Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 10: 1003028, 2022.
Article in English | MEDLINE | ID: mdl-36425528

ABSTRACT

Rationale and Goal: Endothelial cells (ECs) are quiescent and critical for maintaining homeostatic functions of the mature vascular system, while disruption of quiescence is at the heart of endothelial to mesenchymal transition (EndMT) and tumor angiogenesis. Here, we addressed the hypothesis that KLF4 maintains the EC quiescence. Methods and Results: In ECs, KLF4 bound to KLF2, and the KLF4-transctivation domain (TAD) interacted directly with KLF2. KLF4-depletion increased KLF2 expression, accompanied by phosphorylation of SMAD3, increased expression of alpha-smooth muscle actin (αSMA), VCAM-1, TGF-ß1, and ACE2, but decreased VE-cadherin expression. In the absence of Klf4, Klf2 bound to the Klf2-promoter/enhancer region and autoregulated its own expression. Loss of EC-Klf4 in Rosa mT/mG ::Klf4 fl/fl ::Cdh5 CreERT2 engineered mice, increased Klf2 levels and these cells underwent EndMT. Importantly, these mice harboring EndMT was also accompanied by lung inflammation, disruption of lung alveolar architecture, and pulmonary fibrosis. Conclusion: In quiescent ECs, KLF2 and KLF4 partnered to regulate a combinatorial mechanism. The loss of KLF4 disrupted this combinatorial mechanism, thereby upregulating KLF2 as an adaptive response. However, increased KLF2 expression overdrives for the loss of KLF4, giving rise to an EndMT phenotype.

2.
Front Immunol ; 12: 697588, 2021.
Article in English | MEDLINE | ID: mdl-34305934

ABSTRACT

The Toll-interleukin-1 Receptor (TIR) domain-containing adaptor protein (TIRAP) represents a key intracellular signalling molecule regulating diverse immune responses. Its capacity to function as an adaptor molecule has been widely investigated in relation to Toll-like Receptor (TLR)-mediated innate immune signalling. Since the discovery of TIRAP in 2001, initial studies were mainly focused on its role as an adaptor protein that couples Myeloid differentiation factor 88 (MyD88) with TLRs, to activate MyD88-dependent TLRs signalling. Subsequent studies delineated TIRAP's role as a transducer of signalling events through its interaction with non-TLR signalling mediators. Indeed, the ability of TIRAP to interact with an array of intracellular signalling mediators suggests its central role in various immune responses. Therefore, continued studies that elucidate the molecular basis of various TIRAP-protein interactions and how they affect the signalling magnitude, should provide key information on the inflammatory disease mechanisms. This review summarizes the TIRAP recruitment to activated receptors and discusses the mechanism of interactions in relation to the signalling that precede acute and chronic inflammatory diseases. Furthermore, we highlighted the significance of TIRAP-TIR domain containing binding sites for several intracellular inflammatory signalling molecules. Collectively, we discuss the importance of the TIR domain in TIRAP as a key interface involved in protein interactions which could hence serve as a therapeutic target to dampen the extent of acute and chronic inflammatory conditions.


Subject(s)
Inflammation/immunology , Membrane Glycoproteins/immunology , Receptors, Interleukin-1/immunology , Agammaglobulinaemia Tyrosine Kinase/immunology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Carrier Proteins/immunology , Carrier Proteins/metabolism , Class Ia Phosphatidylinositol 3-Kinase/immunology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Humans , Immunity, Innate , Inflammation/metabolism , Membrane Glycoproteins/metabolism , Models, Biological , Protein Interaction Maps , Protein Kinase C-delta/immunology , Protein Kinase C-delta/metabolism , Receptor for Advanced Glycation End Products/immunology , Receptor for Advanced Glycation End Products/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology
3.
Arterioscler Thromb Vasc Biol ; 40(9): 2244-2264, 2020 09.
Article in English | MEDLINE | ID: mdl-32640900

ABSTRACT

OBJECTIVE: Nanog is expressed in adult endothelial cells (ECs) at a low-level, however, its functional significance is not known. The goal of our study was to elucidate the role of Nanog in adult ECs using a genetically engineered mouse model system. Approach and Results: Biochemical analyses showed that Nanog is expressed in both adult human and mouse tissues. Primary ECs isolated from adult mice showed detectable levels of Nanog, Tert (telomerase reverse transcriptase), and eNos (endothelial nitric oxide synthase). Wnt3a (Wnt family member 3A) increased the expression of Nanog and hTERT (human telomerase reverse transcriptase) in ECs and increased telomerase activity in these cells. In a chromatin immunoprecipitation experiment, Nanog directly bound to the hTERT and eNOS promoter/enhancer DNA elements, thereby regulating their transcription. Administration of low-dose tamoxifen to ROSAmT/mG::Nanogfl/+::Cdh5CreERT2 mice induced deletion of a single Nanog allele, simultaneously labeling ECs with green fluorescent protein and resulting in decreased Tert and eNos levels. Histological and morphometric analyses of heart tissue sections prepared from these mice revealed cell death, microvascular rarefaction, and increased fibrosis in cardiac vessels. Accordingly, EC-specific Nanog-haploinsufficiency resulted in impaired EC homeostasis and angiogenesis. Conversely, re-expression of cDNA encoding the hTERT in Nanog-depleted ECs, in part, restored the effect of loss of Nanog. CONCLUSIONS: We showed that low-level Nanog expression is required for normal EC homeostasis and angiogenesis in adulthood.


Subject(s)
Cell Proliferation , Cellular Senescence , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Nanog Homeobox Protein/metabolism , Animals , Apoptosis , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Coronary Vessels/drug effects , Coronary Vessels/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Fibrosis , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Nanog Homeobox Protein/deficiency , Nanog Homeobox Protein/genetics , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Telomerase/genetics , Telomerase/metabolism , Transcriptional Activation , Wnt Signaling Pathway , Wnt3A Protein/pharmacology
4.
Inflamm Res ; 69(5): 435-451, 2020 May.
Article in English | MEDLINE | ID: mdl-32162012

ABSTRACT

BACKGROUND: This review focuses on exosomes derived from various cancer cells. The review discusses the possibility of differentiating macrophages in alternatively activated anti-inflammatory pro-tumorigenic M2 macrophage phenotypes and classically activated pro-inflammatory, anti-tumorigenic M1 macrophage phenotypes in the tumor microenvironment (TME). The review is divided into two main parts, as follows: (1) role of exosomes in alternatively activating M2-like macrophages-breast cancer-derived exosomes, hepatocellular carcinoma (HCC) cell-derived exosomes, lung cancer-derived exosomes, prostate cancer-derived exosomes, Oral squamous cell carcinoma (OSCC)-derived exosomes, epithelial ovarian cancer (EOC)-derived exosomes, Glioblastoma (GBM) cell-derived exosomes, and colorectal cancer-derived exosomes, (2) role of exosomes in classically activating M1-like macrophages, oral squamous cell carcinoma-derived exosomes, breast cancer-derived exosomes, Pancreatic-cancer derived modified exosomes, and colorectal cancer-derived exosomes, and (3) exosomes and antibody-dependent cellular cytotoxicity (ADCC). This review addresses the following subjects: (1) crosstalk between cancer-derived exosomes and recipient macrophages, (2) the role of cancer-derived exosome payload(s) in modulating macrophage fate of differentiation, and (3) intracellular signaling mechanisms in macrophages regarding the exosome's payload(s) upon its uptake and regulation of the TME. EVIDENCE: Under the electron microscope, nanoscale exosomes appear as specialized membranous vesicles that emerge from the endocytic cellular compartments. Exosomes harbor proteins, growth factors, cytokines, lipids, miRNA, mRNA, and DNAs. Exosomes are released by many cell types, including reticulocytes, dendritic cells, B-lymphocytes, platelets, mast cells, and tumor cells. It is becoming clear that exosomes can impinge upon signal transduction pathways, serve as a mediator of signaling crosstalk, thereby regulating cell-to-cell wireless communications. CONCLUSION: Based on the vesicular cargo, the molecular constituents, the exosomes have the potential to change the fate of macrophage phenotypes, either M1, classically activated macrophages, or M2, alternatively activated macrophages. In this review, we discuss and describe the ability of tumor-derived exosomes in the mechanism of macrophage activation and polarization.


Subject(s)
Exosomes/immunology , Macrophages/immunology , Neoplasms/immunology , Animals , Humans , Phenotype
5.
Am J Physiol Cell Physiol ; 318(4): C762-C776, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31995410

ABSTRACT

Hypercholesterolemia is a major risk factor for adverse cardiovascular outcomes, but its effect on angiogenesis and wound healing is not well understood. In this study, using a combination of mass spectrometry and laurdan two-photon imaging, we show that elevated levels of low-density lipoprotein (LDL), like those seen in hypercholesterolemic patients, lead to an increase in both free cholesterol and cholesterol esters, as well as increase in lipid order of endothelial cell membranes. Notably, these effects are distinct and opposite to the lack of cholesterol loading and the disruption of lipid order observed in our earlier studies in response to oxidized LDL (oxLDL). The same pathological level of LDL leads to a significant inhibition of endothelial proliferation and cell cycle arrest in G2/M phase, whereas oxLDL enhances endothelial proliferation in S phase of the cycle. LDL but not oxLDL suppresses the expression of vascular endothelial growth factor receptor-2 while enhancing the expression of vascular endothelial growth factor (VEGF). Furthermore, we show that aged (8-10 mo) hypercholesterolemic apolipoprotein E-deficient (ApoE-/-) mice display delayed wound closure compared with age-matched C57/BL6 wild-type controls following a skin punch biopsy. The delay in wound healing is associated with a decreased expression of cluster of differentiation 31 platelet endothelial cell adhesion molecule endothelial marker and decreased angiogenesis within the wound bed. Furthermore, decreased endothelial responsiveness to the growth factors VEGF and basic fibroblast growth factor is observed in ApoE-/- mice in Matrigel plugs and in Matrigels with high levels of LDL in wild-type mice. We propose that plasma hypercholesterolemia is antiangiogenic due to elevated levels of LDL.


Subject(s)
Cholesterol/metabolism , Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , Wound Healing/physiology , Animals , Cells, Cultured , Collagen , Drug Combinations , Hypercholesterolemia/blood , Hypercholesterolemia/metabolism , Laminin , Mice , Neovascularization, Pathologic/metabolism , Proteoglycans , Vascular Endothelial Growth Factor A/metabolism
6.
Cell Rep ; 29(11): 3472-3487.e4, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31825830

ABSTRACT

The vascular endothelial growth factor-A (VEGF-A)-VEGFR2 pathway drives tumor vascularization by activating proangiogenic signaling in endothelial cells (ECs). Here, we show that EC-sphingosine-1-phosphate receptor 1 (S1PR1) amplifies VEGFR2-mediated angiogenic signaling to enhance tumor growth. We show that cancer cells induce S1PR1 activity in ECs, and thereby, conditional deletion of S1PR1 in ECs (EC-S1pr1-/- mice) impairs tumor vascularization and growth. Mechanistically, we show that S1PR1 engages the heterotrimeric G-protein Gi, which amplifies VEGF-VEGFR2 signaling due to an increase in the activity of the tyrosine kinase c-Abl1. c-Abl1, by phosphorylating VEGFR2 at tyrosine-951, prolongs VEGFR2 retention on the plasmalemma to sustain Rac1 activity and EC migration. Thus, S1PR1 or VEGFR2 antagonists, alone or in combination, reverse the tumor growth in control mice to the level seen in EC-S1pr1-/- mice. Our findings suggest that blocking S1PR1 activity in ECs has the potential to suppress tumor growth by preventing amplification of VEGF-VEGFR2 signaling.


Subject(s)
Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , HEK293 Cells , Humans , Male , Mice , Neoplasms, Experimental/pathology , Neuropeptides/metabolism , Proto-Oncogene Proteins c-abl/metabolism , rac1 GTP-Binding Protein/metabolism
7.
Front Cell Dev Biol ; 7: 353, 2019.
Article in English | MEDLINE | ID: mdl-31998716

ABSTRACT

Exosomes have been described as nanoscale membranous extracellular vesicles that emerge from a variety of cells and tissues and are enriched with biologically active genomic and non-genomic biomolecules capable of transducing cell to cell communication. Exosome release, and exosome mediated signaling and cross-talks have been reported in several pathophysiological states. Therefore, exosomes have the potential to become suitable for the diagnosis, prognosis and treatment of specific diseases, including endothelial cell (EC) dysfunction and regeneration. The role of EC-derived exosomes in the mechanisms of cardiovascular tissue regenerative processes represents currently an area of intense research activity. Recent studies have described the potential of exosomes to influence the pathophysiology of immune signaling, tumor metastasis, and angiogenesis. In this review, we briefly discuss progress made in our understanding of the composition and the roles of exosomes in relation to EC regeneration as well as revascularization of ischemic tissues.

8.
J Biol Chem ; 292(50): 20785-20798, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29070680

ABSTRACT

A key feature of acute myocardial infarction (AMI) is an alteration in cardiac architecture. Signaling events that result in the inhibition of glycogen synthase kinase-3 (GSK-3)ß represent an adaptive response that might limit the extent of adverse remodeling in the aftermath of AMI. Here, we report that an allosteric inhibitor of GSK-3ß, 4-benzyl-2-(naphthalene-1-yl)-1,2,4-thiadiazolidine-3,5-dione (NP12), lessens the magnitude of adverse myocardial remodeling and promotes angiogenesis. Male and female mice 8-10 weeks old were grouped (six animals in each group) into sham surgery (sham group), left anterior descending (LAD) ligation of the coronary artery followed by intramyocardial PBS injections (control group), and LAD ligation followed by NP12 administration (NP12 group). After 7 and 14 days, the extents of fibrosis and integrity of blood vessels were determined. Intramyocardial administration of NP12 increased phosphorylation of GSK-3ß, reduced fibrosis, and restored diastolic function in the mice that had experienced an AMI. Morphometric analyses revealed increased CD31+ and Ki67+ vascular structures and decreased apoptosis in these mice. NP12 administration mediated proliferation of reparative cells in the AMI hearts. In a time-course analysis, Wnt3a and NP12 stabilized ß-catenin and increased expression of both Nanog and VEGFR2. Moreover, NP12 increased the expression of ß-catenin and Nanog in myocardium from AMI mice. Finally, loss- and gain-of-function experiments indicated that the NP12-mediated benefit is, in part, Nanog-specific. These findings indicate that NP12 reduces fibrosis, reestablishes coronary blood flow, and improves ventricular function following an AMI. We conclude that NP12 might be useful for limiting ventricular remodeling after an AMI.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Atrial Remodeling/drug effects , Disease Models, Animal , Glycogen Synthase Kinase 3/antagonists & inhibitors , Myocardial Infarction/drug therapy , Protein Kinase Inhibitors/therapeutic use , Thiadiazoles/therapeutic use , Allosteric Regulation/drug effects , Angiogenesis Inducing Agents/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aorta/surgery , Apoptosis/drug effects , Coronary Vessels/drug effects , Coronary Vessels/pathology , Female , Glycogen Synthase Kinase 3/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , In Vitro Techniques , Ligation , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Thiadiazoles/pharmacology
9.
Am J Physiol Cell Physiol ; 313(3): C340-C351, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28701359

ABSTRACT

Oxidized modifications of LDL (oxLDL) play a key role in the development of endothelial dysfunction and atherosclerosis. However, the underlying mechanisms of oxLDL-mediated cellular behavior are not completely understood. Here, we compared the effects of two major types of oxLDL, copper-oxidized LDL (Cu2+-oxLDL) and lipoxygenase-oxidized LDL (LPO-oxLDL), on proliferation of human aortic endothelial cells (HAECs). Cu2+-oxLDL enhanced HAECs' proliferation in a dose- and degree of oxidation-dependent manner. Similarly, LPO-oxLDL also enhanced HAEC proliferation. Mechanistically, both Cu2+-oxLDL and LPO-oxLDL enhance HAEC proliferation via activation of Rho, Akt phosphorylation, and a decrease in the expression of cyclin-dependent kinase inhibitor 1B (p27kip1). Both Cu2+-oxLDL or LPO-oxLDL significantly increased Akt phosphorylation, whereas an Akt inhibitor, MK2206, blocked oxLDL-induced increase in HAEC proliferation. Blocking Rho with C3 or its downstream target ROCK with Y27632 significantly inhibited oxLDL-induced Akt phosphorylation and proliferation mediated by both Cu2+- and LPO-oxLDL. Activation of RhoA was blocked by Rho-GDI-1, which also abrogated oxLDL-induced Akt phosphorylation and HAEC proliferation. In contrast, blocking Rac1 in these cells had no effect on oxLDL-induced Akt phosphorylation or cell proliferation. Moreover, oxLDL-induced Rho/Akt signaling downregulated cell cycle inhibitor p27kip1 Preloading these cells with cholesterol, however, prevented oxLDL-induced Akt phosphorylation and HAEC proliferation. These findings provide a new understanding of the effects of oxLDL on endothelial proliferation, which is essential for developing new treatments against neovascularization and progression of atherosclerosis.


Subject(s)
Cholesterol/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endothelial Cells/physiology , Lipoproteins, LDL/metabolism , Oncogene Protein v-akt/metabolism , rho-Associated Kinases/metabolism , Cell Proliferation/physiology , Cells, Cultured , Endothelial Cells/cytology , Gene Expression Regulation, Enzymologic/physiology , Humans , Signal Transduction/physiology
10.
PLoS One ; 12(5): e0176496, 2017.
Article in English | MEDLINE | ID: mdl-28467484

ABSTRACT

RATIONALE: The human epigenome is plastic. The goal of this study was to address if fibroblast cells can be epigenetically modified to promote neovessel formation. METHODS AND RESULTS: Here, we used highly abundant human adult dermal fibroblast cells (hADFCs) that were treated with the chromatin-modifying agents 5-aza-2'-deoxycytidine and trichostatin A, and subsequently subjected to differentiation by activating Wnt signaling. Our results show that these epigenetically modified hADFCs increasingly expressed ß-catenin, pluripotency factor octamer-binding transcription factor-4 (OCT4, also known as POU5F1), and endothelial cell (EC) marker called vascular endothelial growth factor receptor-2 (VEGFR-2, also known as Fetal Liver Kinase-1). In microscopic analysis, ß-catenin localized to cell-cell contact points, while OCT4 was found to be localized primarily to the nucleus of these cells. Furthermore, in a chromatin immunoprecipitation experiment, OCT4 bound to the VEGFR-2/FLK1 promoter. Finally, these modified hADFCs also transduced Wnt signaling. Importantly, on a two-dimensional (2D) gel substrate, a subset of the converted cells formed vascular network-like structures in the presence of VEGF. CONCLUSION: Chromatin-modifying agents converted hADFCs to OCT4+ and VEGFR-2+ capillary tube-forming cells in a 2D matrix in VEGF-dependent manner.


Subject(s)
Capillaries/cytology , Chromatin/metabolism , Fibroblasts/metabolism , Octamer Transcription Factor-3/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Humans
11.
Cardiovasc Res ; 111(1): 105-18, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27125875

ABSTRACT

AIMS: Lipid phosphate phosphatase-3 (LPP3) is expressed at high levels in endothelial cells (ECs). Although LPP3 is known to hydrolyse the phosphate group from lysolipids such as spingosine-1-phosphate and its structural homologues, the function of Lpp3 in ECs is not completely understood. In this study, we investigated how tyrosine-protein kinase receptor (TEK or Tie2) promoter-dependent deletion of Lpp3 alters EC activities. METHODS AND RESULTS: Lpp3(fl/fl) mice were crossed with the tg.Tie2(Cre) transgenic line. Vasculogenesis occurred normally in embryos with Tie2(Cre)-mediated deletion of Lpp3 (called Lpp3(ECKO)), but embryonic lethality occurred in two waves, the first wave between E8.5 and E10.5, while the second between E11.5 and E13.5. Lethality in Lpp3(ECKO) embryos after E11.5 was accompanied by vascular leakage and haemorrhage, which likely resulted in insufficient cardiovascular development. Analyses of haematoxylin- and eosin-stained heart sections from E11.5 Lpp3(ECKO) embryos showed insufficient heart growth associated with decreased trabeculation, reduced growth of the compact wall, and absence of cardiac cushions. Staining followed by microscopic analyses of Lpp3(ECKO) embryos revealed the presence of apoptotic ECs. Furthermore, Lpp3-deficient ECs showed decreased gene expression and protein levels of Cyclin-D1, VE-cadherin, Fibronectin, Klf2, and Klf4. To determine the underlying mechanisms of vascular leakage and barrier disruption, we performed knockdown and rescue experiments in cultured ECs. LPP3 knockdown decreased transendothelial electrical resistance and increased permeability. Re-expression of ß-catenin cDNA in LPP3-knockdown ECs partially restored the effect of the LPP3 loss, whereas re-expression of p120ctn cDNA did not. CONCLUSION: These findings demonstrate the essential roles of LPP3 in the maturation of EC barrier integrity and normal cardiovascular development.


Subject(s)
Blood Vessels/enzymology , Capillary Permeability , Endothelial Cells/enzymology , Neovascularization, Physiologic , Phosphatidate Phosphatase/deficiency , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis , Blood Vessels/embryology , Blood Vessels/pathology , Cadherins/genetics , Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , Cells, Cultured , Electric Impedance , Endothelial Cells/pathology , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Heart/embryology , Kruppel-Like Factor 4 , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphatidate Phosphatase/genetics , RNA Interference , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Transfection , beta Catenin/genetics , beta Catenin/metabolism , Delta Catenin
12.
J Lipid Res ; 57(5): 791-808, 2016 05.
Article in English | MEDLINE | ID: mdl-26989083

ABSTRACT

Endothelial biomechanics is emerging as a key factor in endothelial function. Here, we address the mechanisms of endothelial stiffening induced by oxidized LDL (oxLDL) and investigate the role of oxLDL in lumen formation. We show that oxLDL-induced endothelial stiffening is mediated by CD36-dependent activation of RhoA and its downstream target, Rho kinase (ROCK), via inhibition of myosin light-chain phosphatase (MLCP) and myosin light-chain (MLC)2 phosphorylation. The LC-MS/MS analysis identifies 7-ketocholesterol (7KC) as the major oxysterol in oxLDL. Similarly to oxLDL, 7KC induces RhoA activation, MLCP inhibition, and MLC2 phosphorylation resulting in endothelial stiffening. OxLDL also facilitates formation of endothelial branching networks in 3D collagen gels in vitro and induces increased formation of functional blood vessels in a Matrigel plug assay in vivo. Both effects are RhoA and ROCK dependent. An increase in lumen formation was also observed in response to pre-exposing the cells to 7KC, an oxysterol that induces endothelial stiffening, but not to 5α,6α epoxide that does not affect endothelial stiffness. Importantly, loading cells with cholesterol prevented oxLDL-induced RhoA activation and the downstream signaling cascade, and reversed oxLDL-induced lumen formation. In summary, we show that oxLDL-induced endothelial stiffening is mediated by the CD36/RhoA/ROCK/MLCP/MLC2 pathway and is associated with increased endothelial angiogenic activity.


Subject(s)
Endothelial Cells/pathology , Lipoproteins, LDL/physiology , Neovascularization, Pathologic/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cardiac Myosins/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Mice, Nude , Mice, SCID , Myosin Light Chains/metabolism , Signal Transduction , Vascular Stiffness , rho-Associated Kinases/metabolism
13.
Methods Mol Biol ; 1357: 311-27, 2016.
Article in English | MEDLINE | ID: mdl-25687301

ABSTRACT

The study of stem cell behavior and differentiation in a developmental context is complex, time-consuming, and expensive, and for this reason, cell culture remains a method of choice for developmental and regenerative biology and mechanistic studies. Similar to ES cells, iPS cells have the ability to differentiate into endothelial cells (ECs), and the route for differentiation appears to mimic the developmental process that occurs during the formation of an embryo. Traditional EC induction methods from embryonic stem (ES) cells rely mostly on the formation of embryoid body (EB), which employs feeder or feeder-free conditions in the presence or absence of supporting cells. Similar to ES cells, iPS cells can be cultured in feeder layer or feeder-free conditions. Here, we describe the iPS cell culture methods and induction differentiation of these cells into ECs. We use anti-mouse Flk1 and anti-mouse VE-cadherin to isolate and characterize mouse ECs, because these antibodies are commercially available and their use has been described in the literature, including by our group. The ECs produced by this method have been used by our laboratory, and we have demonstrated their in vivo potential. We also discuss how iPS cells differ in their ability to differentiate into endothelial cells in culture.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming Techniques/methods , Induced Pluripotent Stem Cells/cytology , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Cadherins/immunology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Cryopreservation , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Hindlimb/blood supply , Induced Pluripotent Stem Cells/transplantation , Ischemia/therapy , Kruppel-Like Factor 4 , Mice , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/immunology
14.
Stem Cell Reports ; 5(1): 10-21, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26120059

ABSTRACT

Understanding epigenetic mechanisms regulating embryonic stem cell (ESC) differentiation to endothelial cells may lead to increased efficiency of generation of vessel wall endothelial cells needed for vascular engineering. Here we demonstrated that the histone demethylases KDM4A and KDM4C played an indispensable but independent role in mediating the expression of fetal liver kinase (Flk)1 and VE-cadherin, respectively, and thereby the transition of mouse ESCs (mESCs) to endothelial cells. KDM4A was shown to bind to histones associated with the Flk1 promoter and KDM4C to bind to histones associated with the VE-cadherin promoter. KDM4A and KDM4C were also both required for capillary tube formation and vasculogenesis in mice. We observed in zebrafish that KDM4A depletion induced more severe vasculogenesis defects than KDM4C depletion, reflecting the early involvement of KDM4A in specifying endothelial cell fate. These findings together demonstrate the essential role of KDM4A and KDM4C in orchestrating mESC differentiation to endothelial cells through the activation of Flk1 and VE-cadherin promoters, respectively.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cell Differentiation/genetics , Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Antigens, CD/genetics , Cadherins/genetics , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Mice , Promoter Regions, Genetic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish
15.
Stem Cells ; 33(6): 1719-29, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25693840

ABSTRACT

Adhesion of embryonic stem cells (ESCs) to the extracellular matrix may influence differentiation potential and cell fate decisions. Here, we investigated the inductive role of binding of integrin α6ß1 expressed in mouse (m)ESCs to laminin-1 (LN1) in mediating the differentiation of ESCs to endothelial cells (ECs). We observed that α6ß1 binding to LN1 was required for differentiation to ECs. α6ß1 functioned by recruiting the adaptor tetraspanin protein CD151, which activated FAK and Akt signaling and mediated the EC lineage-specifying transcription factor Er71. In contrast, association of the ESC-expressed α3ß1, another highly expressed LN1 binding integrin, with CD151, prevented α6ß1-mediated differentiation. CD151 thus functioned as a bifurcation router to direct ESCs toward ECs when α6ß1 associated with CD151, or prevented transition to ECs when α3ß1 associated with CD151. These observations were recapitulated in mice in which α6 integrin or CD151 knockdown reduced the expression of Er71-regulated angiogenesis genes and development of blood vessels. Thus, interaction of α6ß1 in ESCs with LN1 activates α6ß1/CD151 signaling which programs ESCs toward the EC lineage fate.


Subject(s)
Cell Differentiation/physiology , Endothelial Cells/cytology , Integrin alpha6beta1/metabolism , Laminin/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Cell Adhesion/physiology , Extracellular Matrix/metabolism , Mice , Morphogenesis/physiology , Mouse Embryonic Stem Cells/cytology , Signal Transduction/physiology , Tetraspanin 24/genetics
16.
J Cell Sci ; 127(Pt 20): 4518-30, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128562

ABSTRACT

Although the trafficking of newly synthesized VEGFR2 to the plasma membrane is a key determinant of angiogenesis, the molecular mechanisms of Golgi to plasma membrane trafficking are unknown. Here, we have identified a key role of the kinesin family plus-end molecular motor KIF13B in delivering VEGFR2 cargo from the Golgi to the endothelial cell surface. KIF13B is shown to interact directly with VEGFR2 on microtubules. We also observed that overexpression of truncated versions of KIF13B containing the binding domains that interact with VEGFR2 inhibited VEGF-induced capillary tube formation. KIF13B depletion prevented VEGF-mediated endothelial migration, capillary tube formation and neo-vascularization in mice. Impairment in trafficking induced by knockdown of KIF13B shunted VEGFR2 towards the lysosomal degradation pathway. Thus, KIF13B is an essential molecular motor required for the trafficking of VEGFR2 from the Golgi, and its delivery to the endothelial cell surface mediates angiogenesis.


Subject(s)
Cell Membrane/metabolism , Golgi Apparatus/metabolism , Kinesins/metabolism , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Humans , Kinesins/genetics , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Neovascularization, Physiologic/genetics , Protein Binding , Protein Structure, Tertiary/genetics , Protein Transport/genetics , RNA, Small Interfering/genetics , Sequence Deletion/genetics , Transgenes/genetics
17.
J Biol Chem ; 289(19): 13476-91, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24634221

ABSTRACT

Hepatocyte growth factor (HGF) mediated signaling promotes cell proliferation and migration in a variety of cell types and plays a key role in tumorigenesis. As cell migration is important to angiogenesis, we characterized HGF-mediated effects on the formation of lamellipodia, a pre-requisite for migration using human lung microvascular endothelial cells (HLMVECs). HGF, in a dose-dependent manner, induced c-Met phosphorylation (Tyr-1234/1235, Tyr-1349, Ser-985, Tyr-1003, and Tyr-1313), activation of PI3k (phospho-Yp85) and Akt (phospho-Thr-308 and phospho-Ser-473) and potentiated lamellipodia formation and HLMVEC migration. Inhibition of c-Met kinase by SU11274 significantly attenuated c-Met, PI3k, and Akt phosphorylation, suppressed lamellipodia formation and endothelial cell migration. LY294002, an inhibitor of PI3k, abolished HGF-induced PI3k (Tyr-458), and Akt (Thr-308 and Ser-473) phosphorylation and suppressed lamellipodia formation. Furthermore, HGF stimulated p47(phox)/Cortactin/Rac1 translocation to lamellipodia and ROS generation. Moreover, inhibition of c-Met/PI3k/Akt signaling axis and NADPH oxidase attenuated HGF- induced lamellipodia formation, ROS generation and cell migration. Ex vivo experiments with mouse aortic rings revealed a role for c-Met signaling in HGF-induced sprouting and lamellipodia formation. Taken together, these data provide evidence in support of a significant role for HGF-induced c-Met/PI3k/Akt signaling and NADPH oxidase activation in lamellipodia formation and motility of lung endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Hepatocyte Growth Factor/metabolism , Lung/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Pseudopodia/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Endothelial Cells/cytology , Hepatocyte Growth Factor/genetics , Humans , Lung/cytology , Mice , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Pseudopodia/genetics
18.
Stem Cells ; 32(6): 1538-52, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24496925

ABSTRACT

Endothelial cell (EC) dedifferentiation in relation to neovascularization is a poorly understood process. In this report, we addressed the role of Wnt signaling in the mechanisms of neovascularization in adult tissues. Here, we show that a low-dose of 6-bromoindirubin-3'-oxime (BIO), a competitive inhibitor of glycogen synthase kinase-3ß, induced the stabilization of ß-catenin and its subsequent direct interaction with the transcription factor NANOG in the nucleus of ECs. This event induced loss of VE-cadherin from the adherens junctions, increased EC proliferation accompanied by asymmetric cell division (ACD), and formed cellular aggregates in hanging drop assays indicating the acquisition of a dedifferentiated state. In a chromatin immunoprecipitation assay, nuclear NANOG protein bound to the NANOG- and VEGFR2-promoters in ECs, and the addition of BIO activated the NANOG-promoter-luciferase reporter system in a cell-based assay. Consequently, NANOG-knockdown decreased BIO-induced NOTCH-1 expression, thereby decreasing cell proliferation, ACD, and neovascularization. In a Matrigel plug assay, BIO induced increased neovascularization, secondary to the presence of vascular endothelial growth factor (VEGF). Moreover, in a mouse model of hind limb ischemia, BIO augmented neovascularization that was coupled with increased expression of NOTCH-1 in ECs and increased smooth muscle α-actin(+) cell recruitment around the neovessels. Thus, these results demonstrate the ability of a low-dose of BIO to augment neovascularization secondary to VEGF, a process that was accompanied by a partial dedifferentiation of ECs via ß-catenin and the NANOG signaling pathway.


Subject(s)
Cell Dedifferentiation/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Indoles/pharmacology , Neovascularization, Physiologic/drug effects , Oximes/pharmacology , Angiogenesis Inducing Agents/metabolism , Animals , Cell Aggregation/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Fetal Proteins/genetics , Hindlimb/blood supply , Hindlimb/pathology , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoles/administration & dosage , Ischemia/pathology , Mice , Nanog Homeobox Protein , Oximes/administration & dosage , Phenotype , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Stability/drug effects , T-Box Domain Proteins/genetics , Vascular Endothelial Growth Factor A/pharmacology , beta Catenin/metabolism
19.
PLoS One ; 8(12): e85549, 2013.
Article in English | MEDLINE | ID: mdl-24386480

ABSTRACT

RATIONALE: Induced pluripotent stem (iPS) cells have emerged as a source of potentially unlimited supply of autologous endothelial cells (ECs) for vascularization. However, the regenerative function of these cells relative to adult ECs and ECs derived from embryonic stem (ES) cells is unknown. The objective was to define the differentiation characteristics and vascularization potential of Fetal liver kinase (Flk)1(+) and Vascular Endothelial (VE)-cadherin(+) ECs derived identically from mouse (m)ES and miPS cells. METHODS AND RESULTS: Naive mES and miPS cells cultured in type IV collagen (IV Col) in defined media for 5 days induced the formation of adherent cell populations, which demonstrated similar expression of Flk1 and VE-cadherin and the emergence of EC progenies. FACS purification resulted in 100% Flk1(+) VE-cadherin(+) cells from both mES and miPS cells. Emergence of Flk1(+)VE-cadherin(+) cells entailed expression of the vascular developmental transcription factor Er71, which bound identically to Flk1, VE-cadherin, and CD31 promoters in both populations. Immunostaining with anti-VE-cadherin and anti-CD31 antibodies and microscopy demonstrated the endothelial nature of these cells. Each cell population (unlike mature ECs) organized into well-developed vascular structures in vitro and incorporated into CD31(+) neovessels in matrigel plugs implanted in nude mice in vivo. CONCLUSION: Thus, iPS cell-derived Flk1(+)VE-cadherin(+) cells expressing the Er71 are as angiogenic as mES cell-derived cells and incorporate into CD31(+) neovessels. Their vessel forming capacity highlights the potential of autologous iPS cells-derived EC progeny for therapeutic angiogenesis.


Subject(s)
Antigens, CD/biosynthesis , Cadherins/biosynthesis , Endothelial Cells/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Animals , Cell Line , Endothelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Mice , Transcription Factors/biosynthesis
20.
J Tissue Eng Regen Med ; 7(8): 621-30, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22696416

ABSTRACT

Manipulation of stem cells using physicochemical stimuli has emerged as an important tool in regenerative medicine. While 2D substrates with tunable elasticity have been studied for control of stem cell differentiation, we recently developed a stratified co-culture model of angiogenesis of human mesenchymal stem cells (hMSCs) that differentiate on a tunable polydimethylsiloxane (PDMS) substrate, thereby creating a physiologic context for elasticity-induced differentiation. Endothelial cells (EC) were cultured on top of the hMSC construct on a collagen gel to monitor network formation. Media composition influenced EC invasion due to the conditioning media, the reduction of serum and supplemental growth factors, and the addition of recombinant growth factors. Conditioned media, recombinant growth factors and direct co-culture were compared for endothelial cell invasive response using quantitative image analysis. As anticipated, use of recombinant vascular endothelial growth factor (VEGF) induced the deepest EC invasions while direct co-culture caused shallow invasions compared to other conditions. However, endothelial cells displayed lumen-like morphology, suggesting that cell-cell interaction in the co-culture model could mimic sprouting behaviour. In summary, an engineered suitable biochemical and physical environment facilitated endothelial cells to form 3D vessel structures onto hMSCs. These structures were plated on a stiff surface known to induce osteodifferentiation of stem cells. This low cost co-culture system, with its minimal chemical supplementation and physically controllable matrix, could potentially model in vivo potential in engineered and pre-vascularized bone grafts.


Subject(s)
Cell Communication , Cell Differentiation , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Animals , Cell Line , Coculture Techniques , Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Rats , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...