac stem cells and its importance in cardiac regeneration, in particular in the zebrafish and mouse model systems is discussed.
Collapse
Affiliation(s)
- Jan Schlueter
- 1Harefield Heart Science Centre, National Heart
and Lung Institute, Imperial College London, Hill End Road, Harefield,
Middlesex, UB9 6JH, United Kingdom
| | - Thomas Brand
- 1Harefield Heart Science Centre, National Heart
and Lung Institute, Imperial College London, Hill End Road, Harefield,
Middlesex, UB9 6JH, United Kingdom
| |
Collapse
|
22
|
Zhang J, Liu J, Huang Y, Chang JYF, Liu L, McKeehan WL, Martin JF, Wang F. FRS2α-mediated FGF signals suppress premature differentiation of cardiac stem cells through regulating autophagy activity. Circ Res 2011; 110:e29-39. [PMID: 22207710 DOI: 10.1161/circresaha.111.255950] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE Although the fibroblast growth factor (FGF) signaling axis plays important roles in heart development, the molecular mechanism by which the FGF regulates cardiogenesis is not fully understood. OBJECTIVE To investigate the mechanism by which FGF signaling regulates cardiac progenitor cell differentiation. METHODS AND RESULTS Using mice with tissue-specific ablation of FGF receptors and FGF receptor substrate 2α (Frs2α) in heart progenitor cells, we demonstrate that disruption of FGF signaling leads to premature differentiation of cardiac progenitor cells in mice. Using embryoid body cultures of mouse embryonic stem cells, we reveal that FGF signaling promotes mesoderm differentiation in embryonic stem cells but inhibits cardiomyocyte differentiation of the mesoderm cells at later stages. Furthermore, we also report that inhibiting FRS2α-mediated signals increases autophagy and that activating autophagy promotes myocardial differentiation and vice versa. CONCLUSIONS The results indicate that the FGF/FRS2α-mediated signals prevent premature differentiation of heart progenitor cells through suppressing autophagy. The findings provide the first evidence that autophagy plays a role in heart progenitor differentiation.
Collapse
Affiliation(s)
- Jue Zhang
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030-3303, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Christalla P, Hudson JE, Zimmermann WH. The cardiogenic niche as a fundamental building block of engineered myocardium. Cells Tissues Organs 2011; 195:82-93. [PMID: 21996934 DOI: 10.1159/000331407] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cardiac muscle engineering is evolving rapidly, aiming at the provision of innovative models for drug development and therapeutic myocardium. The progress in this field will depend crucially on the proper exploitation of stem cell technologies. Understanding the processes governing stem cell differentiation towards a desired phenotype and subsequent maturation in an organotypic manner will be key to ultimately providing realistic tissue models or therapeutics. Cardiogenesis is controlled by milieu factors that collectively constitute a so-called cardiogenic niche. The components of the cardiogenic niche are not yet fully defined but include paracrine factors and instructive extracellular matrix. Both are provided by supportive stromal cells under strict spatial and temporal control. Detailed knowledge on the exact composition and functionality of the dynamic cardiogenic niche during development will likely be instrumental to further advance cardiac muscle engineering. This review will discuss the concept of myocardial tissue engineering from the stem cell/developmental biology perspective and put forward the hypothesis of the cardiogenic niche as a fundamental building block of tissue-engineered myocardium.
Collapse
Affiliation(s)
- Peter Christalla
- Department of Pharmacology, University Medical Center Göttingen and Heart Research Center Göttingen, Germany
| | | | | |
Collapse
|
24
|
Abstract
Regulation of organ growth is critical during embryogenesis. At the cellular level, mechanisms controlling the size of individual embryonic organs include cell proliferation, differentiation, migration, and attrition through cell death. All these mechanisms play a role in cardiac morphogenesis, but experimental studies have shown that the major determinant of cardiac size during prenatal development is myocyte proliferation. As this proliferative capacity becomes severely restricted after birth, the number of cell divisions that occur during embryogenesis limits the growth potential of the postnatal heart. We summarize here current knowledge concerning regional control of myocyte proliferation as related to cardiac morphogenesis and dysmorphogenesis. There are significant spatial and temporal differences in rates of cell division, peaking during the preseptation period and then gradually decreasing toward birth. Analysis of regional rates of proliferation helps to explain the mechanics of ventricular septation, chamber morphogenesis, and the development of the cardiac conduction system. Proliferation rates are influenced by hemodynamic loading, and transduced by autocrine and paracrine signaling by means of growth factors. Understanding the biological response of the developing heart to such factors and physical forces will further our progress in engineering artificial myocardial tissues for heart repair and designing optimal treatment strategies for congenital heart disease.
Collapse
Affiliation(s)
- David Sedmera
- Charles University in Prague, First Faculty of Medicine, Institute of Anatomy, Prague, Czech Republic.
| | | |
Collapse
|
25
|
van Weerd JH, Koshiba-Takeuchi K, Kwon C, Takeuchi JK. Epigenetic factors and cardiac development. Cardiovasc Res 2011; 91:203-11. [PMID: 21606181 DOI: 10.1093/cvr/cvr138] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Congenital heart malformations remain the leading cause of death related to birth defects. Recent advances in developmental and regenerative cardiology have shed light on a mechanistic understanding of heart development that is controlled by a transcriptional network of genetic and epigenetic factors. This article reviews the roles of chromatin remodelling factors important for cardiac development with the current knowledge of cardiac morphogenesis, regeneration, and direct cardiac differentiation. In the last 5 years, critical roles of epigenetic factors have been revealed in the cardiac research field.
Collapse
Affiliation(s)
- Jan Hendrick van Weerd
- Cardiovascular Regeneration, Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
26
|
Rajala K, Pekkanen-Mattila M, Aalto-Setälä K. Cardiac differentiation of pluripotent stem cells. Stem Cells Int 2011; 2011:383709. [PMID: 21603143 PMCID: PMC3096314 DOI: 10.4061/2011/383709] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/01/2011] [Accepted: 02/08/2011] [Indexed: 01/12/2023] Open
Abstract
The ability of human pluripotent stem cells to differentiate towards the cardiac lineage has attracted significant interest, initially with a strong focus on regenerative medicine. The ultimate goal to repair the heart by cardiomyocyte replacement has, however, proven challenging. Human cardiac differentiation has been difficult to control, but methods are improving, and the process, to a certain extent, can be manipulated and directed. The stem cell-derived cardiomyocytes described to date exhibit rather immature functional and structural characteristics compared to adult cardiomyocytes. Thus, a future challenge will be to develop strategies to reach a higher degree of cardiomyocyte maturation in vitro, to isolate cardiomyocytes from the heterogeneous pool of differentiating cells, as well as to guide the differentiation into the desired subtype, that is, ventricular, atrial, and pacemaker cells. In this paper, we will discuss the strategies for the generation of cardiomyocytes from pluripotent stem cells and their characteristics, as well as highlight some applications for the cells.
Collapse
Affiliation(s)
- Kristiina Rajala
- Regea - Institute for Regenerative Medicine, University of Tampere, Tampere University Hospital, 33520 Tampere, Finland
| | | | | |
Collapse
|
27
|
Brade T, Kumar S, Cunningham TJ, Chatzi C, Zhao X, Cavallero S, Li P, Sucov HM, Ruiz-Lozano P, Duester G. Retinoic acid stimulates myocardial expansion by induction of hepatic erythropoietin which activates epicardial Igf2. Development 2011; 138:139-48. [PMID: 21138976 DOI: 10.1242/dev.054239] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Epicardial signaling and Rxra are required for expansion of the ventricular myocardial compact zone. Here, we examine Raldh2(-/-) and Rxra(-/-) mouse embryos to investigate the role of retinoic acid (RA) signaling in this developmental process. The heart phenotypes of Raldh2 and Rxra mutants are very similar and are characterized by a prominent defect in ventricular compact zone growth. Although RA activity is completely lost in Raldh2(-/-) epicardium and the adjacent myocardium, RA activity is not lost in Rxra(-/-) hearts, suggesting that RA signaling in the epicardium/myocardium is not required for myocardial compact zone formation. We explored the possibility that RA-mediated target gene transcription in non-cardiac tissues is required for this process. We found that hepatic expression of erythropoietin (EPO), a secreted factor implicated in myocardial expansion, is dependent on both Raldh2 and Rxra. Chromatin immunoprecipitation studies support Epo as a direct target of RA signaling in embryonic liver. Treatment of an epicardial cell line with EPO, but not RA, upregulates Igf2. Furthermore, both Raldh2(-/-) and Rxra(-/-) hearts exhibit downregulation of Igf2 mRNA in the epicardium. EPO treatment of cultured Raldh2(-/-) hearts restores epicardial Igf2 expression and rescues ventricular cardiomyocyte proliferation. We propose a new model for the mechanism of RA-mediated myocardial expansion in which RA directly induces hepatic Epo resulting in activation of epicardial Igf2 that stimulates compact zone growth. This RA-EPO-IGF2 signaling axis coordinates liver hematopoiesis with heart development.
Collapse
Affiliation(s)
- Thomas Brade
- Sanford-Burnham Medical Research Institute, Development and Aging Program, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
The epicardium in cardiac repair: From the stem cell view. Pharmacol Ther 2011; 129:82-96. [DOI: 10.1016/j.pharmthera.2010.09.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 09/09/2010] [Indexed: 12/12/2022]
|
29
|
Ishii Y, Garriock RJ, Navetta AM, Coughlin LE, Mikawa T. BMP signals promote proepicardial protrusion necessary for recruitment of coronary vessel and epicardial progenitors to the heart. Dev Cell 2010; 19:307-16. [PMID: 20708592 DOI: 10.1016/j.devcel.2010.07.017] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 04/21/2010] [Accepted: 07/26/2010] [Indexed: 11/25/2022]
Abstract
The coronary vessels and epicardium arise from an extracardiac rudiment called the proepicardium. Failed fusion of the proepicardium to the heart results in severe coronary and heart defects. However, it is unknown how the proepicardium protrudes toward and attaches to the looping heart tube. Here, we show that ectopic expression of BMP ligands in the embryonic myocardium can cause proepicardial cells to target aberrant regions of the heart. Additionally, misexpression of a BMP antagonist, Noggin, suppresses proepicardium protrusion and contact with the heart. Finally, proepicardium explant preferentially expands toward a cocultured heart segment. This preference can be mimicked by BMP2/4 and suppressed by Noggin. These results support a model in which myocardium-derived BMP signals regulate the entry of coronary progenitors to the specific site of the heart by directing their morphogenetic movement.
Collapse
Affiliation(s)
- Yasuo Ishii
- Cardiovascular Research Institute, University of California, San Francisco, 94158, USA
| | | | | | | | | |
Collapse
|
30
|
Abstract
Recent studies have shown that cardiomyocytes can be induced to differentiate from several types of stem cells. Techniques to purify and transplant stem-cell-derived cardiomyocytes have also been developed, and the transplanted cells have been demonstrated to reside in the recipient hearts for long periods. Recently, cardiomyocyte cell sheets enabling transplantation of viable tissue have been reported. Promising results have also been obtained using cytokines to mobilize stem cells in vivo as a potential treatment for heart failure. However, a number of hurdles remain in the quest to treat heart failure by cell transplantation without the need for a donor at the preclinical stage. In the next phase, the field needs to develop innovations to specifically differentiate cardiomyocytes from stem cells, to purify cardiomyocytes from contaminating cells in a cell mixture using a high-throughput method, and to establish and maintain artificial regenerated tissue using tissue engineering for transplantation.
Collapse
Affiliation(s)
- Keiichi Fukuda
- Keio University School of Medicine , Cardiology Division, Department of Internal Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
31
|
Liao S, Bodmer JR, Azhar M, Newman G, Coffin JD, Doetschman T, Schultz JEJ. The influence of FGF2 high molecular weight (HMW) isoforms in the development of cardiac ischemia-reperfusion injury. J Mol Cell Cardiol 2010; 48:1245-54. [PMID: 20116383 DOI: 10.1016/j.yjmcc.2010.01.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 01/19/2010] [Accepted: 01/20/2010] [Indexed: 12/01/2022]
Abstract
Fibroblast growth factor 2 (FGF2) consists of multiple protein isoforms (low [LMW] and high molecular weight [HMW]), which are localized to different cellular compartments, indicating unique biological activity. We previously showed that the LMW isoform is important in protecting the heart from myocardial dysfunction associated with ischemia-reperfusion (I/R) injury, but the roles of the HMW isoforms remain unknown. To elucidate the role of HMW isoforms in I/R and cardioprotection, hearts from novel mouse models, in which the murine FGF2 HMWs are knocked out (HMWKO) or the human FGF2 24 kDa HMW isoform is overexpressed (HMW Tg) and their wildtype (Wt) or non-transgenic (NTg) cohorts were subjected to an ex vivo work-performing heart model of I/R. There was a significant improvement in post-ischemic recovery of cardiac function in HMWKO hearts (76+/-5%, p<0.05) compared to Wt hearts (55+/-5%), with a corresponding decrease in HMW Tg function (line 20: 38+/-6% and line 28: 33+/-4%, p<0.05) compared to non-transgenic hearts (68+/-9%). FGF2 LMW isoform was secreted from Wt and HMWKO hearts during I/R, and a FGF receptor (FGFR) inhibitor, PD173074 caused a decrease in cardiac function when administered in I/R in Wt and FGF2 HMWKO hearts (p<0.05), indicating that FGFR is involved in FGF2 LMW isoform's biological effect in ischemia-reperfusion injury. Moreover, overexpression of HMW isoform reduced FGFR1 phosphorylation/activation with no further decrease in the phosphorylation state in the presence of the FGFR inhibitor. Overall, our data indicate that HMW isoforms have a detrimental role in the development of post-ischemic myocardial dysfunction.
Collapse
Affiliation(s)
- Siyun Liao
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, ML 0575, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Damon BJ, Rémond MC, Bigelow MR, Trusk TC, Xie W, Perucchio R, Sedmera D, Denslow S, Thompson RP. Patterns of muscular strain in the embryonic heart wall. Dev Dyn 2009; 238:1535-46. [PMID: 19418446 DOI: 10.1002/dvdy.21958] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The hypothesis that inner layers of contracting muscular tubes undergo greater strain than concentric outer layers was tested by numerical modeling and by confocal microscopy of strain within the wall of the early chick heart. We modeled the looped heart as a thin muscular shell surrounding an inner layer of sponge-like trabeculae by two methods: calculation within a two-dimensional three-variable lumped model and simulated expansion of a three-dimensional, four-layer mesh of finite elements. Analysis of both models, and correlative microscopy of chamber dimensions, sarcomere spacing, and membrane leaks, indicate a gradient of strain decreasing across the wall from highest strain along inner layers. Prediction of wall thickening during expansion was confirmed by ultrasonography of beating hearts. Degree of stretch determined by radial position may thus contribute to observed patterns of regional myocardial conditioning and slowed proliferation, as well as to the morphogenesis of ventricular trabeculae and conduction fascicles. Developmental Dynamics 238:1535-1546, 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Brooke J Damon
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ishii Y, Weinberg K, Oda-Ishii I, Coughlin L, Mikawa T. Morphogenesis and cytodifferentiation of the avian retinal pigmented epithelium require downregulation of Group B1 Sox genes. Development 2009; 136:2579-89. [PMID: 19570849 DOI: 10.1242/dev.031344] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The optic vesicle is a multipotential primordium of the retina, which becomes subdivided into the neural retina and retinal pigmented epithelium domains. Although the roles of several paracrine factors in patterning the optic vesicle have been studied extensively, little is known about cell-autonomous mechanisms that regulate coordinated cell morphogenesis and cytodifferentiation of the retinal pigmented epithelium. Here we demonstrate that members of the SoxB1 gene family, Sox1, Sox2 and Sox3, are all downregulated in the presumptive retinal pigmented epithelium. Constitutive maintenance of SoxB1 expression in the presumptive retinal pigmented epithelium both in vivo and in vitro resulted in the absence of cuboidal morphology and pigmentation, and in concomitant induction of neural differentiation markers. We also demonstrate that exogenous Fgf4 inhibits downregulation all SoxB1 family members in the presumptive retinal pigment epithelium. These results suggest that retinal pigment epithelium morphogenesis and cytodifferentiation requires SoxB1 downregulation, which depends on the absence of exposure to an FGF-like signal.
Collapse
Affiliation(s)
- Yasuo Ishii
- University of California San Francisco, Cardiovascular Research Institute, Rock Hall Room 384D, San Francisco, CA 94158, USA
| | | | | | | | | |
Collapse
|
34
|
Pennisi DJ, Mikawa T. FGFR-1 is required by epicardium-derived cells for myocardial invasion and correct coronary vascular lineage differentiation. Dev Biol 2009; 328:148-59. [PMID: 19389363 PMCID: PMC2724599 DOI: 10.1016/j.ydbio.2009.01.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 01/09/2009] [Accepted: 01/16/2009] [Indexed: 12/11/2022]
Abstract
Critical steps in coronary vascular formation include the epithelial-mesenchyme transition (EMT) that epicardial cells undergo to become sub-epicardial; the invasion of the myocardium; and the differentiation of coronary lineages. However, the factors controlling these processes are not completely understood. Epicardial and coronary vascular precursors migrate to the avascular heart tube during embryogenesis via the proepicardium (PE). Here, we show that in the quail embryo fibroblast growth factor receptor (FGFR)-1 is expressed in a spatially and temporally restricted manner in the PE and epicardium-derived cells, including vascular endothelial precursors, and is up-regulated in epicardial cells after EMT. We used replication-defective retroviral vectors to over-express or knock-down FGFR-1 in the PE. FGFR-1 over-expression resulted in increased epicardial EMT. Knock-down of FGFR-1, however, did not inhibit epicardial EMT but greatly compromised the ability of PE progeny to invade the myocardium. The latter could, however, contribute to endothelia and smooth muscle of sub-epicardial vessels. Correct FGFR-1 levels were also important for correct coronary lineage differentiation with, at E12, an increase in the proportion of endothelial cells amongst FGFR-1 over-expressing PE progeny and a decrease in the proportion of smooth muscle cells in antisense FGFR-1 virus-infected PE progeny. Finally, in a heart explant system, constitutive activation of FGFR-1 signaling in epicardial cells resulted in increased delamination from the epicardium, invasion of the sub-epicardium, and invasion of the myocardium. These data reveal novel roles for FGFR-1 signaling in epicardial biology and coronary vascular lineage differentiation, and point to potential new therapeutic avenues.
Collapse
Affiliation(s)
- David J Pennisi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | | |
Collapse
|
35
|
Fibroblast Growth Factor-2 regulates proliferation of cardiac myocytes in normal and hypoplastic left ventricles in the developing chick. Cardiol Young 2009; 19:159-69. [PMID: 19195417 DOI: 10.1017/s1047951109003552] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The developing heart increases its mass predominantly by increasing the number of contained cells through proliferation. We hypothesized that addition of fibroblast growth factor-2, a factor previously shown to stimulate division of the embryonic myocytes, to the left ventricular myocardium in an experimental model of left heart hypoplasia created in the chicken would attenuate phenotypic severity by increasing cellular proliferation. We have established an effective mode of delivery of fibroblast growth factor-2 to the chick embryonic left ventricular myocardium by using adenovirus vectors, which was more efficient and better tolerated than direct injection of recombinant fibroblast growth factor-2 protein. Injection of control adenovirus expressing green fluorescent protein did not result in significant alterations in myocytic proliferation or cell death compared with intact, uninjected, controls. Co-injection of adenoviruses expressing green fluorescent protein and fibroblast growth factor-2 was used for verification of positive injection, and induction of proliferation, respectively. Treatment of both normal and hypoplastic left ventricles with fibroblast growth factor-2 expressing adenovirus resulted in to 2 to 3-fold overexpression of fibroblast growth factor-2, as verified by immunostaining. An increase by 45% in myocytic proliferation was observed following injection of normal hearts, and an increase of 39% was observed in hypoplastic hearts. There was a significant increase in anti-myosin immunostaining in the hypoplastic, but not the normal hearts. We have shown, therefore, that expression of exogenous fibroblast growth factor-2 in the late embryonic heart can exert direct effects on cardiac myocytes, inducing both their proliferation and differentiation. These data suggest potential for a novel therapeutic option in selected cases of congenital cardiac disease, such as hypoplastic left heart syndrome.
Collapse
|
36
|
Maass A, Kajahn J, Guerleyik E, Guldner NW, Rapoport DH, Kruse C. Towards a pragmatic strategy for regenerating infarcted myocardium with glandular stem cells. Ann Anat 2009; 191:51-61. [DOI: 10.1016/j.aanat.2008.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 08/04/2008] [Accepted: 09/07/2008] [Indexed: 12/11/2022]
|
37
|
Yuasa S, Fukuda K. Recent advances in cardiovascular regenerative medicine: the induced pluripotent stem cell era. Expert Rev Cardiovasc Ther 2008; 6:803-10. [PMID: 18570618 DOI: 10.1586/14779072.6.6.803] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Induced pluripotent stem (iPS) cells have recently been established by transfecting mouse and human fibroblasts with the transcription factors Oct3/4, Sox2, Klf4 and c-Myc, known to be expressed at high levels in embryonic stem (ES) cells. These cells have great potential in regenerative medicine as they have the capacity to differentiate into all three germ layer-derived cells and are syngeneic. The differentiation of ES cells into cardiomyocytes mimics the early processes involved in heart development. Recent studies describe the contribution of various growth factors and corresponding inhibitors to heart development during embryogenesis. Bone morphogenetic proteins, Wnt protein and Notch signals play critical roles in heart development in a context- and time-dependent manner. Consistent with ES cells, the exposure of iPS cells to such growth factors is hypothesized to augment differentiation into cardiomyocytes. The combination of iPS cells and appropriate developmental signal information has the potential for providing the foundations for future regenerative medicine.
Collapse
Affiliation(s)
- Shinsuke Yuasa
- Cardiology Division, Department of Medicine, Keio University School of Medicine, 35-Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | | |
Collapse
|
38
|
Sumi T, Tsuneyoshi N, Nakatsuji N, Suemori H. Defining early lineage specification of human embryonic stem cells by the orchestrated balance of canonical Wnt/beta-catenin, Activin/Nodal and BMP signaling. Development 2008; 135:2969-79. [PMID: 18667462 DOI: 10.1242/dev.021121] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The canonical Wnt/beta-catenin signaling has remarkably diverse roles in embryonic development, stem cell self-renewal and cancer progression. Here, we show that stabilized expression of beta-catenin perturbed human embryonic stem (hES)-cell self-renewal, such that up to 80% of the hES cells developed into the primitive streak (PS)/mesoderm progenitors, reminiscent of early mammalian embryogenesis. The formation of the PS/mesoderm progenitors essentially depended on the cooperative action of beta-catenin together with Activin/Nodal and BMP signaling pathways. Intriguingly, blockade of BMP signaling completely abolished mesoderm generation, and induced a cell fate change towards the anterior PS progenitors. The PI3-kinase/Akt, but not MAPK, signaling pathway had a crucial role in the anterior PS specification, at least in part, by enhancing beta-catenin stability. In addition, Activin/Nodal and Wnt/beta-catenin signaling synergistically induced the generation and specification of the anterior PS/endoderm. Taken together, our findings clearly demonstrate that the orchestrated balance of Activin/Nodal and BMP signaling defines the cell fate of the nascent PS induced by canonical Wnt/beta-catenin signaling in hES cells.
Collapse
Affiliation(s)
- Tomoyuki Sumi
- Laboratory of Embryonic Stem Cell Research, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, Shogoin, Kyoto 606-8507, Japan
| | | | | | | |
Collapse
|
39
|
Ream M, Ray AM, Chandra R, Chikaraishi DM. Early fetal hypoxia leads to growth restriction and myocardial thinning. Am J Physiol Regul Integr Comp Physiol 2008; 295:R583-95. [PMID: 18509101 DOI: 10.1152/ajpregu.00771.2007] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hypoxia is necessary for fetal development; however, excess hypoxia is detrimental. Hypoxia has been extensively studied in the near-term fetus, but less is known about earlier fetal effects. The purpose of this study was to determine the window of vulnerability to severe hypoxia, what organ system(s) is most sensitive, and why hypoxic fetuses die. We induced hypoxia by reducing maternal-inspired O2 from 21% to 8%, which decreased fetal tissue oxygenation assessed by pimonidazole binding. The mouse fetus was most vulnerable in midgestation: 24 h of hypoxia killed 89% of embryonic day 13.5 (E13.5) fetuses, but only 5% of E11.5 and 51% of E17.5 fetuses. Sublethal hypoxia at E12.5 caused growth restriction, reducing fetal weight by 26% and protein by 45%. Hypoxia induced HIF-1 target genes, including vascular endothelial growth factor (Vegf), erythropoietin, glucose transporter-1 and insulin-like growth factor binding protein-1 (Igfbp-1), which has been implicated in human intrauterine growth restriction (IUGR). Hypoxia severely compromised the cardiovascular system. Signs of heart failure, including loss of yolk sac circulation, hemorrhage, and edema, were caused by 18-24 h of hypoxia. Hypoxia induced ventricular dilation and myocardial hypoplasia, decreasing ventricular tissue by 50% and proliferation by 21% in vivo and by 40% in isolated cultured hearts. Epicardial detachment was the first sign of hypoxic damage in the heart, although expression of epicardially derived mitogens, such as FGF2, FGF9, and Wnt9b was not reduced. We propose that hypoxia compromises the fetus through myocardial hypoplasia and reduced heart rate.
Collapse
Affiliation(s)
- Margie Ream
- Department of Neurobiology, Box 3209, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
40
|
Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, Henckaerts E, Bonham K, Abbott GW, Linden RM, Field LJ, Keller GM. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 2008; 453:524-8. [PMID: 18432194 DOI: 10.1038/nature06894] [Citation(s) in RCA: 1053] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Accepted: 02/22/2008] [Indexed: 12/15/2022]
Abstract
The functional heart is comprised of distinct mesoderm-derived lineages including cardiomyocytes, endothelial cells and vascular smooth muscle cells. Studies in the mouse embryo and the mouse embryonic stem cell differentiation model have provided evidence indicating that these three lineages develop from a common Flk-1(+) (kinase insert domain protein receptor, also known as Kdr) cardiovascular progenitor that represents one of the earliest stages in mesoderm specification to the cardiovascular lineages. To determine whether a comparable progenitor is present during human cardiogenesis, we analysed the development of the cardiovascular lineages in human embryonic stem cell differentiation cultures. Here we show that after induction with combinations of activin A, bone morphogenetic protein 4 (BMP4), basic fibroblast growth factor (bFGF, also known as FGF2), vascular endothelial growth factor (VEGF, also known as VEGFA) and dickkopf homolog 1 (DKK1) in serum-free media, human embryonic-stem-cell-derived embryoid bodies generate a KDR(low)/C-KIT(CD117)(neg) population that displays cardiac, endothelial and vascular smooth muscle potential in vitro and, after transplantation, in vivo. When plated in monolayer cultures, these KDR(low)/C-KIT(neg) cells differentiate to generate populations consisting of greater than 50% contracting cardiomyocytes. Populations derived from the KDR(low)/C-KIT(neg) fraction give rise to colonies that contain all three lineages when plated in methylcellulose cultures. Results from limiting dilution studies and cell-mixing experiments support the interpretation that these colonies are clones, indicating that they develop from a cardiovascular colony-forming cell. Together, these findings identify a human cardiovascular progenitor that defines one of the earliest stages of human cardiac development.
Collapse
Affiliation(s)
- Lei Yang
- Department of Gene and Cell Medicine, The Black Family Stem Cell Institute, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Barthelery M, Salli U, Vrana KE. Nuclear proteomics and directed differentiation of embryonic stem cells. Stem Cells Dev 2008; 16:905-19. [PMID: 17999636 DOI: 10.1089/scd.2007.0071] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
During the past decade, regenerative medicine has been the subject of intense interest due, in large part, to our growing knowledge of embryonic stem (ES) cell biology. ES cells give rise to cell lineages from the three primordial germ layers--endoderm, mesoderm, and ectoderm. This process needs to be channeled if these cells are to be differentiated efficiently and used subsequently for therapeutic purposes. Indeed, an important area of investigation involves directed differentiation to influence the lineage commitment of these pluripotent cells in vitro. Various strategies involving timely growth factor supplementation, cell co-cultures, and gene transfection are used to drive lineage specific emergence. The underlying goal is to control directly the center of gene expression and cellular programming--the nucleus. Gene expression is enabled, managed, and sustained by the collective actions and interactions of proteins found in the nucleus--the nuclear proteome--in response to extracellular signaling. Nuclear proteomics can inventory these nuclear proteins in differentiating cells and decipher their dynamics during cellular phenotypic commitment. This review details what is currently known about nuclear effectors of stem cell differentiation and describes emerging techniques in the discovery of nuclear proteomics that will illuminate new transcription factors and modulators of gene expression.
Collapse
Affiliation(s)
- Miguel Barthelery
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | |
Collapse
|
42
|
Montano MM, Doughman YQ, Deng H, Chaplin L, Yang J, Wang N, Zhou Q, Ward NL, Watanabe M. Mutation of the HEXIM1 gene results in defects during heart and vascular development partly through downregulation of vascular endothelial growth factor. Circ Res 2007; 102:415-22. [PMID: 18079413 DOI: 10.1161/circresaha.107.157859] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our previous studies and those of others indicated that the transcription factor Hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) is a tumor suppressor and cyclin-dependent kinase inhibitor, and that these HEXIM1 functions are mainly dependent on its C-terminal region. We provide evidence here that the HEXIM1 C-terminal region is critical for cardiovascular development. HEXIM1 protein was detected in the heart during critical time periods in cardiac growth and chamber maturation. We created mice carrying an insertional mutation in the HEXIM1 gene that disrupted its C-terminal region and found that this resulted in prenatal lethality. Heart defects in HEXIM1(1 to 312) mice included abnormal coronary patterning and thin ventricular walls. The thin myocardium can be partly attributed to increased apoptosis. Platelet endothelial cell adhesion molecular precursor-1 staining of HEXIM1(1 to 312) heart sections revealed decreased vascularization of the myocardium despite the presence of coronary vasculature in the epicardium. The expression of vascular endothelial growth factor (VEGF), known to affect angioblast invasion and myocardial proliferation and survival, was decreased in HEXIM1(1 to 312) mice compared with control littermates. We also observed decreased fibroblast growth factor 9 (FGF9) expression, suggesting that effects of HEXIM1 in the myocardium are partly mediated through epicardial FGF9 signaling. Together our results suggest that HEXIM1 plays critical roles in coronary vessel development and myocardial growth. The basis for this role of HEXIM1 is that VEGF is a direct transcriptional target of HEXIM1, and involves attenuation a repressive effects of C/EBPalpha on VEGF gene transcription.
Collapse
Affiliation(s)
- Monica M Montano
- Department of Pharmacology, Case Western Reserve University School of Medicine, H.G. Wood Bldg W307, 2109 Adelbert Rd, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Améen C, Strehl R, Björquist P, Lindahl A, Hyllner J, Sartipy P. Human embryonic stem cells: current technologies and emerging industrial applications. Crit Rev Oncol Hematol 2007; 65:54-80. [PMID: 17689256 DOI: 10.1016/j.critrevonc.2007.06.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 06/11/2007] [Accepted: 06/27/2007] [Indexed: 12/28/2022] Open
Abstract
The efficiency and accuracy of the drug development process is severely restricted by the lack of functional human cell systems. However, the successful derivation of pluripotent human embryonic stem (hES) cell lines in the late 1990s is expected to revolutionize biomedical research in many areas. Due to their growth capacity and unique developmental potential to differentiate into almost any cell type of the human body, hES cells have opened novel avenues both in basic and applied research as well as for therapeutic applications. In this review we describe, from an industrial perspective, the basic science that underlies the hES cell technology and discuss the current and future prospects for hES cells in novel and improved stem cell based applications for drug discovery, toxicity testing as well as regenerative medicine.
Collapse
Affiliation(s)
- Caroline Améen
- Cellartis AB, Arvid Wallgrens Backe 20, 413 46 Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Despite the critical importance of proper cell cycle regulation in establishing the correct morphology of organs and tissues during development, relatively little is known about how cell proliferation is regulated in a tissue-specific manner. The control of cell proliferation within the developing heart is of considerable interest, given the high prevalence of congenital cardiac abnormalities among humans, and recent interest in the isolation of cardiac progenitor populations. We therefore review studies exploring the contribution of cell proliferation to overall cardiac morphology and the molecular mechanisms regulating this process. In addition, we also review recent studies that have identified progenitor cell populations within the adult myocardium, as well as those exploring the capability of differentiated myocardial cells to proliferate post-natally. Thus, the exploration of cardiomyocyte cell cycle regulation, both during development as well as in the adult heart, promises to yield many exciting and important discoveries over the coming years.
Collapse
Affiliation(s)
- Sarah C. Goetz
- Carolina Cardiovascular Biology Center; University of North Carolina at Chapel Hill; Chapel Hill, North Carolina USA
- Department of Biology; University of North Carolina at Chapel Hill; Chapel Hill, North Carolina USA
| | - Frank L. Conlon
- Carolina Cardiovascular Biology Center; University of North Carolina at Chapel Hill; Chapel Hill, North Carolina USA
- Department of Biology; University of North Carolina at Chapel Hill; Chapel Hill, North Carolina USA
- Department of Genetics Fordham Hall; University of North Carolina at Chapel Hill; Chapel Hill, North Carolina USA
- Correspondence to: Frank L. Conlon; Department of Genetics; University of North Carolina at Chapel Hill; 221 Fordham Hall, CB 3280; Chapel Hill, North Carolina 27599 USA; Tel.: 919.843.5500;
| |
Collapse
|
45
|
Ahuja P, Sdek P, Maclellan WR. Cardiac myocyte cell cycle control in development, disease, and regeneration. Physiol Rev 2007; 87:521-44. [PMID: 17429040 PMCID: PMC2708177 DOI: 10.1152/physrev.00032.2006] [Citation(s) in RCA: 418] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiac myocytes rapidly proliferate during fetal life but exit the cell cycle soon after birth in mammals. Although the extent to which adult cardiac myocytes are capable of cell cycle reentry is controversial and species-specific differences may exist, it appears that for the vast majority of adult cardiac myocytes the predominant form of growth postnatally is an increase in cell size (hypertrophy) not number. Unfortunately, this limits the ability of the heart to restore function after any significant injury. Interest in novel regenerative therapies has led to the accumulation of much information on the mechanisms that regulate the rapid proliferation of cardiac myocytes in utero, their cell cycle exit in the perinatal period, and the permanent arrest (terminal differentiation) in adult myocytes. The recent identification of cardiac progenitor cells capable of giving rise to cardiac myocyte-like cells has challenged the dogma that the heart is a terminally differentiated organ and opened new prospects for cardiac regeneration. In this review, we summarize the current understanding of cardiomyocyte cell cycle control in normal development and disease. In addition, we also discuss the potential usefulness of cardiomyocyte self-renewal as well as feasibility of therapeutic manipulation of the cardiac myocyte cell cycle for cardiac regeneration.
Collapse
Affiliation(s)
| | | | - W. Robb Maclellan
- Corresponding author: W. Robb MacLellan, Cardiovascular Research Laboratories, David Geffen school of Medicine at UCLA, 675 C.E. Young Dr., MRL 3-645, Los Angeles, California, 90095-1760; Phone: (310) 825-2556; Fax: (310) 206-5777; e-mail:
| |
Collapse
|
46
|
Cardiac Development: Toward a Molecular Basis for Congenital Heart Disease. CARDIOVASCULAR MEDICINE 2007. [DOI: 10.1007/978-1-84628-715-2_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
47
|
Abstract
The realization of regenerative cardiac medicine depends on the availability of cardiomyocytes in sufficient numbers for transplantation of cardiac tissue and the accompanying blood vessels. Embryonic stem (ES) cells, bone marrow (BM) stem cells, and tissue-derived stem cells are all potential cell sources. Although ES cells are highly proliferative and suitable for mass production, an efficient protocol is yet to be established to ensure selective cardiomyocyte induction using these cells. Recent advances in developmental biology have clarified the involvement of critical factors in cardiomyocyte differentiation, including bone morphogenic protein and Wnt signaling proteins, and such factors have the potential to improve the efficiency of stem cell induction. Initial studies of the intracoronary administration of BM mononuclear cells after myocardial infarction has yielded promising results; however, intensive investigation of the underlying molecular mechanisms at play as well as double-blinded clinical trials will be necessary to establish the extent of both migration of the BM stem cells into the damaged cardiac tissue and their differentiation into cardiomyocytes. Several types of cardiac tissue stem cells have also been reported, but an accurate and extensive comparison of these cells with regard to their characteristics and multipotency remains to be done. An integrative study involving developmental biology, stem cell biology, and tissue engineering is required to achieve the full potential of cardiac regeneration.
Collapse
Affiliation(s)
- Keiichi Fukuda
- Department of Regenerative Medicine and Advanced Cardiac Therapeutics, Keio University School of Medicine, Tokyo, Japan.
| | | |
Collapse
|
48
|
Amann K, Faulhaber J, Campean V, Balajew V, Dono R, Mall G, Ehmke H. Impaired myocardial capillarogenesis and increased adaptive capillary growth in FGF2-deficient mice. J Transl Med 2006; 86:45-53. [PMID: 16258522 DOI: 10.1038/labinvest.3700359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Basic fibroblast growth factor (FGF2) plays a major role in angiogenesis and capillary growth. In contrast to vascular endothelial growth factor, which is required for proliferation and survival of endothelial cells, FGF2 does not seem to be essential since the Fgf2 knockout is not lethal. Therefore, the precise genetic and physiological roles of FGF2 for capillary development and adaptation remain to be determined. Here we show that myocardial capillary supply is normal at birth, but significantly reduced by approximately 25% in adult Fgf2+/- and Fgf2-/- mice as compared with wild-type littermates. In contrast, after induction of myocardial hypertrophy by continuous infusion of angiotensin II (ANG II) for 6 days marked capillary growth was seen in both Fgf2+/- and Fgf2-/- mice, but not in wild-type littermates. These data demonstrate that two intact Fgf2 genes are necessary for normal capillary development after birth, whereas FGF2 seems to be dispensable for adaptive myocardial capillary growth in the adult mouse.
Collapse
Affiliation(s)
- Kerstin Amann
- Department of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The chick embryo is a versatile model system, in which classical embryology can be combined with modern molecular approaches. In the last two decades, several efficient methods have been developed to introduce exogenous genes into the chick embryo. These techniques allow alteration of gene expression levels in a spatially and temporally restricted manner, thereby circumventing embryonic lethality and/or eliminating secondary effects in other tissues. Here, we present the current status of avian somatic transgenic techniques, focusing on electroporation and retrovirus-mediated gene transfer. Electroporation allows quick and efficient gain-of-function studies based on transient misexpression of genes. Retroviral vectors, which are capable of integrating exogenous genes into the host chromosome, permit analysis of long-term effects of gene misexpression. The variety of methods available for somatic transgenesis, along with the recent completion of the chicken genome, are transforming the chick embryo into one of the most attractive model systems to examine function of genes that are important for embryonic development.
Collapse
Affiliation(s)
- Yasuo Ishii
- Department of Cell and Developmental Biology, Cornell University Medical College, New York, New York 10021, USA
| | | |
Collapse
|
50
|
Yuasa S, Itabashi Y, Koshimizu U, Tanaka T, Sugimura K, Kinoshita M, Hattori F, Fukami SI, Shimazaki T, Ogawa S, Okano H, Fukuda K. Transient inhibition of BMP signaling by Noggin induces cardiomyocyte differentiation of mouse embryonic stem cells. Nat Biotechnol 2005; 23:607-11. [PMID: 15867910 DOI: 10.1038/nbt1093] [Citation(s) in RCA: 266] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 03/30/2005] [Indexed: 11/08/2022]
Abstract
Embryonic stem (ES) cells are a promising source of cardiomyocytes, but clinical application of ES cells has been hindered by the lack of reliable selective differentiation methods. Differentiation into any lineage is partly dependent on the regulatory mechanisms of normal early development. Although several signals, including bone morphogenetic protein (BMP), Wnt and FGF, are involved in heart development, scarce evidence is available about the exact signals that mediate cardiomyocyte differentiation. While investigating the involvement of BMP signaling in early heart formation in the mouse, we found that the BMP antagonist Noggin is transiently but strongly expressed in the heart-forming region during gastrulation and acts at the level of induction of mesendoderm to establish conditions conducive to cardiogenesis. We applied this finding to develop an effective protocol for obtaining cardiomyocytes from mouse ES cells by inhibition of BMP signaling.
Collapse
|