Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Aging Cell ; 23(4): e14095, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38348753

ABSTRACT

As the innermost lining of the vasculature, endothelial cells (ECs) are constantly subjected to systemic inflammation and particularly vulnerable to aging. Endothelial health is hence vital to prevent age-related vascular disease. Healthy ECs rely on the proper localization of transcription factors via nuclear pore complexes (NPCs) to govern cellular behavior. Emerging studies report NPC degradation with natural aging, suggesting impaired nucleocytoplasmic transport in age-associated EC dysfunction. We herein identify nucleoporin93 (Nup93), a crucial structural NPC protein, as an indispensable player in vascular protection. Endothelial Nup93 protein levels are significantly reduced in the vasculature of aged mice, paralleling observations of Nup93 loss when using in vitro models of EC senescence. The loss of Nup93 in human ECs induces cell senescence and promotes the expression of inflammatory adhesion molecules, where restoring Nup93 protein in senescent ECs reverses features of endothelial aging. Mechanistically, we find that both senescence and loss of Nup93 impair endothelial NPC transport, leading to nuclear accumulation of Yap and downstream inflammation. Pharmacological studies indicate Yap hyperactivation as the primary consequence of senescence and Nup93 loss in ECs. Collectively, our findings indicate that the maintenance of endothelial Nup93 is a key determinant of EC health, where aging targets endothelial Nup93 levels to impair NPC function as a novel mechanism of EC senescence and vascular aging.


Subject(s)
Cellular Senescence , Endothelial Cells , Humans , Mice , Animals , Endothelial Cells/metabolism , Aging/physiology , Cells, Cultured , Inflammation/metabolism
2.
J Clin Invest ; 134(4)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38175710

ABSTRACT

Blood vessels are continually exposed to circulating lipids, and elevation of ApoB-containing lipoproteins causes atherosclerosis. Lipoprotein metabolism is highly regulated by lipolysis, largely at the level of the capillary endothelium lining metabolically active tissues. How large blood vessels, the site of atherosclerotic vascular disease, regulate the flux of fatty acids (FAs) into triglyceride-rich (TG-rich) lipid droplets (LDs) is not known. In this study, we showed that deletion of the enzyme adipose TG lipase (ATGL) in the endothelium led to neutral lipid accumulation in vessels and impaired endothelial-dependent vascular tone and nitric oxide synthesis to promote endothelial dysfunction. Mechanistically, the loss of ATGL led to endoplasmic reticulum stress-induced inflammation in the endothelium. Consistent with this mechanism, deletion of endothelial ATGL markedly increased lesion size in a model of atherosclerosis. Together, these data demonstrate that the dynamics of FA flux through LD affects endothelial cell homeostasis and consequently large vessel function during normal physiology and in a chronic disease state.


Subject(s)
Atherosclerosis , Lipase , Mice , Animals , Triglycerides/metabolism , Lipase/genetics , Lipase/metabolism , Lipolysis , Lipid Metabolism , Endothelium, Vascular/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism
3.
bioRxiv ; 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38014013

ABSTRACT

Endothelial cells (ECs) form the innermost lining of the vasculature and serve a pivotal role in preventing age-related vascular disease. Endothelial health relies on the proper nucleocytoplasmic shuttling of transcription factors via nuclear pore complexes (NPCs). Emerging studies report NPC degradation with natural aging, suggesting impaired nucleocytoplasmic transport in age-related EC dysfunction. We herein identify nucleoporin93 (Nup93), a crucial structural NPC protein, as an indispensable player for vascular protection. Endothelial Nup93 protein levels are significantly reduced in the vasculature of aged mice, paralleling observations of Nup93 loss when using in vitro models of endothelial aging. Mechanistically, we find that loss of Nup93 impairs NPC transport, leading to the nuclear accumulation of Yap and downstream inflammation. Collectively, our findings indicate maintenance of endothelial Nup93 as a key determinant of EC health, where aging targets endothelial Nup93 levels to impair NPC function as a novel mechanism for EC senescence and vascular aging.

4.
Front Cardiovasc Med ; 10: 1279868, 2023.
Article in English | MEDLINE | ID: mdl-38034389

ABSTRACT

Objective: We have previously demonstrated the in vivo importance of the Akt-eNOS substrate-kinase relationship, as defective postnatal angiogenesis characteristic of global Akt1-null mice is rescued when bred to 'gain-of-function' eNOS S1176D mutant mice. While multiple studies support the vascular protective role of endothelial NO generation, the causal role of Akt1-dependent eNOS S1176 phosphorylation during atherosclerotic plaque formation is not yet clear. Approach and results: We herein bred congenic 'loss-of-function' eNOS S1176A and 'gain-of-function' eNOS S1176D mutant mice to the exacerbated atherogenic Akt1-/-; ApoE-/- double knockout mice to definitively test the importance of Akt-mediated eNOS S1176 phosphorylation during atherogenesis. We find that a single amino acid substitution at the eNOS S1176 phosphorylation site yields divergent effects on atherosclerotic plaque formation, as an eNOS phospho-mimic aspartate (D) substitution at S1176 leads to favorable lipid profiles and decreased indices of atherosclerosis, even when on a proatherogenic Akt1 global deletion background. Conversely, mice harboring an unphosphorylatable mutation to alanine (S1176A) result in increased plasma lipids, increased lesion formation and cellular apoptosis, phenocopying the physiological consequence of eNOS deletion and/or impaired enzyme function. Furthermore, gene expression analyses of whole aortas indicate a combinatorial detriment from NO deficiency and Western Diet challenge, as 'loss-of-function' eNOS S1176A mice on a Western Diet present a unique expression pattern indicative of augmented T-cell activity when compared to eNOS S1176D mice. Conclusions: By using genetic epistasis approaches, we conclusively demonstrate that Akt-mediated eNOS S1176 phosphorylation and subsequent eNOS activation remains to be the most physiologically relevant method of NO production to promote athero-protective effects.

5.
Arterioscler Thromb Vasc Biol ; 38(4): 870-879, 2018 04.
Article in English | MEDLINE | ID: mdl-29449333

ABSTRACT

OBJECTIVE: The importance of PI3K/Akt signaling in the vasculature has been demonstrated in several models, as global loss of Akt1 results in impaired postnatal ischemia- and VEGF-induced angiogenesis. The ubiquitous expression of Akt1, however, raises the possibility of cell-type-dependent Akt1-driven actions, thereby necessitating tissue-specific characterization. APPROACH AND RESULTS: Herein, we used an inducible, endothelial-specific Akt1-deleted adult mouse model (Akt1iECKO) to characterize the endothelial cell autonomous functions of Akt1 in the vascular system. Endothelial-targeted ablation of Akt1 reduces eNOS (endothelial nitric oxide synthase) phosphorylation and promotes both increased vascular contractility in isolated vessels and elevated diastolic blood pressures throughout the diurnal cycle in vivo. Furthermore, Akt1iECKO mice subject to the hindlimb ischemia model display impaired blood flow and decreased arteriogenesis. CONCLUSIONS: Endothelial Akt1 signaling is necessary for ischemic resolution post-injury and likely reflects the consequence of NO insufficiency critical for vascular repair.


Subject(s)
Aorta, Thoracic/enzymology , Endothelial Cells/enzymology , Ischemia/enzymology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt/metabolism , Vasoconstriction , Animals , Blood Flow Velocity , Blood Pressure , Disease Models, Animal , Hindlimb , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Male , Mice, Knockout , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Regional Blood Flow , Signal Transduction
6.
J Biol Chem ; 293(3): 973-983, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29203526

ABSTRACT

Lipid droplets (LD) are dynamic organelles involved in intracellular lipid metabolism in almost all eukaryotic cells, and LD-associated proteins tightly regulate their dynamics. One LD coat protein is caveolin-1 (Cav-1), an essential component for caveola assembly in highly differentiated cells, including adipocytes, smooth muscle cells, and endothelial cells (EC). However, the role of Cav-1 in LD dynamics is unclear. Here we report that EC lacking Cav-1 exhibit impaired LD formation. The decreased LD formation is due to enhanced lipolysis and not caused by reduced triglyceride synthesis or fatty acid uptake. Mechanistically, the absence of Cav-1 increased cAMP/PKA signaling in EC, as indicated by elevated phosphorylation of hormone-sensitive lipase and increased lipolysis. Unexpectedly, we also observed enhanced autocrine production of prostaglandin I2 (PGI2, also called prostacyclin) in Cav-1 KO EC, and this PGI2 increase appeared to stimulate cAMP/PKA pathways, contributing to the enhanced lipolysis in Cav-1 KO cells. Our results reveal an unanticipated role of Cav-1 in regulating lipolysis in non-adipose tissue, indicating that Cav-1 is required for LD metabolism in EC and that it regulates cAMP-dependent lipolysis in part via the autocrine production of PGI2.


Subject(s)
Caveolin 1/metabolism , Cyclic AMP/metabolism , Endothelial Cells/metabolism , Epoprostenol/pharmacology , Lipolysis/drug effects , Animals , Blotting, Western , Caveolin 1/genetics , Cell Line , Cells, Cultured , Lipid Metabolism/drug effects , Mass Spectrometry , Mice , Mice, Mutant Strains , Phosphorylation
7.
Sci Rep ; 7(1): 15386, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29133876

ABSTRACT

Low rates of arteriovenous fistula (AVF) maturation prevent optimal fistula use for hemodialysis; however, the mechanism of venous remodeling in the fistula environment is not well understood. We hypothesized that the embryonic venous determinant Eph-B4 mediates AVF maturation. In human AVF and a mouse aortocaval fistula model, Eph-B4 protein expression increased in the fistula vein; expression of the arterial determinant Ephrin-B2 also increased. Stimulation of Eph-B-mediated signaling with Ephrin-B2/Fc showed improved fistula patency with less wall thickness. Mutagenesis studies showed that tyrosine-774 is critical for Eph-B4 signaling and administration of inactive Eph-B4-Y774F increased fistula wall thickness. Akt1 expression also increased in AVF; Akt1 knockout mice showed reduced fistula diameter and wall thickness. In Akt1 knockout mice, stimulation of Eph-B signaling with Ephrin-B2/Fc showed no effect on remodeling. These results show that AVF maturation is associated with acquisition of dual arteriovenous identity; increased Eph-B activity improves AVF patency. Inhibition of Akt1 function abolishes Eph-B-mediated venous remodeling suggesting that Eph-B4 regulates AVF venous adaptation through an Akt1-mediated mechanism.


Subject(s)
Arteriovenous Shunt, Surgical , Vascular Patency , Vascular Remodeling , Animals , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphB2/genetics , Receptor, EphB2/metabolism , Receptor, EphB4/genetics
8.
Arterioscler Thromb Vasc Biol ; 37(12): 2311-2321, 2017 12.
Article in English | MEDLINE | ID: mdl-29025710

ABSTRACT

OBJECTIVE: Drug-eluting stent delivery of mTORC1 (mechanistic target of rapamycin complex 1) inhibitors is highly effective in preventing intimal hyperplasia after coronary revascularization, but adverse effects limit their use for systemic vascular disease. Understanding the mechanism of action may lead to new treatment strategies. We have shown that rapamycin promotes vascular smooth muscle cell differentiation in an AKT2-dependent manner in vitro. Here, we investigate the roles of AKT (protein kinase B) isoforms in intimal hyperplasia. APPROACH AND RESULTS: We found that germ-line-specific or smooth muscle-specific deletion of Akt2 resulted in more severe intimal hyperplasia compared with control mice after arterial denudation injury. Conversely, smooth muscle-specific Akt1 knockout prevented intimal hyperplasia, whereas germ-line Akt1 deletion caused severe thrombosis. Notably, rapamycin prevented intimal hyperplasia in wild-type mice but had no therapeutic benefit in Akt2 knockouts. We identified opposing roles for AKT1 and AKT2 isoforms in smooth muscle cell proliferation, migration, differentiation, and rapamycin response in vitro. Mechanistically, rapamycin induced MYOCD (myocardin) mRNA expression. This was mediated by AKT2 phosphorylation and nuclear exclusion of FOXO4 (forkhead box O4), inhibiting its binding to the MYOCD promoter. CONCLUSIONS: Our data reveal opposing roles for AKT isoforms in smooth muscle cell remodeling. AKT2 is required for rapamycin's therapeutic inhibition of intimal hyperplasia, likely mediated in part through AKT2-specific regulation of MYOCD via FOXO4. Because AKT2 signaling is impaired in diabetes mellitus, this work has important implications for rapamycin therapy, particularly in diabetic patients.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/pharmacology , Vascular System Injuries/prevention & control , Animals , Binding Sites , Cell Cycle Proteins , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Vascular System Injuries/enzymology , Vascular System Injuries/genetics , Vascular System Injuries/pathology
9.
Circ Res ; 120(8): 1289-1297, 2017 Apr 14.
Article in English | MEDLINE | ID: mdl-28119423

ABSTRACT

RATIONALE: Fatty acids (FA) are transported across the capillary endothelium to parenchymal tissues. However, it is not known how endothelial cells (EC) from large vessels process a postprandial surge of FA. OBJECTIVE: This study was designed to characterize lipid droplet (LD) formation in EC by manipulating pathways leading to the formation and degradation of LD. In addition, several functions of LD-derived FA were assessed. METHODS AND RESULTS: LD were present in EC lining the aorta after the peak in plasma triglycerides initiated by a gavage of olive oil in mice, in vivo. Similarly, in isolated aorta, oleic acid treatment generates LD in EC ex vivo. Cultured EC readily form LD largely via the enzyme DGAT (diacylglycerol O-acyltransferase 1) and degrade LD via ATGL (adipocyte triglyceride lipase) after FA loading. Functionally, LD-derived FA are dynamically regulated and function to protect EC from lipotoxic stress and provide FA for metabolic needs. CONCLUSIONS: Our results delineate endothelial LD dynamics for the first time in vivo and in vitro. Moreover, LD formation protects EC from lipotoxic stress, regulates EC glycolysis, and provides a source of FA for adjacent cells in the vessel wall or tissues.


Subject(s)
Aorta, Thoracic/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Lipid Droplets/metabolism , Olive Oil/metabolism , Administration, Oral , Animals , Cells, Cultured , Diacylglycerol O-Acyltransferase/metabolism , Glycolysis , Humans , Hydrolysis , Intubation, Gastrointestinal , Lipase/metabolism , Lipolysis , Mice, Inbred C57BL , Oleic Acid/metabolism , Olive Oil/administration & dosage , Time Factors , Triglycerides/blood
10.
Nat Commun ; 7: 13516, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869117

ABSTRACT

In humans and animals lacking functional LDL receptor (LDLR), LDL from plasma still readily traverses the endothelium. To identify the pathways of LDL uptake, a genome-wide RNAi screen was performed in endothelial cells and cross-referenced with GWAS-data sets. Here we show that the activin-like kinase 1 (ALK1) mediates LDL uptake into endothelial cells. ALK1 binds LDL with lower affinity than LDLR and saturates only at hypercholesterolemic concentrations. ALK1 mediates uptake of LDL into endothelial cells via an unusual endocytic pathway that diverts the ligand from lysosomal degradation and promotes LDL transcytosis. The endothelium-specific genetic ablation of Alk1 in Ldlr-KO animals leads to less LDL uptake into the aortic endothelium, showing its physiological role in endothelial lipoprotein metabolism. In summary, identification of pathways mediating LDLR-independent uptake of LDL may provide unique opportunities to block the initiation of LDL accumulation in the vessel wall or augment hepatic LDLR-dependent clearance of LDL.


Subject(s)
Activin Receptors, Type II/metabolism , Cholesterol, LDL/metabolism , Endothelial Cells/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/genetics , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Biological Transport , Cells, Cultured , Cholesterol, LDL/genetics , Cloning, Molecular , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Male , Mice , RNA Interference
11.
Circ Res ; 118(1): 48-55, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26602865

ABSTRACT

RATIONALE: Caveolin-1 (Cav-1) negatively regulates endothelial nitric oxide (NO) synthase-derived NO production, and this has been mapped to several residues on Cav-1, including F92. Herein, we reasoned that endothelial expression of an F92ACav-1 transgene would let us decipher the mechanisms and relationships between caveolae structure and intracellular signaling. OBJECTIVE: This study was designed to separate caveolae formation from its downstream signaling effects. METHODS AND RESULTS: An endothelial-specific doxycycline-regulated mouse model for the expression of Cav-1-F92A was developed. Blood pressure by telemetry and nitric oxide bioavailability by electron paramagnetic resonance and phosphorylation of vasodilator-stimulated phosphoprotein were determined. Caveolae integrity in the presence of Cav-1-F92A was measured by stabilization of caveolin-2, sucrose gradient, and electron microscopy. Histological analysis of heart and lung, echocardiography, and signaling were performed. CONCLUSIONS: This study shows that mutant Cav-1-F92A forms caveolae structures similar to WT but leads to increases in NO bioavailability in vivo, thereby demonstrating that caveolae formation and downstream signaling events occur through independent mechanisms.


Subject(s)
Caveolae/metabolism , Caveolin 1/biosynthesis , Caveolin 1/genetics , Intracellular Fluid/metabolism , Signal Transduction/physiology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Caveolae/drug effects , Doxycycline/pharmacology , Humans , Intracellular Fluid/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Signal Transduction/drug effects , Uncoupling Agents/pharmacology
12.
Circ Res ; 118(1): 38-47, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26472816

ABSTRACT

RATIONALE: Several lines of evidence indicate that the regulation of microRNA (miRNA) levels by different stimuli may contribute to the modulation of stimulus-induced responses. The miR-17-92 cluster has been linked to tumor development and angiogenesis, but its role in vascular endothelial growth factor-induced endothelial cell (EC) functions is unclear and its regulation is unknown. OBJECTIVE: The purpose of this study was to elucidate the mechanism by which VEGF regulates the expression of miR-17-92 cluster in ECs and determine its contribution to the regulation of endothelial angiogenic functions, both in vitro and in vivo. This was done by analyzing the effect of postnatal inactivation of miR-17-92 cluster in the endothelium (miR-17-92 iEC-KO mice) on developmental retinal angiogenesis, VEGF-induced ear angiogenesis, and tumor angiogenesis. METHODS AND RESULTS: Here, we show that Erk/Elk1 activation on VEGF stimulation of ECs is responsible for Elk-1-mediated transcription activation (chromatin immunoprecipitation analysis) of the miR-17-92 cluster. Furthermore, we demonstrate that VEGF-mediated upregulation of the miR-17-92 cluster in vitro is necessary for EC proliferation and angiogenic sprouting. Finally, we provide genetic evidence that miR-17-92 iEC-KO mice have blunted physiological retinal angiogenesis during development and diminished VEGF-induced ear angiogenesis and tumor angiogenesis. Computational analysis and rescue experiments show that PTEN (phosphatase and tensin homolog) is a target of the miR-17-92 cluster and is a crucial mediator of miR-17-92-induced EC proliferation. However, the angiogenic transcriptional program is reduced when miR-17-92 is inhibited. CONCLUSIONS: Taken together, our results indicate that VEGF-induced miR-17-92 cluster expression contributes to the angiogenic switch of ECs and participates in the regulation of angiogenesis.


Subject(s)
Endothelium, Vascular/metabolism , MAP Kinase Signaling System/physiology , MicroRNAs/biosynthesis , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Endothelium, Vascular/drug effects , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects
13.
Proc Natl Acad Sci U S A ; 112(41): 12812-7, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26417068

ABSTRACT

The contribution of endothelial-derived miR-17∼92 to ischemia-induced arteriogenesis has not been investigated in an in vivo model. In the present study, we demonstrate a critical role for the endothelial-derived miR-17∼92 cluster in shaping physiological and ischemia-triggered arteriogenesis. Endothelial-specific deletion of miR-17∼92 results in an increase in collateral density limbs and hearts and in ischemic limbs compared with control mice, and consequently improves blood flow recovery. Individual cluster components positively or negatively regulate endothelial cell (EC) functions in vitro, and, remarkably, ECs lacking the cluster spontaneously form cords in a manner rescued by miR-17a, -18a, and -19a. Using both in vitro and in vivo analyses, we identified FZD4 and LRP6 as targets of miR-19a/b. Both of these targets were up-regulated in 17∼92 KO ECs compared with control ECs, and both were shown to be targeted by miR-19 using luciferase assays. We demonstrate that miR-19a negatively regulates FZD4, its coreceptor LRP6, and WNT signaling, and that antagonism of miR-19a/b in aged mice improves blood flow recovery after ischemia and reduces repression of these targets. Collectively, these data provide insights into miRNA regulation of arterialization and highlight the importance of vascular WNT signaling in maintaining arterial blood flow.


Subject(s)
Frizzled Receptors/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , MicroRNAs/metabolism , Multigene Family/physiology , Neovascularization, Physiologic/physiology , Wnt Signaling Pathway/physiology , Animals , Frizzled Receptors/genetics , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Mice , Mice, Knockout , MicroRNAs/genetics
14.
Proc Natl Acad Sci U S A ; 111(35): 12865-70, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136137

ABSTRACT

The PI3K/Akt pathway is necessary for several key endothelial cell (EC) functions, including cell growth, migration, survival, and vascular tone. However, existing literature supports the idea that Akt can be either pro- or antiangiogenic, possibly due to compensation by multiple isoforms in the EC when a single isoform is deleted. Thus, biochemical, genetic, and proteomic studies were conducted to examine isoform-substrate specificity for Akt1 vs. Akt2. In vitro, Akt1 preferentially phosphorylates endothelial nitric oxide synthase (eNOS) and promotes NO release, whereas nonphysiological overexpression of Akt2 can bypass the loss of Akt1. Conditional deletion of Akt1 in the EC, in the absence or presence of Akt2, retards retinal angiogenesis, implying that Akt1 exerts a nonredundant function during physiological angiogenesis. Finally, proteomic analysis of Akt substrates isolated from Akt1- or Akt2-deficient ECs documents that phosphorylation of multiple Akt substrates regulating angiogenic signaling is reduced in Akt1-deficient, but not Akt2-deficient, ECs, including eNOS and Forkhead box proteins. Therefore, Akt1 promotes angiogenesis largely due to phosphorylation and regulation of important downstream effectors that promote aspects of angiogenic signaling.


Subject(s)
Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins c-akt/metabolism , Retinal Vessels/metabolism , Animals , Cell Line, Transformed , Lung/blood supply , Lung/cytology , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Nitrogen Mustard Compounds/metabolism , Phosphorylation/physiology , Proteomics , Proto-Oncogene Proteins c-akt/genetics , Retina/pathology , Retinal Vessels/pathology , Signal Transduction/physiology , Substrate Specificity
15.
Development ; 141(7): 1465-72, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24598168

ABSTRACT

Here we show that dynamin 2 (Dnm2) is essential for angiogenesis in vitro and in vivo. In cultured endothelial cells lacking Dnm2, vascular endothelial growth factor (VEGF) signaling and receptor levels are augmented whereas cell migration and morphogenesis are impaired. Mechanistically, the loss of Dnm2 increases focal adhesion size and the surface levels of multiple integrins and reduces the activation state of ß1 integrin. In vivo, the constitutive or inducible loss of Dnm2 in endothelium impairs branching morphogenesis and promotes the accumulation of ß1 integrin at sites of failed angiogenic sprouting. Collectively, our data show that Dnm2 uncouples VEGF signaling from function and coordinates the endocytic turnover of integrins in a manner that is crucially important for angiogenesis in vitro and in vivo.


Subject(s)
Blood Vessels/embryology , Dynamin II/physiology , Endocytosis/genetics , Integrins/metabolism , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor A/physiology , Animals , Animals, Newborn , Blood Vessels/growth & development , Cells, Cultured , Dynamin II/genetics , Embryo, Mammalian , Female , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Mice , Mice, Transgenic , Signal Transduction/physiology
16.
Nature ; 491(7424): 473-7, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23123858

ABSTRACT

Models of unregulated nitric oxide (NO) diffusion do not consistently account for the biochemistry of NO synthase (NOS)-dependent signalling in many cell systems. For example, endothelial NOS controls blood pressure, blood flow and oxygen delivery through its effect on vascular smooth muscle tone, but the regulation of these processes is not adequately explained by simple NO diffusion from endothelium to smooth muscle. Here we report a new model for the regulation of NO signalling by demonstrating that haemoglobin (Hb) α (encoded by the HBA1 and HBA2 genes in humans) is expressed in human and mouse arterial endothelial cells and enriched at the myoendothelial junction, where it regulates the effects of NO on vascular reactivity. Notably, this function is unique to Hb α and is abrogated by its genetic depletion. Mechanistically, endothelial Hb α haem iron in the Fe(3+) state permits NO signalling, and this signalling is shut off when Hb α is reduced to the Fe(2+) state by endothelial cytochrome b5 reductase 3 (CYB5R3, also known as diaphorase 1). Genetic and pharmacological inhibition of CYB5R3 increases NO bioactivity in small arteries. These data reveal a new mechanism by which the regulation of the intracellular Hb α oxidation state controls NOS signalling in non-erythroid cells. This model may be relevant to haem-containing globins in a broad range of NOS-containing somatic cells.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Hemoglobins/metabolism , Nitric Oxide/metabolism , Peptide Fragments/metabolism , Signal Transduction , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Cells, Cultured , Diffusion , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hemoglobins/genetics , Humans , Iron/chemistry , Mice , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Peptide Fragments/genetics , Phenylephrine/pharmacology
17.
Arterioscler Thromb Vasc Biol ; 31(11): 2665-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21903947

ABSTRACT

OBJECTIVE: Calcineurin (Cn) and the nuclear factor of activated T cells (NFAT) family of transcription factors are critical in vascular smooth muscle cell (SMC) development and pathology. Here, we used a genomics approach to identify and validate NFAT gene targets activated during platelet-derived growth factor-BB (PDGF-BB)-induced SMC phenotypic modulation. METHODS AND RESULTS: Genome-wide expression arrays were used to identify genes both (1) differentially activated in response to PDGF-BB and (2) whose differential expression was reduced by both the Cn inhibitor cyclosporin A and the NFAT inhibitor A-285222. The 20 most pharmacologically sensitive genes were validated by quantitative reverse transcription-polymerase chain reaction analysis of PDGF-BB-stimulated SMCs in the presence of Cn/NFAT inhibitors, including the VIVIT peptide. In all experiments, protein C receptor (PROCR) gene activation was reduced. We showed that PROCR expression was virtually absent in untreated, quiescent SMCs. PDGF-BB stimulation, however, induced significant PROCR promoter activation and downstream protein expression in a Cn/NFAT-dependent manner. Mutation of a species-conserved, NFAT binding motif significantly attenuated PDGF-BB-induced PROCR promoter activity, thereby distinguishing NFAT as the first PROCR transcriptional activator to date. Moreover, SMC PROCR expression was upregulated in the neointima as early as 7 days following acute vascular injury in rat carotid arteries. CONCLUSION: We hereby report PROCR as a novel, NFAT-dependent gene that may be implicated in vascular restenosis and consequent inward remodeling.


Subject(s)
Blood Coagulation Factors/genetics , Calcineurin/genetics , Genome/genetics , Muscle, Smooth, Vascular/pathology , NFATC Transcription Factors/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Receptors, Cell Surface/genetics , Animals , Base Sequence , Becaplermin , Blood Coagulation Factors/metabolism , Calcineurin/metabolism , Calcineurin Inhibitors , Carotid Artery Injuries/metabolism , Catheterization/adverse effects , Cells, Cultured , Cyclosporine/pharmacology , Gene Expression Regulation/drug effects , Models, Animal , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/metabolism , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Pyrazoles/pharmacology , Rats , Receptors, Cell Surface/metabolism
18.
Circ Res ; 107(6): 737-46, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20651288

ABSTRACT

RATIONALE: Macrophages change their phenotype and biological functions depending on the microenvironment. In atherosclerosis, oxidative tissue damage accompanies chronic inflammation; however, macrophage phenotypic changes in response to oxidatively modified molecules are not known. OBJECTIVE: To examine macrophage phenotypic changes in response to oxidized phospholipids that are present in atherosclerotic lesions. METHODS AND RESULTS: We show that oxidized phospholipid-treated murine macrophages develop into a novel phenotype (Mox) that is strikingly different from the conventional M1 and M2 macrophage phenotypes. Compared to M1 and M2, Mox macrophages show a different gene expression pattern, as well as decreased phagocytotic and chemotactic capacity. Treatment with oxidized phospholipids induces both M1 and M2 macrophages to switch to the Mox phenotype. Whole-genome expression array analysis and subsequent gene ontology clustering revealed that the Mox phenotype was characterized by abundant overrepresentation of Nrf2-mediated expression of redox-regulatory genes. In macrophages isolated from Nrf2(-/-) mice, oxidized phospholipid-induced gene expression and regulation of redox status were compromised. Moreover, we found that Mox macrophages comprise 30% of all macrophages in advanced atherosclerotic lesions of low-density lipoprotein receptor knockout (LDLR(-/-)) mice. CONCLUSIONS: Together, we identify Nrf2 as a key regulator in the formation of a novel macrophage phenotype (Mox) that develops in response to oxidative tissue damage. The unique biological properties of Mox macrophages suggest this phenotype may play an important role in atherosclerotic lesion development as well as in other settings of chronic inflammation.


Subject(s)
Atherosclerosis/metabolism , Immunophenotyping , Macrophages/physiology , NF-E2-Related Factor 2/physiology , Phospholipids/physiology , Animals , Atherosclerosis/genetics , Cells, Cultured , Female , Macrophages/classification , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Phospholipids/metabolism
19.
Hum Mol Genet ; 19(3): 468-79, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19926569

ABSTRACT

Vascular smooth muscle cells (SMCs) display remarkable phenotypic plasticity in response to environmental cues. The nuclear factor of activated T-cells (NFAT) family of transcription factors plays a critical role in vascular pathology. However, known functional NFAT gene targets in vascular SMCs are currently limited. Publicly available whole-genome expression array data sets were analyzed to identify differentially expressed genes in human, mouse and rat SMCs. Comparison between vehicle and phenotypic modulatory stimuli identified 63 species-conserved, upregulated genes. Integration of the 63 upregulated genes with an in silico NFAT-ome (a species-conserved list of gene promoters containing at least one NFAT binding site) identified 18 putative NFAT-dependent genes. Further intersection of these 18 potential NFAT target genes with a mouse in vivo vascular injury microarray identified four putative NFAT-dependent, injury-responsive genes. In vitro validations substantiated the NFAT-dependent role of Cyclooxygenase 2 (COX2/PTGS2) in SMC phenotypic modulation and uncovered Down Syndrome Candidate Region 1 (DSCR1/RCAN1) as a novel NFAT target gene in SMCs. We show that induction of DSCR1 inhibits calcineurin/NFAT signaling through a negative feedback mechanism; DSCR1 overexpression attenuates NFAT transcriptional activity and COX2 protein expression, whereas knockdown of endogenous DSCR1 enhances NFAT transcriptional activity. Our integrative genomics approach illustrates how the combination of publicly available gene expression arrays, computational databases and empirical research methods can answer specific questions in any cell type for a transcriptional network of interest. Herein, we report DSCR1 as a novel NFAT-dependent, injury-inducible, early gene that may serve to negatively regulate SMC phenotypic switching.


Subject(s)
Genomics , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , NFATC Transcription Factors/metabolism , Animals , Calcium-Binding Proteins , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA-Binding Proteins , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , NFATC Transcription Factors/genetics , Protein Binding , Rats
20.
J Biol Chem ; 284(48): 33671-82, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19801679

ABSTRACT

Large conductance calcium-activated potassium (MaxiK) channels play a pivotal role in maintaining normal arterial tone by regulating the excitation-contraction coupling process. MaxiK channels comprise alpha and beta subunits encoded by Kcnma and the cell-restricted Kcnmb genes, respectively. Although the functionality of MaxiK channel subunits has been well studied, the molecular regulation of their transcription and modulation in smooth muscle cells (SMCs) is incomplete. Using several model systems, we demonstrate down-regulation of Kcnmb1 mRNA upon SMC phenotypic modulation in vitro and in vivo. As part of a broad effort to define all functional CArG elements in the genome (i.e. the CArGome), we discovered two conserved CArG boxes located in the proximal promoter and first intron of the human KCNMB1 gene. Gel shift and chromatin immunoprecipitation assays confirmed serum response factor (SRF) binding to both CArG elements. A luciferase assay showed myocardin (MYOCD)-mediated transactivation of the KCNMB1 promoter in a CArG element-dependent manner. In vivo analysis of the human KCNMB1 promoter disclosed activity in embryonic heart and aortic SMCs; mutation of both conserved CArG elements completely abolished in vivo promoter activity. Forced expression of MYOCD increased Kcnmb1 expression in a variety of rodent and human non-SMC lines with no effect on expression of the Kcnma1 subunit. Conversely, knockdown of Srf resulted in decreases of endogenous Kcnmb1. Functional studies demonstrated MYOCD-induced, iberiotoxin-sensitive potassium currents in porcine coronary SMCs. These results reveal the first ion channel subunit as a direct target of SRF-MYOCD transactivation, providing further insight into the role of MYOCD as a master regulator of the SMC contractile phenotype.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , Animals , Blotting, Western , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Female , Gene Expression Regulation , HeLa Cells , Humans , In Situ Hybridization , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Protein Binding , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...