Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 218
Filtrer
1.
Atherosclerosis ; 397: 118582, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39260002

RÉSUMÉ

BACKGROUND AND AIMS: Lysyl oxidase (LOX) catalyzes the crosslinking of collagen and elastin to maintain tensile strength and structural integrity of the vasculature. Excessive LOX activity increases vascular stiffness and the severity of occlusive diseases. Herein, we investigated the mechanisms by which LOX controls atherogenesis and osteogenic differentiation of vascular smooth muscle cells (SMC) in hyperlipidemic mice. METHODS: Gene inactivation of Lox in SMC was achieved in conditional knockout mice after tamoxifen injections. Atherosclerosis burden and vascular calcification were assessed in hyperlipidemic conditional [Loxf/fMyh11-CreERT2ApoE-/-] and sibling control mice [Loxwt/wtMyh11-CreERT2ApoE-/-]. Mechanistic studies were performed with primary aortic SMC from Lox mutant and wild type mice. RESULTS: Inactivation of Lox in SMCs decreased > 70 % its RNA expression and protein level in the aortic wall and significantly reduced LOX activity without compromising vascular structure and function. Moreover, LOX deficiency protected mice against atherosclerotic burden (13 ± 2 versus 23 ± 1 %, p < 0.01) and plaque calcification (5 ± 0.4 versus 11.8 ± 3 %, p < 0.05) compared to sibling controls. Interestingly, gene inactivation of Lox in SMCs preserved the contractile phenotype of vascular SMC under hyperlipidemic conditions as demonstrated by single-cell RNA sequencing and immunofluorescence. Mechanistically, the absence of LOX in SMC prevented excessive collagen crosslinking and the subsequent activation of the pro-osteogenic FAK/ß-catenin signaling axis. CONCLUSIONS: Lox inactivation in SMC protects mice against atherosclerosis and plaque calcification by reducing SMC modulation and FAK/ß-catenin signaling.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Hyperlipidémies , Souris knockout , Muscles lisses vasculaires , Myocytes du muscle lisse , Plaque d'athérosclérose , Lysyloxidase , Calcification vasculaire , Animaux , Lysyloxidase/métabolisme , Lysyloxidase/génétique , Myocytes du muscle lisse/enzymologie , Myocytes du muscle lisse/anatomopathologie , Myocytes du muscle lisse/métabolisme , Athérosclérose/génétique , Athérosclérose/enzymologie , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/enzymologie , Muscles lisses vasculaires/métabolisme , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie , Calcification vasculaire/enzymologie , Calcification vasculaire/prévention et contrôle , Calcification vasculaire/métabolisme , Hyperlipidémies/génétique , Hyperlipidémies/enzymologie , Hyperlipidémies/complications , Hyperlipidémies/métabolisme , Souris , Ostéogenèse , Cellules cultivées , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/métabolisme , Aorte/anatomopathologie , Aorte/enzymologie , Aorte/métabolisme , Mâle , Souris de lignée C57BL , bêta-Caténine/métabolisme , Transduction du signal , Protéines de la matrice extracellulaire
2.
Atherosclerosis ; 396: 118544, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39126769

RÉSUMÉ

BACKGROUND AND AIMS: Inflammatory cells within atherosclerotic lesions secrete proteolytic enzymes that contribute to lesion progression and destabilization, increasing the risk for an acute cardiovascular event. Elastase is a serine protease, secreted by macrophages and neutrophils, that may contribute to the development of unstable plaque. We previously reported interaction of endogenous protease-inhibitor proteins with high-density lipoprotein (HDL), including alpha-1-antitrypsin, an inhibitor of elastase. These findings support a potential role for HDL as a modulator of protease activity. In this study, we test the hypothesis that enhancement of HDL-associated elastase inhibitor activity is protective against atherosclerotic lesion progression. METHODS: We designed an HDL-targeting protease inhibitor (HTPI) that binds to HDL and confers elastase inhibitor activity. Lipoprotein binding and the impact of HTPI on atherosclerosis were examined using mouse models. Histology and immunofluorescence staining of aortic root sections were used to examine the impact of HTPI on lesion morphology and inflammatory features. RESULTS: HTPI is a small (1.6 kDa) peptide with an elastase inhibitor domain, a soluble linker, and an HDL-targeting domain. When incubated with human plasma ex vivo, HTPI predominantly binds to HDL. Intravenous administration of HTPI to mice resulted in its binding to plasma HDL and increased elastase inhibitor activity on isolated HDL. Accumulation of HTPI within plaque was observed after administration to Apoe-/- mice. To examine the effect of HTPI treatment on atherosclerosis, prevention and progression studies were performed using Ldlr-/- mice fed Western diet. In both study designs, HTPI-treated mice had reduced lipid deposition in plaque. CONCLUSIONS: These data support the hypothesis that HDL-associated anti-elastase activity can improve the atheroprotective potential of HDL and highlight the potential utility of HDL enrichment with anti-protease activity as an approach for stabilization of atherosclerotic lesions.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Évolution de la maladie , Lipoprotéines HDL , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/prévention et contrôle , Athérosclérose/enzymologie , Athérosclérose/métabolisme , Athérosclérose/traitement médicamenteux , Lipoprotéines HDL/métabolisme , Humains , Souris de lignée C57BL , Souris , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Mâle , Pancreatic elastase/métabolisme , Aorte/anatomopathologie , Aorte/effets des médicaments et des substances chimiques , Aorte/enzymologie , Aorte/métabolisme , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/métabolisme , Inhibiteurs de protéases/pharmacologie , alpha-1-Antitrypsine/pharmacologie , alpha-1-Antitrypsine/métabolisme
3.
Atherosclerosis ; 396: 118531, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38996716

RÉSUMÉ

BACKGROUND AND AIMS: Diabetes is one of the major causes of cardiovascular disease (CVD). As high as 29 % of patients with diabetes develop atherosclerosis. Vascular Smooth Muscle Cells (VSMCs) are a key mediator in the pathogenesis of atherosclerosis, generating pro-inflammatory and proliferative characteristics in atherosclerotic lesions. METHODS: We used human atherosclerotic samples, developed diabetes-induced atherosclerotic mice, and generated loss of function and gain of function in Klotho human aortic smooth muscle cells to investigate the function of Klotho in atherosclerosis. RESULTS: We found that Klotho expression is decreased in smooth muscle actin-positive cells in patients with diabetes and atherosclerosis. Consistent with human data, we found that Apoe knockout mice with streptozotocin-induced diabetes fed on a high-fat diet showed decreased expression of Klotho in SMCs. Additionally, these mice showed increased expression of TGF-ß, MMP9, phosphorylation of ERK and Akt. Further, we utilized primary Human Aortic Smooth Muscle Cells (HASMCs) with d-glucose under dose-response and in time-dependent conditions to study the role of Klotho in these cells. Klotho gain of function and loss of function studies showed that Klotho inversely regulated the expression of atherosclerotic markers TGF-ß, MMP2, MMP9, and Fractalkine. Further, High Glucose (HG) induced Akt, and ERK1/2 phosphorylation were enhanced or mitigated by endogenous Klotho deficiency or its overexpression respectively. PI3K/Akt and MAPK/ERK inhibition partially abolished the HG-induced upregulation of TGF-ß, MMP2, MMP9, and Fractalkine. Additionally, Klotho knockdown increased the proliferation of HASMCs and enhanced α-SMA and TGF-ß expression. CONCLUSIONS: Taken together, these results indicate that local vascular Klotho is involved in diabetes-induced atherosclerosis, which is via PI3K/Akt and ERK1/2-dependent signaling pathways.


Sujet(s)
Athérosclérose , Diabète expérimental , Glucuronidase , Protéines Klotho , Souris invalidées pour les gènes ApoE , Muscles lisses vasculaires , Myocytes du muscle lisse , Protéines Klotho/métabolisme , Animaux , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Athérosclérose/génétique , Glucuronidase/métabolisme , Glucuronidase/génétique , Humains , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Diabète expérimental/métabolisme , Diabète expérimental/complications , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Mâle , Transduction du signal , Cellules cultivées , Aorte/anatomopathologie , Aorte/métabolisme , Système de signalisation des MAP kinases , Souris , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/métabolisme , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Souris de lignée C57BL , Protéines proto-oncogènes c-akt/métabolisme , Prolifération cellulaire
4.
Atherosclerosis ; 392: 117519, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38581737

RÉSUMÉ

BACKGROUND AND AIMS: Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS: We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS: Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS: This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.


Sujet(s)
Aorte , Athérosclérose , Glucuronidase , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Animaux , Mâle , Souris , Aorte/anatomopathologie , Aorte/métabolisme , Aorte/enzymologie , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Maladies de l'aorte/métabolisme , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/métabolisme , Alimentation riche en graisse , Modèles animaux de maladie humaine , Évolution de la maladie , Glucuronidase/déficit , Glucuronidase/génétique , Glucuronidase/métabolisme , Souris de lignée C57BL , Souris knockout , Nécrose , Sinus de l'aorte/anatomopathologie , Molécule-1 d'adhérence des cellules vasculaires/métabolisme
5.
Arterioscler Thromb Vasc Biol ; 44(6): 1346-1364, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38660806

RÉSUMÉ

BACKGROUND: Atherosclerosis is the major underlying pathology of cardiovascular disease and is driven by dyslipidemia and inflammation. Inhibition of the immunoproteasome, a proteasome variant that is predominantly expressed by immune cells and plays an important role in antigen presentation, has been shown to have immunosuppressive effects. METHODS: We assessed the effect of ONX-0914, an inhibitor of the immunoproteasomal catalytic subunits LMP7 (proteasome subunit ß5i/large multifunctional peptidase 7) and LMP2 (proteasome subunit ß1i/large multifunctional peptidase 2), on atherosclerosis and metabolism in LDLr-/- and APOE*3-Leiden.CETP mice. RESULTS: ONX-0914 treatment significantly reduced atherosclerosis, reduced dendritic cell and macrophage levels and their activation, as well as the levels of antigen-experienced T cells during early plaque formation, and Th1 cells in advanced atherosclerosis in young and aged mice in various immune compartments. Additionally, ONX-0914 treatment led to a strong reduction in white adipose tissue mass and adipocyte progenitors, which coincided with neutrophil and macrophage accumulation in white adipose tissue. ONX-0914 reduced intestinal triglyceride uptake and gastric emptying, likely contributing to the reduction in white adipose tissue mass, as ONX-0914 did not increase energy expenditure or reduce total food intake. Concomitant with the reduction in white adipose tissue mass upon ONX-0914 treatment, we observed improvements in markers of metabolic syndrome, including lowered plasma triglyceride levels, insulin levels, and fasting blood glucose. CONCLUSIONS: We propose that immunoproteasomal inhibition reduces 3 major causes underlying cardiovascular disease, dyslipidemia, metabolic syndrome, and inflammation and is a new target in drug development for atherosclerosis treatment.


Sujet(s)
Tissu adipeux blanc , Athérosclérose , Modèles animaux de maladie humaine , Syndrome métabolique X , Souris de lignée C57BL , Proteasome endopeptidase complex , Récepteurs aux lipoprotéines LDL , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/prévention et contrôle , Athérosclérose/traitement médicamenteux , Athérosclérose/immunologie , Athérosclérose/génétique , Athérosclérose/métabolisme , Syndrome métabolique X/traitement médicamenteux , Syndrome métabolique X/immunologie , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Tissu adipeux blanc/anatomopathologie , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/déficit , Proteasome endopeptidase complex/métabolisme , Mâle , Inhibiteurs du protéasome/pharmacologie , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Maladies de l'aorte/immunologie , Maladies de l'aorte/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/immunologie , Plaque d'athérosclérose , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris invalidées pour les gènes ApoE , Souris , Métabolisme énergétique/effets des médicaments et des substances chimiques , Oligopeptides
6.
Microvasc Res ; 140: 104276, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34742813

RÉSUMÉ

PURPOSE: We previously reported that a calpain inhibitor (CAI) prevents the development of atherosclerosis in rats. This study aimed to investigate the effects of CAI (1 mg/kg) on atherosclerosis in apolipoprotein E knockout (ApoE KO) mice that were fed a high-fat diet (HFD) and explore the underlying mechanism by analyzing the expression of genes related to the uptake and efflux of cholesterol. METHODS: Atherosclerotic plaques were evaluated. The activity of calpain in the aorta and that of superoxide dismutase (SOD) in the serum were assessed. Lipid profiles in the serum and liver were examined. Serum oxidized low-density lipoprotein (oxLDL), malondialdehyde (MDA), tumor necrosis factor (TNF-α), and interleukin-6 (IL-6) levels were measured. The mRNA expressions of CD68, TNF-α, IL-6, CD36, scavenger receptor (SR-A), peroxisome proliferator-activated receptor gamma (PPAR-γ), liver-x-receptor alpha (LXR-α), and ATP-binding cassette transporter class A1 (ABCA1) in the aorta and peritoneal macrophages were also evaluated. RESULTS: CAI reduced calpain activity in the aorta. CAI also impeded atherosclerotic lesion formation and mRNA expression of CD68 in the aorta and peritoneal macrophages of ApoE KO mice compared with those of mice receiving HFD. However, CAI had no effect on body weight and lipid levels in both the serum and liver. CAI significantly decreased MDA, oxLDL, TNF-α, and IL-6 levels and increased SOD activity in the serum. Moreover, CAI significantly inhibited the mRNA expression of TNF-α and IL-6 genes in the aorta and peritoneal macrophages. In addition, CAI significantly downregulated the mRNA expression of scavenger receptors CD36 and SR-A and upregulated the expression of genes involved in the cholesterol efflux pathway, i.e., PPAR-γ, LXR-α, and ABCA1 in the aorta and peritoneal macrophages. CONCLUSIONS: CAI inhibited the development of atherosclerotic lesions in ApoE KO mice, and this effect might be related to the reduction of oxidative stress and inflammation and the improvement of cholesterol intake and efflux pathways.


Sujet(s)
Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Calpain/antagonistes et inhibiteurs , Cholestérol/métabolisme , Inhibiteurs de la cystéine protéinase/pharmacologie , Leupeptines/pharmacologie , Métabolisme lipidique/effets des médicaments et des substances chimiques , Macrophages péritonéaux/effets des médicaments et des substances chimiques , ARN messager/métabolisme , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/génétique , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/métabolisme , Animaux , Antigènes CD/génétique , Antigènes CD/métabolisme , Antigènes de différenciation des myélomonocytes/génétique , Antigènes de différenciation des myélomonocytes/métabolisme , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Calpain/métabolisme , Modèles animaux de maladie humaine , Régulation de l'expression des gènes , Métabolisme lipidique/génétique , Récepteurs hépatiques X/génétique , Récepteurs hépatiques X/métabolisme , Macrophages péritonéaux/enzymologie , Macrophages péritonéaux/anatomopathologie , Mâle , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Récepteur PPAR gamma/génétique , Récepteur PPAR gamma/métabolisme , Plaque d'athérosclérose , ARN messager/génétique , Récepteurs éboueurs de classe A/génétique , Récepteurs éboueurs de classe A/métabolisme
7.
Clin Sci (Lond) ; 135(21): 2483-2502, 2021 11 12.
Article de Anglais | MEDLINE | ID: mdl-34643227

RÉSUMÉ

Vascular calcification is highly prevalent in chronic kidney disease (CKD), and is characterized by transdifferentiation from contractile vascular smooth muscle cells (VSMCs) into an osteogenic phenotype. However, no effective and therapeutic option to prevent vascular calcification is yet available. Dihydromyricetin (DMY), a bioactive flavonoid isolated from Ampelopsis grossedentata, has been found to inhibit VSMCs proliferation and the injury-induced neointimal formation. However, whether DMY has an effect on osteogenic differentiation of VSMCs and vascular calcification is still unclear. In the present study, we sought to investigate the effect of DMY on vascular calcification in CKD and the underlying mechanism. DMY treatment significantly attenuated calcium/phosphate-induced calcification of rat and human VSMCs in a dose-dependent manner, as shown by Alizarin Red S staining and calcium content assay, associated with down-regulation of osteogenic markers including type I collagen (COL I), Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2) and osteocalcin (OCN). These results were further confirmed in aortic rings ex vivo. Moreover, DMY ameliorated vascular calcification in rats with CKD. Additionally, we found that AKT signaling was activated during vascular calcification, whereas significantly inhibited by DMY administration. DMY treatment significantly reversed AKT activator-induced vascular calcification. Furthermore, inhibition of AKT signaling efficiently attenuated calcification, which was similar to that after treatment with DMY alone, and DMY had a better inhibitory effect on calcification as compared with AKT inhibitor. The present study demonstrated that DMY has a potent inhibitory role in vascular calcification partially by inhibiting AKT activation, suggesting that DMY may act as a promising therapeutic candidate for patients suffering from vascular calcification.


Sujet(s)
Maladies de l'aorte/prévention et contrôle , Flavonols/pharmacologie , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Ostéogenèse/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Insuffisance rénale chronique/traitement médicamenteux , Calcification vasculaire/prévention et contrôle , Animaux , Aorte/effets des médicaments et des substances chimiques , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/étiologie , Maladies de l'aorte/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Humains , Mâle , Muscles lisses vasculaires/enzymologie , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/enzymologie , Myocytes du muscle lisse/anatomopathologie , Phosphorylation , Protéines proto-oncogènes c-akt/génétique , Rat Sprague-Dawley , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/enzymologie , Insuffisance rénale chronique/anatomopathologie , Transduction du signal , Calcification vasculaire/enzymologie , Calcification vasculaire/étiologie , Calcification vasculaire/anatomopathologie
8.
Physiol Res ; 70(4): 533-542, 2021 08 31.
Article de Anglais | MEDLINE | ID: mdl-34062069

RÉSUMÉ

Valsartan has the potential to attenuate neointimal hyperplasia and to suppress the inflammatory response. This study aimed to evaluate the role of valsartan in neointimal hyperplasia and the toll-like receptor 4 (TLR4)-nitric oxide synthase (NOS) pathway in the balloon-injured rat aorta.Forty-eight Wistar rats were randomly allocated to three groups: sham control (control), balloon-injured group (surgery), and balloon-injured+valsartan-treated group (valsartan). Rats were killed at 14 and 28 days after balloon-injury, and then the aortic tissues were collected for morphometric analysis as well as for measurements of the mRNA or protein expression of angiotensin II, angiotensin II type 1 (AT1) receptor, angiotensin II type 2 (AT2) receptor, TLR4, endothelial nitric oxide synthase (eNOS), inducible NOS (iNOS), serine/arginine-rich splicing factor 1(SRSF1) and extracellular signal regulated kinase (ERK). Valsartan at a dose of 20 mg/kg/day markedly decreased neointimal hyperplasia in the aorta of balloon-injured rats, and significantly reduced the mRNA or protein expression of TLR4, AT1 receptor, SRSF1 and phosphorylated-ERK (p-ERK) as well as the aortic levels of iNOS (all p < 0.05). Moreover, valsartan increased the eNOS level and AT2 receptor mRNA and protein expression levels (all p < 0.05). Valsartan prevented neointimal hyperplasia and inhibited SRSF1 expression and the TLR4-iNOS-ERK-AT1 receptor pathway in the balloon-injured rat aorta.


Sujet(s)
Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/traitement médicamenteux , Extracellular Signal-Regulated MAP Kinases/métabolisme , Néointima , Nitric oxide synthase type II/métabolisme , Récepteur de type 1 à l'angiotensine-II/métabolisme , Facteurs d'épissage riches en sérine-arginine/métabolisme , Récepteur de type Toll-4/métabolisme , Valsartan/pharmacologie , Lésions du système vasculaire/traitement médicamenteux , Animaux , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Modèles animaux de maladie humaine , Hyperplasie , Mâle , Phosphorylation , Rat Wistar , Récepteur de type 1 à l'angiotensine-II/génétique , Transduction du signal , Récepteur de type Toll-4/génétique , Lésions du système vasculaire/enzymologie , Lésions du système vasculaire/génétique , Lésions du système vasculaire/anatomopathologie
10.
Arterioscler Thromb Vasc Biol ; 41(6): e338-e353, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33792343
11.
J Cell Physiol ; 236(6): 4750-4763, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33615471

RÉSUMÉ

Excessively high cholesterol content in the blood leads to nonalcohol fatty liver disease (NAFLD) and arteriosclerosis. Although there are increasing publications and patent applications to lower blood cholesterol with small chemical molecules, limited effective drugs can be available in clinic. It is necessary to uncover new targets and drugs to alleviate high cholesterol. Esterase D (ESD) is abundant in liver and it remains unknown about its role in cholesterol metabolism. Here we reported that small chemical molecule fluorescigenic pyrazoline derivative 5 (FPD5), a new ESD activator, could effectively reverse high blood cholesterol level and prevent fatty liver and arteriosclerosis in apoE-/- mice fed the high-fat diet. We also observed that FPD5 could reduce oxidized low density lipoprotein (oxLDL)-induced formation of foam cells. To further investigate the mechanism of FPD5 action on blood cholesterol modulation, we found that ESD trigged by FPD5 was aggregated in lysosome and interacted with Jun activation domain binding protein 1 (JAB1). ESD served as a deacetylase to remove Thr89 acetylation of JAB1 and increased its activity; thus, promoting the ATP-binding cassette transporters A1 (ABCA1) to accelerate cholesterol efflux. Our findings demonstrate that FPD5 decreases blood cholesterol level to ameliorate NAFLD and arteriosclerosis through ESD/JAB1/ABCA1 pathway, and ESD functions as a novel nonclassical deacetylase that hydrolyzes serine/threonine acetyl group. Our findings not only highlight that FPD5 may be a pioneer drug for alleviating blood cholesterol but also indicate that ESD is a potential drug target that promotes cholesterol metabolism.


Sujet(s)
Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/métabolisme , Anticholestérolémiants/pharmacologie , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Complexe du signalosome COP9/métabolisme , Cholestérol/sang , Antienzymes/pharmacologie , Cellules spumeuses/effets des médicaments et des substances chimiques , Peptide hydrolases/métabolisme , Thiolester hydrolases/antagonistes et inhibiteurs , Acétylation , Animaux , Maladies de l'aorte/sang , Maladies de l'aorte/enzymologie , Maladies de l'aorte/anatomopathologie , Athérosclérose/sang , Athérosclérose/enzymologie , Athérosclérose/anatomopathologie , Marqueurs biologiques/sang , Alimentation riche en graisse , Modèles animaux de maladie humaine , Régulation négative , Cellules spumeuses/enzymologie , Cellules spumeuses/anatomopathologie , Cellules HEK293 , Humains , Mâle , Souris , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Stéatose hépatique non alcoolique/enzymologie , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/prévention et contrôle , Plaque d'athérosclérose , Maturation post-traductionnelle des protéines , Cellules RAW 264.7 , Thiolester hydrolases/métabolisme
12.
Cardiovasc Res ; 117(5): 1295-1308, 2021 04 23.
Article de Anglais | MEDLINE | ID: mdl-32667970

RÉSUMÉ

AIMS: Atherosclerotic vascular disease has an inflammatory pathogenesis. Heme from intraplaque haemorrhage may drive a protective and pro-resolving macrophage M2-like phenotype, Mhem, via AMPK and activating transcription factor 1 (ATF1). The antidiabetic drug metformin may also activate AMPK-dependent signalling. Hypothesis: Metformin systematically induces atheroprotective genes in macrophages via AMPK and ATF1, thereby suppresses atherogenesis. METHODS AND RESULTS: Normoglycaemic Ldlr-/- hyperlipidaemic mice were treated with oral metformin, which profoundly suppressed atherosclerotic lesion development (P < 5 × 10-11). Bone marrow transplantation from AMPK-deficient mice demonstrated that metformin-related atheroprotection required haematopoietic AMPK [analysis of variance (ANOVA), P < 0.03]. Metformin at a clinically relevant concentration (10 µM) evoked AMPK-dependent and ATF1-dependent increases in Hmox1, Nr1h2 (Lxrb), Abca1, Apoe, Igf1, and Pdgf, increases in several M2-markers and decreases in Nos2, in murine bone marrow macrophages. Similar effects were seen in human blood-derived macrophages, in which metformin-induced protective genes and M2-like genes, suppressible by si-ATF1-mediated knockdown. Microarray analysis comparing metformin with heme in human macrophages indicated that the transcriptomic effects of metformin were related to those of heme, but not identical. Metformin-induced lesional macrophage expression of p-AMPK, p-ATF1, and downstream M2-like protective effects. CONCLUSION: Metformin activates a conserved AMPK-ATF1-M2-like pathway in mouse and human macrophages, and results in highly suppressed atherogenesis in hyperlipidaemic mice via haematopoietic AMPK.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Facteur de transcription ATF-1/métabolisme , Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Macrophages/effets des médicaments et des substances chimiques , Metformine/pharmacologie , Plaque d'athérosclérose , AMP-Activated Protein Kinases/génétique , Facteur de transcription ATF-1/génétique , Animaux , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Régulation de l'expression des gènes , Humains , Macrophages/enzymologie , Macrophages/anatomopathologie , Souris knockout , Phénotype , Phosphorylation , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Transduction du signal
13.
Cardiovasc Res ; 117(4): 1060-1069, 2021 03 21.
Article de Anglais | MEDLINE | ID: mdl-32402085

RÉSUMÉ

AIMS: Fibroblast activation protein (FAP) is upregulated at sites of tissue remodelling including chronic arthritis, solid tumours, and fibrotic hearts. It has also been associated with human coronary atherosclerotic plaques. Yet, the causal role of FAP in atherosclerosis remains unknown. To investigate the cause-effect relationship of endogenous FAP in atherogenesis, we assessed the effects of constitutive Fap deletion on plaque formation in atherosclerosis-prone apolipoprotein E (Apoe) or low-density lipoprotein receptor (Ldlr) knockout mice. METHODS AND RESULTS: Using en face analyses of thoraco-abdominal aortae and aortic sinus cross-sections, we demonstrate that Fap deficiency decreased plaque formation in two atherosclerotic mouse models (-46% in Apoe and -34% in Ldlr knockout mice). As a surrogate of plaque vulnerability fibrous cap thickness was used; it was increased in Fap-deficient mice, whereas Sirius red staining demonstrated that total collagen content remained unchanged. Using polarized light, atherosclerotic lesions from Fap-deficient mice displayed increased FAP targets in terms of enhanced collagen birefringence in plaques and increased pre-COL3A1 expression in aortic lysates. Analyses of the Stockholm Atherosclerosis Gene Expression data revealed that FAP expression was increased in human atherosclerotic compared to non-atherosclerotic arteries. CONCLUSIONS: Our data provide causal evidence that constitutive Fap deletion decreases progression of experimental atherosclerosis and increases features of plaque stability with decreased collagen breakdown. Thus, inhibition of FAP expression or activity may not only represent a promising therapeutic target in atherosclerosis but appears safe at the experimental level for FAP-targeted cancer therapies.


Sujet(s)
Aorte/enzymologie , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Endopeptidases/déficit , Protéines membranaires/déficit , Remodelage vasculaire , Animaux , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Études cas-témoins , Collagène/génétique , Collagène/métabolisme , Modèles animaux de maladie humaine , Endopeptidases/génétique , Fibrose , Délétion de gène , Humains , Lipides/sang , Mâle , Protéines membranaires/génétique , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Muscles lisses vasculaires/enzymologie , Muscles lisses vasculaires/anatomopathologie , Plaque d'athérosclérose , Protéome , Récepteurs aux lipoprotéines LDL/déficit , Récepteurs aux lipoprotéines LDL/génétique , Transcriptome
14.
Vascul Pharmacol ; 133-134: 106777, 2020.
Article de Anglais | MEDLINE | ID: mdl-32750408

RÉSUMÉ

Atherosclerosis is a systemic chronic inflammatory disease. Many antioxidants including alpha-lipoic acid (LA), a product of lipoic acid synthase (Lias), have proven to be effective for treatment of this disease. However, the question remains whether LA regulates the immune response as a protective mechanism against atherosclerosis. We initially investigated whether enhanced endogenous antioxidant can retard the development of atherosclerosis via immunomodulation. To explore the impact of enhanced endogenous antioxidant on the retardation of atherosclerosis via immune regulation, our laboratory has recently created a double mutant mouse model, using apolipoprotein E-deficient (Apoe-/-) mice crossbred with mice overexpressing lipoic acid synthase gene (LiasH/H), designated as LiasH/HApoe-/- mice. Their littermates, Lias+/+Apoe-/- mice, served as a control. Distinct redox environments between the two strains of mice have been established and they can be used to facilitate identification of antioxidant targets in the immune response. At 6 months of age, LiasH/HApoe-/- mice had profoundly decreased atherosclerotic lesion size in the aortic sinus compared to their Lias+/+Apoe-/- littermates, accompanied by significantly enhanced numbers of regulatory T cells (Tregs) and anti-oxidized LDL autoantibody in the vascular system, and reduced T cell infiltrates in aortic walls. Our results represent a novel exploration into an environment with increased endogenous antioxidant and its ability to alleviate atherosclerosis, likely through regulation of the immune response. These outcomes shed light on a new therapeutic strategy using antioxidants to lessen atherosclerosis.


Sujet(s)
Aorte/enzymologie , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Plaque d'athérosclérose , Sulfurtransferases/biosynthèse , Animaux , Aorte/immunologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/immunologie , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/immunologie , Athérosclérose/anatomopathologie , Autoanticorps/sang , Modèles animaux de maladie humaine , Induction enzymatique , Lipoprotéines LDL/immunologie , Mâle , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Oxydoréduction , Stress oxydatif , Sulfurtransferases/génétique , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme
15.
BMC Cardiovasc Disord ; 20(1): 133, 2020 03 13.
Article de Anglais | MEDLINE | ID: mdl-32169038

RÉSUMÉ

BACKGROUND: Previous studies have indicated that the JAK/STAT signaling pathway is involved in modulating arterial adventitia inflammation response. In this study, we designed experiments to further investigate the effect of JAK2/STAT3/SOCS3 signaling in rabbit atherosclerosis process. METHODS: Atherosclerosis was induced in the abdominal arteries of rabbits by balloon injury of the aorta supplemented by the atherogenic diet. Simultaneously, in the process of atherosclerosis, animals underwent either ruxolitinib treatment or not for 12 weeks. At the end of the experimental period, all rabbits were sacrificed. The plaque areas in abdominal artery, the lipid burden of plaque and the calcium burden of plaque were detected by H&E staining, Oil Red O staining and Alizarin Red staining, respectively. In addition, rabbit plasma lipids and inflammatory cytokines were measured by biochemical test kits or ELISA kits. Finally, the expression and phosphorylation levels of JAK2/STAT3/SOCS3 pathway-related proteins were detected by RT-qPCR, western blot and immunohistochemistry assays. RESULTS: H&E staining and CT scan analysis showed that rabbit atherosclerosis model was constructed successfully. Ruxolitinib, an inhibitor of the Janus kinase 2 (JAK2), substantially reduced the area of atherosclerotic plaques in rabbits treated with high fat diet and balloon injury of the aorta. Moreover, ruxolitinib significantly decreased IL-6, IL-1ß, IFN-γ and TNF-α, but increased IL-10 and IL-17 levels in plasma of atherosclerotic rabbits. Additionally, ruxolitinib reduced plasma TC, TG and LDL-C contents and AIP value, while enhanced HDL-C level in atherosclerotic rabbits. Furthermore, we found that JAK2 and STAT3 phosphorylation were up-regulated in rabbits with atherosclerosis when compared with those of the control group, followed by the expression of SOCS3 was also increased due to the activation of JAK2 and STAT3. Interestingly, ruxolitinib could inactivate JAK2 and STAT3 pathway and decrease SOCS3 expression. CONCLUSION: Taken together, the inhibition of JAK2/STAT3/SOCS3 signaling pathway may be a novel method for the clinical treatment of artery atherosclerosis.


Sujet(s)
Aorte abdominale/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Kinase Janus-2/antagonistes et inhibiteurs , Inhibiteurs des Janus kinases/pharmacologie , Plaque d'athérosclérose , Pyrazoles/pharmacologie , Facteur de transcription STAT-3/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Animaux , Aorte abdominale/enzymologie , Aorte abdominale/anatomopathologie , Maladies de l'aorte/sang , Maladies de l'aorte/enzymologie , Maladies de l'aorte/anatomopathologie , Athérosclérose/sang , Athérosclérose/enzymologie , Athérosclérose/anatomopathologie , Cytokines/sang , Modèles animaux de maladie humaine , Médiateurs de l'inflammation/sang , Kinase Janus-2/métabolisme , Lipides/sang , Mâle , Nitriles , Phosphorylation , Pyrimidines , Lapins , Transduction du signal
16.
Arterioscler Thromb Vasc Biol ; 40(5): 1155-1167, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32212851

RÉSUMÉ

OBJECTIVES: During the advancement of atherosclerosis, plaque cellularity is governed by the influx of monocyte-derived macrophages and their turnover via apoptotic and nonapoptotic forms of cell death. Previous reports have demonstrated that programmed necrosis, or necroptosis, of plaque macrophages contribute to necrotic core formation. Knockdown or inhibition of the necrosome components RIPK1 (receptor-interacting protein kinase 1) and RIPK3 (receptor-interacting protein kinase 3) slow atherogenesis, and activation of the terminal step of necroptosis, MLKL (mixed lineage kinase domain-like protein), has been demonstrated in advanced human atherosclerotic plaques. However, whether MLKL directly contributes to lesion development and necrotic core formation has not been investigated. Approaches and Results: MLKL expression was knocked down in atherogenic Apoe-knockout mice via the administration of antisense oligonucleotides. During atherogenesis, Mlkl knockdown decreased both programmed cell death and the necrotic core in the plaque. However, total lesion area remained unchanged. Furthermore, treatment with the MLKL antisense oligonucleotide unexpectedly reduced circulating cholesterol levels compared with control antisense oligonucleotide but increased the accumulation of lipids within the plaque and in vitro in macrophage foam cells. MLKL colocalized with the late endosome and multivesicular bodies in peritoneal macrophages incubated with atherogenic lipoproteins. Transfection with MLKL antisense oligonucleotide increased lipid localization with the multivesicular bodies, suggesting that upon Mlkl knockdown, lipid trafficking becomes defective leading to enhanced lipid accumulation in macrophages. CONCLUSIONS: These studies confirm the requirement for MLKL as the executioner of necroptosis, and as such a significant contributor to the necrotic core during atherogenesis. We also identified a previously unknown role for MLKL in regulating endosomal trafficking to facilitate lipid handling in macrophages during atherogenesis.


Sujet(s)
Maladies de l'aorte/enzymologie , Athérosclérose/enzymologie , Cholestérol/métabolisme , Cellules spumeuses/enzymologie , Macrophages péritonéaux/enzymologie , Plaque d'athérosclérose , Protein kinases/déficit , Animaux , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Modèles animaux de maladie humaine , Endosomes/métabolisme , Femelle , Cellules spumeuses/anatomopathologie , Macrophages péritonéaux/anatomopathologie , Mâle , Souris invalidées pour les gènes ApoE , Nécroptose , Nécrose , Oligonucléotides antisens/administration et posologie , Protein kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Transduction du signal
17.
Cardiovasc Drugs Ther ; 34(2): 145-152, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32086626

RÉSUMÉ

OBJECTIVE: Increased myelopoiesis has been linked to risk of atherosclerotic cardiovascular disease (ACD). Excessive myelopoiesis can be driven by dyslipidemia and cholesterol accumulation in hematopoietic stem and progenitor cells (HSPC) and may involve increased signaling via Janus kinase 2 (JAK2). Constitutively activating JAK2 mutants drive biased myelopoiesis and promote development of myeloproliferative neoplasms (MPN) or clonal hematopoiesis, conditions associated with increased risk of ACD. JAK2 inhibitors have been developed as a therapy for MPNs. The potential for JAK2 inhibitors to protect against atherosclerosis has not been tested. We therefore assessed the impact of JAK2 inhibition on atherogenesis. METHODS: A selective JAK2 inhibitor TG101348 (fedratinib) or vehicle was given to high-fat high-cholesterol Western diet (WD)-fed wild-type (WT) or Apoe-/- mice. Hematopoietic cell profiles, cell proliferation, and atherosclerosis in WT or Apoe-/- mice were assessed. RESULTS: TG101348 selectively reversed neutrophilia, monocytosis, HSPC, and granulocyte-macrophage progenitor (GMP) expansion in Apoe-/- mice with decreased cellular phosphorylated STAT5 and ERK1/2 and reduced cell cycling and BrdU incorporation in HSPCs, indicating inhibition of JAK/STAT signaling and cell proliferation. Ten-week WD feeding allowed the development of marked aortic atherosclerosis in Apoe-/- mice which was substantially reduced by TG101348. CONCLUSIONS: Selective JAK2 inhibition reduces atherogenesis by suppressing excessive myelopoiesis in hypercholesterolemic Apoe-/- mice. These findings suggest selective JAK2 inhibition as a potential therapeutic approach to decrease ACD risk in patients with increased myelopoiesis and leukocytosis.


Sujet(s)
Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Kinase Janus-2/antagonistes et inhibiteurs , Inhibiteurs des Janus kinases/pharmacologie , Myélopoïèse/effets des médicaments et des substances chimiques , Pyrrolidines/pharmacologie , Sulfonamides/pharmacologie , Animaux , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Modèles animaux de maladie humaine , Femelle , Cellules souches hématopoïétiques/enzymologie , Cellules souches hématopoïétiques/anatomopathologie , Kinase Janus-2/métabolisme , Hyperleucocytose/enzymologie , Hyperleucocytose/prévention et contrôle , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Transduction du signal
18.
J Cardiovasc Pharmacol ; 75(4): 321-332, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-31895870

RÉSUMÉ

Artemisinin is an endoperoxide sesquiterpene lactone from Artemisia annua L with multiple beneficial effects, including anti-inflammation, antioxidant, and vascular protection. Recent studies have found that inflammation along with autophagy deficiency in macrophages is the possible reason for foam cell accumulation in the intima, which leads to atherosclerotic plaque formation. The primary aims of this study were to explore the inhibiting effect of artemisinin on atherosclerosis in high-fat diet-fed ApoE mice and investigate the probable mechanism. Artemisinin (50 and 100 mg/kg, intragastric administration) treatment effectively inhibited foamy macrophage transformation and decreased atherosclerotic plaque formation in atherosclerotic mice. Moreover, artemisinin promoted AMP-activated protein kinase (AMPK) activation, inhibited mammalian target of rapamycin (mTOR) and uncoordinated-51-like kinase 1 (ULK1) phosphorylation, and increased LC-3II accumulation and P62 degradation, thereby enhancing macrophage autophagy. Besides, the inhibiting effect of artemisinin on mTOR and ULK1 phosphorylation could be abrogated by AMPK knockdown, suggesting AMPK was the essential target of artemisinin on promoting macrophage autophagy. Our study indicated that artemisinin alleviated atherosclerotic lesions by accelerating macrophage autophagy through the AMPK/mTOR/ULK1 pathway.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Anti-inflammatoires/pharmacologie , Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Artémisinines/pharmacologie , Athérosclérose/prévention et contrôle , Autophagie/effets des médicaments et des substances chimiques , Cellules spumeuses/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Animaux , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Homologue de la protéine-1 associée à l'autophagie/métabolisme , Alimentation riche en graisse , Modèles animaux de maladie humaine , Cellules spumeuses/enzymologie , Cellules spumeuses/anatomopathologie , Lipoprotéines LDL/toxicité , Mâle , Souris , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Protéines associées aux microtubules/métabolisme , Phosphorylation , Plaque d'athérosclérose , Cellules RAW 264.7 , Séquestosome-1/métabolisme , Transduction du signal
19.
J Pathol ; 250(1): 30-41, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31509234

RÉSUMÉ

Medial arterial calcification (MAC) is a major complication of chronic kidney disease (CKD) and an indicator of poor prognosis. Aortic overexpression of tissue-nonspecific alkaline phosphatase (TNAP) accelerates MAC formation. The present study aimed to assess whether a TNAP inhibitor, SBI-425, protects against MAC and improves survival probability in a CKD-mineral and bone disorder (MBD) mouse model. CKD-MBD mice were divided in three groups: vehicle, SBI-10, and SBI-30. They were fed a 0.2% adenine and 0.8% phosphorus diet from 14 to 20 weeks of age to induce CKD, followed by a high-phosphorus (0.2% adenine and 1.8% phosphorus) diet for another 6 weeks. At 14-20 weeks of age, mice in the SBI-10 and SBI-30 groups were given 10 and 30 mg/kg SBI-425 by gavage once a day, respectively, while vehicle-group mice were given distilled water as vehicle. Control mice were fed a standard chow (0.8% phosphorus) between the ages of 8 and 20 weeks. Computed tomography imaging, histology, and aortic tissue calcium content revealed that, compared to vehicle animals, SBI-425 nearly halted the formation of MAC. Mice in the control, SBI-10 and SBI-30 groups exhibited 100% survival, which was significantly better than vehicle-treated mice (57.1%). Aortic mRNA expression of Alpl, encoding TNAP, as well as plasma and aortic tissue TNAP activity, were suppressed by SBI-425 administration, whereas plasma pyrophosphate increased. We conclude that a TNAP inhibitor successfully protected the vasculature from MAC and improved survival rate in a mouse CKD-MBD model, without causing any adverse effects on normal skeletal formation and residual renal function. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Sujet(s)
Phosphatase alcaline/antagonistes et inhibiteurs , Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Ostéodystrophie rénale/traitement médicamenteux , Antienzymes/pharmacologie , Nicotinamide/analogues et dérivés , Sulfonamides/pharmacologie , Calcification vasculaire/prévention et contrôle , Phosphatase alcaline/génétique , Phosphatase alcaline/métabolisme , Animaux , Aorte/enzymologie , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/étiologie , Maladies de l'aorte/anatomopathologie , Ostéodystrophie rénale/complications , Ostéodystrophie rénale/enzymologie , Ostéodystrophie rénale/anatomopathologie , Modèles animaux de maladie humaine , Régulation de l'expression des gènes , Mâle , Souris de lignée C57BL , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/enzymologie , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/enzymologie , Myocytes du muscle lisse/anatomopathologie , Nicotinamide/pharmacologie , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/enzymologie , Ostéoblastes/anatomopathologie , Facteurs temps , Calcification vasculaire/enzymologie , Calcification vasculaire/étiologie , Calcification vasculaire/anatomopathologie
20.
Atherosclerosis ; 291: 78-86, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31704554

RÉSUMÉ

BACKGROUND AND AIMS: Atherosclerosis is a chronic inflammatory disorder mediated by macrophage activation. MicroRNA-21 (miR-21) is a key regulator in the macrophage inflammatory response. However, the functional role of miR-21 in atherogenesis is far from clear. METHODS AND RESULTS: Here, we report that miR-21 is significantly upregulated in mouse atherosclerotic plaques and peripheral monocytes from patients with coronary artery disease. Compared with miR-21+/+apoE-/- mice (apoE-/- mice), miR-21-/-apoE-/- (double knockout, DKO) mice showed less atherosclerotic lesions, reduced presence of macrophages, decreased smooth muscle cells(SMC) and collagen content in the aorta. We further explored the role of miR-21 in macrophage activation in vitro. Bone marrow-derived macrophages (BMDMs) from DKO mice not only exhibit impaired function of migration induced by chemokine (C-C motif) ligand 2 (CCL2) but also a weakened macrophage-endothelium interaction activated by tumor necrosis factor-α (TNF-α). However, atherogenic inflammatory cytokine secretion was not affected by miR-21 in vitro or in vivo. Additionally, miR-21 knockdown in BMDMs directly derepressed the expression of dual specificity protein phosphatase 8 (Dusp-8), a previously validated miR-21 target in cardiac fibroblasts, which negatively regulates mitogen-activated protein kinase (MAPK) signaling, particularly the p38-and c-Jun N-terminal kinase (JNK)-related signaling pathways. CONCLUSIONS: These data demonstrate that inhibition of miR-21 may restrict the formation of atherosclerotic plaques partly by regulating macrophage migration and adhesion, while, reduced SMCs and collagen content in plaques may lead to a less stable phenotype with the progression of atherosclerosis. Thus, the absence of miR-21 reduces atherosclerotic lesions but may not represent all benefit in atherosclerosis development.


Sujet(s)
Aorte/enzymologie , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Chimiotaxie , Dual-specificity phosphatases/métabolisme , Activation des macrophages , Macrophages/enzymologie , microARN/métabolisme , Animaux , Aorte/anatomopathologie , Maladies de l'aorte/enzymologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Adhérence cellulaire , Modèles animaux de maladie humaine , Dual-specificity phosphatases/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Humains , JNK Mitogen-Activated Protein Kinases/métabolisme , Macrophages/anatomopathologie , Mâle , Souris , Souris invalidées pour les gènes ApoE , microARN/génétique , Plaque d'athérosclérose , Cellules RAW 264.7 , Transduction du signal , p38 Mitogen-Activated Protein Kinases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE