Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Vis Exp ; (88): e51666, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24998400

ABSTRACT

Primary rat neonatal cardiomyocytes are useful in basic in vitro cardiovascular research because they can be easily isolated in large numbers in a single procedure. Due to advances in microscope technology it is relatively easy to capture live cell images for the purpose of investigating cellular events in real time with minimal concern regarding phototoxicity to the cells. This protocol describes how to take live cell timelapse images of primary rat neonatal cardiomyocytes using a confocal spinning disk microscope following lentiviral and adenoviral transduction to modulate properties of the cell. The application of two different types of viruses makes it easier to achieve an appropriate transduction rate and expression levels for two different genes. Well focused live cell images can be obtained using the microscope's autofocus system, which maintains stable focus for long time periods. Applying this method, the functions of exogenously engineered proteins expressed in cultured primary cells can be analyzed. Additionally, this system can be used to examine the functions of genes through the use of siRNAs as well as of chemical modulators.


Subject(s)
Adenoviridae/genetics , Lentivirus/genetics , Microscopy, Confocal/methods , Myocytes, Cardiac/physiology , Transduction, Genetic/methods , Animals , Image Processing, Computer-Assisted/methods , Myocytes, Cardiac/cytology , Rats
2.
PLoS One ; 9(6): e90736, 2014.
Article in English | MEDLINE | ID: mdl-24603875

ABSTRACT

Cell-cell communications typically involve receptor-mediated signaling initiated by soluble or cell-bound ligands. Here, we report a unique mode of endocytosis: proteins originating from cell-cell junctions and cytosolic cellular components from the neighboring cell are internalized, leading to direct exchange of cellular components between two adjacent endothelial cells. VE-cadherins form transcellular bridges between two endothelial cells that are the basis of adherence junctions. At such adherens junction sites, we observed the movement of the entire VE-cadherin molecule from one endothelial cell into the other with junctional and cytoplasmic components. This phenomenon, here termed trans-endocytosis, requires the establishment of a VE-cadherin homodimer in trans with internalization proceeding in a Rac1-, and actomyosin-dependent manner. Importantly, the trans-endocytosis is not dependent on any known endocytic pathway including clathrin-dependent endocytosis, macropinocytosis or phagocytosis. This novel form of cell-cell communications, leading to a direct exchange of cellular components, was observed in 2D and 3D-cultured endothelial cells as well as in the developing zebrafish vasculature.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endocytosis , Human Umbilical Vein Endothelial Cells/metabolism , Actins/metabolism , Animals , COS Cells , Cell Communication , Chlorocebus aethiops , Coculture Techniques , Humans , Myosins/metabolism , Protein Transport , Vinculin/metabolism , Zebrafish , rac1 GTP-Binding Protein/metabolism
3.
J Clin Invest ; 123(12): 5082-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24177422

ABSTRACT

Aberrant blood vessel formation contributes to a wide variety of pathologies, and factors that regulate angiogenesis are attractive therapeutic targets. Endothelial and smooth muscle cell-derived neuropilin-like protein (ESDN) is a neuropilin-related transmembrane protein expressed in ECs; however, its potential effect on VEGF responses remains undefined. Here, we generated global and EC-specific Esdn knockout mice and demonstrated that ESDN promotes VEGF-induced human and murine EC proliferation and migration. Deletion of Esdn in the mouse interfered with adult and developmental angiogenesis, and knockdown of the Esdn homolog (dcbld2) in zebrafish impaired normal vascular development. Loss of ESDN in ECs blunted VEGF responses in vivo and attenuated VEGF-induced VEGFR-2 signaling without altering VEGF receptor or neuropilin expression. Finally, we found that ESDN associates with VEGFR-2 and regulates its complex formation with negative regulators of VEGF signaling, protein tyrosine phosphatases PTP1B and TC-PTP, and VE-cadherin. These findings establish ESDN as a regulator of VEGF responses in ECs that acts through a mechanism distinct from neuropilins. As such, ESDN may serve as a therapeutic target for angiogenesis regulation.


Subject(s)
Endothelium, Vascular/physiology , Membrane Proteins/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Antigens, CD/physiology , Blood Vessels/embryology , Cadherins/physiology , Cells, Cultured , Ear, External/blood supply , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/physiopathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropilins/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , RNA Interference , RNA, Small Interfering/pharmacology , Retinal Vessels/growth & development , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/physiology
4.
Mol Cells ; 35(2): 166-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23456338

ABSTRACT

Previous studies have identified two zebrafish mutants, cloche and groom of cloche, which lack the majority of the endothelial lineage at early developmental stages. However, at later stages, these avascular mutant embryos generate rudimentary vessels, indicating that they retain the ability to generate endothelial cells despite this initial lack of endothelial progenitors. To further investigate molecular mechanisms that allow the emergence of the endothelial lineage in these avascular mutant embryos, we analyzed the gene expression profile using microarray analysis on isolated endothelial cells. We find that the expression of the genes characteristic of the mesodermal lineages are substantially elevated in the kdrl (+) cells isolated from avascular mutant embryos. Subsequent validation and analyses of the microarray data identifies Sox11b, a zebrafish ortholog of SRY-related HMG box 11 (SOX11), which have not previously implicated in vascular development. We further define the function sox11b during vascular development, and find that Sox11b function is essential for developmental angiogenesis in zebrafish embryos, specifically regulating sprouting angiogenesis. Taken together, our analyses illustrate a complex regulation of endothelial specification and differentiation during vertebrate development.


Subject(s)
Endothelium, Vascular/cytology , SOX Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Bone Morphogenetic Proteins/metabolism , Embryo, Nonmammalian/metabolism , Endothelium, Vascular/growth & development , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Microarray Analysis , Mutation , SOX Transcription Factors/metabolism , Signal Transduction , Zebrafish/metabolism , Zebrafish Proteins/metabolism
5.
Biochem Biophys Res Commun ; 430(4): 1212-6, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23266606

ABSTRACT

Segregation of two axial vessels, the dorsal aorta and caudal vein, is one of the earliest patterning events occur during development of vasculature. Despite the importance of this process and recent advances in our understanding on vascular patterning during development, molecular mechanisms that coordinate the segregation of axial vessels remain largely elusive. In this report, we find that vascular endothelial growth factor-A (Vegf-A) signaling regulates the segregation of dorsal aorta and axial vein during development. Inhibition of Vegf-A pathway components including ligand Vegf-A and its cognate receptor Kdrl, caused failure in segregation of axial vessels in zebrafish embryos. Similarly, chemical inhibition of Mitogen-activated protein kinase kinase (Map2k1)/Extracellular-signal-regulated kinases (Erk) and phosphatidylinositol 3-kinases (PI3K), which are downstream effectors of Vegf-A signaling pathway, led to the fusion of two axial vessels. Moreover, we find that restoring Erk activity by over-expression of constitutively active MEK in embryos with a reduced level of Vegf-A signaling can rescue the defects in axial vessel segregation. Taken together, our data show that segregation of axial vessels requires the function of Vegf-A signaling, and Erk may function as the major downstream effector in this process.


Subject(s)
Aorta/growth & development , Extracellular Signal-Regulated MAP Kinases/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/physiology , Veins/growth & development , Animals , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Zebrafish/growth & development
6.
Mol Cell ; 29(2): 191-206, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18243114

ABSTRACT

Sec14, the major yeast phosphatidylinositol (PtdIns)/phosphatidylcholine (PtdCho) transfer protein, regulates essential interfaces between lipid metabolism and membrane trafficking from the trans-Golgi network (TGN). How Sec14 does so remains unclear. We report that Sec14 binds PtdIns and PtdCho at distinct (but overlapping) sites, and both PtdIns- and PtdCho-binding activities are essential Sec14 activities. We further show both activities must reside within the same molecule to reconstitute a functional Sec14 and for effective Sec14-mediated regulation of phosphoinositide homeostasis in vivo. This regulation is uncoupled from PtdIns-transfer activity and argues for an interfacial presentation mode for Sec14-mediated potentiation of PtdIns kinases. Such a regulatory role for Sec14 is a primary counter to action of the Kes1 sterol-binding protein that antagonizes PtdIns 4-OH kinase activity in vivo. Collectively, these findings outline functional mechanisms for the Sec14 superfamily and reveal additional layers of complexity for regulating phosphoinositide homeostasis in eukaryotes.


Subject(s)
Cell Membrane/metabolism , Homeostasis/physiology , Lipid Metabolism/physiology , Phosphatidylinositols/metabolism , Phospholipid Transfer Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , 1-Phosphatidylinositol 4-Kinase/chemistry , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , Biological Transport/physiology , Cell Membrane/chemistry , Cell Membrane/genetics , Golgi Apparatus/chemistry , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Phosphatidylcholines/chemistry , Phosphatidylcholines/genetics , Phosphatidylcholines/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/genetics , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Protein Structure, Tertiary , Receptors, Steroid , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...