1
|
Kim MS, Lee R, Lee DH, Song H, Ha T, Kim JK, Kang BY, Agger K, Helin K, Shin D, Kang Y, Park C. ETV2/ER71 regulates hematovascular lineage generation and vascularization through an H3K9 demethylase, KDM4A. iScience 2025; 28:111538. [PMID: 39811655 PMCID: PMC11732216 DOI: 10.1016/j.isci.2024.111538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 12/15/2023] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
ETV2/ER71, an ETS (E-twenty six) transcription factor, is critical for hematopoiesis and vascular development. However, research about the molecular mechanisms behind ETV2-mediated gene transcription is limited. Herein, we demonstrate that ETV2 and KDM4A, an H3K9 demethylase, regulate hematopoietic and endothelial genes. Etv2 -/- mouse embryonic stem cells (mESCs), which fail to generate hematopoietic and endothelial cells, exhibit enhanced H3K9me3 levels in hematopoietic and endothelial genes. ETV2 interacts with KDM4A, and the ETV2-mediated transcriptional activation of hematopoietic and endothelial genes depends on KDM4A histone demethylase activity. The ETV2 and KDM4A complex binds to the transcription regulatory regions of genes directly regulated by ETV2. Mice lacking Kdm4a and Etv2 in endothelial cells (Cdh5Cre:Kdm:Etv2 f/f mice) display a more severe perfusion recovery and neovascularization defect, compared with Cdh5Cre:Kdm4a f/f mice, Cdh5Cre:Etv2 f/f mice, and controls. Collectively, we demonstrate that ETV2 interacts with KDM4A, and that this interaction is critical for hematovascular lineage generation and vascular regeneration.
Collapse
Affiliation(s)
- Min Seong Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Raham Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Dong Hun Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biological Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Heesang Song
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju, Republic of Korea
| | - Taekyung Ha
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Joo Kyung Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Bum-Yong Kang
- Department of Medicine, Emory University School of Medicine, and Atlanta VA HCS, Atlanta, GA, USA
| | - Karl Agger
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donghyun Shin
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Changwon Park
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| |
Collapse
|
2
|
Verma M, Asakura Y, Wang X, Zhou K, Ünverdi M, Kann AP, Krauss RS, Asakura A. Endothelial cell signature in muscle stem cells validated by VEGFA-FLT1-AKT1 axis promoting survival of muscle stem cell. eLife 2024; 13:e73592. [PMID: 38842166 PMCID: PMC11216748 DOI: 10.7554/elife.73592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/05/2024] [Indexed: 06/07/2024] Open
Abstract
Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.
Collapse
Affiliation(s)
- Mayank Verma
- Department of Pediatrics & Neurology, Division of Pediatric Neurology, The University of Texas Southwestern Medical CenterDallasUnited States
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Xuerui Wang
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Kasey Zhou
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Mahmut Ünverdi
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Graduate School of Biomedical Sciencesf, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Graduate School of Biomedical Sciencesf, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| |
Collapse
|
3
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
4
|
Shang X, Jin Y, Xue Y, Pan X, Zhu H, Meng X, Cao Z, Rui Y. Overexpression of ETV2 in BMSCs promoted wound healing in cutaneous wound mice by triggering the differentiation of BMSCs into endothelial cells and modulating the transformation of M1 phenotype macrophages to M2 phenotype macrophages. Tissue Cell 2024; 87:102334. [PMID: 38430850 DOI: 10.1016/j.tice.2024.102334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
This study aimed to investigate the effects of E26-transformation-specific variant-2 (ETV2) overexpression on wound healing in a cutaneous wound (CW) model and clarify associated mechanisms. pLVX-ETV2 lentivirus expressing ETV2 was constructed and infected into BMSCs to generate ETV2-overexpressed BMSCs (BMSCs+pLVX+ETV2). The RT-PCR assay was applied to amplify ETV2, VE-cadherin, vWF, ARG-1, IL-6, iNOS, TGF-β, IL-10, TNF-α. Western blot was used to determine expression of VE-cadherin and vWF. ETV2 induced differentiation of BMSCs into ECs by increasing CDH5/CD31, triggering tube-like structures, inducing Dil-Ac-LDL positive BMSCs. ETV2 overexpression increased the gene transcription and expression of VE-cadherin and vWF (P<0.01). Transcription of M1 phenotype specific iNOS gene was lower and transcription of M2 phenotype specific ARG-1 gene was higher in the RAW264.7+BMSCs+ETV2 group compared to the RAW264.7+BMSCs+pLVX group (P<0.01). ETV2 overexpression (RAW264.7+BMSCs+ETV2) downregulated IL-6 and TNF-α, and upregulated IL-10 and TGF-β gene transcription compared to RAW264.7+BMSCs+pLVX group (P<0.01). ETV2-overexpressed BMSCs promoted wound healing in CW mice and triggered the migration of BMSCs to the wound region and macrophage activation. ETV2-overexpressed BMSCs promoted collagen fibers and blood vessel formation in the wound region of CW mice. In conclusion, this study revealed a novel biofunction of ETV2 molecule in the wound healing process. ETV2 overexpression in BMSCs promoted wound healing in CW mice by triggering BMSCs differentiation into endothelial cells and modulating the transformation of M1 pro-inflammatory and M2 anti-inflammatory macrophages in vitro and in vivo.
Collapse
Affiliation(s)
- Xiuchao Shang
- Medical College, Soochow University, Suzhou. China; The First People's Hospital of Lianyungang, Lianyungang, China
| | - Yesheng Jin
- Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, China
| | - Yuan Xue
- Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, China
| | - Xiaoyun Pan
- Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, China
| | - Haiquan Zhu
- The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiangsheng Meng
- The First People's Hospital of Lianyungang, Lianyungang, China
| | - Zhihai Cao
- Medical College, Soochow University, Suzhou. China
| | - Yongjun Rui
- Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, China.
| |
Collapse
|
5
|
Huang NF, Stern B, Oropeza BP, Zaitseva TS, Paukshto MV, Zoldan J. Bioengineering Cell Therapy for Treatment of Peripheral Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:e66-e81. [PMID: 38174560 PMCID: PMC10923024 DOI: 10.1161/atvbaha.123.318126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Peripheral artery disease is an atherosclerotic disease associated with limb ischemia that necessitates limb amputation in severe cases. Cell therapies comprised of adult mononuclear or stromal cells have been clinically tested and show moderate benefits. Bioengineering strategies can be applied to modify cell behavior and function in a controllable fashion. Using mechanically tunable or spatially controllable biomaterials, we highlight examples in which biomaterials can increase the survival and function of the transplanted cells to improve their revascularization efficacy in preclinical models. Biomaterials can be used in conjunction with soluble factors or genetic approaches to further modulate the behavior of transplanted cells and the locally implanted tissue environment in vivo. We critically assess the advances in bioengineering strategies such as 3-dimensional bioprinting and immunomodulatory biomaterials that can be applied to the treatment of peripheral artery disease and then discuss the current challenges and future directions in the implementation of bioengineering strategies.
Collapse
Affiliation(s)
- Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Brett Stern
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | | | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| |
Collapse
|
6
|
Sato Y, Asahi T, Kataoka K. Integrative single-cell RNA-seq analysis of vascularized cerebral organoids. BMC Biol 2023; 21:245. [PMID: 37940920 PMCID: PMC10634128 DOI: 10.1186/s12915-023-01711-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/25/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Cerebral organoids are three-dimensional in vitro cultured brains that mimic the function and structure of the human brain. One of the major challenges for cerebral organoids is the lack of functional vasculature. Without perfusable vessels, oxygen and nutrient supplies may be insufficient for long-term culture, hindering the investigation of the neurovascular interactions. Recently, several strategies for the vascularization of human cerebral organoids have been reported. However, the generalizable trends and variability among different strategies are unclear due to the lack of a comprehensive characterization and comparison of these vascularization strategies. In this study, we aimed to explore the effect of different vascularization strategies on the nervous system and vasculature in human cerebral organoids. RESULTS We integrated single-cell RNA sequencing data of multiple vascularized and vascular organoids and fetal brains from publicly available datasets and assessed the protocol-dependent and culture-day-dependent effects on the cell composition and transcriptomic profiles in neuronal and vascular cells. We revealed the similarities and uniqueness of multiple vascularization strategies and demonstrated the transcriptomic effects of vascular induction on neuronal and mesodermal-like cell populations. Moreover, our data suggested that the interaction between neurons and mesodermal-like cell populations is important for the cerebrovascular-specific profile of endothelial-like cells. CONCLUSIONS This study highlights the current challenges to vascularization strategies in human cerebral organoids and offers a benchmark for the future fabrication of vascularized organoids.
Collapse
Affiliation(s)
- Yuya Sato
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Toru Asahi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.
- Comprehensive Research Organization, Waseda University, Tokyo, Japan.
- Research Organization for Nano & Life Innovation, Waseda University, Tokyo, Japan.
| | - Kosuke Kataoka
- Comprehensive Research Organization, Waseda University, Tokyo, Japan.
| |
Collapse
|
7
|
Sahai-Hernandez P, Pouget C, Eyal S, Svoboda O, Chacon J, Grimm L, Gjøen T, Traver D. Dermomyotome-derived endothelial cells migrate to the dorsal aorta to support hematopoietic stem cell emergence. eLife 2023; 12:e58300. [PMID: 37695317 PMCID: PMC10495111 DOI: 10.7554/elife.58300] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/03/2023] [Indexed: 09/12/2023] Open
Abstract
Development of the dorsal aorta is a key step in the establishment of the adult blood-forming system, since hematopoietic stem and progenitor cells (HSPCs) arise from ventral aortic endothelium in all vertebrate animals studied. Work in zebrafish has demonstrated that arterial and venous endothelial precursors arise from distinct subsets of lateral plate mesoderm. Here, we profile the transcriptome of the earliest detectable endothelial cells (ECs) during zebrafish embryogenesis to demonstrate that tissue-specific EC programs initiate much earlier than previously appreciated, by the end of gastrulation. Classic studies in the chick embryo showed that paraxial mesoderm generates a subset of somite-derived endothelial cells (SDECs) that incorporate into the dorsal aorta to replace HSPCs as they exit the aorta and enter circulation. We describe a conserved program in the zebrafish, where a rare population of endothelial precursors delaminates from the dermomyotome to incorporate exclusively into the developing dorsal aorta. Although SDECs lack hematopoietic potential, they act as a local niche to support the emergence of HSPCs from neighboring hemogenic endothelium. Thus, at least three subsets of ECs contribute to the developing dorsal aorta: vascular ECs, hemogenic ECs, and SDECs. Taken together, our findings indicate that the distinct spatial origins of endothelial precursors dictate different cellular potentials within the developing dorsal aorta.
Collapse
Affiliation(s)
- Pankaj Sahai-Hernandez
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Claire Pouget
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Shai Eyal
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Ondrej Svoboda
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
- Department of Cell Differentiation, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic v.v.i, Prague, Czech Republic
| | - Jose Chacon
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Lin Grimm
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| | - Tor Gjøen
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - David Traver
- Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, United States
| |
Collapse
|
8
|
Steimle JD, Kim C, Rowton M, Nadadur RD, Wang Z, Stocker M, Hoffmann AD, Hanson E, Kweon J, Sinha T, Choi K, Black BL, Cunningham JM, Moskowitz IP, Ikegami K. ETV2 primes hematoendothelial gene enhancers prior to hematoendothelial fate commitment. Cell Rep 2023; 42:112665. [PMID: 37330911 PMCID: PMC10592526 DOI: 10.1016/j.celrep.2023.112665] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 03/14/2023] [Accepted: 06/02/2023] [Indexed: 06/20/2023] Open
Abstract
Mechanisms underlying distinct specification, commitment, and differentiation phases of cell fate determination remain undefined due to difficulties capturing these processes. Here, we interrogate the activity of ETV2, a transcription factor necessary and sufficient for hematoendothelial differentiation, within isolated fate intermediates. We observe transcriptional upregulation of Etv2 and opening of ETV2-binding sites, indicating new ETV2 binding, in a common cardiac-hematoendothelial progenitor population. Accessible ETV2-binding sites are active at the Etv2 locus but not at other hematoendothelial regulator genes. Hematoendothelial commitment coincides with the activation of a small repertoire of previously accessible ETV2-binding sites at hematoendothelial regulators. Hematoendothelial differentiation accompanies activation of a large repertoire of new ETV2-binding sites and upregulation of hematopoietic and endothelial gene regulatory networks. This work distinguishes specification, commitment, and sublineage differentiation phases of ETV2-dependent transcription and suggests that the shift from ETV2 binding to ETV2-bound enhancer activation, not ETV2 binding to target enhancers, drives hematoendothelial fate commitment.
Collapse
Affiliation(s)
- Jeffrey D Steimle
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Chul Kim
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA; Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Megan Rowton
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Rangarajan D Nadadur
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Zhezhen Wang
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Matthew Stocker
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Andrew D Hoffmann
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Erika Hanson
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Junghun Kweon
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John M Cunningham
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Ivan P Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA.
| | - Kohta Ikegami
- Division of Molecular and Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
9
|
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
|
10
|
Kim TM, Lee RH, Kim MS, Lewis CA, Park C. ETV2/ER71, the key factor leading the paths to vascular regeneration and angiogenic reprogramming. Stem Cell Res Ther 2023; 14:41. [PMID: 36927793 PMCID: PMC10019431 DOI: 10.1186/s13287-023-03267-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Extensive efforts have been made to achieve vascular regeneration accompanying tissue repair for treating vascular dysfunction-associated diseases. Recent advancements in stem cell biology and cell reprogramming have opened unforeseen opportunities to promote angiogenesis in vivo and generate autologous endothelial cells (ECs) for clinical use. We have, for the first time, identified a unique endothelial-specific transcription factor, ETV2/ER71, and revealed its essential role in regulating endothelial cell generation and function, along with vascular regeneration and tissue repair. Furthermore, we and other groups have demonstrated its ability to directly reprogram terminally differentiated non-ECs into functional ECs, proposing ETV2/ER71 as an effective therapeutic target for vascular diseases. In this review, we discuss the up-to-date status of studies on ETV2/ER71, spanning from its molecular mechanism to vasculo-angiogenic role and direct cell reprogramming toward ECs. Furthermore, we discuss future directions to deploy the clinical potential of ETV2/ER71 as a novel and potent target for vascular disorders such as cardiovascular disease, neurovascular impairment and cancer.
Collapse
Affiliation(s)
- Tae Min Kim
- Graduate School of International Agricultural Technology and Institutes of Green-Bio Science and Technology, Seoul National University, 1447 Pyeongchang-daero, Pyeongchang, Gangwon-do, 25354, Republic of Korea.
| | - Ra Ham Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Min Seong Kim
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chloe A Lewis
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Changwon Park
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
11
|
Sierra-Pagan JE, Garry DJ. The regulatory role of pioneer factors during cardiovascular lineage specification – A mini review. Front Cardiovasc Med 2022; 9:972591. [PMID: 36082116 PMCID: PMC9445115 DOI: 10.3389/fcvm.2022.972591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease (CVD) remains the number one cause of death worldwide. Ischemic heart disease contributes to heart failure and has considerable morbidity and mortality. Therefore, alternative therapeutic strategies are urgently needed. One class of epigenetic regulators known as pioneer factors has emerged as an important tool for the development of regenerative therapies for the treatment of CVD. Pioneer factors bind closed chromatin and remodel it to drive lineage specification. Here, we review pioneer factors within the cardiovascular lineage, particularly during development and reprogramming and highlight the implications this field of research has for the future development of cardiac specific regenerative therapies.
Collapse
Affiliation(s)
- Javier E. Sierra-Pagan
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Daniel J. Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Daniel J. Garry
| |
Collapse
|
12
|
Koyano-Nakagawa N, Gong W, Das S, Theisen JWM, Swanholm TB, Van Ly D, Dsouza N, Singh BN, Kawakami H, Young S, Chen KQ, Kawakami Y, Garry DJ. Etv2 regulates enhancer chromatin status to initiate Shh expression in the limb bud. Nat Commun 2022; 13:4221. [PMID: 35864091 PMCID: PMC9304341 DOI: 10.1038/s41467-022-31848-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 07/05/2022] [Indexed: 12/02/2022] Open
Abstract
Sonic hedgehog (Shh) is essential for limb development, and the mechanisms that govern the propagation and maintenance of its expression has been well studied; however, the mechanisms that govern the initiation of Shh expression are incomplete. Here we report that ETV2 initiates Shh expression by changing the chromatin status of the developmental limb enhancer, ZRS. Etv2 expression precedes Shh in limb buds, and Etv2 inactivation prevents the opening of limb chromatin, including the ZRS, resulting in an absence of Shh expression. Etv2 overexpression in limb buds causes nucleosomal displacement at the ZRS, ectopic Shh expression, and polydactyly. Areas of nucleosome displacement coincide with ETS binding site clusters. ETV2 also functions as a transcriptional activator of ZRS and is antagonized by ETV4/5 repressors. Known human polydactyl mutations introduce novel ETV2 binding sites in the ZRS, suggesting that ETV2 dosage regulates ZRS activation. These studies identify ETV2 as a pioneer transcription factor (TF) regulating the onset of Shh expression, having both a chromatin regulatory role and a transcriptional activation role.
Collapse
Affiliation(s)
- Naoko Koyano-Nakagawa
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA
- Hamre, Schumann, Mueller & Larson, P.C., Minneapolis, MN, 55402, USA
- Mitchell Hamline School of Law, St. Paul, MN, 55105, USA
| | - Wuming Gong
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Satyabrata Das
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joshua W M Theisen
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Tran B Swanholm
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Daniel Van Ly
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Nikita Dsouza
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Bhairab N Singh
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hiroko Kawakami
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Samantha Young
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Katherine Q Chen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yasuhiko Kawakami
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
13
|
Martin BL. Mesoderm induction and patterning: Insights from neuromesodermal progenitors. Semin Cell Dev Biol 2022; 127:37-45. [PMID: 34840081 PMCID: PMC9130346 DOI: 10.1016/j.semcdb.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 12/23/2022]
Abstract
The discovery of mesoderm inducing signals helped usher in the era of molecular developmental biology, and today the mechanisms of mesoderm induction and patterning are still intensely studied. Mesoderm induction begins during gastrulation, but recent evidence in vertebrates shows that this process continues after gastrulation in a group of posteriorly localized cells called neuromesodermal progenitors (NMPs). NMPs reside within the post-gastrulation embryonic structure called the tailbud, where they make a lineage decision between ectoderm (spinal cord) and mesoderm. The majority of NMP-derived mesoderm generates somites, but also contributes to lateral mesoderm fates such as endothelium. The discovery of NMPs provides a new paradigm in which to study vertebrate mesoderm induction. This review will discuss mechanisms of mesoderm induction within NMPs, and how they have informed our understanding of mesoderm induction more broadly within vertebrates as well as animal species outside of the vertebrate lineage. Special focus will be given to the signaling networks underlying NMP-derived mesoderm induction and patterning, as well as emerging work on the significance of partial epithelial-mesenchymal states in coordinating cell fate and morphogenesis.
Collapse
Affiliation(s)
- Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
14
|
Panara V, Monteiro R, Koltowska K. Epigenetic Regulation of Endothelial Cell Lineages During Zebrafish Development-New Insights From Technical Advances. Front Cell Dev Biol 2022; 10:891538. [PMID: 35615697 PMCID: PMC9125237 DOI: 10.3389/fcell.2022.891538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/10/2022] [Indexed: 01/09/2023] Open
Abstract
Epigenetic regulation is integral in orchestrating the spatiotemporal regulation of gene expression which underlies tissue development. The emergence of new tools to assess genome-wide epigenetic modifications has enabled significant advances in the field of vascular biology in zebrafish. Zebrafish represents a powerful model to investigate the activity of cis-regulatory elements in vivo by combining technologies such as ATAC-seq, ChIP-seq and CUT&Tag with the generation of transgenic lines and live imaging to validate the activity of these regulatory elements. Recently, this approach led to the identification and characterization of key enhancers of important vascular genes, such as gata2a, notch1b and dll4. In this review we will discuss how the latest technologies in epigenetics are being used in the zebrafish to determine chromatin states and assess the function of the cis-regulatory sequences that shape the zebrafish vascular network.
Collapse
Affiliation(s)
- Virginia Panara
- Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rui Monteiro
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Centre of Genome Biology, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
15
|
Zhang H, Yamaguchi T, Kokubu Y, Kawabata K. Transient ETV2 Expression Promotes the Generation of Mature Endothelial Cells from Human Pluripotent Stem Cells. Biol Pharm Bull 2022; 45:483-490. [DOI: 10.1248/bpb.b21-00929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hongyan Zhang
- Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Tomoko Yamaguchi
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health, and Nutrition
| | - Yasuhiro Kokubu
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health, and Nutrition
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health, and Nutrition
| |
Collapse
|
16
|
Abdel-Raouf KMA, Rezgui R, Stefanini C, Teo JCM, Christoforou N. Transdifferentiation of Human Fibroblasts into Skeletal Muscle Cells: Optimization and Assembly into Engineered Tissue Constructs through Biological Ligands. BIOLOGY 2021; 10:biology10060539. [PMID: 34208436 PMCID: PMC8235639 DOI: 10.3390/biology10060539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Engineered human skeletal muscle tissue is a platform tool that can help scientists and physicians better understand human physiology, pharmacology, and disease modeling. Over the past few years this area of research has been actively being pursued by many labs worldwide. Significant challenges remain, including accessing an adequate cell source, and achieving proper physiological-like architecture of the engineered tissue. To address cell resourcing we aimed at further optimizing a process called transdifferentiation which involves the direct conversion of fibroblasts into skeletal muscle cells. The opportunity here is that fibroblasts are readily available and can be expanded sufficiently to meet the needs of a tissue engineering approach. Additionally, we aimed to demonstrate the applicability of transdifferentiation in assembling tissue engineered skeletal muscle. We implemented a screening process of protein ligands in an effort to refine transdifferentiation, and identified that most proteins resulted in a deficit in transdifferentiation efficiency, although one resulted in robust expansion of cultured cells. We were also successful in assembling engineered constructs consisting of transdifferentiated cells. Future directives involve demonstrating that the engineered tissues are capable of contractile and functional activity, and pursuit of optimizing factors such as electrical and chemical exposure, towards achieving physiological parameters observed in human muscle. Abstract The development of robust skeletal muscle models has been challenging due to the partial recapitulation of human physiology and architecture. Reliable and innovative 3D skeletal muscle models recently described offer an alternative that more accurately captures the in vivo environment but require an abundant cell source. Direct reprogramming or transdifferentiation has been considered as an alternative. Recent reports have provided evidence for significant improvements in the efficiency of derivation of human skeletal myotubes from human fibroblasts. Herein we aimed at improving the transdifferentiation process of human fibroblasts (tHFs), in addition to the differentiation of murine skeletal myoblasts (C2C12), and the differentiation of primary human skeletal myoblasts (HSkM). Differentiating or transdifferentiating cells were exposed to single or combinations of biological ligands, including Follistatin, GDF8, FGF2, GDF11, GDF15, hGH, TMSB4X, BMP4, BMP7, IL6, and TNF-α. These were selected for their critical roles in myogenesis and regeneration. C2C12 and tHFs displayed significant differentiation deficits when exposed to FGF2, BMP4, BMP7, and TNF-α, while proliferation was significantly enhanced by FGF2. When exposed to combinations of ligands, we observed consistent deficit differentiation when TNF-α was included. Finally, our direct reprogramming technique allowed for the assembly of elongated, cross-striated, and aligned tHFs within tissue-engineered 3D skeletal muscle constructs. In conclusion, we describe an efficient system to transdifferentiate human fibroblasts into myogenic cells and a platform for the generation of tissue-engineered constructs. Future directions will involve the evaluation of the functional characteristics of these engineered tissues.
Collapse
Affiliation(s)
- Khaled M. A. Abdel-Raouf
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Department of Biology, American University in Cairo, New Cairo 11835, Egypt
- Correspondence: (K.M.A.A.-R.); (N.C.)
| | - Rachid Rezgui
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Cesare Stefanini
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Jeremy C. M. Teo
- Department of Mechanical and Biomedical Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Nicolas Christoforou
- Pfizer Inc., Rare Disease Research Unit, 610 Main Street, Cambridge, MA 02139, USA
- Correspondence: (K.M.A.A.-R.); (N.C.)
| |
Collapse
|
17
|
Ho C, Morsut L. Novel synthetic biology approaches for developmental systems. Stem Cell Reports 2021; 16:1051-1064. [PMID: 33979593 PMCID: PMC8185972 DOI: 10.1016/j.stemcr.2021.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
Recently, developmental systems are investigated with increasing technological power. Still, open questions remain, especially concerning self-organization capacity and its control. Here, we present three areas where synthetic biology tools are used in top-down and bottom-up approaches for studying and constructing developmental systems. First, we discuss how synthetic biology tools can improve stem cell-based organoid models. Second, we discuss recent studies employing user-defined perturbations to study embryonic patterning in model species. Third, we present "toy models" of patterning and morphogenesis using synthetic genetic circuits in non-developmental systems. Finally, we discuss how these tools and approaches can specifically benefit the field of embryo models.
Collapse
Affiliation(s)
- Christine Ho
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Leonardo Morsut
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
18
|
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
|
19
|
Mao A, Zhang M, Li L, Liu J, Ning G, Cao Y, Wang Q. Pharyngeal pouches provide a niche microenvironment for arch artery progenitor specification. Development 2021; 148:dev.192658. [PMID: 33334861 PMCID: PMC7847271 DOI: 10.1242/dev.192658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/10/2020] [Indexed: 11/20/2022]
Abstract
The paired pharyngeal arch arteries (PAAs) are transient blood vessels connecting the heart with the dorsal aorta during embryogenesis. Although PAA malformations often occur along with pharyngeal pouch defects, the functional interaction between these adjacent tissues remains largely unclear. Here, we report that pharyngeal pouches are essential for PAA progenitor specification in zebrafish embryos. We reveal that the segmentation of pharyngeal pouches coincides spatiotemporally with the emergence of PAA progenitor clusters. These pouches physically associate with pharyngeal mesoderm in discrete regions and provide a niche microenvironment for PAA progenitor commitment by expressing BMP proteins. Specifically, pouch-derived BMP2a and BMP5 are the primary niche cues responsible for activating the BMP/Smad pathway in pharyngeal mesoderm, thereby promoting progenitor specification. In addition, BMP2a and BMP5 play an inductive function in the expression of the cloche gene npas4l in PAA progenitors. cloche mutants exhibit a striking failure to specify PAA progenitors and display ectopic expression of head muscle markers in the pharyngeal mesoderm. Therefore, our results support a crucial role for pharyngeal pouches in establishing a progenitor niche for PAA morphogenesis via BMP2a/5 expression.
Collapse
Affiliation(s)
- Aihua Mao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingming Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Linwei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
20
|
Chestnut B, Casie Chetty S, Koenig AL, Sumanas S. Single-cell transcriptomic analysis identifies the conversion of zebrafish Etv2-deficient vascular progenitors into skeletal muscle. Nat Commun 2020; 11:2796. [PMID: 32493965 PMCID: PMC7271194 DOI: 10.1038/s41467-020-16515-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 04/29/2020] [Indexed: 01/09/2023] Open
Abstract
Cell fate decisions involved in vascular and hematopoietic embryonic development are still poorly understood. An ETS transcription factor Etv2 functions as an evolutionarily conserved master regulator of vasculogenesis. Here we report a single-cell transcriptomic analysis of hematovascular development in wild-type and etv2 mutant zebrafish embryos. Distinct transcriptional signatures of different types of hematopoietic and vascular progenitors are identified using an etv2ci32Gt gene trap line, in which the Gal4 transcriptional activator is integrated into the etv2 gene locus. We observe a cell population with a skeletal muscle signature in etv2-deficient embryos. We demonstrate that multiple etv2ci32Gt; UAS:GFP cells differentiate as skeletal muscle cells instead of contributing to vasculature in etv2-deficient embryos. Wnt and FGF signaling promote the differentiation of these putative multipotent etv2 progenitor cells into skeletal muscle cells. We conclude that etv2 actively represses muscle differentiation in vascular progenitors, thus restricting these cells to a vascular endothelial fate.
Collapse
Affiliation(s)
- Brendan Chestnut
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Satish Casie Chetty
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Andrew L Koenig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.,Center for Cardiovascular Research, Washington University School of Medicine, 660S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
21
|
Yan G, Yan R, Chen C, Chen C, Zhao Y, Qin W, Veldman MB, Li S, Lin S. Engineering vascularized skeletal muscle tissue with transcriptional factor ETV2-induced autologous endothelial cells. Protein Cell 2020; 10:217-222. [PMID: 29687363 PMCID: PMC6338622 DOI: 10.1007/s13238-018-0542-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Guanrong Yan
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Ruibin Yan
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Cheng Chen
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Cheng Chen
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Yanqiu Zhao
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Wei Qin
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Matthew B Veldman
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095-1555, USA
| | - Song Li
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China
| | - Shuo Lin
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen University Town, Shenzhen, 518055, China. .,Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095-1555, USA.
| |
Collapse
|
22
|
Lee DH, Kim TM, Kim JK, Park C. ETV2/ER71 Transcription Factor as a Therapeutic Vehicle for Cardiovascular Disease. Theranostics 2019; 9:5694-5705. [PMID: 31534512 PMCID: PMC6735401 DOI: 10.7150/thno.35300] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases have long been the leading cause of mortality and morbidity in the United States as well as worldwide. Despite numerous efforts over the past few decades, the number of the patients with cardiovascular disease still remains high, thereby necessitating the development of novel therapeutic strategies equipped with a better understanding of the biology of the cardiovascular system. Recently, the ETS transcription factor, ETV2 (also known as ER71), has been recognized as a master regulator of the development of the cardiovascular system and plays an important role in pathophysiological angiogenesis and the endothelial cell reprogramming. Here, we discuss the detailed mechanisms underlying ETV2/ER71-regulated cardiovascular lineage development. In addition, recent reports on the novel functions of ETV2/ER71 in neovascularization and direct cell reprogramming are discussed with a focus on its therapeutic potential for cardiovascular diseases.
Collapse
|
23
|
In vivo transduction of ETV2 improves cardiac function and induces vascular regeneration following myocardial infarction. Exp Mol Med 2019; 51:1-14. [PMID: 30755583 PMCID: PMC6372609 DOI: 10.1038/s12276-019-0206-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/27/2018] [Accepted: 10/02/2018] [Indexed: 01/16/2023] Open
Abstract
Vascular regeneration in ischemic hearts has been considered a target for new therapeutic strategies. It has been reported that ETV2 is essential for vascular development, injury-induced neovascularization and direct cell reprogramming of non-endothelial cells into endothelial cells. Thus, the objective of this study was to explore the therapeutic potential of ETV2 in murine models of myocardial infarction in vivo. Direct myocardial delivery of lentiviral ETV2 into rodents undergoing myocardial infarction dramatically upregulated the expression of markers for angiogenesis as well as anti-fibrosis and anti-inflammatory factors in vivo. Consistent with these findings, echocardiography showed significantly improved cardiac function in hearts with induced myocardial infarction upon ETV2 injection compared to that in the control virus-injected group as determined by enhanced ejection fraction and fractional shortening. In addition, ETV2-injected hearts were protected against massive fibrosis with a remarkable increase in capillary density. Interestingly, major fractions of capillaries were stained positive for ETV2. In addition, ECs infected with ETV2 showed enhanced proliferation, suggesting a direct role of ETV2 in vascular regeneration in diseased hearts. Furthermore, culture media from ETV2-overexpressing cardiac fibroblasts promoted endothelial cell migration based on scratch assay. Importantly, intramyocardial injection of the adeno-associated virus form of ETV2 into rat hearts with induced myocardial infarction designed for clinical applicability consistently resulted in significant augmentation of cardiac function. We provide compelling evidence that ETV2 has a robust effect on vascular regeneration and enhanced cardiac repair after myocardial infarction, highlighting a potential therapeutic function of ETV2 as an efficient means to treat failing hearts. A gene therapy strategy that stimulates cardiovascular repair could improve recovery for heart attack patients. Heart attacks inflict severe damage on the heart and blood vessels, tissues with limited capacity for self-repair. Researchers led by Kiwon Ban of the City University of Hong Kong and Hun-Jun Park of the Catholic University of Korea, Seoul, have now demonstrated that a gene responsible for cardiovascular development can also efficiently stimulate heart repair. They used viruses to deliver the gene into a mouse model of heart attack, and showed that treated heart tissues exhibited strong recovery relative to untreated controls. The treatment reduced scar tissue formation and promoted proliferation of the cells lining blood vessels and blood vessel formation, measurably improving heart function. This approach could lay the groundwork for treating a common potentially fatal event.
Collapse
|
24
|
Koyano-Nakagawa N, Garry DJ. Etv2 as an essential regulator of mesodermal lineage development. Cardiovasc Res 2018; 113:1294-1306. [PMID: 28859300 DOI: 10.1093/cvr/cvx133] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/24/2017] [Indexed: 11/14/2022] Open
Abstract
The 'master regulatory factors' that position at the top of the genetic hierarchy of lineage determination have been a focus of intense interest, and have been investigated in various systems. Etv2/Etsrp71/ER71 is such a factor that is both necessary and sufficient for the development of haematopoietic and endothelial lineages. As such, genetic ablation of Etv2 leads to complete loss of blood and vessels, and overexpression can convert non-endothelial cells to the endothelial lineage. Understanding such master regulatory role of a lineage is not only a fundamental quest in developmental biology, but also holds immense possibilities in regenerative medicine. To harness its activity and utility for therapeutic interventions, it is essential to understand the regulatory mechanisms, molecular function, and networks that surround Etv2. In this review, we provide a comprehensive overview of Etv2 biology focused on mouse and human systems.
Collapse
Affiliation(s)
- Naoko Koyano-Nakagawa
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 2231 6th st. SE, Minneapolis, MN 55455, USA
| | - Daniel J Garry
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 2231 6th st. SE, Minneapolis, MN 55455, USA
| |
Collapse
|
25
|
Davis JA, Koenig AL, Lubert A, Chestnut B, Liu F, Palencia Desai S, Winkler T, Pociute K, Choi K, Sumanas S. ETS transcription factor Etsrp / Etv2 is required for lymphangiogenesis and directly regulates vegfr3 / flt4 expression. Dev Biol 2018; 440:40-52. [PMID: 29753018 DOI: 10.1016/j.ydbio.2018.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 01/05/2023]
Abstract
The molecular mechanisms initiating the formation of the lymphatic system, lymphangiogenesis, are still poorly understood. Here we have identified a novel role in lymphangiogenesis for an ETS transcription factor, Etv2/Etsrp, a known regulator of embryonic vascular development. Through the use of fully validated photoactivatable morpholinos we show that inducible Etv2 inhibition in zebrafish embryos at 1 day post-fertilization (dpf) results in significant inhibition of lymphangiogenesis, while development of blood vessels is unaffected. In Etv2-inhibited embryos and larvae, the number of lymphatic progenitors is greatly reduced, the major lymphatic vessel, the thoracic duct, is absent or severely fragmented, and lymphangiogenesis-associated marker expression, including lyve1b, prox1a, and vegfr3/flt4, is strongly downregulated. We also demonstrate that lymphatic progenitors in Etv2 deficient embryos fail to respond to Vegfc signaling. Chromatin immunoprecipitation and sequencing (ChIP-Seq) studies using differentiated mouse embryonic stem (ES) cells as well as luciferase reporter studies in the ES cells and in zebrafish embryos argue that Etv2 directly binds the promoter/enhancer regions of Vegfc receptor Vegfr3/Flt4 and lymphatic marker Lyve1, and promotes their expression. Together these data support a model where Etv2 initiates lymphangiogenesis by directly promoting the expression of flt4 within the posterior cardinal vein.
Collapse
Affiliation(s)
- Jennifer A Davis
- Cancer&Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew L Koenig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Allison Lubert
- Cancer&Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Brendan Chestnut
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Fang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sharina Palencia Desai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tamara Winkler
- Cancer&Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Karolina Pociute
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Graduate School of Biotechnology, Kyung Hee University, Yong In, South Korea
| | - 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.
| |
Collapse
|
26
|
Abstract
Glioblastoma multiforme (GBM) is characterized by extensive endothelial hyperplasia. Recent studies suggest that a subpopulation of endothelial cells originates via vasculogenesis by the transdifferentiation of GBM tumor cells into endothelial cells (endo-transdifferentiation). The molecular mechanism underlying this process remains poorly defined. Here, we show that the expression of ETS variant 2 (ETV2), a master regulator of endothelial cell development, is highly correlated with malignancy. Functional studies demonstrate that ETV2 is sufficient and necessary for the transdifferentiation of a subpopulation of CD133+/Nestin+ GBM/neural stem cells to an endothelial lineage. Combinational studies of ChIP-Seq with gain-of-function RNA-Seq data sets suggest that ETV2, in addition to activating vascular genes, represses proneural genes to direct endo-transdifferentiation. Since endo-transdifferentiation by ETV2 is VEGF-A independent, it likely accounts for the observed resistance of GBM tumor cells to anti-angiogenesis therapy. Further characterization of the regulatory networks mediated by ETV2 in endo-transdifferentiation of GBM tumor cells should lead to the identification of more effective therapeutic targets for GBM.
Collapse
|
27
|
Jia D, Jolly MK, Harrison W, Boareto M, Ben-Jacob E, Levine H. Operating principles of tristable circuits regulating cellular differentiation. Phys Biol 2017; 14:035007. [PMID: 28443829 DOI: 10.1088/1478-3975/aa6f90] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Many cell-fate decisions during embryonic development are governed by a motif comprised of two transcription factors (TFs) A and B that mutually inhibit each other and may self-activate. This motif, called as a self-activating toggle switch (SATS), can typically have three stable states (phenotypes)-two corresponding to differentiated cell fates, each of which has a much higher level of one TF than the other-[Formula: see text] or [Formula: see text]-and the third state corresponding to an 'undecided' stem-like state with similar levels of both A and B-[Formula: see text]. Furthermore, two or more SATSes can be coupled together in various topologies in different contexts, thereby affecting the coordination between multiple cellular decisions. However, two questions remain largely unanswered: (a) what governs the co-existence and relative stability of these three stable states? (b) What orchestrates the decision-making of coupled SATSes? Here, we first demonstrate that the co-existence and relative stability of the three stable states in an individual SATS can be governed by the relative strength of self-activation, external signals activating and/or inhibiting A and B, and mutual degradation between A and B. Simultaneously, we investigate the effects of these factors on the decision-making of two coupled SATSes. Our results offer novel understanding into the operating principles of individual and coupled tristable self-activating toggle switches (SATSes) regulating cellular differentiation and can yield insights into synthesizing three-way genetic circuits and understanding of cellular reprogramming.
Collapse
Affiliation(s)
- Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, United States of America. Program in Systems, Synthetic and Physical Biology, Rice University, Houston, TX 77005-1827, United States of America
| | | | | | | | | | | |
Collapse
|
28
|
Ross JA, George BJ, Bruno M, Ge Y. Chemical-agnostic hazard prediction: statistical inference of in vitro toxicity pathways from proteomics responses to chemical mixtures. ACTA ACUST UNITED AC 2017; 2:39-44. [PMID: 30345409 DOI: 10.1016/j.comtox.2017.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Toxicity pathways have been defined as normal cellular pathways that, when sufficiently perturbed as a consequence of chemical exposure, lead to an adverse outcome. If an exposure alters one or more normal biological pathways to an extent that leads to an adverse toxicity outcome, a significant correlation must exist between the exposure, the extent of pathway alteration, and the degree of adverse outcome. Biological pathways are regulated at multiple levels, including transcriptional, post-transcriptional, post-translational, and targeted degradation, each of which can affect the levels and extents of modification of proteins involved in the pathways. Significant alterations of toxicity pathways resulting from changes in regulation at any of these levels therefore are likely to be detectable as alterations in the proteome. We hypothesize that significant correlations between exposures, adverse outcomes, and changes in the proteome have the potential to identify putative toxicity pathways, facilitating selection of candidate targets for high throughput screening, even in the absence of a priori knowledge of either the specific pathways involved or the specific agents inducing the pathway alterations. We explored this hypothesis in vitro in BEAS-2B human airway epithelial cells exposed to different concentrations of Ni2+, Cd2+, and Cr6+, alone and in defined mixtures. Levels and phosphorylation status of a variety of signaling pathway proteins and cytokines were measured after 48 hours exposure, together with cytotoxicity. Least Absolute Shrinkage and Selection Operator (LASSO) multiple regression was used to identify a subset of these proteins that constitute a putative toxicity pathway capable of predicting cytotoxicity. The putative toxicity pathway for cytotoxicity of these metals and metal mixtures identified by LASSO is composed of phospho-RPS6KB1, phospho-p53, cleaved CASP3, phospho-MAPK8, IL-10, and Hif-1α. As this approach does not depend on knowledge of the chemical composition of the mixtures, it may be generally useful for identifying sets of proteins predictive of adverse effects for a variety of mixtures, including complex environmental mixtures of unknown composition.
Collapse
Affiliation(s)
- Jeffrey A Ross
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Barbara Jane George
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Maribel Bruno
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Yue Ge
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| |
Collapse
|
29
|
Goto H, Kimmey SC, Row RH, Matus DQ, Martin BL. FGF and canonical Wnt signaling cooperate to induce paraxial mesoderm from tailbud neuromesodermal progenitors through regulation of a two-step epithelial to mesenchymal transition. Development 2017; 144:1412-1424. [PMID: 28242612 DOI: 10.1242/dev.143578] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
Abstract
Mesoderm induction begins during gastrulation. Recent evidence from several vertebrate species indicates that mesoderm induction continues after gastrulation in neuromesodermal progenitors (NMPs) within the posteriormost embryonic structure, the tailbud. It is unclear to what extent the molecular mechanisms of mesoderm induction are conserved between gastrula and post-gastrula stages of development. Fibroblast growth factor (FGF) signaling is required for mesoderm induction during gastrulation through positive transcriptional regulation of the T-box transcription factor brachyury We find in zebrafish that FGF is continuously required for paraxial mesoderm (PM) induction in post-gastrula NMPs. FGF signaling represses the NMP markers brachyury (ntla) and sox2 through regulation of tbx16 and msgn1, thereby committing cells to a PM fate. FGF-mediated PM induction in NMPs functions in tight coordination with canonical Wnt signaling during the epithelial to mesenchymal transition (EMT) from NMP to mesodermal progenitor. Wnt signaling initiates EMT, whereas FGF signaling terminates this event. Our results indicate that germ layer induction in the zebrafish tailbud is not a simple continuation of gastrulation events.
Collapse
Affiliation(s)
- Hana Goto
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Samuel C Kimmey
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Richard H Row
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| |
Collapse
|
30
|
Lee S, Park C, Han JW, Kim JY, Cho K, Kim EJ, Kim S, Lee SJ, Oh SY, Tanaka Y, Park IH, An HJ, Shin CM, Sharma S, Yoon YS. Direct Reprogramming of Human Dermal Fibroblasts Into Endothelial Cells Using ER71/ETV2. Circ Res 2016; 120:848-861. [PMID: 28003219 DOI: 10.1161/circresaha.116.309833] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022]
Abstract
RATIONALE Direct conversion or reprogramming of human postnatal cells into endothelial cells (ECs), bypassing stem or progenitor cell status, is crucial for regenerative medicine, cell therapy, and pathophysiological investigation but has remained largely unexplored. OBJECTIVE We sought to directly reprogram human postnatal dermal fibroblasts to ECs with vasculogenic and endothelial transcription factors and determine their vascularizing and therapeutic potential. METHODS AND RESULTS We utilized various combinations of 7 EC transcription factors to transduce human postnatal dermal fibroblasts and found that ER71/ETV2 (ETS variant 2) alone best induced endothelial features. KDR+ (kinase insert domain receptor) cells sorted at day 7 from ER71/ETV2-transduced human postnatal dermal fibroblasts showed less mature but enriched endothelial characteristics and thus were referred to as early reprogrammed ECs (rECs), and did not undergo maturation by further culture. After a period of several weeks' transgene-free culture followed by transient reinduction of ER71/ETV2, early rECs matured during 3 months of culture and showed reduced ETV2 expression, reaching a mature phenotype similar to postnatal human ECs. These were termed late rECs. While early rECs exhibited an immature phenotype, their implantation into ischemic hindlimbs induced enhanced recovery from ischemia. These 2 rECs showed clear capacity for contributing to new vessel formation through direct vascular incorporation in vivo. Paracrine or proangiogenic effects of implanted early rECs played a significant role in repairing hindlimb ischemia. CONCLUSIONS This study for the first time demonstrates that ER71/ETV2 alone can directly reprogram human postnatal cells to functional, mature ECs after an intervening transgene-free period. These rECs could be valuable for cell therapy, personalized disease investigation, and exploration of the reprogramming process.
Collapse
Affiliation(s)
- Sangho Lee
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Changwon Park
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Ji Woong Han
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Ju Young Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Kyuwon Cho
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Eun Jae Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Sangsung Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Shin-Jeong Lee
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Se Yeong Oh
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Yoshiaki Tanaka
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - In-Hyun Park
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Hyo Jae An
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Claire Min Shin
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Shraya Sharma
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Young-Sup Yoon
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| |
Collapse
|
31
|
Craig MP, Sumanas S. ETS transcription factors in embryonic vascular development. Angiogenesis 2016; 19:275-85. [PMID: 27126901 DOI: 10.1007/s10456-016-9511-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/19/2016] [Indexed: 11/25/2022]
Abstract
At least thirteen ETS-domain transcription factors are expressed during embryonic hematopoietic or vascular development and potentially function in the formation and maintenance of the embryonic vasculature or blood lineages. This review summarizes our current understanding of the specific roles played by ETS factors in vasculogenesis and angiogenesis and the implications of functional redundancies between them.
Collapse
Affiliation(s)
- Michael P Craig
- Department of Biochemistry and Molecular Biology, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435, USA.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
32
|
Sumanas S, Choi K. ETS Transcription Factor ETV2/ER71/Etsrp in Hematopoietic and Vascular Development. Curr Top Dev Biol 2016; 118:77-111. [PMID: 27137655 DOI: 10.1016/bs.ctdb.2016.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Effective establishment of the hematopoietic and vascular systems is prerequisite for successful embryogenesis. The ETS transcription factor Etv2 has proven to be essential for hematopoietic and vascular development. Etv2 expression marks the onset of the hematopoietic and vascular development and its deficiency leads to an absolute block in hematopoietic and vascular development. Etv2 is transiently expressed during development and is mainly expressed in testis in adults. Consistent with its expression pattern, Etv2 is transiently required for the generation of the optimal levels of the hemangiogenic cell population. Deletion of this gene after the hemangiogenic progenitor formation leads to normal hematopoietic and vascular development. Mechanistically, ETV2 induces the hemangiogenic program by activating blood and endothelial cell lineage specifying genes and enhancing VEGF signaling. Moreover, ETV2 establishes an ETS hierarchy by directly activating other Ets genes, which in the face of transient Etv2 expression, presumably maintain blood and endothelial cell program initiated by ETV2 through an ETS switching mechanism. Current studies suggest that the hemangiogenic progenitor population is exclusively sensitive to ETV2-dependent FLK1 signaling. Any perturbation in the ETV2, VEGF, and FLK1 balance causing insufficient hemangiogenic progenitor cell generation would lead to defects in hematopoietic and endothelial cell development.
Collapse
Affiliation(s)
- S Sumanas
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - K Choi
- Washington University, School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
33
|
Frasch M. Dedifferentiation, Redifferentiation, and Transdifferentiation of Striated Muscles During Regeneration and Development. Curr Top Dev Biol 2016; 116:331-55. [PMID: 26970627 DOI: 10.1016/bs.ctdb.2015.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
In some rare and striking cases, striated muscle fibers of the skeleton or body wall, which consist of terminally differentiated syncytia with complex ultrastructures, were found to be capable of dedifferentiating and fragmenting into mononucleate cells. Examples of such events will be discussed in which the dedifferentiated cells reenter the cell cycle, proliferate, and rebuilt damaged muscle fibers during limb regeneration or transdifferentiate to generate new types of muscles during normal development.
Collapse
Affiliation(s)
- Manfred Frasch
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
34
|
Row RH, Tsotras SR, Goto H, Martin BL. The zebrafish tailbud contains two independent populations of midline progenitor cells that maintain long-term germ layer plasticity and differentiate in response to local signaling cues. Development 2015; 143:244-54. [PMID: 26674311 DOI: 10.1242/dev.129015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/09/2015] [Indexed: 12/25/2022]
Abstract
Vertebrate body axis formation depends on a population of bipotential neuromesodermal cells along the posterior wall of the tailbud that make a germ layer decision after gastrulation to form spinal cord and mesoderm. Despite exhibiting germ layer plasticity, these cells never give rise to midline tissues of the notochord, floor plate and dorsal endoderm, raising the question of whether midline tissues also arise from basal posterior progenitors after gastrulation. We show in zebrafish that local posterior signals specify germ layer fate in two basal tailbud midline progenitor populations. Wnt signaling induces notochord within a population of notochord/floor plate bipotential cells through negative transcriptional regulation of sox2. Notch signaling, required for hypochord induction during gastrulation, continues to act in the tailbud to specify hypochord from a notochord/hypochord bipotential cell population. Our results lend strong support to a continuous allocation model of midline tissue formation in zebrafish, and provide an embryological basis for zebrafish and mouse bifurcated notochord phenotypes as well as the rare human congenital split notochord syndrome. We demonstrate developmental equivalency between the tailbud progenitor cell populations. Midline progenitors can be transfated from notochord to somite fate after gastrulation by ectopic expression of msgn1, a master regulator of paraxial mesoderm fate, or if transplanted into the bipotential progenitors that normally give rise to somites. Our results indicate that the entire non-epidermal posterior body is derived from discrete, basal tailbud cell populations. These cells remain receptive to extracellular cues after gastrulation and continue to make basic germ layer decisions.
Collapse
Affiliation(s)
- Richard H Row
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Steve R Tsotras
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Hana Goto
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| |
Collapse
|
35
|
Ulrich F, Carretero-Ortega J, Menéndez J, Narvaez C, Sun B, Lancaster E, Pershad V, Trzaska S, Véliz E, Kamei M, Prendergast A, Kidd KR, Shaw KM, Castranova DA, Pham VN, Lo BD, Martin BL, Raible DW, Weinstein BM, Torres-Vázquez J. Reck enables cerebrovascular development by promoting canonical Wnt signaling. Development 2015; 143:147-59. [PMID: 26657775 DOI: 10.1242/dev.123059] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 11/25/2015] [Indexed: 01/03/2023]
Abstract
The cerebral vasculature provides the massive blood supply that the brain needs to grow and survive. By acquiring distinctive cellular and molecular characteristics it becomes the blood-brain barrier (BBB), a selectively permeable and protective interface between the brain and the peripheral circulation that maintains the extracellular milieu permissive for neuronal activity. Accordingly, there is great interest in uncovering the mechanisms that modulate the formation and differentiation of the brain vasculature. By performing a forward genetic screen in zebrafish we isolated no food for thought (nft (y72)), a recessive late-lethal mutant that lacks most of the intracerebral central arteries (CtAs), but not other brain blood vessels. We found that the cerebral vascularization deficit of nft (y72) mutants is caused by an inactivating lesion in reversion-inducing cysteine-rich protein with Kazal motifs [reck; also known as suppressor of tumorigenicity 15 protein (ST15)], which encodes a membrane-anchored tumor suppressor glycoprotein. Our findings highlight Reck as a novel and pivotal modulator of the canonical Wnt signaling pathway that acts in endothelial cells to enable intracerebral vascularization and proper expression of molecular markers associated with BBB formation. Additional studies with cultured endothelial cells suggest that, in other contexts, Reck impacts vascular biology via the vascular endothelial growth factor (VEGF) cascade. Together, our findings have broad implications for both vascular and cancer biology.
Collapse
Affiliation(s)
- Florian Ulrich
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Jorge Carretero-Ortega
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Javier Menéndez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Carlos Narvaez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Belinda Sun
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Eva Lancaster
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Valerie Pershad
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Sean Trzaska
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Evelyn Véliz
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Makoto Kamei
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Prendergast
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Kameha R Kidd
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenna M Shaw
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel A Castranova
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brigid D Lo
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesús Torres-Vázquez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
36
|
Veldman MB, Rios-Galdamez Y, Lu XH, Gu X, Qin W, Li S, Yang XW, Lin S. The N17 domain mitigates nuclear toxicity in a novel zebrafish Huntington's disease model. Mol Neurodegener 2015; 10:67. [PMID: 26645399 PMCID: PMC4673728 DOI: 10.1186/s13024-015-0063-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 11/30/2015] [Indexed: 01/14/2023] Open
Abstract
Background Although the genetic cause for Huntington’s disease (HD) has been known for over 20 years, the mechanisms that cause the neurotoxicity and behavioral symptoms of this disease are not well understood. One hypothesis is that N-terminal fragments of the HTT protein are the causative agents in HD and that peptide sequences adjacent to the poly-glutamine (Q) repeats modify its toxicity. Here we test the function of the N-terminal 17 amino acids (N17) in the context of the exon 1 fragment of HTT in a novel, inducible zebrafish model of HD. Results Deletion of N17 coupled with 97Q expansion (mHTT-ΔN17-exon1) resulted in a robust, rapidly progressing movement deficit, while fish with intact N17 and 97Q expansion (mHTT-exon1) have more delayed-onset movement deficits with slower progression. The level of mHTT-ΔN17-exon1 protein was significantly higher than mHTT-exon1, although the mRNA level of each transgene was marginally different, suggesting that N17 may regulate HTT protein stability in vivo. In addition, cell lineage specific induction of the mHTT-ΔN17-exon1 transgene in neurons was sufficient to recapitulate the consequences of ubiquitous transgene expression. Within neurons, accelerated nuclear accumulation of the toxic HTT fragment was observed in mHTT-ΔN17-exon1 fish, demonstrating that N17 also plays an important role in sub-cellular localization in vivo. Conclusions We have developed a novel, inducible zebrafish model of HD. These animals exhibit a progressive movement deficit reminiscent of that seen in other animal models and human patients. Deletion of the N17 terminal amino acids of the huntingtin fragment results in an accelerated HD-like phenotype that may be due to enhanced protein stability and nuclear accumulation of HTT. These transgenic lines will provide a valuable new tool to study mechanisms of HD at the behavioral, cellular, and molecular levels. Future experiments will be focused on identifying genetic modifiers, mechanisms and therapeutics that alleviate polyQ aggregation in the nucleus of neurons. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0063-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthew B Veldman
- Department of Molecular, Cell and Developmental Biology, University of California-Los Angeles, 621 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Yesenia Rios-Galdamez
- Department of Molecular, Cell and Developmental Biology, University of California-Los Angeles, 621 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Xiao-Hong Lu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, USA.,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiaofeng Gu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, USA.,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Wei Qin
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Song Li
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - X William Yang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, USA.,Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Shuo Lin
- Department of Molecular, Cell and Developmental Biology, University of California-Los Angeles, 621 Charles E. Young Drive South, Los Angeles, CA, 90095, USA. .,Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
37
|
Oh SY, Kim JY, Park C. The ETS Factor, ETV2: a Master Regulator for Vascular Endothelial Cell Development. Mol Cells 2015; 38:1029-36. [PMID: 26694034 PMCID: PMC4696993 DOI: 10.14348/molcells.2015.0331] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 01/15/2023] Open
Abstract
Appropriate vessel development and its coordinated function is essential for proper embryogenesis and homeostasis in the adult. Defects in vessels cause birth defects and are an important etiology of diseases such as cardiovascular disease, tumor and diabetes retinopathy. The accumulative data indicate that ETV2, an ETS transcription factor, performs a potent and indispensable function in mediating vessel development. This review discusses the recent progress of the study of ETV2 with special focus on its regulatory mechanisms and cell fate determining role in developing mouse embryos as well as somatic cells.
Collapse
Affiliation(s)
- Se-Yeong Oh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA,
USA
- Children’s Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA,
USA
| | - Ju Young Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA,
USA
- Children’s Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA,
USA
- Molecular and Systems Pharmacology Program, Emory University School of Medicine, Atlanta, GA,
USA
| | - Changwon Park
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA,
USA
- Children’s Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA,
USA
- Molecular and Systems Pharmacology Program, Emory University School of Medicine, Atlanta, GA,
USA
- Biochemistry, Cell Biology and Developmental Biology Program, Emory University School of Medicine, Atlanta, GA,
USA
| |
Collapse
|
38
|
Park C, Lee TJ, Bhang SH, Liu F, Nakamura R, Oladipupo SS, Pitha-Rowe I, Capoccia B, Choi HS, Kim TM, Urao N, Ushio-Fukai M, Lee DJ, Miyoshi H, Kim BS, Lim DS, Apte RS, Ornitz DM, Choi K. Injury-Mediated Vascular Regeneration Requires Endothelial ER71/ETV2. Arterioscler Thromb Vasc Biol 2015; 36:86-96. [PMID: 26586661 DOI: 10.1161/atvbaha.115.306430] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 11/07/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Comprehensive understanding of the mechanisms regulating angiogenesis might provide new strategies for angiogenic therapies for treating diverse physiological and pathological ischemic conditions. The E-twenty six (ETS) factor Ets variant 2 (ETV2; aka Ets-related protein 71) is essential for the formation of hematopoietic and vascular systems. Despite its indispensable function in vessel development, ETV2 role in adult angiogenesis has not yet been addressed. We have therefore investigated the role of ETV2 in vascular regeneration. APPROACH AND RESULTS We used endothelial Etv2 conditional knockout mice and ischemic injury models to assess the role of ETV2 in vascular regeneration. Although Etv2 expression was not detectable under steady-state conditions, its expression was readily observed in endothelial cells after injury. Mice lacking endothelial Etv2 displayed impaired neovascularization in response to eye injury, wounding, or hindlimb ischemic injury. Lentiviral Etv2 expression in ischemic hindlimbs led to improved recovery of blood perfusion with enhanced vessel formation. After injury, fetal liver kinase 1 (Flk1), aka VEGFR2, expression and neovascularization were significantly upregulated by Etv2, whereas Flk1 expression and vascular endothelial growth factor response were significantly blunted in Etv2-deficient endothelial cells. Conversely, enforced Etv2 expression enhanced vascular endothelial growth factor-mediated endothelial sprouting from embryoid bodies. Lentiviral Flk1 expression rescued angiogenesis defects in endothelial Etv2 conditional knockout mice after hindlimb ischemic injury. Furthermore, Etv2(+/-); Flk1(+/-) double heterozygous mice displayed a more severe hindlimb ischemic injury response compared with Etv2(+/-) or Flk1(+/-) heterozygous mice, revealing an epistatic interaction between ETV2 and FLK1 in vascular regeneration. CONCLUSIONS Our study demonstrates a novel obligatory role for the ETV2 in postnatal vascular repair and regeneration.
Collapse
Affiliation(s)
- Changwon Park
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Tae-Jin Lee
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Suk Ho Bhang
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Fang Liu
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Rei Nakamura
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Sunday S Oladipupo
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Ian Pitha-Rowe
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Benjamin Capoccia
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Hong Seo Choi
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Tae Min Kim
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Norifumi Urao
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Masuko Ushio-Fukai
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Dong Jun Lee
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Hiroyuki Miyoshi
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Byung-Soo Kim
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Dae-Sik Lim
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Rajendra S Apte
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - David M Ornitz
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| | - Kyunghee Choi
- Department of Pediatrics (C.P., H.S.C.), Children's Heart Research and Outcomes Center (C.P.), Molecular and Systems Pharmacology Program (C.P.), Emory University School of Medicine, Atlanta; Department of Pharmacology, College of Medicine, University of Illinois at Chicago, IL (T.M.K., N.U., M.U-F.); School of Chemical Engineering, Sungkyunkwan University, Korea (S.H.B.); School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea (B-S.K.); Korea Advanced Institute of Science and Technology, Korea (D.J.L., D-S.L.); RIKEN BioResource Center, Japan (H.M.); the Departments of Pathology and Immunology (T-J.L., F.L., K.C.), Ophthalmology and Visual Sciences (R.N., I.P-R., R.S.A.), Developmental Biology (S.S.O., D.M.O.), Biochemistry and Molecular Biophysics (B. C.), Developmental, Regenerative, and Stem cell Biology Program (D.M.O., R.S.A., K.C.), Washington University School of Medicine, MO
| |
Collapse
|
39
|
LSD1/KDM1A promotes hematopoietic commitment of hemangioblasts through downregulation of Etv2. Proc Natl Acad Sci U S A 2015; 112:13922-7. [PMID: 26512114 DOI: 10.1073/pnas.1517326112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The hemangioblast is a progenitor cell with the capacity to give rise to both hematopoietic and endothelial progenitors. Currently, the regulatory mechanisms underlying hemangioblast formation are being elucidated, whereas those controllers for the selection of hematopoietic or endothelial fates still remain a mystery. To answer these questions, we screened for zebrafish mutants that have defects in the hemangioblast expression of Gata1, which is never expressed in endothelial progenitors. One of the isolated mutants, it627, showed not only down-regulation of hematopoietic genes but also up-regulation of endothelial genes. We identified the gene responsible for the it627 mutant as the zebrafish homolog of Lys-specific demethylase 1 (LSD1/KDM1A). Surprisingly, the hematopoietic defects in lsd1(it627) embryos were rescued by the gene knockdown of the Ets variant 2 gene (etv2), an essential regulator for vasculogenesis. Our results suggest that the LSD1-dependent shutdown of Etv2 gene expression may be a significant event required for hemangioblasts to initiate hematopoietic differentiation.
Collapse
|
40
|
Koyano-Nakagawa N, Shi X, Rasmussen TL, Das S, Walter CA, Garry DJ. Feedback Mechanisms Regulate Ets Variant 2 (Etv2) Gene Expression and Hematoendothelial Lineages. J Biol Chem 2015; 290:28107-28119. [PMID: 26396195 DOI: 10.1074/jbc.m115.662197] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Indexed: 12/12/2022] Open
Abstract
Etv2 is an essential transcriptional regulator of hematoendothelial lineages during embryogenesis. Although Etv2 downstream targets have been identified, little is known regarding the upstream transcriptional regulation of Etv2 gene expression. In this study, we established a novel methodology that utilizes the differentiating ES cell and embryoid body system to define the modules and enhancers embedded within the Etv2 promoter. Using this system, we defined an autoactivating role for Etv2 that is mediated by two adjacent Ets motifs in the proximal promoter. In addition, we defined the role of VEGF/Flk1-Calcineurin-NFAT signaling cascade in the transcriptional regulation of Etv2. Furthermore, we defined an Etv2-Flt1-Flk1 cascade that serves as a negative feedback mechanism to regulate Etv2 gene expression. To complement and extend these studies, we demonstrated that the Flt1 null embryonic phenotype was partially rescued in the Etv2 conditional knockout background. In summary, these studies define upstream and downstream networks that serve as a transcriptional rheostat to regulate Etv2 gene expression.
Collapse
Affiliation(s)
- Naoko Koyano-Nakagawa
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Xiaozhong Shi
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Tara L Rasmussen
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Satyabrata Das
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Camille A Walter
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, Minnesota 55455.
| |
Collapse
|
41
|
Bouldin CM, Manning AJ, Peng YH, Farr GH, Hung KL, Dong A, Kimelman D. Wnt signaling and tbx16 form a bistable switch to commit bipotential progenitors to mesoderm. Development 2015; 142:2499-507. [PMID: 26062939 DOI: 10.1242/dev.124024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/03/2015] [Indexed: 01/16/2023]
Abstract
Anterior to posterior growth of the vertebrate body is fueled by a posteriorly located population of bipotential neuro-mesodermal progenitor cells. These progenitors have a limited rate of proliferation and their maintenance is crucial for completion of the anterior-posterior axis. How they leave the progenitor state and commit to differentiation is largely unknown, in part because widespread modulation of factors essential for this process causes organism-wide effects. Using a novel assay, we show that zebrafish Tbx16 (Spadetail) is capable of advancing mesodermal differentiation cell-autonomously. Tbx16 locks cells into the mesodermal state by not only activating downstream mesodermal genes, but also by repressing bipotential progenitor genes, in part through a direct repression of sox2. We demonstrate that tbx16 is activated as cells move from an intermediate Wnt environment to a high Wnt environment, and show that Wnt signaling activates the tbx16 promoter. Importantly, high-level Wnt signaling is able to accelerate mesodermal differentiation cell-autonomously, just as we observe with Tbx16. Finally, because our assay for mesodermal commitment is quantitative we are able to show that the acceleration of mesodermal differentiation is surprisingly incomplete, implicating a potential separation of cell movement and differentiation during this process. Together, our data suggest a model in which high levels of Wnt signaling induce a transition to mesoderm by directly activating tbx16, which in turn acts to irreversibly flip a bistable switch, leading to maintenance of the mesodermal fate and repression of the bipotential progenitor state, even as cells leave the initial high-Wnt environment.
Collapse
Affiliation(s)
- Cortney M Bouldin
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alyssa J Manning
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Yu-Hsuan Peng
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Gist H Farr
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - King L Hung
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alice Dong
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - David Kimelman
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
42
|
Org-1-dependent lineage reprogramming generates the ventral longitudinal musculature of the Drosophila heart. Curr Biol 2015; 25:488-94. [PMID: 25660543 DOI: 10.1016/j.cub.2014.12.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 01/27/2023]
Abstract
Only few examples of transdifferentiation, which denotes the conversion of one differentiated cell type to another, are known to occur during normal development, and more often, it is associated with regeneration processes. With respect to muscles, dedifferentiation/redifferentiation processes have been documented during post-traumatic muscle regeneration in blastema of newts as well as during myocardial regeneration. As shown herein, the ventral longitudinal muscles of the adult Drosophila heart arise from specific larval alary muscles in a process that represents the first known example of syncytial muscle transdifferentiation via dedifferentiation into mononucleate myoblasts during normal development. We demonstrate that this unique process depends on the reinitiation of a transcriptional program previously employed for embryonic alary muscle development, in which the factors Org-1 (Drosophila Tbx1) and Tailup (Drosophila Islet1) are key components. During metamorphosis, the action of these factors is combined with cell-autonomous inputs from the ecdysone steroid and the Hox gene Ultrabithorax, which provide temporal and spatial specificity to the transdifferentiation events. Following muscle dedifferentiation, inductive cues, particularly from the remodeling heart tube, are required for the redifferentiation of myoblasts into ventral longitudinal muscles. Our results provide new insights into mechanisms of lineage commitment and cell-fate plasticity during development.
Collapse
|
43
|
Lindgren AG, Veldman MB, Lin S. ETV2 expression increases the efficiency of primitive endothelial cell derivation from human embryonic stem cells. CELL REGENERATION 2015; 4:1. [PMID: 25780560 PMCID: PMC4318149 DOI: 10.1186/s13619-014-0014-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/09/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND Endothelial cells line the luminal surface of blood vessels and form a barrier between the blood and other tissues of the body. Ets variant 2 (ETV2) is transiently expressed in both zebrafish and mice and is necessary and sufficient for vascular endothelial cell specification. Overexpression of this gene in early zebrafish and mouse embryos results in ectopic appearance of endothelial cells. Ectopic expression of ETV2 in later development results in only a subset of cells responding to the signal. FINDINGS We have examined the expression pattern of ETV2 in differentiating human embryonic stem cells (ESCs) to determine when the peak of ETV2 expression occurs. We show that overexpression of ETV2 in differentiating human ESC is able to increase the number of endothelial cells generated when administered during or after the endogenous peak of gene expression. CONCLUSIONS Addition of exogenous ETV2 to human ESCs significantly increased the number of cells expressing angioblast genes without arterial or venous specification. This may be a viable solution to generate in vitro endothelial cells for use in research and in the clinic.
Collapse
Affiliation(s)
- Anne G Lindgren
- Department of Molecular Cellular and Developmental Biology, University of California, 615 Charles E. Young Drive South, Los Angeles, CA 90095 USA
| | - Matthew B Veldman
- Department of Molecular Cellular and Developmental Biology, University of California, 615 Charles E. Young Drive South, Los Angeles, CA 90095 USA
| | - Shuo Lin
- Department of Molecular Cellular and Developmental Biology, University of California, 615 Charles E. Young Drive South, Los Angeles, CA 90095 USA
| |
Collapse
|
44
|
ETS transcription factor ETV2 directly converts human fibroblasts into functional endothelial cells. Proc Natl Acad Sci U S A 2014; 112:160-5. [PMID: 25540418 DOI: 10.1073/pnas.1413234112] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transplantation of endothelial cells (ECs) is a promising therapeutic approach for ischemic disorders. In addition, the generation of ECs has become increasingly important for providing vascular plexus to regenerated organs, such as the liver. Although many attempts have been made to generate ECs from pluripotent stem cells and nonvascular cells, the minimum number of transcription factors that specialize in directly inducing vascular ECs remains undefined. Here, by screening 18 transcription factors that are important for both endothelial and hematopoietic development, we demonstrate that ets variant 2 (ETV2) alone directly converts primary human adult skin fibroblasts into functional vascular endothelial cells (ETVECs). In coordination with endogenous FOXC2 in fibroblasts, transduced ETV2 elicits expression of multiple key endothelial development factors, including FLI1, ERG, and TAL1, and induces expression of endothelial functional molecules, including EGFL7 and von Willebrand factor. Consequently, ETVECs exhibits EC characteristics in vitro and forms mature functional vasculature in Matrigel plugs transplanted in NOD SCID mice. Furthermore, ETVECs significantly improve blood flow recovery in a hind limb ischemic model using BALB/c-nu mice. Our study indicates that the creation of ETVECs provides further understanding of human EC development induced by ETV2.
Collapse
|