Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 552
Filtrer
1.
Genet Test Mol Biomarkers ; 28(6): 223-232, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38708584

RÉSUMÉ

Background: Matrix metalloproteinase (MMP) enzyme gene polymorphisms MMP-2-1575G/A and MMP-9-1562C/T promoter polymorphism, their serum levels, and activity are associated with aortic valve calcification (AVC). Materials and Methods: The synergistic link between the risk of AVC and the alleles T and A of MMP-9 and MMP-2 was investigated, respectively. Ninety-two cases with AVC and 92 healthy individuals from the west of Iran were included, and MMP- 2-1575G/A and MMP-9-1562C/T promoter polymorphisms were detected using PCR-RFLP. The serum levels and activity of MMP-2 and -9 were assessed using ELISA and gelatin zymography methods, respectively. In addition, serum biochemical markers, including FBS, urea and creatinine, cholesterol, triglyceride, HDL, LDL, calcium, phosphorus, and blood pressure: systolic blood pressure and diastolic blood pressure were measured. Results: Heart valve calcification disease was associated with a comparatively higher frequency of the A allele of the MMP2-1575 variation (p = 0.002). In addition, the frequency of T allele of the MMP9-1562 variant was higher than the control group (p = 0.007). Conclusion: MMP-2 and MMP-9 serum levels and activities were observed to be considerably higher in the experimental group than in the control group (p < 0.001). Patients are more susceptible to cardiovascular disease than the control group due to elevated serum levels and activity of MMP-2 and MMP-9.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , Prédisposition génétique à une maladie , Matrix metalloproteinase 2 , Matrix metalloproteinase 9 , Régions promotrices (génétique) , Humains , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 9/sang , Calcinose/génétique , Calcinose/sang , Femelle , Mâle , Iran , Matrix metalloproteinase 2/génétique , Matrix metalloproteinase 2/sang , Valve aortique/anatomopathologie , Régions promotrices (génétique)/génétique , Adulte d'âge moyen , Sténose aortique/génétique , Sténose aortique/sang , Polymorphisme de nucléotide simple/génétique , Sujet âgé , Adulte , Allèles , Études cas-témoins , Fréquence d'allèle/génétique , Valvulopathies/génétique , Valvulopathies/sang , Génotype
2.
Redox Biol ; 73: 103215, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38810422

RÉSUMÉ

The prevalence of calcific aortic valve disease (CAVD) remains substantial while there is currently no medical therapy available. Forkhead box O1 (FOXO1) is known to be involved in the pathogenesis of cardiovascular diseases, including vascular calcification and atherosclerosis; however, its specific role in calcific aortic valve disease remains to be elucidated. In this study, we identified FOXO1 significantly down-regulated in the aortic valve interstitial cells (VICs) of calcified aortic valves by investigating clinical specimens and GEO database analysis. FOXO1 silencing or inhibition promoted VICs osteogenic differentiation in vitro and aortic valve calcification in Apoe-/- mice, respectively. We identified that FOXO1 facilitated the ubiquitination and degradation of RUNX2, which process was mainly mediated by SMAD-specific E3 ubiquitin ligase 2 (SMURF2). Our discoveries unveil a heretofore unacknowledged mechanism involving the FOXO1/SMURF2/RUNX2 axis in CAVD, thereby proposing the potential therapeutic utility of FOXO1 or SMURF2 as viable strategies to impede the progression of CAVD.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , Sous-unité alpha 1 du facteur CBF , Protéine O1 à motif en tête de fourche , Ubiquitin-protein ligases , Ubiquitination , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Animaux , Valve aortique/métabolisme , Valve aortique/anatomopathologie , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Souris , Humains , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Calcinose/métabolisme , Calcinose/anatomopathologie , Calcinose/génétique , Sténose aortique/métabolisme , Sténose aortique/anatomopathologie , Sténose aortique/génétique , Mâle , Ostéogenèse/génétique , Modèles animaux de maladie humaine , Différenciation cellulaire
3.
IET Syst Biol ; 18(3): 77-91, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38566328

RÉSUMÉ

Calcific aortic valve disease (CAVD) and osteoarthritis (OA) are common diseases in the ageing population and share similar pathogenesis, especially in inflammation. This study aims to discover potential diagnostic and therapeutic targets in patients with CAVD and OA. Three CAVD datasets and one OA dataset were obtained from the Gene Expression Omnibus database. We used bioinformatics methods to search for key genes and immune infiltration, and established a ceRNA network. Immunohistochemical staining was performed to verify the expression of candidate genes in human and mice aortic valve tissues. Two key genes obtained, leucine rich repeat containing 15 (LRRC15) and secreted phosphoprotein 1 (SPP1), were further screened using machine learning and verified in human and mice aortic valve tissues. Compared to normal tissues, the infiltration of immune cells in CAVD tissues was significantly higher, and the expressions of LRRC15 and SPP1 were positively correlated with immune cells infiltration. Moreover, the ceRNA network showed extensive regulatory interactions based on LRRC15 and SPP1. The authors' findings identified LRRC15 and SPP1 as hub genes in immunological mechanisms during CAVD and OA initiation and progression, as well as potential targets for drug development.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , Biologie informatique , Arthrose , Ostéopontine , Animaux , Humains , Souris , Valve aortique/anatomopathologie , Valve aortique/métabolisme , Maladie de la valve aortique/génétique , Maladie de la valve aortique/métabolisme , Sténose aortique/génétique , Sténose aortique/métabolisme , Sténose aortique/anatomopathologie , Calcinose/génétique , Calcinose/métabolisme , Calcinose/anatomopathologie , Réseaux de régulation génique , Arthrose/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Ostéopontine/génétique , Ostéopontine/métabolisme
4.
Cardiovasc Toxicol ; 24(5): 499-512, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38589550

RÉSUMÉ

Calcific aortic valve stenosis (CAVS) is characterized by increasing inflammation and progressive calcification in the aortic valve leaflets and is a major cause of death in the aging population. This study aimed to identify the inflammatory proteins involved in CAVS and provide potential therapeutic targets. We investigated the observational and causal associations of 92 inflammatory proteins, which were measured using affinity-based proteomic assays. Firstly, the case-control cohort identified differential proteins associated with the occurrence and progression of CAVS. Subsequently, we delved into exploring the causal impacts of these associated proteins through Mendelian randomization. This involved utilizing genetic instruments derived from cis-protein quantitative loci identified in genome-wide association studies, encompassing a cohort of over 400,000 individuals. Finally, we investigated the gene transcription and protein expression levels of inflammatory proteins by single-cell and immunohistochemistry analysis. Multivariate logistic regression and spearman's correlation analysis showed that five proteins showed a significant positive correlation with disease severity. Mendelian randomization showed that elevated levels of two proteins, namely, matrix metallopeptidase-1 (MMP1) and sirtuin 2 (SIRT2), were associated with an increased risk of CAVS. Immunohistochemistry and single-cell transcriptomes showed that expression levels of MMP1 and SIRT2 at the tissue and cell levels were significantly higher in calcified valves than in non-calcified control valves. These findings indicate that MMP1 and SIRT2 are causally related to CAVS and open up the possibility for identifying novel therapeutic targets.


Sujet(s)
Sténose aortique , Valve aortique , Valve aortique/anatomopathologie , Marqueurs biologiques , Calcinose , Médiateurs de l'inflammation , Matrix metalloproteinase 1 , Analyse de randomisation mendélienne , Protéomique , Humains , Sténose aortique/métabolisme , Sténose aortique/sang , Sténose aortique/anatomopathologie , Sténose aortique/génétique , Calcinose/génétique , Calcinose/métabolisme , Calcinose/sang , Calcinose/anatomopathologie , Valve aortique/métabolisme , Mâle , Femelle , Sujet âgé , Études cas-témoins , Marqueurs biologiques/sang , Médiateurs de l'inflammation/métabolisme , Médiateurs de l'inflammation/sang , Matrix metalloproteinase 1/génétique , Matrix metalloproteinase 1/métabolisme , Adulte d'âge moyen , Facteurs de risque , Indice de gravité de la maladie , Sujet âgé de 80 ans ou plus , Prédisposition génétique à une maladie , Protéines du sang/génétique , Protéines du sang/analyse , Phénotype
5.
J Am Heart Assoc ; 13(9): e034102, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38639330

RÉSUMÉ

BACKGROUND: Large observational studies have demonstrated a clear inverse association between renal function and risk of aortic stenosis (AS). Whether this represents a causal, reverse causal or correlative relationship remains unclear. We investigated this using a bidirectional 2-sample Mendelian randomization approach. METHODS AND RESULTS: We collected summary statistics for the primary analysis of chronic kidney disease (CKD) and AS from genome-wide association study meta-analyses including 480 698 and 653 867 participants, respectively. We collected further genome-wide association study summary statistics from up to 1 004 040 participants for sensitivity analyses involving estimated glomerular filtration rate (eGFR) derived from creatinine, eGFR derived from cystatin C, and serum urea nitrogen. Inverse-variance weighted was the primary analysis method, with weighted-median, weighted-mode, Mendelian randomization-Egger, and Mendelian randomization-Pleiotropy Residual Sum and Outlier as sensitivity analyses. We did not find evidence of a causal relationship between genetically predicted CKD liability as the exposure and AS as the outcome (odds ratio [OR], 0.94 per unit increase in log odds of genetic liability to CKD [95% CI, 0.85-1.04], P=0.26) nor robust evidence of AS liability as the exposure and CKD as the outcome (OR, 1.04 per unit increase in log odds of genetic liability to AS [95% CI, 0.97-1.12], P=0.30). The sensitivity analyses were neutral overall, as were the analyses using eGFR derived from creatinine, eGFR derived from cystatin C, and serum urea nitrogen. All positive controls demonstrated strong significant associations. CONCLUSIONS: The present study did not find evidence of a substantial effect of genetically predicted renal impairment on risk of AS. This has important implications for research efforts that attempt to identify prevention and treatment targets for both CKD and AS.


Sujet(s)
Sténose aortique , Étude d'association pangénomique , Débit de filtration glomérulaire , Analyse de randomisation mendélienne , Insuffisance rénale chronique , Humains , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/physiopathologie , Insuffisance rénale chronique/épidémiologie , Débit de filtration glomérulaire/génétique , Sténose aortique/génétique , Sténose aortique/physiopathologie , Cystatine C/sang , Cystatine C/génétique , Facteurs de risque , Rein/physiopathologie , Prédisposition génétique à une maladie , Créatinine/sang , Appréciation des risques , Polymorphisme de nucléotide simple , Marqueurs biologiques/sang , Azote uréique sanguin
7.
Nat Commun ; 15(1): 2407, 2024 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-38494474

RÉSUMÉ

There is currently no medical therapy to prevent calcific aortic valve stenosis (CAVS). Multi-omics approaches could lead to the identification of novel molecular targets. Here, we perform a genome-wide association study (GWAS) meta-analysis including 14,819 cases among 941,863 participants of European ancestry. We report 32 genomic loci, among which 20 are novel. RNA sequencing of 500 human aortic valves highlights an enrichment in expression regulation at these loci and prioritizes candidate causal genes. Homozygous genotype for a risk variant near TWIST1, a gene involved in endothelial-mesenchymal transition, has a profound impact on aortic valve transcriptomics. We identify five genes outside of GWAS loci by combining a transcriptome-wide association study, colocalization, and Mendelian randomization analyses. Using cross-phenotype and phenome-wide approaches, we highlight the role of circulating lipoproteins, blood pressure and inflammation in the disease process. Our findings pave the way for the development of novel therapies for CAVS.


Sujet(s)
Sténose aortique , Valve aortique , Valve aortique/anatomopathologie , Calcinose , Humains , Valve aortique/métabolisme , Étude d'association pangénomique , Sténose aortique/génétique , Génomique
8.
Adv Sci (Weinh) ; 11(20): e2307319, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38502885

RÉSUMÉ

The senescence of aortic valve interstitial cells (VICs) plays a critical role in the progression of calcific aortic valve disease (CAVD). However, the precise mechanisms underlying the senescence of VICs remain unclear, demanding the identification of a novel target to mitigate this process. Previous studies have highlighted the anti-aging potential of morusin. Thus, this study aimed to explore the therapeutic potential of morusin in CAVD. Cellular experiments reveal that morusin effectively suppresses cellular senescence and cause a shift toward osteogenic differentiation of VICs in vitro. Mechanistically, morusin activate the Nrf2-mediated antiaging signaling pathway by downregulating CCND1 expression and aiding Keap1 degradation through Trim 25. This activation lead to the upregulated expression of antioxidant genes, thus reducing reactive oxygen species production and thereby preventing VIC osteogenic differentiation. In vivo experiments in ApoE-/- mice on a high-fat Western diet demonstrate the positive effect of morusin in mitigating aortic valve calcification. These findings emphasize the antiaging properties of morusin and its potential as a therapeutic agent for CAVD.


Sujet(s)
Sténose aortique , Calcinose , Vieillissement de la cellule , Flavonoïdes , Transduction du signal , Animaux , Humains , Mâle , Souris , Valve aortique/métabolisme , Valve aortique/anatomopathologie , Sténose aortique/métabolisme , Sténose aortique/génétique , Sténose aortique/anatomopathologie , Calcinose/métabolisme , Calcinose/génétique , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Cycline D1/métabolisme , Cycline D1/génétique , Modèles animaux de maladie humaine , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Ostéogenèse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Flavonoïdes/administration et posologie
9.
FASEB J ; 38(6): e23505, 2024 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-38507255

RÉSUMÉ

Aortic stenosis (AS) and hypertrophic cardiomyopathy (HCM) are distinct disorders leading to left ventricular hypertrophy (LVH), but whether cardiac metabolism substantially differs between these in humans remains to be elucidated. We undertook an invasive (aortic root, coronary sinus) metabolic profiling in patients with severe AS and HCM in comparison with non-LVH controls to investigate cardiac fuel selection and metabolic remodeling. These patients were assessed under different physiological states (at rest, during stress induced by pacing). The identified changes in the metabolome were further validated by metabolomic and orthogonal transcriptomic analysis, in separately recruited patient cohorts. We identified a highly discriminant metabolomic signature in severe AS in all samples, regardless of sampling site, characterized by striking accumulation of long-chain acylcarnitines, intermediates of fatty acid transport across the inner mitochondrial membrane, and validated this in a separate cohort. Mechanistically, we identify a downregulation in the PPAR-α transcriptional network, including expression of genes regulating fatty acid oxidation (FAO). In silico modeling of ß-oxidation demonstrated that flux could be inhibited by both the accumulation of fatty acids as a substrate for mitochondria and the accumulation of medium-chain carnitines which induce competitive inhibition of the acyl-CoA dehydrogenases. We present a comprehensive analysis of changes in the metabolic pathways (transcriptome to metabolome) in severe AS, and its comparison to HCM. Our results demonstrate a progressive impairment of ß-oxidation from HCM to AS, particularly for FAO of long-chain fatty acids, and that the PPAR-α signaling network may be a specific metabolic therapeutic target in AS.


Sujet(s)
Sténose aortique , Cardiomyopathie hypertrophique , Humains , Récepteurs activés par les proliférateurs de peroxysomes , Cardiomyopathie hypertrophique/génétique , Hypertrophie ventriculaire gauche/génétique , Sténose aortique/génétique , Acides gras/métabolisme
10.
Comput Biol Med ; 172: 108214, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38508057

RÉSUMÉ

Calcific aortic valve disease (CAVD) is a heart valve disorder characterized primarily by calcification of the aortic valve, resulting in stiffness and dysfunction of the valve. CAVD is prevalent among aging populations and is linked to factors such as hypertension, dyslipidemia, tobacco use, and genetic predisposition, and can result in becoming a growing economic and health burden. Once aortic valve calcification occurs, it will inevitably progress to aortic stenosis. At present, there are no medications available that have demonstrated effectiveness in managing or delaying the progression of the disease. In this study, we mined four publicly available microarray datasets (GSE12644 GSE51472, GSE77287, GSE233819) associated with CAVD from the GEO database with the aim of identifying hub genes associated with the occurrence of CAVD and searching for possible biological targets for the early prevention and diagnosis of CAVD. This study provides preliminary evidence for therapeutic and preventive targets for CAVD and may provide a solid foundation for subsequent biological studies.


Sujet(s)
Sténose aortique , Valve aortique/anatomopathologie , Calcinose , Valvulopathies , Humains , Sténose aortique/génétique , Sténose aortique/diagnostic , Sténose aortique/épidémiologie , Valvulopathies/génétique , Calcinose/génétique
11.
Eur J Pharmacol ; 968: 176423, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38365109

RÉSUMÉ

Calcific aortic valve disease (CAVD) is a progressive cardiovascular disorder involving multiple pathogenesis. Effective pharmacological therapies are currently unavailable. Sirtuin6 (SIRT6) has been shown to protect against aortic valve calcification in CAVD. The exact regulatory mechanism of SIRT6 in osteoblastic differentiation remains to be determined, although it inhibits osteogenic differentiation of aortic valve interstitial cells. We demonstrated that SIRT6 was markedly downregulated in calcific human aortic valves. Mechanistically, SIRT6 suppressed osteogenic differentiation in human aortic valve interstitial cells (HAVICs), as confirmed by loss- and gain-of-function experiments. SIRT6 directly interacted with Runx2, decreased Runx2 acetylation levels, and facilitated Runx2 nuclear export to inhibit the osteoblastic phenotype transition of HAVICs. In addition, the AKT signaling pathway acted upstream of SIRT6. Together, these findings elucidate that SIRT6-mediated Runx2 downregulation inhibits aortic valve calcification and provide novel insights into therapeutic strategies for CAVD.


Sujet(s)
Sténose aortique , Valve aortique/anatomopathologie , Calcinose , Sirtuines , Humains , Valve aortique/métabolisme , Régulation négative , Ostéogenèse/génétique , Cellules cultivées , Sténose aortique/génétique , Sténose aortique/métabolisme , Sténose aortique/anatomopathologie , Sirtuines/génétique , Sirtuines/métabolisme
12.
JAMA Cardiol ; 9(4): 357-366, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38416462

RÉSUMÉ

Importance: Polygenic risk scores (PRSs) have proven to be as strong as or stronger than established clinical risk factors for many cardiovascular phenotypes. Whether this is true for aortic stenosis remains unknown. Objective: To develop a novel aortic stenosis PRS and compare its aortic stenosis risk estimation to established clinical risk factors. Design, Setting, and Participants: This was a longitudinal cohort study using data from the Million Veteran Program (MVP; 2011-2020), UK Biobank (2006-2010), and 6 Thrombolysis in Myocardial Infarction (TIMI) trials, including DECLARE-TIMI 58 (2013-2018), FOURIER (TIMI 59; 2013-2017), PEGASUS-TIMI 54 (2010-2014), SAVOR-TIMI 53 (2010-2013), SOLID-TIMI 52 (2009-2014), and ENGAGE AF-TIMI 48 (2008-2013), which were a mix of population-based and randomized clinical trials. Individuals from UK Biobank and the MVP meeting a previously validated case/control definition for aortic stenosis were included. All individuals from TIMI trials were included unless they had a documented preexisting aortic valve replacement. Analysis took place from January 2022 to December 2023. Exposures: PRS for aortic stenosis (developed using data from MVP and validated in UK Biobank) and other previously validated cardiovascular PRSs, defined either as a continuous variable or as low (bottom 20%), intermediate, and high (top 20%), and clinical risk factors. Main Outcomes: Aortic stenosis (defined using International Classification of Diseases or Current Procedural Terminology codes in UK Biobank and MVP or safety event data in the TIMI trials). Results: The median (IQR) age in MVP was 67 (57-73) years, and 135 140 of 147 104 participants (92%) were male. The median (IQR) age in the TIMI trials was 66 (54-78) years, and 45 524 of 59 866 participants (71%) were male. The best aortic stenosis PRS incorporated 5 170 041 single-nucleotide variants and was associated with aortic stenosis in both the MVP testing sample (odds ratio, 1.41; 95% CI, 1.37-1.45 per 1 SD PRS; P = 4.6 × 10-116) and TIMI trials (hazard ratio, 1.44; 95% CI, 1.27-1.62 per 1 SD PRS; P = 3.2 × 10-9). Among genetic and clinical risk factors, the aortic stenosis PRS performed comparably to most risk factors besides age, and within a given age range, the combination of clinical and genetic risk factors was additive, providing a 3- to 4-fold increased gradient of risk of aortic stenosis. However, the addition of the aortic stenosis PRS to a model including clinical risk factors only improved risk discrimination of aortic stenosis by 0.01 to 0.02 (C index in MVP: 0.78 with clinical risk factors, 0.79 with risk factors and aortic stenosis PRS; C index in TIMI: 0.71 with clinical risk factors, 0.73 with risk factors and aortic stenosis PRS). Conclusions: This study developed and validated 1 of the first aortic stenosis PRSs. While aortic stenosis genetic risk was independent from clinical risk factors and performed comparably to all other risk factors besides age, genetic risk resulted in only a small improvement in overall aortic stenosis risk discrimination beyond age and clinical risk factors. This work sets the stage for further development of an aortic stenosis PRS.


Sujet(s)
Sténose aortique , Infarctus du myocarde , Humains , Mâle , Sujet âgé , Femelle , , Études longitudinales , Prédisposition génétique à une maladie , Facteurs de risque , Sténose aortique/génétique
13.
Ann Lab Med ; 44(3): 279-288, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38205526

RÉSUMÉ

Background: The mechanism and medical treatment target for degenerative aortic valve disease, including aortic stenosis, is not well studied. In this study, we investigated the effect of clonal hematopoiesis of indeterminate potential (CHIP) on the development of aortic valve sclerosis (AVS), a calcified aortic valve without significant stenosis. Methods: Participants with AVS (valves ≥2 mm thick, high echogenicity, and a peak transaortic velocity of <2.5 m/sec) and an age- and sex-matched control group were enrolled. Twenty-four CHIP genes with common variants in cardiovascular disease were used to generate a next-generation sequencing panel. The primary endpoint was the CHIP detection rate between the AVS and control groups. Inverse-probability treatment weighting (IPTW) analysis was performed to adjust for differences in baseline characteristics. Results: From April 2020 to April 2022, 187 participants (125 with AVS and 62 controls) were enrolled; the mean age was 72.6±8.5 yrs, and 54.5% were male. An average of 1.3 CHIP variants was observed. CHIP detection, defined by a variant allele frequency (VAF) of ≥0.5%, was similar between the groups. However, the AVS group had larger CHIP clones: 49 (39.2%) participants had a VAF of ≥1% (vs. 13 [21.0%] in the control group; P=0.020), and 25 (20.0%) had a VAF of ≥2% (vs. 4 [6.5%]; P=0.028). AVS is independently associated with a VAF of ≥1% (adjusted odds ratio: 2.44, 95% confidence interval: 1.11-5.36; P=0.027). This trend was concordant and clearer in the IPTW cohort. Conclusions: Participants with AVS more commonly had larger CHIP clones than age- and sex-matched controls. Further studies are warranted to identify causality between AVS and CHIP.


Sujet(s)
Sténose aortique , Calcinose , Humains , Mâle , Adulte d'âge moyen , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Valve aortique/imagerie diagnostique , Valve aortique/anatomopathologie , Hématopoïèse clonale , Sclérose/anatomopathologie , Sténose aortique/diagnostic , Sténose aortique/génétique , Sténose aortique/anatomopathologie , Calcinose/anatomopathologie
14.
Circulation ; 149(20): 1578-1597, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38258575

RÉSUMÉ

BACKGROUND: Calcification of the aortic valve leads to increased leaflet stiffness and consequently results in the development of calcific aortic valve disease (CAVD). However, the underlying molecular and cellular mechanisms of calcification remain unclear. Here, we identified a novel aortic valve calcification-associated PIWI-interacting RNA (piRNA; AVCAPIR) that increases valvular calcification and promotes CAVD progression. METHODS: Using piRNA sequencing, we identified piRNAs contributing to the pathogenesis of CAVD that we termed AVCAPIRs. High-cholesterol diet-fed ApoE-/- mice with AVCAPIR knockout were used to examine the role of AVCAPIR in aortic valve calcification (AVC). Gain- and loss-of-function assays were conducted to determine the role of AVCAPIR in the induced osteogenic differentiation of human valvular interstitial cells. To dissect the mechanisms underlying AVCAPIR-elicited procalcific effects, we performed various analyses, including an RNA pulldown assay followed by liquid chromatography-tandem mass spectrometry, methylated RNA immunoprecipitation sequencing, and RNA sequencing. RNA pulldown and RNA immunoprecipitation assays were used to study piRNA interactions with proteins. RESULTS: We found that AVCAPIR was significantly upregulated during AVC and exhibited potential diagnostic value for CAVD. AVCAPIR deletion markedly ameliorated AVC in high-cholesterol diet-fed ApoE-/- mice, as shown by reduced thickness and calcium deposition in the aortic valve leaflets, improved echocardiographic parameters (decreased peak transvalvular jet velocity and mean transvalvular pressure gradient, as well as increased aortic valve area), and diminished levels of osteogenic markers (Runx2 and Osterix) in aortic valves. These results were confirmed in osteogenic medium-induced human valvular interstitial cells. Using unbiased protein-RNA screening and molecular validation, we found that AVCAPIR directly interacts with FTO (fat mass and obesity-associated protein), subsequently blocking its N6-methyladenosine demethylase activity. Further transcriptomic and N6-methyladenosine modification epitranscriptomic screening followed by molecular validation confirmed that AVCAPIR hindered FTO-mediated demethylation of CD36 mRNA transcripts, thus enhancing CD36 mRNA stability through the N6-methyladenosine reader IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1). In turn, the AVCAPIR-dependent increase in CD36 stabilizes its binding partner PCSK9 (proprotein convertase subtilisin/kexin type 9), a procalcific gene, at the protein level, which accelerates the progression of AVC. CONCLUSIONS: We identified a novel piRNA that induced AVC through an RNA epigenetic mechanism and provide novel insights into piRNA-directed theranostics in CAVD.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , Petit ARN interférent , Animaux , Calcinose/métabolisme , Calcinose/génétique , Calcinose/anatomopathologie , Valve aortique/métabolisme , Valve aortique/anatomopathologie , Valve aortique/malformations , Humains , Souris , Sténose aortique/métabolisme , Sténose aortique/génétique , Sténose aortique/anatomopathologie , Petit ARN interférent/métabolisme , Petit ARN interférent/génétique , Mâle , Ostéogenèse , Souris de lignée C57BL , Souris knockout , Modèles animaux de maladie humaine , Maladie de la valve aortique/métabolisme , Maladie de la valve aortique/génétique , Maladie de la valve aortique/anatomopathologie , ARN interagissant avec Piwi
15.
Am J Pathol ; 194(4): 539-550, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37517686

RÉSUMÉ

This review focuses on technologies at the core of calcific aortic valve disease (CAVD) and drug target research advancement, including transcriptomics, proteomics, and molecular imaging. We examine how bulk RNA sequencing and single-cell RNA sequencing have engendered organismal genomes and transcriptomes, promoting the analysis of tissue gene expression profiles and cell subpopulations, respectively. We bring into focus how the field is also largely influenced by increasingly accessible proteome profiling techniques. In unison, global transcriptional and protein expression analyses allow for increased understanding of cellular behavior and pathogenic pathways under pathologic stimuli including stress, inflammation, low-density lipoprotein accumulation, increased calcium and phosphate levels, and vascular injury. We also look at how direct investigation of protein signatures paves the way for identification of targetable pathways for pharmacologic intervention. Here, we note that imaging techniques, once a clinical diagnostic tool for late-stage CAVD, have since been refined to address a clinical need to identify microcalcifications using positron emission tomography/computed tomography and even detect in vivo cellular events indicative of early stage CAVD and map the expression of identified proteins in animal models. Together, these techniques generate a holistic approach to CAVD investigation, with the potential to identify additional novel regulatory pathways.


Sujet(s)
Sténose aortique , Valve aortique/anatomopathologie , Calcinose , Animaux , Valve aortique/métabolisme , Sténose aortique/génétique , Sténose aortique/métabolisme , Sténose aortique/anatomopathologie , Analyse de profil d'expression de gènes , Calcinose/génétique , Calcinose/métabolisme
16.
Clin Hemorheol Microcirc ; 86(3): 285-302, 2024.
Article de Anglais | MEDLINE | ID: mdl-37355887

RÉSUMÉ

Calcific aortic valve disease (CAVD) is featured by thickening and calcification of the aortic valve. Osteoblast differentiation is a crucial step in valve calcification. Long non-coding RNAs (LncRNAs) participate in the osteogenic differentiation of mesenchymal cells. However, the character of lncRNA FGD5 antisense RNA 1 (FGD5-AS1) in CAVD is uncertain. After collection of human aortic valve tissue samples, detection of FGD5-AS1, microRNA (miR)-497-5p and Baculovirus inhibitor 5 (BIRC5) was conducted. Valve mesenchymal cells were isolated from CAVD patients and induced to differentiate to osteoblasts, and transfected with FGD5-AS1, miR-497-5p and BIRC5 plasmids. Detection of the alkaline phosphatase activity was after osteogenic induction of human aortic valve interstitial cells (hAVICs); Detection of the degree of calcium nodules and osteoblast differentiation markers (RUNX2 and OPN) was conducted. After establishment of a mouse model of CAVD, detection of the thickness of aortic valve leaflets, and the degree of calcification of the valve leaflets, and evaluation of echocardiographic parameters were implemented. Experimental data manifested in CAVD patients, lncRNAFGD5-AS1 and BIRC5 were reduced, but miR-497-5p was elevated; Enhancing lncRNA FGD5-AS1 or repressing miR-497-5p mitigated CAVD by restraining osteogenic differentiation; LncRNA FGD5-AS1 sponged miR-497-5p to target BIRC5; Repressive BIRC5 turned around the therapeutic action of elevated FGD5-AS1 or depressed miR-497-5p on hAVICs; Enhancive FGD5-AS1 in vivo was available to reduce ApoE-/- mouse CAVD induced via high cholesterol diet. All in all, lncRNAFGD5-AS1 targets BIRC5 via miR-497-5p to alleviate CAVD.


Sujet(s)
Sténose aortique , Valve aortique , Calcinose , microARN , ARN long non codant , Survivine , Animaux , Humains , Souris , Valve aortique/anatomopathologie , Sténose aortique/génétique , Facteurs d'échange de nucléotides guanyliques/génétique , microARN/génétique , Ostéogenèse/génétique , ARN long non codant/génétique , Survivine/génétique , Survivine/métabolisme
17.
Cardiovasc Res ; 120(1): 95-107, 2024 02 27.
Article de Anglais | MEDLINE | ID: mdl-37897683

RÉSUMÉ

AIMS: Adiponectin may play an important protective role in heart failure and associated cardiovascular diseases. We hypothesized that plasma adiponectin is associated observationally and causally, genetically with risk of heart failure, atrial fibrillation, aortic valve stenosis, and myocardial infarction. METHODS AND RESULTS: In the Copenhagen General Population Study, we examined 30 045 individuals with plasma adiponectin measurements observationally and 96 903 individuals genetically in one-sample Mendelian randomization analyses using five genetic variants explaining 3% of the variation in plasma adiponectin. In the HERMES, UK Biobank, The Nord-Trøndelag Health Study (HUNT), deCODE, the Michigan Genomics Initiative (MGI), DiscovEHR, and the AFGen consortia, we performed two-sample Mendelian randomization analyses in up to 1 030 836 individuals using 12 genetic variants explaining 14% of the variation in plasma adiponectin.In observational analyses modelled linearly, a 1 unit log-transformed higher plasma adiponectin was associated with a hazard ratio of 1.51 (95% confidence interval: 1.37-1.66) for heart failure, 1.63 (1.50-1.78) for atrial fibrillation, 1.21 (1.03-1.41) for aortic valve stenosis, and 1.03 (0.93-1.14) for myocardial infarction; levels above the median were also associated with an increased risk of myocardial infarction, and non-linear U-shaped associations were more apparent for heart failure, aortic valve stenosis, and myocardial infarction in less-adjusted models. Corresponding genetic, causal risk ratios were 0.92 (0.65-1.29), 0.87 (0.68-1.12), 1.55 (0.87-2.76), and 0.93 (0.67-1.30) in one-sample Mendelian randomization analyses, and no significant associations were seen for non-linear one-sample Mendelian randomization analyses; corresponding causal risk ratios were 0.99 (0.89-1.09), 1.00 (0.92-1.08), 1.01 (0.79-1.28), and 0.99 (0.86-1.13) in two-sample Mendelian randomization analyses, respectively. CONCLUSION: Observationally, elevated plasma adiponectin was associated with an increased risk of heart failure, atrial fibrillation, aortic valve stenosis, and myocardial infarction. However, genetic evidence did not support causality for these associations.


Sujet(s)
Sténose aortique , Fibrillation auriculaire , Défaillance cardiaque , Infarctus du myocarde , Humains , Adiponectine/génétique , Sténose aortique/épidémiologie , Sténose aortique/génétique , Fibrillation auriculaire/diagnostic , Fibrillation auriculaire/épidémiologie , Fibrillation auriculaire/génétique , Étude d'association pangénomique , Défaillance cardiaque/diagnostic , Défaillance cardiaque/épidémiologie , Défaillance cardiaque/génétique , Analyse de randomisation mendélienne/méthodes , Infarctus du myocarde/diagnostic , Infarctus du myocarde/épidémiologie , Infarctus du myocarde/génétique , Polymorphisme de nucléotide simple , Facteurs de risque
18.
Circulation ; 149(5): 391-401, 2024 01 30.
Article de Anglais | MEDLINE | ID: mdl-37937463

RÉSUMÉ

BACKGROUND: High circulating levels of Lp(a) (lipoprotein[a]) increase the risk of atherosclerosis and calcific aortic valve disease, affecting millions of patients worldwide. Although atherosclerosis is commonly treated with low-density lipoprotein-targeting therapies, these do not reduce Lp(a) or risk of calcific aortic valve disease, which has no available drug therapies. Targeting Lp(a) production and catabolism may provide therapeutic benefit, but little is known about Lp(a) cellular uptake. METHODS: Here, unbiased ligand-receptor capture mass spectrometry was used to identify MFSD5 (major facilitator superfamily domain containing 5) as a novel receptor/cofactor involved in Lp(a) uptake. RESULTS: Reducing MFSD5 expression by a computationally identified small molecule or small interfering RNA suppressed Lp(a) uptake and calcification in primary human valvular endothelial and interstitial cells. MFSD5 variants were associated with aortic stenosis (P=0.027 after multiple hypothesis testing) with evidence suggestive of an interaction with plasma Lp(a) levels. CONCLUSIONS: MFSD5 knockdown suppressing human valvular cell Lp(a) uptake and calcification, along with meta-analysis of MFSD5 variants associating with aortic stenosis, supports further preclinical assessment of MFSD5 in cardiovascular diseases, the leading cause of death worldwide.


Sujet(s)
Maladie de la valve aortique , Sténose aortique , Athérosclérose , Calcinose , Valvulopathies , Humains , Valve aortique/métabolisme , Maladie de la valve aortique/métabolisme , Sténose aortique/traitement médicamenteux , Sténose aortique/génétique , Athérosclérose/métabolisme , Valvulopathies/traitement médicamenteux , Valvulopathies/génétique , Valvulopathies/complications , Lipoprotéine (a) , Facteurs de risque
19.
J Coll Physicians Surg Pak ; 33(12): 1344-1348, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38062586

RÉSUMÉ

OBJECTIVE: To investigate the potential treatments for aortic stenosis and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using bioinformatics and systems biology. STUDY DESIGN: Observational study. Place and Duration of the Study: Jiading District Central Hospital affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China, from August to December 2022. METHODOLOGY: GSE147507 was chosen as the SARS-CoV-2 infection dataset from the Biotechnology Information (NCBI) GEO database, while GSE153555 was chosen as the dataset of patients with aortic stenosis (AS). This analysis predicted protein-drug interactions (PDIs) and found therapeutic compounds for AS and COVID-19. RESULTS: One hundred and four DEGs were shared between the two datasets. Researchers built a PPI network to identify 10 hub genes from the network. Researchers discovered that COVID-19 and AS shared certain pathogenic pathways and found a relationship between hub genes and transcription factors and miRNAs, as well as a connection between hub genes and proposed treatments. CONCLUSION: Hub genes were identified as potential pathogenic pathways in SARS-CoV-2 infection and AS. In addition, new prescription medication options for treating both illnesses were provided. KEY WORDS: SARS-CoV-2 infection, COVID-19, Aortic stenosis, Differentially expressed genes, Hub genes, Gene-disease, Drug molecule.


Sujet(s)
Sténose aortique , COVID-19 , microARN , Humains , SARS-CoV-2 , Chine , Sténose aortique/épidémiologie , Sténose aortique/génétique , Biologie informatique
20.
Sci Rep ; 13(1): 20284, 2023 11 20.
Article de Anglais | MEDLINE | ID: mdl-37985719

RÉSUMÉ

Calcific aortic valve stenosis (CAVS) is associated with an increased risk of atrial fibrillation (AF) in observational studies, but whether these associations are causal has not been determined. This study aimed to explore the potential causal relationship between CAVS and AF via Mendelian randomization (MR). Genetic variants from the genome-wide association study (GWAS) summary data of the European population for CAVS were used to investigate the association with AF. The inverse variance weighted (IVW) approach was used to obtain the primary causal inference, and several sensitivity analysis approaches, such as the MR‒Egger and weighted median (WM), were performed to assess the robustness of the results. A total of nineteen valid and independent genetic SNPs associated with CAVS were obtained from the GWAS database. Genetically predicted CAVS (OR: 1.105; 95% CI: 1.072-1.139; p = 8.60E-11) was associated with an increased risk of AF. Similar results were discovered in the sensitivity analyses by using MR Egger and weighted median approaches. An MR design was used to reduce confounding variables and the potential for reverse causality bias. The results provide genetic evidence that CAVS considerably increased the risk of AF.


Sujet(s)
Sténose aortique , Fibrillation auriculaire , Humains , Fibrillation auriculaire/génétique , Étude d'association pangénomique , Analyse de randomisation mendélienne , Sténose aortique/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...