Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Genes Dev ; 34(13-14): 950-964, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32499402

ABSTRACT

Hematopoietic stem cell (HSC) ontogeny is accompanied by dynamic changes in gene regulatory networks. We performed RNA-seq and histone mark ChIP-seq to define the transcriptomes and epigenomes of cells representing key developmental stages of HSC ontogeny in mice. The five populations analyzed were embryonic day 10.5 (E10.5) endothelium and hemogenic endothelium from the major arteries, an enriched population of prehematopoietic stem cells (pre-HSCs), fetal liver HSCs, and adult bone marrow HSCs. Using epigenetic signatures, we identified enhancers for each developmental stage. Only 12% of enhancers are primed, and 78% are active, suggesting the vast majority of enhancers are established de novo without prior priming in earlier stages. We constructed developmental stage-specific transcriptional regulatory networks by linking enhancers and predicted bound transcription factors to their target promoters using a novel computational algorithm, target inference via physical connection (TIPC). TIPC predicted known transcriptional regulators for the endothelial-to-hematopoietic transition, validating our overall approach, and identified putative novel transcription factors, including the broadly expressed transcription factors SP3 and MAZ. Finally, we validated a role for SP3 and MAZ in the formation of hemogenic endothelium. Our data and computational analyses provide a useful resource for uncovering regulators of HSC formation.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Algorithms , Animals , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/genetics , Gene Editing , Mice , Sp3 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcriptome
2.
Development ; 150(16)2023 08 15.
Article in English | MEDLINE | ID: mdl-37642459

ABSTRACT

The vasculature consists of vessels of different sizes that are arranged in a hierarchical pattern. Two cell populations work in concert to establish this pattern during embryonic development and adopt it to changes in blood flow demand later in life: endothelial cells that line the inner surface of blood vessels, and adjacent vascular mural cells, including smooth muscle cells and pericytes. Despite recent progress in elucidating the signalling pathways controlling their crosstalk, much debate remains with regard to how mural cells influence endothelial cell biology and thereby contribute to the regulation of blood vessel formation and diameters. In this Review, I discuss mural cell functions and their interactions with endothelial cells, focusing on how these interactions ensure optimal blood flow patterns. Subsequently, I introduce the signalling pathways controlling mural cell development followed by an overview of mural cell ontogeny with an emphasis on the distinguishing features of mural cells located on different types of blood vessels. Ultimately, I explore therapeutic strategies involving mural cells to alleviate tissue ischemia and improve vascular efficiency in a variety of diseases.


Subject(s)
Blood Cells , Endothelial Cells , Female , Pregnancy , Humans , Cell Differentiation , Embryonic Development , Biology
3.
Development ; 150(7)2023 04 01.
Article in English | MEDLINE | ID: mdl-36938965

ABSTRACT

Blood vessels form elaborate networks that depend on tissue-specific signalling pathways and anatomical structures to guide their growth. However, it is not clear which morphogenetic principles organize the stepwise assembly of the vasculature. We therefore performed a longitudinal analysis of zebrafish caudal fin vascular assembly, revealing the existence of temporally and spatially distinct morphogenetic processes. Initially, vein-derived endothelial cells (ECs) generated arteries in a reiterative process requiring vascular endothelial growth factor (Vegf), Notch and cxcr4a signalling. Subsequently, veins produced veins in more proximal fin regions, transforming pre-existing artery-vein loops into a three-vessel pattern consisting of an artery and two veins. A distinct set of vascular plexuses formed at the base of the fin. They differed in their diameter, flow magnitude and marker gene expression. At later stages, intussusceptive angiogenesis occurred from veins in distal fin regions. In proximal fin regions, we observed new vein sprouts crossing the inter-ray tissue through sprouting angiogenesis. Together, our results reveal a surprising diversity among the mechanisms generating the mature fin vasculature and suggest that these might be driven by separate local cues.


Subject(s)
Endothelial Cells , Zebrafish , Animals , Zebrafish/genetics , Vascular Endothelial Growth Factor A/metabolism , Neovascularization, Physiologic , Veins/metabolism
4.
Development ; 149(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35297968

ABSTRACT

Vascular networks comprise endothelial cells and mural cells, which include pericytes and smooth muscle cells. To elucidate the mechanisms controlling mural cell recruitment during development and tissue regeneration, we studied zebrafish caudal fin arteries. Mural cells colonizing arteries proximal to the body wrapped around them, whereas those in more distal regions extended protrusions along the proximo-distal vascular axis. Both cell populations expressed platelet-derived growth factor receptor ß (pdgfrb) and the smooth muscle cell marker myosin heavy chain 11a (myh11a). Most wrapping cells in proximal locations additionally expressed actin alpha2, smooth muscle (acta2). Loss of Pdgfrb signalling specifically decreased mural cell numbers at the vascular front. Using lineage tracing, we demonstrate that precursor cells located in periarterial regions and expressing Pgdfrb can give rise to mural cells. Studying tissue regeneration, we did not find evidence that newly formed mural cells were derived from pre-existing cells. Together, our findings reveal conserved roles for Pdgfrb signalling in development and regeneration, and suggest a limited capacity of mural cells to self-renew or contribute to other cell types during tissue regeneration.


Subject(s)
Myocytes, Smooth Muscle , Pericytes , Receptor, Platelet-Derived Growth Factor beta , Zebrafish Proteins , Zebrafish , Animals , Endothelial Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Nat Chem Biol ; 18(2): 152-160, 2022 02.
Article in English | MEDLINE | ID: mdl-34937907

ABSTRACT

We describe single-component optogenetic probes whose activation dynamics depend on both light and temperature. We used the BcLOV4 photoreceptor to stimulate Ras and phosphatidyl inositol-3-kinase signaling in mammalian cells, allowing activation over a large dynamic range with low basal levels. Surprisingly, we found that BcLOV4 membrane translocation dynamics could be tuned by both light and temperature such that membrane localization spontaneously decayed at elevated temperatures despite constant illumination. Quantitative modeling predicted BcLOV4 activation dynamics across a range of light and temperature inputs and thus provides an experimental roadmap for BcLOV4-based probes. BcLOV4 drove strong and stable signal activation in both zebrafish and fly cells, and thermal inactivation provided a means to multiplex distinct blue-light sensitive tools in individual mammalian cells. BcLOV4 is thus a versatile photosensor with unique light and temperature sensitivity that enables straightforward generation of broadly applicable optogenetic tools.


Subject(s)
Cell Communication/physiology , Optogenetics , Phosphatidylinositol 3-Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Line , Drosophila , Embryo, Nonmammalian , Mice , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , Temperature , Zebrafish , ras Proteins/genetics
6.
Dev Biol ; 486: 26-43, 2022 06.
Article in English | MEDLINE | ID: mdl-35337795

ABSTRACT

The formation of appropriately patterned blood vessel networks requires endothelial cell migration and proliferation. Signaling through the Vascular Endothelial Growth Factor A (VEGFA) pathway is instrumental in coordinating these processes. mRNA splicing generates short (diffusible) and long (extracellular matrix bound) Vegfa isoforms. The differences between these isoforms in controlling cellular functions are not understood. In zebrafish, vegfaa generates short and long isoforms, while vegfab only generates long isoforms. We found that mutations in vegfaa had an impact on endothelial cell (EC) migration and proliferation. Surprisingly, mutations in vegfab more strongly affected EC proliferation in distinct blood vessels, such as intersegmental blood vessels in the zebrafish trunk and central arteries in the head. Analysis of downstream signaling pathways revealed no change in MAPK (ERK) activation, while inhibiting PI3 kinase signaling phenocopied vegfab mutant phenotypes in affected blood vessels. Together, these results suggest that extracellular matrix bound Vegfa might act through PI3K signaling to control EC proliferation in a distinct set of blood vessels during angiogenesis.


Subject(s)
Vascular Endothelial Growth Factor A , Zebrafish , Animals , Cell Proliferation , Neovascularization, Physiologic/genetics , Phenotype , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish/metabolism
7.
Bioessays ; 41(3): e1800198, 2019 03.
Article in English | MEDLINE | ID: mdl-30805984

ABSTRACT

A tree-like hierarchical branching structure is present in many biological systems, such as the kidney, lung, mammary gland, and blood vessels. Most of these organs form through branching morphogenesis, where outward growth results in smaller and smaller branches. However, the blood vasculature is unique in that it exists as two trees (arterial and venous) connected at their tips. Obtaining this organization might therefore require unique developmental mechanisms. As reviewed here, recent data indicate that arterial trees often form in reverse order. Accordingly, initial arterial endothelial cell differentiation occurs outside of arterial vessels. These pre-artery cells then build trees by following a migratory path from smaller into larger arteries, a process guided by the forces imparted by blood flow. Thus, in comparison to other branched organs, arteries can obtain their structure through inward growth and coalescence. Here, new information on the underlying mechanisms is discussed, and how defects can lead to pathologies, such as hypoplastic arteries and arteriovenous malformations.


Subject(s)
Arteries/embryology , Arteries/growth & development , Neovascularization, Physiologic , Veins/embryology , Veins/growth & development , Animals , Cell Differentiation/physiology , Cell Movement , Cell Plasticity , Epithelial Cells/physiology , Humans , Mice , Morphogenesis , Receptors, CXCR4/metabolism , Receptors, Notch/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
8.
9.
Curr Opin Hematol ; 25(3): 237-244, 2018 05.
Article in English | MEDLINE | ID: mdl-29438260

ABSTRACT

PURPOSE OF REVIEW: Mutations in the Endoglin (Eng) gene, an auxiliary receptor in the transforming growth factor beta (TGFß)-superfamily signaling pathway, are responsible for the human vascular disorder hereditary hemorrhagic telangiectasia (HHT) type 1, characterized in part by blood vessel enlargement. A growing body of work has uncovered an autonomous role for Eng in endothelial cells. We will highlight the influence of Eng on distinct cellular behaviors, such as migration and shape control, which are ultimately important for the assignment of proper blood vessel diameters. RECENT FINDINGS: How endothelial cells establish hierarchically ordered blood vessel trees is one of the outstanding questions in vascular biology. Mutations in components of the TGFß-superfamily of signaling molecules disrupt this patterning and cause arteriovenous malformations (AVMs). Eng is a TGFß coreceptor enhancing signaling through the type I receptor Alk1. Recent studies identified bone morphogenetic proteins (BMPs) 9 and 10 as the primary ligands for Alk1/Eng. Importantly, Eng potentiated Alk1 pathway activation downstream of hemodynamic forces. New results furthermore revealed how Eng affects endothelial cell migration and cell shape control in response to these forces, thereby providing new avenues for our understanding of AVM cause. SUMMARY: We will discuss the interplay of Eng and hemodynamic forces, such as shear stress, in relation to Alk1 receptor activation. We will furthermore detail how this signaling pathway influences endothelial cell behaviors important for the establishment of hierarchically ordered blood vessel trees. Finally, we will provide an outlook how these insights might help in developing new therapies for the treatment of HHT.


Subject(s)
Endoglin , Endothelial Cells , Hemodynamics , Mutation , Signal Transduction/genetics , Telangiectasia, Hereditary Hemorrhagic , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Movement/genetics , Cell Shape/genetics , Endoglin/genetics , Endoglin/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Growth Differentiation Factor 2 , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Humans , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism , Telangiectasia, Hereditary Hemorrhagic/pathology
10.
Development ; 141(7): 1544-52, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24598161

ABSTRACT

Differentiation of arteries and veins is essential for the development of a functional circulatory system. In vertebrate embryos, genetic manipulation of Notch signaling has demonstrated the importance of this pathway in driving artery endothelial cell differentiation. However, when and where Notch activation occurs to affect endothelial cell fate is less clear. Using transgenic zebrafish bearing a Notch-responsive reporter, we demonstrate that Notch is activated in endothelial progenitors during vasculogenesis prior to blood vessel morphogenesis and is maintained in arterial endothelial cells throughout larval stages. Furthermore, we find that endothelial progenitors in which Notch is activated are committed to a dorsal aorta fate. Interestingly, some arterial endothelial cells subsequently downregulate Notch signaling and then contribute to veins during vascular remodeling. Lineage analysis, together with perturbation of both Notch receptor and ligand function, further suggests several distinct developmental windows in which Notch signaling acts to promote artery commitment and maintenance. Together, these findings demonstrate that Notch acts in distinct contexts to initiate and maintain artery identity during embryogenesis.


Subject(s)
Arteries/embryology , Body Patterning/genetics , Receptors, Notch/physiology , Animals , Animals, Genetically Modified , Arteries/cytology , Cell Differentiation/genetics , Embryo, Nonmammalian , Endothelium, Vascular/embryology , Morphogenesis/genetics , Neovascularization, Physiologic/genetics , Signal Transduction/physiology , Veins/embryology , Zebrafish/embryology , Zebrafish/genetics
11.
Genes Dev ; 23(19): 2272-7, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19797767

ABSTRACT

The aorta traverses the body, yet little is known about how it is patterned in different anatomical locations. Here, we show that the aorta develops from genetically distinct endothelial cells originating from diverse locations within the embryo. Furthermore, chemokine (C-X-C motif) receptor 4a (cxcr4a) is restricted to endothelial cells derived from anterior mesoderm, and is required specifically for formation of the lateral aortae. Cxcl12b, a cxcr4a ligand, is expressed in endoderm underlying the lateral aortae, and loss of cxcl12b phenocopies cxcr4a deficiency. These studies reveal unexpected endothelial diversity within the aorta that is necessary to facilitate its regional patterning by local cues.


Subject(s)
Aorta/embryology , Body Patterning/physiology , Chemokines/physiology , Signal Transduction , Zebrafish/embryology , Animals , Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Receptors, CXCR4/deficiency , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
12.
Development ; 138(9): 1717-26, 2011 May.
Article in English | MEDLINE | ID: mdl-21429983

ABSTRACT

During angiogenic sprouting, newly forming blood vessels need to connect to the existing vasculature in order to establish a functional circulatory loop. Previous studies have implicated genetic pathways, such as VEGF and Notch signaling, in controlling angiogenesis. We show here that both pathways similarly act during vascularization of the zebrafish central nervous system. In addition, we find that chemokine signaling specifically controls arterial-venous network formation in the brain. Zebrafish mutants for the chemokine receptor cxcr4a or its ligand cxcl12b establish a decreased number of arterial-venous connections, leading to the formation of an unperfused and interconnected blood vessel network. We further find that expression of cxcr4a in newly forming brain capillaries is negatively regulated by blood flow. Accordingly, unperfused vessels continue to express cxcr4a, whereas connection of these vessels to the arterial circulation leads to rapid downregulation of cxcr4a expression and loss of angiogenic characteristics in endothelial cells, such as filopodia formation. Together, our findings indicate that hemodynamics, in addition to genetic pathways, influence vascular morphogenesis by regulating the expression of a proangiogenic factor that is necessary for the correct pathfinding of sprouting brain capillaries.


Subject(s)
Arteries/embryology , Brain/blood supply , Brain/embryology , Chemokines/metabolism , Hemodynamics/physiology , Veins/embryology , Animals , Animals, Genetically Modified , Arteries/metabolism , Body Patterning/genetics , Body Patterning/physiology , Chemokines/physiology , Chemokines, CXC/metabolism , Chemokines, CXC/physiology , Computer Simulation , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Hemodynamics/genetics , Models, Biological , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Veins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology
13.
Development ; 138(20): 4555-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21937602

ABSTRACT

Zinc-finger nucleases (ZFNs) allow targeted gene inactivation in a wide range of model organisms. However, construction of target-specific ZFNs is technically challenging. Here, we evaluate a straightforward modular assembly-based approach for ZFN construction and gene inactivation in zebrafish. From an archive of 27 different zinc-finger modules, we assembled more than 70 different zinc-finger cassettes and evaluated their specificity using a bacterial one-hybrid assay. In parallel, we constructed ZFNs from these cassettes and tested their ability to induce lesions in zebrafish embryos. We found that the majority of zinc-finger proteins assembled from these modules have favorable specificities and nearly one-third of modular ZFNs generated lesions at their targets in the zebrafish genome. To facilitate the application of ZFNs within the zebrafish community we constructed a public database of sites in the zebrafish genome that can be targeted using this archive. Importantly, we generated new germline mutations in eight different genes, confirming that this is a viable platform for heritable gene inactivation in vertebrates. Characterization of one of these mutants, gata2a, revealed an unexpected role for this transcription factor in vascular development. This work provides a resource to allow targeted germline gene inactivation in zebrafish and highlights the benefit of a definitive reverse genetic strategy to reveal gene function.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zinc Fingers/genetics , Animals , Animals, Genetically Modified , Base Sequence , DNA/genetics , DNA/metabolism , Databases, Genetic , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Targeting , Mutation , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Zebrafish/embryology
14.
Nature ; 445(7129): 781-4, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17259972

ABSTRACT

Recent evidence indicates that growing blood-vessel sprouts consist of endothelial cells with distinct cell fates and behaviours; however, it is not clear what signals determine these sprout cell characteristics. Here we show that Notch signalling is necessary to restrict angiogenic cell behaviour to tip cells in developing segmental arteries in the zebrafish embryo. In the absence of the Notch signalling component Rbpsuh (recombining binding protein suppressor of hairless) we observed excessive sprouting of segmental arteries, whereas Notch activation suppresses angiogenesis. Through mosaic analysis we find that cells lacking Rbpsuh preferentially localize to the terminal position in developing sprouts. In contrast, cells in which Notch signalling has been activated are excluded from the tip-cell position. In vivo time-lapse analysis reveals that endothelial tip cells undergo a stereotypical pattern of proliferation and migration during sprouting. In the absence of Notch, nearly all sprouting endothelial cells exhibit tip-cell behaviour, leading to excessive numbers of cells within segmental arteries. Furthermore, we find that flt4 (fms-related tyrosine kinase 4, also called vegfr3) is expressed in segmental artery tip cells and becomes ectopically expressed throughout the sprout in the absence of Notch. Loss of flt4 can partially restore normal endothelial cell number in Rbpsuh-deficient segmental arteries. Finally, loss of the Notch ligand dll4 (delta-like 4) also leads to an increased number of endothelial cells within segmental arteries. Together, these studies indicate that proper specification of cell identity, position and behaviour in a developing blood-vessel sprout is required for normal angiogenesis, and implicate the Notch signalling pathway in this process.


Subject(s)
Arteries/cytology , Arteries/embryology , Neovascularization, Physiologic/physiology , Receptors, Notch/metabolism , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism , Animals , Cell Lineage , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Cell Mol Life Sci ; 68(17): 2811-30, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21479594

ABSTRACT

Chemokines are a vertebrate-specific group of small molecules that regulate cell migration and behaviour in diverse contexts. So far, around 50 chemokines have been identified in humans, which bind to 18 different chemokine receptors. These are members of the seven-transmembrane receptor family. Initially, chemokines were identified as modulators of the immune response. Subsequently, they were also shown to regulate cell migration during embryonic development. Here, we discuss the influence of chemokines and their receptors on angiogenesis, or the formation of new blood vessels. We highlight recent advances in our understanding of how chemokine signalling might directly influence endothelial cell migration. We furthermore examine the contributions of chemokine signalling in immune cells during this process. Finally, we explore possible implications for disease settings, such as chronic inflammation and tumour progression.


Subject(s)
Chemokines/physiology , Neovascularization, Pathologic , Neovascularization, Physiologic , Receptors, Chemokine/physiology , Chemokines/metabolism , Embryo, Mammalian/blood supply , Embryo, Mammalian/metabolism , Embryonic Development , Humans , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Chemokine/metabolism , Signal Transduction
16.
Sci Rep ; 12(1): 4795, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35314737

ABSTRACT

Endothelial cells (ECs) lining blood vessels are exposed to mechanical forces, such as shear stress. These forces control many aspects of EC biology, including vascular tone, cell migration and proliferation. Despite a good understanding of the genes responding to shear stress, our insight into the transcriptional regulation of these genes is much more limited. Here, we set out to study alterations in the chromatin landscape of human umbilical vein endothelial cells (HUVEC) exposed to laminar shear stress. To do so, we performed ChIP-Seq for H3K27 acetylation, indicative of active enhancer elements and ATAC-Seq to mark regions of open chromatin in addition to RNA-Seq on HUVEC exposed to 6 h of laminar shear stress. Our results show a correlation of gained and lost enhancers with up and downregulated genes, respectively. DNA motif analysis revealed an over-representation of KLF transcription factor (TF) binding sites in gained enhancers, while lost enhancers contained more ETV/ETS motifs. We validated a subset of flow responsive enhancers using luciferase-based reporter constructs and CRISPR-Cas9 mediated genome editing. Lastly, we characterized the shear stress response in ECs of zebrafish embryos using RNA-Seq. Our results lay the groundwork for the exploration of shear stress responsive elements in controlling EC biology.


Subject(s)
Chromatin , Zebrafish , Animals , Binding Sites , Cells, Cultured , Chromatin/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Stress, Mechanical , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish/metabolism
17.
Dev Dyn ; 239(3): 855-64, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20108354

ABSTRACT

The Notch signaling pathway regulates specification of zebrafish liver progenitor cells towards a biliary cell fate. Here, using staged administration of a pharmacological inhibitor of Notch receptor processing, we show that activation of the Notch pathway is also important for growth and expansion of the intrahepatic biliary network in zebrafish larvae. Biliary expansion is accompanied by extensive cell proliferation and active remodeling of the nascent ductal network, as revealed by time lapse imaging of living zebrafish larvae that express a Notch responsive fluorescent reporter transgene. Together, these data support a model in which the Notch signal functions reiteratively during biliary development; first to specific biliary cells and then to direct remodeling of the nascent biliary network. As the Notch pathway plays a comparable role during mammalian biliary development, including humans, these studies also indicate broad conservation of the molecular mechanisms directing biliary development in vertebrates.


Subject(s)
Bile Ducts/embryology , Gene Expression Regulation, Developmental , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Lineage , Cell Proliferation , Fluorescent Dyes/chemistry , Immunohistochemistry/methods , Liver/embryology , Microscopy, Fluorescence/methods , Models, Biological , Time Factors , Transgenes , Zebrafish
18.
Bioessays ; 30(4): 303-13, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18348190

ABSTRACT

The formation of blood vessels within the vascular system entails a variety of cellular processes, including proliferation, migration and differentiation. In many cases, these diverse processes need to be finely coordinated among neighbouring endothelial cells in order to establish a functional vascular network. For instance, during angiogenic sprouting specialized endothelial tip cells follow guidance cues and migrate extensively into avascular tissues while trailing stalk cells must stay connected to the patent blood vessel. The vascular endothelial growth factor (VEGF) and Notch signalling pathways have emerged as the major players in governing these different cellular behaviours. In particular, recent work indicates an important role for Notch signalling in determining how an endothelial cell responds to VEGF. In this review, we provide an overview of these biochemically distinct pathways and discuss how they may interact during endothelial cell differentiation and angiogenesis.


Subject(s)
Cell Physiological Phenomena , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Lineage , Endothelial Cells/cytology , Humans , Ligands , Models, Biological , Mutation , Neovascularization, Pathologic , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
20.
Cardiovasc Res ; 115(10): 1487-1499, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-30785199

ABSTRACT

AIMS: Oscillatory shear stress (OSS) is an atheroprone haemodynamic force that occurs in areas of vessel irregularities and is implicated in the pathogenesis of atherosclerosis. Changes in signalling and transcriptional programme in response to OSS have been vigorously studied; however, the underlying changes in the chromatin landscape controlling transcription remain to be elucidated. Here, we investigated the changes in the regulatory element (RE) landscape of endothelial cells under atheroprone OSS conditions in an in vitro model. METHODS AND RESULTS: Analyses of H3K27ac chromatin immunoprecipitation-Seq enrichment and RNA-Seq in primary human umbilical vein endothelial cells 6 h after onset of OSS identified 2806 differential responsive REs and 33 differentially expressed genes compared with control cells kept under static conditions. Furthermore, gene ontology analyses of putative RE-associated genes uncovered enrichment of WNT/HIPPO pathway and cytoskeleton reorganization signatures. Transcription factor (TF) binding motif analysis within RE sequences identified over-representation of ETS, Zinc finger, and activator protein 1 TF families that regulate cell cycle, proliferation, and apoptosis, implicating them in the development of atherosclerosis. Importantly, we confirmed the activation of EGR1 as well as the YAP/TAZ complex early (6 h) after onset of OSS in both cultured human vein and artery endothelial cells and, by undertaking luciferase assays, functionally verified their role in RE activation in response to OSS. CONCLUSIONS: Based on the identification and verification of specific responsive REs early upon OSS exposure, we propose an expanded mechanism of how OSS might contribute to the development of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Mechanotransduction, Cellular , Response Elements , Transcription Factors/metabolism , Umbilical Arteries/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Gene Expression Regulation , Gene Regulatory Networks , Human Umbilical Vein Endothelial Cells/pathology , Humans , Plaque, Atherosclerotic , Protein Interaction Maps , Regional Blood Flow , Stress, Mechanical , Transcription Factors/genetics , Umbilical Arteries/pathology , Umbilical Arteries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL