Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nano Lett ; 11(11): 4932-8, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-21967244

ABSTRACT

The exact mechanism of angiogenesis by europium hydroxide nanorods was unclear. In this study we have showed that formation of reactive oxygen species (H(2)O(2) and O(2)·-) is involved in redox signaling pathways during angiogenesis, important for cardiovascular and ischemic diseases. Here we used single-walled carbon nanotube sensor array to measure the single-molecule efflux of H(2)O(2) and a HPLC method for the determination of O(2)·- from endothelial cells in response to proangiogenic factors. Additionally, reactive oxygen species-mediated angiogenesis using inorganic nanorods was observed in transgenic (fli1a:EGFP) zebrafish embryos.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/physiology , Inorganic Chemicals/pharmacology , Nanoparticles/administration & dosage , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Reactive Oxygen Species/metabolism , Cells, Cultured , Humans
2.
J Biol Chem ; 286(35): 30740-30747, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21730073

ABSTRACT

Despite their importance as members of the Roundabout (Robo) family in the control of axonal and vascular patterning, the transcriptional regulation of these genes is poorly understood. In this study, we show that members of the Sry-related high mobility box (Sox) transcription factor family as being transcriptional regulators of roundabout4 (robo4), a Robo gene family member that participates in sprouting angiogenesis in vivo, in zebrafish. Double whole mount in situ hybridization analysis in zebrafish embryos revealed co-localization of the vascular relevant Sox factors sox7 or sox18 mRNA with robo4 transcripts in developing intersomitic vessels. A 3-kb human ROBO4 promoter element was able to drive reporter expression in zebrafish to recapitulate the endogenous temporal intersomitic vessel expression pattern of robo4. EMSA analysis confirmed binding of Sox18 to a canonical Sox binding site (from -1170 bp to -1176 bp) in the ROBO4 promoter (3 kb), and mutation analysis indicated that this site was partially responsible for ROBO4 promoter activity in ECs. A combination of gain- and loss-of-function analysis identified Sox7 and Sox18 co-regulation of robo4 but not fli1a transcripts in zebrafish. Finally, Sox-mediated robo4 transcriptional regulation is conserved across evolution. These studies imply Sox-mediated transcriptional regulation of Robo4 in the developing embryonic vasculature.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Cell Surface/biosynthesis , Zebrafish Proteins/biosynthesis , Animals , Cell Movement , DNA Mutational Analysis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Mice , Mutation , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Receptors, Cell Surface/physiology , SOXF Transcription Factors/metabolism , Transcription, Genetic , Zebrafish , Zebrafish Proteins/physiology
3.
PLoS One ; 6(2): e14732, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21364913

ABSTRACT

BACKGROUND: Vasculogenesis, the de novo formation of blood vessels from precursor cells is critical for a developing embryo. However, the signals and events that dictate the formation of primary axial vessels remain poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we use ets-related protein-1 (etsrp), which is essential for vascular development, to analyze the early stages of vasculogenesis in zebrafish. We found etsrp(+) cells of the head, trunk and tail follow distinct developmental sequences. Using a combination of genetic, molecular and chemical approaches, we demonstrate that fli(+)etsrp(+) hemato-vascular progenitors (FEVPs) are proliferating at the lateral plate mesoderm (LPM). The Shh-VEGF-Notch-Hey2 signaling pathway controls the proliferation process, and experimental modulation of single components of this pathway alters etsrp(+) cell numbers at the LPM. CONCLUSIONS/SIGNIFICANCE: This study for the first time defines factors controlling proliferation, and cell numbers of pre-migratory FEVPs in zebrafish.


Subject(s)
Blood Vessels/embryology , Cell Proliferation , Hematopoietic Stem Cells/physiology , Mesoderm/cytology , Proto-Oncogene Protein c-fli-1/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Blood Vessels/metabolism , Cell Movement/physiology , Embryo, Nonmammalian , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Gene Expression Regulation, Developmental , Head/embryology , Hematopoietic Stem Cells/metabolism , Mesoderm/metabolism , Mesoderm/physiology , Models, Biological , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/physiology , Tail/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
4.
Blood ; 116(24): 5423-33, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20813898

ABSTRACT

Our previous work has shown that axon guidance gene family Nogo-B and its receptor (NgBR) are essential for chemotaxis and morphogenesis of endothelial cells in vitro. To investigate NogoB-NgBR function in vivo, we cloned the zebrafish ortholog of both genes and studied loss of function in vivo using morpholino antisense technology. Zebrafish ortholog of Nogo-B is expressed in somite while expression of zebrafish NgBR is localized in intersomitic vessel (ISV) and axial dorsal aorta during embryonic development. NgBR or Nogo-B knockdown embryos show defects in ISV sprouting in the zebrafish trunk. Mechanistically, we found that NgBR knockdown not only abolished its ligand Nogo-B-stimulated endothelial cell migration but also reduced the vascular endothelial growth factor (VEGF)-stimulated phosphorylation of Akt and vascular endothelial growth factor-induced chemotaxis and morphogenesis of human umbilical vein endothelial cells. Further, constitutively activated Akt (myristoylated [myr]Akt) or human NgBR can rescue the NgBR knockdown umbilical vein endothelial cell migration defects in vitro or NgBR morpholino-caused ISV defects in vivo. These data place Akt at the downstream of NgBR in both Nogo-B- and VEGF-coordinated sprouting of ISVs. In summary, this study identifies the in vivo functional role for Nogo-B and its receptor (NgBR) in angiogenesis in zebrafish.


Subject(s)
Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/physiology , Zebrafish Proteins/physiology , Animals , Chemotaxis , Embryonic Development , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Humans , Phosphorylation , RNA, Antisense/pharmacology , Receptors, Cell Surface/genetics , Vascular Endothelial Growth Factor A/physiology , Zebrafish , Zebrafish Proteins/genetics
5.
Blood ; 115(22): 4614-22, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20086248

ABSTRACT

Endothelial cell-specific chemotaxis receptor (ECSCR) is a cell surface protein expressed by blood endothelial cells with roles in endothelial cell migration and signal transduction. We investigated the function of ecscr in the development of the zebrafish vasculature. Zebrafish ecscr is expressed in angioblasts and in axial vessels during angioblast migration and vasculogenesis. Morpholino-directed ecscr knockdown resulted in defective angioblast migration in the posterior lateral plate mesoderm, a process known to depend on vascular endothelial-derived growth factor (VEGF). In cultured cells, transfected ECSCR localized to actin-rich membrane protrusions, colocalizing with kinase insert domain protein receptor (KDR)/VEGF receptor 2 in these regions. ECSCR-silenced cells show reduced VEGF-induced phosphorylation of KDR but not of FMS-like tyrosine kinase 1 (FLT1)/VEGF receptor 1. Finally, chemical inhibition of VEGF receptor activity in zebrafish resulted in angioblast deficiencies that partially overlap with those seen in ecscr morphants. We propose that ecscr promotes migration of zebrafish angioblasts by enhancing endothelial kdr sensitivity to VEGF.


Subject(s)
Blood Vessels/embryology , Blood Vessels/metabolism , Chemotaxis/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins , Base Sequence , Cell Movement , Cell Proliferation , Cells, Cultured , DNA Primers/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Blood ; 115(1): 133-9, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19880500

ABSTRACT

Recently, messenger RNAs in eukaryotes have shown to associate with antisense (AS) transcript partners that are often referred to as long noncoding RNAs (lncRNAs) whose function is largely unknown. Here, we have identified a natural AS transcript for tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-1 (tie-1), tie-1AS lncRNA in zebrafish, mouse, and humans. In embryonic zebrafish, tie-1AS lncRNA transcript is expressed temporally and spatially in vivo with its native target, the tie-1 coding transcript and in additional locations (ear and brain). The tie-1AS lncRNA selectively binds tie-1 mRNA in vivo and regulates tie-1 transcript levels, resulting in specific defects in endothelial cell contact junctions in vivo and in vitro. The ratio of tie-1 versus tie-1AS lncRNA is altered in human vascular anomaly samples. These results directly implicate noncoding RNA-mediated transcriptional regulation of gene expression as a fundamental control mechanism for physiologic processes, such as vascular development.


Subject(s)
Genetic Loci/genetics , RNA, Antisense/genetics , RNA, Untranslated/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Intercellular Junctions/pathology , Mice , Neovascularization, Physiologic/drug effects , Phenotype , RNA, Antisense/metabolism , RNA, Untranslated/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Species Specificity , Vascular Diseases/genetics , Vascular Diseases/pathology , Vascular Endothelial Growth Factor A/pharmacology , Zebrafish Proteins/genetics
7.
Transgenic Res ; 18(4): 595-605, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19266305

ABSTRACT

The frog Xenopus is a well established vertebrate model to study the processes involved in embryogenesis and organogenesis, as it can be manipulated easily with a whole series of methods. We have expanded these approaches by establishing two transgenic Xenopus strains that allow specific interference with the activity of defined genes using a heat-shock inducible Cre recombinase that can induce upon heat-shock expression of a reporter gene in crossings to a corresponding reporter strain. We have applied this binary technique of gene interference in Xenopus development to overexpress the mutated HNF1 beta transcription factor at distinct developmental stages. Induction of HNF1 beta P328L329del by heat-shock at the gastrula stage resulted in a dramatic phenotype including malformation of the pronephros, gut, stomach, abnormal tail development and massive edemas indicative for kidney dysfunction. Thus, we have established the first binary inducible gene expression system in Xenopus laevis that can be used to study organogenesis.


Subject(s)
Heat-Shock Response , Integrases/biosynthesis , Organogenesis , Xenopus laevis/growth & development , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Gastrula/growth & development , Gene Expression , Genes, Reporter , HSC70 Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Organogenesis/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics
8.
Blood ; 113(5): 1184-91, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-18927432

ABSTRACT

Mitogen-activated protein kinases play an integral role in several cellular processes. To regulate mitogen-activated protein kinases, cells express members of a counteracting group of proteins called phosphatases. In this study, we have identified a specific role that one member of this family of phosphatases, dual-specific phosphatase-5 (Dusp-5) plays in vascular development in vivo. We have determined that dusp-5 is expressed in angioblasts and in established vasculature and that it counteracts the function of a serine threonine kinase, Snrk-1, which also plays a functional role in angioblast development. Together, Dusp-5 and Snrk-1 control angioblast populations in the lateral plate mesoderm with Dusp-5 functioning downstream of Snrk-1. Importantly, mutations in dusp-5 and snrk-1 have been identified in affected tissues of patients with vascular anomalies, implicating the Snrk-1-Dusp-5 signaling pathway in human disease.


Subject(s)
Blood Vessels/embryology , Dual-Specificity Phosphatases/biosynthesis , Gene Expression Regulation , Hemangioma/enzymology , Mesoderm/blood supply , Neoplasm Proteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Blood Vessels/pathology , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation/genetics , Hemangioma/genetics , Hemangioma/pathology , Humans , Mesoderm/embryology , Mesoderm/pathology , Mutation , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Zebrafish Proteins/genetics
9.
Blood ; 113(5): 1192-9, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-18723694

ABSTRACT

In vertebrates, molecular mechanisms dictate angioblasts' migration and subsequent differentiation into arteries and veins. In this study, we used a microarray screen to identify a novel member of the sucrose nonfermenting related kinase (snrk-1) family of serine/threonine kinases expressed specifically in the embryonic zebrafish vasculature and investigated its function in vivo. Using gain- and loss-of-function studies in vivo, we show that Snrk-1 plays an essential role in the migration, maintenance, and differentiation of angioblasts. The kinase function of Snrk-1 is critical for migration and maintenance, but not for the differentiation of angioblasts. In vitro, snrk-1 knockdown endothelial cells show only defects in migration. The snrk-1 gene acts downstream or parallel to notch and upstream of gridlock during artery-vein specification, and the human gene compensates for zebrafish snrk-1 knockdown, suggesting evolutionary conservation of function.


Subject(s)
Arteries/embryology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Protein Serine-Threonine Kinases/biosynthesis , Veins/embryology , Zebrafish/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement/physiology , Endothelial Cells/enzymology , Gene Knockdown Techniques , Humans , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
J Biomol Screen ; 13(10): 960-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19029015

ABSTRACT

Regulation of whole-body metabolism and energy homeostasis has been shown to require signaling between multiple organs. To identify genetic programs that determine metabolic rate, and compounds that can modify it, a whole-animal assay amenable to large-scale screening was developed. The direct correlation of acid production with metabolic rate was exploited to use a noninvasive colorimetric assay for acid secretion by individual zebrafish larvae in a 96-well plate format. A 3-fold increase in metabolic rate was detected that accompanied development between 24 and 96 h postfertilization. Dynamic changes in metabolic rate were also detected in response to different conditions such as temperature and drug treatments, in general agreement with the rate of oxygen consumption measured concomitantly. This assay was used to measure metabolic rate in the progeny of fish known to carry a recessive mutation in a gene required for ribosome biogenesis ( npo(fW07-g)), which would be expected to reduce energy consumption. A strong correlation was found (p < 10(-6) ) between reduced metabolic rate and genotype even before the developmental defect was visually evident. These studies support the conclusion that whole-animal acid secretion can be used as a readout for energy metabolism, thus enabling large-scale screening for genetic and chemical regulators of metabolic rate in a vertebrate.


Subject(s)
Biological Assay/methods , Energy Metabolism , Zebrafish/metabolism , 2,4-Dinitrophenol/pharmacology , Acids , Aging/drug effects , Animals , Energy Metabolism/drug effects , Energy Metabolism/genetics , Genotype , Larva/drug effects , Larva/metabolism , Phenotype , Sirolimus/pharmacology , Temperature
11.
BMC Cell Biol ; 9: 61, 2008 Nov 03.
Article in English | MEDLINE | ID: mdl-18980679

ABSTRACT

BACKGROUND: Roundabouts are axon guidance molecules that have recently been identified to play a role in vascular guidance as well. In this study, we have investigated gene knockdown analysis of endothelial Robos, in particular roundabout 4 (robo4), the predominant Robo in endothelial cells using small interfering RNA technology in vitro. RESULTS: Robo1 and Robo4 knockdown cells display distinct activity in endothelial cell migration assay. The knockdown of robo4 abrogated the chemotactic response of endothelial cells to serum but enhanced a chemokinetic response to Slit2, while robo1 knockdown cells do not display chemotactic response to serum or VEGF. Robo4 knockdown endothelial cells unexpectedly show up regulation of Rho GTPases. Zebrafish Robo4 rescues both Rho GTPase homeostasis and serum reduced chemotaxis in robo4 knockdown cells. Robo1 and Robo4 interact and share molecules such as Slit2, Mena and Vilse, a Cdc42-GAP. In addition, this study mechanistically implicates IRSp53 in the signaling nexus between activated Cdc42 and Mena, both of which have previously been shown to be involved with Robo4 signaling in endothelial cells. CONCLUSION: This study identifies specific components of the Robo signaling apparatus that work together to guide directional migration of endothelial cells.


Subject(s)
Endothelial Cells/metabolism , RNA, Small Interfering/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Cell Movement , Cells, Cultured , Chemotaxis , Endothelial Cells/cytology , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Serum/metabolism , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , Roundabout Proteins
12.
Leuk Res ; 32(5): 781-9, 2008 May.
Article in English | MEDLINE | ID: mdl-17980910

ABSTRACT

While critical steps in the regulation of leukemia cell development have been intensively studied in recent years, less is known about the interactions of leukemic cells with their stroma. Previously, we have shown that human acute myeloid leukemia (AML) cells differentiate upon injection into murine blastocysts. We here describe that human AML Kasumi-1 cells, cocultured with murine aorta-gonad-mesonephros (AGM) region-derived DAS104-4 stromal cells, decrease proliferation and colony formation efficiency; and up-regulate myelo-monocytic cell surface markers. Gene expression analysis showed decreased transcription of the AML1-ETO fusion gene and increased transcription of p16 (INK4A), p21 (WAF1) and C/EBPalpha genes. Coculture can induce myeloid differentiation also in patient-derived AML cells. Our findings strengthen the notion that the embryonic milieu can regulate the proliferation and differentiation of leukemic cells.


Subject(s)
Aorta/embryology , Gonads/embryology , Leukemia, Myeloid, Acute/pathology , Mesonephros/embryology , Stromal Cells/physiology , Aorta/cytology , CD11b Antigen/analysis , Cell Differentiation , Cell Line , Cell Proliferation , Coculture Techniques , Core Binding Factor Alpha 2 Subunit/genetics , Gonads/cytology , Humans , Lipopolysaccharide Receptors/analysis , Mesonephros/cytology , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...