Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 114(3): 443-455, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29206900

ABSTRACT

AIMS: To investigate the role of chromobox protein homolog 3 (Cbx3) in vascular smooth muscle cell (VSMC) proliferation, migration, and neointima formation following vascular injury. METHODS AND RESULTS: Overexpression of Cbx3 led to a significant increase in VSMC contractile gene expression and VSMC apoptosis as well as a dramatic decrease in collagen gene expression, VSMC proliferation, and migration. Meanwhile, the opposite was observed following inhibition of endogenous Cbx3. Luciferase activity assays revealed that Notch signalling, but neither ß-catenin nor NF-κB signalling, is regulated by Cbx3 in VSMCs, and among the four Notch receptors, Notch3 is selectively down-regulated by Cbx3 through a transcriptional repression mechanism. Notch3 gene activation recapitulates the effects of Cbx3 knockdown on VSMC proliferation and migration. Consequently, the inhibitory effects of Cbx3 over-expression on VSMC proliferation and migration were reversed by Notch3 gene reactivation. In a model of vascular damage by carotid wire injury, we observed that Cbx3 expression was dramatically down-regulated in the injured arteries. Local ectopic over-expression of Cbx3 in the injured arteries significantly inhibited Notch3 expression, thereby reducing VSMCs proliferation and causing an overall decrease in neointima formation. Additionally, injury-induced neointimal SMC hyperplasia was significantly reduced by aortic inhibition of Notch3. Importantly, a decreased expression level of Cbx3, but an increased expression level of Notch3, was observed in human femoral arteries with atherosclerotic lesions. CONCLUSION: Cbx3 modulates VSMC contractile and collagen gene expression, as well as VSMC proliferation, migration, and apoptosis via a Notch3 pathway, and plays an important role in controlling injury-induced neointima formation.


Subject(s)
Carotid Artery Injuries/metabolism , Cell Movement , Cell Proliferation , Chromosomal Proteins, Non-Histone/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Animals , Apoptosis , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Femoral Artery/metabolism , Femoral Artery/pathology , Humans , Mice, Inbred C57BL , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/pathology , Plaque, Atherosclerotic , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Signal Transduction , beta Catenin/metabolism
2.
Circulation ; 137(17): 1824-1841, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29246895

ABSTRACT

BACKGROUND: MicroRNA-22 (miR-22) has recently been reported to play a regulatory role during vascular smooth muscle cell (VSMC) differentiation from stem cells, but little is known about its target genes and related pathways in mature VSMC phenotypic modulation or its clinical implication in neointima formation following vascular injury. METHODS: We applied a wire-injury mouse model, and local delivery of AgomiR-22 or miR-22 inhibitor, as well, to explore the therapeutic potential of miR-22 in vascular diseases. Furthermore, normal and diseased human femoral arteries were harvested, and various in vivo, ex vivo, and in vitro models of VSMC phenotype switching were conducted to examine miR-22 expression during VSMC phenotype switching. RESULTS: Expression of miR-22 was closely regulated during VSMC phenotypic modulation. miR-22 overexpression significantly increased expression of VSMC marker genes and inhibited VSMC proliferation and migration, whereas the opposite effect was observed when endogenous miR-22 was knocked down. As expected, 2 previously reported miR-22 target genes, MECP2 (methyl-CpG binding protein 2) and histone deacetylase 4, exhibited a regulatory role in VSMC phenotypic modulation. A transcriptional regulator and oncoprotein, EVI1 (ecotropic virus integration site 1 protein homolog), has been identified as a novel miR-22 target gene in VSMC phenotypic modulation. It is noteworthy that overexpression of miR-22 in the injured vessels significantly reduced the expression of its target genes, decreased VSMC proliferation, and inhibited neointima formation in wire-injured femoral arteries, whereas the opposite effect was observed with local application of a miR-22 inhibitor to injured arteries. We next examined the clinical relevance of miR-22 expression and its target genes in human femoral arteries. We found that miR-22 expression was significantly reduced, whereas MECP2 and EVI1 expression levels were dramatically increased, in diseased in comparison with healthy femoral human arteries. This inverse relationship between miR-22 and MECP2 and EVI1 was evident in both healthy and diseased human femoral arteries. CONCLUSIONS: Our data demonstrate that miR-22 and EVI1 are novel regulators of VSMC function, specifically during neointima hyperplasia, offering a novel therapeutic opportunity for treating vascular diseases.


Subject(s)
MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Vascular System Injuries/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antagomirs/genetics , Antagomirs/metabolism , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Gene Expression Regulation , Humans , Male , Mice , MicroRNAs/genetics , Middle Aged , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction , Tissue Culture Techniques , Vascular System Injuries/genetics , Vascular System Injuries/pathology
3.
J Am Heart Assoc ; 5(12)2016 12 07.
Article in English | MEDLINE | ID: mdl-27927633

ABSTRACT

BACKGROUND: MicroRNA miR-214 has been implicated in many biological cellular functions, but the impact of miR-214 and its target genes on vascular smooth muscle cell (VSMC) proliferation, migration, and neointima smooth muscle cell hyperplasia is unknown. METHODS AND RESULTS: Expression of miR-214 was closely regulated by different pathogenic stimuli in VSMCs through a transcriptional mechanism and decreased in response to vascular injury. Overexpression of miR-214 in serum-starved VSMCs significantly decreased VSMC proliferation and migration, whereas knockdown of miR-214 dramatically increased VSMC proliferation and migration. Gene and protein biochemical assays, including proteomic analyses, showed that NCK associated protein 1 (NCKAP1)-a major component of the WAVE complex that regulates lamellipodia formation and cell motility-was negatively regulated by miR-214 in VSMCs. Luciferase assays showed that miR-214 substantially repressed wild-type but not the miR-214 binding site mutated version of NCKAP1 3' untranslated region luciferase activity in VSMCs. This result confirmed that NCKAP1 is the functional target of miR-214 in VSMCs. NCKAP1 knockdown in VSMCs recapitulates the inhibitory effects of miR-214 overexpression on actin polymerization, cell migration, and proliferation. Data from cotransfection experiments also revealed that inhibition of NCKAP1 is required for miR-214-mediated lamellipodia formation, cell motility, and growth. Importantly, locally enforced expression of miR-214 in the injured vessels significantly reduced NCKAP1 expression levels, inhibited VSMC proliferation, and prevented neointima smooth muscle cell hyperplasia after injury. CONCLUSIONS: We uncovered an important role of miR-214 and its target gene NCKAP1 in modulating VSMC functions and neointima hyperplasia. Our findings suggest that miR-214 represents a potential therapeutic target for vascular diseases.


Subject(s)
Membrane Proteins/physiology , MicroRNAs/physiology , Neointima/pathology , Angiogenesis Inducing Agents/pharmacology , Animals , Becaplermin , Binding Sites/genetics , Cell Line , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation , Femoral Artery/surgery , Gene Knockdown Techniques , Hyperplasia/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Muscle, Smooth, Vascular/physiology , Mutation/genetics , Myocytes, Smooth Muscle , Proteomics , Proto-Oncogene Proteins c-sis/pharmacology , RNA, Small Interfering/physiology , Twist-Related Protein 1/antagonists & inhibitors
4.
Arterioscler Thromb Vasc Biol ; 35(4): 918-29, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25722434

ABSTRACT

OBJECTIVE: In this study, we attempted to uncover the functional impact of microRNA-22 (miR-22) and its target gene in smooth muscle cell (SMC) differentiation and delineate the molecular mechanism involved. APPROACH AND RESULTS: miR-22 was found to be significantly upregulated during SMC differentiation from embryonic stem cells and adventitia stem/progenitor cells. Enforced expression of miR-22 by its mimic, while knockdown of miR-22 by its antagomiR, promotes or inhibits SMC differentiation from embryonic stem cells and adventitia stem/progenitor cells, respectively. Expectedly, miR-22 overexpression in stem cells promoted SMC differentiation in vivo. Methyl CpG-binding protein 2 (MECP2) was predicted as one of the top targets of miR-22. Interestingly, the gene expression levels of MECP2 were significantly decreased during SMC differentiation, and MECP2 was dramatically decreased in miR-22 overexpressing cells but significantly increased when miR-22 was knockdown in the differentiating stem cells. Importantly, luciferase assay showed that miR-22 substantially inhibited wild-type, but not mutant MECP2-3' untranslated region-luciferase activity. In addition, modulation of MECP2 expression levels affects multiple SMC-specific gene expression in differentiated embryonic stem cells. Mechanistically, our data showed that MECP2 could transcriptionally repress SMC gene expression through modulating various SMC transcription factors, as well as several proven SMC differentiation regulators. Evidence also revealed that enrichment of H3K9 trimethylation around the promoter regions of the SMC differentiation regulators genes were significantly increased by MECP2 overexpression. Finally, miR-22 was upregulated by platelet-derived growth factor-BB and transforming growth factor-ß through a transcriptional mechanism during SMC differentiation. CONCLUSIONS: miR-22 plays an important role in SMC differentiation, and epigenetic regulation through MECP2 is required for miR-22 mediated SMC differentiation.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Methyl-CpG-Binding Protein 2/metabolism , MicroRNAs/metabolism , Myocytes, Smooth Muscle/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , 3' Untranslated Regions , Animals , Becaplermin , Binding Sites , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/drug effects , Epigenesis, Genetic , Gene Expression Regulation , Histones/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methylation , Mice , MicroRNAs/genetics , Mutation , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotides/metabolism , Phospholipases A2/genetics , Phospholipases A2/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , RNA Interference , Serum Response Element , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , Transfection , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...