Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Biomaterials ; 309: 122600, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38718614

ABSTRACT

Engineering vascularized tissues remains a promising approach for treating ischemic cardiovascular diseases. The availability of 3D-bioprinted vascular grafts that induce therapeutic angiogenesis can help avoid necrosis and excision of ischemic tissues. Here, using a combination of living cells and biodegradable hydrogels, we fabricated 3D-printed biocompatible proangiogenic patches from endothelial cell-laden photo-crosslinked gelatin (EC-PCG) bioink and smooth muscle cell-encapsulated polyurethane (SMC-PU) bioink. Implantation of 3D-bioprinted proangiogenic patches in a mouse model showed that EC-PCG served as an angiogenic capillary bed, whereas patterned SMC-PU increased the density of microvessels. Moreover, the assembled patterns between EC-PCG and SMC-PU induced the geometrically guided generation of microvessels with blood perfusion. In a rodent model of hindlimb ischemia, the vascular patches rescued blood flow to distal tissues, prevented toe/foot necrosis, promoted muscle remodeling, and increased the capillary density, thereby improving the heat-escape behavior of ischemic animals. Thus, our 3D-printed vascular cell-laden bioinks constitute efficient and scalable biomaterials that facilitate the engineering of vascular patches capable of directing therapeutic angiogenesis for treating ischemic vascular diseases.


Subject(s)
Gelatin , Hydrogels , Ischemia , Neovascularization, Physiologic , Polyurethanes , Printing, Three-Dimensional , Animals , Gelatin/chemistry , Polyurethanes/chemistry , Hydrogels/chemistry , Ischemia/therapy , Neovascularization, Physiologic/drug effects , Mice , Humans , Myocytes, Smooth Muscle/cytology , Cross-Linking Reagents/chemistry , Human Umbilical Vein Endothelial Cells , Hindlimb/blood supply , Hindlimb/pathology , Male , Tissue Engineering/methods , Bioprinting/methods
2.
Proc Natl Acad Sci U S A ; 121(5): e2318718121, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38252820

ABSTRACT

Several compounds have been used for atherosclerosis treatment, including clinical trials; however, no anti-atherosclerotic drugs based on hemodynamic force-mediated atherogenesis have been discovered. Our previous studies demonstrated that "small mothers against decapentaplegic homolog 1/5" (Smad1/5) is a convergent signaling molecule for chemical [e.g., bone morphogenetic proteins (BMPs)] and mechanical (e.g., disturbed flow) stimulations and hence may serve as a promising hemodynamic-based target for anti-atherosclerosis drug development. The goal of this study was to develop a high-throughput screening (HTS) platform to identify potential compounds that can inhibit disturbed flow- and BMP-induced Smad1/5 activation and atherosclerosis. Through HTS using a Smad1/5 downstream target inhibitor of DNA binding 1 (Id-1) as a luciferase reporter, we demonstrated that KU-55933 and Apicidin suppressed Id-1 expression in AD-293 cells. KU-55933 (10 µM), Apicidin (10 µM), and the combination of half doses of each [1/2(K + A)] inhibited disturbed flow- and BMP4-induced Smad1/5 activation in human vascular endothelial cells (ECs). KU-55933, Apicidin, and 1/2(K + A) treatments caused 50.6%, 47.4%, and 73.3% inhibitions of EC proliferation induced by disturbed flow, respectively, whereas EC inflammation was only suppressed by KU-55933 and 1/2(K + A), but not Apicidin alone. Administrations of KU-55933 and 1/2(K + A) to apolipoprotein E-deficient mice inhibited Smad1/5 activation in ECs in athero-susceptible regions, thereby suppressing endothelial proliferation and inflammation, with the attenuation of atherosclerotic lesions in these mice. A unique drug screening platform has been developed to demonstrate that KU-55933 and its combination with Apicidin are promising therapeutic compounds for atherosclerosis based on hemodynamic considerations.


Subject(s)
Atherosclerosis , Endothelial Cells , Morpholines , Pyrones , Humans , Animals , Mice , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Atherosclerosis/drug therapy , Hemodynamics , Inflammation
3.
J Adv Res ; 43: 187-203, 2023 01.
Article in English | MEDLINE | ID: mdl-36585108

ABSTRACT

INTRODUCTION: Atherosclerotic complications represent the leading cause of cardiovascular mortality globally. Dysfunction of endothelial cells (ECs) often initiates the pathological events in atherosclerosis. OBJECTIVES: In this study, we sought to investigate the transcriptional profile of atherosclerotic aortae, identify novel regulator in dysfunctional ECs and hence provide mechanistic insights into atherosclerotic progression. METHODS: We applied single-cell RNA sequencing (scRNA-seq) on aortic cells from Western diet-fed apolipoprotein E-deficient (ApoE-/-) mice to explore the transcriptional landscape and heterogeneity of dysfunctional ECs. In vivo validation of SOX4 upregulation in ECs were performed in atherosclerotic tissues, including mouse aortic tissues, human coronary arteries, and human renal arteries. Single-cell analysis on human aortic aneurysmal tissue was also performed. Downstream vascular abnormalities induced by EC-specific SOX4 overexpression, and upstream modulators of SOX4 were revealed by biochemical assays, immunostaining, and wire myography. Effects of shear stress on endothelial SOX4 expression was investigated by in vitro hemodynamic study. RESULTS: Among the compendium of aortic cells, mesenchymal markers in ECs were significantly enriched. Two EC subsets were subsequently distinguished, as the 'endothelial-like' and 'mesenchymal-like' subsets. Conventional assays consistently identified SOX4 as a novel atherosclerotic marker in mouse and different human arteries, additional to a cancer marker. EC-specific SOX4 overexpression promoted atherogenesis and endothelial-to-mesenchymal transition (EndoMT). Importantly, hyperlipidemia-associated cytokines and oscillatory blood flow upregulated, whereas the anti-diabetic drug metformin pharmacologically suppressed SOX4 level in ECs. CONCLUSION: Our study unravels SOX4 as a novel phenotypic regulator during endothelial dysfunction, which exacerbates atherogenesis. Our study also pinpoints hyperlipidemia-associated cytokines and oscillatory blood flow as endogenous SOX4 inducers, providing more therapeutic insights against atherosclerotic diseases.


Subject(s)
Atherosclerosis , Endothelial Cells , Humans , Mice , Animals , Endothelial Cells/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Aorta/metabolism , Cytokines/metabolism , Single-Cell Analysis , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
4.
Eur Heart J ; 44(4): 304-318, 2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36380599

ABSTRACT

BACKGROUND AND AIMS: Atherosclerosis preferentially develops in arterial branches and curvatures where vascular endothelium is exposed to disturbed flow. In this study, the effects of disturbed flow on the regulation of vascular endothelial phosphoproteins and their contribution to therapeutic application in atherogenesis were elucidated. METHODS: Porcine models, large-scale phosphoproteomics, transgenic mice, and clinical specimens were used to discover novel site-specific phosphorylation alterations induced by disturbed flow in endothelial cells (ECs). RESULTS: A large-scale phosphoproteomics analysis of native endothelium from disturbed (athero-susceptible) vs. pulsatile flow (athero-resistant) regions of porcine aortas led to the identification of a novel atherosclerosis-related phosphoprotein vinculin (VCL) with disturbed flow-induced phosphorylation at serine 721 (VCLS721p). The induction of VCLS721p was mediated by G-protein-coupled receptor kinase 2 (GRK2)S29p and resulted in an inactive form of VCL with a closed conformation, leading to the VE-cadherin/catenin complex disruption to enhance endothelial permeability and atherogenesis. The generation of novel apolipoprotein E-deficient (ApoE-/-) mice overexpressing S721-non-phosphorylatable VCL mutant in ECs confirmed the critical role of VCLS721p in promoting atherosclerosis. The administration of a GRK2 inhibitor to ApoE-/- mice suppressed plaque formation by inhibiting endothelial VCLS721p. Studies on clinical specimens from patients with coronary artery disease (CAD) revealed that endothelial VCLS721p is a critical clinicopathological biomarker for atherosclerosis progression and that serum VCLS721p level is a promising biomarker for CAD diagnosis. CONCLUSIONS: The findings of this study indicate that endothelial VCLS721p is a valuable hemodynamic-based target for clinical assessment and treatment of vascular disorders resulting from atherosclerosis.


Subject(s)
Atherosclerosis , Endothelial Cells , Vinculin , Animals , Mice , Atherosclerosis/pathology , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Mice, Knockout, ApoE , Phosphorylation , Swine , Humans
5.
Circ Res ; 129(12): 1158-1174, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34747636

ABSTRACT

RATIONALE: Disturbed flow occurring in arterial branches and curvatures induces vascular endothelial cell (EC) dysfunction and atherosclerosis. We postulated that disturbed flow plays important role in modulating phosphoprotein expression profiles to regulate endothelial functions and atherogenesis. OBJECTIVE: The goal of this study is to discover novel site-specific phosphorylation alterations induced by disturbed flow in ECs to contribute to atherosclerosis. METHODS AND RESULTS: Quantitative phosphoproteomics analysis of ECs exposed to disturbed flow with low and oscillatory shear stress (0.5±4 dynes/cm2) versus pulsatile shear stress (12±4 dynes/cm2) revealed that oscillatory shear stress induces phospho-YY1S118 (serine [S]118 phosphorylation of Yin Yang 1) in ECs. Elevated phospho-YY1S118 level in ECs was further confirmed to be present in the disturbed flow regions in experimental animals and human atherosclerotic arteries. This disturbed flow-induced EC phospho-YY1S118 is mediated by CK2α (casein kinase 2α) through its direct interaction with YY1. Yeast 2-hybrid library screening and in situ proximity ligation assays demonstrate that phospho-YY1S118 directly binds ZKSCAN4 (zinc finger with KRAB [krüppel-associated box] and SCAN [SRE-ZBP, CTfin51, AW-1 and Number 18 cDNA] domains 4) to induce promoter activity and gene expression of HDM2 (human double minute 2), which consequently induces EC proliferation through downregulation of p53 and p21CIP1. Administration of apoE-deficient (ApoE-/-) mice with CK2-specific inhibitor tetrabromocinnamic acid or atorvastatin inhibits atherosclerosis formation through downregulations of EC phospho-YY1S118 and HDM2. Generation of novel transgenic mice bearing EC-specific overexpression of S118-nonphosphorylatable mutant of YY1 in ApoE-/- mice confirms the critical role of phospho-YY1S118 in promoting atherosclerosis through EC HDM2. CONCLUSIONS: Our findings provide new insights into the mechanisms by which disturbed flow induces endothelial phospho-YY1S118 to promote atherosclerosis, thus indicating phospho-YY1S118 as a potential molecular target for atherosclerosis treatment.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , YY1 Transcription Factor/metabolism , Animals , Atherosclerosis/physiopathology , Binding Sites , Blood Circulation , Casein Kinase II/metabolism , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Rats, Sprague-Dawley , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/genetics , Zinc Fingers
6.
Cardiol Res Pract ; 2021: 1452917, 2021.
Article in English | MEDLINE | ID: mdl-34336268

ABSTRACT

BACKGROUND: MicroRNA-10a (miR-10a) inhibits transcriptional factor GATA6 to repress inflammatory GATA6/VCAM-1 signaling, which is regulated by blood flow to affect endothelial function/dysfunction. This study aimed to identify the expression patterns of miR-10a/GATA6/VCAM-1 in vivo and study their implications in the pathophysiology of human coronary artery disease (CAD), i.e., atherosclerosis. METHODS: Human atherosclerotic coronary arteries and nondiseased arteries were used to detect the expressions of miR-10a/GATA6/VCAM-1 in pathogenic vs. normal conditions. In addition, sera from CAD patients and healthy subjects were collected to detect the level of circulating miR-10a. RESULTS: The comparison of human atherosclerotic coronary arteries with nondiseased arteries demonstrated that lower levels of endothelial miR-10a are related to human atherogenesis. Moreover, GATA6/VCAM-1 (a downstream target of miR-10a) was highly expressed in the endothelium, accompanied by the reduced levels of miR-10a during the development of human atherosclerosis. In addition, CAD patients had a significantly lower concentration of miR-10a in their serum compared to healthy subjects. CONCLUSIONS: Our findings suggest that low miR-10a and high GATA6/VCAM-1 in the cardiovascular endothelium correlates to the development of human atherosclerotic lesions, suggesting that miR-10a signaling has the potential to be developed as a biomarker for human atherosclerosis.

7.
Front Cell Dev Biol ; 9: 697539, 2021.
Article in English | MEDLINE | ID: mdl-34262908

ABSTRACT

BACKGROUND: Pathophysiological vascular remodeling in response to disturbed flow with low and oscillatory shear stress (OSS) plays important roles in atherosclerosis progression. Pomegranate extraction (PE) was reported having anti-atherogenic effects. However, whether it can exert a beneficial effect against disturbed flow-induced pathophysiological vascular remodeling to inhibit atherosclerosis remains unclear. The present study aims at investigating the anti-atherogenic effects of pomegranate peel polyphenols (PPP) extraction and its purified compound punicalagin (PU), as well as their protective effects on disturbed flow-induced vascular dysfunction and their underlying molecular mechanisms. METHODS: The anti-atherogenic effects of PPP/PU were examined on low-density lipoprotein receptor knockout mice fed with a high fat diet. The vaso-protective effects of PPP/PU were examined in rat aortas using myograph assay. A combination of in vivo experiments on rats and in vitro flow system with human endothelial cells (ECs) was used to investigate the pharmacological actions of PPP/PU on EC dysfunction induced by disturbed flow. In addition, the effects of PPP/PU on vascular smooth muscle cell (VSMC) dysfunction were also examined. RESULTS: PU is the effective component in PPP against atherosclerosis. PPP/PU evoked endothelium-dependent relaxation in rat aortas. PPP/PU inhibited the activation of Smad1/5 in the EC layers at post-stenotic regions of rat aortas exposed to disturbed flow with OSS. PPP/PU suppressed OSS-induced expression of cell cycle regulatory and pro-inflammatory genes in ECs. Moreover, PPP/PU inhibited inflammation-induced VSMC dysfunction. CONCLUSION: PPP/PU protect against OSS-induced vascular remodeling through inhibiting force-specific activation of Smad1/5 in ECs and this mechanism contributes to their anti-atherogenic effects.

8.
Front Cell Dev Biol ; 9: 647714, 2021.
Article in English | MEDLINE | ID: mdl-33959608

ABSTRACT

MicroRNAs (miRs) and bone morphogenetic protein receptor-specific Smads are mechano-responsive molecules that play vital roles in modulating endothelial cell (EC) functions in response to blood flow. However, the roles of interplay between these molecules in modulating EC functions under flows remain unclear. We elucidated the regulatory roles of the interplay between miR-487a and Smad5 in EC proliferation in response to different flow patterns. Microarray and quantitative RT-PCR showed that disturbed flow with low and oscillatory shear stress (OS, 0.5 ± 4 dynes/cm2) upregulates EC miR-487a in comparison to static controls and pulsatile shear stress (12 ± 4 dynes/cm2). MiR-487a expression was higher in ECs in the inner curvature (OS region) than the outer curvature of the rat aortic arch and thoracic aorta and also elevated in diseased human coronary arteries. MiR-487a expression was promoted by nuclear phospho-Smad5, which bound to primary-miR-487a to facilitate miR-487a processing. Algorithm prediction and luciferase reporter and argonaute 2-immunoprecipitation assays demonstrated that miR-487a binds to 3'UTR of CREB binding protein (CBP) and p53. Knockdown and overexpression of miR-487a decreased and increased, respectively, phospho-Rb and cyclin A expressions through CBP and p53. A BrdU incorporation assay showed that miR-487a enhanced EC proliferation under OS in vitro and in disturbed flow regions of experimentally stenosed rat abdominal aorta in vivo. These results demonstrate that disturbed flow with OS induces EC expression of miR-487a through its enhanced processing by activated-Smad5. MiR-487 inhibits its direct targets CBP and p53 to induce EC cycle progression and proliferation. Our findings suggest that EC miR-487 may serve as an important molecular target for intervention against disturbed flow-associated vascular disorders resulting from atherosclerosis.

9.
Cardiovasc Drugs Ther ; 35(1): 61-71, 2021 02.
Article in English | MEDLINE | ID: mdl-32902737

ABSTRACT

PURPOSE: Little is known about the molecular interactions among inflammatory responses that damage venous endothelial cells (vECs) during venous-to-arterial flow transition in vein graft diseases. Because arterial flow triggers excessive autophagy and inflammation in vECs, this study aimed to investigate the mediator of inflammation and methods to prevent vEC damage. METHODS: Arterial laminar shear stress (ALSS; 12 dynes/cm2) was applied to vECs via in vitro and ex vivo perfusion systems. Inflammation in vECs was measured using inflammatory protein markers, NFκB translocation, cyclooxygenase-2 (COX-2) and COX-2 and NFκB promoter assays. The involvement of microRNA-4488 (miR-4488) was measured and confirmed by altering the specific miR using a miR-4488 mimic or inhibitor. The potential anti-inflammatory drugs and/or nitric oxide (NO) donor L-arginine (L-Arg) to prevent damage to vECs under ALSS was investigated. RESULTS: ALSS triggered reactive oxygen species production, excessive autophagy, COX-2 protein expression, and NFκB translocation during vEC inflammation. Reduction in miR-4488 expression was detected in inflamed vECs treated with LPS, lipopolysaccharide (LPS) TNFα, and ALSS. Transfection of miR-4488 mimic (50 nM) prior to ALSS application inhibited the accumulation of inflammatory proteins as well as the translocation of NFκB. Combined treatment of vECs with COX-2-specific inhibitor (SC-236) and L-Arg alleviated the ALSS-induced inflammatory responses. Protective effects of the combined treatment on vECs against ALSS-induced damage were abolished by the application of miR-4488 inhibitor. CONCLUSION: We showed that ALSS triggered the COX-2/NFκB pathway to induce vEC inflammation with a reduction in miR-4488. Combination of SC-236 and L-Arg prevented ALSS-induced vEC damage, thus, shows high potential for preventing vein graft diseases.


Subject(s)
Endothelium, Vascular/metabolism , Inflammation Mediators/metabolism , MicroRNAs/biosynthesis , NF-kappa B/metabolism , Anti-Inflammatory Agents/pharmacology , Autophagy/drug effects , Coronary Artery Bypass/adverse effects , Coronary Vessels/physiopathology , Cyclooxygenase 2/drug effects , Hemodynamics , Human Umbilical Vein Endothelial Cells , Humans , Lipopolysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Saphenous Vein/physiopathology , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis
10.
Cardiovasc Res ; 116(1): 226-236, 2020 01 01.
Article in English | MEDLINE | ID: mdl-30785200

ABSTRACT

AIMS: Disturbed blood flow at arterial branches and curvatures modulates endothelial function and predisposes the region to endothelial inflammation and subsequent development of atherosclerotic lesions. Activation of the endothelial Toll-like receptors (TLRs), in particular TLR4, contributes to vascular inflammation. Therefore, we investigate whether TLR4 can sense disturbed flow (DF) to mediate the subsequent endothelial inflammation. METHODS AND RESULTS: En face staining of endothelium revealed that TLR4 expression, activation, and its downstream inflammatory markers were elevated in mouse aortic arch compared with thoracic aorta, which were absent in Tlr4mut mice. Similar results were observed in the partial carotid ligation model where TLR4 signalling was activated in response to ligation-induced flow disturbance in mouse carotid arteries, and such effect was attenuated in Tlr4mut mice. DF in vitro increased TLR4 expression and activation in human endothelial cells (ECs) and promoted monocyte-EC adhesion, which were inhibited in TLR4-knockdown ECs. Among endogenous TLR4 ligands examined as candidate mediators of DF-induced TLR4 activation, fibronectin containing the extra domain A (FN-EDA) expressed by ECs was increased by DF and was revealed to directly interact with and activate TLR4. CONCLUSION: Our findings demonstrate the indispensable role of TLR4 in DF-induced endothelial inflammation and pinpoint FN-EDA as the endogenous TLR4 activator in this scenario. This novel mechanism of vascular inflammation under DF condition may serve as a critical initiating step in atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Carotid Arteries/metabolism , Endothelial Cells/metabolism , Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Blood Flow Velocity , Carotid Arteries/physiopathology , Carotid Arteries/surgery , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/physiopathology , Cell Adhesion , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Fibronectins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/physiopathology , Ligands , Ligation , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/metabolism , Stress, Mechanical , Toll-Like Receptor 4/genetics
11.
J Cell Mol Med ; 24(1): 910-920, 2020 01.
Article in English | MEDLINE | ID: mdl-31680453

ABSTRACT

In type 1 and type 2 diabetes mellitus, increased cardiac fibrosis, stiffness and associated diastolic dysfunction may be the earliest pathological phenomena in diabetic cardiomyopathy. Endothelial-mesenchymal transition (EndMT) in endothelia cells (ECs) is a critical cellular phenomenon that increases cardiac fibroblasts (CFs) and cardiac fibrosis in diabetic hearts. The purpose of this paper is to explore the molecular mechanism of miR-21 regulating EndMT and cardiac perivascular fibrosis in diabetic cardiomyopathy. In vivo, hyperglycaemia up-regulated the mRNA level of miR-21, aggravated cardiac dysfunction and collagen deposition. The condition was recovered by inhibition of miR-21 following with improving cardiac function and decreasing collagen deposition. miR-21 inhibition decreased cardiac perivascular fibrosis by suppressing EndMT and up-regulating SMAD7 whereas activating p-SMAD2 and p-SMAD3. In vitro, high glucose (HG) up-regulated miR-21 and induced EndMT in ECs, which was decreased by inhibition of miR-21. A highly conserved binding site of NF-κB located in miR-21 5'-UTR was identified. In ECs, SMAD7 is directly regulated by miR-21. In conclusion, the pathway of NF-κB/miR-21/SMAD7 regulated the process of EndMT in T1DM, in diabetic cardiomyopathy, which may be regarded as a potential clinical therapeutic target for cardiac perivascular fibrosis.


Subject(s)
Coronary Artery Disease/prevention & control , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Endothelium, Vascular/metabolism , Epithelial-Mesenchymal Transition , Fibrosis/prevention & control , MicroRNAs/antagonists & inhibitors , Animals , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics
12.
J Biomed Sci ; 26(1): 56, 2019 Aug 07.
Article in English | MEDLINE | ID: mdl-31387590

ABSTRACT

BACKGROUND: Endothelial cell (EC) dysfunctions, including turnover enrichment, gap junction disruption, inflammation, and oxidation, play vital roles in the initiation of vascular disorders and atherosclerosis. Hemodynamic forces, i.e., atherprotective pulsatile (PS) and pro-atherogenic oscillatory shear stress (OS), can activate mechanotransduction to modulate EC function and dysfunction. This review summarizes current studies aiming to elucidate the roles of epigenetic factors, i.e., histone deacetylases (HDACs), non-coding RNAs, and DNA methyltransferases (DNMTs), in mechanotransduction to modulate hemodynamics-regulated EC function and dysfunction. OS enhances the expression and nuclear accumulation of class I and class II HDACs to induce EC dysfunction, i.e., proliferation, oxidation, and inflammation, whereas PS induces phosphorylation-dependent nuclear export of class II HDACs to inhibit EC dysfunction. PS induces overexpression of the class III HDAC Sirt1 to enhance nitric oxide (NO) production and prevent EC dysfunction. In addition, hemodynamic forces modulate the expression and acetylation of transcription factors, i.e., retinoic acid receptor α and krüppel-like factor-2, to transcriptionally regulate the expression of microRNAs (miRs). OS-modulated miRs, which stimulate proliferative, pro-inflammatory, and oxidative signaling, promote EC dysfunction, whereas PS-regulated miRs, which induce anti-proliferative, anti-inflammatory, and anti-oxidative signaling, inhibit EC dysfunction. PS also modulates the expression of long non-coding RNAs to influence EC function. i.e., turnover, aligmant, and migration. On the other hand, OS enhances the expression of DNMT-1 and -3a to induce EC dysfunction, i.e., proliferation, inflammation, and NO repression. CONCLUSION: Overall, epigenetic factors play vital roles in modulating hemodynamic-directed EC dysfunction and vascular disorders, i.e., atherosclerosis. Understanding the detailed mechanisms through which epigenetic factors regulate hemodynamics-directed EC dysfunction and vascular disorders can help us to elucidate the pathogenic mechanisms of atherosclerosis and develop potential therapeutic strategies for atherosclerosis treatment.


Subject(s)
Atherosclerosis/physiopathology , Endothelial Cells/physiology , Epigenesis, Genetic , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , DNA Methylation/genetics , Endothelial Cells/enzymology , Endothelial Cells/pathology , Hemodynamics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mechanotransduction, Cellular/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
13.
Methods Mol Biol ; 2019: 143-169, 2019.
Article in English | MEDLINE | ID: mdl-31359395

ABSTRACT

The protocols in this chapter describe methods for identifying the functional roles of retinoic acid receptor (RAR) and retinoid X receptor (RXR) signaling in atherosclerosis and developing RARα/RXRα-specific agonists as hemodynamics-based therapeutic components for atherosclerosis treatment. In vitro cell culture flow system is used to elucidate the effects of different flow patterns and shear stresses, i.e., atherogenic oscillatory shear stress (OS) vs. atheroprotective pulsatile shear stress (PS), on RAR/RXR signaling and inflammatory responses in vascular endothelial cells (ECs). Western blotting, nuclear and cytoplasmic protein extraction, immunoprecipitation, and in situ proximity ligation assay are used to examine the expression, location, and association of RARs (i.e., RARα, RARß, and RARγ) and RXRs (i.e., RXRα, RXRß, and RXRγ) in ECs in response to OS vs. PS. Chromatin immunoprecipitation is used to examine the binding activity of RARα/RA-responsive elements (RARE). RT-microRNA (miR) quantitative real-time PCR and RT-PCR are used to detect the expressions of miR-10a and pro-inflammatory molecules, respectively. Specific siRNAs of RARα and RXRα, precursor miR-10a (PreR-10a), and antagomiR-10a (AMR-10a) are used to elucidate the regulatory roles of RARα, RXRα, and miR-10a in pro-inflammatory signaling in ECs. RARα/RXRα-specific agonists are used to induce miR-10a expression and inhibit OS-induced pro-inflammatory signaling in ECs in vitro. Apolipoprotein E-deficient (ApoE-/-) mice are used as an atherosclerotic animal model. Administration of ApoE-/- mice with RARα/RXRα-specific agonists results in inhibitions in atherosclerotic lesion formation. Co-administration of ApoE-/- mice with RARα/RXRα agonists and AMR-10a is performed to identify the role of miR-10a in RARα/RXRα agonists-mediated inhibition in atherosclerotic lesions. Oil Red O staining and H&E staining are used to examine the levels of atherosclerotic lesions in the vessel wall. In situ miR hybridization and immunohistochemical staining are used to detect the expression of miR-10a and pro-inflammatory molecules and the infiltration of inflammatory cells in the vessel wall. RARα/RXRα-specific agonists are used to mimic the atheroprotective effects of PS to induce endothelial miR-10a and hence repress OS-induced pro-inflammatory signaling and atherosclerotic lesion formation in vivo. The results indicate that RAR/RXR-specific agonists have great potential to be developed as hemodynamics-based therapeutic components for atherosclerosis treatment.


Subject(s)
Antagomirs/administration & dosage , Atherosclerosis/drug therapy , Benzoates/administration & dosage , Coumaric Acids/administration & dosage , MicroRNAs/genetics , Tetrahydronaphthalenes/administration & dosage , Animals , Antagomirs/pharmacology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Benzoates/pharmacology , Cell Line , Coumaric Acids/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Hemodynamics , Humans , Mice , MicroRNAs/antagonists & inhibitors , Retinoic Acid Receptor alpha/agonists , Retinoid X Receptor alpha/agonists , Signal Transduction/drug effects , Tetrahydronaphthalenes/pharmacology
14.
Clin J Am Soc Nephrol ; 13(11): 1712-1720, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30242025

ABSTRACT

BACKGROUND AND OBJECTIVES: There is increasing evidence that microRNAs (miRNAs) play crucial roles in the regulation of neointima formation. However, the translational evidence of the role of miRNAs in dialysis vascular access is limited. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: miRNA expression in tissues was assessed by using venous tissues harvested from ten patients on dialysis who received revision or removal surgery, and ten patients who were predialysis and received creation surgery of arteriovenous fistulas served as controls. To extend these findings, 60 patients who received angioplasty of dialysis access were enrolled and the levels of circulating miRNAs were determined before and 2 days after angioplasty. Clinical follow-up was continued monthly for 6 months. The primary outcome of angioplasty cohort was target lesion restenosis within 6 months after angioplasty. RESULTS: In the surgery cohort, the expressions of miR-21, miR-130a, and miR-221 were upregulated in stenotic tissues, whereas those of miR-133 and miR-145 were downregulated. In situ hybridization revealed similar expression patterns of these miRNAs, localized predominantly in the neointima region. Twenty eight patients in the angioplasty cohort developed restenosis within 6 months. The levels of circulating miR-21, miR-130a, miR-221, miR-133, and miR-145 significantly increased 2 days after angioplasty. Kaplan-Meier plots showed that patients with an increase of miR-21 expression level >0.35 have a higher risk of patency loss (hazard ratio, 4.45; 95% confidence interval, 1.68 to 11.7). In a multivariable analysis, postangioplasty increase of miR-21 expression was independently associated with restenosis (hazard ratio, 1.20; 95% confidence interval, 1.07 to 1.35 per one unit increase of miR-21 expression level; P=0.001). CONCLUSIONS: Certain miRNAs are differentially expressed in the stenotic venous segments of dialysis accesses. An increase in blood miR-21 level with angioplasty is associated with a higher risk of restenosis.


Subject(s)
Arteriovenous Shunt, Surgical/adverse effects , MicroRNAs/blood , Neointima/metabolism , Neointima/pathology , Veins/pathology , Aged , Aged, 80 and over , Angioplasty , Case-Control Studies , Constriction, Pathologic/etiology , Constriction, Pathologic/metabolism , Constriction, Pathologic/therapy , Down-Regulation , Female , Humans , Hyperplasia , Male , MicroRNAs/metabolism , Middle Aged , Neointima/etiology , Recurrence , Renal Dialysis , Risk Factors , Up-Regulation , Veins/metabolism
15.
Atherosclerosis ; 271: 36-44, 2018 04.
Article in English | MEDLINE | ID: mdl-29459264

ABSTRACT

BACKGROUND AND AIMS: MicroRNA (miR)-10a is a shear-regulated miR with the lowest expression in vascular endothelial cells (ECs) in athero-susceptible regions with oscillatory shear stress (OS). The aim of this study is to elucidate the relationship between EC miR-10a and atherosclerosis and develop a hemodynamics-based strategy for atherosclerosis treatment. METHODS: A combination of in vitro flow system and in vivo experimental animals was used to examine the functional roles of EC miR-10a and its clinical applications in atherosclerosis. RESULTS: En face staining showed that EC miR-10a is down-regulated in the inner curvature (OS region) of aortic arch in rats. Co-administration with retinoic acid receptor-α (RARα)- and retinoid X receptor-α (RXRα)-specific agonists rescued EC miR-10a expression in this OS region. These effects of OS and RARα/RXRα-specific agonists on EC miR-10a expression were confirmed by the in vitro flow system, and were modulated by the RARα-histone deacetylases complex, with the consequent modulation in the downstream GATA6/vascular cell adhesion molecule (VCAM)-1 signaling cascade. Animal studies showed that miR-10a levels are decreased in both aortic endothelium of atherosclerotic lesions and blood plasma from apolipoprotein E-deficient (ApoE-/-) mice. In vivo induction of EC miR-10a by administration of RARα/RXRα-specific agonists protects ApoE-/- mice from atherosclerosis through inhibition of GATA6/VCAM-1 signaling and inflammatory cell infiltration. CONCLUSIONS: Our findings indicate that down-regulation of miR-10a in aortic endothelium and blood serum is associated with atherosclerosis, and miR-10a has potential to be developed as diagnostic molecule for atherosclerosis. Moreover, EC miR-10a induction by RARα/RXRα-specific agonists is a potential hemodynamics-based strategy for atherosclerosis treatment.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Benzoates/pharmacology , Coumaric Acids/pharmacology , MicroRNAs/metabolism , Plaque, Atherosclerotic , Retinoic Acid Receptor alpha/agonists , Retinoid X Receptor alpha/agonists , Tetrahydronaphthalenes/pharmacology , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , GATA6 Transcription Factor/metabolism , Hemodynamics , Humans , Mechanotransduction, Cellular/drug effects , Mice, Knockout, ApoE , MicroRNAs/genetics , Rats , Regional Blood Flow , Retinoic Acid Receptor alpha/metabolism , Retinoid X Receptor alpha/metabolism , Stress, Mechanical , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
16.
J Biomed Sci ; 25(1): 1, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29295709

ABSTRACT

BACKGROUND: Atherosclerosis occurs in arterial curvatures and branches, where the flow is disturbed with low and oscillatory shear stress (OSS). The remodeling and alterations of extracellular matrices (ECMs) and their composition is the critical step in atherogenesis. In this study, we investigated the effects of different ECM proteins on the regulation of mechanotransduction in vascular endothelial cells (ECs) in response to OSS. METHODS: Through the experiments ranging from in vitro cell culture studies on effects of OSS on molecular signaling to in vivo examinations on clinical specimens from patients with coronary artery disease (CAD), we elucidated the roles of integrins and different ECMs, i.e., fibronectin (FN) and laminin (LM), in transforming growth factor (TGF)-ß receptor (TßR)-mediated Smad2 activation and nuclear factor-κB (NF-κB) signaling in ECs in response to OSS and hence atherogenesis. RESULTS: OSS at 0.5±12 dynes/cm2 induces sustained increases in the association of types I and II TßRs with ß1 and ß3 integrins in ECs grown on FN, but it only transient increases in ECs grown on LM. OSS induces a sustained activation of Smad2 in ECs on FN, but only a transient activation of Smad2 in ECs on LM. OSS-activation of Smad2 in ECs on FN regulates downstream NF-κB signaling and pro-inflammatory gene expression through the activation of ß1 integrin and its association with TßRs. In contrast, OSS induces transient activations of ß1 and ß3 integrins in ECs on LM, which associate with type I TßR to regulate Smad2 phosphorylation, resulting in transient induction of NF-κB and pro-inflammatory gene expression. In vivo investigations on diseased human coronary arteries from CAD patients revealed that Smad2 is highly activated in ECs of atherosclerotic lesions, which is accompanied by the concomitant increase of FN rather than LM in the EC layer and neointimal region of atherosclerotic lesions. CONCLUSIONS: Our findings provide new insights into the mechanisms of how OSS regulates Smad2 signaling and pro-inflammatory genes through the complex signaling networks of integrins, TßRs, and ECMs, thus illustrating the molecular basis of regional pro-inflammatory activation within disturbed flow regions in the arterial tree.


Subject(s)
Endothelial Cells/physiology , Mechanotransduction, Cellular , Smad2 Protein/genetics , Biomechanical Phenomena , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Humans , Laminin/genetics , Laminin/metabolism , Smad2 Protein/metabolism , Stress, Mechanical
17.
Proc Natl Acad Sci U S A ; 115(1): 133-138, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29255056

ABSTRACT

Leukocyte transmigration across vessel walls is a critical step in the innate immune response. Upon their activation and firm adhesion to vascular endothelial cells (VECs), leukocytes preferentially extravasate across junctional gaps in the endothelial monolayer (paracellular diapedesis). It has been hypothesized that VECs facilitate paracellular diapedesis by opening their cell-cell junctions in response to the presence of an adhering leukocyte. However, it is unclear how leukocytes interact mechanically with VECs to open the VEC junctions and migrate across the endothelium. In this study, we measured the spatial and temporal evolution of the 3D traction stresses generated by the leukocytes and VECs to elucidate the sequence of mechanical events involved in paracellular diapedesis. Our measurements suggest that the contractile stresses exerted by the leukocytes and the VECs can separately perturb the junctional tensions of VECs to result in the opening of gaps before the initiation of leukocyte transmigration. Decoupling the stresses exerted by the transmigrating leukocytes and the VECs reveals that the leukocytes actively contract the VECs to open a junctional gap and then push themselves across the gap by generating strong stresses that push into the matrix. In addition, we found that diapedesis is facilitated when the tension fluctuations in the VEC monolayer were increased by proinflammatory thrombin treatment. Our findings demonstrate that diapedesis can be mechanically regulated by the transmigrating leukocytes and by proinflammatory signals that increase VEC contractility.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Leukocytes/metabolism , Models, Biological , Transendothelial and Transepithelial Migration/physiology , HL-60 Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , Leukocytes/cytology
18.
Biomaterials ; 155: 203-216, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29182961

ABSTRACT

Cells perceive the physical cues such as perturbations of extracellular matrix (ECM) stiffness, and translate these stimuli into biochemical signals controlling various aspects of cell behavior, which contribute to the physiological and pathological processes of multiple organs. In this study, we tested the hypothesis that during arterial stiffening, vascular smooth muscle cells (SMCs) sense the increase of ECM stiffness, which modulates the cellular phenotype through the regulation in DNA methyltransferases 1 (DNMT1) expression. Moreover, we hypothesized that the mechanisms involve intrinsic stiffening and deficiency in contractility of vascular SMCs. Substrate stiffening was mimicked in vitro with polyacrylamide gels. A contractile-to-synthetic phenotypic transition was induced by substrate stiffening in vascular SMCs through the down-regulation of DNMT1 expression. DNMT1 repression was also observed in the tunica media of mice aortas in an acute aortic injury model and a chronic kidney failure model, as well as in the tunica intima of human carotid arteries with calcified atherosclerotic lesions. DNMT1 inhibition facilitates arterial stiffening in vivo and promotes osteogenic transdifferentiation, calcification and cellular stiffening of vascular SMCs in vitro. These effects may be attributable, at least in part, to the role of DNMT1 in regulating the promoter activities of Transgelin (SM22α) and α-smooth muscle actin (SMA) and the functional contractility of SMCs. We conclude that DNMT1 is a critical regulator that negatively regulates arterial stiffening via maintaining the contractile phenotype of vascular SMCs. This research may facilitate elucidation of the complex crosstalk between vascular SMCs and their surrounding matrix in healthy and in pathological conditions and provide new insights into the implications for potential targeting of the phenotypic regulatory mechanisms in material-related therapeutic applications.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Actins/metabolism , Animals , DNA Methylation/physiology , Extracellular Matrix/metabolism , Mice , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Vascular Stiffness/physiology
19.
Proc Natl Acad Sci U S A ; 114(8): 2072-2077, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167758

ABSTRACT

Histone deacetylases (HDACs) and microRNAs (miRs) have emerged as two important epigenetic factors in the regulation of vascular physiology. This study aimed to elucidate the relationship between HDACs and miRs in the hemodynamic modulation of endothelial cell (EC) dysfunction. We found that miR-10a has the lowest expression among all examined shear-responsive miRs in ECs under oscillatory shear stress (OS), and a relatively high expression under pulsatile shear stress (PS). PS and OS alter EC miR-10a expression to regulate the expression of its direct target GATA6 and downstream vascular cell adhesion molecule (VCAM)-1. PS induces the expression, nuclear accumulation, and association of retinoid acid receptor-α (RARα) and retinoid X receptor-α (RXRα). RARα and RXRα serve as a "director" and an "enhancer," respectively, to enhance RARα binding to RA-responsive element (RARE) and hence miR-10a expression, thus down-regulating GATA6/VCAM-1 signaling in ECs. In contrast, OS induces associations of "repressors" HDAC-3/5/7 with RARα to inhibit the RARα-directed miR-10a signaling. The flow-mediated miR-10a expression is regulated by Krüppel-like factor 2 through modulation in RARα-RARE binding, with the consequent regulation in GATA6/VCAM-1 in ECs. These results are confirmed in vivo by en face staining on the aortic arch vs. the straight thoracic aorta of rats. Our findings identify a mechanism by which HDACs and RXRα modulate the hormone receptor RARα to switch miR-10a expression and hence the proinflammatory vs. anti-inflammatory responses of vascular endothelium under different hemodynamic forces.


Subject(s)
Endothelium, Vascular/physiology , GATA6 Transcription Factor/genetics , Histone Deacetylases/metabolism , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Retinoic Acid Receptor alpha/metabolism , Retinoid X Receptor alpha/metabolism , Animals , Aorta/cytology , Aorta/physiology , Atherosclerosis/physiopathology , Cells, Cultured , Down-Regulation , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , GATA6 Transcription Factor/metabolism , Humans , RNA Interference , RNA, Small Interfering/metabolism , Rats , Retinoic Acid Receptor alpha/genetics , Retinoid X Receptor alpha/genetics , Signal Transduction/genetics , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
20.
Nature ; 540(7634): 579-582, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27926730

ABSTRACT

The Yorkie homologues YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif, also known as WWTR1), effectors of the Hippo pathway, have been identified as mediators for mechanical stimuli. However, the role of YAP/TAZ in haemodynamics-induced mechanotransduction and pathogenesis of atherosclerosis remains unclear. Here we show that endothelial YAP/TAZ activity is regulated by different patterns of blood flow, and YAP/TAZ inhibition suppresses inflammation and retards atherogenesis. Atheroprone-disturbed flow increases whereas atheroprotective unidirectional shear stress inhibits YAP/TAZ activity. Unidirectional shear stress activates integrin and promotes integrin-Gα13 interaction, leading to RhoA inhibition and YAP phosphorylation and suppression. YAP/TAZ inhibition suppresses JNK signalling and downregulates pro-inflammatory genes expression, thereby reducing monocyte attachment and infiltration. In vivo endothelial-specific YAP overexpression exacerbates, while CRISPR/Cas9-mediated Yap knockdown in endothelium retards, plaque formation in ApoE-/- mice. We also show several existing anti-atherosclerotic agents such as statins inhibit YAP/TAZ transactivation. On the other hand, simvastatin fails to suppress constitutively active YAP/TAZ-induced pro-inflammatory gene expression in endothelial cells, indicating that YAP/TAZ inhibition could contribute to the anti-inflammatory effect of simvastatin. Furthermore, activation of integrin by oral administration of MnCl2 reduces plaque formation. Taken together, our results indicate that integrin-Gα13-RhoA-YAP pathway holds promise as a novel drug target against atherosclerosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...