Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.153
Filtrer
2.
Cells ; 13(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38994980

RÉSUMÉ

The Ectonucleotide Pyrophosphatase/Phosphodiesterase 1 (ENPP1) ectoenzyme regulates vascular intimal proliferation and mineralization of bone and soft tissues. ENPP1 variants cause Generalized Arterial Calcification of Infancy (GACI), a rare genetic disorder characterized by ectopic calcification, intimal proliferation, and stenosis of large- and medium-sized arteries. ENPP1 hydrolyzes extracellular ATP to pyrophosphate (PPi) and AMP. AMP is the precursor of adenosine, which has been implicated in the control of neointimal formation. Herein, we demonstrate that an ENPP1-Fc recombinant therapeutic inhibits proliferation of vascular smooth muscle cells (VSMCs) in vitro and in vivo. Addition of ENPP1 and ATP to cultured VSMCs generated AMP, which was metabolized to adenosine. It also significantly decreased cell proliferation. AMP or adenosine alone inhibited VSMC growth. Inhibition of ecto-5'-nucleotidase CD73 decreased adenosine accumulation and suppressed the anti-proliferative effects of ENPP1/ATP. Addition of AMP increased cAMP synthesis and phosphorylation of VASP at Ser157. This AMP-mediated cAMP increase was abrogated by CD73 inhibitors or by A2aR and A2bR antagonists. Ligation of the carotid artery promoted neointimal hyperplasia in wild-type mice, which was exacerbated in ENPP1-deficient ttw/ttw mice. Prophylactic or therapeutic treatments with ENPP1 significantly reduced intimal hyperplasia not only in ttw/ttw but also in wild-type mice. These findings provide the first insight into the mechanism of the anti-proliferative effect of ENPP1 and broaden its potential therapeutic applications beyond enzyme replacement therapy.


Sujet(s)
5'-Nucleotidase , Adénosine , Prolifération cellulaire , Muscles lisses vasculaires , Myocytes du muscle lisse , Phosphodiesterases , Pyrophosphatases , Transduction du signal , Phosphodiesterases/métabolisme , Phosphodiesterases/génétique , Pyrophosphatases/métabolisme , Pyrophosphatases/génétique , 5'-Nucleotidase/métabolisme , 5'-Nucleotidase/génétique , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Adénosine/métabolisme , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Souris , Humains , AMP/métabolisme , Souris de lignée C57BL , AMP cyclique/métabolisme , Mâle , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Calcification vasculaire/génétique
3.
Ren Fail ; 46(2): 2367708, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38973391

RÉSUMÉ

BACKGROUND: Cellular senescence, macrophages infiltration, and vascular smooth muscle cells (VSMCs) osteogenic transdifferentiation participate in the pathophysiology of vascular calcification in chronic kidney disease (CKD). Senescent macrophages are involved in the regulation of inflammation in pathological diseases. In addition, senescent cells spread senescence to neighboring cells via Interferon-induced transmembrane protein3 (IFITM3). However, the role of senescent macrophages and IFITM3 in VSMCs calcification remains unexplored. AIMS: To explore the hypothesis that senescent macrophages contribute to the calcification and senescence of VSMCs via IFITM3. METHODS: Here, the macrophage senescence model was established using Lipopolysaccharides (LPS). The VSMCs were subjected to supernatants from macrophages (MCFS) or LPS-induced macrophages (LPS-MCFS) in the presence or absence of calcifying media (CM). Senescence-associated ß-galactosidase (SA-ß-gal), Alizarin red (AR), immunofluorescent staining, and western blot were used to identify cell senescence and calcification. RESULTS: The expression of IFITM3 was significantly increased in LPS-induced macrophages and the supernatants. The VSMCs transdifferentiated into osteogenic phenotype, expressing higher osteogenic differentiation markers (RUNX2) and lower VSMCs constructive makers (SM22α) when cultured with senescent macrophages supernatants. Also, senescence markers (p16 and p21) in VSMCs were significantly increased by senescent macrophages supernatants treated. However, IFITM3 knockdown inhibited this process. CONCLUSIONS: Our study showed that LPS-induced senescence of macrophages accelerated the calcification of VSMCs via IFITM3. These data provide a new perspective linking VC and aging, which may provide clues for diagnosing and treating accelerated vascular aging in patients with CKD.


Sujet(s)
Vieillissement de la cellule , Lipopolysaccharides , Macrophages , Protéines membranaires , Muscles lisses vasculaires , Protéines de liaison à l'ARN , Calcification vasculaire , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Lipopolysaccharides/pharmacologie , Calcification vasculaire/anatomopathologie , Calcification vasculaire/métabolisme , Macrophages/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines de liaison à l'ARN/métabolisme , Humains , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/anatomopathologie , Cellules cultivées , Animaux , Ostéogenèse , Transdifférenciation cellulaire
4.
Sci Rep ; 14(1): 16323, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009669

RÉSUMÉ

Vascular calcification, which is a major complication of diabetes mellitus, is an independent risk factor for cardiovascular disease. Osteogenic differentiation of vascular smooth muscle cells (VSMCs) is one of the key mechanisms underlying vascular calcification. Emerging evidence suggests that macrophage-derived extracellular vesicles (EVs) may be involved in calcification within atherosclerotic plaques in patients with diabetes mellitus. However, the role of macrophage-derived EVs in the progression of vascular calcification is largely unknown. In this study, we investigated whether macrophage-derived EVs contribute to the osteogenic differentiation of VSMCs under high glucose conditions. We isolated EVs that were secreted by murine peritoneal macrophages under normal glucose (EVs-NG) or high glucose (EVs-HG) conditions. miRNA array analysis in EVs from murine macrophages showed that miR-17-5p was significantly increased in EVs-HG compared with EVs-NG. Prediction analysis with miRbase identified transforming growth factor ß receptor type II (TGF-ß RII) as a potential target of miR-17-5p. EVs-HG as well as miR-17-5p overexpression with lipid nanoparticles inhibited the gene expression of Runx2, and TGF-ß RII. Furthermore, we demonstrated that VSMCs transfected with miR-17-5p mimic inhibited calcium deposition. Our findings reveal a novel role of macrophage-derived EVs in the negative regulation of osteogenic differentiation in VSMCs under high glucose conditions.


Sujet(s)
Différenciation cellulaire , Vésicules extracellulaires , Glucose , microARN , Muscles lisses vasculaires , Myocytes du muscle lisse , Ostéogenèse , Transduction du signal , Facteur de croissance transformant bêta , microARN/génétique , microARN/métabolisme , Animaux , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/cytologie , Glucose/pharmacologie , Glucose/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéogenèse/génétique , Facteur de croissance transformant bêta/métabolisme , Souris , Myocytes du muscle lisse/métabolisme , Vésicules extracellulaires/métabolisme , Calcification vasculaire/métabolisme , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Mâle , Souris de lignée C57BL , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique
5.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000533

RÉSUMÉ

Vascular calcification (VC) is a cardiovascular disease characterized by calcium salt deposition in vascular smooth muscle cells (VSMCs). Standard in vitro models used in VC investigations are based on VSMC monocultures under static conditions. Although these platforms are easy to use, the absence of interactions between different cell types and dynamic conditions makes these models insufficient to study key aspects of vascular pathophysiology. The present study aimed to develop a dynamic endothelial cell-VSMC co-culture that better mimics the in vivo vascular microenvironment. A double-flow bioreactor supported cellular interactions and reproduced the blood flow dynamic. VSMC calcification was stimulated with a DMEM high glucose calcification medium supplemented with 1.9 mM NaH2PO4/Na2HPO4 (1:1) for 7 days. Calcification, cell viability, inflammatory mediators, and molecular markers (SIRT-1, TGFß1) related to VSMC differentiation were evaluated. Our dynamic model was able to reproduce VSMC calcification and inflammation and evidenced differences in the modulation of effectors involved in the VSMC calcified phenotype compared with standard monocultures, highlighting the importance of the microenvironment in controlling cell behavior. Hence, our platform represents an advanced system to investigate the pathophysiologic mechanisms underlying VC, providing information not available with the standard cell monoculture.


Sujet(s)
Différenciation cellulaire , Techniques de coculture , Muscles lisses vasculaires , Myocytes du muscle lisse , Calcification vasculaire , Humains , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/cytologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Cellules cultivées , Survie cellulaire , Facteur de croissance transformant bêta-1/métabolisme , Sirtuine-1/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Bioréacteurs
6.
Radiol Cardiothorac Imaging ; 6(4): e230328, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39023373

RÉSUMÉ

Purpose To investigate the impact of plaque size and density on virtual noncontrast (VNC)-based coronary artery calcium scoring (CACS) using photon-counting detector CT and to provide safety net reconstructions for improved detection of subtle plaques in patients whose VNC-based CACS would otherwise be erroneously zero when compared with true noncontrast (TNC)-based CACS. Materials and Methods In this prospective study, CACS was evaluated in a phantom containing calcifications with different diameters (5, 3, and 1 mm) and densities (800, 400, and 200 mg/cm3) and in participants who underwent TNC and contrast-enhanced cardiac photon-counting detector CT (July 2021-March 2022). VNC images were reconstructed at different virtual monoenergetic imaging (55-80 keV) and quantum iterative reconstruction (QIR) levels (QIR,1-4). TNC scans at 70 keV with QIR off served as the reference standard. In vitro CACS was analyzed using standard settings (3.0-mm sections, kernel Qr36, 130-HU threshold). Calcification detectability and CACS of small and low-density plaques were also evaluated using 1.0-mm sections, kernel Qr44, and 120- or 110-HU thresholds. Safety net reconstructions were defined based on background Agatston scores and evaluated in vivo in TNC plaques initially nondetectable using standard VNC reconstructions. Results The in vivo cohort included 63 participants (57.8 years ± 15.5 [SD]; 37 [59%] male, 26 [41%] female). Correlation and agreement between standard CACSVNC and CACSTNC were higher in large- and medium-sized and high- and medium-density than in low-density plaques (in vitro: intraclass correlation coefficient [ICC] ≥ 0.90; r > 0.9 vs ICC = 0.20-0.48; r = 0.5-0.6). Small plaques were not detectable using standard VNC reconstructions. Calcification detectability was highest using 1.0-mm sections, kernel Qr44, 120- and 110-HU thresholds, and QIR level of 2 or less VNC reconstructions. Compared with standard VNC, using safety net reconstructions (55 keV, QIR 2, 110-HU threshold) for in vivo subtle plaque detection led to higher detection (increased by 89% [50 of 56]) and improved correlation and agreement of CACSVNC with CACSTNC (in vivo: ICC = 0.51-0.61; r = 0.6). Conclusion Compared with TNC-based calcium scoring, VNC-based calcium scoring was limited for small and low-density plaques but improved using safety net reconstructions, which may be particularly useful in patients with low calcium scores who would otherwise be treated based on potentially false-negative results. Keywords: Coronary Artery Calcium CT, Photon-Counting Detector CT, Virtual Noncontrast, Plaque Size, Plaque Density Supplemental material is available for this article. © RSNA, 2024.


Sujet(s)
Maladie des artères coronaires , Fantômes en imagerie , Plaque d'athérosclérose , Humains , Mâle , Femelle , Études prospectives , Plaque d'athérosclérose/imagerie diagnostique , Plaque d'athérosclérose/anatomopathologie , Adulte d'âge moyen , Maladie des artères coronaires/imagerie diagnostique , Maladie des artères coronaires/anatomopathologie , Sujet âgé , Photons , Vaisseaux coronaires/imagerie diagnostique , Vaisseaux coronaires/anatomopathologie , Calcification vasculaire/imagerie diagnostique , Calcification vasculaire/anatomopathologie , Tomodensitométrie/méthodes , Interprétation d'images radiographiques assistée par ordinateur/méthodes , Coronarographie/méthodes , Produits de contraste
9.
Nat Commun ; 15(1): 4985, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862515

RÉSUMÉ

Hyperglycemia accelerates calcification of atherosclerotic plaques in diabetic patients, and the accumulation of advanced glycation end products (AGEs) is closely related to the atherosclerotic calcification. Here, we show that hyperglycemia-mediated AGEs markedly increase vascular smooth muscle cells (VSMCs) NF90/110 activation in male diabetic patients with atherosclerotic calcified samples. VSMC-specific NF90/110 knockout in male mice decreases obviously AGEs-induced atherosclerotic calcification, along with the inhibitions of VSMC phenotypic changes to osteoblast-like cells, apoptosis, and matrix vesicle release. Mechanistically, AGEs increase the activity of NF90, which then enhances ubiquitination and degradation of AGE receptor 1 (AGER1) by stabilizing the mRNA of E3 ubiquitin ligase FBXW7, thus causing the accumulation of more AGEs and atherosclerotic calcification. Collectively, our study demonstrates the effects of VSMC NF90 in mediating the metabolic imbalance of AGEs to accelerate diabetic atherosclerotic calcification. Therefore, inhibition of VSMC NF90 may be a potential therapeutic target for diabetic atherosclerotic calcification.


Sujet(s)
Athérosclérose , Protéine-7 contenant une boite F et des répétitions WD , Produits terminaux de glycation avancée , Souris knockout , Muscles lisses vasculaires , Myocytes du muscle lisse , Facteurs nucléaires-90 , Récepteur spécifique des produits finaux de glycosylation avancée , Animaux , Mâle , Souris , Produits terminaux de glycation avancée/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Humains , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/génétique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Facteurs nucléaires-90/métabolisme , Facteurs nucléaires-90/génétique , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/génétique , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Calcification vasculaire/génétique , Souris de lignée C57BL , Ubiquitination , Diabète expérimental/métabolisme , Diabète expérimental/complications , Diabète expérimental/génétique , Diabète expérimental/anatomopathologie , Hyperglycémie/métabolisme , Hyperglycémie/génétique , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/génétique , Apoptose
10.
Biomacromolecules ; 25(7): 4329-4343, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38833553

RÉSUMÉ

The development of nanotherapy targeting mitochondria to alleviate oxidative stress is a critical therapeutic strategy for vascular calcification (VC) in diabetes. In this study, we engineered mitochondria-targeted nanodrugs (T4O@TPP/PEG-PLGA) utilizing terpinen-4-ol (T4O) as a natural antioxidant and mitochondrial protector, PEG-PLGA as the nanocarrier, and triphenylphosphine (TPP) as the mitochondrial targeting ligand. In vitro assessments demonstrated enhanced cellular uptake of T4O@TPP/PEG-PLGA, with effective mitochondrial targeting. This nanodrug successfully reduced oxidative stress induced by high glucose levels in vascular smooth muscle cells. In vivo studies showed prolonged retention of the nanomaterials in the thoracic aorta for up to 24 h. Importantly, experiments in diabetic VC models underscored the potent antioxidant properties of T4O@TPP/PEG-PLGA, as evidenced by its ability to mitigate VC and restore mitochondrial morphology. These results suggest that these nanodrugs could be a promising strategy for managing diabetic VC.


Sujet(s)
Antioxydants , Mitochondries , Stress oxydatif , Calcification vasculaire , Animaux , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Antioxydants/pharmacologie , Antioxydants/composition chimique , Calcification vasculaire/traitement médicamenteux , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Composés organiques du phosphore/composition chimique , Composés organiques du phosphore/pharmacologie , Diabète expérimental/traitement médicamenteux , Nanoparticules/composition chimique , Souris , Mâle , Polyéthylène glycols/composition chimique , Rats , Humains , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/métabolisme
11.
Int J Biochem Cell Biol ; 173: 106613, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909746

RÉSUMÉ

Vascular calcification in diabetes patients is a major independent risk factor for developing diabetic cardiovascular complications. However, the mechanisms by which diabetes leads to vascular calcification are complex and not yet fully understood. Our previous study revealed that miR-32-5p is a potential new diagnostic marker for coronary artery calcification. In this study, we found that miR-32-5p levels were significantly greater in the plasma of type 2 diabetes patients with coronary artery calcification and were positively correlated with the coronary artery calcification score. In type 2 diabetic mice, miR-32-5p levels were also elevated in the aorta, and knockout of miR-32-5p inhibited the osteogenic differentiation of vascular smooth muscle cells in vivo. Furthermore, overexpression of miR-32-5p promoted vascular smooth muscle cell calcification, while antagonism of miR-32-5p inhibited vascular smooth muscle cell calcification under high-glucose conditions. GATA binding protein 6 (GATA6) was identified as the key target gene through which miR-32-5p promotes vascular smooth muscle cell calcification. Overexpression of GATA6 antagonized the effects of miR-32-5p on vascular calcification. Additionally, high glucose levels were shown to induce the upregulation of miR-32-5p by activating CCAAT/enhancer binding protein beta (CEBPB). These results suggest that miR-32-5p is an important procalcification factor in vascular calcification associated with type 2 diabetes and identify the CEBPB/miR-32-5p/GATA6 axis as a potential biomarker and therapeutic target for preventing and treating vascular calcification in type 2 diabetes.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT , Diabète de type 2 , Facteur de transcription GATA-6 , microARN , Calcification vasculaire , Diabète de type 2/complications , Diabète de type 2/métabolisme , Diabète de type 2/anatomopathologie , Diabète de type 2/génétique , microARN/génétique , microARN/métabolisme , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Calcification vasculaire/génétique , Animaux , Humains , Souris , Mâle , Facteur de transcription GATA-6/métabolisme , Facteur de transcription GATA-6/génétique , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Souris de lignée C57BL , Adulte d'âge moyen , Femelle , Souris knockout
12.
Exp Cell Res ; 440(2): 114147, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38944174

RÉSUMÉ

Coronary artery calcification (CAC) is a hallmark event in the pathogenesis of cardiovascular disease, involving the phenotypic transformation of vascular smooth muscle cells (VSMC) towards an osteogenic state. Despite this understanding, the molecular mechanisms governing the VSMC osteogenic switch remain incompletely elucidated. Here, we sought to examine the potential role of circular RNA (circRNA) in the context of CAC. Through transcriptome analysis of circRNA-seq, we identified circTOP1 as a potential candidate circRNA in individuals with CAC. Furthermore, we observed that overexpression of circTOP1 exacerbated vascular calcification in a CAC model. Subsequent pull-down assays revealed an interaction between circTOP1 and PTBP1, a putative target gene of circTOP1 in the context of CAC. In both in vivo and in vitro experiments, we observed heightened expression of circTOP1 and PTBP1 in the CAC model, and noted that reducing circTOP1 expression effectively reduced calcium salt deposits and mineralized nodules in model mice. Additionally, in vitro experiments demonstrated that overexpression of PTBP1 reversed the weakening of signaling caused by silencing circTOP1, thereby exacerbating the osteogenic transition and calcification of VSMC. Collectively, our findings suggested that circTOP1 promotes CAC by modulating PTBP1 expression to mediate VSMC transdifferentiation.


Sujet(s)
Ribonucléoprotéines nucléaires hétérogènes , Muscles lisses vasculaires , Myocytes du muscle lisse , Protéine PTB , ARN circulaire , Calcification vasculaire , Protéine PTB/génétique , Protéine PTB/métabolisme , Animaux , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Souris , ARN circulaire/génétique , ARN circulaire/métabolisme , Humains , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie , Calcification vasculaire/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Mâle , Maladie des artères coronaires/génétique , Maladie des artères coronaires/anatomopathologie , Maladie des artères coronaires/métabolisme , Souris de lignée C57BL , Vaisseaux coronaires/anatomopathologie , Vaisseaux coronaires/métabolisme , Ostéogenèse/génétique , Évolution de la maladie , Régulation de l'expression des gènes/génétique
13.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891922

RÉSUMÉ

Vascular calcification has a global health impact that is closely linked to bone loss. The Receptor Activator of Nuclear Factor Kappa B (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, fundamental for bone metabolism, also plays an important role in vascular calcification. The Leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), a novel receptor for RANKL, regulates bone remodeling, and it appears to be involved in vascular calcification. Besides RANKL, LGR4 interacts with R-spondins (RSPOs), which are known for their roles in bone but are less understood in vascular calcification. Studies were conducted in rats with chronic renal failure fed normal or high phosphorus diets for 18 weeks, with and without control of circulating parathormone (PTH) levels, resulting in different degrees of aortic calcification. Additionally, vascular smooth muscle cells (VSMCs) were cultured under non-calcifying (1 mM phosphate) and calcifying (3 mM phosphate) media with different concentrations of PTH. To explore the role of RANKL in VSMC calcification, increasing concentrations of soluble RANKL were added to non-calcifying and calcifying media. The effects mediated by RANKL binding to its receptor LGR4 were investigated by silencing the LGR4 receptor in VSMCs. Furthermore, the gene expression of the RANK/RANKL/OPG system and the ligands of LGR4 was assessed in human epigastric arteries obtained from kidney transplant recipients with calcification scores (Kauppila Index). Increased aortic calcium in rats coincided with elevated systolic blood pressure, upregulated Lgr4 and Rankl gene expression, downregulated Opg gene expression, and higher serum RANKL/OPG ratio without changes in Rspos gene expression. Elevated phosphate in vitro increased calcium content and expression of Rankl and Lgr4 while reducing Opg. Elevated PTH in the presence of high phosphate exacerbated the increase in calcium content. No changes in Rspos were observed under the conditions employed. The addition of soluble RANKL to VSMCs induced genotypic differentiation and calcification, partly prevented by LGR4 silencing. In the epigastric arteries of individuals presenting vascular calcification, the gene expression of RANKL was higher. While RSPOs show minimal impact on VSMC calcification, RANKL, interacting with LGR4, drives osteogenic differentiation in VSMCs, unveiling a novel mechanism beyond RANKL-RANK binding.


Sujet(s)
Muscles lisses vasculaires , Ligand de RANK , Récepteurs couplés aux protéines G , Calcification vasculaire , Ligand de RANK/métabolisme , Ligand de RANK/génétique , Animaux , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Rats , Humains , Mâle , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Ostéoprotégérine/métabolisme , Ostéoprotégérine/génétique , Hormone parathyroïdienne/métabolisme , Cellules cultivées , Rat Sprague-Dawley
14.
Front Immunol ; 15: 1395596, 2024.
Article de Anglais | MEDLINE | ID: mdl-38919629

RÉSUMÉ

Vascular calcification (VC) is considered a common pathological process in various vascular diseases. Accumulating studies have confirmed that VC is involved in the inflammatory response in heart disease, and SPP1+ macrophages play an important role in this process. In VC, studies have focused on the physiological and pathological functions of macrophages, such as pro-inflammatory or anti-inflammatory cytokines and pro-fibrotic vesicles. Additionally, macrophages and activated lymphocytes highly express SPP1 in atherosclerotic plaques, which promote the formation of fatty streaks and plaque development, and SPP1 is also involved in the calcification process of atherosclerotic plaques that results in heart failure, but the crosstalk between SPP1-mediated immune cells and VC has not been adequately addressed. In this review, we summarize the regulatory effect of SPP1 on VC in T cells, macrophages, and dendritic cells in different organs' VC, which could be a potential therapeutic target for VC.


Sujet(s)
Macrophages , Ostéopontine , Calcification vasculaire , Animaux , Humains , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Ostéopontine/métabolisme , Plaque d'athérosclérose/immunologie , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Calcification vasculaire/immunologie , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie
15.
Vascul Pharmacol ; 155: 107376, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38692418

RÉSUMÉ

Cardiovascular disease and osteoporosis, major causes of morbidity and mortality, are associated with hyperlipidemia. Recent studies show that empagliflozin (EMPA), an inhibitor of sodium-glucose cotransporter-2 (SGLT2), improves cardiovascular health. In preclinical animal studies, EMPA mitigates vascular calcification in the males but its effects in the females are not known. Thus, we used female mice to test the effects of EMPA on calcification in the artery wall, cardiac function, and skeletal bone. By serial in vivo microCT imaging, we followed the progression of aortic calcification and bone mineral density in young and older female Apoe-/- mice fed a high-fat diet with or without EMPA. The two different age groups were used to compare early vs. advanced stages of aortic calcification. Results show that EMPA treatment increased urine glucose levels. Aortic calcium content increased in both the controls and the EMPA-treated mice, and EMPA did not affect progression of aortic calcium content in both young and older mice. However, 3-D segmentation analysis of aortic calcium deposits on microCT images revealed that EMPA-treated mice had significantly less surface area and volume of calcified deposits as well as fewer numbers of deposits than the control mice. To test for direct effects on vascular cell calcification, we treated murine aortic smooth muscle cells with EMPA, and results showed a slight inhibition of alkaline phosphatase activity and inflammatory matrix calcification. As for skeletal bone, EMPA-treated mice had significantly lower BMD than the controls in both the lumbar vertebrae and femoral bones in both young and older mice. The findings suggest that, in hyperlipidemic female mice, unlike males, SGLT2 inhibition with empagliflozin does not mitigate progression of aortic calcification and may even lower skeletal bone density.


Sujet(s)
Composés benzhydryliques , Densité osseuse , Modèles animaux de maladie humaine , Glucosides , Hyperlipidémies , Souris invalidées pour les gènes ApoE , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Calcification vasculaire , Microtomographie aux rayons X , Animaux , Glucosides/pharmacologie , Composés benzhydryliques/pharmacologie , Femelle , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Calcification vasculaire/anatomopathologie , Calcification vasculaire/traitement médicamenteux , Calcification vasculaire/prévention et contrôle , Calcification vasculaire/métabolisme , Hyperlipidémies/traitement médicamenteux , Densité osseuse/effets des médicaments et des substances chimiques , Aorte/effets des médicaments et des substances chimiques , Aorte/anatomopathologie , Aorte/métabolisme , Aorte/imagerie diagnostique , Aorte/physiopathologie , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/métabolisme , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/traitement médicamenteux , Maladies de l'aorte/physiopathologie , Maladies de l'aorte/imagerie diagnostique , Souris de lignée C57BL , Alimentation riche en graisse , Souris , Facteurs âges , Cellules cultivées
16.
Cell Signal ; 120: 111211, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38705504

RÉSUMÉ

Vascular calcification (VC) is a characteristic feature in patients with diabetes mellitus (DM) and is closely associated with the osteogenic differentiation of vascular smooth muscle cells (VSMCs). Ubiquitin-Specific Protease 10 (USP10) has been shown to regulate multiple cellular processes; however, its relationship with diabetic VC remains unclear. This study aims to elucidate the role of USP10 in VC development and the underlying regulatory mechanisms. Nε-carboxymethyl lysine (CML) was significantly increased in calcified ateries from diabetic atherosclerosis ApoE-/- mice fed with high-fat diets. CML downregulated USP10 expression in VSMCs and calcified mice coronary arteries, as assessd by Western blotting, RT-qPCR,immunofluorescence and immunohistochemistry. Loss-and gain-of-function experiments were conducted both in vitro and in vivo to verify the biological functions of USP10. Ectopic expression of USP10 mitigated the severity of VC. With regard to the mechanism, the interaction between USP10 and AMPKα was investigated through double-label immunofluorescence and Co-immunoprecipitation. In vitro ubiquitination assay revealed that USP10 was capable of mediating AMPKα ubiquitination and caused increased AMPKα phosphorylation level at Thr172. Moreover, the anticalcification effect of USP10 was reversed by pharmacological inhibition of AMPK signaling pathway. The current fundings suggest an important role of USP10 in diabetic VC progression, at least in part, via mediating the ubiquitination and activation of AMPKα. USP10 may serve as a novel therapeutic target for the treatment of diabetic VC.


Sujet(s)
AMP-Activated Protein Kinases , Athérosclérose , Lysine , Ubiquitin thiolesterase , Calcification vasculaire , Animaux , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Souris , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Lysine/métabolisme , Lysine/analogues et dérivés , AMP-Activated Protein Kinases/métabolisme , Mâle , Ubiquitination , Souris de lignée C57BL , Humains , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Diabète expérimental/métabolisme , Diabète expérimental/complications , Diabète expérimental/anatomopathologie
17.
Cardiovasc Diabetol ; 23(1): 186, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38812011

RÉSUMÉ

BACKGROUND: Vascular calcification (VC) is an independent risk factor for cardiovascular diseases. Recently, ferroptosis has been recognised as a novel therapeutic target for cardiovascular diseases. Although an association between ferroptosis and vascular calcification has been reported, the role and mechanism of iron overload in vascular calcification are still poorly understood. Specifically, further in-depth research is required on whether metalloproteins SLC39a14 and SLC39a8 are involved in ferroptosis induced by iron overload. METHODS: R language was employed for the differential analysis of the dataset, revealing the correlation between ferroptosis and calcification. The experimental approaches encompassed both in vitro and in vivo studies, incorporating the use of iron chelators and models of iron overload. Additionally, gain- and loss-of-function experiments were conducted to investigate iron's effects on vascular calcification comprehensively. Electron microscopy, immunofluorescence, western blotting, and real-time polymerase chain reaction were used to elucidate how Slc39a14 and Slc39a8 mediate iron overload and promote calcification. RESULTS: Ferroptosis was observed in conjunction with vascular calcification (VC); the association was consistently confirmed by in vitro and in vivo studies. Our results showed a positive correlation between iron overload in VSMCs and calcification. Iron chelators are effective in reversing VC and iron overload exacerbates this process. The expression levels of the metal transport proteins Slc39a14 and Slc39a8 were significantly upregulated during calcification; the inhibition of their expression alleviated VC. Conversely, Slc39a14 overexpression exacerbates calcification and promotes intracellular iron accumulation in VSMCs. CONCLUSIONS: Our research demonstrates that iron overload occurs during VC, and that inhibition of Slc39a14 and Slc39a8 significantly relieves VC by intercepting iron overload-induced ferroptosis in VSMCs, providing new insights into the VC treatment.


Sujet(s)
Transporteurs de cations , Modèles animaux de maladie humaine , Ferroptose , Agents chélateurs du fer , Souris de lignée C57BL , Muscles lisses vasculaires , Myocytes du muscle lisse , Calcification vasculaire , Ferroptose/effets des médicaments et des substances chimiques , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Animaux , Transporteurs de cations/métabolisme , Transporteurs de cations/génétique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Agents chélateurs du fer/pharmacologie , Agents chélateurs du fer/usage thérapeutique , Transduction du signal , Mâle , Humains , Fer/métabolisme , Surcharge en fer/métabolisme , Surcharge en fer/anatomopathologie
18.
Redox Biol ; 73: 103183, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38759418

RÉSUMÉ

AIMS: Vascular calcification is strongly linked to the development of major adverse cardiovascular events, but effective treatments are lacking. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an emerging category of oral hypoglycemic drugs that have displayed marked effects on metabolic and cardiovascular diseases, including recently reported vascular medial calcification. However, the roles and underlying mechanisms of SGLT2 inhibitors in vascular calcification have not been fully elucidated. Thus, we aimed to further determine whether SGLT2 inhibitors protect against vascular calcification and to investigate the mechanisms involved. METHODS AND RESULTS: A computed tomography angiography investigation of coronary arteries from 1554 patients with type 2 diabetes revealed that SGLT2 inhibitor use was correlated with a lower Agatston calcification score. In the vitamin D3 overdose, 5/6 nephrectomy chronic kidney disease-induced medial calcification and Western diet-induced atherosclerotic intimal calcification models, dapagliflozin (DAPA) substantially alleviated vascular calcification in the aorta. Furthermore, we showed that DAPA reduced vascular calcification via Runx2-dependent osteogenic transdifferentiation in vascular smooth muscle cells (VSMCs). Transcriptome profiling revealed that thioredoxin domain containing 5 (TXNDC5) was involved in the attenuation of vascular calcification by DAPA. Rescue experiments showed that DAPA-induced TXNDC5 downregulation in VSMCs blocked the protective effect on vascular calcification. Furthermore, TXNDC5 downregulation disrupted protein folding-dependent Runx2 stability and promoted subsequent proteasomal degradation. Moreover, DAPA downregulated TXNDC5 expression via amelioration of oxidative stress and ATF6-dependent endoplasmic reticulum stress. Consistently, the class effects of SGLT2 inhibitors on vascular calcification were validated with empagliflozin in intimal and medial calcification models. CONCLUSIONS: SGLT2 inhibitors ameliorate vascular calcification through blocking endoplasmic reticulum stress-dependent TXNDC5 upregulation and promoting subsequent Runx2 proteasomal degradation, suggesting that SGLT2 inhibitors are potentially beneficial for vascular calcification treatment and prevention.


Sujet(s)
Glucosides , Ostéogenèse , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Calcification vasculaire , Calcification vasculaire/métabolisme , Calcification vasculaire/traitement médicamenteux , Calcification vasculaire/anatomopathologie , Calcification vasculaire/étiologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Animaux , Humains , Ostéogenèse/effets des médicaments et des substances chimiques , Souris , Glucosides/pharmacologie , Mâle , Thiorédoxines/métabolisme , Thiorédoxines/génétique , Composés benzhydryliques/pharmacologie , Diabète de type 2/métabolisme , Diabète de type 2/complications , Diabète de type 2/traitement médicamenteux , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Rats , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Modèles animaux de maladie humaine , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/cytologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Femelle
19.
Gut Microbes ; 16(1): 2351532, 2024.
Article de Anglais | MEDLINE | ID: mdl-38727248

RÉSUMÉ

Emerging evidence indicates that alteration of gut microbiota plays an important role in chronic kidney disease (CKD)-related vascular calcification (VC). We aimed to investigate the specific gut microbiota and the underlying mechanism involved in CKD-VC. We identified an increased abundance of Prevotella copri (P. copri) in the feces of CKD rats (induced by using 5/6 nephrectomy followed by a high calcium and phosphate diet) with aortic calcification via amplicon sequencing of 16S rRNA genes. In patients with CKD, we further confirmed a positive correlation between abundance of P. copri and aortic calcification scores. Moreover, oral administration of live P. copri aggravated CKD-related VC and osteogenic differentiation of vascular smooth muscle cells in vivo, accompanied by intestinal destruction, enhanced expression of Toll-like receptor-4 (TLR4), and elevated lipopolysaccharide (LPS) levels. In vitro and ex vivo experiments consistently demonstrated that P. copri-derived LPS (Pc-LPS) accelerated high phosphate-induced VC and VSMC osteogenic differentiation. Mechanistically, Pc-LPS bound to TLR4, then activated the nuclear factor κB (NF-κB) and nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome signals during VC. Inhibition of NF-κB reduced NLRP3 inflammasome and attenuated Pc-LPS-induced VSMC calcification. Our study clarifies a novel role of P. copri in CKD-related VC, by the mechanisms involving increased inflammation-regulating metabolites including Pc-LPS, and activation of the NF-κB/NLRP3 signaling pathway. These findings highlight P. copri and its-derived LPS as potential therapeutic targets for VC in CKD.


Sujet(s)
Microbiome gastro-intestinal , Lipopolysaccharides , Facteur de transcription NF-kappa B , Prevotella , Transduction du signal , Calcification vasculaire , Animaux , Humains , Mâle , Rats , Fèces/microbiologie , Inflammasomes/métabolisme , Lipopolysaccharides/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Ostéogenèse/effets des médicaments et des substances chimiques , Prevotella/métabolisme , Rat Sprague-Dawley , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/microbiologie , Insuffisance rénale chronique/anatomopathologie , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Calcification vasculaire/métabolisme , Calcification vasculaire/microbiologie , Calcification vasculaire/anatomopathologie
20.
Cells ; 13(9)2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38727287

RÉSUMÉ

Currently, more and more people are suffering from chronic kidney disease (CKD). It is estimated that CKD affects over 10% of the population worldwide. This is a significant issue, as the kidneys largely contribute to maintaining homeostasis by, among other things, regulating blood pressure, the pH of blood, and the water-electrolyte balance and by eliminating unnecessary metabolic waste products from blood. What is more, this disease does not show any specific symptoms at the beginning. The development of CKD is predisposed by certain conditions, such as diabetes mellitus or hypertension. However, these disorders are not the only factors promoting the onset and progression of CKD. The primary purpose of this review is to examine renin-angiotensin-aldosterone system (RAAS) activity, transforming growth factor-ß1 (TGF-ß1), vascular calcification (VC), uremic toxins, and hypertension in the context of their impact on the occurrence and the course of CKD. We firmly believe that a deeper comprehension of the cellular and molecular mechanisms underlying CKD can lead to an enhanced understanding of the disease. In the future, this may result in the development of medications targeting specific mechanisms involved in the decline of kidney function. Our paper unveils the selected processes responsible for the deterioration of renal filtration abilities.


Sujet(s)
Évolution de la maladie , Insuffisance rénale chronique , Système rénine-angiotensine , Humains , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Système rénine-angiotensine/physiologie , Animaux , Hypertension artérielle/physiopathologie , Hypertension artérielle/anatomopathologie , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Calcification vasculaire/physiopathologie , Facteur de croissance transformant bêta-1/métabolisme , Rein/anatomopathologie , Rein/métabolisme , Rein/physiopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE