1
|
Paredes-Espinosa MB, Paluh JL. Synthetic embryology of the human heart. Front Cell Dev Biol 2025; 12:1478549. [PMID: 39935786 PMCID: PMC11810959 DOI: 10.3389/fcell.2024.1478549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
The evolution of stem cell-based heart models from cells and tissues to organoids and assembloids and recently synthetic embryology gastruloids, is poised to revolutionize our understanding of cardiac development, congenital to adult diseases, and patient customized therapies. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have already been integrated into transplantable patches and are in preclinical efforts to reverse fibrotic scarring from myocardial infarctions. To inform on the complexity of heart diseases, multi-tissue morphogenic heart models are needed that replicate fundamental components of heart function to heart organogenesis in vitro and which require a deep understanding of heart development. Organoid and assembloid models capture selected multicellular cardiac processes, such as chamber formation and priming events for vascularization. Gastruloid heart models offer deeper insights as synthetic embryology to mimic multi-staged developmental events of in vivo heart organogenesis including established heart fields, crescent formation and heart tube development along with vascular systemic foundation and even further steps. The human Elongating Multi-Lineage Organized Cardiac (EMLOC) gastruloid model captures these stages and additional events including chamber genesis, patterned vascularization, and extrinsic central and intrinsic cardiac nervous system (CNS-ICNS) integration guided by spatiotemporal and morphogenic processes with neural crest cells. Gastruloid synthetic embryology heart models offer new insights into previously hidden processes of development and provide powerful platforms for addressing heart disease that extends beyond cardiomyocytes, such as arrhythmogenic diseases, congenital defects, and systemic injury interactions, as in spinal cord injuries. The holistic view that is emerging will reveal heart development and disease in unprecedented detail to drive transformative state-of-the-art innovative applications for heart health.
Collapse
Affiliation(s)
- Maria Belen Paredes-Espinosa
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science and Engineering, University at Albany, Albany, NY, United States
| | - Janet L Paluh
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science and Engineering, University at Albany, Albany, NY, United States
| |
Collapse
|
2
|
Prill K, Windsor Reid P, Pilgrim D. Heart Morphogenesis Requires Smyd1b for Proper Incorporation of the Second Heart Field in Zebrafish. Genes (Basel) 2025; 16:52. [PMID: 39858599 PMCID: PMC11764850 DOI: 10.3390/genes16010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Abnormal development of the second heart field significantly contributes to congenital heart defects, often caused by disruptions in tightly regulated molecular pathways. Smyd1, a gene encoding a protein with SET and MYND domains, is essential for heart and skeletal muscle development. Mutations in SMYD1 result in severe cardiac malformations and misregulation of Hand2 expression in mammals. This study examines the role of Smyd1b in zebrafish cardiac morphogenesis to elucidate its function and the mechanisms underlying congenital heart defects. Methods: Smyd1b (still heart) mutant embryos were analyzed for cardiac defects, and changes in gene expression related to heart development using live imaging, in situ hybridization, quantitative PCR and immunofluorescent comparisons and analysis. Results: Smyd1b mutants displayed severe cardiac defects, including failure to loop, severe edema, and an expansion of cardiac jelly linked to increased has2 expression. Additionally, the expression of key cardiac transcription factors, such as gata4, gata5, and nkx2.5, was notably reduced, indicating disrupted transcriptional regulation. The migration of cardiac progenitors was impaired and the absence of Islet-1-positive cells in the mutant hearts suggests a failed contribution of SHF progenitor cells. Conclusions: These findings underscore the essential role of Smyd1b in regulating cardiac morphogenesis and the development of the second heart field. This study highlights the potential of Smyd1b as a key factor in understanding the genetic and molecular mechanisms underlying congenital heart defects and cardiac development.
Collapse
Affiliation(s)
- Kendal Prill
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; (K.P.); (P.W.R.)
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 1Y2, Canada
| | - Pamela Windsor Reid
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; (K.P.); (P.W.R.)
- Department of Biological Science, MacEwan University, Edmonton, AB T5J 4S2, Canada
| | - Dave Pilgrim
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; (K.P.); (P.W.R.)
| |
Collapse
|
3
|
Holman AR, Tran S, Destici E, Farah EN, Li T, Nelson AC, Engler AJ, Chi NC. Single-cell multi-modal integrative analyses highlight functional dynamic gene regulatory networks directing human cardiac development. CELL GENOMICS 2024; 4:100680. [PMID: 39437788 PMCID: PMC11605693 DOI: 10.1016/j.xgen.2024.100680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Illuminating the precise stepwise genetic programs directing cardiac development provides insights into the mechanisms of congenital heart disease and strategies for cardiac regenerative therapies. Here, we integrate in vitro and in vivo human single-cell multi-omic studies with high-throughput functional genomic screening to reveal dynamic, cardiac-specific gene regulatory networks (GRNs) and transcriptional regulators during human cardiomyocyte development. Interrogating developmental trajectories reconstructed from single-cell data unexpectedly reveal divergent cardiomyocyte lineages with distinct gene programs based on developmental signaling pathways. High-throughput functional genomic screens identify key transcription factors from inferred GRNs that are functionally relevant for cardiomyocyte lineages derived from each pathway. Notably, we discover a critical heat shock transcription factor 1 (HSF1)-mediated cardiometabolic GRN controlling cardiac mitochondrial/metabolic function and cell survival, also observed in fetal human cardiomyocytes. Overall, these multi-modal genomic studies enable the systematic discovery and validation of coordinated GRNs and transcriptional regulators controlling the development of distinct human cardiomyocyte populations.
Collapse
Affiliation(s)
- Alyssa R Holman
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shaina Tran
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eugin Destici
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elie N Farah
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ting Li
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Aileena C Nelson
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, USA
| | - Neil C Chi
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
4
|
Sam J, Torregroza I, Evans T. Gata6 functions in zebrafish endoderm to regulate late differentiating arterial pole cardiogenesis. Development 2024; 151:dev202895. [PMID: 39133135 PMCID: PMC11423812 DOI: 10.1242/dev.202895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Mutations in GATA6 are associated with congenital heart disease, most notably conotruncal structural defects. However, how GATA6 regulates cardiac morphology during embryogenesis is undefined. We used knockout and conditional mutant zebrafish alleles to investigate the spatiotemporal role of gata6 during cardiogenesis. Loss of gata6 specifically impacts atrioventricular valve formation and recruitment of epicardium, with a prominent loss of arterial pole cardiac cells, including those of the ventricle and outflow tract. However, there are no obvious defects in cardiac progenitor cell specification, proliferation or death. Conditional loss of gata6 starting at 24 h is sufficient to disrupt the addition of late differentiating cardiomyocytes at the arterial pole, with decreased expression levels of anterior secondary heart field (SHF) markers spry4 and mef2cb. Conditional loss of gata6 in the endoderm is sufficient to phenocopy the straight knockout, resulting in a significant loss of ventricular and outflow tract tissue. Exposure to a Dusp6 inhibitor largely rescues the loss of ventricular cells in gata6-/- larvae. Thus, gata6 functions in endoderm are mediated by FGF signaling to regulate the addition of anterior SHF progenitor derivatives during heart formation.
Collapse
Affiliation(s)
- Jessica Sam
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Ingrid Torregroza
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
- Hartman Institute for Therapeutic Organ Regeneration, Weill Cornell Medicine, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
5
|
Mensah IK, Gowher H. Signaling Pathways Governing Cardiomyocyte Differentiation. Genes (Basel) 2024; 15:798. [PMID: 38927734 PMCID: PMC11202427 DOI: 10.3390/genes15060798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiomyocytes are the largest cell type that make up the heart and confer beating activity to the heart. The proper differentiation of cardiomyocytes relies on the efficient transmission and perception of differentiation cues from several signaling pathways that influence cardiomyocyte-specific gene expression programs. Signaling pathways also mediate intercellular communications to promote proper cardiomyocyte differentiation. We have reviewed the major signaling pathways involved in cardiomyocyte differentiation, including the BMP, Notch, sonic hedgehog, Hippo, and Wnt signaling pathways. Additionally, we highlight the differences between different cardiomyocyte cell lines and the use of these signaling pathways in the differentiation of cardiomyocytes from stem cells. Finally, we conclude by discussing open questions and current gaps in knowledge about the in vitro differentiation of cardiomyocytes and propose new avenues of research to fill those gaps.
Collapse
Affiliation(s)
| | - Humaira Gowher
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
6
|
Liang J, Ul Hassan I, Yee Cheung M, Feng L, Lin YJ, Long Q, Wang C, Ding Y, Wang Z, Zhang Y, Li Y, Guo D, Guo X, Chi Bun Wong T, Kaleem Samma M, Rong Z, Qi X, Cai D, Ngai SM, Zhao H. Mechanistic study of transcription factor Sox18 during heart development. Gen Comp Endocrinol 2024; 350:114472. [PMID: 38373462 DOI: 10.1016/j.ygcen.2024.114472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/18/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Heart development is a delicate and complex process regulated by coordination of various signaling pathways. In this study, we investigated the role of sox18 in heart development by modulating Wnt/β-Catenin signaling pathways. Our spatiotemporal expression analysis revealed that sox18 is mainly expressed in the heart, branchial arch, pharyngeal arch, spinal cord, and intersegmental vessels at the tailbud stage of Xenopus tropicalis embryo. Overexpression of sox18 in the X. tropicalis embryos causes heart edema, while loss-of-function of sox18 can change the signal of developmental heart marker gata4 at different stages, suggesting that sox18 plays an essential role in the development of the heart. Knockdown of SOX18 in human umbilical vein endothelial cells suggests a link between Sox18 and β-CATENIN, a key regulator of the Wnt signaling pathway. Sox18 negatively regulates islet1 and tbx3, the downstream factors of Wnt/β-Catenin signaling, during the linear heart tube formation and the heart looping stage. Taken together, our findings highlight the crucial role of Sox18 in the development of the heart via inhibiting Wnt/β-Catenin signaling.
Collapse
Affiliation(s)
- Jianxin Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Imtiaz Ul Hassan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Man Yee Cheung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lei Feng
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China; Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yi-Jyun Lin
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qi Long
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chengdong Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuyue Ding
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ziqing Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuan Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yulong Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Donghao Guo
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaofang Guo
- School of Life Sciences, Jinan University, Guangzhou, China
| | - Thomas Chi Bun Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Muhammad Kaleem Samma
- Department of Biology and Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Zixin Rong
- Department of Gene Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - Xufeng Qi
- School of Life Sciences, Jinan University, Guangzhou, China
| | - Dongqing Cai
- School of Life Sciences, Jinan University, Guangzhou, China
| | - Sai-Ming Ngai
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Zhao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
7
|
Cai S, Dai Q. Progress in preclinical research on induced pluripotent stem cell therapy for acute myocardial infarction. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:244-253. [PMID: 38594961 PMCID: PMC11057988 DOI: 10.3724/zdxbyxb-2023-0402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
Induced pluripotent stem cells (iPSCs) are obtained by introducing exogenous genes or adding chemicals to the culture medium to induce somatic cell differentiation. Similarly to embryonic stem cells, iPSCs have the ability to differentiate into all three embryonic cell lines. iPSCs can differentiate into cardiac muscle cells through two-dimensional differentiation methods such as monolayer cell culture and co-culture, or through embryoid body and scaffold-based three-dimensional differentiation methods. In addition, the process of iPSCs differentiation into cardiac muscle cells also requires activation or inhibition of specific signaling pathways,such as Wnt, BMP, Notch signaling pathways to mimic the development of the heart in vivo. In recent years, suspension culturing in bioreactors has been shown to produce large number of iPSCs derived cardiac muscle cells (iPSC-CMs). Before transplantation, it is necessary to purify iPSC-CMs through metabolic regulation or cell sorting to eliminate undifferentiated iPSCs, which may lead to teratoma formation. The transplantation methods for iPSC-CMs are mainly injection of cell suspension and transplantation of cell patches into the infarcted myocardium. Animal studies have shown that transplantation of iPSC-CMs into the infarcted myocardium can improve cardiac function. This article reviews the progress in preclinical studies on iPSC-CMs therapy for acute myocardial infarction and discusses the limitations and challenges of its clinical application to provide references for further clinical research and application.
Collapse
Affiliation(s)
- Songyan Cai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Qingyuan Dai
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
8
|
Chi C, Roland TJ, Song K. Differentiation of Pluripotent Stem Cells for Disease Modeling: Learning from Heart Development. Pharmaceuticals (Basel) 2024; 17:337. [PMID: 38543122 PMCID: PMC10975450 DOI: 10.3390/ph17030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
Heart disease is a pressing public health problem and the leading cause of death worldwide. The heart is the first organ to gain function during embryogenesis in mammals. Heart development involves cell determination, expansion, migration, and crosstalk, which are orchestrated by numerous signaling pathways, such as the Wnt, TGF-β, IGF, and Retinoic acid signaling pathways. Human-induced pluripotent stem cell-based platforms are emerging as promising approaches for modeling heart disease in vitro. Understanding the signaling pathways that are essential for cardiac development has shed light on the molecular mechanisms of congenital heart defects and postnatal heart diseases, significantly advancing stem cell-based platforms to model heart diseases. This review summarizes signaling pathways that are crucial for heart development and discusses how these findings improve the strategies for modeling human heart disease in vitro.
Collapse
Affiliation(s)
- Congwu Chi
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Truman J. Roland
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Kunhua Song
- Heart Institute, University of South Florida, Tampa, FL 33602, USA; (C.C.); (T.J.R.)
- Department of Internal Medicine, University of South Florida, Tampa, FL 33602, USA
- Center for Regenerative Medicine, University of South Florida, Tampa, FL 33602, USA
| |
Collapse
|
9
|
Nakanishi-Koakutsu M, Miki K, Naka Y, Sasaki M, Wakimizu T, Napier SC, Okubo C, Narita M, Nishikawa M, Hata R, Chonabayashi K, Hotta A, Imahashi K, Nishimoto T, Yoshida Y. CD151 expression marks atrial- and ventricular- differentiation from human induced pluripotent stem cells. Commun Biol 2024; 7:231. [PMID: 38418926 PMCID: PMC10901864 DOI: 10.1038/s42003-024-05809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
Current differentiation protocols for human induced pluripotent stem cells (hiPSCs) produce heterogeneous cardiomyocytes (CMs). Although chamber-specific CM selection using cell surface antigens enhances biomedical applications, a cell surface marker that accurately distinguishes between hiPSC-derived atrial CMs (ACMs) and ventricular CMs (VCMs) has not yet been identified. We have developed an approach for obtaining functional hiPSC-ACMs and -VCMs based on CD151 expression. For ACM differentiation, we found that ACMs are enriched in the CD151low population and that CD151 expression is correlated with the expression of Notch4 and its ligands. Furthermore, Notch signaling inhibition followed by selecting the CD151low population during atrial differentiation leads to the highly efficient generation of ACMs as evidenced by gene expression and electrophysiology. In contrast, for VCM differentiation, VCMs exhibiting a ventricular-related gene signature and uniform action potentials are enriched in the CD151high population. Our findings enable the production of high-quality ACMs and VCMs appropriate for hiPSC-derived chamber-specific disease models and other applications.
Collapse
Affiliation(s)
- Misato Nakanishi-Koakutsu
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Kenji Miki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
- Center for Organ Engineering, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02114, USA.
- Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita, 565-0871, Japan.
| | - Yuki Naka
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Masako Sasaki
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Takayuki Wakimizu
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Stephanie C Napier
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Global Advanced Platform, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan
| | - Chikako Okubo
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Megumi Narita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Misato Nishikawa
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Reo Hata
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Kazuhisa Chonabayashi
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
| | - Kenichi Imahashi
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Global Advanced Platform, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Japan
| | - Tomoyuki Nishimoto
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan
- Orizuru Therapeutics Incorporated, Fujisawa, 251-8555, Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
- Takeda-CiRA Joint program (T-CiRA), Fujisawa, 251-8555, Japan.
| |
Collapse
|
10
|
Devendran A, Liu C. Subtype and Lineage-Mediated Protocol for Standardizing Activin/Nodal and BMP Signaling for hiPSC-Derived Cardiomyocyte Differentiation. Methods Mol Biol 2024; 2803:13-33. [PMID: 38676882 DOI: 10.1007/978-1-0716-3846-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
The adept and systematic differentiation of embryonic stem cells (ESCs) and human-induced pluripotent stem cells (hiPSCs) to diverse lineage-prone cell types involves crucial step-by-step process that mimics the vital strategic commitment phase that is usually observed during the process of embryo development. The development of precise tissue-specific cell types from these stem cells indeed plays an important role in the advancement of imminent stem cell-based therapeutic strategies. Therefore, the usage of hiPSC-derived cell types for subsequent cardiovascular disease modeling, drug screening, and therapeutic drug development undeniably entails an in-depth understanding of each and every step to proficiently stimulate these stem cells into desired cardiomyogenic lineage. Thus, to accomplish this definitive and decisive fate, it is essential to efficiently induce the mesoderm or pre-cardiac mesoderm, succeeded by the division of cells into cardiovascular and ultimately ensuing with the cardiomyogenic lineage outcome. This usually commences from the earliest phases of pluripotent cell induction. In this chapter, we discuss our robust and reproducible step-wise protocol that will describe the subtype controlled, precise lineage targeted standardization of activin/nodal, and BMP signaling molecules/cytokines, for the efficient differentiation of ventricular cardiomyocytes from hiPSCs via the embryoid body method. In addition, we also describe techniques to dissociate hiPSCs, hiPSC-derived early cardiomyocytes for mesoderm and pre-cardiac mesoderm assessment, and hiPSC-derived cardiomyocytes for early and mature markers assessment.
Collapse
Affiliation(s)
- Anichavezhi Devendran
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clifford Liu
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Maas RGC, van den Dolder FW, Yuan Q, van der Velden J, Wu SM, Sluijter JPG, Buikema JW. Harnessing developmental cues for cardiomyocyte production. Development 2023; 150:dev201483. [PMID: 37560977 PMCID: PMC10445742 DOI: 10.1242/dev.201483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Developmental research has attempted to untangle the exact signals that control heart growth and size, with knockout studies in mice identifying pivotal roles for Wnt and Hippo signaling during embryonic and fetal heart growth. Despite this improved understanding, no clinically relevant therapies are yet available to compensate for the loss of functional adult myocardium and the absence of mature cardiomyocyte renewal that underlies cardiomyopathies of multiple origins. It remains of great interest to understand which mechanisms are responsible for the decline in proliferation in adult hearts and to elucidate new strategies for the stimulation of cardiac regeneration. Multiple signaling pathways have been identified that regulate the proliferation of cardiomyocytes in the embryonic heart and appear to be upregulated in postnatal injured hearts. In this Review, we highlight the interaction of signaling pathways in heart development and discuss how this knowledge has been translated into current technologies for cardiomyocyte production.
Collapse
Affiliation(s)
- Renee G. C. Maas
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
| | - Floor W. van den Dolder
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Qianliang Yuan
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Sean M. Wu
- Department of Medicine, Division of Cardiovascular Medicine,Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joost P. G. Sluijter
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
| | - Jan W. Buikema
- Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3508 GA Utrecht, the Netherlands
- Amsterdam Cardiovascular Sciences, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
- Department of Cardiology, Amsterdam Heart Center, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
12
|
Kocere A, Lalonde RL, Mosimann C, Burger A. Lateral thinking in syndromic congenital cardiovascular disease. Dis Model Mech 2023; 16:dmm049735. [PMID: 37125615 PMCID: PMC10184679 DOI: 10.1242/dmm.049735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Syndromic birth defects are rare diseases that can present with seemingly pleiotropic comorbidities. Prime examples are rare congenital heart and cardiovascular anomalies that can be accompanied by forelimb defects, kidney disorders and more. Whether such multi-organ defects share a developmental link remains a key question with relevance to the diagnosis, therapeutic intervention and long-term care of affected patients. The heart, endothelial and blood lineages develop together from the lateral plate mesoderm (LPM), which also harbors the progenitor cells for limb connective tissue, kidneys, mesothelia and smooth muscle. This developmental plasticity of the LPM, which founds on multi-lineage progenitor cells and shared transcription factor expression across different descendant lineages, has the potential to explain the seemingly disparate syndromic defects in rare congenital diseases. Combining patient genome-sequencing data with model organism studies has already provided a wealth of insights into complex LPM-associated birth defects, such as heart-hand syndromes. Here, we summarize developmental and known disease-causing mechanisms in early LPM patterning, address how defects in these processes drive multi-organ comorbidities, and outline how several cardiovascular and hematopoietic birth defects with complex comorbidities may be LPM-associated diseases. We also discuss strategies to integrate patient sequencing, data-aggregating resources and model organism studies to mechanistically decode congenital defects, including potentially LPM-associated orphan diseases. Eventually, linking complex congenital phenotypes to a common LPM origin provides a framework to discover developmental mechanisms and to anticipate comorbidities in congenital diseases affecting the cardiovascular system and beyond.
Collapse
Affiliation(s)
- Agnese Kocere
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
- Department of Molecular Life Science, University of Zurich, 8057 Zurich, Switzerland
| | - Robert L. Lalonde
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Alexa Burger
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| |
Collapse
|
13
|
The Exciting Realities and Possibilities of iPS-Derived Cardiomyocytes. Bioengineering (Basel) 2023; 10:bioengineering10020237. [PMID: 36829731 PMCID: PMC9952364 DOI: 10.3390/bioengineering10020237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have become a prevalent topic after their discovery, advertised as an ethical alternative to embryonic stem cells (ESCs). Due to their ability to differentiate into several kinds of cells, including cardiomyocytes, researchers quickly realized the potential for differentiated cardiomyocytes to be used in the treatment of heart failure, a research area with few alternatives. This paper discusses the differentiation process for human iPSC-derived cardiomyocytes and the possible applications of said cells while answering some questions regarding ethical issues.
Collapse
|
14
|
Pan Z, Liang P. Human-Induced Pluripotent Stem Cell-Based Differentiation of Cardiomyocyte Subtypes for Drug Discovery and Cell Therapy. Handb Exp Pharmacol 2023; 281:209-233. [PMID: 37421443 DOI: 10.1007/164_2023_663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Drug attrition rates have increased over the past few years, accompanied with growing costs for the pharmaceutical industry and consumers. Lack of in vitro models connecting the results of toxicity screening assays with clinical outcomes accounts for this high attrition rate. The emergence of cardiomyocytes derived from human pluripotent stem cells provides an amenable source of cells for disease modeling, drug discovery, and cardiotoxicity screening. Functionally similar to to embryonic stem cells, but with fewer ethical concerns, induced pluripotent stem cells (iPSCs) can recapitulate patient-specific genetic backgrounds, which would be a huge revolution for personalized medicine. The generated iPSC-derived cardiomyocytes (iPSC-CMs) represent different subtypes including ventricular-, atrial-, and nodal-like cardiomyocytes. Purifying these subtypes for chamber-specific drug screening presents opportunities and challenges. In this chapter, we discuss the strategies for the purification of iPSC-CMs, the use of iPSC-CMs for drug discovery and cardiotoxicity test, and the current limitations of iPSC-CMs that should be overcome for wider and more precise cardiovascular applications.
Collapse
Affiliation(s)
- Ziwei Pan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
15
|
Genome Editing and Myocardial Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1396:53-73. [PMID: 36454459 DOI: 10.1007/978-981-19-5642-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Congenital heart disease (CHD) has a strong genetic etiology, making it a likely candidate for therapeutic intervention using genetic editing. Complex genetics involving an orchestrated series of genetic events and over 400 genes are responsible for myocardial development. Cooperation is required from a vast series of genetic networks, and mutations in such can lead to CHD and cardiovascular abnormalities, affecting up to 1% of all live births. Genome editing technologies are becoming better studied and with time and improved logistics, CHD could be a prime therapeutic target. Syndromic, nonsyndromic, and cases of familial inheritance all involve identifiable causative mutations and thus have the potential for genome editing therapy. Mouse models are well-suited to study and predict clinical outcome. This review summarizes the anatomical and genetic timeline of myocardial development in both mice and humans, the potential of gene editing in typical CHD categories, as well as the use of mice thus far in reproducing models of human CHD and correcting the mutations that create them.
Collapse
|
16
|
Leowattana W, Leowattana T, Leowattana P. Human-induced pluripotent stem cell-atrial-specific cardiomyocytes and atrial fibrillation. World J Clin Cases 2022; 10:9588-9601. [PMID: 36186184 PMCID: PMC9516943 DOI: 10.12998/wjcc.v10.i27.9588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 02/05/2023] Open
Abstract
Patient-specific human-induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCMs) may be produced, genome-edited, and differentiated into multiple cell types for regenerative medicine, disease modeling, drug testing, toxicity screening, and three-dimensional tissue fabrication. There is presently no complete model of atrial fibrillation (AF) available for studying human pharmacological responses and evaluating the toxicity of potential medication candidates. It has been demonstrated that hiPSC-aCMs can replicate the electrophysiological disease phenotype and genotype of AF. The hiPSC-aCMs, however, are immature and do not reflect the maturity of aCMs in the native myocardium. Numerous laboratories utilize a variety of methodologies and procedures to improve and promote aCM maturation, including electrical stimulation, culture duration, biophysical signals, and changes in metabolic variables. This review covers the current methods being explored for use in the maturation of patient-specific hiPSC-aCMs and their application towards a personalized approach to the pharmacologic therapy of AF.
Collapse
Affiliation(s)
- Wattana Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tawithep Leowattana
- Department of Medicine, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Pathomthep Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Cardiovascular diseases are the leading cause of death worldwide, largely due to the limited regenerative capacity of the adult human heart. In contrast, teleost zebrafish hearts possess natural regeneration capacity by proliferation of pre-existing cardiomyocytes after injury. Hearts of mice can regenerate if injured in a few days after birth, which coincides with the transient capacity for cardiomyocyte proliferation. This review tends to elaborate the roles and mechanisms of Wnt/β-catenin signaling in heart development and regeneration in mammals and non-mammalian vertebrates. RECENT FINDINGS Studies in zebrafish, mice, and human embryonic stem cells demonstrate the binary effect for Wnt/β-catenin signaling during heart development. Both Wnts and Wnt antagonists are induced in multiple cell types during cardiac development and injury repair. In this review, we summarize composites of the Wnt signaling pathway and their different action routes, followed by the discussion of their involvements in cardiac specification, proliferation, and patterning. We provide overviews about canonical and non-canonical Wnt activity during heart homeostasis, remodeling, and regeneration. Wnt/β-catenin signaling exhibits biphasic and antagonistic effects on cardiac specification and differentiation depending on the stage of embryogenesis. Inhibition of Wnt signaling is beneficial for cardiac wound healing and functional recovery after injury. Understanding of the roles and mechanisms of Wnt signaling pathway in injured animal hearts will contribute to the development of potential therapeutics for human diseased hearts.
Collapse
Affiliation(s)
- Dongliang Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jianjian Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China
| | - Tao P Zhong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
18
|
Hsu Y, Huang K, Cheng K. Resuscitating the Field of Cardiac Regeneration: Seeking Answers from Basic Biology. Adv Biol (Weinh) 2021; 6:e2101133. [PMID: 34939372 DOI: 10.1002/adbi.202101133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is one of the leading causes for hospital admissions worldwide. HF patients are classified based on the chronic changes in left ventricular ejection fraction (LVEF) as preserved (LVEF ≥ 50%), reduced (LVEF ≤ 40%), or mid-ranged (40% < LVEF < 50%) HFs. Treatments nowadays can prevent HFrEF progress, whereas only a few of the treatments have been proven to be effective in improving the survival of HFpEF. In this review, numerous mediators involved in the pathogenesis of HF are summarized. The regional upstream signaling and their diagnostic and therapeutic potential are also discussed. Additionally, the recent challenges and development in cardiac regenerative therapy that hold opportunities for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Yaching Hsu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|
19
|
Miyamoto M, Andersen P, Sulistio E, Liu X, Murphy S, Kannan S, Nam L, Miyamoto W, Tampakakis E, Hibino N, Uosaki H, Kwon C. Noncanonical Notch signals have opposing roles during cardiac development. Biochem Biophys Res Commun 2021; 577:12-16. [PMID: 34487959 PMCID: PMC8484041 DOI: 10.1016/j.bbrc.2021.08.094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
The Notch pathway is an ancient intercellular signaling system with crucial roles in numerous cell-fate decision processes across species. While the canonical pathway is activated by ligand-induced cleavage and nuclear localization of membrane-bound Notch, Notch can also exert its activity in a ligand/transcription-independent fashion, which is conserved in Drosophila, Xenopus, and mammals. However, the noncanonical role remains poorly understood in in vivo processes. Here we show that increased levels of the Notch intracellular domain (NICD) in the early mesoderm inhibit heart development, potentially through impaired induction of the second heart field (SHF), independently of the transcriptional effector RBP-J. Similarly, inhibiting Notch cleavage, shown to increase noncanonical Notch activity, suppressed SHF induction in embryonic stem cell (ESC)-derived mesodermal cells. In contrast, NICD overexpression in late cardiac progenitor cells lacking RBP-J resulted in an increase in heart size. Our study suggests that noncanonical Notch signaling has stage-specific roles during cardiac development.
Collapse
Affiliation(s)
- Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Edrick Sulistio
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Xihe Liu
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Lucy Nam
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD,Present address: Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - William Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Emmanouil Tampakakis
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Narutoshi Hibino
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA; Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
20
|
Abstract
Cardiomyogenesis, the process by which the body generates cardiomyocytes, is poorly understood. We have recently shown that Sfrp2 promotes cardiomyogenesis in vitro. The objective of this study was to determine if Sfrp2 would similarly promote cardiomyogenesis in vivo. To test this hypothesis, we tracked multipotent cKit(+) cells in response to Sfrp2 treatment. In control adult mice, multipotent cKit(+) cells typically differentiated into endothelial cells but not cardiomyocytes. In contrast, Sfrp2 switched the fate of these cells. Following Sfrp2 injection, multipotent cKit(+) cells differentiated solely into cardiomyocytes. Sfrp2-derived cardiomyocytes integrated into the myocardium and exhibited identical physiological properties to preexisting native cardiomyocytes. The ability of Sfrp2 to promote cardiomyogenesis was further supported by tracking EdU-labeled cells. In addition, Sfrp2 did not promote the formation of new cardiomyocytes when the cKit(+) cell population was selectively ablated in vivo using a diphtheria toxin receptor-diphtheria toxin model. Notably, Sfrp2-induced cardiomyogenesis was associated with significant functional improvements in a cardiac injury model. In summary, our study further demonstrates the importance of Sfrp2 in cardiomyogenesis.
Collapse
|
21
|
Characterization of Ventricular and Atrial Cardiomyocyte Subtypes from Human-Induced Pluripotent Stem Cells. Methods Mol Biol 2021. [PMID: 34302655 DOI: 10.1007/978-1-0716-1484-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Human iPSC-derived cardiomyocytes (hiPSC-CMs) are expected to be used in regenerative therapies and drug discovery for heart failure. hiPSC-CMs are a mixture of mainly ventricular CMs (VCMs) and also of atrial CMs (ACMs) and pacemaker cells. Here we describe a method to enrich VCM and ACM differentiation and to characterize these subtypes by gene expression analysis using qRT-PCR and by electrophysiological properties using the patch-clamp method. The differentiated VCMs and ACMs highly express VCM and ACM marker genes, respectively. Furthermore, both subtypes show specific properties of action potentials.
Collapse
|
22
|
Yamada D, Nakamura M, Takao T, Takihira S, Yoshida A, Kawai S, Miura A, Ming L, Yoshitomi H, Gozu M, Okamoto K, Hojo H, Kusaka N, Iwai R, Nakata E, Ozaki T, Toguchida J, Takarada T. Induction and expansion of human PRRX1 + limb-bud-like mesenchymal cells from pluripotent stem cells. Nat Biomed Eng 2021; 5:926-940. [PMID: 34373601 DOI: 10.1038/s41551-021-00778-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/01/2021] [Indexed: 02/05/2023]
Abstract
Current protocols for the differentiation of human pluripotent stem cells (hPSCs) into chondrocytes do not allow for the expansion of intermediate progenitors so as to prospectively assess their chondrogenic potential. Here we report a protocol that leverages PRRX1-tdTomato reporter hPSCs for the selective induction of expandable and ontogenetically defined PRRX1+ limb-bud-like mesenchymal cells under defined xeno-free conditions, and the prospective assessment of the cells' chondrogenic potential via the cell-surface markers CD90, CD140B and CD82. The cells, which proliferated stably and exhibited the potential to undergo chondrogenic differentiation, formed hyaline cartilaginous-like tissue commensurate to their PRRX1-expression levels. Moreover, we show that limb-bud-like mesenchymal cells derived from patient-derived induced hPSCs can be used to identify therapeutic candidates for type II collagenopathy and we developed a method to generate uniformly sized hyaline cartilaginous-like particles by plating the cells on culture dishes coated with spots of a zwitterionic polymer. PRRX1+ limb-bud-like mesenchymal cells could facilitate the mass production of chondrocytes and cartilaginous tissues for applications in drug screening and tissue engineering.
Collapse
Affiliation(s)
- Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Nakamura
- Precision Health, Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomoka Takao
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shota Takihira
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Aki Yoshida
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shunsuke Kawai
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akihiro Miura
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Lu Ming
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Yoshitomi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mai Gozu
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kumi Okamoto
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyuki Kusaka
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama, Japan
| | - Ryosuke Iwai
- Institute of Frontier Science and Technology, Okayama University of Science, Okayama, Japan
| | - Eiji Nakata
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Department Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
23
|
Abstract
Congenital birth defects result from an abnormal development of an embryo and have detrimental effects on children's health. Specifically, congenital heart malformations are a leading cause of death among pediatric patients and often require surgical interventions within the first year of life. Increased efforts to navigate the human genome provide an opportunity to discover multiple candidate genes in patients suffering from birth defects. These efforts, however, fail to provide an explanation regarding the mechanisms of disease pathogenesis and emphasize the need for an efficient platform to screen candidate genes. Xenopus is a rapid, cost effective, high-throughput vertebrate organism to model the mechanisms behind human disease. This review provides numerous examples describing the successful use of Xenopus to investigate the contribution of patient mutations to complex phenotypes including congenital heart disease and heterotaxy. Moreover, we describe a variety of unique methods that allow us to rapidly recapitulate patients' phenotypes in frogs: gene knockout and knockdown strategies, the use of fate maps for targeted manipulations, and novel imaging modalities. The combination of patient genomics data and the functional studies in Xenopus will provide necessary answers to the patients suffering from birth defects. Furthermore, it will allow for the development of better diagnostic methods to ensure early detection and intervention. Finally, with better understanding of disease pathogenesis, new treatment methods can be tailored specifically to address patient's phenotype and genotype.
Collapse
Affiliation(s)
- Valentyna Kostiuk
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, United States
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
24
|
Nomura S, Komuro I. Precision medicine for heart failure based on molecular mechanisms: The 2019 ISHR Research Achievement Award Lecture. J Mol Cell Cardiol 2021; 152:29-39. [PMID: 33275937 DOI: 10.1016/j.yjmcc.2020.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Heart failure is a leading cause of death, and the number of patients with heart failure continues to increase worldwide. To realize precision medicine for heart failure, its underlying molecular mechanisms must be elucidated. In this review summarizing the "The Research Achievement Award Lecture" of the 2019 XXIII ISHR World Congress held in Beijing, China, we would like to introduce our approaches for investigating the molecular mechanisms of cardiac hypertrophy, development, and failure, as well as discuss future perspectives.
Collapse
Affiliation(s)
- Seitaro Nomura
- Department of Cardiovascular Medicine, The University of Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo, Japan.
| |
Collapse
|
25
|
Lin L, Xu W, Li Y, Zhu P, Yuan W, Liu M, Shi Y, Chen Y, Liang J, Chen J, Yang B, Cai W, Wen Y, Zhu X, Peng X, Zhou Z, Mo X, Wan Y, Yuan H, Li F, Ye X, Jiang Z, Wang Y, Zhuang J, Fan X, Wu X. Pygo1 regulates pathological cardiac hypertrophy via a β-catenin-dependent mechanism. Am J Physiol Heart Circ Physiol 2021; 320:H1634-H1645. [PMID: 33635162 DOI: 10.1152/ajpheart.00538.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Wnt/β-catenin signaling plays a key role in pathological cardiac remodeling in adults. The identification of a tissue-specific Wnt/β-catenin interaction factor may provide a tissue-specific clinical targeting strategy. Drosophila Pygo encodes the core interaction factor of Wnt/β-catenin. Two Pygo homologs (Pygo1 and Pygo2) have been identified in mammals. Different from the ubiquitous expression profile of Pygo2, Pygo1 is enriched in cardiac tissue. However, the role of Pygo1 in mammalian cardiac disease is yet to be elucidated. In this study, we found that Pygo1 was upregulated in human cardiac tissues with pathological hypertrophy. Cardiac-specific overexpression of Pygo1 in mice spontaneously led to cardiac hypertrophy accompanied by declined cardiac function, increased heart weight/body weight and heart weight/tibial length ratios, and increased cell size. The canonical β-catenin/T-cell transcription factor 4 (TCF4) complex was abundant in Pygo1-overexpressing transgenic (Pygo1-TG) cardiac tissue, and the downstream genes of Wnt signaling, that is, Axin2, Ephb3, and c-Myc, were upregulated. A tail vein injection of β-catenin inhibitor effectively rescued the phenotype of cardiac failure and pathological myocardial remodeling in Pygo1-TG mice. Furthermore, in vivo downregulated pygo1 during cardiac hypertrophic condition antagonized agonist-induced cardiac hypertrophy. Therefore, our study is the first to present in vivo evidence demonstrating that Pygo1 regulates pathological cardiac hypertrophy in a canonical Wnt/β-catenin-dependent manner, which may provide new clues for tissue-specific clinical treatment via targeting this pathway.NEW & NOTEWORTHY In this study, we found that Pygo1 is associated with human pathological hypertrophy. Cardiac-specific overexpression of Pygo1 in mice spontaneously led to cardiac hypertrophy. Meanwhile, cardiac function was improved when expression of Pygo1 was interfered in hypertrophy-model mice. Our study is the first to present in vivo evidence demonstrating that Pygo1 regulates pathological cardiac hypertrophy in a canonical Wnt/β-catenin-dependent manner, which may provide new clues for a tissue-specific clinical treatment targeting this pathway.
Collapse
Affiliation(s)
- Li Lin
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Wei Xu
- Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yongqing Li
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wuzhou Yuan
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ming Liu
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yan Shi
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu Chen
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jifeng Liang
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jimei Chen
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Boyu Yang
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Wanwan Cai
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yao Wen
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaolan Zhu
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiyang Peng
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zuoqiong Zhou
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoyang Mo
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yongqi Wan
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Haiyun Yuan
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fang Li
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiangli Ye
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhigang Jiang
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yuequn Wang
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiongwei Fan
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiushan Wu
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
26
|
Procopio MC, Lauro R, Nasso C, Carerj S, Squadrito F, Bitto A, Di Bella G, Micari A, Irrera N, Costa F. Role of Adenosine and Purinergic Receptors in Myocardial Infarction: Focus on Different Signal Transduction Pathways. Biomedicines 2021; 9:biomedicines9020204. [PMID: 33670488 PMCID: PMC7922652 DOI: 10.3390/biomedicines9020204] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Myocardial infarction (MI) is a dramatic event often caused by atherosclerotic plaque erosion or rupture and subsequent thrombotic occlusion of a coronary vessel. The low supply of oxygen and nutrients in the infarcted area may result in cardiomyocytes necrosis, replacement of intact myocardium with non-contractile fibrous tissue and left ventricular (LV) function impairment if blood flow is not quickly restored. In this review, we summarized the possible correlation between adenosine system, purinergic system and Wnt/β-catenin pathway and their role in the pathogenesis of cardiac damage following MI. In this context, several pathways are involved and, in particular, the adenosine receptors system shows different interactions between its members and purinergic receptors: their modulation might be effective not only for a normal functional recovery but also for the treatment of heart diseases, thus avoiding fibrosis, reducing infarcted area and limiting scaring. Similarly, it has been shown that Wnt/β catenin pathway is activated following myocardial injury and its unbalanced activation might promote cardiac fibrosis and, consequently, LV systolic function impairment. In this regard, the therapeutic benefits of Wnt inhibitors use were highlighted, thus demonstrating that Wnt/β-catenin pathway might be considered as a therapeutic target to prevent adverse LV remodeling and heart failure following MI.
Collapse
Affiliation(s)
- Maria Cristina Procopio
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Chiara Nasso
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Antonio Micari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, A.O.U. Policlinic “G. Martino”, 98165 Messina, Italy;
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
- Correspondence: ; Tel.: +39-090-221-3093; Fax: +39-090-221-23-81
| | - Francesco Costa
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| |
Collapse
|
27
|
Ashok P, Parikh A, Du C, Tzanakakis ES. Xenogeneic-Free System for Biomanufacturing of Cardiomyocyte Progeny From Human Pluripotent Stem Cells. Front Bioeng Biotechnol 2020; 8:571425. [PMID: 33195131 PMCID: PMC7644809 DOI: 10.3389/fbioe.2020.571425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/28/2020] [Indexed: 01/14/2023] Open
Abstract
Functional heart cells and tissues sourced from human pluripotent stem cells (hPSCs) have great potential for substantially advancing treatments of cardiovascular maladies. Realization of this potential will require the development of cost-effective and tunable bioprocesses for manufacturing hPSC-based cell therapeutics. Here, we report the development of a xeno-free platform for guiding the cardiogenic commitment of hPSCs. The system is based on a fully defined, open-source formulation without complex supplements, which have varied and often undetermined effects on stem cell physiology. The formulation was used to systematically investigate factors inducing the efficient commitment to cardiac mesoderm of three hPSC lines. Contractile clusters of cells appeared within a week of differentiation in planar cultures and by day 13 over 80% of the cells expressed cardiac progeny markers such as TNNT2. In conjunction with expansion, this differentiation strategy was employed in stirred-suspension cultures of hPSCs. Scalable differentiation resulted in 0.4-2 million CMs/ml or ∼5-20 TNNT2-positive cells per seeded hPSC without further enrichment. Our findings will contribute to the engineering of bioprocesses advancing the manufacturing of stem cell-based therapeutics for heart diseases.
Collapse
Affiliation(s)
- Preeti Ashok
- Chemical and Biological Engineering, Tufts University, Medford, MA, United States
| | | | - Chuang Du
- Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Emmanuel S. Tzanakakis
- Chemical and Biological Engineering, Tufts University, Medford, MA, United States
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
- Developmental Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
28
|
Yan Y, Liu F, Hou X, Wan J, Xiong Q, Zhou R, Liao B. [Joint regulation of Wnt and bone morphogenetic protein signaling pathways to promote differentiation of human induced pluripotent stem cells into cardiomyocytes]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:1313-1321. [PMID: 33063499 DOI: 10.7507/1002-1892.201912087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To explore the role of joint regulation of Wnt and bone morphogenetic protein (BMP) signaling pathways in the differentiation of human induced pluripotent stem cells (hiPSCs) into cardiomyocytes. Methods HiPSCs were cultured and observed under inverted phase contrast microscope. Immunofluorescence staining was used to observe the expressions of hiPSCs pluripotent markers (OCT3/4, NANOG, and TRA-1-60). HiPSCs were passaged which were taken for subsequent experiments within the 35th passage. When the fusion degree of hiPSCs was close to 100%, the CHIR99021 (Wnt pathway activator) was added on the 0th day of differentiation. Different concentrations of IWP4 (inhibitor of Wnt production) were added on the 3rd day of differentiation, and the best concentration of IWP4 was added at different time points. The optimal concentration and the best effective period of IWP4 were obtained by detecting the expression of troponin T (TNNT2) mRNA by real-time fluorescence quantitative PCR. Then, on the basis of adding CHIR99021 and IWP4, different concentrations of BMP-4 were added on the 5th day of differentiation, and the best concentration of BMP-4 was added at different time points. The optimal concentration and best effective period of BMP-4 were obtained by detecting the expression of TNNT2 mRNA. Finally, hiPSCs were divided into three groups: Wnt group, BMP group, and Wnt+BMP group. On the basis of adding CHIR99021 on the 0th day of differentiation, IWP4, BMP-4, and IWP4+BMP-4 were added into Wnt group, BMP group, and Wnt+BMP group respectively according to the screening results. Cells were collected on the 7th and the 15th days of differentiation. The expressions of myocardial precursor cell markers [ISL LIM homeobox 1 (ISL1), NK2 homeobox 5 (NKX2-5)] and cardiomyocyte specific markers [myocyte enhancer factor 2C (MEF2C), myosin light chain 2 (MYL2), MYL7, and TNNT2] were detected by real-time fluorescent quantitative PCR. Cells were collected on the 28th day of differentiation, and the expression of cardiac troponin T (cTnT) was detected by flow cytometry and immunofluorescence staining. Results The results of cell mophology and immunoflurescence staining showed that the OCT3/4, NANOG, and TRA-1-60 were highly expressed in hiPSCs, which suggested that hiPSCs had characteristics of pluripotency. The optimal concentration of IWP4 was 10.0 μmol/L ( P<0.05) and the best effective period was the 3rd day ( P<0.05) in inducing hiPSCs to differentiate into cardiomyocytes. The optimal concentration of BMP-4 was 20.0 ng/mL ( P<0.05) and the best effective period was the 3rd day ( P<0.05). The relative expressions of ISL1, NKX2-5, MEF2C, MYL2, MYL7, and TNNT2 mRNAs, the positive expression ratio of cTnT detected by flow cytometry, and sarcomere structure detected by immunofluorescence staining of Wnt+BMP group were superior to those of Wnt group ( P<0.05). Conclusion Joint regulation of Wnt and BMP signaling pathways can improve the differentiation efficiency of hiPSCs into cardiomyocytes.
Collapse
Affiliation(s)
- Ying Yan
- Clinical Medicine College of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China;College of Integrative Chinese and Western Medicine, Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Feng Liu
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Xiaojie Hou
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Juyi Wan
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Qi Xiong
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Rui Zhou
- Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000, P.R.China
| |
Collapse
|
29
|
Protze SI, Lee JH, Keller GM. Human Pluripotent Stem Cell-Derived Cardiovascular Cells: From Developmental Biology to Therapeutic Applications. Cell Stem Cell 2020; 25:311-327. [PMID: 31491395 DOI: 10.1016/j.stem.2019.07.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in our understanding of cardiovascular development have provided a roadmap for the directed differentiation of human pluripotent stem cells (hPSCs) to the major cell types found in the heart. In this Perspective, we review the state of the field in generating and maturing cardiovascular cells from hPSCs based on our fundamental understanding of heart development. We then highlight their applications for studying human heart development, modeling disease-performing drug screening, and cell replacement therapy. With the advancements highlighted here, the promise that hPSCs will deliver new treatments for degenerative and debilitating diseases may soon be fulfilled.
Collapse
Affiliation(s)
- Stephanie I Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jee Hoon Lee
- BlueRock Therapeutics ULC, Toronto, ON M5G 1L7, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
30
|
D’Antonio-Chronowska A, D’Antonio M, Frazer KA. In vitro Differentiation of Human iPSC-derived Cardiovascular Progenitor Cells (iPSC-CVPCs). Bio Protoc 2020; 10:e3755. [PMID: 33659414 PMCID: PMC7853936 DOI: 10.21769/bioprotoc.3755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cell derived cardiovascular progenitor cells (iPSC-CVPCs) provide an unprecedented platform for examining the molecular underpinnings of cardiac development and disease etiology, but also have great potential to play pivotal roles in the future of regenerative medicine and pharmacogenomic studies. Biobanks like iPSCORE ( Stacey et al., 2013 ; Panopoulos et al., 2017 ), which contain iPSCs generated from hundreds of genetically and ethnically diverse individuals, are an invaluable resource for conducting these studies. Here, we present an optimized, cost-effective and highly standardized protocol for large-scale derivation of human iPSC-CVPCs using small molecules and purification using metabolic selection. We have successfully applied this protocol to derive iPSC-CVPCs from 154 different iPSCORE iPSC lines obtaining large quantities of highly pure cardiac cells. An important component of our protocol is Cell confluency estimates (ccEstimate), an automated methodology for estimating the time when an iPSC monolayer will reach 80% confluency, which is optimal for initiating iPSC-CVPC derivation, and enables the protocol to be readily used across iPSC lines with different growth rates. Moreover, we showed that cellular heterogeneity across iPSC-CVPCs is due to varying proportions of two distinct cardiac cell types: cardiomyocytes (CMs) and epicardium-derived cells (EPDCs), both of which have been shown to have a critical function in heart regeneration. This protocol eliminates the need of iPSC line-to-line optimization and can be easily adapted and scaled to high-throughput studies or to generate large quantities of cells suitable for regenerative medicine applications.
Collapse
Affiliation(s)
| | - Matteo D’Antonio
- Department of Pediatrics, University of California, San Diego, La Jolla, USA
| | - Kelly A. Frazer
- Department of Pediatrics, University of California, San Diego, La Jolla, USA
- Institue for Genomic Medicine, University of California, San Diego, La Jolla, USA
| |
Collapse
|
31
|
Swedlund B, Lescroart F. Cardiopharyngeal Progenitor Specification: Multiple Roads to the Heart and Head Muscles. Cold Spring Harb Perspect Biol 2020; 12:a036731. [PMID: 31818856 PMCID: PMC7397823 DOI: 10.1101/cshperspect.a036731] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During embryonic development, the heart arises from various sources of undifferentiated mesodermal progenitors, with an additional contribution from ectodermal neural crest cells. Mesodermal cardiac progenitors are plastic and multipotent, but are nevertheless specified to a precise heart region and cell type very early during development. Recent findings have defined both this lineage plasticity and early commitment of cardiac progenitors, using a combination of single-cell and population analyses. In this review, we discuss several aspects of cardiac progenitor specification. We discuss their markers, fate potential in vitro and in vivo, early segregation and commitment, and also intrinsic and extrinsic cues regulating lineage restriction from multipotency to a specific cell type of the heart. Finally, we also discuss the subdivisions of the cardiopharyngeal field, and the shared origins of the heart with other mesodermal derivatives, including head and neck muscles.
Collapse
Affiliation(s)
- Benjamin Swedlund
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | | |
Collapse
|
32
|
Hoppler S, Conlon FL. Xenopus: Experimental Access to Cardiovascular Development, Regeneration Discovery, and Cardiovascular Heart-Defect Modeling. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037200. [PMID: 31767648 DOI: 10.1101/cshperspect.a037200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Xenopus has been used to study a wide array of developmental processes, benefiting from vast quantities of relatively large, externally developing eggs. Xenopus is particularly amenable to examining the cardiac system because many of the developmental processes and genes involved in cardiac specification, differentiation, and growth are conserved between Xenopus and human and have been characterized in detail. Furthermore, compared with other higher vertebrate models, Xenopus embryos can survive longer without a properly functioning heart or circulatory system, enabling investigation of later consequences of early embryological manipulations. This biology is complemented by experimental technology, such as embryonic explants to study the heart, microinjection of overexpression constructs, and, most recently, the generation of genetic mutations through gene-editing technologies. Recent investigations highlight Xenopus as a powerful experimental system for studying injury/repair and regeneration and for congenital heart disease (CHD) modeling, which reinforces why this model system remains ideal for studying heart development.
Collapse
Affiliation(s)
- Stefan Hoppler
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, United Kingdom
| | - Frank L Conlon
- Department of Biology and Genetics, University of North Carolina McAllister Heart Institute, Chapel Hill, North Carolina 27599-3280, USA
| |
Collapse
|
33
|
Vyas B, Nandkishore N, Sambasivan R. Vertebrate cranial mesoderm: developmental trajectory and evolutionary origin. Cell Mol Life Sci 2020; 77:1933-1945. [PMID: 31722070 PMCID: PMC11105048 DOI: 10.1007/s00018-019-03373-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023]
Abstract
Vertebrate cranial mesoderm is a discrete developmental unit compared to the mesoderm below the developing neck. An extraordinary feature of the cranial mesoderm is that it includes a common progenitor pool contributing to the chambered heart and the craniofacial skeletal muscles. This striking developmental potential and the excitement it generated led to advances in our understanding of cranial mesoderm developmental mechanism. Remarkably, recent findings have begun to unravel the origin of its distinct developmental characteristics. Here, we take a detailed view of the ontogenetic trajectory of cranial mesoderm and its regulatory network. Based on the emerging evidence, we propose that cranial and posterior mesoderm diverge at the earliest step of the process that patterns the mesoderm germ layer along the anterior-posterior body axis. Further, we discuss the latest evidence and their impact on our current understanding of the evolutionary origin of cranial mesoderm. Overall, the review highlights the findings from contemporary research, which lays the foundation to probe the molecular basis of unique developmental potential and evolutionary origin of cranial mesoderm.
Collapse
Affiliation(s)
- Bhakti Vyas
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nitya Nandkishore
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
- SASTRA University, Thirumalaisamudram, Thanjavur, 613401, India
| | - Ramkumar Sambasivan
- Indian Institute of Science Education and Research (IISER) Tirupati, Transit Campus, Karakambadi Road, Rami Reddy Nagar, Mangalam, Tirupati, Andhra Pradesh, 517507, India.
| |
Collapse
|
34
|
IGF1 Knockdown Hinders Myocardial Development through Energy Metabolism Dysfunction Caused by ROS-Dependent FOXO Activation in the Chicken Heart. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7838754. [PMID: 31949883 PMCID: PMC6948330 DOI: 10.1155/2019/7838754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/16/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022]
Abstract
Insulin-like growth factor 1 (IGF1) is a multifunctional cellular regulatory factor that can regulate cell growth and development by mediating growth hormone stimulation. However, the mechanism of IGF1 dysfunction in cardiomyocyte development is seldom reported. To study this, we employed the models of IGF1 knockdown in chicken embryo in vivo and in cardiomyocytes in vitro. We detected the antioxidant capacity, PI3K/Akt pathway, energy metabolism-related genes, and myocardial development-related genes. Our results revealed that the low expression of IGF1 can significantly suppress the antioxidant capacity and increase the ROS (P < 0.05) levels, activating the AMPK and PI3K pathway by inhibiting the expression of IRS1. We also found that myocardial energy metabolism is blocked through IGF1, GLUT, and IGFBP inhibition, further inducing myocardial developmental disorder by inhibiting Mesp1, GATA, Nkx2.5, and MyoD expression. Altogether, we conclude that low IGF1 expression can hinder myocardial development through the dysfunction of energy metabolism caused by ROS-dependent FOXO activation.
Collapse
|
35
|
Hamad S, Derichsweiler D, Papadopoulos S, Nguemo F, Šarić T, Sachinidis A, Brockmeier K, Hescheler J, Boukens BJ, Pfannkuche K. Generation of human induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer and scalable 3D suspension bioreactor cultures with reduced batch-to-batch variations. Theranostics 2019; 9:7222-7238. [PMID: 31695764 PMCID: PMC6831300 DOI: 10.7150/thno.32058] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are promising candidates to treat myocardial infarction and other cardiac diseases. Such treatments require pure cardiomyocytes (CMs) in large quantities. Methods: In the present study we describe an improved protocol for production of hiPSC-CMs in which hiPSCs are first converted into mesodermal cells by stimulation of wingless (Wnt) signaling using CHIR99021, which are then further differentiated into CM progenitors by simultaneous inhibition of porcupine and tankyrase pathways using IWP2 and XAV939 under continuous supplementation of ascorbate during the entire differentiation procedure. Results: The protocol resulted in reproducible generation of >90% cardiac troponin T (TNNT2)-positive cells containing highly organized sarcomeres. In 2D monolayer cultures CM yields amounted to 0.5 million cells per cm2 growth area, and on average 72 million cells per 100 mL bioreactor suspension culture without continuous perfusion. The differentiation efficiency was hardly affected by the initial seeding density of undifferentiated hiPSCs. Furthermore, batch-to-batch variations were reduced by combinatorial use of ascorbate, IWP2, and XAV939. Conclusion: Combined inhibition of porcupine and tankyrase sub-pathways of Wnt signaling and continuous ascorbate supplementation, enable robust and efficient production of hiPSC-CMs.
Collapse
Affiliation(s)
- Sarkawt Hamad
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Biology Department, Faculty of Science, Soran University, Soran, Kurdistan region-Iraq
| | - Daniel Derichsweiler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Symeon Papadopoulos
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Center for Physiology and Pathophysiology, Institute for Vegetative Physiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Agapios Sachinidis
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Konrad Brockmeier
- Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kurt Pfannkuche
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| |
Collapse
|
36
|
Hwang WY, Marquez J, Khokha MK. Xenopus: Driving the Discovery of Novel Genes in Patient Disease and Their Underlying Pathological Mechanisms Relevant for Organogenesis. Front Physiol 2019; 10:953. [PMID: 31417417 PMCID: PMC6682594 DOI: 10.3389/fphys.2019.00953] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 07/09/2019] [Indexed: 12/16/2022] Open
Abstract
Frog model organisms have been appreciated for their utility in exploring physiological phenomena for nearly a century. Now, a vibrant community of biologists that utilize this model organism has poised Xenopus to serve as a high throughput vertebrate organism to model patient-driven genetic diseases. This has facilitated the investigation of effects of patient mutations on specific organs and signaling pathways. This approach promises a rapid investigation into novel mechanisms that disrupt normal organ morphology and function. Considering that many disease states are still interrogated in vitro to determine relevant biological processes for further study, the prospect of interrogating genetic disease in Xenopus in vivo is an attractive alternative. This model may more closely capture important aspects of the pathology under investigation such as cellular micro environments and local forces relevant to a specific organ's development and homeostasis. This review aims to highlight recent methodological advances that allow investigation of genetic disease in organ-specific contexts in Xenopus as well as provide examples of how these methods have led to the identification of novel mechanisms and pathways important for understanding human disease.
Collapse
Affiliation(s)
| | | | - Mustafa K. Khokha
- Department of Pediatrics and Genetics, The Pediatric Genomics Discovery Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
37
|
Kaplan NA, Wang W, Christiaen L. Initial characterization of Wnt-Tcf functions during Ciona heart development. Dev Biol 2019; 448:199-209. [PMID: 30635127 PMCID: PMC6487219 DOI: 10.1016/j.ydbio.2018.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 12/16/2022]
Abstract
In vertebrate embryos, the cardiopharyngeal mesoderm gives rise to both cardiac and branchiomeric head muscles. The canonical Wnt signaling pathway regulates many aspects of cardiomyocyte specification, and modulates a balance between skeletal and cardiac myogenesis during vertebrate head muscle development. However, the role of Wnt signaling during ascidian cardiopharyngeal development remains elusive. Here, we documented the expression of Wnt pathway components during cardiopharyngeal development in Ciona, and generated tools to investigate potential roles for Wnt signaling, and its transcriptional effector Tcf, on heart vs. pharyngeal muscle fate specification. Neither focused functional analyses nor lineage-specific transcriptome profiling uncovered a significant role for Tcf during early cardiac vs. pharyngeal muscle fate choice. By contrast, Wnt gene expression patterns of Frizzled4 and Lrp4/8 and CRISPR/Cas9-mediated Tcf knock-down suggested a later requirement for Wnt signaling during heart morphogenesis and/or cardiomyocyte differentiation. This study provides a provisional set of reagents to study Wnt signaling function in Ciona, and promising insights for future analyses of Wnt functions during heart organogenesis.
Collapse
Affiliation(s)
- Nicole A Kaplan
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Wei Wang
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA.
| |
Collapse
|
38
|
Foley TE, Hess B, Savory JGA, Ringuette R, Lohnes D. Role of Cdx factors in early mesodermal fate decisions. Development 2019; 146:146/7/dev170498. [PMID: 30936115 DOI: 10.1242/dev.170498] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/01/2019] [Indexed: 12/30/2022]
Abstract
Murine cardiac and hematopoietic progenitors are derived from Mesp1+ mesoderm. Cdx function impacts both yolk sac hematopoiesis and cardiogenesis in zebrafish, suggesting that Cdx family members regulate early mesoderm cell fate decisions. We found that Cdx2 occupies a number of transcription factor loci during embryogenesis, including key regulators of both cardiac and blood development, and that Cdx function is required for normal expression of the cardiogenic transcription factors Nkx2-5 and Tbx5 Furthermore, Cdx and Brg1, an ATPase subunit of the SWI/SNF chromatin remodeling complex, co-occupy a number of loci, suggesting that Cdx family members regulate target gene expression through alterations in chromatin architecture. Consistent with this, we demonstrate loss of Brg1 occupancy and altered chromatin structure at several cardiogenic genes in Cdx-null mutants. Finally, we provide evidence for an onset of Cdx2 expression at E6.5 coinciding with egression of cardiac progenitors from the primitive streak. Together, these findings suggest that Cdx functions in multi-potential mesoderm to direct early cell fate decisions through transcriptional regulation of several novel target genes, and provide further insight into a potential epigenetic mechanism by which Cdx influences target gene expression.
Collapse
Affiliation(s)
- Tanya E Foley
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Bradley Hess
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Joanne G A Savory
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Randy Ringuette
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - David Lohnes
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
39
|
Kim YH, Kim BJ, Kim SM, Kim SU, Ryu BY. Induction of cardiomyocyte‑like cells from hair follicle cells in mice. Int J Mol Med 2019; 43:2230-2240. [PMID: 30864673 DOI: 10.3892/ijmm.2019.4133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 03/08/2019] [Indexed: 11/05/2022] Open
Abstract
Hair follicles (HFs) are a well‑characterized niche for adult stem cells (SCs), and include epithelial and melanocytic SCs. HF cells are an accessible source of multipotent adult SCs for the generation of the interfollicular epidermis, HF structures and sebaceous glands in addition to the reconstitution of novel HFs in vivo. In the present study, it was demonstrated that HF cells are able to be induced to differentiate into cardiomyocyte‑like cells in vitro under specific conditions. It was determined that HF cells cultured on OP9 feeder cells in KnockOut‑Dulbecco's modified Eagle's medium/B27 in the presence of vascular endothelial growth factors differentiated into cardiomyocyte‑like cells that express markers specific to cardiac lineage, but do not express non‑cardiac lineage markers including neural stem/progenitor cell, HF bulge cells or undifferentiated spermatogonia markers. These cardiomyocyte‑like cells exhibited a spindle‑ and filament‑shaped morphology similar to that presented by cardiac muscles and exhibited spontaneous beating that persisted for over 3 months. These results demonstrate that SC reprogramming and differentiation may be induced without resulting in any genetic modification, which is important for the clinical applications of SCs including tissue and organ regeneration.
Collapse
Affiliation(s)
- Yong-Hee Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| | - Bang-Jin Kim
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seok-Man Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| | - Sun-Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung‑Ang University, Anseong, Gyeonggi‑do 17546, Republic of Korea
| |
Collapse
|
40
|
Haworth K, Samuel L, Black S, Kirilenko P, Latinkic B. Liver Specification in the Absence of Cardiac Differentiation Revealed by Differential Sensitivity to Wnt/β Catenin Pathway Activation. Front Physiol 2019; 10:155. [PMID: 30890948 PMCID: PMC6411699 DOI: 10.3389/fphys.2019.00155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/08/2019] [Indexed: 12/30/2022] Open
Abstract
Embryonic precursors of liver and heart, whilst not sharing cellular origin, develop in close proximity through a dynamic series of inductive signaling events. During gastrulation anterior endoderm (AE) provides cardiogenic signals that act on adjacent mesoderm, resulting in induction of cardiac precursors. Subsequently cardiogenic mesoderm generates a FGF signal that acts on adjacent AE to induce foregut organ specification. Additional signals such as BMP and Wnt provide further information required for liver specification. Most findings on liver specification were derived from mouse explant studies as well as experiments with Xenopus and zebrafish embryos. To address some of the limitations of these models, here we used two complementary ex vivo models based on Xenopus embryos: pluripotent animal cap explants expressing Gata4 transcription factor and conjugates of gastrula-stage AE with animal caps (AC). We show that in these models liver specification is not sensitive to Wnt signaling manipulation, in contrast to the requirement for Wnt antagonism shown in vivo. FGF pathway is not necessary for Gata4-induced liver specification in animal cap explants but is required for prolonged period in sandwiches of AE and AC. In contrast, BMP signaling is shown to be essential for Gata4-induced liver specification. Our findings may have implications for research on liver differentiation from embryonic stem cells.
Collapse
Affiliation(s)
- Kim Haworth
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Lee Samuel
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Sarah Black
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Pavel Kirilenko
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Branko Latinkic
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
41
|
Guo Y, Dorn T, Kühl SJ, Linnemann A, Rothe M, Pfister AS, Vainio S, Laugwitz KL, Moretti A, Kühl M. The Wnt inhibitor Dkk1 is required for maintaining the normal cardiac differentiation program in Xenopus laevis. Dev Biol 2019; 449:1-13. [PMID: 30797757 PMCID: PMC6496975 DOI: 10.1016/j.ydbio.2019.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/15/2019] [Accepted: 02/16/2019] [Indexed: 12/15/2022]
Abstract
Wnt proteins can activate different intracellular signaling pathways. These pathways need to be tightly regulated for proper cardiogenesis. The canonical Wnt/β-catenin inhibitor Dkk1 has been shown to be sufficient to trigger cardiogenesis in gain-of-function experiments performed in multiple model systems. Loss-of-function studies however did not reveal any fundamental function for Dkk1 during cardiogenesis. Using Xenopus laevis as a model we here show for the first time that Dkk1 is required for proper differentiation of cardiomyocytes, whereas specification of cardiomyocytes remains unaffected in absence of Dkk1. This effect is at least in part mediated through regulation of non-canonical Wnt signaling via Wnt11. In line with these observations we also found that Isl1, a critical regulator for specification of the common cardiac progenitor cell (CPC) population, acts upstream of Dkk1. Dkk1 is required for cardiac development in Xenopus laevis. The Wnt inhibitor Dkk1 acts downstream of Isl1 during cardiac development in vivo. Loss of Dkk1 has no impact on cardiac specification in Xenopus. Normal cardiac differentiation is impaired upon Dkk1 inhibition in Xenopus. Dkk1 regulates canonical Wnt/β-catenin signaling during Xenopus cardiogenesis.
Collapse
Affiliation(s)
- Yanchun Guo
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; International Graduate School in Molecular Medicine Ulm, Ulm University, 89081 Ulm, Germany
| | - Tatjana Dorn
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar der Technischen Universität München, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Susanne J Kühl
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Alexander Linnemann
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Melanie Rothe
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; International Graduate School in Molecular Medicine Ulm, Ulm University, 89081 Ulm, Germany
| | - Astrid S Pfister
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Seppo Vainio
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, InfoTech Oulu, Oulu University and Biobank Borealis of Northern Finland, Oulu University Hospital, Aapistie 5, FIN-90014, University of Oulu, Finland
| | - Karl-Ludwig Laugwitz
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar der Technischen Universität München, Ismaninger Strasse 22, 81675 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - Partner Site Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar der Technischen Universität München, Ismaninger Strasse 22, 81675 Munich, Germany; DZHK (German Centre for Cardiovascular Research) - Partner Site Munich Heart Alliance, Munich, Germany.
| | - Michael Kühl
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
42
|
A novel high-content imaging-based technique for measuring binding of Dickkopf-1 to low-density lipoprotein receptor-related protein 6. J Pharmacol Toxicol Methods 2019; 95:47-55. [DOI: 10.1016/j.vascn.2018.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/09/2018] [Accepted: 11/21/2018] [Indexed: 01/05/2023]
|
43
|
Rasouli SJ, El-Brolosy M, Tsedeke AT, Bensimon-Brito A, Ghanbari P, Maischein HM, Kuenne C, Stainier DY. The flow responsive transcription factor Klf2 is required for myocardial wall integrity by modulating Fgf signaling. eLife 2018; 7:e38889. [PMID: 30592462 PMCID: PMC6329608 DOI: 10.7554/elife.38889] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 12/24/2018] [Indexed: 12/17/2022] Open
Abstract
Complex interplay between cardiac tissues is crucial for their integrity. The flow responsive transcription factor KLF2, which is expressed in the endocardium, is vital for cardiovascular development but its exact role remains to be defined. To this end, we mutated both klf2 paralogues in zebrafish, and while single mutants exhibit no obvious phenotype, double mutants display a novel phenotype of cardiomyocyte extrusion towards the abluminal side. This extrusion requires cardiac contractility and correlates with the mislocalization of N-cadherin from the lateral to the apical side of cardiomyocytes. Transgenic rescue data show that klf2 expression in endothelium, but not myocardium, prevents this cardiomyocyte extrusion phenotype. Transcriptome analysis of klf2 mutant hearts reveals that Fgf signaling is affected, and accordingly, we find that inhibition of Fgf signaling in wild-type animals can lead to abluminal cardiomyocyte extrusion. These studies provide new insights into how Klf2 regulates cardiovascular development and specifically myocardial wall integrity.
Collapse
Affiliation(s)
- Seyed Javad Rasouli
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Mohamed El-Brolosy
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Ayele Taddese Tsedeke
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Anabela Bensimon-Brito
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Parisa Ghanbari
- Department of Cardiac Development and RemodelingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Hans-Martin Maischein
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Carsten Kuenne
- Bioinformatics Core UnitMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier Y Stainier
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
44
|
Nandkishore N, Vyas B, Javali A, Ghosh S, Sambasivan R. Divergent early mesoderm specification underlies distinct head and trunk muscle programmes in vertebrates. Development 2018; 145:dev.160945. [PMID: 30237317 DOI: 10.1242/dev.160945] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 07/31/2018] [Indexed: 01/19/2023]
Abstract
Head and trunk muscles have discrete embryological origins and are governed by distinct regulatory programmes. Whereas the developmental route of trunk muscles from mesoderm is well studied, that of head muscles is ill defined. Here, we show that, unlike the myogenic trunk paraxial mesoderm, head mesoderm development is independent of the T/Tbx6 network in mouse. We reveal that, in contrast to Wnt and FGF-driven trunk mesoderm, dual inhibition of Wnt/β-catenin and Nodal specifies head mesoderm. Remarkably, the progenitors derived from embryonic stem cells by dual inhibition efficiently differentiate into cardiac and skeletal muscle cells. This twin potential is the defining feature of cardiopharyngeal mesoderm: the head subtype giving rise to heart and branchiomeric head muscles. Therefore, our findings provide compelling evidence that dual inhibition specifies head mesoderm and unravel the mechanism that diversifies head and trunk muscle programmes during early mesoderm fate commitment. Significantly, this is the first report of directed differentiation of pluripotent stem cells, without transgenes, into progenitors with muscle/heart dual potential. Ability to generate branchiomeric muscle in vitro could catalyse efforts in modelling myopathies that selectively involve head muscles.
Collapse
Affiliation(s)
- Nitya Nandkishore
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru 560065, India.,SASTRA University, Thirumalaisamudram, Thanjavur 613401, India
| | - Bhakti Vyas
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru 560065, India.,Manipal Academy of Higher Education, Manipal 576104, India
| | - Alok Javali
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru 560065, India.,National Centre for Biological Sciences, TIFR, GKVK Campus, Bellary Road, Bengaluru 560065, India
| | - Subho Ghosh
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru 560065, India
| | - Ramkumar Sambasivan
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru 560065, India
| |
Collapse
|
45
|
Janesick A, Tang W, Shioda T, Blumberg B. RARγ is required for mesodermal gene expression prior to gastrulation in Xenopus. Development 2018; 145:dev147769. [PMID: 30111657 DOI: 10.1242/dev.147769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 07/31/2018] [Indexed: 12/12/2022]
Abstract
The developing vertebrate embryo is exquisitely sensitive to retinoic acid (RA) concentration, particularly during anteroposterior patterning. In contrast to Nodal and Wnt signaling, RA was not previously considered to be an instructive signal in mesoderm formation during gastrulation. Here, we show in Xenopus that RARγ is indispensable for the expression of early mesoderm markers and is, therefore, an obligatory factor in mesodermal competence and/or maintenance. We identified several novel targets upregulated by RA receptor signaling in the early gastrula that are expressed in the circumblastoporal ring and linked to mesodermal development. Despite overlapping expression patterns of the genes encoding the RA-synthesizing enzyme Aldh1a2 and the RA-degrading enzyme Cyp26a1, RARγ1 functions as a transcriptional activator in early mesoderm development, suggesting that RA ligand is available to the embryo earlier than previously appreciated. RARγ1 is required for cellular adhesion, as revealed by spontaneous dissociation and depletion of ncam1 mRNA in animal caps harvested from RARγ1 knockdown embryos. RARγ1 knockdown obliterates somite boundaries, and causes loss of Myod protein in the presomitic mesoderm, but ectopic, persistent expression of Myod protein in the trunk. Thus, RARγ1 is required for stabilizing the mesodermal fate, myogenic commitment, somite boundary formation, and terminal skeletal muscle differentiation.
Collapse
Affiliation(s)
- Amanda Janesick
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, 92697-2300, USA
| | - Weiyi Tang
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, 92697-2300, USA
| | - Toshi Shioda
- Center for Cancer Research, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, MA 02129, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, 92697-2300, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697-2300, USA
| |
Collapse
|
46
|
Ming Z, Wang Y, Gong AY, Zhang XT, Li M, Chen T, Mathy NW, Strauss-Soukup JK, Chen XM. Attenuation of Intestinal Epithelial Cell Migration During Cryptosporidium parvum Infection Involves Parasite Cdg7_FLc_1030 RNA-Mediated Induction and Release of Dickkopf-1. J Infect Dis 2018; 218:1336-1347. [PMID: 30052999 PMCID: PMC6129111 DOI: 10.1093/infdis/jiy299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/17/2022] Open
Abstract
Intestinal infection by Cryptosporidium is known to cause epithelial cell migration disorder but the underlying mechanisms are unclear. Previous studies demonstrated that a panel of parasite RNA transcripts of low protein-coding potential are delivered into infected epithelial cells. Using multiple models of intestinal cryptosporidiosis, we report here that C. parvum infection induces expression and release of the dickkopf protein 1 (Dkk1) from intestinal epithelial cells. Delivery of parasite Cdg7_FLc_1030 RNA to intestinal epithelial cells triggers transactivation of host Dkk1 gene during C. parvum infection. Release of Dkk1 is involved in C. parvum-induced inhibition of cell migration of epithelial cells, including noninfected bystander cells. Moreover, Dkk1-mediated suppression of host cell migration during C. parvum infection involves inhibition of Cdc42/Par6 signaling. Our data support the hypothesis that attenuation of intestinal epithelial cell migration during Cryptosporidium infection involves parasite Cdg7_FLc_1030 RNA-mediated induction and release of Dkk1 from infected cells.
Collapse
Affiliation(s)
- Zhenping Ming
- Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| | - Yang Wang
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| | - Ai-Yu Gong
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| | - Xin-Tian Zhang
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| | - Min Li
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| | - Ting Chen
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
- Department of Gastroenterology, Hubei University of Science and Technology, Hubei, China
| | - Nicholas W Mathy
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| | | | - Xian-Ming Chen
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska
| |
Collapse
|
47
|
Insights from molecular signature of in vivo cardiac c-Kit(+) cells following cardiac injury and β-catenin inhibition. J Mol Cell Cardiol 2018; 123:64-74. [PMID: 30171847 DOI: 10.1016/j.yjmcc.2018.08.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/03/2018] [Accepted: 08/28/2018] [Indexed: 01/26/2023]
Abstract
There is much interest over resident c-Kit(+) cells in tissue regeneration. Their role in cardiac regeneration has been controversial. In this study we aim to understand the in vivo behavior of cardiac c-Kit(+) cells at baseline and after myocardial infarction and in response to Sfrp2. This approach can accurately study the in vivo transcript expressions of these cells in temporal response to injury and overcomes the limitations of the in vitro approach. RNA-seq was performed with c-Kit(+) cells and cardiomyocytes from healthy non-injured mice as well as c-Kit(+) cells from 1 day post-MI and 12 days post-MI mice. When compared to in vivo c-Kit(+) cells isolated from a healthy non-injured mouse heart, cardiomyocytes were enriched in transcripts that express anion channels, cation channels, developmental/differentiation pathway components, as well as proteins that inhibit canonical Wnt/β-catenin signaling. Myocardial infarction (MI) induced in vivo c-Kit(+) cells to transiently adopt the cardiomyocyte-specific signature: expression of a number of cardiomyocyte-specific transcripts was maximal 1 day post-MI and declined by 12 days post-MI. We next studied the effect of β-catenin inhibition on in vivo c-Kit(+) cells by administering the Wnt inhibitor Sfrp2 into the infarct border zone. Sfrp2 both enhanced and sustained cardiomyocyte-specific gene expression in the in vivo c-Kit(+) cells: expression of cardiomyocyte-specific transcripts was higher and there was no decline in expression by 12 days post-MI. Further analysis of the biology of c-Kit(+) cells identified that culture induced a significant and irreversible change in their molecular signature raising questions about reliability of in vitro studies. Our findings provide evidence that MI induces in vivo c-Kit(+) cells to adopt transiently a cardiomyocyte-specific pattern of gene expression, and Sfrp2 further enhances and induces sustained gene expression. Our approach is important for understanding c-Kit(+) cells in cardiac regeneration and also has broad implications in the investigation of in vivo resident stem cells in other areas of tissue regeneration.
Collapse
|
48
|
Kim BJ, Kim YH, Lee YA, Jung SE, Hong YH, Lee EJ, Kim BG, Hwang S, Do JT, Pang MG, Ryu BY. Platelet-derived growth factor receptor-alpha positive cardiac progenitor cells derived from multipotent germline stem cells are capable of cardiomyogenesis in vitro and in vivo. Oncotarget 2018; 8:29643-29656. [PMID: 28410244 PMCID: PMC5444692 DOI: 10.18632/oncotarget.16772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 02/28/2017] [Indexed: 01/12/2023] Open
Abstract
Cardiac cell therapy has the potential to revolutionize treatment of heart diseases, but its success hinders on the development of a stem cell therapy capable of efficiently producing functionally differentiated cardiomyocytes. A key to unlocking the therapeutic application of stem cells lies in understanding the molecular mechanisms that govern the differentiation process. Here we report that a population of platelet-derived growth factor receptor alpha (PDGFRA) cells derived from mouse multipotent germline stem cells (mGSCs) were capable of undergoing cardiomyogenesis in vitro. Cells derived in vitro from PDGFRA positive mGSCs express significantly higher levels of cardiac marker proteins compared to PDGFRA negative mGSCs. Using Pdgfra shRNAs to investigate the dependence of Pdgfra on cardiomyocyte differentiation, we observed that Pdgfra silencing inhibited cardiac differentiation. In a rat myocardial infarction (MI) model, transplantation of a PDGFRAenriched cell population into the rat heart readily underwent functional differentiation into cardiomyocytes and reduced areas of fibrosis associated with MI injury. Together, these results suggest that mGSCs may provide a unique source of cardiac stem/progenitor cells for future regenerative therapy of damaged heart tissue.
Collapse
Affiliation(s)
- Bang-Jin Kim
- Department of Animal Science & Technology, Chung-Ang University, Anseong, Republic of Korea.,Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yong-Hee Kim
- Department of Animal Science & Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Yong-An Lee
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Sang-Eun Jung
- Department of Animal Science & Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Yeong Ho Hong
- Department of Animal Science & Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Eun-Ju Lee
- Department of Internal medicine, Seoul National University, Seoul, Republic of Korea
| | - Byung-Gak Kim
- Bio Environment Technology Research Institute, Chung-Ang University, Anseong, Republic of Korea
| | - Seongsoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, Jeollabuk-do, Republic of Korea
| | - Jeong Tae Do
- Department of Stem Cell and Regenerative Biology, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| | - Myung-Geol Pang
- Department of Animal Science & Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Buom-Yong Ryu
- Department of Animal Science & Technology, Chung-Ang University, Anseong, Republic of Korea
| |
Collapse
|
49
|
Reichman DE, Park L, Man L, Redmond D, Chao K, Harvey RP, Taketo MM, Rosenwaks Z, James D. Wnt inhibition promotes vascular specification of embryonic cardiac progenitors. Development 2018; 145:dev.159905. [PMID: 29217753 PMCID: PMC5825863 DOI: 10.1242/dev.159905] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/26/2017] [Indexed: 01/29/2023]
Abstract
Several studies have demonstrated a multiphasic role for Wnt signaling during embryonic cardiogenesis and developed protocols that enrich for cardiac derivatives during in vitro differentiation of human pluripotent stem cells (hPSCs). However, few studies have investigated the role of Wnt signaling in the specification of cardiac progenitor cells (CPCs) toward downstream fates. Using transgenic mice and hPSCs, we tracked endothelial cells (ECs) that originated from CPCs expressing NKX2.5. Analysis of EC-fated CPCs at discrete phenotypic milestones during hPSC differentiation identified reduced Wnt activity as a hallmark of EC specification, and the enforced activation or inhibition of Wnt reduced or increased, respectively, the degree of vascular commitment within the CPC population during both hPSC differentiation and mouse embryogenesis. Wnt5a, which has been shown to exert an inhibitory influence on Wnt signaling during cardiac development, was dynamically expressed during vascular commitment of hPSC-derived CPCs, and ectopic Wnt5a promoted vascular specification of hPSC-derived and mouse embryonic CPCs.
Collapse
Affiliation(s)
- David E Reichman
- Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Laura Park
- Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Limor Man
- Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - David Redmond
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kenny Chao
- Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia.,School of Biological and Biomolecular Sciences, University of New South Wales, Kensington 2052, Australia
| | - Makoto M Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto 606-8501, Japan
| | - Zev Rosenwaks
- Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Daylon James
- Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065, USA .,Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
50
|
Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A, Blankesteijn WM. WNT Signaling in Cardiac and Vascular Disease. Pharmacol Rev 2018; 70:68-141. [PMID: 29247129 PMCID: PMC6040091 DOI: 10.1124/pr.117.013896] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-β and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.
Collapse
Affiliation(s)
- Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Gentian Lluri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Arjun Deb
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| |
Collapse
|