1
|
Savage AM, Wagner AC, Kim RT, Gilbert P, Singer HD, Chen E, Kim EM, Lopez N, Dooling KE, Paoli JC, Wu SYC, Böhm S, Froitzheim T, Chilambi R, Blair SJ, Powell CJ, Abouelela A, Luong AG, Thornton KN, Harake N, Karabacak A, Tajer B, Payzin-Dogru D, Whited JL. VEGF signaling promotes blastema growth and proliferation of vascular and non-vascular cells during axolotl limb regeneration. Dev Biol 2025; 525:206-215. [PMID: 40480306 DOI: 10.1016/j.ydbio.2025.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 05/27/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025]
Abstract
Salamanders are capable of regenerating whole limbs throughout life, a feat that is unmatched by other tetrapods. Limb regeneration is dependent upon the formation of a blastema containing progenitor cells which give rise to most tissues of the regenerated limb. Many signaling pathways, including FGF, BMP and Wnt, are required for regeneration, but the role of VEGF signaling during salamander limb regeneration is not well understood, particularly outside of angiogenesis. Here we show that VEGF signaling is essential for limb regeneration and that blastema cells and limb fibroblasts display impaired proliferation in the absence of VEGF signaling. Loss of VEGF signaling reduces expression of EMT-associated genes, suggesting VEGF signaling promotes expression of EMT-associated transcription factors, including Snai2, during axolotl limb regeneration. These findings highlight potential roles for VEGF signaling during regeneration which may extend beyond its expected pro-angiogenic function.
Collapse
Affiliation(s)
- Aaron M Savage
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Alexandra C Wagner
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Ryan T Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Paul Gilbert
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Hani D Singer
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Erica Chen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Elane M Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Noah Lopez
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Kelly E Dooling
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Julia C Paoli
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - S Y Celeste Wu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Sebastian Böhm
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Tim Froitzheim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Rachna Chilambi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Steven J Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Connor J Powell
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Adnan Abouelela
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Anna G Luong
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Kara N Thornton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Noora Harake
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Alparslan Karabacak
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Benjamin Tajer
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Duygu Payzin-Dogru
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
| |
Collapse
|
2
|
Senevirathne G, Shubin NH. Molecular basis of urostyle development in frogs: genes and gene regulation underlying an evolutionary novelty. Open Biol 2024; 14:240111. [PMID: 39191278 DOI: 10.1098/rsob.240111] [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: 04/30/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Evolutionary novelties entail the origin of morphologies that enable new functions. These features can arise through changes to gene function and regulation. One key novelty is the fused rod at the end of the vertebral column in anurans, the urostyle. This feature is composed of a coccyx and a hypochord, both of which ossify during metamorphosis. To elucidate the genetic basis of these features, we used laser capture microdissection of these tissues and did RNA-seq and ATAC-seq at three developmental stages in tadpoles of Xenopus tropicalis. RNA-seq reveals that the coccyx and hypochord have two different molecular signatures. Neuronal (TUBB3) and muscle markers (MYH3) are upregulated in coccygeal tissues, whereas T-box genes (TBXT, TBXT.2), corticosteroid stress hormones (CRCH.1) and matrix metallopeptidases (MMP1, MMP8 and MMP13) are upregulated in the hypochord. ATAC-seq reveals potential regulatory regions that are observed in proximity to candidate genes that regulate ossification identified from RNA-seq. Even though an ossifying hypochord is only present in anurans, this ossification between the vertebral column and the notochord resembles a congenital vertebral anomaly seen prenatally in humans caused by an ectopic expression of the TBXT/TBXT.2 gene. This work opens the way to functional studies that can elucidate anuran bauplan evolution.
Collapse
Affiliation(s)
- Gayani Senevirathne
- Human Evolutionary Biology, Harvard University , Cambridge, MA 02138, USA
- Organismal Biology and Anatomy, University of Chicago Biological Sciences Division , Chicago, IL 60637, USA
| | - Neil H Shubin
- Organismal Biology and Anatomy, University of Chicago Biological Sciences Division , Chicago, IL 60637, USA
| |
Collapse
|
3
|
Curtis GH, Reeve RE, Crespi EJ. Leptin signaling promotes blood vessel formation in the Xenopus tail during the embryo-larval transition. Dev Biol 2024; 512:26-34. [PMID: 38705558 DOI: 10.1016/j.ydbio.2024.05.001] [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: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The signals that regulate peripheral blood vessel formation during development are still under investigation. The hormone leptin promotes blood vessel formation, adipose tissue establishment and expansion, tumor growth, and wound healing, but the underlying mechanisms for these actions are currently unknown. We investigated whether leptin promotes angiogenesis in the developing tail fin using embryonic transgenic xflk-1:GFP Xenopus laevis, which express a green fluorescent protein on vascular endothelial cells to mark blood vessels. We found that leptin protein is expressed in endothelial cells of developing blood vessels and that leptin treatment via injection increased phosphorylated STAT3 signaling, which is indicative of leptin activation of its receptor, in blood vessels of the larval tail fin. Leptin administration via media increased vessel length, branching, and reconnection with the cardinal vein, while decreased leptin signaling via immunoneutralization had an opposing effect on vessel development. We also observed disorganization of major vessels and microvessels of the tail fin and muscle when leptin signaling was decreased. Reduced leptin signaling lowered mRNA expression of cenpk, gpx1, and mmp9, markers for cell proliferation, antioxidation, and extracellular matrix remodeling/cell migration, respectively, in the developing tail, providing insight into three possible mechanisms underlying leptin's promotion of angiogenesis. Together these results illustrate that leptin levels are correlated with embryonic angiogenesis and that leptin coordinates multiple aspects of blood vessel growth and development, showing that leptin is an important morphogen during embryonic development.
Collapse
Affiliation(s)
- Grace H Curtis
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA, USA, 99164.
| | - Robyn E Reeve
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA, USA, 99164
| | - Erica J Crespi
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, WA, USA, 99164
| |
Collapse
|
4
|
Wang Y, Li C, Wang R, Zhao X, Pan Y, Zhang Q, Li S, Fan J, Wang Y, Sun X. PIEZO1 Promotes the Migration of Endothelial Cells via Enhancing CXCR4 Expression under Simulated Microgravity. Int J Mol Sci 2024; 25:7254. [PMID: 39000362 PMCID: PMC11242226 DOI: 10.3390/ijms25137254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Exposure to microgravity during spaceflight induces the alterations in endothelial cell function associated with post-flight cardiovascular deconditioning. PIEZO1 is a major mechanosensitive ion channel that regulates endothelial cell function. In this study, we used a two-dimensional clinostat to investigate the expression of PIEZO1 and its regulatory mechanism on human umbilical vein endothelial cells (HUVECs) under simulated microgravity. Utilizing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis, we observed that PIEZO1 expression was significantly increased in response to simulated microgravity. Moreover, we found microgravity promoted endothelial cells migration by increasing expression of PIEZO1. Proteomics analysis highlighted the importance of C-X-C chemokine receptor type 4(CXCR4) as a main target molecule of PIEZO1 in HUVECs. CXCR4 protein level was increased with simulated microgravity and decreased with PIEZO1 knock down. The mechanistic study showed that PIEZO1 enhances CXCR4 expression via Ca2+ influx. In addition, CXCR4 could promote endothelial cell migration under simulated microgravity. Taken together, these results suggest that the upregulation of PIEZO1 in response to simulated microgravity regulates endothelial cell migration due to enhancing CXCR4 expression via Ca2+ influx.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an 710032, China; (Y.W.); (C.L.); (R.W.); (X.Z.); (Y.P.); (Q.Z.); (S.L.); (J.F.); (Y.W.)
| |
Collapse
|
5
|
Jadon J, Yelin R, Arraf AA, Asleh MA, Zaher M, Schultheiss TM. Regulation of aortic morphogenesis and VE-cadherin dynamics by VEGF. Dev Biol 2023; 497:1-10. [PMID: 36841503 DOI: 10.1016/j.ydbio.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/04/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
In amniote vertebrates, the definitive dorsal aorta is formed by the fusion of two primordial aortic endothelial tubes. Formation of the definitive dorsal aorta requires extensive cellular migrations and rearrangements of the primordial tubes in order to generate a single vessel located at the embryonic ventral midline. This study examines the role of VEGF signaling in the generation of the definitive dorsal aorta. Through gain- and loss-of-function studies in vivo in the chick embryo, we document a requirement for VEGF signaling in growth and remodeling of the paired primordia. We find that regions of the aorta are differentially sensitive to levels of VEGF signaling, and present evidence that areas of low blood flow are more sensitive to the loss of VEGF signaling. We also find that VEGF signaling regulates the intracellular distribution between membrane and cytoplasm of the cell-cell adhesion molecule VE-cadherin in aortic endothelial cells in vivo. Together, these finding identify mechanisms that likely contribute to the dynamic behavior of endothelial cells during aorta morphogenesis.
Collapse
Affiliation(s)
- Julian Jadon
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Alaa A Arraf
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Manar Abboud Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Mira Zaher
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096, Israel.
| |
Collapse
|
6
|
Yvernogeau L, Dainese G, Jaffredo T. Dorsal aorta polarization and haematopoietic stem cell emergence. Development 2023; 150:286251. [PMID: 36602140 DOI: 10.1242/dev.201173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recent studies have highlighted the crucial role of the aorta microenvironment in the generation of the first haematopoietic stem cells (HSCs) from specialized haemogenic endothelial cells (HECs). Despite more than two decades of investigations, we require a better understanding of the cellular and molecular events driving aorta formation and polarization, which will be pivotal to establish the mechanisms that operate during HEC specification and HSC competency. Here, we outline the early mechanisms involved in vertebrate aorta formation by comparing four different species: zebrafish, chicken, mouse and human. We highlight how this process, which is tightly controlled in time and space, requires a coordinated specification of several cell types, in particular endothelial cells originating from distinct mesodermal tissues. We also discuss how molecular signals originating from the aorta environment result in its polarization, creating a unique entity for HSC generation.
Collapse
Affiliation(s)
- Laurent Yvernogeau
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Giovanna Dainese
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Thierry Jaffredo
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| |
Collapse
|
7
|
Abstract
Formation of the vasculature is a critical step within the developing embryo and its disruption causes early embryonic lethality. This complex process is driven by a cascade of signaling events that controls differentiation of mesodermal progenitors into primordial endothelial cells and their further specification into distinct subtypes (arterial, venous, hemogenic) that are needed to generate a blood circulatory network. Hemogenic endothelial cells give rise to hematopoietic stem and progenitor cells that generate all blood cells in the body during embryogenesis and postnatally. We focus our discussion on the regulation of endothelial cell differentiation, and subsequent hemogenic specification, and highlight many of the signaling pathways involved in these processes, which are conserved across vertebrates. Gaining a better understanding of the regulation of these processes will yield insights needed to optimize the treatment of vascular and hematopoietic disease and generate human stem cell-derived vascular and hematopoietic cells for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jordon W Aragon
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
- Departments of Medicine and Genetics, Yale University School of Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut 06520, USA
| |
Collapse
|
8
|
Wiltbank AT, Steinson ER, Criswell SJ, Piller M, Kucenas S. Cd59 and inflammation regulate Schwann cell development. eLife 2022; 11:e76640. [PMID: 35748863 PMCID: PMC9232220 DOI: 10.7554/elife.76640] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Efficient neurotransmission is essential for organism survival and is enhanced by myelination. However, the genes that regulate myelin and myelinating glial cell development have not been fully characterized. Data from our lab and others demonstrates that cd59, which encodes for a small GPI-anchored glycoprotein, is highly expressed in developing zebrafish, rodent, and human oligodendrocytes (OLs) and Schwann cells (SCs), and that patients with CD59 dysfunction develop neurological dysfunction during early childhood. Yet, the function of Cd59 in the developing nervous system is currently undefined. In this study, we demonstrate that cd59 is expressed in a subset of developing SCs. Using cd59 mutant zebrafish, we show that developing SCs proliferate excessively and nerves may have reduced myelin volume, altered myelin ultrastructure, and perturbed node of Ranvier assembly. Finally, we demonstrate that complement activity is elevated in cd59 mutants and that inhibiting inflammation restores SC proliferation, myelin volume, and nodes of Ranvier to wildtype levels. Together, this work identifies Cd59 and developmental inflammation as key players in myelinating glial cell development, highlighting the collaboration between glia and the innate immune system to ensure normal neural development.
Collapse
Affiliation(s)
- Ashtyn T Wiltbank
- Neuroscience Graduate Program, University of VirginiaCharlottesvilleUnited States
- Program in Fundamental Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Emma R Steinson
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Stacey J Criswell
- Department of Cell Biology, University of VirginiaCharlottesvilleUnited States
| | - Melanie Piller
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Sarah Kucenas
- Neuroscience Graduate Program, University of VirginiaCharlottesvilleUnited States
- Program in Fundamental Neuroscience, University of VirginiaCharlottesvilleUnited States
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
9
|
Li-Villarreal N, Wong RLY, Garcia MD, Udan RS, Poché RA, Rasmussen TL, Rhyner AM, Wythe JD, Dickinson ME. FOXO1 represses sprouty 2 and sprouty 4 expression to promote arterial specification and vascular remodeling in the mouse yolk sac. Development 2022; 149:274922. [PMID: 35297995 PMCID: PMC8995087 DOI: 10.1242/dev.200131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/04/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Establishing a functional circulatory system is required for post-implantation development during murine embryogenesis. Previous studies in loss-of-function mouse models showed that FOXO1, a Forkhead family transcription factor, is required for yolk sac (YS) vascular remodeling and survival beyond embryonic day (E) 11. Here, we demonstrate that at E8.25, loss of Foxo1 in Tie2-cre expressing cells resulted in increased sprouty 2 (Spry2) and Spry4 expression, reduced arterial gene expression and reduced Kdr (also known as Vegfr2 and Flk1) transcripts without affecting overall endothelial cell identity, survival or proliferation. Using a Dll4-BAC-nlacZ reporter line, we found that one of the earliest expressed arterial genes, delta like 4, is significantly reduced in Foxo1 mutant YS without being substantially affected in the embryo proper. We show that FOXO1 binds directly to previously identified Spry2 gene regulatory elements (GREs) and newly identified, evolutionarily conserved Spry4 GREs to repress their expression. Furthermore, overexpression of Spry4 in transient transgenic embryos largely recapitulates the reduced expression of arterial genes seen in conditional Foxo1 mutants. Together, these data reveal a novel role for FOXO1 as a key transcriptional repressor regulating both pre-flow arterial specification and subsequent vessel remodeling within the murine YS.
Collapse
Affiliation(s)
- Nanbing Li-Villarreal
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Rebecca Lee Yean Wong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Monica D. Garcia
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ryan S. Udan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ross A. Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Tara L. Rasmussen
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Alexander M. Rhyner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Joshua D. Wythe
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Mary E. Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
10
|
|
11
|
Paulissen E, Palmisano NJ, Waxman J, Martin BL. Somite morphogenesis is required for axial blood vessel formation during zebrafish embryogenesis. eLife 2022; 11:74821. [PMID: 35137687 PMCID: PMC8863375 DOI: 10.7554/elife.74821] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Angioblasts that form the major axial blood vessels of the dorsal aorta and cardinal vein migrate toward the embryonic midline from distant lateral positions. Little is known about what controls the precise timing of angioblast migration and their final destination at the midline. Using zebrafish, we found that midline angioblast migration requires neighboring tissue rearrangements generated by somite morphogenesis. The somitic shape changes cause the adjacent notochord to separate from the underlying endoderm, creating a ventral midline cavity that provides a physical space for the angioblasts to migrate into. The anterior to posterior progression of midline angioblast migration is facilitated by retinoic acid-induced anterior to posterior somite maturation and the subsequent progressive opening of the ventral midline cavity. Our work demonstrates a critical role for somite morphogenesis in organizing surrounding tissues to facilitate notochord positioning and angioblast migration, which is ultimately responsible for creating a functional cardiovascular system.
Collapse
Affiliation(s)
- Eric Paulissen
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Nicholas J Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Joshua Waxman
- Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Benjamin Louis Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| |
Collapse
|
12
|
Della Gaspera B, Weill L, Chanoine C. Evolution of Somite Compartmentalization: A View From Xenopus. Front Cell Dev Biol 2022; 9:790847. [PMID: 35111756 PMCID: PMC8802780 DOI: 10.3389/fcell.2021.790847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Somites are transitory metameric structures at the basis of the axial organization of vertebrate musculoskeletal system. During evolution, somites appear in the chordate phylum and compartmentalize mainly into the dermomyotome, the myotome, and the sclerotome in vertebrates. In this review, we summarized the existing literature about somite compartmentalization in Xenopus and compared it with other anamniote and amniote vertebrates. We also present and discuss a model that describes the evolutionary history of somite compartmentalization from ancestral chordates to amniote vertebrates. We propose that the ancestral organization of chordate somite, subdivided into a lateral compartment of multipotent somitic cells (MSCs) and a medial primitive myotome, evolves through two major transitions. From ancestral chordates to vertebrates, the cell potency of MSCs may have evolved and gave rise to all new vertebrate compartments, i.e., the dermomyome, its hypaxial region, and the sclerotome. From anamniote to amniote vertebrates, the lateral MSC territory may expand to the whole somite at the expense of primitive myotome and may probably facilitate sclerotome formation. We propose that successive modifications of the cell potency of some type of embryonic progenitors could be one of major processes of the vertebrate evolution.
Collapse
|
13
|
Takino JI, Miyazaki S, Nagamine K, Hori T. The Role of RASGRP2 in Vascular Endothelial Cells-A Mini Review. Int J Mol Sci 2021; 22:ijms222011129. [PMID: 34681791 PMCID: PMC8537898 DOI: 10.3390/ijms222011129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
RAS guanyl nucleotide-releasing proteins (RASGRPs) are important proteins that act as guanine nucleotide exchange factors, which activate small GTPases and function as molecular switches for intracellular signals. The RASGRP family is composed of RASGRP1-4 proteins and activates the small GTPases, RAS and RAP. Among them, RASGRP2 has different characteristics from other RASGRPs in that it targets small GTPases and its localizations are different. Many studies related to RASGRP2 have been reported in cells of the blood cell lineage. Furthermore, RASGRP2 has also been reported to be associated with Huntington's disease, tumors, and rheumatoid arthritis. In addition, we also recently reported RASGRP2 expression in vascular endothelial cells, and clarified the involvement of xenopus Rasgrp2 in the vasculogenesis process and multiple signaling pathways of RASGRP2 in human vascular endothelial cells with stable expression of RASGRP2. Therefore, this article outlines the existing knowledge of RASGRP2 and focuses on its expression and role in vascular endothelial cells, and suggests that RASGRP2 functions as a protective factor for maintaining healthy blood vessels.
Collapse
Affiliation(s)
- Jun-ichi Takino
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
- Correspondence: ; Tel.: +81-823-73-8584
| | - Shouhei Miyazaki
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
| | - Kentaro Nagamine
- Faculty of Health Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan;
| | - Takamitsu Hori
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
| |
Collapse
|
14
|
Abstract
Phospholipase C γ1 (PLCγ1) is a member of the PLC family that functions as signal transducer by hydrolyzing membrane lipid to generate second messengers. The unique protein structure of PLCγ1 confers a critical role as a direct effector of VEGFR2 and signaling mediated by other receptor tyrosine kinases. The distinct vascular phenotypes in PLCγ1-deficient animal models and the gain-of-function mutations of PLCγ1 found in human endothelial cancers point to a major physiological role of PLCγ1 in the endothelial system. In this review, we discuss aspects of physiological and molecular function centering around PLCγ1 in the context of endothelial cells and provide a perspective for future investigation.
Collapse
Affiliation(s)
- Dongying Chen
- Yale Cardiovascular Research Center, Departments of Internal Medicine and Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Departments of Internal Medicine and Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
15
|
Oonuma K, Yamamoto M, Moritsugu N, Okawa N, Mukai M, Sotani M, Tsunemi S, Sugimoto H, Nakagome E, Hasegawa Y, Shimai K, Horie T, Kusakabe TG. Evolution of Developmental Programs for the Midline Structures in Chordates: Insights From Gene Regulation in the Floor Plate and Hypochord Homologues of Ciona Embryos. Front Cell Dev Biol 2021; 9:704367. [PMID: 34235159 PMCID: PMC8256262 DOI: 10.3389/fcell.2021.704367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 05/25/2021] [Indexed: 11/24/2022] Open
Abstract
In vertebrate embryos, dorsal midline tissues, including the notochord, the prechordal plate, and the floor plate, play important roles in patterning of the central nervous system, somites, and endodermal tissues by producing extracellular signaling molecules, such as Sonic hedgehog (Shh). In Ciona, hedgehog.b, one of the two hedgehog genes, is expressed in the floor plate of the embryonic neural tube, while none of the hedgehog genes are expressed in the notochord. We have identified a cis-regulatory region of hedgehog.b that was sufficient to drive a reporter gene expression in the floor plate. The hedgehog.b cis-regulatory region also drove ectopic expression of the reporter gene in the endodermal strand, suggesting that the floor plate and the endodermal strand share a part of their gene regulatory programs. The endodermal strand occupies the same topographic position of the embryo as does the vertebrate hypochord, which consists of a row of single cells lined up immediately ventral to the notochord. The hypochord shares expression of several genes with the floor plate, including Shh and FoxA, and play a role in dorsal aorta development. Whole-embryo single-cell transcriptome analysis identified a number of genes specifically expressed in both the floor plate and the endodermal strand in Ciona tailbud embryos. A Ciona FoxA ortholog FoxA.a is shown to be a candidate transcriptional activator for the midline gene battery. The present findings suggest an ancient evolutionary origin of a common developmental program for the midline structures in Olfactores.
Collapse
Affiliation(s)
- Kouhei Oonuma
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Maho Yamamoto
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Naho Moritsugu
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Nanako Okawa
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Megumi Mukai
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Miku Sotani
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Shuto Tsunemi
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Haruka Sugimoto
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Eri Nakagome
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Yuichi Hasegawa
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Kotaro Shimai
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| | - Takeo Horie
- Shimoda Marine Research Center, University of Tsukuba, Shimoda, Japan
| | - Takehiro G Kusakabe
- Department of Biology, Faculty of Science and Engineering, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Graduate School of Natural Science, Konan University, Kobe, Japan
| |
Collapse
|
16
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Collart C, Ciccarelli A, Ivanovitch K, Rosewell I, Kumar S, Kelly G, Edwards A, Smith JC. The migratory pathways of the cells that form the endocardium, dorsal aortae, and head vasculature in the mouse embryo. BMC DEVELOPMENTAL BIOLOGY 2021; 21:8. [PMID: 33752600 PMCID: PMC7986287 DOI: 10.1186/s12861-021-00239-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/12/2021] [Indexed: 11/25/2022]
Abstract
Background Vasculogenesis in amniotes is often viewed as two spatially and temporally distinct processes, occurring in the yolk sac and in the embryo. However, the spatial origins of the cells that form the primary intra-embryonic vasculature remain uncertain. In particular, do they obtain their haemato-endothelial cell fate in situ, or do they migrate from elsewhere? Recently developed imaging techniques, together with new Tal1 and existing Flk1 reporter mouse lines, have allowed us to investigate this question directly, by visualising cell trajectories live and in three dimensions. Results We describe the pathways that cells follow to form the primary embryonic circulatory system in the mouse embryo. In particular, we show that Tal1-positive cells migrate from within the yolk sac, at its distal border, to contribute to the endocardium, dorsal aortae and head vasculature. Other Tal1 positive cells, similarly activated within the yolk sac, contribute to the yolk sac vasculature. Using single-cell transcriptomics and our imaging, we identify VEGF and Apela as potential chemo-attractants that may regulate the migration into the embryo. The dorsal aortae and head vasculature are known sites of secondary haematopoiesis; given the common origins that we observe, we investigate whether this is also the case for the endocardium. We discover cells budding from the wall of the endocardium with high Tal1 expression and diminished Flk1 expression, indicative of an endothelial to haematopoietic transition. Conclusions In contrast to the view that the yolk sac and embryonic circulatory systems form by two separate processes, our results indicate that Tal1-positive cells from the yolk sac contribute to both vascular systems. It may be that initial Tal1 activation in these cells is through a common mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s12861-021-00239-3.
Collapse
Affiliation(s)
- C Collart
- Developmental Biology Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - A Ciccarelli
- Advanced Light Microscopy Facility, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - K Ivanovitch
- Developmental Biology Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - I Rosewell
- Genetic Modification Service, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - S Kumar
- Advanced Light Microscopy Facility, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.,Photonics Group, 606 Blackett Laboratory, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - G Kelly
- Bioinformatics and Biostatistics Facility, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - A Edwards
- Advanced Sequencing Facility, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - J C Smith
- Developmental Biology Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| |
Collapse
|
18
|
Ontogeny of the anuran urostyle and the developmental context of evolutionary novelty. Proc Natl Acad Sci U S A 2020; 117:3034-3044. [PMID: 31988131 PMCID: PMC7022158 DOI: 10.1073/pnas.1917506117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fusion of caudal vertebrae has evolved multiple times independently: the pygostyle of birds, coccyx in apes and humans, ural plate of fish, and the urostyle of frogs. The anuran urostyle, however, is structurally and developmentally distinct because of the contribution of an ossifying hypochord. To date, the developmental mechanisms behind an ossifying hypochord have remained obscure. Here, we provide a detailed analysis of the development of this evolutionary innovative structure and of how neuromusculature, cell death, and proliferation paved their way to facilitate its formation. Finally, we propose that the ossifying hypochord plays a role in tail loss in anurans and reorganizing the dorsal aorta and thus is pivotal in the evolution of the anuran bauplan. Developmental novelties often underlie the evolutionary origins of key metazoan features. The anuran urostyle, which evolved nearly 200 MYA, is one such structure. It forms as the tail regresses during metamorphosis, when locomotion changes from an axial-driven mode in larvae to a limb-driven one in adult frogs. The urostyle comprises of a coccyx and a hypochord. The coccyx forms by fusion of caudal vertebrae and has evolved repeatedly across vertebrates. However, the contribution of an ossifying hypochord to the coccyx in anurans is unique among vertebrates and remains a developmental enigma. Here, we focus on the developmental changes that lead to the anuran urostyle, with an emphasis on understanding the ossifying hypochord. We find that the coccyx and hypochord have two different developmental histories: First, the development of the coccyx initiates before metamorphic climax whereas the ossifying hypochord undergoes rapid ossification and hypertrophy; second, thyroid hormone directly affects hypochord formation and appears to have a secondary effect on the coccygeal portion of the urostyle. The embryonic hypochord is known to play a significant role in the positioning of the dorsal aorta (DA), but the reason for hypochordal ossification remains obscure. Our results suggest that the ossifying hypochord plays a role in remodeling the DA in the newly forming adult body by partially occluding the DA in the tail. We propose that the ossifying hypochord-induced loss of the tail during metamorphosis has enabled the evolution of the unique anuran bauplan.
Collapse
|
19
|
Ciau-Uitz A, Patient R. Gene Regulatory Networks Governing the Generation and Regeneration of Blood. J Comput Biol 2019; 26:719-725. [DOI: 10.1089/cmb.2019.0114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Aldo Ciau-Uitz
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Roger Patient
- Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Dissecting BMP signaling input into the gene regulatory networks driving specification of the blood stem cell lineage. Proc Natl Acad Sci U S A 2018; 114:5814-5821. [PMID: 28584091 DOI: 10.1073/pnas.1610615114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) that sustain lifelong blood production are created during embryogenesis. They emerge from a specialized endothelial population, termed hemogenic endothelium (HE), located in the ventral wall of the dorsal aorta (DA). In Xenopus, we have been studying the gene regulatory networks (GRNs) required for the formation of HSCs, and critically found that the hemogenic potential is defined at an earlier time point when precursors to the DA express hematopoietic as well as endothelial genes, in the definitive hemangioblasts (DHs). The GRN for DH programming has been constructed and, here, we show that bone morphogenetic protein (BMP) signaling is essential for the initiation of this GRN. BMP2, -4, and -7 are the principal ligands expressed in the lineage forming the HE. To investigate the requirement and timing of all BMP signaling in HSC ontogeny, we have used a transgenic line, which inducibly expresses an inhibitor of BMP signaling, Noggin, as well as a chemical inhibitor of BMP receptors, DMH1, and described the inputs from BMP signaling into the DH GRN and the HE, as well as into primitive hematopoiesis. BMP signaling is required in at least three points in DH programming: first to initiate the DH GRN through gata2 expression, then for kdr expression to enable the DH to respond to vascular endothelial growth factor A (VEGFA) ligand from the somites, and finally for gata2 expression in the DA, but is dispensable for HE specification after hemangioblasts have been formed.
Collapse
|
21
|
Ni R, Luo L. A noncanonical function of histidyl-tRNA synthetase: inhibition of vascular hyperbranching during zebrafish development. FEBS Open Bio 2018; 8:722-731. [PMID: 29744287 PMCID: PMC5929932 DOI: 10.1002/2211-5463.12420] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 02/21/2018] [Accepted: 03/14/2018] [Indexed: 11/09/2022] Open
Abstract
Histidyl‐tRNA synthetase (Hars) catalyzes the ligation of histidine residues to cognate tRNA. Here, we demonstrate a noncanonical function of Hars in vascular development in zebrafish. We obtained a novel zebrafish cq34 mutant which exhibited hyperbranching of cranial and intersegmental blood vessels 48 h after fertilization. The gene responsible for this phenotype was identified as hars. We found the increased expression of cdh5 and vegfa in the harscq34 mutant. Knockdown of cdh5 in the mutant reduced disordered connections of the hindbrain capillaries. Inhibition of vascular endothelial growth factor signaling suppressed the abnormal vascular branching observed in the mutant. Moreover, the human HARSmRNA rescued the vascular defects in the cq34 mutant. Thus, the noncanonical function of Hars regulates vascular development, mainly by modulating expression of cdh5 and vegfa.
Collapse
Affiliation(s)
- Rui Ni
- Key Laboratory of Freshwater Fish Reproduction and Development Ministry of Education Laboratory of Molecular Developmental Biology School of Life Sciences Southwest University Chongqing China
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development Ministry of Education Laboratory of Molecular Developmental Biology School of Life Sciences Southwest University Chongqing China
| |
Collapse
|
22
|
Roman BL, Hinck AP. ALK1 signaling in development and disease: new paradigms. Cell Mol Life Sci 2017; 74:4539-4560. [PMID: 28871312 PMCID: PMC5687069 DOI: 10.1007/s00018-017-2636-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/01/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Activin A receptor like type 1 (ALK1) is a transmembrane serine/threonine receptor kinase in the transforming growth factor-beta receptor family that is expressed on endothelial cells. Defects in ALK1 signaling cause the autosomal dominant vascular disorder, hereditary hemorrhagic telangiectasia (HHT), which is characterized by development of direct connections between arteries and veins, or arteriovenous malformations (AVMs). Although previous studies have implicated ALK1 in various aspects of sprouting angiogenesis, including tip/stalk cell selection, migration, and proliferation, recent work suggests an intriguing role for ALK1 in transducing a flow-based signal that governs directed endothelial cell migration within patent, perfused vessels. In this review, we present an updated view of the mechanism of ALK1 signaling, put forth a unified hypothesis to explain the cellular missteps that lead to AVMs associated with ALK1 deficiency, and discuss emerging roles for ALK1 signaling in diseases beyond HHT.
Collapse
Affiliation(s)
- Beth L Roman
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto St, Pittsburgh, PA, 15261, USA.
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Sato Y, Nagatoshi K, Hamano A, Imamura Y, Huss D, Uchida S, Lansford R. Basal filopodia and vascular mechanical stress organize fibronectin into pillars bridging the mesoderm-endoderm gap. Development 2017; 144:281-291. [PMID: 28096216 DOI: 10.1242/dev.141259] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/29/2016] [Indexed: 12/23/2022]
Abstract
Cells may exchange information with other cells and tissues by exerting forces on the extracellular matrix (ECM). Fibronectin (FN) is an important ECM component that forms fibrils through cell contacts and creates directionally biased geometry. Here, we demonstrate that FN is deposited as pillars between widely separated germ layers, namely the somitic mesoderm and the endoderm, in quail embryos. Alongside the FN pillars, long filopodia protrude from the basal surfaces of somite epithelial cells. Loss-of-function of Ena/VASP, α5β1-integrins or talin in the somitic cells abolished the FN pillars, indicating that FN pillar formation is dependent on the basal filopodia through these molecules. The basal filopodia and FN pillars are also necessary for proper somite morphogenesis. We identified a new mechanism contributing to FN pillar formation by focusing on cyclic expansion of adjacent dorsal aorta. Maintenance of the directional alignment of the FN pillars depends on pulsatile blood flow through the dorsal aortae. These results suggest that the FN pillars are specifically established through filopodia-mediated and pulsating force-related mechanisms.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan .,JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Kei Nagatoshi
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ayumi Hamano
- Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0385, Japan
| | - Yuko Imamura
- Graduate School of Science, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - David Huss
- Department of Radiology and Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.,Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Seiichi Uchida
- Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0385, Japan
| | - Rusty Lansford
- Department of Radiology and Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.,Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
24
|
Erkenbrack EM, Petsios E. A Conserved Role for VEGF Signaling in Specification of Homologous Mesenchymal Cell Types Positioned at Spatially Distinct Developmental Addresses in Early Development of Sea Urchins. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2017; 328:423-432. [PMID: 28544452 DOI: 10.1002/jez.b.22743] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/04/2017] [Accepted: 04/05/2017] [Indexed: 11/06/2022]
Abstract
Comparative studies of early development in echinoderms are revealing the tempo and mode of alterations to developmental gene regulatory networks and to the cell types they specify. In euechinoid sea urchins, skeletogenic mesenchyme (SM) ingresses prior to gastrulation at the vegetal pole and aligns into a ring-like array with two bilateral pockets of cells, the sites where spiculogenesis will later occur. In cidaroid sea urchins, the anciently diverged sister clade to euechinoid sea urchins, a homologous SM cell type ingresses later in development, after gastrulation has commenced, and consequently at a distinct developmental address. Thus, a heterochronic shift of ingression of the SM cell type occurred in one of the echinoid lineages. In euechinoids, specification and migration of SM are facilitated by vascular endothelial growth factor (VEGF) signaling. We describe spatiotemporal expression of vegf and vegfr and experimental manipulations targeting VEGF signaling in the cidaroid Eucidaris tribuloides. Spatially, vegf and vegfr mRNA localizes similarly as in euechinoids, suggesting conserved deployment in echinoids despite their spatially distinct development addresses of ingression. Inhibition of VEGF signaling in E. tribuloides suggests its role in SM specification is conserved in echinoids. Temporal discrepancies between the onset of vegf expression and SM ingression likely result in previous observations of SM "random wandering" behavior. Our results indicate that, although the SM cell type in echinoids ingresses into distinct developmental landscapes, it retains a signaling mechanism that restricts their spatial localization to a conserved developmental address where spiculogenesis later occurs.
Collapse
Affiliation(s)
- Eric M Erkenbrack
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California.,Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut
| | - Elizabeth Petsios
- Department of Earth Sciences, University of Southern California, Los Angeles, California
| |
Collapse
|
25
|
Zhu ZX, Sun CC, Ting Zhu Y, Wang Y, Wang T, Chi LS, Cai WH, Zheng JY, Zhou X, Cong WT, Li XK, Jin LT. Hedgehog signaling contributes to basic fibroblast growth factor-regulated fibroblast migration. Exp Cell Res 2017; 355:83-94. [PMID: 28363830 DOI: 10.1016/j.yexcr.2017.03.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022]
Abstract
Fibroblast migration is a central process in skin wound healing, which requires the coordination of several types of growth factors. bFGF, a well-known fibroblast growth factor (FGF), is able to accelerate fibroblast migration; however, the underlying mechanism of bFGF regulation fibroblast migration remains unclear. Through the RNA-seq analysis, we had identified that the hedgehog (Hh) canonical pathway genes including Smoothened (Smo) and Gli1, were regulated by bFGF. Further analysis revealed that activation of the Hh pathway via up-regulation of Smo promoted fibroblast migration, invasion, and skin wound healing, but which significantly reduced by GANT61, a selective antagonist of Gli1/Gli2. Western blot analyses and siRNA transfection assays demonstrated that Smo acted upstream of phosphoinositide 3-kinase (PI3K)-c-Jun N-terminal kinase (JNK)-β-catenin to promote cell migration. Moreover, RNA-seq and qRT-PCR analyses revealed that Hh pathway genes including Smo and Gli1 were under control of β-catenin, suggesting that β-catenin turn feedback activates Hh signaling. Taken together, our analyses identified a new bFGF-regulating mechanism by which Hh signaling regulates human fibroblast migration, and the data presented here opens a new avenue for the wound healing therapy.
Collapse
Affiliation(s)
- Zhong Xin Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong Cong Sun
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Wenzhou People's Hospital, Wenzhou, Zhejiang, China
| | - Yu Ting Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Li Sha Chi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wan Hui Cai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Xuan Zhou
- Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Wei Tao Cong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiao Kun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Li Tai Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
26
|
Genthe JR, Clements WK. R-spondin 1 is required for specification of hematopoietic stem cells through Wnt16 and Vegfa signaling pathways. Development 2017; 144:590-600. [PMID: 28087636 DOI: 10.1242/dev.139956] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 12/22/2016] [Indexed: 01/18/2023]
Abstract
Hematopoietic stem cells (HSCs) are the therapeutic component of bone marrow transplants, but finding immune-compatible donors limits treatment availability and efficacy. Recapitulation of endogenous specification during development is a promising approach to directing HSC specification in vitro, but current protocols are not capable of generating authentic HSCs with high efficiency. Across phyla, HSCs arise from hemogenic endothelium in the ventral floor of the dorsal aorta concurrent with arteriovenous specification and intersegmental vessel (ISV) sprouting, processes regulated by Notch and Wnt. We hypothesized that coordination of HSC specification with vessel patterning might involve modulatory regulatory factors such as R-spondin 1 (Rspo1), an extracellular protein that enhances β-catenin-dependent Wnt signaling and has previously been shown to regulate ISV patterning. We find that Rspo1 is required for HSC specification through control of parallel signaling pathways controlling HSC specification: Wnt16/DeltaC/DeltaD and Vegfa/Tgfβ1. Our results define Rspo1 as a key upstream regulator of two crucial pathways necessary for HSC specification.
Collapse
Affiliation(s)
- Jamie R Genthe
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wilson K Clements
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
27
|
Ciau-Uitz A, Patient R. The embryonic origins and genetic programming of emerging haematopoietic stem cells. FEBS Lett 2016; 590:4002-4015. [PMID: 27531714 DOI: 10.1002/1873-3468.12363] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/26/2016] [Accepted: 08/12/2016] [Indexed: 11/10/2022]
Abstract
Haematopoietic stem cells (HSCs) emerge from the haemogenic endothelium (HE) localised in the ventral wall of the embryonic dorsal aorta (DA). The HE generates HSCs through a process known as the endothelial to haematopoietic transition (EHT), which has been visualised in live embryos and is currently under intense study. However, EHT is the culmination of multiple programming events, which are as yet poorly understood, that take place before the specification of HE. A number of haematopoietic precursor cells have been described before the emergence of definitive HSCs, but only one haematovascular progenitor, the definitive haemangioblast (DH), gives rise to the DA, HE and HSCs. DHs emerge in the lateral plate mesoderm (LPM) and have a distinct origin and genetic programme compared to other, previously described haematovascular progenitors. Although DHs have so far only been established in Xenopus embryos, evidence for their existence in the LPM of mouse and chicken embryos is discussed here. We also review the current knowledge of the origins, lineage relationships, genetic programming and differentiation of the DHs that leads to the generation of HSCs. Importantly, we discuss the significance of the gene regulatory network (GRN) that controls the programming of DHs, a better understanding of which may aid in the establishment of protocols for the de novo generation of HSCs in vitro.
Collapse
Affiliation(s)
- Aldo Ciau-Uitz
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, UK
| | - Roger Patient
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, UK
| |
Collapse
|
28
|
Syndecan-4 regulates the bFGF-induced chemotactic migration of endothelial cells. J Mol Histol 2016; 47:503-9. [PMID: 27541034 DOI: 10.1007/s10735-016-9693-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 08/13/2016] [Indexed: 12/23/2022]
Abstract
Chemotactic migration of endothelial cells (ECs) guided by extracellular attractants is essential for blood vessel formation. Synd4 is a ubiquitous heparin sulfate proteoglycan receptor on the cell surface that has been identified to promote angiogenesis during tissue repair. Here, the role synd4 played in chemotactic migration of ECs was investigated in vitro and in vivo. Human umbilical vein endothelial cells (HUVECs) were transfected with Lenti-synd4-RNAi or Lenti-null. Cell migration was observed in a 2D-chemotaxis slide with a stable gradient of basic fibroblast growth factor (bFGF) for 18 h using time-lapse microscopy. Synd4 knockdown HUVECs showed reduced mobility compared with the control. In animal studies, Matrigel premixed with bFGF was used to induce the migration of ECs. The cells migrated less distance from the skin in the Matrigel plugs of synd4 null mice compared with the control mice. Then recombinant adenoviruses containing the synd4 gene (Ad-synd4) or null (Ad-null) were constructed to enhance the synd4 expression of migratory cells in Matrigel plugs of wild-type mice. Migratory cells with synd4 overexpression did not invade further in the Matrigel plugs of wild-type mice, but showed a high ability to proliferate.
Collapse
|
29
|
Koenig AL, Baltrunaite K, Bower NI, Rossi A, Stainier DYR, Hogan BM, Sumanas S. Vegfa signaling promotes zebrafish intestinal vasculature development through endothelial cell migration from the posterior cardinal vein. Dev Biol 2016; 411:115-27. [PMID: 26769101 DOI: 10.1016/j.ydbio.2016.01.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 12/12/2015] [Accepted: 01/04/2016] [Indexed: 10/22/2022]
Abstract
The mechanisms underlying organ vascularization are not well understood. The zebrafish intestinal vasculature forms early, is easily imaged using transgenic lines and in-situ hybridization, and develops in a stereotypical pattern thus making it an excellent model for investigating mechanisms of organ specific vascularization. Here, we demonstrate that the sub-intestinal vein (SIV) and supra-intestinal artery (SIA) form by a novel mechanism from angioblasts that migrate out of the posterior cardinal vein and coalesce to form the intestinal vasculature in an anterior to posterior wave with the SIA forming after the SIV. We show that vascular endothelial growth factor aa (vegfaa) is expressed in the endoderm at the site where intestinal vessels form and therefore likely provides a guidance signal. Vegfa/Vegfr2 signaling is required for early intestinal vasculature development with mutation in vegfaa or loss of Vegfr2 homologs causing nearly complete inhibition of the formation of the intestinal vasculature. Vegfc and Vegfr3 function, however, are dispensable for intestinal vascularization. Interestingly, ubiquitous overexpression of Vegfc resulted in an overgrowth of the SIV, suggesting that Vegfc is sufficient to induce SIV development. These results argue that Vegfa signaling directs endothelial cells to migrate out of existing vasculature and coalesce to form the intestinal vessels. It is likely that a similar mechanism is utilized during vascularization of other organs.
Collapse
Affiliation(s)
- Andrew L Koenig
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Kristina Baltrunaite
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Neil I Bower
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4073, Australia.
| | - Andrea Rossi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4073, Australia.
| | - Saulius Sumanas
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
30
|
Rana U, Liu Z, Kumar SN, Zhao B, Hu W, Bordas M, Cossette S, Szabo S, Foeckler J, Weiler H, Chrzanowska-Wodnicka M, Holtz ML, Misra RP, Salato V, North PE, Ramchandran R, Miao QR. Nogo-B receptor deficiency causes cerebral vasculature defects during embryonic development in mice. Dev Biol 2015; 410:190-201. [PMID: 26746789 DOI: 10.1016/j.ydbio.2015.12.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/21/2015] [Accepted: 12/21/2015] [Indexed: 01/07/2023]
Abstract
Nogo-B receptor (NgBR) was identified as a receptor specific for Nogo-B. Our previous work has shown that Nogo-B and its receptor (NgBR) are essential for chemotaxis and morphogenesis of endothelial cells in vitro and intersomitic vessel formation via Akt pathway in zebrafish. Here, we further demonstrated the roles of NgBR in regulating vasculature development in mouse embryo and primitive blood vessel formation in embryoid body culture systems, respectively. Our results showed that NgBR homozygous knockout mice are embryonically lethal at E7.5 or earlier, and Tie2Cre-mediated endothelial cell-specific NgBR knockout (NgBR ecKO) mice die at E11.5 and have severe blood vessel assembly defects in embryo. In addition, mutant embryos exhibit dilation of cerebral blood vessel, resulting in thin-walled endothelial caverns. The similar vascular defects also were detected in Cdh5(PAC)-CreERT2 NgBR inducible ecKO mice. Murine NgBR gene-targeting embryonic stem cells (ESC) were generated by homologous recombination approaches. Homozygous knockout of NgBR in ESC results in cell apoptosis. Heterozygous knockout of NgBR does not affect ESC cell survival, but reduces the formation and branching of primitive blood vessels in embryoid body culture systems. Mechanistically, NgBR knockdown not only decreases both Nogo-B and VEGF-stimulated endothelial cell migration by abolishing Akt phosphorylation, but also decreases the expression of CCM1 and CCM2 proteins. Furthermore, we performed immunofluorescence (IF) staining of NgBR in human cerebral cavernous malformation patient tissue sections. The quantitative analysis results showed that NgBR expression levels in CD31 positive endothelial cells is significantly decreased in patient tissue sections. These results suggest that NgBR may be one of important genes coordinating the cerebral vasculature development.
Collapse
Affiliation(s)
- Ujala Rana
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Zhong Liu
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Suresh N Kumar
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Baofeng Zhao
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wenquan Hu
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michelle Bordas
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stephanie Cossette
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sara Szabo
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jamie Foeckler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; BloodCenter of Wisconsin, Milwaukee, WI 53226, USA
| | - Hartmut Weiler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; BloodCenter of Wisconsin, Milwaukee, WI 53226, USA
| | | | - Mary L Holtz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ravindra P Misra
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Valerie Salato
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Paula E North
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ramani Ramchandran
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Qing Robert Miao
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
31
|
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
|
32
|
Butko E, Pouget C, Traver D. Complex regulation of HSC emergence by the Notch signaling pathway. Dev Biol 2015; 409:129-138. [PMID: 26586199 DOI: 10.1016/j.ydbio.2015.11.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/11/2015] [Accepted: 11/12/2015] [Indexed: 01/13/2023]
Abstract
Hematopoietic stem cells are formed during embryonic development, and serve as the foundation of the definitive blood program for life. Notch signaling has been well established as an essential direct contributor to HSC specification. However, several recent studies have indicated that the contribution of Notch signaling is complex. HSC specification requires multiple Notch signaling inputs, some received directly by hematopoietic precursors, and others that occur indirectly within neighboring somites. Of note, proinflammatory signals provided by primitive myeloid cells are needed for HSC specification via upregulation of the Notch pathway in hemogenic endothelium. In addition to multiple requirements for Notch activation, recent studies indicate that Notch signaling must subsequently be repressed to permit HSC emergence. Finally, Notch must then be reactivated to maintain HSC fate. In this review, we discuss the growing understanding of the dynamic contributions of Notch signaling to the establishment of hematopoiesis during development.
Collapse
Affiliation(s)
- Emerald Butko
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Claire Pouget
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
33
|
Hasan SS, Siekmann AF. The same but different: signaling pathways in control of endothelial cell migration. Curr Opin Cell Biol 2015; 36:86-92. [DOI: 10.1016/j.ceb.2015.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/16/2015] [Accepted: 07/19/2015] [Indexed: 11/24/2022]
|
34
|
Adachi H, Tominaga H, Maruyama Y, Yoneda K, Maruyama K, Yoshii K, Kinoshita S, Nakano M, Tashiro K. Stage-specific reference genes significant for quantitative PCR during mouse retinal development. Genes Cells 2015; 20:625-35. [PMID: 26059597 DOI: 10.1111/gtc.12254] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/27/2015] [Indexed: 11/26/2022]
Abstract
Developing mouse retina has been serving as an ideal model for investigating the molecular mechanism of neural development and angiogenesis, because several significant events associated with these physiological phenomena are drastically occurring in conjunction with retinal development. However, as many genes are influencing on each other to establish mature retina within 21 days from E10 to P12, we must carefully design the experiments, such as in the case of quantitating the amount of altered gene expression toward the establishment of retina by quantitative PCR. As we have seen considerable variations of quantitative results in different developmental stages of retina depending on the reference genes used for compensation, we here attempted to determine a reliable reference gene to accurately quantitate the target genes in each stage. According to the results of in silico prediction and comparison with a database of SAGE, we found that the most stable gene from early to late stages was Sdha, whereas one of the most popular housekeeping genes, Actb, was the one that could mislead the quantitative results even in the adult stage. Consequently, we pointed out the importance of selecting an appropriate reference gene, especially to quantitate the amount of gene expression in the developmental stages of a certain tissue.
Collapse
Affiliation(s)
- Hiroko Adachi
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hiroyuki Tominaga
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yuko Maruyama
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazuhito Yoneda
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kengo Yoshii
- Department of Medical Statistics, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Shigeru Kinoshita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masakazu Nakano
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| |
Collapse
|
35
|
Meadows SM, Cleaver O. Vascular patterning: coordinated signals keep blood vessels on track. Curr Opin Genet Dev 2015; 32:86-91. [DOI: 10.1016/j.gde.2015.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 11/24/2022]
|
36
|
Helker CSM, Schuermann A, Pollmann C, Chng SC, Kiefer F, Reversade B, Herzog W. The hormonal peptide Elabela guides angioblasts to the midline during vasculogenesis. eLife 2015; 4. [PMID: 26017639 PMCID: PMC4468421 DOI: 10.7554/elife.06726] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022] Open
Abstract
A key step in the de novo formation of the embryonic vasculature is the migration of endothelial precursors, the angioblasts, to the position of the future vessels. To form the first axial vessels, angioblasts migrate towards the midline and coalesce underneath the notochord. Vascular endothelial growth factor has been proposed to serve as a chemoattractant for the angioblasts and to regulate this medial migration. Here we challenge this model and instead demonstrate that angioblasts rely on their intrinsic expression of Apelin receptors (Aplr, APJ) for their migration to the midline. We further show that during this angioblast migration Apelin receptor signaling is mainly triggered by the recently discovered ligand Elabela (Ela). As neither of the ligands Ela or Apelin (Apln) nor their receptors have previously been implicated in regulating angioblast migration, we hereby provide a novel mechanism for regulating vasculogenesis, with direct relevance to physiological and pathological angiogenesis. DOI:http://dx.doi.org/10.7554/eLife.06726.001 The circulatory system enables blood to move around the body and deliver substances including nutrients and oxygen to the cells that need them. In the embryos of animals with a backbone, blood flows from the heart through the aorta into branching smaller vessels to the cells. The blood then gets collected by progressively bigger vessels and flows back to the heart via the cardinal vein. The cells that make up these blood vessels develop from cells called angioblasts—but first, during development these angioblasts must move to the place where the vessels will form. A protein called Vascular endothelial growth factor (VEGF) had been suggested to help guide and align the angioblasts as the embryo develops. Now, Helker, Schuermann et al. have examined developing zebrafish embryos using new technologies. This revealed that VEGF is in fact not essential for the dorsal aorta and cardinal vein to develop. Instead, the angioblasts only move to the correct part of the embryo if they can produce the Apelin receptor protein, which forms part of a signaling pathway. There are two hormones—called Apelin and Elabela—that can bind to and activate the Apelin receptor. Helker, Schuermann et al. show that Elabela alone is needed to guide the angioblasts to the right part of the embryo during blood vessel development. However, in embryos where there is not enough Elabela, the Apelin hormone can compensate for this deficiency and the first blood vessels will later develop correctly. Future research will address whether this signaling pathway not only guides angioblasts to establish a circulatory system, but also guides blood vessel growth. As blood vessel growth is very relevant to human disease, identifying the mechanisms that regulate it will have an impact on biomedical research. DOI:http://dx.doi.org/10.7554/eLife.06726.002
Collapse
Affiliation(s)
| | | | - Cathrin Pollmann
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Serene C Chng
- Institute of Medical Biology, Human Genetics and Embryology Laboratory, A*STAR, Singapore, Singapore
| | | | - Bruno Reversade
- Institute of Medical Biology, Human Genetics and Embryology Laboratory, A*STAR, Singapore, Singapore
| | | |
Collapse
|
37
|
Simons M, Alitalo K, Annex BH, Augustin HG, Beam C, Berk BC, Byzova T, Carmeliet P, Chilian W, Cooke JP, Davis GE, Eichmann A, Iruela-Arispe ML, Keshet E, Sinusas AJ, Ruhrberg C, Woo YJ, Dimmeler S. State-of-the-Art Methods for Evaluation of Angiogenesis and Tissue Vascularization: A Scientific Statement From the American Heart Association. Circ Res 2015; 116:e99-132. [PMID: 25931450 DOI: 10.1161/res.0000000000000054] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
38
|
Chang SL, Cavnar SP, Takayama S, Luker GD, Linderman JJ. Cell, isoform, and environment factors shape gradients and modulate chemotaxis. PLoS One 2015; 10:e0123450. [PMID: 25909600 PMCID: PMC4409393 DOI: 10.1371/journal.pone.0123450] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/04/2015] [Indexed: 12/02/2022] Open
Abstract
Chemokine gradient formation requires multiple processes that include ligand secretion and diffusion, receptor binding and internalization, and immobilization of ligand to surfaces. To understand how these events dynamically shape gradients and influence ensuing cell chemotaxis, we built a multi-scale hybrid agent-based model linking gradient formation, cell responses, and receptor-level information. The CXCL12/CXCR4/CXCR7 signaling axis is highly implicated in metastasis of many cancers. We model CXCL12 gradient formation as it is impacted by CXCR4 and CXCR7, with particular focus on the three most highly expressed isoforms of CXCL12. We trained and validated our model using data from an in vitro microfluidic source-sink device. Our simulations demonstrate how isoform differences on the molecular level affect gradient formation and cell responses. We determine that ligand properties specific to CXCL12 isoforms (binding to the migration surface and to CXCR4) significantly impact migration and explain differences in in vitro chemotaxis data. We extend our model to analyze CXCL12 gradient formation in a tumor environment and find that short distance, steep gradients characteristic of the CXCL12-γ isoform are effective at driving chemotaxis. We highlight the importance of CXCL12-γ in cancer cell migration: its high effective affinity for both extracellular surface sites and CXCR4 strongly promote CXCR4+ cell migration. CXCL12-γ is also more difficult to inhibit, and we predict that co-inhibition of CXCR4 and CXCR7 is necessary to effectively hinder CXCL12-γ-induced migration. These findings support the growing importance of understanding differences in protein isoforms, and in particular their implications for cancer treatment.
Collapse
Affiliation(s)
- S. Laura Chang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Stephen P. Cavnar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gary D. Luker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jennifer J. Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
39
|
Charpentier MS, Tandon P, Trincot CE, Koutleva EK, Conlon FL. A distinct mechanism of vascular lumen formation in Xenopus requires EGFL7. PLoS One 2015; 10:e0116086. [PMID: 25705891 PMCID: PMC4338030 DOI: 10.1371/journal.pone.0116086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/04/2014] [Indexed: 01/03/2023] Open
Abstract
During vertebrate blood vessel development, lumen formation is the critical process by which cords of endothelial cells transition into functional tubular vessels. Here, we use Xenopus embryos to explore the cellular and molecular mechanisms underlying lumen formation of the dorsal aorta and the posterior cardinal veins, the primary major vessels that arise via vasculogenesis within the first 48 hours of life. We demonstrate that endothelial cells are initially found in close association with one another through the formation of tight junctions expressing ZO-1. The emergence of vascular lumens is characterized by elongation of endothelial cell shape, reorganization of junctions away from the cord center to the periphery of the vessel, and onset of Claudin-5 expression within tight junctions. Furthermore, unlike most vertebrate vessels that exhibit specialized apical and basal domains, we show that early Xenopus vessels are not polarized. Moreover, we demonstrate that in embryos depleted of the extracellular matrix factor Epidermal Growth Factor-Like Domain 7 (EGFL7), an evolutionarily conserved factor associated with vertebrate vessel development, vascular lumens fail to form. While Claudin-5 localizes to endothelial tight junctions of EGFL7-depleted embryos in a timely manner, endothelial cells of the aorta and veins fail to undergo appropriate cell shape changes or clear junctions from the cell-cell contact. Taken together, we demonstrate for the first time the mechanisms by which lumens are generated within the major vessels in Xenopus and implicate EGFL7 in modulating cell shape and cell-cell junctions to drive proper lumen morphogenesis.
Collapse
Affiliation(s)
- Marta S. Charpentier
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Panna Tandon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Claire E. Trincot
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Elitza K. Koutleva
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Frank L. Conlon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
40
|
Lee Y, Manegold JE, Kim AD, Pouget C, Stachura DL, Clements WK, Traver D. FGF signalling specifies haematopoietic stem cells through its regulation of somitic Notch signalling. Nat Commun 2014; 5:5583. [PMID: 25428693 PMCID: PMC4271318 DOI: 10.1038/ncomms6583] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/16/2014] [Indexed: 01/07/2023] Open
Abstract
Hematopoietic stem cells (HSCs) derive from hemogenic endothelial cells of the primitive dorsal aorta (DA) during vertebrate embryogenesis. The molecular mechanisms governing this unique endothelial to hematopoietic transition remain unclear. Here, we demonstrate a novel requirement for fibroblast growth factor (FGF) signaling in HSC emergence. This requirement is non-cell-autonomous, and acts within the somite to bridge the Wnt and Notch signaling pathways. We previously demonstrated that Wnt16 regulates the somitic expression of two Notch ligands, deltaC (dlc) and deltaD (dld), whose combined function is required for HSC fate. How Wnt16 connects to Notch function has remained an open question. Our current studies demonstrate that FGF signaling, via FGF receptor 4 (Fgfr4), mediates a signal transduction pathway between Wnt16 and Dlc, but not Dld, to regulate HSC specification. Our findings demonstrate that FGF signaling acts as a key molecular relay within the developmental HSC niche to instruct HSC fate.
Collapse
Affiliation(s)
- Yoonsung Lee
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Jennifer E Manegold
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Albert D Kim
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Claire Pouget
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - David L Stachura
- 1] Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA [2] Department of Biological Sciences, California State University, Chico, California 95929, USA
| | - Wilson K Clements
- 1] Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA [2] Department of Hematology, St Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - David Traver
- Department of Cellular and Molecular Medicine and Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
41
|
Michaelis UR. Mechanisms of endothelial cell migration. Cell Mol Life Sci 2014; 71:4131-48. [PMID: 25038776 PMCID: PMC11113960 DOI: 10.1007/s00018-014-1678-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/23/2014] [Accepted: 07/07/2014] [Indexed: 01/13/2023]
Abstract
Cell migration plays a central role in a variety of physiological and pathological processes during our whole life. Cellular movement is a complex, tightly regulated multistep process. Although the principle mechanisms of migration follow a defined general motility cycle, the cell type and the context of moving influences the detailed mode of migration. Endothelial cells migrate during vasculogenesis and angiogenesis but also in a damaged vessel to restore vessel integrity. Depending on the situation they migrate individually, in chains or sheets and complex signaling, intercellular signals as well as environmental cues modulate the process. Here, the different modes of cell migration, the peculiarities of endothelial cell migration and specific guidance molecules controlling this process will be reviewed.
Collapse
Affiliation(s)
- U Ruth Michaelis
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany,
| |
Collapse
|
42
|
Tanaka Y, Sanchez V, Takata N, Yokomizo T, Yamanaka Y, Kataoka H, Hoppe P, Schroeder T, Nishikawa SI. Circulation-Independent Differentiation Pathway from Extraembryonic Mesoderm toward Hematopoietic Stem Cells via Hemogenic Angioblasts. Cell Rep 2014; 8:31-9. [DOI: 10.1016/j.celrep.2014.05.055] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 03/15/2014] [Accepted: 05/29/2014] [Indexed: 10/25/2022] Open
|
43
|
Developmental hematopoiesis: ontogeny, genetic programming and conservation. Exp Hematol 2014; 42:669-83. [PMID: 24950425 DOI: 10.1016/j.exphem.2014.06.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/15/2014] [Accepted: 06/09/2014] [Indexed: 02/01/2023]
Abstract
Hematopoietic stem cells (HSCs) sustain blood production throughout life and are of pivotal importance in regenerative medicine. Although HSC generation from pluripotent stem cells would resolve their shortage for clinical applications, this has not yet been achieved mainly because of the poor mechanistic understanding of their programming. Bone marrow HSCs are first created during embryogenesis in the dorsal aorta (DA) of the midgestation conceptus, from where they migrate to the fetal liver and, eventually, the bone marrow. It is currently accepted that HSCs emerge from specialized endothelium, the hemogenic endothelium, localized in the ventral wall of the DA through an evolutionarily conserved process called the endothelial-to-hematopoietic transition. However, the endothelial-to-hematopoietic transition represents one of the last steps in HSC creation, and an understanding of earlier events in the specification of their progenitors is required if we are to create them from naïve pluripotent cells. Because of their ready availability and external development, zebrafish and Xenopus embryos have enormously facilitated our understanding of the early developmental processes leading to the programming of HSCs from nascent lateral plate mesoderm to hemogenic endothelium in the DA. The amenity of the Xenopus model to lineage tracing experiments has also contributed to the establishment of the distinct origins of embryonic (yolk sac) and adult (HSC) hematopoiesis, whereas the transparency of the zebrafish has allowed in vivo imaging of developing blood cells, particularly during and after the emergence of HSCs in the DA. Here, we discuss the key contributions of these model organisms to our understanding of developmental hematopoiesis.
Collapse
|
44
|
Lim JC, Kurihara S, Tamaki R, Mashima Y, Maéno M. Expression and localization of Rdd proteins in Xenopus embryo. Anat Cell Biol 2014; 47:18-27. [PMID: 24693479 PMCID: PMC3968263 DOI: 10.5115/acb.2014.47.1.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/10/2013] [Accepted: 02/07/2014] [Indexed: 11/27/2022] Open
Abstract
The previous study has shown that repeated D domain-like (Rdd) proteins, a group of novel secretory proteins consisting of repeated domains of a cysteine-rich sequence, are involved in the process of blood vessel formation in Xenopus embryo. We performed further experiments to examine the localization of Rdd proteins in embryogenesis. Detection of tagged Rdd proteins expressed in blastomeres showed that Rdd proteins formed a high molecular weight complex and existed in the extracellular space. A rabbit antibody against the Rdd synthetic peptide identified a single band of 28 kD in embryonic tissue extract. By whole-mount immunostaining analysis, signal was detected in the regions of inter-somites, vitelline veins, and branchial arches at the tailbud stage. Staining of Rdd was remarkably reduced in the embryos injected with vascular endothelial growth factor Morpholino. We suggest that Rdd proteins interact with a molecule(s) associated with vascular precursor cells.
Collapse
Affiliation(s)
- Jong-Chan Lim
- Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Sayaka Kurihara
- Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Rie Tamaki
- Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Yutaka Mashima
- Graduate School of Science and Technology, Niigata University, Niigata, Japan
| | - Mitsugu Maéno
- Graduate School of Science and Technology, Niigata University, Niigata, Japan
| |
Collapse
|
45
|
Wilkinson RN, van Eeden FJ. The Zebrafish as a Model of Vascular Development and Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:93-122. [DOI: 10.1016/b978-0-12-386930-2.00005-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
46
|
Köhn-Luque A, de Back W, Yamaguchi Y, Yoshimura K, Herrero MA, Miura T. Dynamics of VEGF matrix-retention in vascular network patterning. Phys Biol 2013; 10:066007. [DOI: 10.1088/1478-3975/10/6/066007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Ciau-Uitz A, Wang L, Patient R, Liu F. ETS transcription factors in hematopoietic stem cell development. Blood Cells Mol Dis 2013; 51:248-55. [DOI: 10.1016/j.bcmd.2013.07.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/04/2013] [Indexed: 01/08/2023]
|
48
|
Li F, Xu L, Gai X, Zhou Z, Wang L, Zhang H, Gai Y, Song L, Yu J, Liang C. The involvement of PDGF/VEGF related factor in regulation of immune and neuroendocrine in Chinese mitten crab Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2013; 35:1240-1248. [PMID: 23933264 DOI: 10.1016/j.fsi.2013.07.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/28/2013] [Accepted: 07/28/2013] [Indexed: 06/02/2023]
Abstract
Members of the platelet-derived growth factor/vascular endothelial growth factor (PDGF/VEGF) family have been implicated in cell proliferation, cell differentiation, and cell migration, vascular development, angiogenesis and neural development. In the present study, a novel PDGF/VEGF related factor gene was cloned and identified in Chinese mitten crab Eriocheir sinensis (designated as EsPVF1). The full-length cDNA of EsPVF1 was of 1173 bp, consisting a 5' untranslated region (UTR) of 54 bp, a 3' UTR of 1131 bp with a poly (A) tail, and an open reading frame (ORF) of 588 bp encoding 196 amino acid residues. A signal peptide of 20 amino acid residues, a PDGF/VEGF homology growth factor domain of 81 amino acids, and a typical cysteine knot motif (CXCXC) were identified in the deduced amino acid sequence of EsPVF1. By fluorescent quantitative real-time PCR, the EsPVF1 mRNA was detected ubiquitously in the select tissues of hemocytes, gonad, heart, muscle, hepatopancreas and gill, with the high abundance in hemocytes and gonad. The mRNA expression level of EsPVF1 was up-regulated and reached the highest at 24 h after Vibrio anguillarum challenge, while it was induced at 3 h, 6 h, 12 h, 24 h and 48 h compared with the untreated group after Pichia pastoris GS115 challenge. Tissue injury also induced the mRNA expression of EsPVF1 in hemocytes of crabs, and the expression level increased obviously at 8 h. The cDNA fragment encoding mature peptide of EsPVF1 was recombined and expressed in Escherichia coli BL21 (DE3) pLysS. Biogenic amine in hemolymph pre-incubated with recombinant protein of EsPVF1 (rEsPVF1) was detected by fluorimetric method. Norepinephrine and dopamine in hemolymph incubated with rEsPVF1 were higher than that in the blank group. Therefore, EsPVF1 could significantly provoke the release of norepinephrine and dopamine. The results collectively indicated that EsPVF1 was involved in regulation of the immune response and neuroendocrine system in crabs.
Collapse
Affiliation(s)
- Fengmei Li
- Qingdao University of Science and Technology, Qingdao 266042, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lewis C, Krieg PA. Reagents for developmental regulation of Hedgehog signaling. Methods 2013; 66:390-7. [PMID: 23981360 DOI: 10.1016/j.ymeth.2013.08.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/10/2013] [Accepted: 08/13/2013] [Indexed: 12/11/2022] Open
Abstract
We have examined a number of reagents for their ability to modulate activity of the Hh signaling pathway during embryonic development of Xenopus. In particular we have focused on regulation of events occurring during tailbud stages and later. Two inducible protein reagents based on the Gli1 and Gli3 transcription factors were generated and the activity of these proteins was compared to the Hh signaling pathway inhibitor, cyclopamine, and the activators, Smoothened agonist (SAG) and purmorphamine (PMA). Effectiveness of reagents was assayed using both molecular biological techniques and biological readouts. We found that the small molecule modulators of the Hh pathway were highly specific and effective and produced results generally superior to the more conventional protein reagents for examination of later stage developmental processes.
Collapse
Affiliation(s)
- Cristy Lewis
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Paul A Krieg
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ, United States.
| |
Collapse
|
50
|
miR-142-3p Controls the Specification of Definitive Hemangioblasts during Ontogeny. Dev Cell 2013; 26:237-49. [DOI: 10.1016/j.devcel.2013.06.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 05/07/2013] [Accepted: 06/23/2013] [Indexed: 02/03/2023]
|