Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Diabetes ; 73(2): 225-236, 2024 Feb 01.
Article En | MEDLINE | ID: mdl-37976214

Endothelial cells (EC) play essential roles in retinal vascular homeostasis. This study aimed to characterize retinal EC heterogeneity and functional diversity using single-cell RNA sequencing. Systematic analysis of cellular compositions and cell-cell interaction networks identified a unique EC cluster with high inflammatory gene expression in diabetic retina; sphingolipid metabolism is a prominent aspect correlated with changes in retinal function. Among sphingolipid-related genes, alkaline ceramidase 2 (ACER2) showed the most significant increase. Plasma samples of patients with nonproliferative diabetic retinopathy (NPDR) with diabetic macular edema (DME) or without DME (NDME) and active proliferative DR (PDR) were collected for mass spectrometry analysis. Metabolomic profiling revealed that the ceramide levels were significantly elevated in NPDR-NDME/DME and further increased in active PDR compared with control patients. In vitro analyses showed that ACER2 overexpression retarded endothelial barrier breakdown induced by ceramide, while silencing of ACER2 further disrupted the injury. Moreover, intravitreal injection of the recombinant ACER2 adeno-associated virus rescued diabetes-induced vessel leakiness, inflammatory response, and neurovascular disease in diabetic mouse models. Together, this study revealed a new diabetes-specific retinal EC population and a negative feedback regulation pathway that reduces ceramide content and endothelial dysfunction by upregulating ACER2 expression. These findings provide insights into cell-type targeted interventions for diabetic retinopathy.


Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Animals , Mice , Humans , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Retina/metabolism , Ceramides , Sphingolipids
2.
Circ Res ; 131(9): 748-764, 2022 10 14.
Article En | MEDLINE | ID: mdl-36164986

BACKGROUND: Atherosclerosis occurs mainly at arterial branching points exposed to disturbed blood flow. How MST1 (mammalian sterile 20-like kinase 1), the primary kinase in the mechanosensitive Hippo pathway modulates disturbed flow induced endothelial cells (ECs) activation and atherosclerosis remains unclear. METHODS: To assess the role of MST1 in vivo, mice with EC-specific Mst1 deficiency on ApoE-/- background (Mst1iECKOApoE-/-) were used in an atherosclerosis model generated by carotid artery ligation. Mass spectrometry, immunoprecipitation, proximity ligation assay, and dye uptake assay were used to identify the functional substrate of MST1. Human umbilical vein endothelial cells and human aortic endothelial cells were subjected to oscillatory shear stress that mimic disturbed flow in experiments conducted in vitro. RESULTS: We found that the phosphorylation of endothelial MST1 was significantly inhibited in oscillatory shear stress-exposed regions of human and mouse arteries and ECs. Ectopic lenti-mediated overexpression of wild-type MST1, but not a kinase-deficient mutant of MST1, reversed disturbed flow-caused EC activation and atherosclerosis in EC-specific Mst1 deficiency on ApoE-/- background (Mst1iECKOApoE-/-). Inhibition of MST1 by oscillatory shear stress led to reduced phosphorylation of Cx43 (connexin 43) at Ser255, the Cx43 hemichannel open, EC activation, and atherosclerosis, which were blocked by TAT-GAP19, a Cx43 hemichannel inhibitory peptide. Mass spectrometry studies identified that Filamin B fueled the translocation of Cx43 to lipid rafts for further hemichannel open. Finally, lenti-mediated overexpression of the Cx43S255 mutant into glutamate to mimic phosphorylation blunted disturbed flow-induced EC activation, thereby inhibiting the atherogenesis in both ApoE-/- and Mst1 iECKOApoE-/- mice. CONCLUSIONS: Our study reveals that inhibition of the MST1-Cx43 axis is an essential driver of oscillatory shear stress-induced endothelial dysfunction and atherosclerosis, which provides a new therapeutic target for the treatment of atherosclerosis.


Atherosclerosis , Connexin 43 , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Cells, Cultured , Connexin 43/metabolism , Filamins/metabolism , Glutamates/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mammals , Mice , Stress, Mechanical
3.
Hypertension ; 79(3): 549-561, 2022 03.
Article En | MEDLINE | ID: mdl-34983194

BACKGROUND: Vascular endothelial cells are critical for maintaining blood pressure (BP) by releasing biologically active molecules, such as nitric oxide. A non-endothelial cell resident matricellular protein, COMP (cartilage oligomeric matrix protein), plays a pivotal role in maintaining cardiovascular homeostasis, but little is known about its regulatory effect on BP. METHODS: Mice were infused with AngII (angiotensin II; 450 ng/kg per minute) for 3 days via an osmotic minipump, and BP was monitored by a tail-cuff system. Second-order mesenteric arteries were isolated from mice for microvascular tension measurement. Nitric oxide was detected by an electron paramagnetic resonance technique. Small-interfering RNA transfection, co-immunoprecipitation, bioluminescence resonance energy transfer assays, and patch-clamp electrophysiology experiments were used for further detailed mechanism investigation. RESULTS: COMP-/- mice displayed elevated BP and impaired acetylcholine-induced endothelium-dependent relaxation compared with wild-type mice with or without AngII. Inhibition of eNOS (endothelial nitric oxide synthase) abolished the difference in endothelium-dependent relaxation between wild-type and COMP-/- mice. Furthermore, COMP directly interacted with the C-terminus of Piezo1 via its C-terminus and activated the endogenous Piezo1 currents, which induced intracellular Ca2+ influx, Ca2+/calmodulin-dependent protein kinase type II and eNOS activation, and nitric oxide production. The Piezo1 activator, Yoda1, reduced the difference in endothelium-dependent relaxation and BP in wild-type and COMP-/- mice. Moreover, COMP overexpression increased eNOS activation and improved endothelium-dependent relaxation and BP. CONCLUSIONS: Our study demonstrated that COMP is a novel Piezo1 regulator that plays a protective role in BP regulation by increasing cellular Ca2+ influx, eNOS activity, and nitric oxide production.


Blood Pressure/physiology , Cartilage Oligomeric Matrix Protein/metabolism , Endothelium, Vascular/metabolism , Ion Channels/metabolism , Acetylcholine/pharmacology , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Calcium/metabolism , Cartilage Oligomeric Matrix Protein/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Ion Channels/genetics , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Vasodilation/drug effects
4.
J Mol Cell Cardiol ; 162: 158-165, 2022 01.
Article En | MEDLINE | ID: mdl-34547259

Cells are constantly exposed to various mechanical forces, including hydrostatic pressure, cyclic stretch, fluid shear stress, and extracellular matrix stiffness. Mechanical cues can be translated into the cell-specific transcriptional process by a cellular mechanic-transducer. Evidence suggests that mechanical signals assist activated intracellular signal transduction pathways and the relative phenotypic adaptation to coordinate cell behavior and disease appropriately. The Hippo/yes-associated protein (YAP) signaling pathway is regulated in response to numerous mechanical stimuli. It plays an important role in the mechanotransduction mechanism, which converts mechanical forces to cascades of molecular signaling to modulate gene expression. This review summarizes the recent findings relevant to the Hippo/YAP pathway-based mechanotransduction in cell behavior and maintaining blood vessels, as well as cardiovascular disease.


Adaptor Proteins, Signal Transducing , Mechanotransduction, Cellular , Adaptor Proteins, Signal Transducing/metabolism , Homeostasis , Signal Transduction/genetics , YAP-Signaling Proteins
6.
Angew Chem Int Ed Engl ; 60(11): 5718-5722, 2021 Mar 08.
Article En | MEDLINE | ID: mdl-33320989

NH4 + ions as charge carriers show potential for aqueous rechargeable batteries. Studied here for the first time is the NH4 + -storage chemistry using electrodeposited manganese oxide (MnOx ). MnOx experiences morphology and phase transformations during charge/discharge in dilute ammonium acetate (NH4 Ac) electrolyte. The NH4 Ac concentration plays an important role in NH4 + storage for MnOx . The transformed MnOx with a layered structure delivers a high specific capacity (176 mAh g-1 ) at a current density of 0.5 A g-1 , and exhibits good cycling stability over 10 000 cycles in 0.5 M NH4 Ac, outperforming the state-of-the-art NH4 + hosting materials. Experimental results suggest a solid-solution behavior associated with NH4 + migration in layered MnOx . Spectroscopy studies and theoretical calculations show that the reversible NH4 + insertion/deinsertion is accompanied by hydrogen-bond formation/breaking between NH4 + and the MnOx layers. These findings provide a new prototype (i.e., layered MnOx ) for NH4 + -based energy storage and contributes to the fundamental understanding of the NH4 + -storage mechanism for metal oxides.

7.
Matrix Biol ; 95: 32-51, 2021 01.
Article En | MEDLINE | ID: mdl-33068727

Disturbed flow leads to increased inflammatory responses of endothelial cells (ECs) prone to atherogenic state. Currently, little is known about the physiological mechanisms protecting vasculature against disturbed flow-activated ECs leading to atherosclerosis. Understanding the protective mediators involved in EC activation could provide novel therapeutic strategies for atherosclerosis. The extracellular matrix microenvironment profoundly regulates cellular homeostasis. A non-EC resident ECM protein, cartilage oligomeric matrix protein (COMP), has diverse protective roles in the cardiovascular system. To determine whether COMP could protect against disturbed flow-activated EC and atherosclerosis, we compared oscillatory shear stress (OSS) induced EC activation coated with various ECM proteins. Purified COMP inhibited EC activation caused by OSS. EC activation was upregulated in the aortic arch where the flow is disturbed in COMP-/- mice as compared with wild-type mice under physiological conditions or pathologically in partially ligated mouse carotid arteries. Mechanistically, co-immunoprecipitation, mammalian two-hybrid and FRET assay results suggest that COMP bound directly to integrin α5 via its C-terminus. We next synthesized a COMP-derived peptidomimetics (CCPep24) mimicking a specific COMP-integrin α5 interaction and found that CCPep24 protected against EC activation and atherogenesis in vivo. This study extends our current understanding of how ECM and flow coordinately fine-tune EC homeostasis and reveals the potential therapeutic effect of COMP or COMP-derived peptidomimetics on blocking aberrant integrin α5 activation, inflammatory EC activation and atherosclerosis pathogenesis.


Atherosclerosis/genetics , Cartilage Oligomeric Matrix Protein/genetics , Integrin alpha5/genetics , Animals , Atherosclerosis/pathology , Cardiovascular System/metabolism , Cardiovascular System/pathology , Cellular Microenvironment/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Matrix/genetics , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Peptidomimetics/pharmacology , Protein Interaction Maps/genetics
8.
Aging (Albany NY) ; 12(21): 22019-22045, 2020 11 07.
Article En | MEDLINE | ID: mdl-33186123

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder worldwide. Multiple metabolic disorders, such as hyperlipidemia, hyperglycemia, insulin resistance and obesity, have been reportedly associated with NAFLD, but little is known about the detailed mechanisms. METHODS AND RESULTS: Here, we explored the effects of multiple metabolic disorders, especially hyperglycemia on lipid accumulation in liver using several well-established animal models. We found that liver lipid deposition was increased in both type 1 diabetes and high-fat diet (HFD) induced hyperlipidemia models, suggesting that either hyperglycemia or hyperlipidemia alone or together was able to trigger NAFLD. Moreover, we tested whether miR-320, a miRNA promoting lipid accumulation in heart revealed by our previous study, also participated in NAFLD. Though miR-320 treatment further increased liver lipid deposition in type 1 diabetes and HFD-feeding mice, it showed no effect in leptin-receptor deficient db/db mice. Interestingly, miR-320 affected different target genes in cytosol and nucleus, respectively, which collectively led to liver lipid overload. CONCLUSIONS: Our findings illustrated the complex roles of miRNAs in subcellular fractions including nucleus and cytoplasm, which may lead to new insights into the mechanisms and treatment strategies for NAFLD in the future.


Hyperglycemia/metabolism , Hyperlipidemias/metabolism , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Diabetes Mellitus, Experimental , Diet, High-Fat/adverse effects , Disease Models, Animal , Hyperglycemia/complications , Hyperlipidemias/complications , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
9.
Cell Rep ; 32(5): 107990, 2020 08 04.
Article En | MEDLINE | ID: mdl-32755583

The Hippo/Yes-associated protein (YAP) pathway has pivotal roles in innate immune responses against pathogens in macrophages. However, the role of YAP in macrophages during atherosclerosis and its mechanism of YAP activation remain unknown. Here, we find that YAP overexpression in myeloid cells aggravates atherosclerotic lesion size and infiltration of macrophages, whereas YAP deficiency reduces atherosclerotic plaque. Tumor necrosis factor receptor-associated factor 6 (TRAF6), a downstream effector of interleukin-1ß (IL-1ß), triggers YAP ubiquitination at K252, which interrupts the interaction between YAP and angiomotin and results in enhanced YAP nuclear translocation. The recombinant IL-1 receptor antagonist anakinra reduces atherosclerotic lesion formation, which is abrogated by YAP overexpression. YAP level is increased in human and mouse atherosclerotic vessels, and plasma IL-1ß level in patients with STEMI is correlated with YAP protein level in peripheral blood mononuclear cells. These findings elucidate a mechanism of YAP activation, which might be a therapeutic target for atherosclerosis.


Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Lysine/metabolism , Macrophages/metabolism , Transcription Factors/metabolism , Ubiquitination , Animals , Cell Line , Cell Movement , Chemokines/metabolism , Humans , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Monocytes/metabolism , Plaque, Atherosclerotic/metabolism , Protein Binding , Protein Stability , Protein Transport , TNF Receptor-Associated Factor 6/metabolism , Up-Regulation , YAP-Signaling Proteins
10.
Circ Res ; 127(8): 1074-1090, 2020 09 25.
Article En | MEDLINE | ID: mdl-32673515

RATIONALE: Atherosclerosis preferentially occurs at specific sites of the vasculature where endothelial cells (ECs) are exposed to disturbed blood flow. Translocation of integrin α5 to lipid rafts promotes integrin activation and ligation, which is critical for oscillatory shear stress (OSS)-induced EC activation. However, the underlying mechanism of OSS promoted integrin α5 lipid raft translocation has remained largely unknown. OBJECTIVE: The objective of this study was to specify the mechanotransduction mechanism of OSS-induced integrin α5 translocation and subsequent EC activation. METHODS AND RESULTS: Mass spectrometry studies identified endothelial ANXA2 (annexin A2) as a potential carrier allowing integrin α5ß1 to traffic in response to OSS. Interference by siRNA of AnxA2 in ECs greatly decreased OSS-induced integrin α5ß1 translocation to lipid rafts, EC activation, and monocyte adhesion. Pharmacological and genetic inhibition of PTP1B (protein tyrosine phosphatase 1B) blunted OSS-induced integrin α5ß1 activation, which is dependent on Piezo1-mediated calcium influx in ECs. Furthermore, ANXA2 was identified as a direct substrate of activated PTP1B by mass spectrometry. Using bioluminescence resonance energy transfer assay, PTP1B-dephosphorylated ANXA2 at Y24 was found to lead to conformational freedom of the C-terminal core domain from the N-terminal domain of ANXA2. Immunoprecipitation assays showed that this unmasked ANXA2-C-terminal core domain specifically binds to an integrin α5 nonconserved cytoplasmic domain but not ß1. Importantly, ectopic lentiviral overexpression of an ANXA2Y24F mutant increased and shRNA against Ptp1B decreased integrin α5ß1 ligation, inflammatory signaling, and progression of plaques at atheroprone sites in apolipoprotein E (ApoE)-/- mice. However, the antiatherosclerotic effect of Ptp1B shRNA was abolished in AnxA2-/-ApoE-/- mice. CONCLUSIONS: Our data elucidate a novel endothelial mechanotransduction molecular mechanism linking atheroprone flow and activation of integrin α5ß1, thereby identifying a class of potential therapeutic targets for atherosclerosis. Graphic Abstract: An graphic abstract is available for this article.


Annexin A2/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Integrin alpha5/metabolism , Integrin alpha5beta1/metabolism , Membrane Microdomains/metabolism , Animals , Annexin A2/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Disease Models, Animal , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Integrin alpha5/genetics , Integrin alpha5beta1/genetics , Integrins , Ion Channels/metabolism , Male , Mechanotransduction, Cellular , Membrane Microdomains/pathology , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Protein Interaction Domains and Motifs , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Regional Blood Flow , Stress, Mechanical , THP-1 Cells
11.
Nanomicro Lett ; 12(1): 118, 2020 May 30.
Article En | MEDLINE | ID: mdl-34138149

The article reviews the recent progress of electrochemical techniques on synthesizing nano-/microstructures as supercapacitor electrodes. With a history of more than a century, electrochemical techniques have evolved from metal plating since their inception to versatile synthesis tools for electrochemically active materials of diverse morphologies, compositions, and functions. The review begins with tutorials on the operating mechanisms of five commonly used electrochemical techniques, including cyclic voltammetry, potentiostatic deposition, galvanostatic deposition, pulse deposition, and electrophoretic deposition, followed by thorough surveys of the nano-/microstructured materials synthesized electrochemically. Specifically, representative synthesis mechanisms and the state-of-the-art electrochemical performances of exfoliated graphene, conducting polymers, metal oxides, metal sulfides, and their composites are surveyed. The article concludes with summaries of the unique merits, potential challenges, and associated opportunities of electrochemical synthesis techniques for electrode materials in supercapacitors.

12.
Hypertension ; 74(4): 936-946, 2019 10.
Article En | MEDLINE | ID: mdl-31378107

Endothelial cells regulate vascular tone by producing both relaxing and contracting factors to control the local blood flow. Hypertension is a common side effect of mTORC1 (mammalian target of rapamycin complex 1) inhibitors. However, the role of endothelial mTORC1 in hypertension remains elusive. The present study aimed to determine the role of endothelial mTORC1 in Ang II (angiotensin II)-induced hypertension and the underlying mechanism. Endothelial mTORC1 activity was increased by Ang II both in vitro and in vivo. Blood pressure was higher in Tie-2-Cre-mediated regulatory associated protein of mTOR (mammalian target of rapamycin; Raptor) heterozygous-deficient (Tie2Cre-RaptorKD) mice than control mice both before and after Ang II infusion. Acetylcholine-evoked endothelium-dependent relaxation of mesenteric arteries was impaired in Tie2Cre-RaptorKD mice. Treatment with indomethacin or a specific COX (cyclooxygenase)-2 inhibitor, NS-398, but not L-NG-nitroarginine methyl ester reduced endothelium-dependent relaxation in Raptorflox/- mice to a similar extent as in Tie2Cre-RaptorKD mice. Metabolomic profiling revealed that the plasma content of prostaglandin E2 was reduced in Tie2Cre-RaptorKD mice with or without Ang II infusion. In endothelial cells, reduction of the protein level of YAP (yes-associated protein) with siRNA-mediated RPTOR deficiency was autophagy dependent and transcriptionally regulated the expression of COX-2 and mPGES-1 (microsomal prostaglandin E synthase-1). Hence, overexpression of YAP in endothelial cells enhanced the mRNA and protein levels of COX-2 and mPGES-1 and reversed the endothelial dysfunction and hypertension in Tie2Cre-RaptorKD mice. The present results demonstrate that suppression of mTORC1 activity in endothelial cells reduces prostaglandin E2 production and causes hypertension by reducing YAP-mediated COX-2/mPGES-1 expression.


Adaptor Proteins, Signal Transducing/metabolism , Blood Pressure/physiology , Endothelium, Vascular/metabolism , Hypertension/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Angiotensin II , Animals , Blood Pressure/drug effects , Cell Cycle Proteins , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Hypertension/chemically induced , Indomethacin/pharmacology , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Prostaglandin-E Synthases/metabolism , Signal Transduction/drug effects , Vasodilation/drug effects , Vasodilation/physiology , YAP-Signaling Proteins
13.
J Clin Invest ; 129(3): 1167-1179, 2019 03 01.
Article En | MEDLINE | ID: mdl-30629551

Local flow patterns determine the uneven distribution of atherosclerotic lesions. This research aims to elucidate the mechanism of regulation of nuclear translocation of Yes-associated protein (YAP) under oscillatory shear stress (OSS) in the atheroprone phenotype of endothelial cells (ECs). We report here that OSS led to tyrosine phosphorylation and strong, continuous nuclear translocation of YAP in ECs that is dependent on integrin α5ß1 activation. YAP overexpression in ECs blunted the anti-atheroprone effect of an integrin α5ß1-blocking peptide (ATN161) in Apoe-/- mice. Activation of integrin α5ß1 induced tyrosine, but not serine, phosphorylation of YAP in ECs. Blockage of integrin α5ß1 with ATN161 abolished the phosphorylation of YAP at Y357 induced by OSS. Mechanistic studies showed that c-Abl inhibitor attenuated the integrin α5ß1-induced YAP tyrosine phosphorylation. Furthermore, the phosphorylation of c-Abl and YAPY357 was significantly increased in ECs in atherosclerotic vessels of mice and in human plaques versus normal vessels. Finally, bosutinib, a tyrosine kinase inhibitor, markedly reduced the level of YAPY357 and the development of atherosclerosis in Apoe-/- mice. The c-Abl/YAPY357 pathway serves as a mechanism for the activation of integrin α5ß1 and the atherogenic phenotype of ECs in response to OSS, and provides a potential therapeutic strategy for atherogenesis.


Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Cell Cycle Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Plaque, Atherosclerotic/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Blood Flow Velocity , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Mice , Mice, Knockout, ApoE , Phosphorylation , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Proto-Oncogene Proteins c-abl/genetics , Shear Strength , THP-1 Cells , Transcription Factors/genetics , YAP-Signaling Proteins
14.
J Mol Cell Biol ; 11(6): 509-520, 2019 06 01.
Article En | MEDLINE | ID: mdl-30295821

Endothelial NO synthase (eNOS) expression is regulated by a number of transcriptional and post-transcriptional mechanisms, but the effects of competing endogenous RNAs (ceRNAs) on eNOS mRNA and the underlying mechanisms are still unknown. Our bioinformatic analysis revealed three highly expressed eNOS-targeting miRNAs (miR-15b, miR-16, and miR-30b) in human endothelial cells (ECs). Among the 1103 mRNA targets of these three miRNAs, 15 mRNAs share a common disease association with eNOS. Gene expression and correlation analysis in patients with cardiovascular diseases identified insulin receptor substrate 2 (IRS2) as the most correlated eNOS-ceRNA. The expression levels of eNOS and IRS2 were coincidentally increased by application of laminar shear but reduced with eNOS or IRS2 siRNA transfection in human ECs, which was impeded by Dicer siRNA treatment. Moreover, luciferase reporter assay showed that these three miRNAs directly target the 3'UTR of eNOS and IRS2. Overexpression of these three miRNAs decreased, whereas inhibition of them increased, both mRNA and protein levels of eNOS and IRS2. Functionally, silencing eNOS suppressed the Akt signal pathway, while IRS2 knockdown reduced NO production in ECs. Thus, we identified eNOS and IRS2 as ceRNAs and revealed a novel mechanism explaining the coincidence of metabolic and cardiovascular diseases.


3' Untranslated Regions , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Insulin Receptor Substrate Proteins/biosynthesis , MicroRNAs/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Signal Transduction , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/pathology , Humans , Insulin Receptor Substrate Proteins/genetics , MicroRNAs/genetics , Nitric Oxide Synthase Type III/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
15.
Circ Res ; 122(4): 591-605, 2018 02 16.
Article En | MEDLINE | ID: mdl-29298775

RATIONALE: Angiogenesis is a complex process regulating endothelial cell (EC) functions. Emerging lines of evidence support that YAP (Yes-associated protein) plays an important role in regulating the angiogenic activity of ECs. OBJECTIVE: The objective of this study was to specify the effect of EC YAP on angiogenesis and its underlying mechanisms. METHOD AND RESULTS: In ECs, vascular endothelial growth factor reduced YAP phosphorylation time and dose dependently and increased its nuclear accumulation. Using Tie2Cre-mediated YAP transgenic mice, we found that YAP promoted angiogenesis in the postnatal retina and tumor tissues. Mass spectrometry revealed signal transducer and activator of transcription 3 (STAT3) as a potential binding partner of YAP in ECs. Western blot and immunoprecipitation assays indicated that binding with YAP prolonged interleukin 6-induced STAT3 nuclear accumulation by blocking chromosomal maintenance 1-mediated STAT3 nuclear export without affecting its phosphorylation. Moreover, angiopoietin-2 expression induced by STAT3 was enhanced by YAP overexpression in ECs. Finally, a selective STAT3 inhibitor or angiopoietin-2 blockage partly attenuated retinal angiogenesis in Tie2Cre-mediated YAP transgenic mice. CONCLUSIONS: YAP binding sustained STAT3 in the nucleus to enhance the latter's transcriptional activity and promote angiogenesis via regulation of angiopoietin-2.


Adaptor Proteins, Signal Transducing/metabolism , Neovascularization, Physiologic , Phosphoproteins/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Cell Cycle Proteins , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding , Retinal Vessels/growth & development , Retinal Vessels/metabolism , STAT3 Transcription Factor/metabolism , YAP-Signaling Proteins
16.
J Mol Cell Cardiol ; 98: 128-37, 2016 09.
Article En | MEDLINE | ID: mdl-27496380

RATIONALE: Circulating monocytes play pivotal roles in chronic inflammatory diseases. Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid, are known to have anti-inflammatory effects and are hydrolyzed by soluble epoxide hydrolase (sEH). OBJECTIVE: We aimed to investigate the effect of sEH inhibition in atherogenesis. METHODS AND RESULTS: Mice with low-density lipoprotein receptor deficiency (Ldlr(-/-)) with or without sEH inhibitor, and Ldlr/sEH double-knockout (DK) mice were fed a Western-type diet (WTD) for 6weeks to induce arteriosclerosis. Both sEH inhibition and gene depletion decreased the WTD-induced hyperlipidemia, plaque area and macrophage infiltration in mice arterial wall. Ly6C(hi) infiltration of monocytes remained similar in blood, spleen and bone marrow of DK mice, but was decreased in aortic lesions. To further assess the role of sEH or EETs in monocyte/macrophage infiltration in atherogenesis, we transplanted DK bone marrow into Ldlr(-/-) recipients, and then fed mice the WTD. Aortic lesions and Ly6C(hi) monocyte infiltration were reduced in mice with transplanted bone marrow of DK mice without diminishing the cholesterol level. Furthermore, sEH inhibition or gene depletion increased the ratio of EETs/DHETs and diminished the expression of P-selectin glycoprotein ligand 1 (PSGL-1) in mice peripheral-blood mononuclear cells. Monocyte adhesion to P-selectin and to tumor necrosis factor α-activated endothelial cells was also diminished by sEH inhibition. CONCLUSION: sEH inhibition and gene depletion attenuated atherosclerosis in mice by decreasing the infiltration of monocytes into the artery wall. EET and PSGL-1 may play pivotal roles in monocyte/macrophage infiltration and atherogenesis.


Atherosclerosis/etiology , Atherosclerosis/metabolism , Epoxide Hydrolases/antagonists & inhibitors , Monocytes/immunology , Monocytes/metabolism , Receptors, LDL/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/pathology , Biomarkers , Cell Adhesion , Collagen/metabolism , Disease Models, Animal , Gene Expression , Hyperlipidemias , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism , Lipids/blood , Macrophages , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Monocytes/pathology , P-Selectin/metabolism
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(3): 376-81, 2013 Mar.
Article Zh | MEDLINE | ID: mdl-23529235

OBJECTIVE: To explore whether strontium ranelate (Sr) promotes osteoblast lineage differentiation of rat bone mesenchymal stem cells (BMSCs) through the bone morphogenetic protein-2 (BMP-2)/Smad signaling pathway. METHODS: Cultured rat BMSCs were exposed to different concentrations of Sr, noggin (an inhibitor of BMP-2) or Smad1 siRNA. The activity of alkaline phosphatase (ALP) in the exposed cells was detected by colorimetry, and the formation of mineralized nodules was observed with alizarin red staining. The expressions of phosphorylated (p) Smad1/5/8 and Runt-related transcription factor 2 (Runx2) in the cells were detected by Western blotting. RESULTS: Exposure to Sr at 0.1 to 10 mmol/L for 1 h markedly increased the expression of p-Smad1/5/8 in the BMSCs, and the increment was the most obvious following 1 mmol/L Sr exposure. Preconditioning with 100 ng/ml noggin for 2 h inhibited Sr-induced up-regulation of p-Smad1/5/8 expressions. Exposure of the cells to 0.1 to 5 mmol/L Sr for 6 h significantly enhanced Runx2 expression, and the peak enhancement occurred following 1 mmol/L Sr exposure. Transfection of the BMSCs with Smad1 siRNA decreased the basal level of Smad1/5/8 protein expression, and also inhibited Sr-induced up-regulation of p-Smad1/5/8 and Runx2 expressions as well as Sr-induced enhancement of ALP activity and formation of mineralized nodules. CONCLUSION: The BMP-2/Smad pathway is involved in Sr-induced osteoblast differentiation of rat BMSCs.


Cell Differentiation/drug effects , Mesenchymal Stem Cells/cytology , Osteogenesis , Signal Transduction , Thiophenes/pharmacology , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/cytology , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured , Rats , Rats, Sprague-Dawley , Smad1 Protein/metabolism , Strontium/pharmacology
...