1
|
Phng LK, Hogan BM. Endothelial cell transitions in zebrafish vascular development. Dev Growth Differ 2024; 66:357-368. [PMID: 39072708 PMCID: PMC11457512 DOI: 10.1111/dgd.12938] [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: 05/07/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
In recent decades, developmental biologists have come to view vascular development as a series of progressive transitions. Mesoderm differentiates into endothelial cells; arteries, veins and lymphatic endothelial cells are specified from early endothelial cells; and vascular networks diversify and invade developing tissues and organs. Our understanding of this elaborate developmental process has benefitted from detailed studies using the zebrafish as a model system. Here, we review a number of key developmental transitions that occur in zebrafish during the formation of the blood and lymphatic vessel networks.
Collapse
Affiliation(s)
- Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Benjamin M Hogan
- Organogenesis and Cancer Programme, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology and the Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
McCracken IR, Baker AH, Smart N, De Val S. Transcriptional regulators of arterial and venous identity in the developing mammalian embryo. CURRENT OPINION IN PHYSIOLOGY 2023; 35:None. [PMID: 38328689 PMCID: PMC10844100 DOI: 10.1016/j.cophys.2023.100691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The complex and hierarchical vascular network of arteries, veins, and capillaries features considerable endothelial heterogeneity, yet the regulatory pathways directing arteriovenous specification, differentiation, and identity are still not fully understood. Recent advances in analysis of endothelial-specific gene-regulatory elements, single-cell RNA sequencing, and cell lineage tracing have both emphasized the importance of transcriptional regulation in this process and shed considerable light on the mechanism and regulation of specification within the endothelium. In this review, we discuss recent advances in our understanding of how endothelial cells acquire arterial and venous identity and the role different transcription factors play in this process.
Collapse
Affiliation(s)
- Ian R McCracken
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, United Kingdom
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, United Kingdom
| | - Sarah De Val
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, United Kingdom
| |
Collapse
|
3
|
Hagedorn EJ, Perlin JR, Freeman RJ, Wattrus SJ, Han T, Mao C, Kim JW, Fernández-Maestre I, Daily ML, D'Amato C, Fairchild MJ, Riquelme R, Li B, Ragoonanan DAVE, Enkhbayar K, Henault EL, Wang HG, Redfield SE, Collins SH, Lichtig A, Yang S, Zhou Y, Kunar B, Gomez-Salinero JM, Dinh TT, Pan J, Holler K, Feldman HA, Butcher EC, van Oudenaarden A, Rafii S, Junker JP, Zon LI. Transcription factor induction of vascular blood stem cell niches in vivo. Dev Cell 2023; 58:1037-1051.e4. [PMID: 37119815 PMCID: PMC10330626 DOI: 10.1016/j.devcel.2023.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/08/2023] [Accepted: 04/07/2023] [Indexed: 05/01/2023]
Abstract
The hematopoietic niche is a supportive microenvironment composed of distinct cell types, including specialized vascular endothelial cells that directly interact with hematopoietic stem and progenitor cells (HSPCs). The molecular factors that specify niche endothelial cells and orchestrate HSPC homeostasis remain largely unknown. Using multi-dimensional gene expression and chromatin accessibility analyses in zebrafish, we define a conserved gene expression signature and cis-regulatory landscape that are unique to sinusoidal endothelial cells in the HSPC niche. Using enhancer mutagenesis and transcription factor overexpression, we elucidate a transcriptional code that involves members of the Ets, Sox, and nuclear hormone receptor families and is sufficient to induce ectopic niche endothelial cells that associate with mesenchymal stromal cells and support the recruitment, maintenance, and division of HSPCs in vivo. These studies set forth an approach for generating synthetic HSPC niches, in vitro or in vivo, and for effective therapies to modulate the endogenous niche.
Collapse
Affiliation(s)
- Elliott J Hagedorn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA; Section of Hematology and Medical Oncology and Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Julie R Perlin
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Rebecca J Freeman
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Samuel J Wattrus
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Tianxiao Han
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Clara Mao
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Ji Wook Kim
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Inés Fernández-Maestre
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Madeleine L Daily
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Christopher D'Amato
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Michael J Fairchild
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Raquel Riquelme
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Brian Li
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Dana A V E Ragoonanan
- Section of Hematology and Medical Oncology and Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Khaliun Enkhbayar
- Section of Hematology and Medical Oncology and Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Emily L Henault
- Section of Hematology and Medical Oncology and Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Helen G Wang
- Section of Hematology and Medical Oncology and Center for Regenerative Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Shelby E Redfield
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Samantha H Collins
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Asher Lichtig
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Song Yang
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Yi Zhou
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA
| | - Balvir Kunar
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jesus Maria Gomez-Salinero
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Thanh T Dinh
- Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research and the Department of Pathology, Stanford University, Stanford, CA, USA
| | - Junliang Pan
- Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research and the Department of Pathology, Stanford University, Stanford, CA, USA
| | - Karoline Holler
- Berlin Institute for Medical Systems Biology, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Henry A Feldman
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | - Eugene C Butcher
- Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research and the Department of Pathology, Stanford University, Stanford, CA, USA
| | - Alexander van Oudenaarden
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - J Philipp Junker
- Berlin Institute for Medical Systems Biology, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA, USA.
| |
Collapse
|
4
|
Dougherty EJ, Chen LY, Awad KS, Ferreyra GA, Demirkale CY, Keshavarz A, Gairhe S, Johnston KA, Hicks ME, Sandler AB, Curran CS, Krack JM, Ding Y, Suffredini AF, Solomon MA, Elinoff JM, Danner RL. Inflammation and DKK1-induced AKT activation contribute to endothelial dysfunction following NR2F2 loss. Am J Physiol Lung Cell Mol Physiol 2023; 324:L783-L798. [PMID: 37039367 PMCID: PMC10202490 DOI: 10.1152/ajplung.00171.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 03/28/2023] [Accepted: 04/01/2023] [Indexed: 04/12/2023] Open
Abstract
NR2F2 is expressed in endothelial cells (ECs) and Nr2f2 knockout produces lethal cardiovascular defects. In humans, reduced NR2F2 expression is associated with cardiovascular diseases including congenital heart disease and atherosclerosis. Here, NR2F2 silencing in human primary ECs led to inflammation, endothelial-to-mesenchymal transition (EndMT), proliferation, hypermigration, apoptosis-resistance, and increased production of reactive oxygen species. These changes were associated with STAT and AKT activation along with increased production of DKK1. Co-silencing DKK1 and NR2F2 prevented NR2F2-loss-induced STAT and AKT activation and reversed EndMT. Serum DKK1 concentrations were elevated in patients with pulmonary arterial hypertension (PAH) and DKK1 was secreted by ECs in response to in vitro loss of either BMPR2 or CAV1, which are genetic defects associated with the development of PAH. In human primary ECs, NR2F2 suppressed DKK1, whereas its loss conversely induced DKK1 and disrupted endothelial homeostasis, promoting phenotypic abnormalities associated with pathologic vascular remodeling. Activating NR2F2 or blocking DKK1 may be useful therapeutic targets for treating chronic vascular diseases associated with EC dysfunction.NEW & NOTEWORTHY NR2F2 loss in the endothelial lining of blood vessels is associated with cardiovascular disease. Here, NR2F2-silenced human endothelial cells were inflammatory, proliferative, hypermigratory, and apoptosis-resistant with increased oxidant stress and endothelial-to-mesenchymal transition. DKK1 was induced in NR2F2-silenced endothelial cells, while co-silencing NR2F2 and DKK1 prevented NR2F2-loss-associated abnormalities in endothelial signaling and phenotype. Activating NR2F2 or blocking DKK1 may be useful therapeutic targets for treating vascular diseases associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Edward J Dougherty
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Li-Yuan Chen
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Keytam S Awad
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Gabriela A Ferreyra
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Cumhur Y Demirkale
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Ali Keshavarz
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Salina Gairhe
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Kathryn A Johnston
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Madelyn E Hicks
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Alexis B Sandler
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Colleen S Curran
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Janell M Krack
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Yi Ding
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Anthony F Suffredini
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Michael A Solomon
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Jason M Elinoff
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| | - Robert L Danner
- Clinical Center/Critical Care Medicine Department, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
5
|
Parab S, Setten E, Astanina E, Bussolino F, Doronzo G. The tissue-specific transcriptional landscape underlines the involvement of endothelial cells in health and disease. Pharmacol Ther 2023; 246:108418. [PMID: 37088448 DOI: 10.1016/j.pharmthera.2023.108418] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/23/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Endothelial cells (ECs) that line vascular and lymphatic vessels are being increasingly recognized as important to organ function in health and disease. ECs participate not only in the trafficking of gases, metabolites, and cells between the bloodstream and tissues but also in the angiocrine-based induction of heterogeneous parenchymal cells, which are unique to their specific tissue functions. The molecular mechanisms regulating EC heterogeneity between and within different tissues are modeled during embryogenesis and become fully established in adults. Any changes in adult tissue homeostasis induced by aging, stress conditions, and various noxae may reshape EC heterogeneity and induce specific transcriptional features that condition a functional phenotype. Heterogeneity is sustained via specific genetic programs organized through the combinatory effects of a discrete number of transcription factors (TFs) that, at the single tissue-level, constitute dynamic networks that are post-transcriptionally and epigenetically regulated. This review is focused on outlining the TF-based networks involved in EC specialization and physiological and pathological stressors thought to modify their architecture.
Collapse
Affiliation(s)
- Sushant Parab
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elisa Setten
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elena Astanina
- Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy.
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| |
Collapse
|
6
|
Single-cell RNA-seq reveals vascular endothelial cell heterogeneity and potential vascular dysfunction in hypertrophic scars. Acta Biochim Biophys Sin (Shanghai) 2023; 55:165-168. [PMID: 36655484 PMCID: PMC10157513 DOI: 10.3724/abbs.2023001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
7
|
Shi HY, Xie MS, Yang CX, Huang RT, Xue S, Liu XY, Xu YJ, Yang YQ. Identification of SOX18 as a New Gene Predisposing to Congenital Heart Disease. Diagnostics (Basel) 2022; 12:diagnostics12081917. [PMID: 36010266 PMCID: PMC9406965 DOI: 10.3390/diagnostics12081917] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital heart disease (CHD) is the most frequent kind of birth deformity in human beings and the leading cause of neonatal mortality worldwide. Although genetic etiologies encompassing aneuploidy, copy number variations, and mutations in over 100 genes have been uncovered to be involved in the pathogenesis of CHD, the genetic components predisposing to CHD in most cases remain unclear. We recruited a family with CHD from the Chinese Han population in the present investigation. Through whole-exome sequencing analysis of selected family members, a new SOX18 variation, namely NM_018419.3:c.349A>T; p.(Lys117*), was identified and confirmed to co-segregate with the CHD phenotype in the entire family by Sanger sequencing analysis. The heterozygous variant was absent from the 384 healthy volunteers enlisted as control individuals. Functional exploration via luciferase reporter analysis in cultivated HeLa cells revealed that Lys117*-mutant SOX18 lost transactivation on its target genes NR2F2 and GATA4, two genes responsible for CHD. Moreover, the genetic variation terminated the synergistic activation between SOX18 and NKX2.5, another gene accountable for CHD. The findings strongly indicate SOX18 as a novel gene contributing to CHD, which helps address challenges in the clinical genetic diagnosis and prenatal prophylaxis of CHD.
Collapse
Affiliation(s)
- Hong-Yu Shi
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Meng-Shi Xie
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai 200940, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| |
Collapse
|
8
|
Gurung S, Restrepo NK, Chestnut B, Klimkaite L, Sumanas S. Single-cell transcriptomic analysis of vascular endothelial cells in zebrafish embryos. Sci Rep 2022; 12:13065. [PMID: 35906287 PMCID: PMC9338088 DOI: 10.1038/s41598-022-17127-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular endothelial cells exhibit substantial phenotypic and transcriptional heterogeneity which is established during early embryogenesis. However, the molecular mechanisms involved in establishing endothelial cell diversity are still not well understood. Zebrafish has emerged as an advantageous model to study vascular development. Despite its importance, the single-cell transcriptomic profile of vascular endothelial cells during zebrafish development is still missing. To address this, we applied single-cell RNA-sequencing (scRNA-seq) of vascular endothelial cells isolated from zebrafish embryos at the 24 hpf stage. Six distinct clusters or subclusters related to vascular endothelial cells were identified which include arterial, two venous, cranial, endocardial and endothelial progenitor cell subtypes. Furthermore, we validated our findings by characterizing novel markers for arterial, venous, and endocardial cells. We experimentally confirmed the presence of two transcriptionally different venous cell subtypes, demonstrating heterogeneity among venous endothelial cells at this early developmental stage. This dataset will be a valuable resource for future functional characterization of vascular endothelial cells and interrogation of molecular mechanisms involved in the establishment of their heterogeneity and cell-fate decisions.
Collapse
Affiliation(s)
- Suman Gurung
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL, 33602, USA
| | - Nicole K Restrepo
- Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL, 33602, USA
| | - Brendan Chestnut
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Laurita Klimkaite
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA. .,Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL, 33602, USA.
| |
Collapse
|
9
|
Cissy Yu Q, Bai L, Chen Y, Chen Y, Peng G, Wang D, Yang G, Cui G, Jing N, Arial Zeng Y. Embryonic vascular establishment requires protein C receptor-expressing endothelial progenitors. Development 2022; 149:275466. [DOI: 10.1242/dev.200419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/05/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Vascular establishment is one of the early events in embryogenesis. It is believed that vessel-initiating endothelial progenitors cluster to form the first primitive vessel. Understanding the molecular identity of these progenitors is crucial in order to elucidate lineage hierarchy. In this study, we identify protein C receptor (Procr) as an endothelial progenitor marker and investigate the role of Procr+ progenitors during embryonic vascular development. Using a ProcrmGFP-2A-lacZ reporter, we reveal a much earlier Procr expression (embryonic day 7.5) than previously acknowledged (embryonic day 13.5). Genetic fate-mapping experiments using ProcrCre and ProcrCreER demonstrate that Procr+ cells give rise to blood vessels throughout the entire embryo proper. Single-cell RNA-sequencing analyses place Procr+ cells at the start of endothelial commitment and maturation. Furthermore, targeted ablation of Procr+ cells results in failure of vessel formation and early embryonic lethality. Notably, genetic fate mapping and scRNA-seq pseudotime analysis support the view that Procr+ progenitors can give rise to hemogenic endothelium. In this study, we establish a Procr expression timeline and identify Procr+ vessel-initiating progenitors, and demonstrate their indispensable role in establishment of the vasculature during embryo development.
Collapse
Affiliation(s)
- Qing Cissy Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Lanyue Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Yujie Chen
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS 3 Key Laboratory of Computational Biology , , Shanghai 200031 , China
| | - Guangdun Peng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Institutes of Biomedicine and Health, Chinese Academy of Sciences 4 , Guangzhou 510530 , China
| | - Daisong Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Guowei Yang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences 2 , 310024 Hangzhou , China
| | - Guizhong Cui
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Institutes of Biomedicine and Health, Chinese Academy of Sciences 4 , Guangzhou 510530 , China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Institutes of Biomedicine and Health, Chinese Academy of Sciences 4 , Guangzhou 510530 , China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences 5 , Beijing 100101 , China
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences 2 , 310024 Hangzhou , China
| |
Collapse
|
10
|
Li RF, Wang YS, Lu FI, Huang YS, Chiu CC, Tai MH, Wu CY. Identification of Novel Vascular Genes Downstream of Islet2 and Nr2f1b Transcription Factors. Biomedicines 2022; 10:biomedicines10061261. [PMID: 35740282 PMCID: PMC9220758 DOI: 10.3390/biomedicines10061261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/21/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
The genetic regulation of vascular development is not elucidated completely. We previously characterized the transcription factors Islet2 (Isl2) and Nr2f1b as being critical for vascular growth. In this study, we further performed combinatorial microarrays to identify genes that are potentially regulated by these factors. We verified the changed expression of several targets in isl2/nr2f1b morphants. Those genes expressed in vessels during embryogenesis suggested their functions in vascular development. We selectively assayed a potential target follistatin a (fsta). Follistatin is known to inhibit BMP, and BMP signaling has been shown to be important for angiogenesis. However, the fsta’s role in vascular development has not been well studied. Here, we showed the vascular defects in ISV growth and CVP patterning while overexpressing fsta in the embryo, which mimics the phenotype of isl2/nr2f1b morphants. The vascular abnormalities are likely caused by defects in migration and proliferation. We further observed the altered expression of vessel markers consistent with the vascular defects in (fli:fsta) embryos. We showed that the knockdown of fsta can rescue the vascular defects in (fli:fsta) fish, suggesting the functional specificity of fsta. Moreover, the decreased expression of fsta rescues abnormal vessel growth in isl2 and nr2f1b morphants, indicating that fsta functions downstream of isl2/nr2f1b. Lastly, we showed that Isl2/Nr2f1b control vascular development, via Fsta–BMP signaling in part. Collectively, our microarray data identify many interesting genes regulated by isl2/nr2f1b, which likely function in the vasculature. Our research provides useful information on the genetic control of vascular development.
Collapse
Affiliation(s)
- Ru-Fang Li
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.-F.L.); (Y.-S.W.); (Y.-S.H.); (C.-C.C.); (M.-H.T.)
| | - Yi-Shan Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.-F.L.); (Y.-S.W.); (Y.-S.H.); (C.-C.C.); (M.-H.T.)
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei 115, Taiwan
| | - Fu-I Lu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan;
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Yi-Shan Huang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.-F.L.); (Y.-S.W.); (Y.-S.H.); (C.-C.C.); (M.-H.T.)
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.-F.L.); (Y.-S.W.); (Y.-S.H.); (C.-C.C.); (M.-H.T.)
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hong Tai
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.-F.L.); (Y.-S.W.); (Y.-S.H.); (C.-C.C.); (M.-H.T.)
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.-F.L.); (Y.-S.W.); (Y.-S.H.); (C.-C.C.); (M.-H.T.)
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei 115, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Correspondence: ; Tel.: +886-7-5252000 (ext. 3627)
| |
Collapse
|
11
|
Wang YS, Huang YS, Chiu CC, Wu TY, Zhou JQ, Liang SR, Tai MH, Wu CY. Interaction of transcription factors Islet2 and Nr2f1b to control vascular patterning during zebrafish development. Biochem Biophys Res Commun 2022; 604:123-129. [PMID: 35303678 DOI: 10.1016/j.bbrc.2022.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022]
Abstract
Many regulators controlling arterial identity are well described; however, transcription factors that promote vein identity and vascular patterning have remained largely unknown. We previously identified the transcription factors Islet2 (Isl2) and Nr2f1b required for specification of the vein and tip cell identity mediated by notch pathway in zebrafish. However, the interaction between Isl2 and Nr2f1b is not known. In this study, we report that Nr2f2 plays minor roles on vein and intersegmental vessels (ISV) growth and dissect the genetic interactions among the three transcription factors Isl2, Nr2f1b, and Nr2f2 using a combinatorial knockdown strategy. The double knockdown of isl2/nr2f1b, isl2/nr2f2, and nr2f1b/nr2f2 showed the enhanced defects in vasculature including less completed ISV, reduced veins, and ISV cells. We further tested the genetic relationship among these three transcription factors. We found isl2 can regulate the expression of nr2f1b and nr2f2, suggesting a model where Isl2 functions upstream of Nr2f1b and Nr2f2. We hypothsized that Isl2 and Nr2f1b can function together through cis-regulatory binding motifs. In-vitro luciferase assay results, we showed that Isl2 and Nr2f1b can cooperatively enhance gene expression. Moreover, co-immunoprecipitation results indicated that Isl2 and Nr2f1b interact physically. Together, we showed that the interaction of the Nr2f1b and Nr2f2 transcription factors in combination with the Islet2 play coordinated roles in the vascular development of zebrafish.
Collapse
Affiliation(s)
- Yi-Shan Wang
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan; Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - Yi-Shan Huang
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Taiwan
| | - Ting-Yun Wu
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan
| | - Jun-Qing Zhou
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan
| | - Shuo-Rong Liang
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan
| | - Ming-Hong Tai
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan; Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Taiwan; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan; Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Taiwan; Institute of Medical Science and Technology, National Sun Yat-sen University, Taiwan.
| |
Collapse
|
12
|
McCracken IR, Dobie R, Bennett M, Passi R, Beqqali A, Henderson NC, Mountford JC, Riley PR, Ponting CP, Smart N, Brittan M, Baker AH. Mapping the developing human cardiac endothelium at single-cell resolution identifies MECOM as a regulator of arteriovenous gene expression. Cardiovasc Res 2022; 118:2960-2972. [PMID: 35212715 PMCID: PMC9648824 DOI: 10.1093/cvr/cvac023] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
AIMS Coronary vasculature formation is a critical event during cardiac development, essential for heart function throughout perinatal and adult life. However, current understanding of coronary vascular development has largely been derived from transgenic mouse models. The aim of this study was to characterize the transcriptome of the human foetal cardiac endothelium using single-cell RNA sequencing (scRNA-seq) to provide critical new insights into the cellular heterogeneity and transcriptional dynamics that underpin endothelial specification within the vasculature of the developing heart. METHODS AND RESULTS We acquired scRNA-seq data of over 10 000 foetal cardiac endothelial cells (ECs), revealing divergent EC subtypes including endocardial, capillary, venous, arterial, and lymphatic populations. Gene regulatory network analyses predicted roles for SMAD1 and MECOM in determining the identity of capillary and arterial populations, respectively. Trajectory inference analysis suggested an endocardial contribution to the coronary vasculature and subsequent arterialization of capillary endothelium accompanied by increasing MECOM expression. Comparative analysis of equivalent data from murine cardiac development demonstrated that transcriptional signatures defining endothelial subpopulations are largely conserved between human and mouse. Comprehensive characterization of the transcriptional response to MECOM knockdown in human embryonic stem cell-derived EC (hESC-EC) demonstrated an increase in the expression of non-arterial markers, including those enriched in venous EC. CONCLUSIONS scRNA-seq of the human foetal cardiac endothelium identified distinct EC populations. A predicted endocardial contribution to the developing coronary vasculature was identified, as well as subsequent arterial specification of capillary EC. Loss of MECOM in hESC-EC increased expression of non-arterial markers, suggesting a role in maintaining arterial EC identity.
Collapse
Affiliation(s)
- Ian R McCracken
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK,Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Ross Dobie
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Matthew Bennett
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Rainha Passi
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Abdelaziz Beqqali
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK,MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | | | - Paul R Riley
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Chris P Ponting
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Nicola Smart
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
13
|
Chen D, Schwartz MA, Simons M. Developmental Perspectives on Arterial Fate Specification. Front Cell Dev Biol 2021; 9:691335. [PMID: 34249941 PMCID: PMC8269928 DOI: 10.3389/fcell.2021.691335] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Blood vessel acquisition of arterial or venous fate is an adaptive phenomenon in response to increasing blood circulation during vascular morphogenesis. The past two decades of effort in this field led to development of a widely accepted paradigm of molecular regulators centering on VEGF and Notch signaling. More recent findings focused on shear stress-induced cell cycle arrest as a prerequisite for arterial specification substantially modify this traditional understanding. This review aims to summarize key molecular mechanisms that work in concert to drive the acquisition of arterial fate in two distinct developmental settings of vascular morphogenesis: de novo vasculogenesis of the dorsal aorta and postnatal retinal angiogenesis. We will also discuss the questions and conceptual controversies that potentially point to novel directions of investigation and possible clinical relevance.
Collapse
Affiliation(s)
- Dongying Chen
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
14
|
Greenspan LJ, Weinstein BM. To be or not to be: endothelial cell plasticity in development, repair, and disease. Angiogenesis 2021; 24:251-269. [PMID: 33449300 PMCID: PMC8205957 DOI: 10.1007/s10456-020-09761-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Endothelial cells display an extraordinary plasticity both during development and throughout adult life. During early development, endothelial cells assume arterial, venous, or lymphatic identity, while selected endothelial cells undergo additional fate changes to become hematopoietic progenitor, cardiac valve, and other cell types. Adult endothelial cells are some of the longest-lived cells in the body and their participation as stable components of the vascular wall is critical for the proper function of both the circulatory and lymphatic systems, yet these cells also display a remarkable capacity to undergo changes in their differentiated identity during injury, disease, and even normal physiological changes in the vasculature. Here, we discuss how endothelial cells become specified during development as arterial, venous, or lymphatic endothelial cells or convert into hematopoietic stem and progenitor cells or cardiac valve cells. We compare findings from in vitro and in vivo studies with a focus on the zebrafish as a valuable model for exploring the signaling pathways and environmental cues that drive these transitions. We also discuss how endothelial plasticity can aid in revascularization and repair of tissue after damage- but may have detrimental consequences under disease conditions. By better understanding endothelial plasticity and the mechanisms underlying endothelial fate transitions, we can begin to explore new therapeutic avenues.
Collapse
Affiliation(s)
- Leah J Greenspan
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
Wu Y, Wharton J, Walters R, Vasilaki E, Aman J, Zhao L, Wilkins MR, Rhodes CJ. The pathophysiological role of novel pulmonary arterial hypertension gene SOX17. Eur Respir J 2021; 58:13993003.04172-2020. [PMID: 33632800 DOI: 10.1183/13993003.04172-2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/08/2021] [Indexed: 11/05/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease predominantly targeting pre-capillary blood vessels. Adverse structural remodelling and increased pulmonary vascular resistance result in cardiac hypertrophy and ultimately failure of the right ventricle. Recent whole-genome and whole-exome sequencing studies have identified SOX17 as a novel risk gene in PAH, with a dominant mode of inheritance and incomplete penetrance. Rare deleterious variants in the gene and more common variants in upstream enhancer sites have both been associated with the disease, and a deficiency of SOX17 expression may predispose to PAH. This review aims to consolidate the evidence linking genetic variants in SOX17 to PAH, and explores the numerous targets and effects of the transcription factor, focusing on the pulmonary vasculature and the pathobiology of PAH.
Collapse
Affiliation(s)
- Yukyee Wu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Rachel Walters
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Eleni Vasilaki
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jurjan Aman
- National Heart and Lung Institute, Imperial College London, London, UK.,VUmc, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lan Zhao
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
16
|
Gunawan F, Gentile A, Gauvrit S, Stainier DYR, Bensimon-Brito A. Nfatc1 Promotes Interstitial Cell Formation During Cardiac Valve Development in Zebrafish. Circ Res 2020; 126:968-984. [PMID: 32070236 DOI: 10.1161/circresaha.119.315992] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE The transcription factor NFATC1 (nuclear factor of activated T-cell 1) has been implicated in cardiac valve formation in humans and mice, but we know little about the underlying mechanisms. To gain mechanistic understanding of cardiac valve formation at single-cell resolution and insights into the role of NFATC1 in this process, we used the zebrafish model as it offers unique attributes for live imaging and facile genetics. OBJECTIVE To understand the role of Nfatc1 in cardiac valve formation. METHODS AND RESULTS Using the zebrafish atrioventricular valve, we focus on the valve interstitial cells (VICs), which confer biomechanical strength to the cardiac valve leaflets. We find that initially atrioventricular endocardial cells migrate collectively into the cardiac jelly to form a bilayered structure; subsequently, the cells that led this migration invade the ECM (extracellular matrix) between the 2 endocardial cell monolayers, undergo endothelial-to-mesenchymal transition as marked by loss of intercellular adhesion, and differentiate into VICs. These cells proliferate and are joined by a few neural crest-derived cells. VIC expansion and a switch from a promigratory to an elastic ECM drive valve leaflet elongation. Functional analysis of Nfatc1 reveals its requirement during VIC development. Zebrafish nfatc1 mutants form significantly fewer VICs due to reduced proliferation and impaired recruitment of endocardial and neural crest cells during the early stages of VIC development. With high-speed microscopy and echocardiography, we show that reduced VIC formation correlates with valvular dysfunction and severe retrograde blood flow that persist into adulthood. Analysis of downstream effectors reveals that Nfatc1 promotes the expression of twist1b-a well-known regulator of epithelial-to-mesenchymal transition. CONCLUSIONS Our study sheds light on the function of Nfatc1 in zebrafish cardiac valve development and reveals its role in VIC formation. It also further establishes the zebrafish as a powerful model to carry out longitudinal studies of valve formation and function.
Collapse
Affiliation(s)
- Felix Gunawan
- From the Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (F.G., A.G., S.G., D.Y.R.S., A.B.-B.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim (F.G., S.G., D.Y.R.S., A.B.-B.)
| | - Alessandra Gentile
- From the Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (F.G., A.G., S.G., D.Y.R.S., A.B.-B.)
| | - Sébastien Gauvrit
- From the Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (F.G., A.G., S.G., D.Y.R.S., A.B.-B.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim (F.G., S.G., D.Y.R.S., A.B.-B.)
| | - Didier Y R Stainier
- From the Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (F.G., A.G., S.G., D.Y.R.S., A.B.-B.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim (F.G., S.G., D.Y.R.S., A.B.-B.)
| | - Anabela Bensimon-Brito
- From the Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (F.G., A.G., S.G., D.Y.R.S., A.B.-B.).,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim (F.G., S.G., D.Y.R.S., A.B.-B.)
| |
Collapse
|
17
|
Iwasaki R, Tsuge K, Kishimoto K, Hayashi Y, Iwaana T, Hohjoh H, Inazumi T, Kawahara A, Tsuchiya S, Sugimoto Y. Essential role of prostaglandin E 2 and the EP3 receptor in lymphatic vessel development during zebrafish embryogenesis. Sci Rep 2019; 9:7650. [PMID: 31114004 PMCID: PMC6529442 DOI: 10.1038/s41598-019-44095-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
Lymphatic endothelial cells arise from the venous endothelial cells in embryonic lymphatic development. However, the molecular mechanisms remain to be elucidated. We here report that prostaglandin (PG) E2 plays essential roles in the embryonic lymphatic development through the EP3 receptor, one of the PGE2 receptors. Knockdown of the EP3 receptor or inhibition of cyclooxygenases (COX; rate-limiting enzymes for PG synthesis) impaired lymphatic development by perturbing lymphatic specification during zebrafish development. These impairments by COX inhibition were recovered by treatment with sulprostone (EP1/3 agonist). Knockdown of the EP3 receptor further demonstrated its requirement in the expression of sex determining region Y-box 18 (sox18) and nuclear receptor subfamily 2, group F, member 2 (nr2f2), essential factors of the lymphatic specification. The EP3 receptor was expressed in the posterior cardinal vein (region of embryonic lymphatic development) and the adjacent intermediate cell mass (ICM) during the lymphatic specification. COX1 was expressed in the region more upstream of the posterior cardinal vein relative to the EP3 receptor, and the COX1-selective inhibitor impaired the lymphatic specification. On the other hand, two COX2 subtypes did not show distinct sites of expression around the region of expression of the EP3 receptor. Finally, we generated EP3-deficient zebrafish, which also showed defect in lymphatic specification and development. Thus, we demonstrated that COX1-derived PGE2-EP3 pathway is required for embryonic lymphatic development by upregulating the expression of key factors for the lymphatic specification.
Collapse
Affiliation(s)
- Ryo Iwasaki
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan
| | - Kyoshiro Tsuge
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan
| | - Koichiro Kishimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan
| | - Yuta Hayashi
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan
| | - Takuya Iwaana
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan
| | - Hirofumi Hohjoh
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan
| | - Tomoaki Inazumi
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), 1-7-1 Otemachi, Chiyoda-ku, 100-0004, Tokyo, Japan
| | - Atsuo Kawahara
- Laboratory for Developmental Biology, Center for Medical Education and Sciences, Graduate School of Medical Science, University of Yamanashi, 1110 Shimokato, Chuo, 409-3898, Yamanashi, Japan
| | - Soken Tsuchiya
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan. .,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), 1-7-1 Otemachi, Chiyoda-ku, 100-0004, Tokyo, Japan.
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, 862-0973, Kumamoto, Japan. .,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), 1-7-1 Otemachi, Chiyoda-ku, 100-0004, Tokyo, Japan.
| |
Collapse
|
18
|
Mitchell CA, Dasgupta S, Zhang S, Stapleton HM, Volz DC. Disruption of Nuclear Receptor Signaling Alters Triphenyl Phosphate-Induced Cardiotoxicity in Zebrafish Embryos. Toxicol Sci 2019. [PMID: 29529285 DOI: 10.1093/toxsci/kfy037] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Triphenyl phosphate (TPHP) is an unsubstituted aryl phosphate ester used as a flame retardant and plasticizer within the United States. Using zebrafish as a model, the objectives of this study were to rely on (1) mRNA-sequencing to uncover pathways disrupted following embryonic TPHP exposure and (2) high-content screening to identify nuclear receptor ligands that enhance or mitigate TPHP-induced cardiotoxicity. Based on mRNA-sequencing, TPHP exposure from 24 to 72-h postfertilization (hpf) resulted in a concentration-dependent increase in the number of transcripts significantly affected at 72 hpf, and pathway analysis revealed that 5 out of 9 nuclear receptor pathways were associated with the retinoid X receptor (RXR). Based on a screen of 74 unique nuclear receptor ligands as well as follow-up experiments, 2 compounds-ciglitazone (a peroxisome proliferator-activated receptor gamma, or PPARγ, agonist) and fenretinide (a pan-retinoic acid receptor, or RAR, agonist)-reliably mitigated TPHP-induced cardiotoxicity in the absence of effects on TPHP uptake or metabolism. As these data suggested that TPHP may be activating RXR (a heterodimer for both RARs and PPARγ), we coexposed embryos to HX 531-a pan-RXR antagonist-from 24 to 72 hpf and, contrary to our hypothesis, found that coexposure to HX 531 significantly enhanced TPHP-induced cardiotoxicity. Using a luciferase reporter assay, we also found that TPHP did not activate nor inhibit chimeric human RXRα, RXRβ, or RXRγ, suggesting that TPHP does not directly bind nor interact with RXRs. Overall, our data suggest that TPHP may interfere with RXR-dependent pathways involved in cardiac development.
Collapse
Affiliation(s)
- Constance A Mitchell
- Environmental Toxicology Graduate Program.,Department of Environmental Sciences, University of California, Riverside, California
| | - Subham Dasgupta
- Department of Environmental Sciences, University of California, Riverside, California
| | - Sharon Zhang
- Division of Environmental Sciences and Policy, Duke University, Durham, North Carolina
| | - Heather M Stapleton
- Division of Environmental Sciences and Policy, Duke University, Durham, North Carolina
| | - David C Volz
- Department of Environmental Sciences, University of California, Riverside, California
| |
Collapse
|
19
|
Chai Y, Liu W, Wang C, Rao M, Zhang Y. Prognostic Role of Chicken Ovalbumin Upstream Promoter Transcription Factor II in Isocitrate Dehydrogenase-Mutant Glioma with 1p19q Co-Deletion. J Mol Neurosci 2019; 68:234-242. [PMID: 30929126 DOI: 10.1007/s12031-019-01281-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chicken ovalbumin upstream promoter transcription factor II is known to play a crucial role in the tumor microenvironment. However, the role of NR2F2 in gliomas is unknown. METHODS The genomic and clinical data of 530 cases of lower grade gliomas (LGGs) patients and 167 cases of glioblastoma (GBM) patients in The Cancer Genome Atlas (TCGA) were extracted for analysis. R2 and UCSC Xena browser were used for Kaplan-Meier survival in the GSE16011 dataset and TCGA dataset, respectively. GraphPad Prism 7 was used to compare the differences in NR2F2 expression between various groups and subtypes. RESULTS LGG patients with low NR2F2 expression had a significantly favorable outcome compared with those with high NR2F2 expression (p < 0.05). By matching histological subtypes and gene expression profiles of LGG patients, grade II glioma group showed lowest levels of NR2F2 expression compared with grade III gliomas and GBM. Patients diagnosed with astrocytoma have highest expression of NR2F2 but lowest OS (p < 0.05). In LGGs, NR2F2 expression was significantly downregulated in patient group with IDH mutation and 1p19q co-deletion (p < 0.05). CONCLUSION Our study suggests that NR2F2 can be used as a prognostic marker in LGG patients with IDH mutation and 1p19 co-deletion.
Collapse
Affiliation(s)
- Yi Chai
- School of Clinical Medicine and Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Wei Liu
- School of Clinical Medicine and Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Caixia Wang
- School of General Practice and Continuing Education, Capital Medical University, Beijing, China
| | - Minchao Rao
- Department of Oncology, Shangrao People Hospital, Shangrao, Jiangxi, China
| | - Yuqi Zhang
- School of Clinical Medicine and Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China. .,Department of Neurosurgery, Yuquan Hospital, Tsinghua University, No. 5 Shijingshan Road, Shijingshan District, Beijing, 100040, China.
| |
Collapse
|
20
|
Wolf K, Hu H, Isaji T, Dardik A. Molecular identity of arteries, veins, and lymphatics. J Vasc Surg 2019; 69:253-262. [PMID: 30154011 PMCID: PMC6309638 DOI: 10.1016/j.jvs.2018.06.195] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Arteries, veins, and lymphatic vessels are distinguished by structural differences that correspond to their different functions. Each of these vessels is also defined by specific molecular markers that persist throughout adult life; these markers are some of the molecular determinants that control the differentiation of embryonic undifferentiated cells into arteries, veins, or lymphatics. METHODS This is a review of experimental literature. RESULTS The Eph-B4 receptor and its ligand, ephrin-B2, are critical molecular determinants of vessel identity, arising on endothelial cells early in embryonic development. Eph-B4 and ephrin-B2 continue to be expressed on adult vessels and mark vessel identity. However, after vascular surgery, vessel identity can change and is marked by altered Eph-B4 and ephrin-B2 expression. Vein grafts show loss of venous identity, with less Eph-B4 expression. Arteriovenous fistulas show gain of dual arterial-venous identity, with both Eph-B4 and ephrin-B2 expression, and manipulation of Eph-B4 improves arteriovenous fistula patency. Patches used to close arteries and veins exhibit context-dependent gain of identity, that is, patches in the arterial environment gain arterial identity, whereas patches in the venous environment gain venous identity; these results show the importance of the host infiltrating cells in determining vascular identity after vascular surgery. CONCLUSIONS Changes in the vessel's molecular identity after vascular surgery correspond to structural changes that depend on the host's postsurgical environment. Regulation of vascular identity and the underlying molecular mechanisms may allow new therapeutic approaches to improve vascular surgical procedures.
Collapse
Affiliation(s)
- Katharine Wolf
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Haidi Hu
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn; VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
21
|
Thomas JM, Surendran S, Abraham M, Sasankan D, Bhaadri S, Rajavelu A, Kartha CC. Gene expression analysis of nidus of cerebral arteriovenous malformations reveals vascular structures with deficient differentiation and maturation. PLoS One 2018; 13:e0198617. [PMID: 29897969 PMCID: PMC5999265 DOI: 10.1371/journal.pone.0198617] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 05/22/2018] [Indexed: 12/19/2022] Open
Abstract
Objective Arteriovenous malformations (AVMs) are characterised by tangles of dysplastic blood vessels which shunt blood from arteries to veins with no intervening capillary bed. It is not known at what stage of development and differentiation, AVM vessels became aberrant. To address this, we have analysed the expression of vascular differentiation, vascular maturation and brain capillary specific genes in AVM nidus. Methodology We performed immunohistochemistry and western blot analysis of vascular differentiation (HEY2, DLL4, EFNB2, and COUP-TFII), vascular maturation (ENG and KLF2) and brain capillary specific genes (GGTP and GLUT1) on ten surgically excised human brain AVMs and ten normal human brain tissues. Results Immunohistochemical analysis revealed that AVM vessels co-express both artery and vein differentiation genes. H-score analysis revealed that there is statistically significant (P < 0.0001) increase in expression of these proteins in AVM vessels compared to control vessels. These findings were further confirmed by western blot analysis and found to be statistically significant (P < 0.0001 and P < 0.001) for all proteins except Hey2. Both immunostaining and western blot analysis revealed that AVM vessels express GGTP and GLUT1, markers specific to brain capillaries. Immunofluorescent staining demonstrated that expression of KLF2, a vascular maturation marker is significantly (P <0.001) decreased in AVM vessels and was further confirmed by western blot analysis (P < 0.001). Immunohistochemical and western blot analysis demonstrated that another vascular maturation protein Endoglin had high expression in AVM vessels compared to control vessels. The results were found to be statistically significant (P < 0.0001). Summary Our findings suggest that vascular structures of AVMs co-express markers specific for arteries, veins and capillaries. We conclude that AVM nidus constitutes of aberrant vessels which are not terminally differentiated and inadequately matured.
Collapse
Affiliation(s)
- Jaya Mary Thomas
- Cardio Vascular Diseases and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sumi Surendran
- Cardio Vascular Diseases and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala, India
| | - Mathew Abraham
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Dhakshmi Sasankan
- Cardio Vascular Diseases and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala, India
| | - Sridutt Bhaadri
- Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, India
| | - Arumugam Rajavelu
- Cardio Vascular Diseases and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala, India
- Tropical Disease Biology Program, Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala, India
- * E-mail: (AR); (CCK)
| | - Chandrasekharan C. Kartha
- Cardio Vascular Diseases and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala, India
- * E-mail: (AR); (CCK)
| |
Collapse
|
22
|
Control of Blood Vessel Formation by Notch Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:319-338. [PMID: 30030834 DOI: 10.1007/978-3-319-89512-3_16] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Blood vessels span throughout the body to nourish tissue cells and to provide gateways for immune surveillance. Endothelial cells that line capillaries have the remarkable capacity to be quiescent for years but to switch rapidly into the activated state once new blood vessels need to be formed. In addition, endothelial cells generate niches for progenitor and tumor cells and provide organ-specific paracrine (angiocrine) factors that control organ development and regeneration, maintenance of homeostasis and tumor progression. Recent data indicate a pivotal role for blood vessels in responding to metabolic changes and that endothelial cell metabolism is a novel regulator of angiogenesis. The Notch pathway is the central signaling mode that cooperates with VEGF, WNT, BMP, TGF-β, angiopoietin signaling and cell metabolism to orchestrate angiogenesis, tip/stalk cell selection and arteriovenous specification. Here, we summarize the current knowledge and implications regarding the complex roles of Notch signaling during physiological and tumor angiogenesis, the dynamic nature of tip/stalk cell selection in the nascent vessel sprout and arteriovenous differentiation. Furthermore, we shed light on recent work on endothelial cell metabolism, perfusion-independent angiocrine functions of endothelial cells in organ-specific vascular beds and how manipulation of Notch signaling may be used to target the tumor vasculature.
Collapse
|
23
|
Roman BL, Hinck AP. ALK1 signaling in development and disease: new paradigms. Cell Mol Life Sci 2017; 74:4539-4560. [PMID: 28871312 PMCID: PMC5687069 DOI: 10.1007/s00018-017-2636-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/01/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Activin A receptor like type 1 (ALK1) is a transmembrane serine/threonine receptor kinase in the transforming growth factor-beta receptor family that is expressed on endothelial cells. Defects in ALK1 signaling cause the autosomal dominant vascular disorder, hereditary hemorrhagic telangiectasia (HHT), which is characterized by development of direct connections between arteries and veins, or arteriovenous malformations (AVMs). Although previous studies have implicated ALK1 in various aspects of sprouting angiogenesis, including tip/stalk cell selection, migration, and proliferation, recent work suggests an intriguing role for ALK1 in transducing a flow-based signal that governs directed endothelial cell migration within patent, perfused vessels. In this review, we present an updated view of the mechanism of ALK1 signaling, put forth a unified hypothesis to explain the cellular missteps that lead to AVMs associated with ALK1 deficiency, and discuss emerging roles for ALK1 signaling in diseases beyond HHT.
Collapse
Affiliation(s)
- Beth L Roman
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto St, Pittsburgh, PA, 15261, USA.
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
24
|
Wnt9a Is Required for the Aortic Amplification of Nascent Hematopoietic Stem Cells. Cell Rep 2017; 17:1595-1606. [PMID: 27806298 PMCID: PMC6309681 DOI: 10.1016/j.celrep.2016.10.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/07/2016] [Accepted: 10/10/2016] [Indexed: 01/08/2023] Open
Abstract
All mature blood cell types in the adult animal arise from hematopoietic stem and progenitor cells (HSPCs). However, the developmental cues regulating HSPC ontogeny are incompletely understood. In particular, the details surrounding a requirement for Wnt/β-catenin signaling in the development of mature HSPCs are controversial and difficult to consolidate. Using zebrafish, we demonstrate that Wnt signaling is required to direct an amplification of HSPCs in the aorta. Wnt9a is specifically required for this process and cannot be replaced by Wnt9b or Wnt3a. This proliferative event occurs independently of initial HSPC fate specification, and the Wnt9a input is required prior to aorta formation. HSPC arterial amplification occurs prior to seeding of secondary hematopoietic tissues and proceeds, in part, through the cell cycle regulator myca (c-myc). Our results support a general paradigm, in which early signaling events, including Wnt, direct later HSPC developmental processes. Hematopoietic stem and progenitor cells (HSPCs) give rise to all of the blood cells of the adult organism; however, how these cells are derived in vivo is still incompletely understood. Using zebrafish, Grainger et al. find that Wnt9a mediates amplification of HSPCs prior to their migration to secondary hematopoietic sites.
Collapse
|
25
|
Baldwin WS, Boswell WT, Ginjupalli G, Litoff EJ. Annotation of the Nuclear Receptors in an Estuarine Fish species, Fundulus heteroclitus. NUCLEAR RECEPTOR RESEARCH 2017; 4. [PMID: 28804711 DOI: 10.11131/2017/101285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The nuclear receptors (NRs) are ligand-dependent transcription factors that respond to various internal as well as external cues such as nutrients, pheromones, and steroid hormones that play crucial roles in regulation and maintenance of homeostasis and orchestrating the physiological and stress responses of an organism. We annotated the Fundulus heteroclitus (mummichog; Atlantic killifish) nuclear receptors. Mummichog are a non-migratory, estuarine fish with a limited home range often used in environmental research as a field model for studying ecological and evolutionary responses to variable environmental conditions such as salinity, oxygen, temperature, pH, and toxic compounds because of their hardiness. F. heteroclitus have at least 74 NRs spanning all seven gene subfamilies. F. heteroclitus is unique in that no RXRα member was found within the genome. Interestingly, some of the NRs are highly conserved between species, while others show a higher degree of divergence such as PXR, SF1, and ARα. Fundulus like other fish species show expansion of the RAR (NR1B), Rev-erb (NR1D), ROR (NR1F), COUPTF (NR2F), ERR (NR3B), RXR (NR2B), and to a lesser extent the NGF (NR4A), and NR3C steroid receptors (GR/AR). Of particular interest is the co-expansion of opposing NRs, Reverb-ROR, and RAR/RXR-COUPTF.
Collapse
Affiliation(s)
- William S Baldwin
- Biological Sciences, Clemson University, Clemson, SC 29634.,Environmental Toxicology Program, Clemson University, Clemson, SC 29634
| | | | - Gautam Ginjupalli
- Environmental Toxicology Program, Clemson University, Clemson, SC 29634
| | | |
Collapse
|
26
|
Jung HM, Castranova D, Swift MR, Pham VN, Venero Galanternik M, Isogai S, Butler MG, Mulligan TS, Weinstein BM. Development of the larval lymphatic system in zebrafish. Development 2017; 144:2070-2081. [PMID: 28506987 PMCID: PMC5482986 DOI: 10.1242/dev.145755] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 04/24/2017] [Indexed: 12/16/2022]
Abstract
The lymphatic vascular system is a hierarchically organized complex network essential for tissue fluid homeostasis, immune trafficking and absorption of dietary fats in the human body. Despite its importance, the assembly of the lymphatic network is still not fully understood. The zebrafish is a powerful model organism that enables study of lymphatic vessel development using high-resolution imaging and sophisticated genetic and experimental manipulation. Although several studies have described early lymphatic development in the fish, lymphatic development at later stages has not been completely elucidated. In this study, we generated a new Tg(mrc1a:egfp)y251 transgenic zebrafish that uses a mannose receptor, C type 1 (mrc1a) promoter to drive strong EGFP expression in lymphatic vessels at all stages of development and in adult zebrafish. We used this line to describe the assembly of the major vessels of the trunk lymphatic vascular network, including the later-developing collateral cardinal, spinal, superficial lateral and superficial intersegmental lymphatics. Our results show that major trunk lymphatic vessels are conserved in the zebrafish, and provide a thorough and complete description of trunk lymphatic vessel assembly.
Collapse
Affiliation(s)
- Hyun Min Jung
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew R Swift
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sumio Isogai
- Department of Anatomy, School of Medicine, Iwate Medical University, Morioka 020-8505, Japan
| | - Matthew G Butler
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy S Mulligan
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Comparison of Zebrafish tmem88a mutant and morpholino knockdown phenotypes. PLoS One 2017; 12:e0172227. [PMID: 28192479 PMCID: PMC5305201 DOI: 10.1371/journal.pone.0172227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 02/01/2017] [Indexed: 01/13/2023] Open
Abstract
Tmem88a is a transmembrane protein that is thought to be a negative regulator of the Wnt signalling pathway. Several groups have used antisense morpholino oligonucleotides in an effort to characterise the role of tmem88a in zebrafish cardiovascular development, but they have not obtained consistent results. Here, we generate an 8 bp deletion in the coding region of the tmem88a locus using TALENs, and we have gone on to establish a viable homozygous tmem88aΔ8 mutant line. Although tmem88aΔ8 mutants have reduced expression of some key haematopoietic genes, differentiation of erythrocytes and neutrophils is unaffected, contradicting our previous study using antisense morpholino oligonucleotides. We find that expression of the tmem88a paralogue tmem88b is not significantly changed in tmem88aΔ8 mutants and injection of the tmem88a splice-blocking morpholino oligonucleotide into tmem88aΔ8 mutants recapitulates the reduction of erythrocytes observed in morphants using o-Dianisidine. This suggests that there is a partial, but inessential, requirement for tmem88a during haematopoiesis and that morpholino injection exacerbates this phenotype in tmem88a morpholino knockdown embryos.
Collapse
|
28
|
Chu M, Li T, Shen B, Cao X, Zhong H, Zhang L, Zhou F, Ma W, Jiang H, Xie P, Liu Z, Dong N, Xu Y, Zhao Y, Xu G, Lu P, Luo J, Wu Q, Alitalo K, Koh GY, Adams RH, He Y. Angiopoietin receptor Tie2 is required for vein specification and maintenance via regulating COUP-TFII. eLife 2016; 5:21032. [PMID: 28005008 PMCID: PMC5218530 DOI: 10.7554/elife.21032] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 12/21/2016] [Indexed: 12/24/2022] Open
Abstract
Mechanisms underlying the vein development remain largely unknown. Tie2 signaling mediates endothelial cell (EC) survival and vascular maturation and its activating mutations are linked to venous malformations. Here we show that vein formation are disrupted in mouse skin and mesentery when Tie2 signals are diminished by targeted deletion of Tek either ubiquitously or specifically in embryonic ECs. Postnatal Tie2 attenuation resulted in the degeneration of newly formed veins followed by the formation of haemangioma-like vascular tufts in retina and venous tortuosity. Mechanistically, Tie2 insufficiency compromised venous EC identity, as indicated by a significant decrease of COUP-TFII protein level, a key regulator in venogenesis. Consistently, angiopoietin-1 stimulation increased COUP-TFII in cultured ECs, while Tie2 knockdown or blockade of Tie2 downstream PI3K/Akt pathway reduced COUP-TFII which could be reverted by the proteasome inhibition. Together, our results imply that Tie2 is essential for venous specification and maintenance via Akt mediated stabilization of COUP-TFII.
Collapse
Affiliation(s)
- Man Chu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Taotao Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Bin Shen
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, Nanjing, China
| | - Xudong Cao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haoyu Zhong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Luqing Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, Nanjing, China
| | - Fei Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,MOE Key Laboratory for Model Animal and Disease Study, Model Animal Research Institute, Nanjing University, Nanjing, China
| | - Wenjuan Ma
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haijuan Jiang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Pancheng Xie
- Cam-Su Genomic Resources Center, Soochow University, Suzhou, China
| | - Zhengzheng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China
| | - Ying Xu
- Cam-Su Genomic Resources Center, Soochow University, Suzhou, China
| | - Yun Zhao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China
| | - Guoqiang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Peirong Lu
- The First Affiliated Hospital, Soochow University, Suzhou, China
| | - Jincai Luo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Center of Excellence, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| | - Gou Young Koh
- Center for Vascular Research, Institute of Basic Science and Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Faculty of Medicine, University of Münster, Münster, Germany
| | - Yulong He
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Cam-Su Genomic Resources Center, Soochow University, Suzhou, China.,Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
29
|
Noyes PD, Garcia GR, Tanguay RL. ZEBRAFISH AS AN IN VIVO MODEL FOR SUSTAINABLE CHEMICAL DESIGN. GREEN CHEMISTRY : AN INTERNATIONAL JOURNAL AND GREEN CHEMISTRY RESOURCE : GC 2016; 18:6410-6430. [PMID: 28461781 PMCID: PMC5408959 DOI: 10.1039/c6gc02061e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Heightened public awareness about the many thousands of chemicals in use and present as persistent contaminants in the environment has increased the demand for safer chemicals and more rigorous toxicity testing. There is a growing recognition that the use of traditional test models and empirical approaches is impractical for screening for toxicity the many thousands of chemicals in the environment and the hundreds of new chemistries introduced each year. These realities coupled with the green chemistry movement have prompted efforts to implement more predictive-based approaches to evaluate chemical toxicity early in product development. While used for many years in environmental toxicology and biomedicine, zebrafish use has accelerated more recently in genetic toxicology, high throughput screening (HTS), and behavioral testing. This review describes major advances in these testing methods that have positioned the zebrafish as a highly applicable model in chemical safety evaluations and sustainable chemistry efforts. Many toxic responses have been shown to be shared among fish and mammals owing to their generally well-conserved development, cellular networks, and organ systems. These shared responses have been observed for chemicals that impair endocrine functioning, development, and reproduction, as well as those that elicit cardiotoxicity and carcinogenicity, among other diseases. HTS technologies with zebrafish enable screening large chemical libraries for bioactivity that provide opportunities for testing early in product development. A compelling attribute of the zebrafish centers on being able to characterize toxicity mechanisms across multiple levels of biological organization from the genome to receptor interactions and cellular processes leading to phenotypic changes such as developmental malformations. Finally, there is a growing recognition of the links between human and wildlife health and the need for approaches that allow for assessment of real world multi-chemical exposures. The zebrafish is poised to be an important model in bridging these two conventionally separate areas of toxicology and characterizing the biological effects of chemical mixtures that could augment its role in sustainable chemistry.
Collapse
Affiliation(s)
- Pamela D. Noyes
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Gloria R. Garcia
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| | - Robert L. Tanguay
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331
| |
Collapse
|
30
|
Venero Galanternik M, Stratman AN, Jung HM, Butler MG, Weinstein BM. Building the drains: the lymphatic vasculature in health and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:689-710. [PMID: 27576003 DOI: 10.1002/wdev.246] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 02/06/2023]
Abstract
The lymphatic vasculature is comprised of a network of endothelial vessels found in close proximity to but separated from the blood vasculature. An essential tissue component of all vertebrates, lymphatics are responsible for the maintenance of fluid homeostasis, dissemination of immune cells, and lipid reabsorption under healthy conditions. When lymphatic vessels are impaired due to invasive surgery, genetic disorders, or parasitic infections, severe fluid build-up accumulates in the affected tissues causing a condition known as lymphedema. Malignant tumors can also directly activate lymphangiogenesis and use these vessels to promote the spread of metastatic cells. Although their first description goes back to the times of Hippocrates, with subsequent anatomical characterization at the beginning of the 20th-century, the lack of identifying molecular markers and tools to visualize these translucent vessels meant that investigation of lymphatic vessels fell well behind research of blood vessels. However, after years under the shadow of the blood vasculature, recent advances in imaging technologies and new genetic and molecular tools have accelerated the pace of research on lymphatic vessel development. These new tools have facilitated both work in classical mammalian models and the emergence of new powerful vertebrate models like zebrafish, quickly driving the field of lymphatic development back into the spotlight. In this review, we summarize the highlights of recent research on the development and function of the lymphatic vascular network in health and disease. WIREs Dev Biol 2016, 5:689-710. doi: 10.1002/wdev.246 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Marina Venero Galanternik
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Amber N Stratman
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hyun Min Jung
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Matthew G Butler
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Brant M Weinstein
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
31
|
Abstract
The zebrafish model is the only available high-throughput vertebrate assessment system, and it is uniquely suited for studies of in vivo cell biology. A sequenced and annotated genome has revealed a large degree of evolutionary conservation in comparison to the human genome. Due to our shared evolutionary history, the anatomical and physiological features of fish are highly homologous to humans, which facilitates studies relevant to human health. In addition, zebrafish provide a very unique vertebrate data stream that allows researchers to anchor hypotheses at the biochemical, genetic, and cellular levels to observations at the structural, functional, and behavioral level in a high-throughput format. In this review, we will draw heavily from toxicological studies to highlight advances in zebrafish high-throughput systems. Breakthroughs in transgenic/reporter lines and methods for genetic manipulation, such as the CRISPR-Cas9 system, will be comprised of reports across diverse disciplines.
Collapse
Affiliation(s)
- Gloria R Garcia
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Pamela D Noyes
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Robert L Tanguay
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA.
| |
Collapse
|
32
|
Chávez MN, Aedo G, Fierro FA, Allende ML, Egaña JT. Zebrafish as an Emerging Model Organism to Study Angiogenesis in Development and Regeneration. Front Physiol 2016; 7:56. [PMID: 27014075 PMCID: PMC4781882 DOI: 10.3389/fphys.2016.00056] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/05/2016] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis is the process through which new blood vessels are formed from preexisting ones and plays a critical role in several conditions including embryonic development, tissue repair and disease. Moreover, enhanced therapeutic angiogenesis is a major goal in the field of regenerative medicine and efficient vascularization of artificial tissues and organs is one of the main hindrances in the implementation of tissue engineering approaches, while, on the other hand, inhibition of angiogenesis is a key therapeutic target to inhibit for instance tumor growth. During the last decades, the understanding of cellular and molecular mechanisms involved in this process has been matter of intense research. In this regard, several in vitro and in vivo models have been established to visualize and study migration of endothelial progenitor cells, formation of endothelial tubules and the generation of new vascular networks, while assessing the conditions and treatments that either promote or inhibit such processes. In this review, we address and compare the most commonly used experimental models to study angiogenesis in vitro and in vivo. In particular, we focus on the implementation of the zebrafish (Danio rerio) as a model to study angiogenesis and discuss the advantages and not yet explored possibilities of its use as model organism.
Collapse
Affiliation(s)
- Myra N Chávez
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität MünchenMunich, Germany; Department of Biology, FONDAP Center for Genome Regulation, Faculty of Science, Universidad de ChileSantiago, Chile; Department of Biochemistry and Molecular Biology, FONDAP Advanced Center for Chronic Diseases (ACCDiS) and Center for Molecular Studies of the Cell (CEMC), Faculty of Chemical and Pharmaceutical Sciences, Faculty of Medicine, University of ChileSantiago, Chile
| | - Geraldine Aedo
- Department of Biology, FONDAP Center for Genome Regulation, Faculty of Science, Universidad de Chile Santiago, Chile
| | - Fernando A Fierro
- Department of Cell Biology and Human Anatomy, University of California Davis, Sacramento, CA, USA
| | - Miguel L Allende
- Department of Biology, FONDAP Center for Genome Regulation, Faculty of Science, Universidad de Chile Santiago, Chile
| | - José T Egaña
- Institute for Medical and Biological Engineering, Schools of Engineering, Biological Sciences and Medicine, Pontifícia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
33
|
Wu SP, Yu CT, Tsai SY, Tsai MJ. Choose your destiny: Make a cell fate decision with COUP-TFII. J Steroid Biochem Mol Biol 2016; 157:7-12. [PMID: 26658017 PMCID: PMC4724268 DOI: 10.1016/j.jsbmb.2015.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 06/04/2015] [Accepted: 11/15/2015] [Indexed: 02/06/2023]
Abstract
Cell fate specification is a critical process to generate cells with a wide range of characteristics from stem and progenitor cells. Emerging evidence demonstrates that the orphan nuclear receptor COUP-TFII serves as a key regulator in determining the cell identity during embryonic development. The present review summarizes our current knowledge on molecular mechanisms by which COUP-TFII employs to define the cell fates, with special emphasis on cardiovascular and renal systems. These novel insights pave the road for future studies of regenerative medicine.
Collapse
Affiliation(s)
- San-Pin Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Cheng-Tai Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
34
|
Astin JW, Crosier PS. Lymphatics, Cancer and Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:199-218. [DOI: 10.1007/978-3-319-30654-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Li RF, Wu TY, Mou YZ, Wang YS, Chen CL, Wu CY. Nr2f1b control venous specification and angiogenic patterning during zebrafish vascular development. J Biomed Sci 2015; 22:104. [PMID: 26572615 PMCID: PMC4647328 DOI: 10.1186/s12929-015-0209-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/16/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The specification of vein and the patterning of intersegmental vessels (ISV) controlled by transcription factor is not fully characterized. The orphan nuclear receptor Chicken ovalbumin upstream promoter transcription factor II (CoupTFII, a.k.a NR2F2) positively regulates vein identity in mice. In this study, we show that nr2f1b is important for vein and tip cell identity during zebrafish development. RESULTS Nr2f1b mRNA is expressed in ventral lateral mesoderm at 15S stage and in vessels at 24 hpf consistent with a role in early vascular specification. Morpholino knockdown of nr2f1b results in a decrease in both vein cell number and expression of the vein specific marker flt4 and mrc1, suggested its role in venous specification. We also show loss of nr2f1b reduced ISV cell number and impairs ISV growth, which is likely due to the impairment of angiogenic cells migration and/or proliferation by time-lapse imaging. Consequently, nr2f1b morphants showed pericardial edema and circulation defects. Overexpression of nr2f1b under the fli promoter increases the number of venous cells and ISV endothelial cells indicated the function of nr2f1b is required and necessary for vascular development. We further showed that nr2f1b likely interact with Notch signalling. nr2f1b expression is increased in rbpsuh morphants and DAPT-treatment embryos suggested nr2f1b is negatively regulated by Notch activity. CONCLUSIONS We show nr2f1b control venous specification and angiogenic patterning during zebrafish vascular development, which is mediated by Notch signalings.
Collapse
Affiliation(s)
- Ru-Fang Li
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ting-Yun Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yu-Zheng Mou
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yi-Shan Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, Taiwan. .,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
36
|
Fish JE, Wythe JD. The molecular regulation of arteriovenous specification and maintenance. Dev Dyn 2015; 244:391-409. [PMID: 25641373 DOI: 10.1002/dvdy.24252] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/02/2015] [Accepted: 01/04/2015] [Indexed: 12/21/2022] Open
Abstract
The formation of a hierarchical vascular network, composed of arteries, veins, and capillaries, is essential for embryogenesis and is required for the production of new functional vasculature in the adult. Elucidating the molecular mechanisms that orchestrate the differentiation of vascular endothelial cells into arterial and venous cell fates is requisite for regenerative medicine, as the directed formation of perfused vessels is desirable in a myriad of pathological settings, such as in diabetes and following myocardial infarction. Additionally, this knowledge will enhance our understanding and treatment of vascular anomalies, such as arteriovenous malformations (AVMs). From studies in vertebrate model organisms, such as mouse, zebrafish, and chick, a number of key signaling pathways have been elucidated that are required for the establishment and maintenance of arterial and venous fates. These include the Hedgehog, Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor-β (TGF-β), Wnt, and Notch signaling pathways. In addition, a variety of transcription factor families acting downstream of, or in concert with, these signaling networks play vital roles in arteriovenous (AV) specification. These include Notch and Notch-regulated transcription factors (e.g., HEY and HES), SOX factors, Forkhead factors, β-Catenin, ETS factors, and COUP-TFII. It is becoming apparent that AV specification is a highly coordinated process that involves the intersection and carefully orchestrated activity of multiple signaling cascades and transcriptional networks. This review will summarize the molecular mechanisms that are involved in the acquisition and maintenance of AV fate, and will highlight some of the limitations in our current knowledge of the molecular machinery that directs AV morphogenesis.
Collapse
Affiliation(s)
- Jason E Fish
- Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada
| | | |
Collapse
|
37
|
Cui X, Lu YW, Lee V, Kim D, Dorsey T, Wang Q, Lee Y, Vincent P, Schwarz J, Dai G. Venous Endothelial Marker COUP-TFII Regulates the Distinct Pathologic Potentials of Adult Arteries and Veins. Sci Rep 2015; 5:16193. [PMID: 26537113 PMCID: PMC4633649 DOI: 10.1038/srep16193] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/05/2015] [Indexed: 12/21/2022] Open
Abstract
Arteries and veins have very different susceptibility to certain vascular diseases such as atherosclerosis and vascular calcification. The molecular mechanisms of these differences are not fully understood. In this study, we discovered that COUP-TFII, a transcription factor critical for establishing the venous identity during embryonic vascular development, also regulates the pathophysiological functions of adult blood vessels, especially those directly related to vascular diseases. Specifically, we found that suppression of COUP-TFII in venous ECs switched its phenotype toward pro-atherogenic by up-regulating the expression of inflammatory genes and down-regulating anti-thrombotic genes. ECs with COUP-TFII knockdown also readily undergo endothelial-to-mesenchymal transition (EndoMT) and subsequent osteogenic differentiation with dramatically increased osteogenic transcriptional program and calcium deposition. Consistently, over-expression of COUP-TFII led to the completely opposite effects. In vivo validation of these pro-atherogenic and osteogenic genes also demonstrates a broad consistent differential expression pattern in mouse aorta vs. vena cava ECs, which cannot be explained by the difference in hemodynamic flow. These data reveal phenotypic modulation by different levels of COUP-TFII in arterial and venous ECs, and suggest COUP-TFII may play an important role in the different susceptibilities of arteries and veins to vascular diseases such as atherosclerosis and vascular calcification.
Collapse
Affiliation(s)
- Xiaofeng Cui
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China 430070.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Yao Wei Lu
- The Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Vivian Lee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Diana Kim
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Taylor Dorsey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Qingjie Wang
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Young Lee
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Peter Vincent
- The Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - John Schwarz
- The Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
38
|
Kashiwada T, Fukuhara S, Terai K, Tanaka T, Wakayama Y, Ando K, Nakajima H, Fukui H, Yuge S, Saito Y, Gemma A, Mochizuki N. β-Catenin-dependent transcription is central to Bmp-mediated formation of venous vessels. Development 2015; 142:497-509. [PMID: 25564648 DOI: 10.1242/dev.115576] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
β-catenin regulates the transcription of genes involved in diverse biological processes, including embryogenesis, tissue homeostasis and regeneration. Endothelial cell (EC)-specific gene-targeting analyses in mice have revealed that β-catenin is required for vascular development. However, the precise function of β-catenin-mediated gene regulation in vascular development is not well understood, since β-catenin regulates not only gene expression but also the formation of cell-cell junctions. To address this question, we have developed a novel transgenic zebrafish line that allows the visualization of β-catenin transcriptional activity specifically in ECs and discovered that β-catenin-dependent transcription is central to the bone morphogenetic protein (Bmp)-mediated formation of venous vessels. During caudal vein (CV) formation, Bmp induces the expression of aggf1, a putative causative gene for Klippel-Trenaunay syndrome, which is characterized by venous malformation and hypertrophy of bones and soft tissues. Subsequently, Aggf1 potentiates β-catenin transcriptional activity by acting as a transcriptional co-factor, suggesting that Bmp evokes β-catenin-mediated gene expression through Aggf1 expression. Bmp-mediated activation of β-catenin induces the expression of Nr2f2 (also known as Coup-TFII), a member of the nuclear receptor superfamily, to promote the differentiation of venous ECs, thereby contributing to CV formation. Furthermore, β-catenin stimulated by Bmp promotes the survival of venous ECs, but not that of arterial ECs. Collectively, these results indicate that Bmp-induced activation of β-catenin through Aggf1 regulates CV development by promoting the Nr2f2-dependent differentiation of venous ECs and their survival. This study demonstrates, for the first time, a crucial role of β-catenin-mediated gene expression in the development of venous vessels.
Collapse
Affiliation(s)
- Takeru Kashiwada
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Shigetomo Fukuhara
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Kenta Terai
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Toru Tanaka
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Yuki Wakayama
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Koji Ando
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Shinya Yuge
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Yoshinobu Saito
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan JST-CREST, National Cerebral and Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| |
Collapse
|
39
|
Reverse genetic screening reveals poor correlation between morpholino-induced and mutant phenotypes in zebrafish. Dev Cell 2014; 32:97-108. [PMID: 25533206 DOI: 10.1016/j.devcel.2014.11.018] [Citation(s) in RCA: 579] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 08/19/2014] [Accepted: 11/10/2014] [Indexed: 12/16/2022]
Abstract
The widespread availability of programmable site-specific nucleases now enables targeted gene disruption in the zebrafish. In this study, we applied site-specific nucleases to generate zebrafish lines bearing individual mutations in more than 20 genes. We found that mutations in only a small proportion of genes caused defects in embryogenesis. Moreover, mutants for ten different genes failed to recapitulate published Morpholino-induced phenotypes (morphants). The absence of phenotypes in mutant embryos was not likely due to maternal effects or failure to eliminate gene function. Consistently, a comparison of published morphant defects with the Sanger Zebrafish Mutation Project revealed that approximately 80% of morphant phenotypes were not observed in mutant embryos, similar to our mutant collection. Based on these results, we suggest that mutant phenotypes become the standard metric to define gene function in zebrafish, after which Morpholinos that recapitulate respective phenotypes could be reliably applied for ancillary analyses.
Collapse
|
40
|
Corada M, Morini MF, Dejana E. Signaling pathways in the specification of arteries and veins. Arterioscler Thromb Vasc Biol 2014; 34:2372-7. [PMID: 25169934 DOI: 10.1161/atvbaha.114.303218] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The establishment of arterial and venous identity of endothelial cells is critical for the proper anatomic configuration and function of the vascular tree. Arterial and venous specification of endothelial cells is determined by genetic factors, although surrounding cells and hemodynamic forces may also contribute to vascular remodeling. This review provides an overview of the signaling pathways and related transcription factors implicated in differentiation of endothelial cells. We will discuss, in particular, the role of upstream and downstream effectors of Wnt, Sox, and Notch pathways. The understanding of the molecular mechanisms that orchestrate endothelial differentiation may have therapeutic relevance for diseases such as atherosclerosis, arteriovenous malformations, aneurysms, and others.
Collapse
Affiliation(s)
- Monica Corada
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.C., M.F.M., E.D.); and Department of Biosciences, University of Milan, Milan, Italy (E.D.)
| | - Marco Francesco Morini
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.C., M.F.M., E.D.); and Department of Biosciences, University of Milan, Milan, Italy (E.D.)
| | - Elisabetta Dejana
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.C., M.F.M., E.D.); and Department of Biosciences, University of Milan, Milan, Italy (E.D.).
| |
Collapse
|
41
|
Wu BJ, Chiu CC, Chen CL, Wang WD, Wang JH, Wen ZH, Liu W, Chang HW, Wu CY. Nuclear receptor subfamily 2 group F member 1a (nr2f1a) is required for vascular development in zebrafish. PLoS One 2014; 9:e105939. [PMID: 25157918 PMCID: PMC4144922 DOI: 10.1371/journal.pone.0105939] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 07/31/2014] [Indexed: 12/17/2022] Open
Abstract
Genetic regulators and signaling pathways are important for the formation of blood vessels. Transcription factors controlling vein identity, intersegmental vessels (ISV) growth and caudal vein plexus (CVP) formation in zebrafish are little understood as yet. Here, we show the importance of the nuclear receptor subfamily member 1A (nr2f1a) in zebrafish vascular development. Amino acid sequence alignment and phylogenetic analysis of nr2f1a is highly conserved among the vertebrates. Our in situ hybridization results showed nr2f1a mRNA is expressed in the lateral plate mesoderm at 18 somite stage and in vessels at 24-30 hpf, suggesting its roles in vasculization. Consistent with this morpholino-based knockdown of nr2fla impaired ISV growth and failed to develop fenestrated vascular structure in CVP, suggesting that nr2f1a has important roles in controlling ISV and CVP growth. Consequently, nr2f1a morphants showed pericardial edema and circulation defects. We further demonstrated reduced ISV cells and decreased CVP endothelial cells sprouting in nr2f1a morphants, indicating the growth impairment of ISV and CVP is due to a decrease of cell proliferation and migration, but not results from cell death in endothelial cells after morpholino knockdown. To test molecular mechanisms and signals that are associated with nr2f1a, we examined the expression of vascular markers. We found that a loss of nr2f1a results in a decreased expression of vein/ISV specific markers, flt4, mrc1, vascular markers stabilin and ephrinb2. This indicates the regulatory role of nr2f1a in controlling vascular development. We further showed that nr2f1a likely interact with Notch signaling by examining nr2f1a expression in rbpsuh morphants and DAPT-treatment embryos. Together, we show nr2f1a plays a critical role for vascular development in zebrafish.
Collapse
Affiliation(s)
- Bao-Jueng Wu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Marine Biotechnology Doctoral Program, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wen-Der Wang
- Department of Bioagricultural Science, National Chiayi University, Chiayi, Taiwan
| | - Jia-Hong Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Marine Biotechnology Doctoral Program, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Marine Biotechnology Doctoral Program, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|