Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 325
Filter
1.
Int J Cardiol ; 408: 132067, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38714235

ABSTRACT

AIMS: The aim of this study is to evaluate the effect of beta-blockers and angiotensin receptor blockers in reducing the aortic growth rate in children with bicuspid aortic valve (BAV)-related aortopathy and ascending phenotype. METHODS: Consecutive paediatric patients (≤16 years) with BAV and ascending aorta (AsAo) dilation (z-score > 3) were enrolled in this observational retrospective cohort study. Patients receiving prophylactic treatment with either atenolol (0.5 to 1.0 mg/kg/daily) or losartan (0.7 to 1.4 mg/kg/daily) were compared with those who did not receive medical prophylaxis (control group). The primary outcome of interest was the annual rate of change in maximal AsAo diameter z-score in the treatment and control groups. RESULTS: From a cohort of 1005 patients, 120 (mean age 11.3 ± 4.5 years, 82% males) fulfilled the inclusion criteria and were included in the study. Patients in the treatment and control group had similar age, sex, family history of BAV, BAV morphology, and baseline AsAo diameter. During a median follow-up of 7.1 years (interquartile range 3.8-10.2), no differences were observed in the annual growth rate of aortic diameter z-score between patients on treatment and controls. The prevalence of aortic diameter progression was similar in the treatment and control groups, and treatment with atenolol or losartan was not associated with a lower rate of aortic disease progression. CONCLUSIONS: The findings revealed no significant difference in the annual aortic growth rate between treated and untreated patients. Larger cohort studies or, ideally, randomized clinical controlled trials are needed to validate these findings.


Subject(s)
Adrenergic beta-Antagonists , Aortic Valve , Bicuspid Aortic Valve Disease , Humans , Male , Female , Child , Retrospective Studies , Adolescent , Adrenergic beta-Antagonists/therapeutic use , Aortic Valve/abnormalities , Aortic Valve/diagnostic imaging , Aortic Valve/pathology , Aortic Valve/drug effects , Angiotensin Receptor Antagonists/therapeutic use , Losartan/therapeutic use , Follow-Up Studies , Cohort Studies , Atenolol/therapeutic use , Treatment Outcome , Aorta/drug effects , Aorta/diagnostic imaging , Aortic Valve Disease/drug therapy , Heart Valve Diseases/drug therapy , Heart Valve Diseases/complications , Angiotensin II Type 1 Receptor Blockers/therapeutic use
2.
Circulation ; 149(25): 1938-1948, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38804148

ABSTRACT

BACKGROUND: Ascending aorta dilation and aortic valve degeneration are common complications in patients with bicuspid aortic valve. Several retrospective studies have suggested the benefit of statins in reducing these complications. This study aimed to determine whether atorvastatin treatment is effective in reducing the growth of aortic diameters in bicuspid aortic valve and if it slows the progression of valve calcification. METHODS: In a randomized clinical trial, 220 patients with bicuspid aortic valve (43 women; 46±13 years of age) were included and treated with either 20 mg of atorvastatin per day or placebo for 3 years. Inclusion criteria were ≥18 years of age, nonsevere valvular dysfunction, nonsevere valve calcification, and ascending aorta diameter ≤50 mm. Computed tomography and echocardiography studies were performed at baseline and after 3 years of treatment. RESULTS: During follow-up, 28 patients (12.7%) discontinued medical treatment (15 on atorvastatin and 13 taking placebo). Thus, 192 patients completed the 36 months of treatment. Low-density lipoprotein cholesterol levels decreased significantly in the atorvastatin group (median [interquartile range], -30 mg/dL [-51.65 to -1.75 mg/dL] versus 6 mg/dL [-4, 22.5 mg/dL]; P<0.001). The maximum ascending aorta diameter increased with no differences between groups: 0.65 mm (95% CI, 0.45-0.85) in the atorvastatin group and 0.74 mm (95% CI, 0.45-1.04) in the placebo group (P=0.613). Similarly, no significant differences were found for the progression of the aortic valve calcium score (P=0.167) or valvular dysfunction. CONCLUSIONS: Among patients with bicuspid aortic valve without severe valvular dysfunction, atorvastatin treatment was not effective in reducing the progression of ascending aorta dilation and aortic valve calcification during 3 years of treatment despite a significant reduction in low-density lipoprotein cholesterol levels. REGISTRATION: URL: https://www.clinicaltrialsregister.eu; Unique identifier: 2015-001808-57. URL: https://www.clinicaltrials.gov; Unique identifier: NCT02679261.


Subject(s)
Aortic Valve , Atorvastatin , Bicuspid Aortic Valve Disease , Calcinosis , Disease Progression , Heart Valve Diseases , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Atorvastatin/therapeutic use , Female , Male , Middle Aged , Aortic Valve/diagnostic imaging , Aortic Valve/pathology , Aortic Valve/abnormalities , Aortic Valve/drug effects , Calcinosis/drug therapy , Calcinosis/diagnostic imaging , Calcinosis/pathology , Bicuspid Aortic Valve Disease/diagnostic imaging , Bicuspid Aortic Valve Disease/drug therapy , Heart Valve Diseases/drug therapy , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/pathology , Adult , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Dilatation, Pathologic/drug therapy , Follow-Up Studies , Double-Blind Method , Treatment Outcome , Aorta/diagnostic imaging , Aorta/pathology , Aorta/drug effects , Aortic Valve Disease/drug therapy , Aortic Valve Stenosis
3.
Br J Pharmacol ; 181(12): 1843-1856, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38378175

ABSTRACT

BACKGROUND AND PURPOSE: Our previous studies have found that andrographolide (AGP) alleviates calcific aortic valve disease (CAVD), but the underlying mechanism is unclear. This study explores the molecular target and signal mechanisms of AGP in inhibiting CAVD. EXPERIMENTAL APPROACH: The anti-calcification effects of the aortic valve with AGP treatment were evaluated by alizarin red staining in vitro and ultrasound and histopathological assessment of a high-fat (HF)-fed ApoE-/- mouse valve calcification model. A correlation between the H3 histone lactylation (H3Kla) and calcification was detected. Molecular docking and surface plasmon resonance (SPR) experiments were further used to confirm p300 as a target for AGP. Overexpression (oe) and silencing (si) of p300 were used to verify the inhibitory effect of AGP targeting p300 on the H3Kla in vitro and ex vivo. KEY RESULTS: AGP significantly inhibited calcium deposition in valve interstitial cells (VICs) and ameliorated aortic valve calcification. The multi-omics analysis revealed the glycolysis pathway involved in CAVD, indicating that AGP interfered with lactate production by regulating lactate dehydrogenase A (LDHA). In addition, lactylation, a new post-translational modification, was shown to have a role in promoting aortic valve calcification. Furthermore, H3Kla and H3K9la site were shown to correlate with Runx2 expression inhibition by AGP treatment. Importantly, we found that p300 transferase was the molecular target of AGP in inhibiting H3Kla. CONCLUSIONS AND IMPLICATIONS: Our findings, for the first time, demonstrated that AGP alleviates calcification by interfering with H3Kla via p300, which might be a powerful drug to prevent CAVD.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Diterpenes , Histones , Animals , Humans , Male , Mice , Aortic Valve/pathology , Aortic Valve/metabolism , Aortic Valve/drug effects , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/metabolism , Calcinosis/drug therapy , Calcinosis/pathology , Diterpenes/pharmacology , Diterpenes/chemistry , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/antagonists & inhibitors , Histones/metabolism , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors
4.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35181609

ABSTRACT

Aortic valve stenosis (AVS) patients experience pathogenic valve leaflet stiffening due to excessive extracellular matrix (ECM) remodeling. Numerous microenvironmental cues influence pathogenic expression of ECM remodeling genes in tissue-resident valvular myofibroblasts, and the regulation of complex myofibroblast signaling networks depends on patient-specific extracellular factors. Here, we combined a manually curated myofibroblast signaling network with a data-driven transcription factor network to predict patient-specific myofibroblast gene expression signatures and drug responses. Using transcriptomic data from myofibroblasts cultured with AVS patient sera, we produced a large-scale, logic-gated differential equation model in which 11 biochemical and biomechanical signals were transduced via a network of 334 signaling and transcription reactions to accurately predict the expression of 27 fibrosis-related genes. Correlations were found between personalized model-predicted gene expression and AVS patient echocardiography data, suggesting links between fibrosis-related signaling and patient-specific AVS severity. Further, global network perturbation analyses revealed signaling molecules with the most influence over network-wide activity, including endothelin 1 (ET1), interleukin 6 (IL6), and transforming growth factor ß (TGFß), along with downstream mediators c-Jun N-terminal kinase (JNK), signal transducer and activator of transcription (STAT), and reactive oxygen species (ROS). Lastly, we performed virtual drug screening to identify patient-specific drug responses, which were experimentally validated via fibrotic gene expression measurements in valvular interstitial cells cultured with AVS patient sera and treated with or without bosentan-a clinically approved ET1 receptor inhibitor. In sum, our work advances the ability of computational approaches to provide a mechanistic basis for clinical decisions including patient stratification and personalized drug screening.


Subject(s)
Aortic Valve/metabolism , Gene Expression Profiling/methods , Precision Medicine/methods , Actins/metabolism , Aortic Valve/drug effects , Aortic Valve/physiology , Aortic Valve Stenosis/metabolism , Biomarkers, Pharmacological , Calcinosis/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Cicatrix/metabolism , Computational Biology/methods , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibrosis , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , Models, Genetic , Myofibroblasts/metabolism , Myofibroblasts/physiology , Serum/metabolism , Signal Transduction , Transcriptome/genetics
5.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638942

ABSTRACT

Calcific aortic valve disease (CAVD) is an athero-inflammatory process. Growing evidence supports the inflammation-driven calcification model, mediated by cytokines such as interferons (IFNs) and tumor necrosis factor (TNF)-α. Our goal was investigating IFNs' effects in human aortic valve endothelial cells (VEC) and the potential differences between aortic (aVEC) and ventricular (vVEC) side cells. The endothelial phenotype was analyzed by Western blot, qPCR, ELISA, monocyte adhesion, and migration assays. In mixed VEC populations, IFNs promoted the activation of signal transducers and activators of transcription-1 and nuclear factor-κB, and the subsequent up-regulation of pro-inflammatory molecules. Side-specific VEC were activated with IFN-γ and TNF-α in an orbital shaker flow system. TNF-α, but not IFN-γ, induced hypoxia-inducible factor (HIF)-1α stabilization or endothelial nitric oxide synthase downregulation. Additionally, IFN-γ inhibited TNF-α-induced migration of aVEC. Also, IFN-γ triggered cytokine secretion and adhesion molecule expression in aVEC and vVEC. Finally, aVEC were more prone to cytokine-mediated monocyte adhesion under multiaxial flow conditions as compared with uniaxial flow. In conclusion, IFNs promote inflammation and reduce TNF-α-mediated migration in human VEC. Moreover, monocyte adhesion was higher in inflamed aVEC sheared under multiaxial flow, which may be relevant to understanding the initial stages of CAVD.


Subject(s)
Aortic Valve/metabolism , Endothelial Cells/metabolism , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Signal Transduction/drug effects , Stress, Physiological/immunology , Aortic Valve/drug effects , Aortic Valve/immunology , Aortic Valve/pathology , Aortic Valve Stenosis/immunology , Calcinosis/immunology , Cell Adhesion/drug effects , Cell Movement/drug effects , Endothelial Cells/drug effects , Heart Transplantation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/chemically induced , Inflammation/immunology , Monocytes/metabolism , NF-kappa B/metabolism , Phenotype , STAT1 Transcription Factor/metabolism , THP-1 Cells , Transplant Recipients , Tumor Necrosis Factor-alpha/pharmacology
6.
J Cardiovasc Pharmacol ; 79(1): e103-e115, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654784

ABSTRACT

ABSTRACT: Aortic valve replacement for severe stenosis is a standard procedure in cardiovascular medicine. However, the use of biological prostheses has limitations especially in young patients because of calcifying degeneration, resulting in implant failure. Pioglitazone, a peroxisome proliferator-activated receptor gamma (PPAR-gamma) agonist, was shown to decrease the degeneration of native aortic valves. In this study, we aim to examine the impact of pioglitazone on inflammation and calcification of aortic valve conduits (AoC) in a rat model. Cryopreserved AoC (n = 40) were infrarenally implanted into Wistar rats treated with pioglitazone (75 mg/kg chow; n = 20, PIO) or untreated (n = 20, controls). After 4 or 12 weeks, AoC were explanted and analyzed by histology, immunohistology, and polymerase chain reaction. Pioglitazone significantly decreased the expression of inflammatory markers and reduced the macrophage-mediated inflammation in PIO compared with controls after 4 (P = 0.03) and 12 weeks (P = 0.012). Chondrogenic transformation was significantly decreased in PIO after 12 weeks (P = 0.001). Calcification of the intima and media was significantly reduced after 12 weeks in PIO versus controls (intima: P = 0.008; media: P = 0.025). Moreover, echocardiography revealed significantly better functional outcome of the AoC in PIO after 12 weeks compared with control. Interestingly, significantly increased intima hyperplasia could be observed in PIO compared with controls after 12 weeks (P = 0.017). Systemic PPAR-gamma activation prevents inflammation as well as intima and media calcification in AoC and seems to inhibit functional impairment of the implanted aortic valve. To further elucidate the therapeutic role of PPAR-gamma regulation for graft durability, translational studies and long-term follow-up data should be striven for.


Subject(s)
Aortic Valve Insufficiency/surgery , Aortic Valve/drug effects , Aortic Valve/transplantation , Bioprosthesis , Heart Valve Prosthesis Implantation/instrumentation , Heart Valve Prosthesis , PPAR gamma/agonists , Pioglitazone/pharmacology , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve Insufficiency/metabolism , Aortic Valve Insufficiency/pathology , Calcinosis/metabolism , Calcinosis/pathology , Calcinosis/prevention & control , Chondrogenesis/drug effects , Cryopreservation , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Heart Valve Prosthesis Implantation/adverse effects , Humans , Inflammation Mediators/metabolism , Osteogenesis/drug effects , PPAR gamma/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction
7.
Ann Biomed Eng ; 49(12): 3550-3562, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34704164

ABSTRACT

The renin-angiotensin system (RAS) is activated in aortic valve disease, yet little is understood about how it affects the acute functional response of valve interstitial cells (VICs). Herein, we developed a gelatin-based valve thin film (vTF) platform to investigate whether the contractile response of VICs can be regulated via RAS mediators and inhibitors. First, the impact of culture medium (quiescent, activated, and osteogenic medium) on VIC phenotype and function was assessed. Contractility of VICs was measured upon treatment with angiotensin I (Ang I), angiotensin II (Ang II), angiotensin-converting enzyme (ACE) inhibitor, and Angiotensin II type 1 receptor (AT1R) inhibitor. Anisotropic cell alignment on gelatin vTF was achieved independent of culture conditions. Cells cultured in activated and osteogenic conditions were found to be more elongated than in quiescent medium. Increased α-SMA expression was observed in activated medium and no RUNX2 expression were observed in cells. VIC contractile stress increased with increasing concentrations (from 10-10 to 10-6 M) of Ang I and Ang II. Moreover, cell contraction was significantly reduced in all ACE and AT1R inhibitor-treated groups. Together, these findings suggest that local RAS is active in VICs, and our vTF may provide a powerful platform for valve drug screening and development.


Subject(s)
Aortic Valve/cytology , Renin-Angiotensin System/physiology , Angiotensin I/pharmacology , Angiotensin I/physiology , Angiotensin II/pharmacology , Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aortic Valve/drug effects , Cells, Cultured , Fluorescent Antibody Technique , Losartan/pharmacology , Myofibroblasts/physiology , Peptidyl-Dipeptidase A/physiology , Receptor, Angiotensin, Type 1/physiology , Swine , Tetrahydroisoquinolines/pharmacology
8.
Biomed Pharmacother ; 139: 111674, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34243610

ABSTRACT

OBJECTIVES: In calcific aortic valve disease (CAVD), the valve interstitial cells (VIC) osteogenic phenotype changes can lead to thickening and calcification of the valve leaflets,eventually lead to restricted valve movement and life-threatening. This study aims to investigate the effect and mechanism of dihydrotanshinone I (DHI) on osteogenic medium (OM) induced osteogenic phenotypic transition of porcine valve interstitial cells (PVICs), which can provide theoretical and scientific basis for clinical intervention in CAVD. METHODS AND RESULTS: Immunohistochemical methods were used to detect the expression of osteogenic indicators Runx2, OPN and inflammation indicators IL-1ß and p-NF-κB in valve specimens of CAVD patients(N = 3) and normal controls(N = 1). PVICs stimulated by osteoblastic medium (OM) were treated with or without DHI. CCK8, ALP and Alizarin Red S staining were used to detect cell growth and calcification, respectively. The results showed that under the treated with DHI, compared with OM, the formation of calcium nodules was reduced, and the expression of calcification-related markers Runx2 and OPN were down-regulated, which quantified by qRT-PCR and western blot. In addition, on the basis of OM induction, DHI also inhibited the phosphorylation of the NF-κB/ERK1/2 and SMAD1/5/8 signaling pathway. CONCLUSION: DHI (10 µM) treatment can reverse the osteogenic phenotypic transition of PVICs induced by osteogenic medium, and the mechanism may be related to NF-κB、ERK 1/2 and Smad1/5/8 pathways.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/drug effects , Aortic Valve/pathology , Calcinosis/drug therapy , Furans/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Phenanthrenes/pharmacology , Quinones/pharmacology , Signal Transduction/drug effects , Smad Proteins/metabolism , Animals , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Calcinosis/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Humans , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Swine
9.
J Cardiovasc Pharmacol ; 78(3): 411-421, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34132687

ABSTRACT

ABSTRACT: Calcific aortic valve disease is a common heart disease that contributes to increased cardiovascular morbidity and mortality. There is a lack of effective pharmaceutical therapy because its mechanisms are not yet fully known. Ginkgo biloba extract (EGB761) is reported to alleviate vascular calcification. However, whether EGB761 protects against aortic valve calcification, a disease whose pathogenesis shares many similarities with vascular calcification, and potential molecular mechanisms remain unknown. In this study, porcine aortic valve interstitial cell (pAVIC) calcification was induced by warfarin with or without the presence of EGB761. Immunostaining was performed to establish and characterize the pAVIC phenotype. Calcium deposition and calcium content were examined by Alizarin Red S staining and an intracellular calcium content assay. Alkaline phosphatase activity was detected by the p-nitrophenyl phosphate method. The expression levels of bone morphogenetic protein-2 (BMP2), Runt-related transcription factor 2 (Runx2), homeobox protein MSX-2, and phosphorylated (p)-Smad1/5 were detected by reverse transcription-quantitative polymerase chain reaction (PCR) and Western blot analysis. Consistent with these in vitro data, we also confirmed the suppression of in vivo calcification by EGB761 in the warfarin-induced C57/Bl6 mice. The results indicated that both pAVICs and aortic valves tissue of mice stimulated with warfarin showed increased calcium deposition and expression of osteogenic markers (alkaline phosphatase, BMP2, homeobox protein MSX-2, and Runx2) and promoted p-Smad1/5 translocation from the cytoplasm to the nucleus. The addition of EGB761 significantly inhibited p-Smad1/5 translocation from the cytoplasm to the nucleus, thus suppressing calcification. In conclusion, EGB761 could ameliorate warfarin-induced aortic valve calcification through the inhibition of the BMP2-medicated Smad1/5/Runx2 signaling pathway.


Subject(s)
Aortic Valve/drug effects , Bone Morphogenetic Protein 2/metabolism , Calcinosis/prevention & control , Core Binding Factor Alpha 1 Subunit/metabolism , Heart Valve Diseases/prevention & control , Plant Extracts/pharmacology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Active Transport, Cell Nucleus , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/chemically induced , Calcinosis/metabolism , Calcinosis/pathology , Calcium/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Disease Models, Animal , Ginkgo biloba , Heart Valve Diseases/chemically induced , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Osteogenesis/drug effects , Phosphorylation , Signal Transduction , Sus scrofa , Warfarin
10.
FEBS J ; 288(22): 6528-6542, 2021 11.
Article in English | MEDLINE | ID: mdl-34009721

ABSTRACT

Calcific aortic valve disease (CAVD) is the most prevalent valvulopathy worldwide. Growing evidence supports a role for viral and cell-derived double-stranded (ds)-RNA in cardiovascular pathophysiology. Poly(I:C), a dsRNA surrogate, has been shown to induce inflammation, type I interferon (IFN) responses, and osteogenesis through Toll-like receptor 3 in aortic valve interstitial cells (VIC). Here, we aimed to determine whether IFN signaling via Janus kinase (JAK)/Signal transducers and activators of transcription (STAT) mediates dsRNA-induced responses in primary human VIC. Western blot, ELISA, qPCR, calcification, flow cytometry, and enzymatic assays were performed to evaluate the mechanisms of dsRNA-induced inflammation and calcification. Poly(I:C) triggered a type I IFN response characterized by IFN-regulatory factors gene upregulation, IFN-ß secretion, and STAT1 activation. Additionally, Poly(I:C) promoted VIC inflammation via NF-κB and subsequent adhesion molecule expression, and cytokine secretion. Pretreatment with ruxolitinib, a clinically used JAK inhibitor, abrogated these responses. Moreover, Poly(I:C) promoted a pro-osteogenic phenotype and increased VIC calcification to a higher extent in cells from males. Inhibition of JAK with ruxolitinib or a type I IFN receptor blocking antibody blunted Poly(I:C)-induced calcification. Mechanistically, Poly(I:C) promoted VIC apoptosis in calcification medium, which was inhibited by ruxolitinib. Moreover, Poly(I:C) co-operated with IFN-γ to increase VIC calcification by synergistically activating extracellular signal-regulated kinases and hypoxia-inducible factor-1α pathways. In conclusion, JAK/STAT signaling mediates dsRNA-triggered inflammation, apoptosis, and calcification and may contribute to a positive autocrine loop in human VIC in the presence of IFN-γ. Blockade of dsRNA responses with JAK inhibitors may be a promising therapeutic avenue for CAVD.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/drug effects , Aortic Valve/pathology , Calcinosis/drug therapy , Inflammation/drug therapy , Janus Kinase Inhibitors/pharmacology , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , Adolescent , Adult , Aged , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Janus Kinase Inhibitors/chemistry , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Male , Middle Aged , Nitriles/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , RNA, Double-Stranded/metabolism , Young Adult
11.
Cells ; 10(1)2021 01 01.
Article in English | MEDLINE | ID: mdl-33401457

ABSTRACT

Calcific aortic valve disease (CAVD) accompanies inflammatory cell infiltration, fibrosis, and ultimately calcification of the valve leaflets. We previously demonstrated that dipeptidyl peptidase-4 (DPP-4) is responsible for the progression of aortic valvular calcification in CAVD animal models. As evogliptin, one of the DPP-4 inhibitors displays high specific accumulation in cardiac tissue, we here evaluated its therapeutic potency for attenuating valvular calcification in CAVD animal models. Evogliptin administration markedly reduced calcific deposition accompanied by a reduction in proinflammatory cytokine expression in endothelial nitric oxide synthase-deficient mice in vivo, and significantly ameliorated the mineralization of the primary human valvular interstitial cells (VICs), with a reduction in the mRNA expression of bone-associated and fibrosis-related genes in vitro. In addition, evogliptin ameliorated the rate of change in the transaortic peak velocity and mean pressure gradients in our rabbit model as assessed by echocardiography. Importantly, evogliptin administration in a rabbit model was found to suppress the effects of a high-cholesterol diet and of vitamin D2-driven fibrosis in association with a reduction in macrophage infiltration and calcific deposition in aortic valves. These results have indicated that evogliptin prohibits inflammatory cytokine expression, fibrosis, and calcification in a CAVD animal model, suggesting its potential as a selective therapeutic agent for the inhibition of valvular calcification during CAVD progression.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Inflammation/drug therapy , Piperazines/therapeutic use , Animals , Aortic Valve/drug effects , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/genetics , Calcinosis/complications , Calcinosis/genetics , Cytokines/metabolism , Disease Models, Animal , Fibrosis , Gene Expression Regulation/drug effects , Humans , Inflammation/complications , Inflammation/genetics , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Piperazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits
12.
Trends Cardiovasc Med ; 31(5): 305-311, 2021 07.
Article in English | MEDLINE | ID: mdl-32525013

ABSTRACT

Elevated levels of lipoprotein(a) [Lp(a)] have been identified as an independent and causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and, more recently, calcific aortic valve disease (CAVD). CAVD is a slow, progressive disorder presenting as severe trileaflet calcification known as aortic valve stenosis (AS) that impairs valve motion and restricts ventricular outflow. AS afflicts 2% of the aging population (≥ 65 years) and tends to be quite advanced by the time it presents clinical symptoms of exertional angina, syncope, or heart failure. Currently, the only effective clinical therapy for AS patients is surgical or transcatheter aortic valve replacement. Evidence is accumulating that Lp(a) can exacerbate pathophysiological processes in CAVD, specifically, endothelial dysfunction, formation of foam cells, and promotion of a pro-inflammatory state. In the valve milieu, the pro-inflammatory effects of Lp(a) are manifested in valve thickening and mineralization through pro-osteogenic signaling and changes in gene expression in valve interstitial cells that is primarily facilitated by the oxidized phospholipid content of Lp(a). In AS pathogenesis, an incomplete understanding of the role of Lp(a) at the molecular level and the absence of appropriate animal models are barriers for the development of specific and effective clinical interventions designed to mitigate the role of Lp(a) in AS. However, the advent of effective therapies that dramatically lower Lp(a) provides the possibility of the first medical treatment to halt AS progression.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Lipoprotein(a)/metabolism , Aged , Animals , Aortic Valve/drug effects , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/genetics , Calcinosis/diagnosis , Calcinosis/drug therapy , Calcinosis/genetics , Disease Models, Animal , Female , Foam Cells/metabolism , Foam Cells/pathology , Humans , Hypolipidemic Agents/therapeutic use , Lipoprotein(a)/genetics , Male , Oligonucleotides/therapeutic use , Oxidation-Reduction , Phospholipids/metabolism
13.
J Thorac Cardiovasc Surg ; 161(4): e261-e271, 2021 04.
Article in English | MEDLINE | ID: mdl-31924353

ABSTRACT

BACKGROUND: Aortic valve interstitial cells have been implicated in the pathogenesis of aortic stenosis. In response to proinflammatory stimuli, aortic valve interstitial cells undergo an osteogenic phenotypic change. The purpose of this study was to determine whether the anti-inflammatory effects of statins prevent osteogenic activity in cultured aortic valve interstitial cells. METHODS: Human aortic valve interstitial cells were isolated from hearts explanted for cardiac transplantation. To test whether simvastatin down-regulates TLR4-induced osteogenic response, aortic valve interstitial cells were treated with simvastatin with and without TLR4 agonist lipopolysaccharide (LPS), and osteogenic markers were measured. Simvastatin's influence on in vitro calcium deposition was assessed by alizarin red staining. Knockdown of postreceptor signaling proteins (MyD88 and TRIF) was performed to determine which of 2 TLR4-associated pathways mediates the osteogenic response. Expression levels of TLR4-induced nuclear factor kappa light chain enhancer of activated B cells (NF-κB) and TLR4 expression were assessed after treatment with simvastatin. Statistical testing was done by analysis of variance (P < .05). RESULTS: Simvastatin decreased LPS-induced ALP and Runx2 expression and inhibited in vitro calcium deposition in aortic valve interstitial cells. Knockdown of MyD88 and TRIF attenuated the osteogenic response. Simvastatin attenuated TLR4-dependent NF-κB signaling and down-regulated TLR4 levels. CONCLUSIONS: Simvastatin prevented TLR4-induced osteogenic phenotypic changes in isolated aortic valve interstitial cells via down-regulation of TLR4 and inhibition of NF-κB signaling. These data offer mechanistic insight into a possible therapeutic role for simvastatin in the prevention of aortic stenosis.


Subject(s)
Aortic Valve/drug effects , Aortic Valve/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Osteogenesis/drug effects , Simvastatin/pharmacology , Adaptor Proteins, Vesicular Transport/physiology , Alkaline Phosphatase/metabolism , Aortic Valve/metabolism , Cell Culture Techniques , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Lipopolysaccharides/physiology , Myeloid Differentiation Factor 88/physiology , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 4/physiology
14.
Cardiovasc Res ; 117(3): 820-835, 2021 02 22.
Article in English | MEDLINE | ID: mdl-32259211

ABSTRACT

AIMS: Calcific aortic valve disease (CAVD) is the most common heart valve disease in the Western world. It has been reported that zinc is accumulated in calcified human aortic valves. However, whether zinc directly regulates CAVD is yet to be elucidated. The present study sought to determine the potential role of zinc in the pathogenesis of CAVD. METHODS AND RESULTS: Using a combination of a human valve interstitial cell (hVIC) calcification model, human aortic valve tissues, and blood samples, we report that 20 µM zinc supplementation attenuates hVIC in vitro calcification, and that this is mediated through inhibition of apoptosis and osteogenic differentiation via the zinc-sensing receptor GPR39-dependent ERK1/2 signalling pathway. Furthermore, we report that GPR39 protein expression is dramatically reduced in calcified human aortic valves, and there is a significant reduction in zinc serum levels in patients with CAVD. Moreover, we reveal that 20 µM zinc treatment prevents the reduction of GPR39 observed in calcified hVICs. We also show that the zinc transporter ZIP13 and ZIP14 are significantly increased in hVICs in response to zinc treatment. Knockdown of ZIP13 or ZIP14 significantly inhibited hVIC in vitro calcification and osteogenic differentiation. CONCLUSIONS: Together, these findings suggest that zinc is a novel inhibitor of CAVD, and report that zinc transporter ZIP13 and ZIP14 are important regulators of hVIC in vitro calcification and osteogenic differentiation. Zinc supplementation may offer a potential therapeutic strategy for CAVD.


Subject(s)
Aortic Valve/drug effects , Calcinosis/drug therapy , Heart Valve Diseases/drug therapy , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, G-Protein-Coupled/metabolism , Zinc Sulfate/pharmacology , Aortic Valve/enzymology , Aortic Valve/pathology , Apoptosis/drug effects , Calcinosis/enzymology , Calcinosis/pathology , Case-Control Studies , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cells, Cultured , Female , Heart Valve Diseases/enzymology , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Humans , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Osteogenesis/drug effects , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Zinc Sulfate/metabolism
15.
Science ; 371(6530)2021 02 12.
Article in English | MEDLINE | ID: mdl-33303684

ABSTRACT

Mapping the gene-regulatory networks dysregulated in human disease would allow the design of network-correcting therapies that treat the core disease mechanism. However, small molecules are traditionally screened for their effects on one to several outputs at most, biasing discovery and limiting the likelihood of true disease-modifying drug candidates. Here, we developed a machine-learning approach to identify small molecules that broadly correct gene networks dysregulated in a human induced pluripotent stem cell (iPSC) disease model of a common form of heart disease involving the aortic valve (AV). Gene network correction by the most efficacious therapeutic candidate, XCT790, generalized to patient-derived primary AV cells and was sufficient to prevent and treat AV disease in vivo in a mouse model. This strategy, made feasible by human iPSC technology, network analysis, and machine learning, may represent an effective path for drug discovery.


Subject(s)
Aortic Valve Disease/drug therapy , Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Gene Regulatory Networks/drug effects , Machine Learning , Nitriles/pharmacology , Nitriles/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , Algorithms , Animals , Aortic Valve/drug effects , Aortic Valve/metabolism , Aortic Valve/physiopathology , Aortic Valve Disease/genetics , Aortic Valve Disease/physiopathology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/physiopathology , Calcinosis/genetics , Calcinosis/physiopathology , Disease Models, Animal , Drug Discovery , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Haploinsufficiency , Humans , Induced Pluripotent Stem Cells , Mice, Inbred C57BL , RNA-Seq , Receptor, Notch1/genetics , Small Molecule Libraries
16.
Arterioscler Thromb Vasc Biol ; 41(1): 11-19, 2021 01.
Article in English | MEDLINE | ID: mdl-33232199

ABSTRACT

Mineralization of cardiovascular structures including blood vessels and heart valves is a common feature. We postulate that ectopic mineralization is a response-to-injury in which signals delivered to cells trigger a chain of events to restore and repair tissues. Maladaptive response to external or internal signals promote the expression of danger-associated molecular patterns, which, in turn, promote, when expressed chronically, a procalcifying gene program. Growing evidence suggest that danger-associated molecular patterns such as oxyphospholipids and small lipid mediators, generated by enzyme activity, are involved in the transition of vascular smooth muscle cells and valve interstitial cells to an osteoblast-like phenotype. Understanding the regulation and the molecular processes underpinning the mineralization of atherosclerotic plaques and cardiac valves are providing valuable mechanistic insights, which could lead to the development of novel therapies. Herein, we provide a focus account on the role oxyphospholipids and their mediators in the development of mineralization in plaques and calcific aortic valve disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Arteries/metabolism , Calcinosis/metabolism , Phospholipids/metabolism , Vascular Calcification/metabolism , Animals , Aortic Valve/drug effects , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/pathology , Arteries/drug effects , Arteries/pathology , Calcinosis/drug therapy , Calcinosis/pathology , Cell Plasticity , Humans , Oxidation-Reduction , Plaque, Atherosclerotic , Signal Transduction , Vascular Calcification/drug therapy , Vascular Calcification/pathology
17.
J Mol Cell Cardiol ; 150: 54-64, 2021 01.
Article in English | MEDLINE | ID: mdl-33045251

ABSTRACT

AIMS: Calcific aortic valve disease (CAVD) is a primary cause of cardiovascular mortality; however, its mechanisms are unknown. Currently, no effective pharmacotherapy is available for CAVD. Aldo-keto reductase family 1 member B (Akr1B1) has been identified as a potential therapeutic target for valve interstitial cell calcification. Herein, we hypothesized that inhibition of Akr1B1 can attenuate aortic valve calcification. METHODS AND RESULTS: Normal and degenerative tricuspid calcific valves from human samples were analyzed by immunoblotting and immunohistochemistry. The results showed significant upregulation of Akr1B1 in CAVD leaflets. Akr1B1 inhibition attenuated calcification of aortic valve interstitial cells in osteogenic medium. In contrast, overexpression of Akr1B1 aggravated calcification in osteogenic medium. Mechanistically, using RNA sequencing (RNAseq), we revealed that Hippo-YAP signaling functions downstream of Akr1B1. Furthermore, we established that the protein level of the Hippo-YAP signaling effector active-YAP had a positive correlation with Akr1B1. Suppression of YAP reversed Akr1B1 overexpression-induced Runx2 upregulation. Moreover, YAP activated the Runx2 promoter through TEAD1 in a manner mediated by ChIP and luciferase reporter systems. Animal experiments showed that the Akr1B1 inhibitor epalrestat attenuated aortic valve calcification induced by a Western diet in LDLR-/- mice. CONCLUSION: This study demonstrates that inhibition of Akr1B1 can attenuate the degree of calcification both in vitro and in vivo. The Akr1B1 inhibitor epalrestat may be a potential treatment option for CAVD.


Subject(s)
Aldehyde Reductase/metabolism , Aldo-Keto Reductases/metabolism , Aortic Valve Stenosis/enzymology , Aortic Valve Stenosis/pathology , Aortic Valve/enzymology , Aortic Valve/pathology , Calcinosis/enzymology , Calcinosis/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Aldehyde Reductase/antagonists & inhibitors , Animals , Aortic Valve/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Humans , Lentivirus/metabolism , Mice , Osteogenesis/drug effects , Signal Transduction/drug effects , Transcription Factors/metabolism , YAP-Signaling Proteins
18.
Arterioscler Thromb Vasc Biol ; 41(1): 117-127, 2021 01.
Article in English | MEDLINE | ID: mdl-33115271

ABSTRACT

Cardiovascular calcification is an insidious form of ectopic tissue mineralization that presents as a frequent comorbidity of atherosclerosis, aortic valve stenosis, diabetes, renal failure, and chronic inflammation. Calcification of the vasculature and heart valves contributes to mortality in these diseases. An inability to clinically image or detect early microcalcification coupled with an utter lack of pharmaceutical therapies capable of inhibiting or regressing entrenched and detectable macrocalcification has led to a prominent and deadly gap in care for a growing portion of our rapidly aging population. Recognition of this mounting concern has arisen over the past decade and led to a series of revolutionary works that has begun to pull back the curtain on the pathogenesis, mechanistic basis, and causative drivers of cardiovascular calcification. Central to this progress is the discovery that calcifying extracellular vesicles act as active precursors of cardiovascular microcalcification in diverse vascular beds. More recently, the omics revolution has resulted in the collection and quantification of vast amounts of molecular-level data. As the field has become poised to leverage these resources for drug discovery, new means of deriving relevant biological insights from these rich and complex datasets have come into focus through the careful application of systems biology and network medicine approaches. As we look onward toward the next decade, we envision a growing need to standardize approaches to study this complex and multifaceted clinical problem and expect that a push to translate mechanistic findings into therapeutics will begin to finally provide relief for those impacted by this disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Arteries/metabolism , Calcinosis/metabolism , Extracellular Vesicles/metabolism , Osteogenesis , Vascular Calcification/metabolism , Animals , Aortic Valve/drug effects , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Arteries/drug effects , Arteries/pathology , Calcinosis/drug therapy , Calcinosis/genetics , Calcinosis/pathology , Drug Development , Drug Discovery , Extracellular Vesicles/pathology , Gene Expression Regulation , Genomics , Humans , Osteogenesis/drug effects , Signal Transduction , Vascular Calcification/drug therapy , Vascular Calcification/genetics , Vascular Calcification/pathology
19.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255450

ABSTRACT

Calcific aortic valve disease (CAVD) is the result of maladaptive fibrocalcific processes leading to a progressive thickening and stiffening of aortic valve (AV) leaflets. CAVD is the most common cause of aortic stenosis (AS). At present, there is no effective pharmacotherapy in reducing CAVD progression; when CAVD becomes symptomatic it can only be treated with valve replacement. Inflammation has a key role in AV pathological remodeling; hence, anti-inflammatory therapy has been proposed as a strategy to prevent CAVD. Cyclooxygenase 2 (COX-2) is a key mediator of the inflammation and it is the target of widely used anti-inflammatory drugs. COX-2-inhibitor celecoxib was initially shown to reduce AV calcification in a murine model. However, in contrast to these findings, a recent retrospective clinical analysis found an association between AS and celecoxib use. In the present study, we investigated whether variations in COX-2 expression levels in human AVs may be linked to CAVD. We extracted total RNA from surgically explanted AVs from patients without CAVD or with CAVD. We found that COX-2 mRNA was higher in non-calcific AVs compared to calcific AVs (0.013 ± 0.002 vs. 0.006 ± 0.0004; p < 0.0001). Moreover, we isolated human aortic valve interstitial cells (AVICs) from AVs and found that COX-2 expression is decreased in AVICs from calcific valves compared to AVICs from non-calcific AVs. Furthermore, we observed that COX-2 inhibition with celecoxib induces AVICs trans-differentiation towards a myofibroblast phenotype, and increases the levels of TGF-ß-induced apoptosis, both processes able to promote the formation of calcific nodules. We conclude that reduced COX-2 expression is a characteristic of human AVICs prone to calcification and that COX-2 inhibition may promote aortic valve calcification. Our findings support the notion that celecoxib may facilitate CAVD progression.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Cyclooxygenase 2/genetics , Inflammation/drug therapy , Transforming Growth Factor beta/genetics , Aged , Aged, 80 and over , Animals , Aortic Valve/drug effects , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Apoptosis/drug effects , Calcinosis/genetics , Calcinosis/pathology , Celecoxib/administration & dosage , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Male , Mice , Middle Aged , RNA, Messenger/genetics
20.
Oxid Med Cell Longev ; 2020: 2043762, 2020.
Article in English | MEDLINE | ID: mdl-33149806

ABSTRACT

Warfarin, a vitamin K antagonist (VKA), is known to promote arterial calcification (AC). In the present study, we conducted a case-cohort study within the Multi-Ethnic Study of Atherosclerosis (MESA); 6655 participants were included. From MESA data, we found that AC was related to both age and vitamin K; furthermore, the score of AC increased with SASP marker including interlukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) rising. Next, a total of 79 warfarin users in our center developed significantly more calcified coronary plaques as compared to non-VKA users. We investigated the role of warfarin in phosphate-induced AC in different ages by in vitro experimental study. Furthermore, dose-time-response of warfarin was positively correlated with AC score distribution and plasma levels of the SASP maker IL-6 among patients < 65 years, but not among patients ≥ 65 years. In addition, in vitro research suggested that warfarin treatment tended to deteriorate calcification in young VSMC at the early stage of calcification. Our results suggested that aging and warfarin-treatment were independently related to increased AC. Younger patients were more sensitive to warfarin-related AC than older patients, which was possibly due to accumulated warfarin-induced cellular senescence.


Subject(s)
Aortic Valve/pathology , Biomarkers/metabolism , Cellular Senescence/drug effects , Vascular Calcification/pathology , Warfarin/pharmacology , Abdomen/pathology , Aged , Animals , Aortic Valve/diagnostic imaging , Aortic Valve/drug effects , Atrial Fibrillation/diagnostic imaging , Atrial Fibrillation/pathology , Dose-Response Relationship, Drug , Electrocardiography , Factor Analysis, Statistical , Female , Humans , Interleukin-6/metabolism , Logistic Models , Male , Middle Aged , Multivariate Analysis , Phosphates , Rats, Sprague-Dawley , Risk Factors , Time Factors , Up-Regulation/drug effects , Vascular Calcification/diagnostic imaging , Vitamin K/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...