Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 430
Filtrer
1.
Int J Cardiol Heart Vasc ; 54: 101500, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39280692

RÉSUMÉ

Abdominal aortic aneurysms (AAAs) are characterized by permanent dilatation of the abdominal aorta, which is accompanied by inflammation, degradation of the extracellular matrix (ECM) and disruption of vascular smooth muscle cell (VSMC) homeostasis. Endoplasmic reticulum (ER) stress is involved in the regulation of inflammation, oxidative stress and VSMC apoptosis, all of which are critical factors in AAA development. Although several studies have revealed the occurrence of ER stress in AAA development, the specific biological functions of ER stress in AAA development remain largely unknown. Given that targeting ER stress is a promising strategy for treating AAAs, further investigation of the physiological and pathological roles of ER stress in AAA development is warranted.

2.
Cardiovasc Res ; 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39189621

RÉSUMÉ

AIMS: Vascular smooth muscle cell (VSMC) plasticity is a state in which VSMCs undergo phenotypic switching from a quiescent contractile phenotype into other functionally distinct phenotypes. Although emerging evidence suggest that VSMC plasticity plays critical roles in the development of vascular diseases, little is known about the key determinant for controlling VSMC plasticity and fate. METHODS AND RESULTS: We found that smooth muscle cell-specific deletion of Lkb1 in tamoxifen-inducible Lkb1flox/flox; Myh11-Cre/ERT2 mice spontaneously and progressively induced aortic/arterial dilation, aneurysm, rupture, and premature death. Single-cell RNA sequencing and imaging-based lineage tracing showed that Lkb1-deficient VSMCs transdifferentiated gradually from early modulated VSMCs to fibroblast-like and chondrocyte-like cells, leading to ossification and blood-vessel rupture. Mechanistically, Lkb1 regulates polypyrimidine tract binding protein 1 (Ptbp1) expression and controls alternative splicing of pyruvate kinase muscle (PKM) isoforms 1 and 2. Lkb1 loss in VSMC results in an increased PKM2/PKM1 ratio and alters the metabolic profile by promoting aerobic glycolysis. Treatment with PKM2 activator TEPP-46 rescues VSMC transformation and aortic dilation in Lkb1flox/flox; Myh11-Cre/ERT2 mice. Furthermore, we found that Lkb1 expression decreased in human aortic aneurysm tissue compared to control tissue, along with changes in markers of VSMC fate. CONCLUSIONS: Lkb1, via its regulation of Ptbp1-dependent alterative splicing of PKM, maintains VSMC in contractile states by suppressing VSMC plasticity.

3.
Cardiovasc Toxicol ; 24(10): 1077-1089, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39126580

RÉSUMÉ

Human aortic vascular smooth muscle cells (HA-VSMCs) play vital roles in the pathogenesis of vascular diseases, including Atherosclerosis (AS). Circular RNAs (circRNAs) have been reported to regulate the biological functions of HA-VSMCs. Therefore, this study aimed to explore the role and mechanism of hsa_circRNA_102353 (circ_0007765) in platelet-derived growth factor-BB (PDGF-BB)-induced HA-VSMCs. Circ_0007765, microRNA-654-3p (miR-654-3p), and Fibroblast Growth Factor Receptor Substrate 2 (FRS2) expression were measured using real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferative ability, invasion, and migration were detected by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), Transwell, and wound healing assays. CyclinD1, MMP2, and FRS2 protein levels were assessed using a Western blot assay. Binding between miR-654-3p and circ_0007765 or FRS2 was predicted by Circinteractome or TargetScan, and verified using dual-luciferase reporter and RNA pull-down assays. PDGF-BB induced HA-VSMC proliferation, invasion, and migration. Circ_0007765 and FRS2 expression levels were increased in PDGF-BB-treated HA-VSMCs, and the miR-654-3p level was reduced. Moreover, circ_0007765 absence hindered PDGF-BB-induced HA-VSMC proliferation, invasion, and migration in vitro. At the molecular level, circ_0007765 increased FRS2 expression by acting as a sponge for miR-654-3p. Our findings revealed that circ_0007765 boosted PDGF-BB-induced HA-VSMC proliferation and migration through elevating FRS2 expression via adsorbing miR-654-3p, providing a feasible therapeutic strategy for AS.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Athérosclérose , Bécaplermine , Mouvement cellulaire , Prolifération cellulaire , Protéines membranaires , microARN , Muscles lisses vasculaires , Myocytes du muscle lisse , ARN circulaire , Transduction du signal , Humains , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , ARN circulaire/métabolisme , ARN circulaire/génétique , Bécaplermine/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , microARN/métabolisme , microARN/génétique , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Cellules cultivées , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Aorte/anatomopathologie , Aorte/métabolisme , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 2/génétique , Régulation de l'expression des gènes , Souris invalidées pour les gènes ApoE , Animaux
4.
Function (Oxf) ; 5(5)2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38984978

RÉSUMÉ

Cantú syndrome (CS), a multisystem disease with a complex cardiovascular phenotype, is caused by gain-of-function (GoF) variants in the Kir6.1/SUR2 subunits of ATP-sensitive potassium (KATP) channels and is characterized by low systemic vascular resistance, as well as tortuous, dilated, vessels, and decreased pulse-wave velocity. Thus, CS vascular dysfunction is multifactorial, with both hypomyotonic and hyperelastic components. To dissect whether such complexities arise cell autonomously within vascular smooth muscle cells (VSMCs) or as secondary responses to the pathophysiological milieu, we assessed electrical properties and gene expression in human induced pluripotent stem cell-derived VSMCs (hiPSC-VSMCs), differentiated from control and CS patient-derived hiPSCs, and in native mouse control and CS VSMCs. Whole-cell voltage clamp of isolated aortic and mesenteric arterial VSMCs isolated from wild-type (WT) and Kir6.1[V65M] (CS) mice revealed no clear differences in voltage-gated K+ (Kv) or Ca2+ currents. Kv and Ca2+ currents were also not different between validated hiPSC-VSMCs differentiated from control and CS patient-derived hiPSCs. While pinacidil-sensitive KATP currents in control hiPSC-VSMCs were similar to those in WT mouse VSMCs, they were considerably larger in CS hiPSC-VSMCs. Under current-clamp conditions, CS hiPSC-VSMCs were also hyperpolarized, consistent with increased basal K conductance and providing an explanation for decreased tone and decreased vascular resistance in CS. Increased compliance was observed in isolated CS mouse aortae and was associated with increased elastin mRNA expression. This was consistent with higher levels of elastin mRNA in CS hiPSC-VSMCs and suggesting that the hyperelastic component of CS vasculopathy is a cell-autonomous consequence of vascular KATP GoF. The results show that hiPSC-VSMCs reiterate expression of the same major ion currents as primary VSMCs, validating the use of these cells to study vascular disease. Results in hiPSC-VSMCs derived from CS patient cells suggest that both the hypomyotonic and hyperelastic components of CS vasculopathy are cell-autonomous phenomena driven by KATP overactivity within VSMCs .


Sujet(s)
Hypertrichose , Cellules souches pluripotentes induites , Canaux KATP , Muscles lisses vasculaires , Myocytes du muscle lisse , Humains , Cellules souches pluripotentes induites/métabolisme , Muscles lisses vasculaires/métabolisme , Hypertrichose/génétique , Hypertrichose/métabolisme , Hypertrichose/physiopathologie , Hypertrichose/anatomopathologie , Animaux , Souris , Myocytes du muscle lisse/métabolisme , Canaux KATP/génétique , Canaux KATP/métabolisme , Ostéochondrodysplasies/génétique , Ostéochondrodysplasies/métabolisme , Ostéochondrodysplasies/anatomopathologie , Ostéochondrodysplasies/physiopathologie , Mutation , Différenciation cellulaire/génétique , Techniques de patch-clamp , Cardiomégalie , Récepteurs des sulfonylurées
5.
Ecotoxicol Environ Saf ; 282: 116695, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38981390

RÉSUMÉ

Microplastics (MPs) are recognized as a major environmental problem due to their ubiquitous presence in ecosystems and bioaccumulation in food chains. Not only humans are continuously exposed to these pollutants through ingestion and inhalation, but recent findings suggest they may trigger vascular inflammation and potentially worsen the clinical conditions of cardiovascular patients. Here we combine headspace analysis by needle trap microextraction-gas chromatography-mass spectrometry (HS-NTME-GC-MS) and biological assays to evaluate the effects of polystyrene, high- and low-density polyethylene MPs on phenotype, metabolic activity, and pro-inflammatory status of Vascular Smooth Muscle Cells (VSMCs) the most prominent cells in vascular walls. Virgin and artificially aged MPs (4 weeks at 40 °C and 750 W/m2 simulated solar irradiation) were comparatively tested at 1 mg/mL to simulate a realistic exposure scenario. Our results clearly show the activation of oxidative stress and inflammatory processes when VSMCs were cultured with aged polymers, with significant overexpression of IL-6 and TNF-α. In addition, volatile organic compounds (VOCs), including pentane, acrolein, propanal, and hexanal as the main components, were released by VSMCs into the headspace. Type-specific VOC response profiles were induced on vascular cells from different MPs.


Sujet(s)
Chromatographie gazeuse-spectrométrie de masse , Inflammation , Interleukine-6 , Microplastiques , Stress oxydatif , Microplastiques/toxicité , Inflammation/induit chimiquement , Stress oxydatif/effets des médicaments et des substances chimiques , Interleukine-6/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Composés organiques volatils/toxicité , Polystyrènes/toxicité , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/cytologie , Humains , Polyéthylène/toxicité , Cellules cultivées , Myocytes du muscle lisse/effets des médicaments et des substances chimiques
6.
Ecotoxicol Environ Saf ; 281: 116681, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964063

RÉSUMÉ

Fluoride exposure has been implicated as a potential risk factor for hypertension, but the underlying mechanisms remain unclear. This study investigated the role of the RhoA/ROCK signaling pathway in fluoride-induced hypertension. Male Wistar rats were divided into different groups and exposed to varying concentrations of sodium fluoride (NaF) or sodium chloride (NaCl) via drinking water. The rats' blood pressure was measured, and their aortic tissue was utilized for high-throughput sequencing analysis. Additionally, rat and A7r5 cell models were established using NaF and/or Fasudil. The study evaluated the effects of fluoride exposure on blood pressure, pathological changes in the aorta, as well as the protein/mRNA expression levels of phenotypic transformation indicators (a-SMA, calp, OPN) in vascular smooth muscle cells (VSMCs), along with the RhoA/ROCK signaling pathway (RhoA, ROCK1, ROCK2, MLC/p-MLC). The results demonstrated that fluoride exposure in rats led to increased blood pressure. High-throughput sequencing analysis revealed differential gene expression associated with vascular smooth muscle contraction, with the RhoA/ROCK signaling pathway emerging as a key regulator. Pathological changes in the rat aorta, such as elastic membrane rupture and collagen fiber deposition, were observed following NaF exposure. However, fasudil, a ROCK inhibitor, mitigated these pathological changes. Both in vitro and in vivo models confirmed the activation of the RhoA/ROCK signaling pathway and the phenotypic transformation of VSMCs from a contractile to a synthetic state upon fluoride exposure. Fasudil effectively inhibited the activities of ROCK1 and ROCK2 and attenuated the phenotypic transformation of VSMCs. In conclusion, fluoride has the potential to induce hypertension through the activation of the RhoA/ROCK signaling pathway and phenotypic changes in vascular smooth muscle cells. These results provide new insights into the mechanism of fluoride-induced hypertension.


Sujet(s)
Hypertension artérielle , Muscles lisses vasculaires , Rat Wistar , Transduction du signal , rho-Associated Kinases , Animaux , rho-Associated Kinases/métabolisme , Mâle , Hypertension artérielle/induit chimiquement , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/anatomopathologie , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Protéine G RhoA/métabolisme , Fluorure de sodium/toxicité , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Phénotype , Pression sanguine/effets des médicaments et des substances chimiques , Fluorures/toxicité , Protéines G rho
7.
Redox Biol ; 73: 103203, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38823208

RÉSUMÉ

Vascular smooth muscle cells (VSMCs), known for their remarkable lifelong phenotypic plasticity, play a pivotal role in vascular pathologies through their ability to transition between different phenotypes. Our group discovered that the deficiency of the mitochondrial protein Poldip2 induces VSMC differentiation both in vivo and in vitro. Further comprehensive biochemical investigations revealed Poldip2's specific interaction with the mitochondrial ATPase caseinolytic protease chaperone subunit X (CLPX), which is the regulatory subunit for the caseinolytic protease proteolytic subunit (ClpP) that forms part of the ClpXP complex - a proteasome-like protease evolutionarily conserved from bacteria to humans. This interaction limits the protease's activity, and reduced Poldip2 levels lead to ClpXP complex activation. This finding prompted the hypothesis that ClpXP complex activity within the mitochondria may regulate the VSMC phenotype. Employing gain-of-function and loss-of-function strategies, we demonstrated that ClpXP activity significantly influences the VSMC phenotype. Notably, both genetic and pharmacological activation of ClpXP inhibits VSMC plasticity and fosters a quiescent, differentiated, and anti-inflammatory VSMC phenotype. The pharmacological activation of ClpP using TIC10, currently in phase III clinical trials for cancer, successfully replicates this phenotype both in vitro and in vivo and markedly reduces aneurysm development in a mouse model of elastase-induced aortic aneurysms. Our mechanistic exploration indicates that ClpP activation regulates the VSMC phenotype by modifying the cellular NAD+/NADH ratio and activating Sirtuin 1. Our findings reveal the crucial role of mitochondrial proteostasis in the regulation of the VSMC phenotype and propose the ClpP protease as a novel, actionable target for manipulating the VSMC phenotype.


Sujet(s)
Endopeptidase Clp , Mitochondries , Muscles lisses vasculaires , Myocytes du muscle lisse , Phénotype , Sirtuine-1 , Animaux , Humains , Souris , Différenciation cellulaire , Endopeptidase Clp/métabolisme , Endopeptidase Clp/génétique , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/cytologie , Myocytes du muscle lisse/métabolisme , Sirtuine-1/métabolisme , Sirtuine-1/génétique
8.
Biomed Pharmacother ; 176: 116858, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38850669

RÉSUMÉ

The roles and mechanisms of A-kinase anchoring protein 1 (AKAP1) in vascular smooth muscle cell (VSMC) phenotypic modulation and neointima formation are currently unknown. AKAP1 is a mitochondrial PKA-anchored protein and maintains mitochondrial homeostasis. This study aimed to investigate how AKAP1/PKA signaling plays a protective role in inhibiting VSMC phenotypic transformation and neointima formation by regulating mitochondrial fission. The results showed that both PDGF-BB treatment and balloon injury reduced the transcription, expression, and mitochondrial anchoring of AKAP1. In vitro, the overexpression of AKAP1 significantly inhibited PDGF-BB mediated VSMC proliferation and migration, whereas AKAP1 knockdown further aggravated VSMC phenotypic transformation. Additionally, in the balloon injury model in vivo, AKAP1 overexpression reduced neointima formation, the muscle fiber area ratio, and rat VSMC proliferation and migration. Furthermore, PDGF-BB and balloon injury inhibited Drp1 phosphorylation at Ser637 and promoted Drp1 activity and mitochondrial midzone fission; AKAP1 overexpression reversed these effects. AKAP1 overexpression also inhibited the distribution of mitochondria at the plasma membrane and the reduction of PKARIIß expression induced by PDGF-BB, as evidenced by an increase in mitochondria-plasma membrane distance as well as PKARIIß protein levels. Moreover, the PKA agonist promoted Drp1 phosphorylation (Ser637) and inhibited PDGF-BB-mediated mitochondrial fission, cell proliferation, and migration. The PKA antagonist reversed the increase in Drp1 phosphorylation (Ser637) and the decline in mitochondrial midzone fission and VSMC phenotypic transformation caused by AKAP1 overexpression. The results of this study reveal that AKAP1 protects VSMCs against phenotypic modulation by improving Drp1 phosphorylation at Ser637 through PKA and inhibiting mitochondrial fission, thereby preventing neointima formation.


Sujet(s)
Protéines d'ancrage aux protéines kinases A , Dynamines , Muscles lisses vasculaires , Néointima , Animaux , Mâle , Rats , Protéines d'ancrage aux protéines kinases A/métabolisme , Protéines d'ancrage aux protéines kinases A/génétique , Bécaplermine/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cyclic AMP-Dependent Protein Kinases/métabolisme , Dynamines/métabolisme , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Néointima/métabolisme , Néointima/anatomopathologie , Phénotype , Phosphorylation , Rat Sprague-Dawley , Transduction du signal
9.
Article de Anglais | MEDLINE | ID: mdl-38822974

RÉSUMÉ

Vascular remodeling is the adaptive response of the vessel wall to physiological and pathophysiological changes, closely linked to vascular diseases. Vascular smooth muscle cells (VSMCs) play a crucial role in this process. Pyroptosis, a form of programmed cell death characterized by excessive release of inflammatory factors, can cause phenotypic transformation of VSMCs, leading to their proliferation, migration, and calcification-all of which accelerate vascular remodeling. Inhibition of VSMC pyroptosis can delay this process. This review summarizes the impact of pyroptosis on VSMCs and the pathogenic role of VSMC pyroptosis in vascular remodeling. We also discuss inhibitors of key proteins in pyroptosis pathways and their effects on VSMC pyroptosis. These findings enhance our understanding of the pathogenesis of vascular remodeling and provide a foundation for the development of novel medications that target the control of VSMC pyroptosis as a potential treatment strategy for vascular diseases.

10.
Aging (Albany NY) ; 16(11): 9369-9385, 2024 05 27.
Article de Anglais | MEDLINE | ID: mdl-38809515

RÉSUMÉ

Vascular aging exacerbates diabetes-associated vascular damage, a major cause of microvascular and macrovascular complications. This study aimed to elucidate key genes and pathways underlying vascular aging in diabetes using integrated bioinformatics and machine learning approaches. Gene expression datasets related to vascular smooth muscle cell (VSMC) senescence and diabetic vascular aging were analyzed. Differential expression analysis identified 428 genes associated with VSMC senescence. Functional enrichment revealed their involvement in cellular senescence, ECM-receptor interaction, PI3K-Akt and AGE-RAGE signaling pathways. Further analysis of diabetic vascular aging datasets revealed 52 differentially expressed genes, enriched in AMPK signaling, AGE-RAGE signaling, cellular senescence, and VEGF signaling pathways. Machine learning algorithms, including LASSO regression and SVM-RFE, pinpointed six key genes: TFB1M, FOXRED2, LY75, DALRD3, PI4K2B, and NDOR1. Immune cell infiltration analysis demonstrated correlations between diabetic vascular aging, the identified key genes, and infiltration levels of plasma cells, M1 macrophages, CD8+ T cells, eosinophils, and regulatory T cells. In conclusion, this study identified six pivotal genes (TFB1M, FOXRED2, LY75, DALRD3, PI4K2B, and NDOR1) closely associated with diabetic vascular aging through integrative bioinformatics and machine learning approaches. These genes are linked to alterations in the immune microenvironment during diabetic vascular aging. This study provides a reference and basis for molecular mechanism research, biomarker mining, and diagnosis and treatment evaluation of diabetes-related vascular aging.


Sujet(s)
Vieillissement , Biologie informatique , Apprentissage machine , Humains , Vieillissement/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Transduction du signal/génétique , Diabète/génétique , Vieillissement de la cellule/génétique
11.
Cells ; 13(9)2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38727271

RÉSUMÉ

Vascular smooth muscle cells (VSMCs) play a key role in aortic aneurysm formation. Bone morphogenetic proteins (BMPs) have been implicated as important regulators of VSMC phenotype, and dysregulation of the BMP pathway has been shown to be associated with vascular diseases. The aim of this study was to investigate for the first time the effects of BMP-4 on the VSMC phenotype and to understand its role in the development of thoracic aortic aneurysms (TAAs). Using the angiotensin II (AngII) osmotic pump model in mice, aortas from mice with VSMC-specific BMP-4 deficiency showed changes similar to AngII-infused aortas, characterised by a loss of contractile markers, increased fibrosis, and activation of matrix metalloproteinase 9. When BMP-4 deficiency was combined with AngII infusion, there was a significantly higher rate of apoptosis and aortic dilatation. In vitro, VSMCs with mRNA silencing of BMP-4 displayed a dedifferentiated phenotype with activated canonical BMP signalling. In contrast, BMP-2-deficient VSMCs exhibited the opposite phenotype. The compensatory regulation between BMP-2 and BMP-4, with BMP-4 promoting the contractile phenotype, appeared to be independent of the canonical signalling pathway. Taken together, these results demonstrate the impact of VSMC-specific BMP-4 deficiency on TAA development.


Sujet(s)
Anévrysme de l'aorte thoracique , Protéine morphogénétique osseuse de type 2 , Protéine morphogénétique osseuse de type 4 , Muscles lisses vasculaires , Myocytes du muscle lisse , Animaux , Mâle , Souris , Angiotensine-II/pharmacologie , Anévrysme de l'aorte thoracique/métabolisme , Anévrysme de l'aorte thoracique/anatomopathologie , Anévrysme de l'aorte thoracique/génétique , Apoptose/effets des médicaments et des substances chimiques , Protéine morphogénétique osseuse de type 2/métabolisme , Protéine morphogénétique osseuse de type 4/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Phénotype , Transduction du signal
12.
Phytomedicine ; 130: 155704, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38759316

RÉSUMÉ

BACKGROUND: Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties. PURPOSE: This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism. METHODS: In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation. RESULTS: We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-ß. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-ß-MEK -ERK-ELK-1/KLF4 signaling pathway. CONCLUSION: Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-ß-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.


Sujet(s)
Anthraquinones , Dédifférenciation cellulaire , Facteur-4 de type Kruppel , Muscles lisses vasculaires , Néointima , Animaux , Mâle , Souris , Rats , Anthraquinones/pharmacologie , Artériosclérose/traitement médicamenteux , Artériosclérose/prévention et contrôle , Athérosclérose/traitement médicamenteux , Bécaplermine/pharmacologie , Lésions traumatiques de l'artère carotide/traitement médicamenteux , Dédifférenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Alimentation riche en graisse , Modèles animaux de maladie humaine , Facteurs de transcription Krüppel-like/métabolisme , Souris de lignée C57BL , Simulation de docking moléculaire , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Néointima/traitement médicamenteux , Rat Sprague-Dawley , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
13.
Arch Intern Med Res ; 7(2): 73-79, 2024.
Article de Anglais | MEDLINE | ID: mdl-38737892

RÉSUMÉ

Atherosclerosis, a critical contributor to coronary artery diseases, involves the accumulation of cholesterol, fibrin, and lipids within arterial walls, inciting inflammatory reactions culminating in plaque formation. This multifaceted interplay encompasses excessive fibrosis, fatty plaque development, vascular smooth muscle cell (VSMC) proliferation, and leukocyte migration in response to inflammatory pathways. While stable plaques demonstrate resilience against complications, vulnerable ones, with lipid-rich cores, necrosis, and thin fibrous caps, lead to thrombosis, myocardial infarction, stroke, and acute cerebrovascular accidents. The nuanced phenotypes of VSMCs, modulated by gene regulation and environmental cues, remain pivotal. Essential markers like alpha-SMA, myosin heavy chain, and calponin regulate VSMC migration and contraction, exhibiting diminished expression during VSMC de-differentiation and proliferation. p27kip, a CDK inhibitor, shows promise in regulating VSMC proliferation and appears associated with TNF-α-induced pathways impacting unstable plaques. Oncostatin M (OSM), an IL-6 family cytokine, correlates with MMP upregulation and foam cell formation, influencing plaque development. Efforts targeting mammalian target of rapamycin (mTOR) inhibition, notably using rapamycin and its analogs, demonstrate potential but pose challenges due to associated adverse effects. Exploration of the impact of p27kip impact on plaque macrophages presents promising avenues, yet its complete therapeutic potential remains untapped. Similarly, while OSM has exhibited potential in inducing cell cycle arrest via p27kip, direct links necessitate further investigation. This critical review discusses the role of mTOR, p27kip, and OSM in VSMC proliferation and differentiation followed by the therapeutic potential of targeting these mediators in atherosclerosis to attenuate plaque vulnerability.

14.
J Adv Res ; 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38821358

RÉSUMÉ

INTRODUCTION: Sympathetic hyperinnervation plays an important role in modulating the vascular smooth muscle cell (VSMC) phenotype and vascular diseases, but its role in abdominal aortic aneurysm (AAA) is still unknown. OBJECTIVES: This study aimed to investigate the role of sympathetic hyperinnervation in promoting AAA development and the underlying mechanism involved. METHODS: Western blotting and immunochemical staining were used to detect sympathetic hyperinnervation. We performed sympathetic denervation through coeliac ganglionectomy (CGX) and 6-OHDA administration to understand the role of sympathetic hyperinnervation in AAA and investigated the underlying mechanisms through transcriptome and functional studies. Sema4D knockout (Sema4D-/-) mice were utilized to determine the involvement of Sema4D in inducing sympathetic hyperinnervation and AAA development. RESULTS: We observed sympathetic hyperinnervation, the most important form of sympathetic neural remodeling, in both mouse AAA models and AAA patients. Elimination of sympathetic hyperinnervation by CGX or 6-OHDA significantly inhibited AAA development and progression. We further revealed that sympathetic hyperinnervation promoted VSMC phenotypic switching in AAA by releasing extracellular ATP (eATP) and activating eATP-P2rx4-p38 signaling. Moreover, single-cell RNA sequencing revealed that Sema4D secreted by osteoclast-like cells induces sympathetic nerve diffusion and hyperinnervation through binding to Plxnb1. We consistently observed that AAA progression was significantly ameliorated in Sema4D-deficient mice. CONCLUSIONS: Sympathetic hyperinnervation driven by osteoclast-like cell-derived Sema4D promotes VSMC phenotypic switching and accelerates pathological aneurysm progression by activating the eATP/P2rx4/p38 pathway. Inhibition of sympathetic hyperinnervation emerges as a potential novel therapeutic strategy for preventing and treating AAA.

15.
Cells ; 13(8)2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38667273

RÉSUMÉ

Vascular smooth muscle cells (VSMCs), in their contractile and differentiated state, are fundamental for maintaining vascular function. Upon exposure to cholesterol (CHO), VSMCs undergo dedifferentiation, adopting characteristics of foam cells-lipid-laden, macrophage-like cells pivotal in atherosclerotic plaque formation. CHO uptake by VSMCs leads to two primary pathways: ABCA1-mediated efflux or storage in lipid droplets as cholesterol esters (CEs). CE formation, involving the condensation of free CHO and fatty acids, is catalyzed by sterol O-acyltransferase 1 (SOAT1). The necessary fatty acids are synthesized by the lipogenic enzyme fatty acid synthase (FASN), which we found to be upregulated in atherosclerotic human coronary arteries. This observation led us to hypothesize that FASN-mediated fatty acid biosynthesis is crucial in the transformation of VSMCs into foam cells. Our study reveals that CHO treatment upregulates FASN in human aortic SMCs, concurrent with increased expression of CD68 and upregulation of KLF4, markers associated with the foam cell transition. Crucially, downregulation of FASN inhibits the CHO-induced upregulation of CD68 and KLF4 in VSMCs. Additionally, FASN-deficient VSMCs exhibit hindered lipid accumulation and an impaired transition to the foam cell phenotype following CHO exposure, while the addition of the fatty acid palmitate, the main FASN product, exacerbates this transition. FASN-deficient cells also show decreased SOAT1 expression and elevated ABCA1. Notably, similar effects are observed in KLF4-deficient cells. Our findings demonstrate that FASN plays an essential role in the CHO-induced upregulation of KLF4 and the VSMC to foam cell transition and suggest that targeting FASN could be a novel therapeutic strategy to regulate VSMC phenotypic modulation.


Sujet(s)
Cellules spumeuses , Facteur-4 de type Kruppel , Muscles lisses vasculaires , Animaux , Humains , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Cholestérol/métabolisme , Fatty acid synthases/métabolisme , Fatty acid synthases/génétique , Acides gras/métabolisme , Cellules spumeuses/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/cytologie , Myocytes du muscle lisse/métabolisme
16.
Am J Physiol Cell Physiol ; 326(6): C1721-C1734, 2024 06 01.
Article de Anglais | MEDLINE | ID: mdl-38646788

RÉSUMÉ

Atherosclerosis (AS) is a significant contributor to cardio-cerebrovascular ischemia diseases, resulting in high mortality rates worldwide. During AS, vascular smooth muscle cells (VSMCs) play a crucial role in plaque formation by undergoing phenotypic and osteogenic switching. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has previously been identified as a nuclear regulator that promotes tumorigenesis and metastasis, but its role in regulating VSMCs in AS remains unclear. Our study aimed to investigate the biological functions and specific mechanisms of NEAT1 in regulating VSMCs in AS. We found that NEAT1 was upregulated in the aortas of AS mouse models and dedifferentiated primary VSMCs. Silencing NEAT1 in vitro attenuated the proliferation, migration, and osteogenic differentiation of VSMCs, while NEAT1 overexpression had the opposite effect. Furthermore, NEAT1 promoted VSMC osteogenic differentiation and vascular calcification in both in vivo and in vitro vascular calcification models. We also discovered that NEAT1 directly activates enhancer of zeste homolog 2 (EZH2), an epigenetic enzyme that suppresses the expression of senescence- and antimigration-related genes, by translocating it into the nucleus. CUT&Tag assay revealed that NEAT1 guides EZH2 to the promoters of senescence-related genes (P16, P21, and TIMP3), methylating local histones to reduce their transcription. Our findings suggest that NEAT1 functions in AS by modulating the epigenetic function of EZH2, which enhances the proliferation, migration, and osteogenic differentiation of VSMCs. This study provides new insights into the molecular mechanisms underlying the pathogenesis of AS and highlights the potential of NEAT1 as a therapeutic target of AS.NEW & NOTEWORTHY Our study demonstrates that the upregulation of long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) promotes proliferation and migration during phenotypic switching of vascular smooth muscle cells in atherosclerosis. We also provide in vivo and in vitro evidence that NEAT1 accelerates vascular calcification. Our findings identified the direct interaction between enhancer of zeste homolog 2 (EZH2) and NEAT1 during atherosclerosis. NEAT1 is necessary for EZH2 to translocate from the cytoplasm to the nucleus, where EZH2 epigenetically inhibits the expression of genes related to senescence and antimigration.


Sujet(s)
Athérosclérose , Différenciation cellulaire , Protéine-2 homologue de l'activateur de Zeste , Muscles lisses vasculaires , Myocytes du muscle lisse , Ostéogenèse , ARN long non codant , Calcification vasculaire , ARN long non codant/génétique , ARN long non codant/métabolisme , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Animaux , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Ostéogenèse/génétique , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Calcification vasculaire/anatomopathologie , Calcification vasculaire/génétique , Calcification vasculaire/métabolisme , Souris , Mâle , Souris de lignée C57BL , Prolifération cellulaire , Phénotype , Cellules cultivées , Humains , Mouvement cellulaire
17.
Cardiovasc Diagn Ther ; 14(1): 72-83, 2024 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-38434568

RÉSUMÉ

Background: In recent years, a mass of studies have shown that pyroptosis plays an important role in the proliferation of vascular smooth muscle cells (VSMCs). We investigated whether angiotensin II (Ang II) induces the pyroptosis of rat aortic VSMCs and the role of NOD-like receptor family pyrin domain containing 3 (NLRP3) in this process. Additionally, we explored the effect and related mechanism of recombinant tissue factor pathway inhibitor (rTFPI) in Ang II-induced VSMC pyroptosis. Methods: Cultured VSMCs were divided into five groups: control group, Ang II group (1×10-5 mol/L), MCC950 group (NLRP3 inhibitor, 15 nmol/L), Ang II + MCC950 group and Ang II + rTFPI (50 µg/L) group. Cell viability was measured by cell counting kit-8 (CCK8) assays and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays. Propidium iodide (PI) staining and immunofluorescence were performed to determine the pyroptosis of VSMCs. Changes in VSMC ultrastructure were evaluated through transmission electron microscopy. The expression levels of NLRP3, pro-caspase-1, gasdermin D-N (GSDMD-N), and interleukin-1ß (IL-1ß) were determined by western blot analysis. Results: The cell viability, the positive rate of PI staining, and the expression level of GSDMD detected by immunofluorescence in the Ang II group were higher than that in the control group, whereas they all decreased in Ang II + MCC950 group and Ang II + rTFPI group compared with Ang II group (P<0.05). Electron microscopy analysis revealed less extracellular matrix, increased myofilaments, and decreased endoplasmic reticulum, Golgi complex, and mitochondria in Ang II + rTFPI-treated VSMCs than in Ang II-treated VSMCs. The protein expression levels of the pyroptosis-related molecules NLRP3, pro-caspase-1, GSDMD-N, and IL-1ß in Ang II group showed an increasing trend compared with those in control group (P<0.05); however, these expression levels in Ang II + MCC950 and Ang II + rTFPI groups were significantly lower than those in Ang II group (P<0.05). Conclusions: Ang II may induce pyroptosis in VSMCs by activating NLRP3. rTFPI can inhibit Ang II-induced VSMC pyroptosis. Furthermore, rTFPI might exert this effect by inhibiting the NLRP3 pathway and therefore play an important role in the treatment of vascular remodeling induced by hypertension.

18.
Front Cardiovasc Med ; 11: 1276066, 2024.
Article de Anglais | MEDLINE | ID: mdl-38374991

RÉSUMÉ

Background: Carotid artery atherosclerosis is a major cause of ischemic stroke, and ischemic stroke is the leading cause of morbidity and mortality worldwide. Unfortunately, the reason for the build-up of atherosclerosis plaque is unknown. The miRNA-29c was reported to promote the phenotype transformation of vascular smooth muscle cells (VSMCs) in diabetes mice, eventually leading to plaque formation and bleeding. However, such studies are rare and limited to animal experiments. Methods: In our study, 40 patients were divided into a diabetic mellitus (DM) group and a non-DM group according to whether they were diagnosed with DM. Then, the real-time quantitative PCR was applied to examine the miRNA-29c level in human carotid plaque tissue derived from 40 subjects receiving carotid endarterectomy. Results: Briefly, diabetes patients had a decreased miRNA-29c level as compared with non-DM subjects, and this comparison was statistically significant (P = 0.02). Notably, variable miRNA-29c level was negatively associated with HbA1c level, although no statistical significance was observed. Moreover, there was an increased miRNA-29c level in patients with cerebral stroke. Conclusion: Collectively, the miRNA-29c level in the carotid plaque is closely associated with DM and cerebral stroke, which may contribute to atherosclerosis formation.

19.
Eur J Pharmacol ; 968: 176397, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38331337

RÉSUMÉ

Abdominal aortic aneurysm (AAA), a vascular degenerative disease, is a potentially life-threatening condition characterised by the loss of vascular smooth muscle cells (VSMCs), degradation of extracellular matrix (ECM), inflammation, and oxidative stress. Despite the severity of AAA, effective drugs for treatment are scarce. At low doses, terazosin (TZ) exerts antiapoptotic and anti-inflammatory effects in several diseases, but its potential to protect against AAA remains unexplored. Herein, we investigated the effects of TZ in two AAA animal models: Angiotensin II (Ang II) infusion in Apoe-/- mice and calcium chloride application in C57BL/6J mice. Mice were orally administered with TZ (100 or 1000 µg/kg/day). The in vivo results indicated that low-dose TZ alleviated AAA formation in both models. Low-dose TZ significantly reduced aortic pulse wave velocity without exerting an apparent antihypertensive effect in the Ang II-induced AAA model. Paternally expressed gene 3 (Peg3) was identified via RNA sequencing as a novel TZ target. PEG3 expression was significantly elevated in both mouse and human AAA tissues. TZ suppressed PEG3 expression and reduced the abundance of matrix metalloproteinases (MMP2/MMP9) in the tunica media. Functional experiments and molecular analyses revealed that TZ (10 nM) treatment and Peg3 knockdown effectively prevented Ang II-induced VSMC senescence and apoptosis in vitro. Thus, Peg3, a novel target of TZ, mediates inflammation-induced VSMC apoptosis and senescence. Low-dose TZ downregulates Peg3 expression to attenuate AAA formation and ECM degradation, suggesting a promising therapeutic strategy for AAA.


Sujet(s)
Anévrysme de l'aorte abdominale , Muscles lisses vasculaires , Prazosine/analogues et dérivés , Souris , Humains , Animaux , Analyse de l'onde de pouls , Souris knockout , Souris de lignée C57BL , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/traitement médicamenteux , Anévrysme de l'aorte abdominale/génétique , Apoptose , Inflammation/métabolisme , Angiotensine-II/pharmacologie , Angiotensine-II/métabolisme , Modèles animaux de maladie humaine , Myocytes du muscle lisse , Facteurs de transcription Krüppel-like/métabolisme
20.
Clin Exp Pharmacol Physiol ; 51(4): e13845, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38382550

RÉSUMÉ

Abnormalities in vascular smooth muscle cells (VSMCs) are pivotal in the pathogenesis of cardiovascular pathologies such as atherosclerosis and hypertension. Scutellarin (Scu), a flavonoid derived from marigold flowers, exhibits a spectrum of biological activities including anti-inflammatory, antioxidant, antitumor, immunomodulatory and antimicrobial effects. Notably, Scu has demonstrated the capacity to mitigate vascular endothelial damage and prevent atherosclerosis via its antioxidative properties. Nevertheless, the influence of Scu on the formation of VSMC-derived foam cells remains underexplored. In this study, Scu was evidenced to efficaciously attenuate oleic acid (OA)-induced lipid accumulation and the upregulation of adipose differentiation-associated protein Plin2 in a dose- and time-responsive manner. We elucidated that Scu effectively diminishes OA-provoked VSMC foam cell formation. Further, it was established that Scu pretreatment augments the protein expression of LC3B-II and the mRNA levels of Map1lc3b and Becn1, concurrently diminishing the protein levels of the NLRP3 inflammasome compared to the OA group. Activation of autophagy through rapamycin attenuated NLRP3 inflammasome protein expression, intracellular lipid droplet content and Plin2 mRNA levels. Scu also counteracted the OA-induced decrement of LC3B-II levels in the presence of bafilomycin-a1, facilitating the genesis of autophagosomes and autolysosomes. Complementarily, in vivo experiments revealed that Scu administration substantially reduced arterial wall thickness, vessel wall cross-sectional area, wall-to-lumen ratio and serum total cholesterol levels in comparison to the high-fat diet model group. Collectively, our findings suggest that Scu attenuates OA-induced VSMC foam cell formation through the induction of autophagy and the suppression of NLRP3 inflammasome activation.


Sujet(s)
Apigénine , Athérosclérose , Glucuronates , Inflammasomes , Humains , Inflammasomes/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Cellules spumeuses/métabolisme , Cellules spumeuses/anatomopathologie , Muscles lisses vasculaires/métabolisme , Acide oléique/pharmacologie , Acide oléique/métabolisme , Athérosclérose/métabolisme , Autophagie , ARN messager/métabolisme , Myocytes du muscle lisse/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE