1
|
Inoue S, Nosetani M, Nakajima Y, Sakaki S, Kato H, Saba R, Takeshita N, Nishikawa K, Ueyama A, Matsuo K, Shigeta M, Kobayashi D, Iehara T, Yashiro K. Sonic Hedgehog signaling regulates the optimal differentiation pace from early-stage mesoderm to cardiogenic mesoderm in mice. Dev Growth Differ 2025; 67:75-84. [PMID: 39783159 PMCID: PMC11842887 DOI: 10.1111/dgd.12955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025]
Abstract
Sonic Hedgehog (Shh), encoding an extracellular signaling molecule, is vital for heart development. Shh null mutants show congenital heart disease due to left-right asymmetry defects stemming from functional anomaly in the midline structure in mice. Shh signaling is also known to affect cardiomyocyte differentiation, endocardium development, and heart morphogenesis, particularly in second heart field (SHF) cardiac progenitor cells that contribute to the right ventricle, outflow tract, and parts of the atrium. Despite extensive studies, our understanding remains incomplete. Notably, Shh signaling is suggested to promote cardiac differentiation, while paradoxically preventing premature differentiation of SHF progenitors. In this study, we elucidate the role of Shh signaling in the earliest phase of cardiac differentiation. Our meta-analysis of single-cell RNA sequencing suggests that cardiogenic nascent mesoderm cells expressing the bHLH transcription factor Mesp1 interact with axial mesoderm via Hh signaling. Activation of Hh signaling using a Smoothened agonist delayed or suppressed the differentiation of primitive streak cells expressing T-box transcription factor T to Mesp1+ nascent mesoderm cells both in vitro and ex vivo. Conversely, inhibition of Hh signaling by cyclopamine facilitated cardiac differentiation. The reduction of Eomes, an inducer of Mesp1, by Hh signaling appears to be the underlying mechanism of this phenomenon. Our data suggest that SHH secreted from axial mesoderm inhibits premature differentiation of T+ cells to Mesp1+ nascent mesoderm cells, thereby regulating the pace of cardiac differentiation. These findings enhance our comprehension of Shh signaling in cardiac development, underscoring its crucial role in early cardiac differentiation.
Collapse
Affiliation(s)
- Satoshi Inoue
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pediatrics, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Moe Nosetani
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yoshiro Nakajima
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Shinichiro Sakaki
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pediatrics, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroki Kato
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Rie Saba
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Radiology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Naoki Takeshita
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pediatrics, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kosuke Nishikawa
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pediatrics, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Atsuko Ueyama
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pediatrics, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Kazuhiko Matsuo
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masaki Shigeta
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Daisuke Kobayashi
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kenta Yashiro
- Division of Anatomy and Developmental Biology, Department of Anatomy, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
2
|
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)
| | - Janet L. Paluh
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science and Engineering, University at Albany, Albany, NY, United States
| |
Collapse
|
3
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2025; 60:62-84.e7. [PMID: 39413783 PMCID: PMC11706711 DOI: 10.1016/j.devcel.2024.09.017] [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: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
4
|
Zhao C, Plaza Reyes A, Schell JP, Weltner J, Ortega NM, Zheng Y, Björklund ÅK, Baqué-Vidal L, Sokka J, Trokovic R, Cox B, Rossant J, Fu J, Petropoulos S, Lanner F. A comprehensive human embryo reference tool using single-cell RNA-sequencing data. Nat Methods 2025; 22:193-206. [PMID: 39543283 PMCID: PMC11725501 DOI: 10.1038/s41592-024-02493-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024]
Abstract
Stem cell-based embryo models offer unprecedented experimental tools for studying early human development. The usefulness of embryo models hinges on their molecular, cellular and structural fidelities to their in vivo counterparts. To authenticate human embryo models, single-cell RNA sequencing has been utilized for unbiased transcriptional profiling. However, an organized and integrated human single-cell RNA-sequencing dataset, serving as a universal reference for benchmarking human embryo models, remains unavailable. Here we developed such a reference through the integration of six published human datasets covering development from the zygote to the gastrula. Lineage annotations are contrasted and validated with available human and nonhuman primate datasets. Using stabilized Uniform Manifold Approximation and Projection, we constructed an early embryogenesis prediction tool, where query datasets can be projected on the reference and annotated with predicted cell identities. Using this reference tool, we examined published human embryo models, highlighting the risk of misannotation when relevant references are not utilized for benchmarking and authentication.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Alvaro Plaza Reyes
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
- Department of Integrative Pathophysiology and Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Seville, Spain
| | - John Paul Schell
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Jere Weltner
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Nicolás M Ortega
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
| | - Åsa K Björklund
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Laura Baqué-Vidal
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Joonas Sokka
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Ras Trokovic
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Brian Cox
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sophie Petropoulos
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden.
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada.
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Axe Immunopathologie, Montreal, Quebec, Canada.
| | - Fredrik Lanner
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, and Division of Obstetrics and Gynecology, Karolinska Universitetssjukhuset, Stockholm, Sweden.
- Ming Wai Lau Center for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Yuri S, Arisawa N, Kitamuro K, Isotani A. Blastocyst complementation-based rat-derived heart generation reveals cardiac anomaly barriers to interspecies chimera development. iScience 2024; 27:111414. [PMID: 39687030 PMCID: PMC11647242 DOI: 10.1016/j.isci.2024.111414] [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: 04/15/2024] [Revised: 08/27/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
The use of pluripotent stem cells (PSCs) to generate functional organs via blastocyst complementation is a cutting-edge strategy in regenerative medicine. However, existing models that use this method for heart generation do not meet expectations owing to the complexity of heart development. Here, we investigated a Mesp1/2 deficient mouse model, which is characterized by abnormalities in the cardiac mesodermal cells. The injection of either mouse or rat PSCs into Mesp1/2 deficient mouse blastocysts led to successful heart generation. In chimeras, the resulting hearts were predominantly composed of rat cells; however, their functionality was limited to the embryonic developmental stage on day 12.5. These results present the functional limitation of the xenogeneic heart, which poses a significant challenge to the development in mouse-rat chimeras.
Collapse
Affiliation(s)
- Shunsuke Yuri
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan
- Laboratory of Experimental Animals, Research Institution, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
| | - Norie Arisawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan
| | - Kohei Kitamuro
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan
| | - Ayako Isotani
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan
- Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
6
|
Kokity L, Czimmerer Z, Benyhe-Kis B, Poscher A, Belai E, Steinbach G, Lipinszki Z, Pirity MK. Brachyury co-operates with polycomb protein RYBP to regulate gastrulation and axial elongation in vitro. Front Cell Dev Biol 2024; 12:1498346. [PMID: 39676794 PMCID: PMC11638158 DOI: 10.3389/fcell.2024.1498346] [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: 09/19/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Early embryonic development is a complex process where undifferentiated cells lose their pluripotency and start to gastrulate. During gastrulation, three germ layers form, giving rise to different cell lineages and organs. This process is regulated by transcription factors and epigenetic regulators, including non-canonical polycomb repressive complex 1s (ncPRC1s). Previously, we reported that ncPRC1-member RYBP (RING1 and YY1 binding protein) is crucial for embryonic implantation and cardiac lineage commitment in mice. However, the role of RYBP in gastrulation and mesoderm formation has not yet been defined. In this study, we used 2D and 3D in vitro model systems, to analyze the role of RYBP in mesoderm formation. First, we showed that cardiac and endothelial progenitors-both derived from mesoderm-are underrepresented in the Rybp -/- cardiac colonies. In the absence of RYBP, the formation of major germ layers was also disrupted, and the expression of mesoderm- (Brachyury, Eomes, and Gsc) and endoderm-specific (Sox17, Gata4) genes was significantly downregulated. Using 3D embryoid bodies as gastrulation models, we showed that RYBP can co-localize with mesoderm lineage marker protein BRACHYURY and endoderm marker protein GATA4 and both proteins. In mutants, both proteins were detected at low levels and showed altered distribution. Additionally, we compared our in vitro results to available in vivo single-cell transcriptomes and showed that Rybp and Brachyury co-expressed in the primitive streak and six mesodermal clusters. Since caudal mesoderm exhibited one of the strongest co-expressions, we tested axial elongation in wt and Rybp -/- gastruloids. In the absence of RYBP, gastruloids exhibited shortened tails and low BRACHYURY levels in the tailbud. Finally, we identified BRACHYURY as a novel binding partner of RYBP and presented evidence of possible cooperative function during mesoderm formation and axial elongation. Together, our results demonstrate the previously unknown role of RYBP in mesoderm formation. We believe our findings will contribute to better understanding of the highly conserved process of gastrulation.
Collapse
Affiliation(s)
- Lilla Kokity
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Zsolt Czimmerer
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
| | - Bernadett Benyhe-Kis
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Anna Poscher
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Emese Belai
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
- Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gábor Steinbach
- Cellular Imaging Laboratory, Core Facility, Biological Research Centre, Hungarian Research Network, Szeged, Hungary
| | - Zoltan Lipinszki
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Hungarian Research Network, Szeged, Hungary
- National Laboratory for Biotechnology, Institute of Genetics, Biological Research Centre, Hungarian Research Network, Szeged, Hungary
| | - Melinda Katalin Pirity
- Biological Research Centre, Institute of Genetics, Hungarian Research Network, Szeged, Hungary
| |
Collapse
|
7
|
Shaikh Qureshi WM, Hentges KE. Functions of cilia in cardiac development and disease. Ann Hum Genet 2024; 88:4-26. [PMID: 37872827 PMCID: PMC10952336 DOI: 10.1111/ahg.12534] [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: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023]
Abstract
Errors in embryonic cardiac development are a leading cause of congenital heart defects (CHDs), including morphological abnormalities of the heart that are often detected after birth. In the past few decades, an emerging role for cilia in the pathogenesis of CHD has been identified, but this topic still largely remains an unexplored area. Mouse forward genetic screens and whole exome sequencing analysis of CHD patients have identified enrichment for de novo mutations in ciliary genes or non-ciliary genes, which regulate cilia-related pathways, linking cilia function to aberrant cardiac development. Key events in cardiac morphogenesis, including left-right asymmetric development of the heart, are dependent upon cilia function. Cilia dysfunction during left-right axis formation contributes to CHD as evidenced by the substantial proportion of heterotaxy patients displaying complex CHD. Cilia-transduced signaling also regulates later events during heart development such as cardiac valve formation, outflow tract septation, ventricle development, and atrioventricular septa formation. In this review, we summarize the role of motile and non-motile (primary cilia) in cardiac asymmetry establishment and later events during heart development.
Collapse
Affiliation(s)
- Wasay Mohiuddin Shaikh Qureshi
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
8
|
Lin CY, Chang YM, Tseng HY, Shih YL, Yeh HH, Liao YR, Tang HH, Hsu CL, Chen CC, Yan YT, Kao CF. Epigenetic regulator RNF20 underlies temporal hierarchy of gene expression to regulate postnatal cardiomyocyte polarization. Cell Rep 2023; 42:113416. [PMID: 37967007 DOI: 10.1016/j.celrep.2023.113416] [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: 12/08/2022] [Revised: 09/19/2023] [Accepted: 10/25/2023] [Indexed: 11/17/2023] Open
Abstract
Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.
Collapse
Affiliation(s)
- Chia-Yeh Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsin-Yi Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yen-Ling Shih
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsiao-Hui Yeh
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - You-Rou Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Han-Hsuan Tang
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chia-Ling Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| |
Collapse
|
9
|
Shi L, Song H, Zhou B, Morrow BE. Crk/Crkl regulates early angiogenesis in mouse embryos by accelerating endothelial cell maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548782. [PMID: 37503032 PMCID: PMC10369973 DOI: 10.1101/2023.07.12.548782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Rationale Ubiquitously expressed cytoplasmic adaptors CRK and CRKL mediate multiple signaling pathways in mammalian embryogenesis. They are also associated with cardiovascular defects occurring in Miller-Dieker syndrome and 22q11.2 deletion syndrome, respectively. The embryonic mesoderm contributes to the formation of the cardiovascular system, yet the roles that Crk and Crkl play there are not understood on a single cell level. Objectives To determine functions of Crk and Crkl in the embryonic mesoderm during early mouse vascular development. Secondly, we will examine the molecular mechanisms responsible for early embryonic endothelial cell (EC) defects by performing single cell RNA-sequencing (scRNA-seq) and in vivo validation experiments. Methods and Results Inactivation of both Crk and Crkl together using Mesp1 Cre resulted embryonic lethality with severe vascular defects. Although vasculogenesis appeared normal, angiogenesis was disrupted both in the yolk sac and embryo proper, leading to disorganized vascular networks. We performed scRNA-seq of the Mesp1 Cre mesodermal lineage and found that there was upregulation of a great number of angiogenesis and cell migration related genes in ECs in the mutants, including NOTCH signaling genes such as Dll4 and Hey1 . Further bioinformatic analysis of EC subpopulations identified a relative increase in the number of more differentiated angiogenic ECs and decrease in EC progenitors. Consistent with this, we identified an expansion of Dll4 expressing cells within abnormal arteries, in vivo . Also, our bioinformatic data indicates that there is dysregulated expression of lineage genes that promote EC differentiation causing accelerated cell fate progression during EC differentiation. Conclusions Our results show that Crk and Crkl are crucial for regulating early embryonic angiogenesis. Combined inactivation of Crk/Crkl caused precocious EC maturation with an increase of atypical differentiated angiogenic ECs and failed vascular remodeling. This is in part due to increased NOTCH signaling and altered expression of cell migration genes.
Collapse
|
10
|
Tyser RCV. Formation of the Heart: Defining Cardiomyocyte Progenitors at Single-Cell Resolution. Curr Cardiol Rep 2023; 25:495-503. [PMID: 37119451 PMCID: PMC10188409 DOI: 10.1007/s11886-023-01880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE OF REVIEW Formation of the heart requires the coordinated addition of multiple progenitor sources which have undergone different pathways of specification and differentiation. In this review, I aim to put into context how recent studies defining cardiac progenitor heterogeneity build on our understanding of early heart development and also discuss the questions raised by this new insight. RECENT FINDINGS With the development of sequencing technologies and imaging approaches, it has been possible to define, at high temporal resolution, the molecular profile and anatomical location of cardiac progenitors at the single-cell level, during the formation of the mammalian heart. Given the recent progress in our understanding of early heart development and technical advances in high-resolution time-lapse imaging and lineage analysis, we are now in a position of great potential, allowing us to resolve heart formation at previously impossible levels of detail. Understanding how this essential organ forms not only addresses questions of fundamental biological significance but also provides a blueprint for strategies to both treat and model heart disease.
Collapse
Affiliation(s)
- Richard C V Tyser
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, CB2 0AW, UK.
| |
Collapse
|
11
|
Krup AL, Winchester SAB, Ranade SS, Agrawal A, Devine WP, Sinha T, Choudhary K, Dominguez MH, Thomas R, Black BL, Srivastava D, Bruneau BG. A Mesp1-dependent developmental breakpoint in transcriptional and epigenomic specification of early cardiac precursors. Development 2023; 150:dev201229. [PMID: 36994838 PMCID: PMC10259516 DOI: 10.1242/dev.201229] [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: 08/23/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Transcriptional networks governing cardiac precursor cell (CPC) specification are incompletely understood owing, in part, to limitations in distinguishing CPCs from non-cardiac mesoderm in early gastrulation. We leveraged detection of early cardiac lineage transgenes within a granular single-cell transcriptomic time course of mouse embryos to identify emerging CPCs and describe their transcriptional profiles. Mesp1, a transiently expressed mesodermal transcription factor, is canonically described as an early regulator of cardiac specification. However, we observed perdurance of CPC transgene-expressing cells in Mesp1 mutants, albeit mislocalized, prompting us to investigate the scope of the role of Mesp1 in CPC emergence and differentiation. Mesp1 mutant CPCs failed to robustly activate markers of cardiomyocyte maturity and crucial cardiac transcription factors, yet they exhibited transcriptional profiles resembling cardiac mesoderm progressing towards cardiomyocyte fates. Single-cell chromatin accessibility analysis defined a Mesp1-dependent developmental breakpoint in cardiac lineage progression at a shift from mesendoderm transcriptional networks to those necessary for cardiac patterning and morphogenesis. These results reveal Mesp1-independent aspects of early CPC specification and underscore a Mesp1-dependent regulatory landscape required for progression through cardiogenesis.
Collapse
Affiliation(s)
- Alexis Leigh Krup
- Biomedical Sciences Program, University of California, San Francisco, CA 94158, USA
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sarah A. B. Winchester
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sanjeev S. Ranade
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ayushi Agrawal
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - W. Patrick Devine
- Department of Pathology, University of California, San Francisco, CA 94158, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Krishna Choudhary
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Martin H. Dominguez
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
- Department of Medicine, Division of Cardiology, University of California, San Francisco, CA 94158, USA
- Cardiovascular Institute and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Reuben Thomas
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Brian L. Black
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Benoit G. Bruneau
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA
- Institute of Human Genetics, University of California, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
12
|
Doyle ME, Premathilake HU, Yao Q, Mazucanti CH, Egan JM. Physiology of the tongue with emphasis on taste transduction. Physiol Rev 2023; 103:1193-1246. [PMID: 36422992 PMCID: PMC9942923 DOI: 10.1152/physrev.00012.2022] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The tongue is a complex multifunctional organ that interacts and senses both interoceptively and exteroceptively. Although it is easily visible to almost all of us, it is relatively understudied and what is in the literature is often contradictory or is not comprehensively reported. The tongue is both a motor and a sensory organ: motor in that it is required for speech and mastication, and sensory in that it receives information to be relayed to the central nervous system pertaining to the safety and quality of the contents of the oral cavity. Additionally, the tongue and its taste apparatus form part of an innate immune surveillance system. For example, loss or alteration in taste perception can be an early indication of infection as became evident during the present global SARS-CoV-2 pandemic. Here, we particularly emphasize the latest updates in the mechanisms of taste perception, taste bud formation and adult taste bud renewal, and the presence and effects of hormones on taste perception, review the understudied lingual immune system with specific reference to SARS-CoV-2, discuss nascent work on tongue microbiome, as well as address the effect of systemic disease on tongue structure and function, especially in relation to taste.
Collapse
Affiliation(s)
- Máire E Doyle
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Hasitha U Premathilake
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Qin Yao
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Caio H Mazucanti
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M Egan
- Diabetes Section/Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
13
|
Hahner F, Moll F, Warwick T, Hebchen DM, Buchmann GK, Epah J, Abplanalp W, Schader T, Günther S, Gilsbach R, Brandes RP, Schröder K. Nox4 promotes endothelial differentiation through chromatin remodeling. Redox Biol 2022; 55:102381. [PMID: 35810713 PMCID: PMC9287364 DOI: 10.1016/j.redox.2022.102381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/20/2022] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Nox4 is a constitutively active NADPH oxidase that constantly produces low levels of H2O2. Thereby, Nox4 contributes to cell homeostasis and long-term processes, such as differentiation. The high expression of Nox4 seen in endothelial cells contrasts with the low abundance of Nox4 in stem cells, which are accordingly characterized by low levels of H2O2. We hypothesize that Nox4 is a major contributor to endothelial differentiation, is induced during the process of differentiation, and facilitates homeostasis of the resulting endothelial cells. OBJECTIVE To determine the role of No×4 in differentiation of murine inducible pluripotent stem cells (miPSC) into endothelial cells (ECs). METHODS AND RESULTS miPSC, generated from mouse embryonic wildtype (WT) and Nox4-/- fibroblasts, were differentiated into endothelial cells (miPSC-EC) by stimulation with BMP4 and VEGF. During this process, Nox4 expression increased and knockout of Nox4 prolonged the abundance of pluripotency markers, while expression of endothelial markers was delayed in differentiating Nox4-depleted iPSCs. Eventually, angiogenic capacity of iPSC-ECs is reduced in Nox4 deficient cells, indicating that an absence of Nox4 diminishes stability of the reached phenotype. As an underlying mechanism, we identified JmjD3 as a redox target of Nox4. iPSC-ECs lacking Nox4 display a lower nuclear abundance of the histone demethylase JmjD3, resulting in an increased triple methylation of histone 3 (H3K27me3), which serves as a repressive mark for several genes involved in differentiation. CONCLUSIONS Nox4 promotes differentiation of miPSCs into ECs by oxidation of JmjD3 and subsequent demethylation of H3K27me3, which forced endothelial differentiation and stability.
Collapse
Affiliation(s)
- F Hahner
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - F Moll
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - T Warwick
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - D M Hebchen
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - G K Buchmann
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - J Epah
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - W Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - T Schader
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - S Günther
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - R Gilsbach
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - R P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - K Schröder
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany.
| |
Collapse
|
14
|
Cheng S, Mittnenzweig M, Mayshar Y, Lifshitz A, Dunjić M, Rais Y, Ben-Yair R, Gehrs S, Chomsky E, Mukamel Z, Rubinstein H, Schlereth K, Reines N, Orenbuch AH, Tanay A, Stelzer Y. The intrinsic and extrinsic effects of TET proteins during gastrulation. Cell 2022; 185:3169-3185.e20. [PMID: 35908548 PMCID: PMC9432429 DOI: 10.1016/j.cell.2022.06.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/18/2022] [Accepted: 06/25/2022] [Indexed: 12/17/2022]
Abstract
Mice deficient for all ten-eleven translocation (TET) genes exhibit early gastrulation lethality. However, separating cause and effect in such embryonic failure is challenging. To isolate cell-autonomous effects of TET loss, we used temporal single-cell atlases from embryos with partial or complete mutant contributions. Strikingly, when developing within a wild-type embryo, Tet-mutant cells retain near-complete differentiation potential, whereas embryos solely comprising mutant cells are defective in epiblast to ectoderm transition with degenerated mesoderm potential. We map de-repressions of early epiblast factors (e.g., Dppa4 and Gdf3) and failure to activate multiple signaling from nascent mesoderm (Lefty, FGF, and Notch) as likely cell-intrinsic drivers of TET loss phenotypes. We further suggest loss of enhancer demethylation as the underlying mechanism. Collectively, our work demonstrates an unbiased approach for defining intrinsic and extrinsic embryonic gene function based on temporal differentiation atlases and disentangles the intracellular effects of the demethylation machinery from its broader tissue-level ramifications. Chimeras with full or partial Tet deficiency are mapped over the course of gastrulation Tet-TKO cells disrupt signaling, leading to skewed whole-embryo mutant gastrulation Tet-TKO cells retain near-complete differentiation potential in a chimera context Loss of TET leads to pervasive hypermethylation and mildly perturbed gene expression
Collapse
Affiliation(s)
- Saifeng Cheng
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Markus Mittnenzweig
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yoav Mayshar
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Aviezer Lifshitz
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Marko Dunjić
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yoach Rais
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Raz Ben-Yair
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Stephanie Gehrs
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Elad Chomsky
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Zohar Mukamel
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Hernan Rubinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Katharina Schlereth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Netta Reines
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | | | - Amos Tanay
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| | - Yonatan Stelzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
15
|
Mesp1 controls the chromatin and enhancer landscapes essential for spatiotemporal patterning of early cardiovascular progenitors. Nat Cell Biol 2022; 24:1114-1128. [PMID: 35817961 PMCID: PMC7613098 DOI: 10.1038/s41556-022-00947-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/25/2022] [Indexed: 01/13/2023]
Abstract
The mammalian heart arises from various populations of Mesp1-expressing cardiovascular progenitors (CPs) that are specified during the early stages of gastrulation. Mesp1 is a transcription factor that acts as a master regulator of CP specification and differentiation. However, how Mesp1 regulates the chromatin landscape of nascent mesodermal cells to define the temporal and spatial patterning of the distinct populations of CPs remains unknown. Here, by combining ChIP-seq, RNA-seq and ATAC-seq during mouse pluripotent stem cell differentiation, we defined the dynamic remodelling of the chromatin landscape mediated by Mesp1. We identified different enhancers that are temporally regulated to erase the pluripotent state and specify the pools of CPs that mediate heart development. We identified Zic2 and Zic3 as essential cofactors that act with Mesp1 to regulate its transcription-factor activity at key mesodermal enhancers, thereby regulating the chromatin remodelling and gene expression associated with the specification of the different populations of CPs in vivo. Our study identifies the dynamics of the chromatin landscape and enhancer remodelling associated with temporal patterning of early mesodermal cells into the distinct populations of CPs that mediate heart development.
Collapse
|
16
|
Gonzalez DM, Schrode N, Ebrahim TAM, Broguiere N, Rossi G, Drakhlis L, Zweigerdt R, Lutolf MP, Beaumont KG, Sebra R, Dubois NC. Dissecting mechanisms of chamber-specific cardiac differentiation and its perturbation following retinoic acid exposure. Development 2022; 149:275658. [DOI: 10.1242/dev.200557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/26/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The specification of distinct cardiac lineages occurs before chamber formation and acquisition of bona fide atrial or ventricular identity. However, the mechanisms underlying these early specification events remain poorly understood. Here, we performed single cell analysis at the murine cardiac crescent, primitive heart tube and heart tube stages to uncover the transcriptional mechanisms underlying formation of atrial and ventricular cells. We find that progression towards differentiated cardiomyocytes occurs primarily based on heart field progenitor identity, and that progenitors contribute to ventricular or atrial identity through distinct differentiation mechanisms. We identify new candidate markers that define such differentiation processes and examine their expression dynamics using computational lineage trajectory methods. We further show that exposure to exogenous retinoic acid causes defects in ventricular chamber size, dysregulation in FGF signaling and a shunt in differentiation towards orthogonal lineages. Retinoic acid also causes defects in cell-cycle exit resulting in formation of hypomorphic ventricles. Collectively, our data identify, at a single cell level, distinct lineage trajectories during cardiac specification and differentiation, and the precise effects of manipulating cardiac progenitor patterning via retinoic acid signaling.
Collapse
Affiliation(s)
- David M. Gonzalez
- Icahn School of Medicine at Mount Sinai 1 Department of Cell, Developmental, and Regenerative Biology , , New York, NY 10029 , USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai 2 , New York, NY 10029 , USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai 4 , New York, NY 10029 , USA
| | - Nadine Schrode
- Icahn School of Medicine at Mount Sinai 5 Department of Genetics and Genomic Sciences , , New York, NY 10029 , USA
| | - Tasneem A. M. Ebrahim
- Icahn School of Medicine at Mount Sinai 1 Department of Cell, Developmental, and Regenerative Biology , , New York, NY 10029 , USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai 2 , New York, NY 10029 , USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai 4 , New York, NY 10029 , USA
| | - Nicolas Broguiere
- School of Life Sciences, EPFL 6 Laboratory of Stem Cell Bioengineering , , Lausanne CH-1015 , Switzerland
| | - Giuliana Rossi
- School of Life Sciences, EPFL 6 Laboratory of Stem Cell Bioengineering , , Lausanne CH-1015 , Switzerland
| | - Lika Drakhlis
- Roche Institute for Translational Bioengineering 7 , Roche Pharma Research and Early Development , Basel 4052 , Switzerland
| | - Robert Zweigerdt
- Roche Institute for Translational Bioengineering 7 , Roche Pharma Research and Early Development , Basel 4052 , Switzerland
| | - Matthias P. Lutolf
- School of Life Sciences, EPFL 6 Laboratory of Stem Cell Bioengineering , , Lausanne CH-1015 , Switzerland
- Roche Institute for Translational Bioengineering 7 , Roche Pharma Research and Early Development , Basel 4052 , Switzerland
| | - Kristin G. Beaumont
- Icahn School of Medicine at Mount Sinai 5 Department of Genetics and Genomic Sciences , , New York, NY 10029 , USA
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO) 8 , Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG) , , Hannover , Germany
- REBIRTH–Research Center for Translational Regenerative Medicine, Hannover Medical School 8 , Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG) , , Hannover , Germany
| | - Robert Sebra
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Icahn School of Medicine at Mount Sinai 5 Department of Genetics and Genomic Sciences , , New York, NY 10029 , USA
- Sema4, a Mount Sinai venture 9 , Stamford, CT 06902 , USA
| | - Nicole C. Dubois
- Icahn School of Medicine at Mount Sinai 1 Department of Cell, Developmental, and Regenerative Biology , , New York, NY 10029 , USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai 2 , New York, NY 10029 , USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai 3 , New York, NY 10029 , USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai 4 , New York, NY 10029 , USA
| |
Collapse
|
17
|
Ramirez-Calderon G, Colombo G, Hernandez-Bautista CA, Astro V, Adamo A. Heart in a Dish: From Traditional 2D Differentiation Protocols to Cardiac Organoids. Front Cell Dev Biol 2022; 10:855966. [PMID: 35252213 PMCID: PMC8893312 DOI: 10.3389/fcell.2022.855966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) constitute a valuable model to study the complexity of early human cardiac development and investigate the molecular mechanisms involved in heart diseases. The differentiation of hPSCs into cardiac lineages in vitro can be achieved by traditional two-dimensional (2D) monolayer approaches or by adopting innovative three-dimensional (3D) cardiac organoid protocols. Human cardiac organoids (hCOs) are complex multicellular aggregates that faithfully recapitulate the cardiac tissue’s transcriptional, functional, and morphological features. In recent years, significant advances in the field have dramatically improved the robustness and efficiency of hCOs derivation and have promoted the application of hCOs for drug screening and heart disease modeling. This review surveys the current differentiation protocols, focusing on the most advanced 3D methods for deriving hCOs from hPSCs. Furthermore, we describe the potential applications of hCOs in the pharmaceutical and tissue bioengineering fields, including their usage to investigate the consequences of Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV2) infection in the heart.
Collapse
|
18
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
19
|
Ajima R, Sakakibara Y, Sakurai-Yamatani N, Muraoka M, Saga Y. Formal proof of the requirement of MESP1 and MESP2 in mesoderm specification and their transcriptional control via specific enhancers in mice. Development 2021; 148:272544. [PMID: 34679163 DOI: 10.1242/dev.194613] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/20/2021] [Indexed: 12/26/2022]
Abstract
MESP1 and MESP2 are transcriptional factors involved in mesoderm specification, somite boundary formation and somite polarity regulation. However, Mesp quadruple mutant zebrafish displayed only abnormal somite polarity without mesoderm specification defects. In order to re-evaluate Mesp1/Mesp2 mutants in mice, Mesp1 and Mesp2 single knockouts (KOs), and a Mesp1/Mesp2 double KO were established using genome-editing techniques without introducing selection markers commonly used before. The Mesp1/Mesp2 double KO embryos exhibited markedly severe mesoderm formation defects that were similar to the previously reported Mesp1/Mesp2 double KO embryos, indicating species differences in the function of MESP family proteins. However, the Mesp1 KO did not display any phenotype, including heart formation defects, which have been reported previously. We noted upregulation of Mesp2 in the Mesp1 KO embryos, suggesting that MESP2 rescues the loss of MESP1 in mesoderm specification. We also found that Mesp1 and Mesp2 expression in the early mesoderm is regulated by the cooperation of two independent enhancers containing T-box- and TCF/Lef-binding sites. Deletion of both enhancers caused the downregulation of both genes, resulting in heart formation defects. This study suggests dose-dependent roles of MESP1 and MESP2 in early mesoderm formation.
Collapse
Affiliation(s)
- Rieko Ajima
- Mammalian Development Laboratory, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Division for Development of Genetic-engineered Mouse Resource, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Yuko Sakakibara
- Division for Development of Genetic-engineered Mouse Resource, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Noriko Sakurai-Yamatani
- Division for Development of Genetic-engineered Mouse Resource, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Masafumi Muraoka
- Mammalian Development Laboratory, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Yumiko Saga
- Mammalian Development Laboratory, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Division for Development of Genetic-engineered Mouse Resource, Research Organization of Information and Systems (ROIS), Yata 1111, Mishima, Shizuoka 411-8540, Japan.,Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Yata 1111, Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
20
|
Han L, Yang L. Multidimensional Mechanistic Spectrum of Long Non-coding RNAs in Heart Development and Disease. Front Cardiovasc Med 2021; 8:728746. [PMID: 34604357 PMCID: PMC8483262 DOI: 10.3389/fcvm.2021.728746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
With the large-scale genome-wide sequencing, long non-coding RNAs (lncRNAs) have been found to compose of a large portion of the human transcriptome. Recent studies demonstrated the multidimensional functions of lncRNAs in heart development and disease. The subcellular localization of lncRNA is considered as a key factor that determines lncRNA function. Cytosolic lncRNAs mainly regulate mRNA stability, mRNA translation, miRNA processing and function, whereas nuclear lncRNAs epigenetically regulate chromatin remodeling, structure, and gene transcription. In this review, we summarize the molecular mechanisms of cytosolic and nuclear lncRNAs in heart development and disease separately, and emphasize the recent progress to dictate the crosstalk of cytosolic and nuclear lncRNAs in orchestrating the same biological process. Given the low evolutionary conservation of most lncRNAs, deeper understanding of human lncRNA will uncover a new layer of human regulatory mechanism underlying heart development and disease, and benefit the future clinical treatment for human heart disease.
Collapse
Affiliation(s)
- Lei Han
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lei Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
21
|
The Key Lnc (RNA)s in Cardiac and Skeletal Muscle Development, Regeneration, and Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8080084. [PMID: 34436226 PMCID: PMC8397000 DOI: 10.3390/jcdd8080084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/29/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
Non-coding RNAs (ncRNAs) play a key role in the regulation of transcriptional and epigenetic activity in mammalian cells. Comprehensive analysis of these ncRNAs has revealed sophisticated gene regulatory mechanisms which finely tune the proper gene output required for cellular homeostasis, proliferation, and differentiation. However, this elaborate circuitry has also made it vulnerable to perturbations that often result in disease. Among the many types of ncRNAs, long non-coding RNAs (lncRNAs) appear to have the most diverse mechanisms of action including competitive binding to miRNA targets, direct binding to mRNA, interactions with transcription factors, and facilitation of epigenetic modifications. Moreover, many lncRNAs display tissue-specific expression patterns suggesting an important regulatory role in organogenesis, yet the molecular mechanisms through which these molecules regulate cardiac and skeletal muscle development remains surprisingly limited. Given the structural and metabolic similarities of cardiac and skeletal muscle, it is likely that several lncRNAs expressed in both of these tissues have conserved functions in establishing the striated muscle phenotype. As many aspects of regeneration recapitulate development, understanding the role lncRNAs play in these processes may provide novel insights to improve regenerative therapeutic interventions in cardiac and skeletal muscle diseases. This review highlights key lncRNAs that function as regulators of development, regeneration, and disease in cardiac and skeletal muscle. Finally, we highlight lncRNAs encoded by imprinted genes in striated muscle and the contributions of these loci on the regulation of gene expression.
Collapse
|
22
|
Rossi G, Broguiere N, Miyamoto M, Boni A, Guiet R, Girgin M, Kelly RG, Kwon C, Lutolf MP. Capturing Cardiogenesis in Gastruloids. Cell Stem Cell 2020; 28:230-240.e6. [PMID: 33176168 DOI: 10.1016/j.stem.2020.10.013] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023]
Abstract
Organoids are powerful models for studying tissue development, physiology, and disease. However, current culture systems disrupt the inductive tissue-tissue interactions needed for the complex morphogenetic processes of native organogenesis. Here, we show that mouse embryonic stem cells (mESCs) can be coaxed to robustly undergo fundamental steps of early heart organogenesis with an in-vivo-like spatiotemporal fidelity. These axially patterned embryonic organoids (gastruloids) mimic embryonic development and support the generation of cardiovascular progenitors, including first and second heart fields. The cardiac progenitors self-organize into an anterior domain reminiscent of a cardiac crescent before forming a beating cardiac tissue near a putative primitive gut-like tube, from which it is separated by an endocardial-like layer. These findings unveil the surprising morphogenetic potential of mESCs to execute key aspects of organogenesis through the coordinated development of multiple tissues. This platform could be an excellent tool for studying heart development in unprecedented detail and throughput.
Collapse
Affiliation(s)
- Giuliana Rossi
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015 Vaud, Switzerland
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015 Vaud, Switzerland
| | - Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrea Boni
- Viventis Microscopy Sàrl, EPFL Innovation Park, Building C, Lausanne, 1015 Vaud, Switzerland
| | - Romain Guiet
- Faculté des Sciences de la Vie, Bioimaging and Optics Platform, École Polytechnique Fédérale de Lausanne (EPFL), Bâtiment AI, Station 15, Lausanne, 1015 Vaud, Switzerland
| | - Mehmet Girgin
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015 Vaud, Switzerland
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015 Vaud, Switzerland; Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015 Vaud, Switzerland.
| |
Collapse
|
23
|
Kidwai F, Mui BWH, Arora D, Iqbal K, Hockaday M, de Castro Diaz LF, Cherman N, Martin D, Myneni VD, Ahmad M, Futrega K, Ali S, Merling RK, Kaufman DS, Lee J, Robey PG. Lineage-specific differentiation of osteogenic progenitors from pluripotent stem cells reveals the FGF1-RUNX2 association in neural crest-derived osteoprogenitors. Stem Cells 2020; 38:1107-1123. [PMID: 32442326 PMCID: PMC7484058 DOI: 10.1002/stem.3206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (hPSCs) can provide a platform to model bone organogenesis and disease. To reflect the developmental process of the human skeleton, hPSC differentiation methods should include osteogenic progenitors (OPs) arising from three distinct embryonic lineages: the paraxial mesoderm, lateral plate mesoderm, and neural crest. Although OP differentiation protocols have been developed, the lineage from which they are derived, as well as characterization of their genetic and molecular differences, has not been well reported. Therefore, to generate lineage-specific OPs from human embryonic stem cells and human induced pluripotent stem cells, we employed stepwise differentiation of paraxial mesoderm-like cells, lateral plate mesoderm-like cells, and neural crest-like cells toward their respective OP subpopulation. Successful differentiation, confirmed through gene expression and in vivo assays, permitted the identification of transcriptomic signatures of all three cell populations. We also report, for the first time, high FGF1 levels in neural crest-derived OPs-a notable finding given the critical role of fibroblast growth factors (FGFs) in osteogenesis and mineral homeostasis. Our results indicate that FGF1 influences RUNX2 levels, with concomitant changes in ERK1/2 signaling. Overall, our study further validates hPSCs' power to model bone development and disease and reveals new, potentially important pathways influencing these processes.
Collapse
Affiliation(s)
- Fahad Kidwai
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Byron W. H. Mui
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Deepika Arora
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
- Biosystems and Biomaterials DivisionNational Institute of Standards and TechnologyGaithersburgMarylandUSA
| | - Kulsum Iqbal
- Department of Health and Human ServicesDental Consult Services, National Institute of Health Dental ClinicBethesdaMarylandUSA
| | - Madison Hockaday
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Luis Fernandez de Castro Diaz
- Department of Health and Human ServicesSkeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Natasha Cherman
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Daniel Martin
- Department of Health and Human ServicesGenomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Vamsee D. Myneni
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch/Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Moaz Ahmad
- Department of Health and Human ServicesMolecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Katarzyna Futrega
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Sania Ali
- Biology of Global Health, Department of BiologyGeorgetown UniversityWashingtonDistrict of ColumbiaUSA
| | - Randall K. Merling
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Dan S. Kaufman
- Department of MedicineUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Janice Lee
- Department of Health and Human ServicesCraniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| | - Pamela G. Robey
- Department of Health and Human ServicesCraniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
24
|
Liu J, Liu S, Gao H, Han L, Chu X, Sheng Y, Shou W, Wang Y, Liu Y, Wan J, Yang L. Genome-wide studies reveal the essential and opposite roles of ARID1A in controlling human cardiogenesis and neurogenesis from pluripotent stem cells. Genome Biol 2020; 21:169. [PMID: 32646524 PMCID: PMC7350744 DOI: 10.1186/s13059-020-02082-4] [Citation(s) in RCA: 27] [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: 03/16/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Early human heart and brain development simultaneously occur during embryogenesis. Notably, in human newborns, congenital heart defects strongly associate with neurodevelopmental abnormalities, suggesting a common gene or complex underlying both cardiogenesis and neurogenesis. However, due to lack of in vivo studies, the molecular mechanisms that govern both early human heart and brain development remain elusive. RESULTS Here, we report ARID1A, a DNA-binding subunit of the SWI/SNF epigenetic complex, controls both neurogenesis and cardiogenesis from human embryonic stem cells (hESCs) through distinct mechanisms. Knockout-of-ARID1A (ARID1A-/-) leads to spontaneous differentiation of neural cells together with globally enhanced expression of neurogenic genes in undifferentiated hESCs. Additionally, when compared with WT hESCs, cardiac differentiation from ARID1A -/- hESCs is prominently suppressed, whereas neural differentiation is significantly promoted. Whole genome-wide scRNA-seq, ATAC-seq, and ChIP-seq analyses reveal that ARID1A is required to open chromatin accessibility on promoters of essential cardiogenic genes, and temporally associated with key cardiogenic transcriptional factors T and MEF2C during early cardiac development. However, during early neural development, transcription of most essential neurogenic genes is dependent on ARID1A, which can interact with a known neural restrictive silencer factor REST/NRSF. CONCLUSIONS We uncover the opposite roles by ARID1A to govern both early cardiac and neural development from pluripotent stem cells. Global chromatin accessibility on cardiogenic genes is dependent on ARID1A, whereas transcriptional activity of neurogenic genes is under control by ARID1A, possibly through ARID1A-REST/NRSF interaction.
Collapse
Affiliation(s)
- Juli Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lei Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yi Sheng
- Department of Obstetrics, Gynecology & Reproductive Sciences, Magee-Women's Research Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W Walnut Street, R4 272, Indianapolis, IN, 46202, USA.
| |
Collapse
|
25
|
Wu T, Kamikawa YF, Donohoe ME. Brd4's Bromodomains Mediate Histone H3 Acetylation and Chromatin Remodeling in Pluripotent Cells through P300 and Brg1. Cell Rep 2019; 25:1756-1771. [PMID: 30428346 DOI: 10.1016/j.celrep.2018.10.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/10/2016] [Accepted: 09/28/2018] [Indexed: 12/22/2022] Open
Abstract
The pluripotent state of embryonic stem cells (ESCs) is defined by its transcriptome and epigenome. The chromatin reader Brd4 determines ESC identity. Although Brd4 regulation in gene transcription has been well described, its contribution to the chromatin landscape is less known. Here, we show that Brd4's bromodomains partner with the histone acetyltransferase P300, increasing its enzymatic activities. Augmenting histone acetylation by Brd4-P300 interaction recruits the chromatin remodeler Brg1 altering chromatin structure. This pathway is important for maintaining the expression and chromatin patterns of pluripotency-associated genes, such as Oct4, Nanog, and the X chromosome regulatory long noncoding RNAs Tsix and Xite. Furthermore, we show that the Brd4-P300 interaction regulates the de novo formation of chromatin marks during ESC differentiation, as exemplified by controlling the master regulators of mesoderm formation. Collectively, we delineate the function of Brd4 in organizing the chromatin structure that contributes to gene transcriptional regulation and cell fate determination.
Collapse
Affiliation(s)
- Tao Wu
- Burke Medical Research Institute, White Plains, NY 10605, USA; Department of Neuroscience, Brain Mind Research Institute, Department of Cell & Development, Weill Cornell Medical College, New York, NY 10065, USA
| | - Yasunao F Kamikawa
- Burke Medical Research Institute, White Plains, NY 10605, USA; Department of Neuroscience, Brain Mind Research Institute, Department of Cell & Development, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mary E Donohoe
- Burke Medical Research Institute, White Plains, NY 10605, USA; Department of Neuroscience, Brain Mind Research Institute, Department of Cell & Development, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
26
|
Ma H, Zhai J, Wan H, Jiang X, Wang X, Wang L, Xiang Y, He X, Zhao ZA, Zhao B, Zheng P, Li L, Wang H. In vitro culture of cynomolgus monkey embryos beyond early gastrulation. Science 2019; 366:science.aax7890. [DOI: 10.1126/science.aax7890] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Gastrulation is a key event in embryonic development when the germ layers are specified and the basic animal body plan is established. The complexities of primate gastrulation remain a mystery because of the difficulties in accessing primate embryos at this stage. Here, we report the establishment of an in vitro culture (IVC) system that supports the continuous development of cynomolgus monkey blastocysts beyond early gastrulation up to 20 days after fertilization. The IVC embryos highly recapitulated the key events of in vivo early postimplantation development, including segregation of the epiblast and hypoblast, formation of the amniotic and yolk sac cavities, appearance of the primordial germ cells, and establishment of the anterior-posterior axis. Single-cell RNA-sequencing analyses of the IVC embryos provide information about lineage specification during primate early postimplantation development. This system provides a platform with which to explore the characteristics and mechanisms of early postimplantation embryogenesis in primates with possible conservation of cell movements and lineages in human embryogenesis.
Collapse
Affiliation(s)
- Huaixiao Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Jinglei Zhai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Haifeng Wan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiangxiang Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoxiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Yunlong Xiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiechao He
- Primate Research Center, Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Zhen-Ao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Zhao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Ping Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Primate Research Center, Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
Penaloza JS, Pappas MP, Hagen HR, Xie N, Chan SSK. Single-cell RNA-seq analysis of Mesp1-induced skeletal myogenic development. Biochem Biophys Res Commun 2019; 520:284-290. [PMID: 31590918 DOI: 10.1016/j.bbrc.2019.09.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Abstract
The Mesp1 lineage contributes to cardiac, hematopoietic and skeletal myogenic development. Interestingly, muscle stem cells residing in craniofacial skeletal muscles primarily arise from Mesp1+ progenitors, but those in trunk and limb skeletal muscles do not. To gain insights into the difference between the head and trunk/limb muscle developmental processes, we studied Mesp1+ skeletal myogenic derivatives via single-cell RNA-seq and other strategies. Using a doxycycline-inducible Mesp1-expressing mouse embryonic stem cell line, we found that the development of Mesp1-induced skeletal myogenic progenitors can be characterized by dynamic expression of PDGFRα and VCAM1. Single-cell RNA-seq analysis further revealed the heterogeneous nature of these Mesp1+ derivatives, spanning pluripotent and mesodermal to mesenchymal and skeletal myogenic. We subsequently reconstructed the single-cell trajectories of these subpopulations. Our data thereby provide a cell fate projection of Mesp1-induced skeletal myogenesis.
Collapse
Affiliation(s)
| | | | | | - Ning Xie
- Department of Pediatrics, Minneapolis, MN, 55455, USA.
| | - Sunny S K Chan
- Department of Pediatrics, Minneapolis, MN, 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
28
|
Abstract
Cardiogenesis is a complex developmental process involving multiple overlapping stages of cell fate specification, proliferation, differentiation, and morphogenesis. Precise spatiotemporal coordination between the different cardiogenic processes is ensured by intercellular signalling crosstalk and tissue-tissue interactions. Notch is an intercellular signalling pathway crucial for cell fate decisions during multicellular organismal development and is aptly positioned to coordinate the complex signalling crosstalk required for progressive cell lineage restriction during cardiogenesis. In this Review, we describe the role of Notch signalling and the crosstalk with other signalling pathways during the differentiation and patterning of the different cardiac tissues and in cardiac valve and ventricular chamber development. We examine how perturbation of Notch signalling activity is linked to congenital heart diseases affecting the neonate and adult, and discuss studies that shed light on the role of Notch signalling in heart regeneration and repair after injury.
Collapse
|
29
|
Ritter N, Ali T, Kopitchinski N, Schuster P, Beisaw A, Hendrix DA, Schulz MH, Müller-McNicoll M, Dimmeler S, Grote P. The lncRNA Locus Handsdown Regulates Cardiac Gene Programs and Is Essential for Early Mouse Development. Dev Cell 2019; 50:644-657.e8. [PMID: 31422919 DOI: 10.1016/j.devcel.2019.07.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 05/10/2019] [Accepted: 07/10/2019] [Indexed: 11/29/2022]
Abstract
Precisely controlled gene regulatory networks are required during embryonic development to give rise to various structures, including those of the cardiovascular system. Long non-coding RNA (lncRNA) loci are known to be important regulators of these genetic programs. We have identified a novel and essential lncRNA locus Handsdown (Hdn), active in early heart cells, and show by genetic inactivation that it is essential for murine development. Hdn displays haploinsufficiency for cardiac development as Hdn-heterozygous adult mice exhibit hyperplasia in the right ventricular wall. Transcriptional activity of the Hdn locus, independent of its RNA, suppresses its neighboring gene Hand2. We reveal a switch in a topologically associated domain in differentiation of the cardiac lineage, allowing the Hdn locus to directly interact with regulatory elements of the Hand2 locus.
Collapse
Affiliation(s)
- Nicole Ritter
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tamer Ali
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Faculty of Science, Benha University, Benha 13518, Egypt
| | - Nina Kopitchinski
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Peggy Schuster
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Arica Beisaw
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - David A Hendrix
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA; School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR, USA
| | - Marcel H Schulz
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; German Center for Cardiovascular Regeneration (DZHK), Partner site Rhein-Main, 60590 Frankfurt am Main, Germany
| | - Michaela Müller-McNicoll
- RNA Regulation Group, Institute of Cell Biology and Neuroscience, Goethe University, Max-von-Laue-Strasse 13, 60438 Frankfurt am Main, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; German Center for Cardiovascular Regeneration (DZHK), Partner site Rhein-Main, 60590 Frankfurt am Main, Germany
| | - Phillip Grote
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
30
|
Yang N, Wu N, Zhang L, Zhao Y, Liu J, Liang X, Ren X, Li W, Chen W, Dong S, Zhao S, Lin J, Xiang H, Xue H, Chen L, Sun H, Zhang J, Shi J, Zhang S, Lu D, Wu X, Jin L, Ding J, Qiu G, Wu Z, Lupski JR, Zhang F. TBX6 compound inheritance leads to congenital vertebral malformations in humans and mice. Hum Mol Genet 2019; 28:539-547. [PMID: 30307510 DOI: 10.1093/hmg/ddy358] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022] Open
Abstract
Congenital vertebral malformations (CVMs) are associated with human TBX6 compound inheritance that combines a rare null allele and a common hypomorphic allele at the TBX6 locus. Our previous in vitro evidence suggested that this compound inheritance resulted in a TBX6 gene dosage of less than haploinsufficiency (i.e. <50%) as a potential mechanism of TBX6-associated CVMs. To further investigate this pathogenetic model, we ascertained and collected 108 Chinese CVM cases and found that 10 (9.3%) of them carried TBX6 null mutations in combination with common hypomorphic variants at the second TBX6 allele. For in vivo functional verification and genetic analysis of TBX6 compound inheritance, we generated both null and hypomorphic mutations in mouse Tbx6 using the CRISPR-Cas9 method. These Tbx6 mutants are not identical to the patient variants at the DNA sequence level, but instead functionally mimic disease-associated TBX6 variants. Intriguingly, as anticipated by the compound inheritance model, a high penetrance of CVM phenotype was only observed in the mice with combined null and hypomorphic alleles of Tbx6. These findings are consistent with our experimental observations in humans and supported the dosage effect of TBX6 in CVM etiology. In conclusion, our findings in the newly collected human CVM subjects and Tbx6 mouse models consistently support the contention that TBX6 compound inheritance causes CVMs, potentially via a gene dosage-dependent mechanism. Furthermore, mouse Tbx6 mutants mimicking human CVM-associated variants will be useful models for further mechanistic investigations of CVM pathogenesis in the cases associated with TBX6.
Collapse
Affiliation(s)
- Nan Yang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Nan Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, China.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, USA
| | - Ling Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Yanxue Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Jiaqi Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Xiangyu Liang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China
| | - Xiaojun Ren
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Weiyu Li
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Weisheng Chen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Shuangshuang Dong
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Sen Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Jiachen Lin
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Hang Xiang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lu Chen
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Hao Sun
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguo Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiangang Shi
- Second Department of Spine Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Shuyang Zhang
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Daru Lu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaohui Wu
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Institute of Developmental Biology and Molecular Medicine, Fudan University, Shanghai, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Department of Central Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
31
|
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
|
32
|
Chagraoui H, Kristiansen MS, Ruiz JP, Serra-Barros A, Richter J, Hall-Ponselé E, Gray N, Waithe D, Clark K, Hublitz P, Repapi E, Otto G, Sopp P, Taylor S, Thongjuea S, Vyas P, Porcher C. SCL/TAL1 cooperates with Polycomb RYBP-PRC1 to suppress alternative lineages in blood-fated cells. Nat Commun 2018; 9:5375. [PMID: 30560907 PMCID: PMC6299140 DOI: 10.1038/s41467-018-07787-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 11/23/2018] [Indexed: 01/03/2023] Open
Abstract
During development, it is unclear if lineage-fated cells derive from multilineage-primed progenitors and whether active mechanisms operate to restrict cell fate. Here we investigate how mesoderm specifies into blood-fated cells. We document temporally restricted co-expression of blood (Scl/Tal1), cardiac (Mesp1) and paraxial (Tbx6) lineage-affiliated transcription factors in single cells, at the onset of blood specification, supporting the existence of common progenitors. At the same time-restricted stage, absence of SCL results in expansion of cardiac/paraxial cell populations and increased cardiac/paraxial gene expression, suggesting active suppression of alternative fates. Indeed, SCL normally activates expression of co-repressor ETO2 and Polycomb-PRC1 subunits (RYBP, PCGF5) and maintains levels of Polycomb-associated histone marks (H2AK119ub/H3K27me3). Genome-wide analyses reveal ETO2 and RYBP co-occupy most SCL target genes, including cardiac/paraxial loci. Reduction of Eto2 or Rybp expression mimics Scl-null cardiac phenotype. Therefore, SCL-mediated transcriptional repression prevents mis-specification of blood-fated cells, establishing active repression as central to fate determination processes.
Collapse
Affiliation(s)
- Hedia Chagraoui
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Maiken S Kristiansen
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Medimmune, Granta Park, CB21 6GH, Cambridge, UK
| | - Juan Pablo Ruiz
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Haematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ana Serra-Barros
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Johanna Richter
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Elisa Hall-Ponselé
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- MRC Centre for Regenerative Medicine, SCRM Building, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Nicki Gray
- Computational Biology Research Group, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Dominic Waithe
- Wolfson Imaging Centre, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Kevin Clark
- FACS Facility, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Philip Hublitz
- Genome Engineering Facility, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Emmanouela Repapi
- Computational Biology Research Group, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Georg Otto
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Oxford National Institute for Health Research, Biomedical Research Centre, Haematology Theme, Oxford University Hospital, Oxford, OX3 9DU, UK
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Paul Sopp
- FACS Facility, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Stephen Taylor
- Computational Biology Research Group, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Supat Thongjuea
- Computational Biology Research Group, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Oxford National Institute for Health Research, Biomedical Research Centre, Haematology Theme, Oxford University Hospital, Oxford, OX3 9DU, UK
| | - Paresh Vyas
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- Oxford National Institute for Health Research, Biomedical Research Centre, Haematology Theme, Oxford University Hospital, Oxford, OX3 9DU, UK
| | - Catherine Porcher
- Medical Research Council Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.
| |
Collapse
|
33
|
Heude E, Tesarova M, Sefton EM, Jullian E, Adachi N, Grimaldi A, Zikmund T, Kaiser J, Kardon G, Kelly RG, Tajbakhsh S. Unique morphogenetic signatures define mammalian neck muscles and associated connective tissues. eLife 2018; 7:40179. [PMID: 30451684 PMCID: PMC6310459 DOI: 10.7554/elife.40179] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/17/2018] [Indexed: 12/16/2022] Open
Abstract
In vertebrates, head and trunk muscles develop from different mesodermal populations and are regulated by distinct genetic networks. Neck muscles at the head-trunk interface remain poorly defined due to their complex morphogenesis and dual mesodermal origins. Here, we use genetically modified mice to establish a 3D model that integrates regulatory genes, cell populations and morphogenetic events that define this transition zone. We show that the evolutionary conserved cucullaris-derived muscles originate from posterior cardiopharyngeal mesoderm, not lateral plate mesoderm, and we define new boundaries for neural crest and mesodermal contributions to neck connective tissue. Furthermore, lineage studies and functional analysis of Tbx1- and Pax3-null mice reveal a unique developmental program for somitic neck muscles that is distinct from that of somitic trunk muscles. Our findings unveil the embryological and developmental requirements underlying tetrapod neck myogenesis and provide a blueprint to investigate how muscle subsets are selectively affected in some human myopathies.
Collapse
Affiliation(s)
- Eglantine Heude
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Elizabeth M Sefton
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Estelle Jullian
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Alexandre Grimaldi
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS UMR 3738, Paris, France
| |
Collapse
|
34
|
Koyano-Nakagawa N, Garry DJ. Etv2 as an essential regulator of mesodermal lineage development. Cardiovasc Res 2018; 113:1294-1306. [PMID: 28859300 DOI: 10.1093/cvr/cvx133] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/24/2017] [Indexed: 11/14/2022] Open
Abstract
The 'master regulatory factors' that position at the top of the genetic hierarchy of lineage determination have been a focus of intense interest, and have been investigated in various systems. Etv2/Etsrp71/ER71 is such a factor that is both necessary and sufficient for the development of haematopoietic and endothelial lineages. As such, genetic ablation of Etv2 leads to complete loss of blood and vessels, and overexpression can convert non-endothelial cells to the endothelial lineage. Understanding such master regulatory role of a lineage is not only a fundamental quest in developmental biology, but also holds immense possibilities in regenerative medicine. To harness its activity and utility for therapeutic interventions, it is essential to understand the regulatory mechanisms, molecular function, and networks that surround Etv2. In this review, we provide a comprehensive overview of Etv2 biology focused on mouse and human systems.
Collapse
Affiliation(s)
- Naoko Koyano-Nakagawa
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 2231 6th st. SE, Minneapolis, MN 55455, USA
| | - Daniel J Garry
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 2231 6th st. SE, Minneapolis, MN 55455, USA
| |
Collapse
|
35
|
Guo X, Xu Y, Wang Z, Wu Y, Chen J, Wang G, Lu C, Jia W, Xi J, Zhu S, Jiapaer Z, Wan X, Liu Z, Gao S, Kang J. A Linc1405/Eomes Complex Promotes Cardiac Mesoderm Specification and Cardiogenesis. Cell Stem Cell 2018; 22:893-908.e6. [PMID: 29754779 DOI: 10.1016/j.stem.2018.04.013] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/07/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022]
Abstract
Large intergenic non-coding RNAs (lincRNAs) play widespread roles in epigenetic regulation during multiple differentiation processes, but little is known about their mode of action in cardiac differentiation. Here, we identified the key roles of a lincRNA, termed linc1405, in modulating the core network of cardiac differentiation by functionally interacting with Eomes. Chromatin- and RNA-immunoprecipitation assays showed that exon 2 of linc1405 physically mediates a complex consisting of Eomes, trithorax group (TrxG) subunit WDR5, and histone acetyltransferase GCN5 binding at the enhancer region of Mesp1 gene and activates its expression during cardiac mesoderm specification of embryonic stem cells. Importantly, linc1405 co-localizes with Eomes, WDR5, and GCN5 at the primitive streak, and linc1405 depletion impairs heart development and function in vivo. In summary, linc1405 mediates a Eomes/WDR5/GCN5 complex that contributes to cardiogenesis, highlighting the critical roles of lincRNA-based complexes in the epigenetic regulation of cardiogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Institute of Regenerative Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yanxin Xu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zikang Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yukang Wu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiayu Chen
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Guiying Wang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Chenqi Lu
- Department of Biostatistics and Computational Biology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Wenwen Jia
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiajie Xi
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Songcheng Zhu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zeyidan Jiapaer
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Zhongmin Liu
- Institute of Regenerative Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
36
|
Abstract
During mammalian heart development, restricted myocardial Bmp2 expression is a key patterning signal for atrioventricular canal specification and the epithelial-mesenchyme transition that gives rise to the valves. Using a mouse transgenic line conditionally expressing Bmp2, we show that widespread Bmp2 expression in the myocardium leads to valve and chamber dysmorphogenesis and embryonic death by E15.5. Transgenic embryos show thickened valves, ventricular septal defect, enlarged trabeculae and dilated ventricles, with an endocardium able to undergo EMT both in vivo and in vitro. Gene profiling and marker analysis indicate that cellular proliferation is increased in transgenic embryos, whereas chamber maturation and patterning are impaired. Similarly, forced Bmp2 expression stimulates proliferation and blocks cardiomyocyte differentiation of embryoid bodies. These data show that widespread myocardial Bmp2 expression directs ectopic valve primordium formation and maintains ventricular myocardium and cardiac progenitors in a primitive, proliferative state, identifying the potential of Bmp2 in the expansion of immature cardiomyocytes.
Collapse
|
37
|
Ivanovitch K, Temiño S, Torres M. Live imaging of heart tube development in mouse reveals alternating phases of cardiac differentiation and morphogenesis. eLife 2017; 6:30668. [PMID: 29202929 PMCID: PMC5731822 DOI: 10.7554/elife.30668] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/26/2017] [Indexed: 12/15/2022] Open
Abstract
During vertebrate heart development, two progenitor populations, first and second heart fields (FHF, SHF), sequentially contribute to longitudinal subdivisions of the heart tube (HT), with the FHF contributing the left ventricle and part of the atria, and the SHF the rest of the heart. Here, we study the dynamics of cardiac differentiation and morphogenesis by tracking individual cells in live analysis of mouse embryos. We report that during an initial phase, FHF precursors differentiate rapidly to form a cardiac crescent, while limited morphogenesis takes place. In a second phase, no differentiation occurs while extensive morphogenesis, including splanchnic mesoderm sliding over the endoderm, results in HT formation. In a third phase, cardiac precursor differentiation resumes and contributes to SHF-derived regions and the dorsal closure of the HT. These results reveal tissue-level coordination between morphogenesis and differentiation during HT formation and provide a new framework to understand heart development. We all start life as a single cell, which – over the course of nine months – multiplies to generate the billions of cells that can be found in a newborn. As an embryo develops, the cells need to achieve two major tasks: they need to diversify into different types of cells, such as blood cells or muscle cells, and they need to organize themselves in space to form tissues and organs. The heart of an embryo, for example, first forms a simple structure called the heart tube that can pump blood and later develops into the four chambers that we see in adults. The tube is made up of cells from two different origins, known as the first and second heart fields. Unlike other organs, the heart has to start beating while it is still developing, and until now, it was unclear how the heart manages this difficult task. Here, Ivanovich et al. studied mouse embryos grown outside the womb by using a combination of advanced microscopy and genetic labeling to track how single cells turn into beating cells and move while the heart forms. The results showed that specializing into beating cells and forming the heart tube shape happened during alternating phases. The first heart-field cells turned into beating cells and began to contract at an early stage before the heart tube was formed. Next, the cells of the second heart field did not instantly develop into beating cells, but instead, helped the first heart-field cells to acquire the shape of a heart tube. Once this was completed, the second heart-field cells started to specialize into beating cells and created the additional parts of the more complex adult heart. This research shows that the second heart field plays an active role in helping the heart tube form. The alternating phases of cell specialization and tissue formation allow the heart to become active whilst it is still developing. A better insight into how the heart forms may help us to create new treatments for various genetic heart conditions. The methods used here could also help to study how cells build other organs.
Collapse
Affiliation(s)
- Kenzo Ivanovitch
- Developmental Biology Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Susana Temiño
- Developmental Biology Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Miguel Torres
- Developmental Biology Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
38
|
Cunningham TJ, Yu MS, McKeithan WL, Spiering S, Carrette F, Huang CT, Bushway PJ, Tierney M, Albini S, Giacca M, Mano M, Puri PL, Sacco A, Ruiz-Lozano P, Riou JF, Umbhauer M, Duester G, Mercola M, Colas AR. Id genes are essential for early heart formation. Genes Dev 2017; 31:1325-1338. [PMID: 28794185 PMCID: PMC5580654 DOI: 10.1101/gad.300400.117] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/17/2017] [Indexed: 01/08/2023]
Abstract
Deciphering the fundamental mechanisms controlling cardiac specification is critical for our understanding of how heart formation is initiated during embryonic development and for applying stem cell biology to regenerative medicine and disease modeling. Using systematic and unbiased functional screening approaches, we discovered that the Id family of helix-loop-helix proteins is both necessary and sufficient to direct cardiac mesoderm formation in frog embryos and human embryonic stem cells. Mechanistically, Id proteins specify cardiac cell fate by repressing two inhibitors of cardiogenic mesoderm formation-Tcf3 and Foxa2-and activating inducers Evx1, Grrp1, and Mesp1. Most importantly, CRISPR/Cas9-mediated ablation of the entire Id (Id1-4) family in mouse embryos leads to failure of anterior cardiac progenitor specification and the development of heartless embryos. Thus, Id proteins play a central and evolutionarily conserved role during heart formation and provide a novel means to efficiently produce cardiovascular progenitors for regenerative medicine and drug discovery applications.
Collapse
Affiliation(s)
- Thomas J Cunningham
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Michael S Yu
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Department of Bioengineering, University of California at San Diego, La Jolla, California 92037, USA
| | - Wesley L McKeithan
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA.,Department of Medicine and Cardiovascular Institute, Stanford University, Palo Alto, California 94305, USA
| | - Sean Spiering
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Florent Carrette
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Chun-Teng Huang
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Paul J Bushway
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92037, USA
| | - Matthew Tierney
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Sonia Albini
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Miguel Mano
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Pier Lorenzo Puri
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Istituti di Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, 00179 Rome, Italy
| | - Alessandra Sacco
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Pilar Ruiz-Lozano
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Regencor, Inc., Los Altos, California 94022, USA
| | - Jean-Francois Riou
- UMR 7622 Developmental Biology, Sorbonne Universités, University Pierre and Marie Curie, F- 75005 Paris, France
| | - Muriel Umbhauer
- UMR 7622 Developmental Biology, Sorbonne Universités, University Pierre and Marie Curie, F- 75005 Paris, France
| | - Gregg Duester
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| | - Mark Mercola
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Department of Medicine and Cardiovascular Institute, Stanford University, Palo Alto, California 94305, USA
| | - Alexandre R Colas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA
| |
Collapse
|
39
|
Lange L, Marks M, Liu J, Wittler L, Bauer H, Piehl S, Bläß G, Timmermann B, Herrmann BG. Patterning and gastrulation defects caused by the tw18 lethal are due to loss of Ppp2r1a. Biol Open 2017; 6:752-764. [PMID: 28619992 PMCID: PMC5483016 DOI: 10.1242/bio.023200] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The mouse t haplotype, a variant 20 cM genomic region on Chromosome 17, harbors 16 embryonic control genes identified by recessive lethal mutations isolated from wild mouse populations. Due to technical constraints so far only one of these, the tw5 lethal, has been cloned and molecularly characterized. Here we report the molecular isolation of the tw18 lethal. Embryos carrying the tw18 lethal die from major gastrulation defects commencing with primitive streak formation at E6.5. We have used transcriptome and marker gene analyses to describe the molecular etiology of the tw18 phenotype. We show that both WNT and Nodal signal transduction are impaired in the mutant epiblast, causing embryonic patterning defects and failure of primitive streak and mesoderm formation. By using a candidate gene approach, gene knockout by homologous recombination and genetic rescue, we have identified the gene causing the tw18 phenotype as Ppp2r1a, encoding the PP2A scaffolding subunit PR65alpha. Our work highlights the importance of phosphatase 2A in embryonic patterning, primitive streak formation, gastrulation, and mesoderm formation downstream of WNT and Nodal signaling.
Collapse
Affiliation(s)
- Lisette Lange
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany.,Free University Berlin, Department of Biology, Chemistry and Pharmacy, Takustrasse 3, Berlin 14195, Germany
| | - Matthias Marks
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany
| | - Jinhua Liu
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany
| | - Lars Wittler
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany
| | - Hermann Bauer
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany
| | - Sandra Piehl
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany
| | - Gabriele Bläß
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany
| | - Bernd Timmermann
- Max Planck Institute for Molecular Genetics, Sequencing Core Facility, Ihnestraße 63-73, Berlin 14195, Germany
| | - Bernhard G Herrmann
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestraße 63-73, Berlin 14195, Germany .,Charité-University Medicine Berlin, Institute for Medical Genetics, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin 12203, Germany
| |
Collapse
|
40
|
Liu Y. Earlier and broader roles of Mesp1 in cardiovascular development. Cell Mol Life Sci 2017; 74:1969-1983. [PMID: 28050627 PMCID: PMC11107530 DOI: 10.1007/s00018-016-2448-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022]
Abstract
Mesoderm posterior 1 is one of earliest markers of the nascent mesoderm. Its best-known function is driving the onset of the cardiovascular system. In the past decade, new evidence supports that Mesp1 acts earlier with greater breadth in cell fate decisions, and through cell-autonomous and cell non-autonomous mechanisms. This review summarizes these new aspects, with an emphasis on the upstream and downstream regulation around Mesp1 and how they may guide cell fate reprogramming.
Collapse
Affiliation(s)
- Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
41
|
Shen X, Bao W, Yu W, Liang R, Nguyen B, Liu Y. An improved method with high sensitivity and low background in detecting low β-galactosidase expression in mouse embryos. PLoS One 2017; 12:e0176915. [PMID: 28475610 PMCID: PMC5419561 DOI: 10.1371/journal.pone.0176915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 04/19/2017] [Indexed: 11/25/2022] Open
Abstract
LacZ is widely used as a reporter in studies of gene expression patterns. β-galactosidase, the product of LacZ gene, is usually detected by X-gal/FeCN staining. In X-gal/FeCN staining, β-galactosidase catalyzes X-gal to produce blue precipitates, which indicate the expression patterns of the gene of interest. A newer LacZ detection method using S-gal/TNBT is more sensitive but plagued by high background. Here, we describe an improved procedure that combines advantageous steps from the two methods. By comparing with X-gal/FeCN and S-gal/TNBT methods in detecting the expression patterns of miR-322/503 and miR-451 at a series of developmental stages, the improved method showed higher sensitivity and lower background. Thus, the improved method could be an alternative way of β-galactosidase staining in low gene expression situations.
Collapse
Affiliation(s)
- Xiaopeng Shen
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States of America
- The College of Life Science, Anhui Normal University, Anhui, China
- * E-mail: (YL); (XS)
| | - Wenjing Bao
- Department of Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wei Yu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States of America
| | - Rui Liang
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States of America
| | - Bao Nguyen
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States of America
| | - Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States of America
- * E-mail: (YL); (XS)
| |
Collapse
|
42
|
Zhang F, Wang L, Li Y, Liu W, Duan F, Huang R, Chen X, Chang SCN, Du Y, Na J. Optimizing mesoderm progenitor selection and three-dimensional microniche culture allows highly efficient endothelial differentiation and ischemic tissue repair from human pluripotent stem cells. Stem Cell Res Ther 2017; 8:6. [PMID: 28114972 PMCID: PMC5259899 DOI: 10.1186/s13287-016-0455-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/05/2016] [Accepted: 12/10/2016] [Indexed: 12/19/2022] Open
Abstract
Background Generation of large quantities of endothelial cells is highly desirable for vascular research, for the treatment of ischemia diseases, and for tissue regeneration. To achieve this goal, we developed a simple, chemically defined culture system to efficiently and rapidly differentiate endothelial cells from human pluripotent stem cells by going through an MESP1 mesoderm progenitor stage. Methods Mesp1 is a key transcription factor that regulates the development of early cardiovascular tissue. Using an MESP1-mTomato knock-in reporter human embryonic stem cell line, we compared the gene expression profiles of MESP1+ and MESP1− cells and identified new signaling pathways that may promote endothelial differentiation. We also used a 3D scaffold to mimic the in vivo microenvironment to further improve the efficiency of endothelial cell generation. Finally, we performed cell transplantation into a critical limb ischemia mouse model to test the repairing potential of endothelial-primed MESP1+ cells. Results MESP1+ mesoderm progenitors, but not MESP1− cells, have strong endothelial differentiation potential. Global gene expression analysis revealed that transcription factors essential for early endothelial differentiation were enriched in MESP1+ cells. Interestingly, MESP1 cells highly expressed Sphingosine-1-phosphate (S1P) receptor and the addition of S1P significantly increased the endothelial differentiation efficiency. Upon seeding in a novel 3D microniche and priming with VEGF and bFGF, MESP1+ cells markedly upregulated genes related to vessel development and regeneration. 3D microniches also enabled long-term endothelial differentiation and proliferation from MESP1+ cells with minimal medium supplements. Finally, we showed that transplanting a small number of endothelial-primed MESP1+ cells in 3D microniches was sufficient to mediate rapid repair of a mouse model of critical limb ischemia. Conclusions Our study demonstrates that combining MESP1+ mesoderm progenitor cells with tissue-engineered 3D microniche and a chemically defined endothelial induction medium is a promising route to maximizing the production of endothelial cells in vitro and augment their regenerative power in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0455-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fengzhi Zhang
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Lin Wang
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yaqian Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Fuyu Duan
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Rujin Huang
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Xi Chen
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Sophia Chia-Ning Chang
- Department of Plastic Surgery, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.,School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Jie Na
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
43
|
Warrier S, Nuwayhid S, Sabatino JA, Sugrue KF, Zohn IE. Supt20 is required for development of the axial skeleton. Dev Biol 2016; 421:245-257. [PMID: 27894818 DOI: 10.1016/j.ydbio.2016.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 09/08/2016] [Accepted: 11/11/2016] [Indexed: 10/20/2022]
Abstract
Somitogenesis and subsequent axial skeletal development is regulated by the interaction of pathways that determine the periodicity of somite formation, rostrocaudal somite polarity and segment identity. Here we use a hypomorphic mutant mouse line to demonstrate that Supt20 (Suppressor of Ty20) is required for development of the axial skeleton. Supt20 hypomorphs display fusions of the ribs and vertebrae at lower thoracic levels along with anterior homeotic transformation of L1 to T14. These defects are preceded by reduction of the rostral somite and posterior shifts in Hox gene expression. While cycling of Notch target genes in the posterior presomitic mesoderm (PSM) appeared normal, expression of Lfng was reduced. In the anterior PSM, Mesp2 expression levels and cycling were unaffected; yet, expression of downstream targets such as Lfng, Ripply2, Mesp1 and Dll3 in the prospective rostral somite was reduced accompanied by expansion of caudal somite markers such as EphrinB2 and Hes7. Supt20 interacts with the Gcn5-containing SAGA histone acetylation complex. Gcn5 hypomorphic mutant embryos show similar defects in axial skeletal development preceded by posterior shift of Hoxc8 and Hoxc9 gene expression. We demonstrate that Gcn5 and Supt20 hypomorphs show similar defects in rostral-caudal somite patterning potentially suggesting shared mechanisms.
Collapse
Affiliation(s)
- Sunita Warrier
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Samer Nuwayhid
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Julia A Sabatino
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Kelsey F Sugrue
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA; Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052, USA
| | - Irene E Zohn
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.
| |
Collapse
|
44
|
Niibe K, Zhang M, Nakazawa K, Morikawa S, Nakagawa T, Matsuzaki Y, Egusa H. The potential of enriched mesenchymal stem cells with neural crest cell phenotypes as a cell source for regenerative dentistry. JAPANESE DENTAL SCIENCE REVIEW 2016; 53:25-33. [PMID: 28479933 PMCID: PMC5405184 DOI: 10.1016/j.jdsr.2016.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/30/2016] [Accepted: 09/16/2016] [Indexed: 01/14/2023] Open
Abstract
Effective regenerative treatments for periodontal tissue defects have recently been demonstrated using mesenchymal stromal/stem cells (MSCs). Furthermore, current bioengineering techniques have enabled de novo fabrication of tooth-perio dental units in mice. These cutting-edge technologies are expected to address unmet needs within regenerative dentistry. However, to achieve efficient and stable treatment outcomes, preparation of an appropriate stem cell source is essential. Many researchers are investigating the use of adult stem cells for regenerative dentistry; bone marrow-derived MSCs (BM-MSCs) are particularly promising and presently used clinically. However, current BM-MSC isolation techniques result in a heterogeneous, non-reproducible cell population because of a lack of identified distinct BM-MSC surface markers. Recently, specific subsets of cell surface markers for BM-MSCs have been reported in mice (PDGFRα+ and Sca-1+) and humans (LNGFR+, THY-1+ and VCAM-1+), facilitating the isolation of unique enriched BM-MSCs (so-called “purified MSCs”). Notably, the enriched BM-MSC population contains neural crest-derived cells, which can differentiate into cells of neural crest- and mesenchymal lineages. In this review, characteristics of the enriched BM-MSCs are outlined with a focus on their potential application within future regenerative dentistry.
Collapse
Affiliation(s)
- Kunimichi Niibe
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Maolin Zhang
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kosuke Nakazawa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Satoru Morikawa
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Taneaki Nakagawa
- Department of Dentistry and Oral Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yumi Matsuzaki
- Department of Cancer Biology, Faculty of Medicine, Shimane University, 89-1 Enya-cho Izumo, Shimane 693-8501, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan.,Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
45
|
Tyser RC, Miranda AM, Chen CM, Davidson SM, Srinivas S, Riley PR. Calcium handling precedes cardiac differentiation to initiate the first heartbeat. eLife 2016; 5. [PMID: 27725084 PMCID: PMC5059139 DOI: 10.7554/elife.17113] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/13/2016] [Indexed: 11/30/2022] Open
Abstract
The mammalian heartbeat is thought to begin just prior to the linear heart tube stage of development. How the initial contractions are established and the downstream consequences of the earliest contractile function on cardiac differentiation and morphogenesis have not been described. Using high-resolution live imaging of mouse embryos, we observed randomly distributed spontaneous asynchronous Ca2+-oscillations (SACOs) in the forming cardiac crescent (stage E7.75) prior to overt beating. Nascent contraction initiated at around E8.0 and was associated with sarcomeric assembly and rapid Ca2+ transients, underpinned by sequential expression of the Na+-Ca2+ exchanger (NCX1) and L-type Ca2+ channel (LTCC). Pharmacological inhibition of NCX1 and LTCC revealed rapid development of Ca2+ handling in the early heart and an essential early role for NCX1 in establishing SACOs through to the initiation of beating. NCX1 blockade impacted on CaMKII signalling to down-regulate cardiac gene expression, leading to impaired differentiation and failed crescent maturation. DOI:http://dx.doi.org/10.7554/eLife.17113.001 The heart is the first organ to form and to begin working in an embryo during pregnancy. It must begin pumping early to supply oxygen and nutrients to the developing embryo. Coordinated contractions of specialised muscle cells in the heart, called cardiomyocytes, generate the force needed to pump blood. The flow of calcium ions into and out of the cardiomyocytes triggers these heartbeats. In addition to triggering heart contractions, calcium ions also act as a messenger that drives changes in which genes are active in the cardiomyocytes and how these cells behave. Scientists commonly think of the first heartbeat as occurring after a tube-like structure forms in the embryo that will eventually develop into the heart. However, it is not yet clear how the first heartbeat starts or how the initial heartbeats affect further heart development. Tyser, Miranda et al. now show that the first heartbeat actually occurs much earlier in embryonic development than widely appreciated. In the experiments, videos of live mouse embryos showed that prior to the first heartbeat the flow of calcium ions between different cardiomyocytes is not synchronised. However, as the heart grows these calcium flows become coordinated leading to the first heartbeat. The heartbeats also become faster as the heart grows. Using drugs to block the movement of calcium ions, Tyser, Miranda et al. also show that a protein called NCX1 is required to trigger the calcium flows prior to the first heartbeat. Moreover, the experiments revealed that these early heartbeats help drive the growth of cardiomyocytes and shape the developing heart. Together, the experiments show that the first heartbeats are essential for normal heart development. Future studies are needed to determine what controls the speed of the first heartbeats, and what organises the calcium flows that trigger the first heartbeat. Such studies may help scientists better understand birth defects of the heart, and may suggest strategies to rebuild hearts that have been damaged by a heart attack or other injury. DOI:http://dx.doi.org/10.7554/eLife.17113.002
Collapse
Affiliation(s)
- Richard Cv Tyser
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,The Hatter Cardiovascular Institute, University College London and Medical School, London, United Kingdom
| | - Antonio Ma Miranda
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London and Medical School, London, United Kingdom
| | - Shankar Srinivas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
46
|
Mespaa can potently induce cardiac fates in zebrafish. Dev Biol 2016; 418:17-27. [PMID: 27554166 DOI: 10.1016/j.ydbio.2016.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/12/2016] [Accepted: 08/18/2016] [Indexed: 01/04/2023]
Abstract
The Mesp family of transcription factors have been implicated in the early formation and migration of the cardiac lineage, although the precise molecular mechanisms underlying this process remain unknown. In this study we examine the function of Mesp family members in zebrafish cardiac development and find that Mespaa is remarkably efficient at promoting cardiac fates in normally non-cardiogenic cells. However, Mespaa is dispensable for normal cardiac formation. Despite no overt defects in cardiovascular specification, we find a consistent defect in cardiac laterality in mespaa null embryos. This is further exacerbated by the depletion of other mesp paralogues, highlighting a conserved role for the mesp family in left-right asymmetry, distinct from a function in cardiac specification. Despite an early requirement for mespaa to promote cardiogenesis, cells over-expressing mespaa are found to both exhibit unique cellular behaviors and activate the transcription of gata5 only after the completion of gastrulation. We propose that while mespaa remains capable of driving cardiac progenitor formation in zebrafish, it may not play an essential role in the cardiac regulatory network. Furthermore, the late activation of migration and cardiac gene transcription in mespaa over-expressing cells challenges previous studies on the timing of these events and provides intriguing questions for future study.
Collapse
|
47
|
Liu Y, Chen L, Diaz AD, Benham A, Xu X, Wijaya CS, Fa'ak F, Luo W, Soibam B, Azares A, Yu W, Lyu Q, Stewart MD, Gunaratne P, Cooney A, McConnell BK, Schwartz RJ. Mesp1 Marked Cardiac Progenitor Cells Repair Infarcted Mouse Hearts. Sci Rep 2016; 6:31457. [PMID: 27538477 PMCID: PMC4990963 DOI: 10.1038/srep31457] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/18/2016] [Indexed: 12/15/2022] Open
Abstract
Mesp1 directs multipotential cardiovascular cell fates, even though it's transiently induced prior to the appearance of the cardiac progenitor program. Tracing Mesp1-expressing cells and their progeny allows isolation and characterization of the earliest cardiovascular progenitor cells. Studying the biology of Mesp1-CPCs in cell culture and ischemic disease models is an important initial step toward using them for heart disease treatment. Because of Mesp1's transitory nature, Mesp1-CPC lineages were traced by following EYFP expression in murine Mesp1(Cre/+); Rosa26(EYFP/+) ES cells. We captured EYFP+ cells that strongly expressed cardiac mesoderm markers and cardiac transcription factors, but not pluripotent or nascent mesoderm markers. BMP2/4 treatment led to the expansion of EYFP+ cells, while Wnt3a and Activin were marginally effective. BMP2/4 exposure readily led EYFP+ cells to endothelial and smooth muscle cells, but inhibition of the canonical Wnt signaling was required to enter the cardiomyocyte fate. Injected mouse pre-contractile Mesp1-EYFP+ CPCs improved the survivability of injured mice and restored the functional performance of infarcted hearts for at least 3 months. Mesp1-EYFP+ cells are bona fide CPCs and they integrated well in infarcted hearts and emerged de novo into terminally differentiated cardiac myocytes, smooth muscle and vascular endothelial cells.
Collapse
Affiliation(s)
- Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Li Chen
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Andrea Diaz Diaz
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Ashley Benham
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Xueping Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cori S Wijaya
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Faisal Fa'ak
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Weijia Luo
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Houston, 77002, USA
| | - Alon Azares
- Stem Cell Engineering, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Wei Yu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Qiongying Lyu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - M David Stewart
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Stem Cell Engineering, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Austin Cooney
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bradley K McConnell
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX 77204, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.,Stem Cell Engineering, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX 77030, USA
| |
Collapse
|
48
|
Yeo HC, Ting S, Brena RM, Koh G, Chen A, Toh SQ, Lim YM, Oh SKW, Lee DY. Genome-Wide Transcriptome and Binding Sites Analyses Identify Early FOX Expressions for Enhancing Cardiomyogenesis Efficiency of hESC Cultures. Sci Rep 2016; 6:31068. [PMID: 27501774 PMCID: PMC4977469 DOI: 10.1038/srep31068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 07/14/2016] [Indexed: 01/19/2023] Open
Abstract
The differentiation efficiency of human embryonic stem cells (hESCs) into heart muscle cells (cardiomyocytes) is highly sensitive to culture conditions. To elucidate the regulatory mechanisms involved, we investigated hESCs grown on three distinct culture platforms: feeder-free Matrigel, mouse embryonic fibroblast feeders, and Matrigel replated on feeders. At the outset, we profiled and quantified their differentiation efficiency, transcriptome, transcription factor binding sites and DNA-methylation. Subsequent genome-wide analyses allowed us to reconstruct the relevant interactome, thereby forming the regulatory basis for implicating the contrasting differentiation efficiency of the culture conditions. We hypothesized that the parental expressions of FOXC1, FOXD1 and FOXQ1 transcription factors (TFs) are correlative with eventual cardiomyogenic outcome. Through WNT induction of the FOX TFs, we observed the co-activation of WNT3 and EOMES which are potent inducers of mesoderm differentiation. The result strengthened our hypothesis on the regulatory role of the FOX TFs in enhancing mesoderm differentiation capacity of hESCs. Importantly, the final proportions of cells expressing cardiac markers were directly correlated to the strength of FOX inductions within 72 hours after initiation of differentiation across different cell lines and protocols. Thus, we affirmed the relationship between early FOX TF expressions and cardiomyogenesis efficiency.
Collapse
Affiliation(s)
- Hock Chuan Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore.,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Sherwin Ting
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Romulo Martin Brena
- USC Epigenome Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Geoffrey Koh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Allen Chen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Siew Qi Toh
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Yu Ming Lim
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Steve Kah Weng Oh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore.,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| |
Collapse
|
49
|
Msx1 and Msx2 function together in the regulation of primordial germ cell migration in the mouse. Dev Biol 2016; 417:11-24. [PMID: 27435625 PMCID: PMC5407493 DOI: 10.1016/j.ydbio.2016.07.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 11/23/2022]
Abstract
Primordial germ cells (PGCs) are a highly migratory cell population that gives rise to eggs and sperm. Much is known about PGC specification, but less about the processes that control PGC migration. In this study, we document a deficiency in PGC development in embryos carrying global homozygous null mutations in Msx1 and Msx2, both immediate downstream effectors of Bmp signaling pathway. We show that Msx1−/−;Msx2−/− mutant embryos have defects in PGC migration as well as a reduced number of PGCs. These phenotypes are also evident in a Mesp1-Cre-mediated mesoderm-specific mutant line of Msx1 and Msx2. Since PGCs are not marked in Mesp1-lineage tracing, our results suggest that Msx1 and Msx2 function cell non-autonomously in directing PGC migration. Consistent with this hypothesis, we noted an upregulation of fibronectin, well known as a mediator of cell migration, in tissues through which PGCs migrate. We also noted a reduction in the expression of Wnt5a and an increase in the expression in Bmp4 in such tissues in Msx1−/−;Msx2−/− mutants, both known effectors of PGC development.
Collapse
|
50
|
Vascular Development and Regeneration in the Mammalian Heart. J Cardiovasc Dev Dis 2016; 3:jcdd3020023. [PMID: 29367569 PMCID: PMC5715682 DOI: 10.3390/jcdd3020023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/31/2016] [Accepted: 06/14/2016] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases including coronary artery disease are the leading cause of death worldwide. Unraveling the developmental origin of coronary vessels could offer important therapeutic implications for treatment of cardiovascular diseases. The recent identification of the endocardial source of coronary vessels reveals a heterogeneous origin of coronary arteries in the adult heart. In this review, we will highlight recent advances in finding the sources of coronary vessels in the mammalian heart from lineage-tracing models as well as differentiation studies using pluripotent stem cells. Moreover, we will also discuss how we induce neovascularization in the damaged heart through transient yet highly efficient expression of VEGF-modified mRNAs as a potentially therapeutic delivery platform.
Collapse
|