Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 319
Filter
1.
Int J Cardiol ; 352: 104-114, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35074490

ABSTRACT

OBJECTIVE: To explore the mechanism of CircANKRD36 regulating cell heterogeneity and endothelial mesenchymal transition in aortic valve stromal cells by regulating miR-599 and TGF-ß signaling pathway. METHODS: Human tissue specimens were divided into Control group (n = 25) and CAVD group (n = 25). The mRNA expressions of CircANKRD36 and miR-599 in tissue samples were analyzed by qRT-PCR. Western blot was used to analyze the protein expression of osteogenic differentiation related factors induced by OM.The expressions of ALP, osteocalcin, osteopontin, Runx2 and Cadherin11 were detected by Western blot. RESULTS: The expression of CircANKRD36mRNA in CAVD tissue was lower than that in Control tissue (P < 0.05), and the expression of miR-599mRNA in CAVD tissue was higher than that in Control tissue (P < 0.05). CircANKRD36 was negatively correlated with ALP, osteocalcin, osteopontin, Runx2, Cadherin11 expression level after OM induced osteogenic differentiation. The expression level of miR-599 was positively correlated with ALP, osteocalcin, osteopontin, Runx2 and Cadherin11 after OM induced osteogenic differentiation.The expression of ALP, osteocalcin, osteopontin, Runx2 and Cadherin11 protein in circ+miR-599 group was lower than that in circ+miR-NC group (P < 0.05). Compared with Vector+miR-NC group, the protein expressions of TGF-ß1, TGF-ß2 and SMAD4 in circ+miR-NC group decreased (P < 0.05), while the protein expressions of TGF-ß1, TGF-ß2 and SMAD4 in circ+miR-599 group increased (P < 0.05). CONCLUSION: CircANKRD36 can inhibit the expression of miR-599 and the activation of TGF-ß signaling pathway, thus inhibiting the expression of differentiation-related factors of VIC osteogenesis and the formation of calcified nodules. Therefore, circANKRD36-miR-599-TGF-ß axis can be a new theoretical basis for treating CAVD.


Subject(s)
MicroRNAs , Nuclear Proteins , Osteogenesis , Stromal Cells , Aortic Diseases , Aortic Valve/cytology , Aortic Valve/metabolism , Calcinosis , Cell Differentiation , Cells, Cultured , Humans , Nuclear Proteins/genetics , Osteogenesis/genetics , RNA, Circular , Signal Transduction/genetics , Stromal Cells/cytology , Stromal Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
2.
Circulation ; 145(7): 513-530, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35000411

ABSTRACT

BACKGROUND: Aortic valve stenosis is a sexually dimorphic disease, with women often presenting with sustained fibrosis and men with more extensive calcification. However, the intracellular molecular mechanisms that drive these clinically important sex differences remain underexplored. METHODS: Hydrogel biomaterials were designed to recapitulate key aspects of the valve tissue microenvironment and to serve as a culture platform for sex-specific valvular interstitial cells (VICs; precursors to profibrotic myofibroblasts). The hydrogel culture system was used to interrogate intracellular pathways involved in sex-dependent VIC-to-myofibroblast activation and deactivation. RNA sequencing was used to define pathways involved in driving sex-dependent activation. Interventions with small molecule inhibitors and siRNA transfections were performed to provide mechanistic insight into sex-specific cellular responses to microenvironmental cues, including matrix stiffness and exogenously delivered biochemical factors. RESULTS: In both healthy porcine and human aortic valves, female leaflets had higher baseline activation of the myofibroblast marker α-smooth muscle actin compared with male leaflets. When isolated and cultured, female porcine and human VICs had higher levels of basal α-smooth muscle actin stress fibers that further increased in response to the hydrogel matrix stiffness, both of which were higher than in male VICs. A transcriptomic analysis of male and female porcine VICs revealed Rho-associated protein kinase signaling as a potential driver of this sex-dependent myofibroblast activation. Furthermore, we found that genes that escape X-chromosome inactivation such as BMX and STS (encoding for Bmx nonreceptor tyrosine kinase and steroid sulfatase, respectively) partially regulate the elevated female myofibroblast activation through Rho-associated protein kinase signaling. This finding was confirmed by treating male and female VICs with endothelin-1 and plasminogen activator inhibitor-1, factors that are secreted by endothelial cells and known to drive myofibroblast activation through Rho-associated protein kinase signaling. CONCLUSIONS: Together, in vivo and in vitro results confirm sex dependencies in myofibroblast activation pathways and implicate genes that escape X-chromosome inactivation in regulating sex differences in myofibroblast activation and subsequent aortic valve stenosis progression. Our results underscore the importance of considering sex as a biological variable to understand the molecular mechanisms of aortic valve stenosis and to help guide sex-based precision therapies.


Subject(s)
Aortic Valve/cytology , Gene Expression , Genes, X-Linked , Myofibroblasts/metabolism , X Chromosome Inactivation , Actins/genetics , Actins/metabolism , Animals , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Biomarkers , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Male , Myofibroblasts/drug effects , Sex Factors , Signal Transduction , Swine , Transcriptome
3.
Mol Med ; 27(1): 156, 2021 12 11.
Article in English | MEDLINE | ID: mdl-34895136

ABSTRACT

BACKGROUND: Calcific aortic valve disease (CAVD) is the most prevalent valvular disease worldwide. However, no effective treatment could delay or prevent the progression of the disease due to the poor understanding of its pathological mechanism. Many studies showed that metformin exerted beneficial effects on multiple cardiovascular diseases by mediating multiple proteins such as AMPK, NF-κB, and AKT. This study aims to verify whether metformin can inhibit aortic calcification through the PI3K/AKT signaling pathway. METHODS: We first analyzed four microarray datasets to screen differentially expressed genes (DEGs) and signaling pathways related to CAVD. Then aortic valve samples were used to verify selected genes and pathways through immunohistochemistry (IHC) and western blot (WB) assays. Aortic valve interstitial cells (AVICs) were isolated from non-calcific aortic valves and then cultured with phosphate medium (PM) with or without metformin to verify whether metformin can inhibit the osteogenic differentiation and calcification of AVICs. Finally, we used inhibitors and siRNA targeting AMPK, NF-κB, and AKT to study the mechanism of metformin. RESULTS: We screened 227 DEGs; NF-κB and PI3K/AKT signaling pathways were implicated in the pathological mechanism of CAVD. IHC and WB experiments showed decreased AMPK and AKT and increased Bax in calcific aortic valves. PM treatment significantly reduced AMPK and PI3K/AKT signaling pathways, promoted Bax/Bcl2 ratio, and induced AVICs calcification. Metformin treatment ameliorated AVICs calcification and apoptosis by activating the PI3K/AKT signaling pathway. AMPK activation and NF-κB inhibition could inhibit AVICs calcification induced by PM treatment; however, AMPK and AKT inhibition reversed the protective effect of metformin. CONCLUSIONS: This study, for the first time, demonstrates that metformin can inhibit AVICs in vitro calcification by activating the PI3K/AKT signaling pathway; this suggests that metformin may provide a potential target for the treatment of CAVD. And the PI3K/AKT signaling pathway emerges as an important regulatory axis in the pathological mechanism of CAVD.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , AMP-Activated Protein Kinases/metabolism , Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Calcinosis/genetics , Calcinosis/metabolism , Cells, Cultured , Humans , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
4.
Food Funct ; 12(23): 11808-11818, 2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34766179

ABSTRACT

Cardamonin (CDM) is a natural chalcone with strong anti-inflammatory properties. Inflammation-induced osteogenic changes in valve interstitial cells (VICs) play crucial roles in the development of calcific aortic valve disease (CAVD), a degenerative disease characterized by degeneration, thickening, fibrosis, and calcification of the heart valve tissues. To investigate the anti-osteogenic differentiation role of CDM in human valve interstitial cells (hVICs), which consequently reverses the calcification of the aortic valve, human VICs were exposed to osteogenic induction medium (OM) with CDM for further cell viability, osteogenic gene and protein expression analyses, and anti-calcification testing. mRNA sequencing was utilized to analyze the differentially expressed genes (DEGs) and related signaling pathways as potential molecular targets involved in CDM's anti-calcification activity. Human aortic valve leaflet ex vivo calcific cultures were used to investigate the CDM inhibition of osteogenic differentiation of hVICs at the tissue level. ApoE-/- mice fed with a high-fat (HF) diet were used to evaluate the effect of CDM on aortic valve calcification. No significant CDM cytotoxicity was seen in the hVICs at 10 µM. The addition of CDM to OM prevented calcified nodule accumulation, and a decrease in the gene/protein expression levels of BMP2, RUNX2, SPP1, TNF-α, and COL1A2 was observed. Venn diagram analysis of the DEGs identified 666 common DEGs and highlighted the NOD-like receptor signaling pathway (ko04621) as an anti-calcification target of CDM. CDM also repressed the activation of p-AKT, p-ERK1/2, and p-IκBα, and prevented the OM-induced nuclear transcription of NF-κB p65. In the in vitro and ex vivo calcific conditional culture experiments, CDM exhibited anti-inflammatory and anti-calcification effects by suppressing the activation of the NLRP3 inflammasome and downregulating IL-1ß expression. In vivo, CDM ameliorated aortic valve calcification by interfering with NLRP3 expression. Our study demonstrated that CDM inhibited the phenotypical calcific transformation of hVICs by mediating the inactivation of the NF-κB/NLRP3 inflammasome. Therefore, it is considered to be a promising natural compound for use in preventing the progression of heart valve calcification disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Cell Differentiation/drug effects , Chalcones/pharmacology , Inflammasomes/drug effects , Osteogenesis/drug effects , Animals , Aortic Valve/cytology , Aortic Valve/metabolism , Cells, Cultured , Humans , Inflammasomes/metabolism , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
5.
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
6.
Exp Biol Med (Maywood) ; 246(21): 2278-2289, 2021 11.
Article in English | MEDLINE | ID: mdl-34260291

ABSTRACT

Valvular endothelial cells line the outer layer of heart valves and can withstand shear forces caused by blood flow. In contrast to vascular endothelial cells, there is limited amount of research over valvular endothelial cells. For this reason, the exact physiologic behavior of valvular endothelial cells is unclear. Prior studies have concluded that valvular endothelial cells align perpendicularly to the direction of blood flow, while vascular endothelial cells align parallel to blood flow. Other studies have suggested that different ranges of shear stress uniquely impact the behavior of valvular endothelial cells. The goal of this study was to characterize the response of valvular endothelial cell under different types, magnitudes, and durations of shear stress. In this work, the results demonstrated that with increased shear rate and duration of exposure, valvular endothelial cells no longer possessed the traditional cuboidal morphology. Instead through the change in cell circularity and aspect ratio, valvular endothelial cells aligned in an organized manner. In addition, different forms of shear exposure caused the area and circularity of valvular endothelial cells to decrease while inducing mesenchymal transformation validated through αSMA and TGFß1 expression. This is the first investigation showing that valvular endothelial cells alignment is not as straightforward as once thought (perpendicular to flow). Different types and magnitudes of shear induce different local behaviors. This is also the first demonstration of valvular endothelial cells undergoing EndMT without chemical inducers on a soft surface in vitro. Findings from this study provide insights to understanding the pathophysiology of valvular endothelial cells which can potentially propel future artificial engineered heart valves.


Subject(s)
Aortic Valve/cytology , Cell Differentiation/physiology , Endothelial Cells/cytology , Shear Strength/physiology , Animals , Aortic Valve/anatomy & histology , Aortic Valve/physiology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Fluorescent Antibody Technique , Swine
7.
Circ Res ; 128(9): 1344-1370, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33914601

ABSTRACT

Calcific aortic valve disease is dramatically increasing in global burden, yet no therapy exists outside of prosthetic replacement. The increasing proportion of younger and more active patients mandates alternative therapies. Studies suggest a window of opportunity for biologically based diagnostics and therapeutics to alleviate or delay calcific aortic valve disease progression. Advancement, however, has been hampered by limited understanding of the complex mechanisms driving calcific aortic valve disease initiation and progression towards clinically relevant interventions.


Subject(s)
Aortic Valve Stenosis/etiology , Aortic Valve/cytology , Aortic Valve/pathology , Calcinosis/etiology , Disease Progression , Endothelial Cells/physiology , Aortic Valve/immunology , Aortic Valve/physiology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/therapy , Calcinosis/diagnosis , Calcinosis/immunology , Calcinosis/therapy , Cell Adhesion Molecules/metabolism , Homeostasis , Humans , Immune System/physiology , Inflammation Mediators/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Prognosis , Reactive Oxygen Species , Risk Factors , Vasculitis/etiology
8.
J Vis Exp ; (168)2021 02 04.
Article in English | MEDLINE | ID: mdl-33616094

ABSTRACT

Calcific aortic valve disease (CAVD), an active disease process ranging from mild thickening of the valve to severe calcification, is associated with high mortality, despite new therapeutic options such as transcatheter aortic valve replacement (TAVR). The complete pathways that start with valve calcification and lead to severe aortic stenosis remain only partly understood. By providing a close representation of the aortic valve cells in vivo, the assaying of T lymphocytes from stenotic valve tissue could be an efficient way to clarify their role in the development of calcification. After surgical excision, the fresh aortic valve sample is dissected in small pieces and the T lymphocytes are cultured, cloned then analyzed using fluorescence activated cell sorting (FACS). The staining procedure is simple and the stained tubes can also be fixed using 0.5% of paraformaldehyde and analyzed up to 15 days later. The results generated from the staining panel can be used to track changes in T cell concentrations over time in relation to intervention and could easily be further developed to assess activation states of specific T cell subtypes of interest. In this study, we show the isolation of T cells, performed on fresh calcified aortic valve samples and the steps of analyzing T cell clones using flow cytometry to further understand the role of adaptive immunity in CAVD pathophysiology.


Subject(s)
Aortic Valve Stenosis/pathology , Aortic Valve/cytology , Aortic Valve/pathology , Blood Buffy Coat/radiation effects , Calcinosis/pathology , Cell Separation/methods , Feeder Cells/cytology , Flow Cytometry/methods , T-Lymphocytes/cytology , Aortic Valve/metabolism , Cells, Cultured , Feeder Cells/metabolism , Humans , T-Lymphocytes/metabolism
9.
J Pharmacol Sci ; 145(2): 213-221, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33451756

ABSTRACT

Since aortic valve stenosis (AVS) is the most frequent and serious valvular heart disease in the elderly, and is accompanied by irreversible valve calcification, medicinal prevention of AVS is important. Although we recently demonstrated that human aortic valve interstitial cells (HAVICs) obtained from patients with AVS were highly sensitive to ectopic calcification stimulation, the cell types contributing to calcification are unknown. We aimed to immunocytochemically characterize HAVICs and identify their contribution to valve calcification. HAVICs were isolated from patients with AVS and cultured on non-coated dishes. Immunocytochemical features and HAVIC differentiation were analyzed in passage 1 (P1). The immunohistochemical features of the calcified aortic valve were analyzed. Most cultured P1 HAVICs were CD73-, CD90-, and CD105-positive, and CD45-and CD34-negative. HAVICs were vascular endothelial growth factor receptor 2 (VEGFR2)-positive; however, approximately half were α-smooth muscle actin (SMA)-positive, colonized, and easily differentiated into osteoblastic cells. Calcified aortic valve immunohistochemistry showed that all cells were positive for VEGFR2 and partly α-SMA. Further, VEGFR2-positive cells were more sensitive to tumor necrosis factor-α-induced ectopic calcification with or without α-SMA positivity. We conclude that HAVICs obtained from patients with AVS are VEGFR2-positive undifferentiated mesenchymal cells and may contribute to aortic valve ectopic calcification.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Calcinosis/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Actins/metabolism , Aged , Aortic Valve Stenosis/etiology , Calcinosis/etiology , Cells, Cultured , Female , Humans , Male , Tumor Necrosis Factor-alpha/metabolism
10.
Eur J Clin Invest ; 51(5): e13470, 2021 May.
Article in English | MEDLINE | ID: mdl-33296074

ABSTRACT

BACKGROUND: Calcific aortic valve disease is associated with ageing and high mortality. However, no effective pharmacological treatment has been developed. Vascular endothelial growth factor (VEGF) and its receptor are overexpressed in the calcified aortic valve tissue. However, the role of VEGF in calcific aortic valve disease pathogenesis and its underlying mechanisms remain unclear. MATERIALS AND METHODS: Runt-related transcription factor 2 expression and calcium-related signalling were investigated in porcine valvular interstitial cells with or without human VEGF-A recombinant protein (VEGF165 , 1-100 ng/mL) treatment and/or calmodulin-dependent kinase II (CaMKII) inhibitor (KN93, 10 µmol/L) and inositol triphosphate receptor inhibitor (2-aminoethyldiphenyl borate, 30 µmol/L) for 5 days. RESULTS: VEGF165 -treated cells had higher Runt-related transcription factor 2 expression and CaMKII/ adenosine 3',5'-monophosphate response element-binding protein (CREB) signalling activation than did control cells. KN93 reduced Runt-related transcription factor 2 expression and CREB phosphorylation in VEGF165 -treated cells. The 2-aminoethyldiphenyl borate also reduced Runt-related transcription factor 2 expression in VICs treated with VEGF165 . CONCLUSION: VEGF upregulated Runt-related transcription factor 2 expression in VICs by activating the IP3R/CaMKII/CREB signalling pathway.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/cytology , Aortic Valve/pathology , Calcinosis/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aortic Valve/metabolism , Benzylamines/pharmacology , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Core Binding Factor Alpha 1 Subunit/drug effects , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Swine , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
11.
J Thorac Cardiovasc Surg ; 161(2): e163-e185, 2021 02.
Article in English | MEDLINE | ID: mdl-31761339

ABSTRACT

OBJECTIVE: Human aortic valve interstitial cells redifferentiate into an osteoblast-like phenotype, which is the key cellular mechanism of aortic valve calcification. Methyltransferase-like 3, the N6-methyladenosine methylation writer, has emerged as a new layer of epigenetic regulation for osteogenic differentiation of bone mesenchymal stem cells. The current study sought to determine whether methyltransferase-like 3 also plays a role in the osteogenic differentiation of human aortic valve interstitial cells. METHODS: Aortic valves from patients with aortic stenosis (n = 50) and normal controls (n = 50) were subjected to determination of methyltransferase-like 3 expression. Mineralized bone matrix formation was assessed by Alizarin Red staining. The interaction between methyltransferase-like 3 and twist-related protein 1 was confirmed via luciferase reporter and N6-methyladenosine methylated RNA immunoprecipitation quantitative reverse-transcription polymerase chain reaction. RESULTS: Methyltransferase-like 3 was highly expressed in human calcified aortic valves (1.61 ± 0.50) versus normal valves (3.07 ± 0.62; P < .0001). Osteogenic stimulation for 7 days resulted in a 2.15 ± 0.16-fold increase (P < .0001) in methyltransferase-like 3 protein level compared with the control group in human aortic valve interstitial cells. Functionally, methyltransferase-like 3 acted as a positive regulator of osteogenic differentiation of human aortic valve interstitial cells. Mechanistically, methylated RNA immunoprecipitation quantitative reverse-transcription polymerase chain reaction identified twist-related protein 1 as a target of methyltransferase-like 3-mediated m6A modification. Moreover, N6-methyladenosine-mediated twist-related protein 1 mRNA inhibition relied on the m6A binding protein YTH-domain family member 2-dependent pathway. CONCLUSIONS: Methyltransferase-like 3 promotes osteogenic differentiation of human aortic valve interstitial cells by inhibiting twist-related protein 1 through an N6-methyladenosine YTH-domain family member 2-dependent pathway. Our findings provide novel mechanistic insights into a critical role of methyltransferase-like 3 in the aortic valve calcification progression and shed new light on N6-methyladenosine-directed diagnostics and therapeutics in aortic valve calcification.


Subject(s)
Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve Stenosis/metabolism , Calcinosis/metabolism , Cell Differentiation , Female , Humans , Immunoprecipitation , Male , Methyltransferases/metabolism , Middle Aged , Osteoblasts/metabolism , Osteoblasts/physiology , Reverse Transcriptase Polymerase Chain Reaction , Twist-Related Protein 1/metabolism
12.
J Bone Miner Metab ; 39(3): 360-371, 2021 May.
Article in English | MEDLINE | ID: mdl-33070258

ABSTRACT

INTRODUCTION: Calcified aortic valve disease (CAVD) is characterized by valve thickening and calcification. Osteoblast differentiation is one of the key steps of valve calcification. CircRNAs is involved in osteogenic differentiation of multiple mesenchymal cells. However, the function of circRNA TGFBR2 (TGFBR2) in CAVD remained unclear. We explored the effect and mechanism of TGFBR2 in modulating CAVD. MATERIALS AND METHODS: Human aortic valve interstitial cells (VICs) were subjected to osteogenic induction, and transfected with TGFBR2, miR-25-3p mimic and siTWIST1. The relationship between miR-25-3p and GFBR2 was predicted by starBase and confirmed by luciferase reporter and Person's correlation test. The relationship between miR-25-3p and TWIST1 was predicted by TargetScan and confirmed by luciferase reporter assay. The expressions of TGFBR2, miR-25-3p, TWIST1, osteoblast markers (RUNX2 and OPN) were detected by Western blot or/and qRT-PCR. Alkaline phosphatase (ALP) activity and calcium nodule was determined by colorimetric method and Alizarin Red S staining. RESULTS: The expression of TGFBR2 was down-regulated and that of miR-25-3p was up-regulated in calcific valves and osteogenic VICs. TGFBR2 was inversely correlated with miR-25-3p expression in calcific valves. TGFBR2 sponged miR-25-3p to regulate TWIST1 expression in osteogenic VICs. During osteogenic differentiation, ALP activity, calcium nodule, the levels of osteoblast markers were increased in VICs. MiR-25-3p overexpression or TWIST1 knockdown reversed the inhibitory effect of TGFBR2 overexpression on ALP activity, calcium nodule, the expressions of RUNX2 and OPN in osteogenic VICs. CONCLUSION: The findings indicated that TGFBR2/miR-25-3p/TWIST1 axis regulates osteoblast differentiation in VICs, supporting the fact that TGFBR2 is a miRNA sponge in CAVD.


Subject(s)
Aortic Valve/cytology , Cell Differentiation/genetics , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , RNA, Circular/metabolism , Signal Transduction , Twist-Related Protein 1/metabolism , Aortic Valve/pathology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Base Sequence , Binding Sites , Calcinosis/genetics , Calcinosis/pathology , Cells, Cultured , Down-Regulation/genetics , Gene Knockdown Techniques , Humans , MicroRNAs/genetics , Osteogenesis/genetics , RNA, Circular/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/genetics
13.
Int J Med Sci ; 17(18): 3065-3072, 2020.
Article in English | MEDLINE | ID: mdl-33173427

ABSTRACT

Objectives: This study amied to whether IL-21 promotes osteoblast transdifferentiation of cultured human Valvular interstitial cells (VICs). Methods: We first confirmed that IL-21 alters gene expression between CAVD aortic valve tissue and normal samples by immunohistochemistry, qPCR, and western blotting. VICs were cultured and treated with IL-21. Gene and protein expression levels of the osteoblastic markers ALP and Runx2, which can be blocked by specific JAK3 inhibitors and/or siRNA of STAT3, were measured. Results: IL-21 expression was upregulated in calcified aortic valves and promotes osteogenic differentiation of human VICs. IL-21 accelerated VIC calcification through the JAK3/STAT3 pathway. Conclusion: Our data suggest that IL-21 is a key factor in valve calcification and a promising candidate for targeted therapeutics for CAVD.


Subject(s)
Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Calcinosis/pathology , Interleukins/metabolism , Osteoblasts/pathology , Adult , Aortic Valve/cytology , Case-Control Studies , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , Cells, Cultured , Female , Gene Knockdown Techniques , Healthy Volunteers , Humans , Interleukin-21 Receptor alpha Subunit/genetics , Interleukin-21 Receptor alpha Subunit/metabolism , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Male , Middle Aged , Primary Cell Culture , Quinazolines/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation
14.
Int J Biol Sci ; 16(15): 3062-3074, 2020.
Article in English | MEDLINE | ID: mdl-33061818

ABSTRACT

Background and Objectives: Chronic valvular inflammation associated with monocyte infiltration promotes calcific aortic valve disease (CAVD) progression. Further, innate immunity in aortic valve interstitial cells (AVICs), mediated by Toll-like receptors (TLRs), up-regulates cellular inflammatory, fibrogenic and osteogenic activities. Currently, the pro-inflammatory communication between monocytes and AVICs and the underlying mechanism are unclear. We hypothesized that monocytes up-regulate AVIC inflammatory activity. This study sought to characterize the interaction between monocytes and AVICs and to elucidate the mechanism underlying cell-to-cell communication. Methods and Results: AVICs, monocytes and co-cultures were exposed to a low concentration of TLR2 activator Pam3CSK4 (0.03 µg/ml). The TLR2 activator at this dose induced a marked increase in AVIC production of ICAM-1 and VCAM-1 only when co-cultured with monocytes. Adding conditioned medium from Pam3CSK4-treated monocytes (Pam3 CM, containing 0.1 µg/ml of Pam3CSK4) to AVIC culture (30% vol/vol; diluting Pam3CSK4 to 0.03 µg/ml) greatly increased the expression of adhesion molecules while adding conditioned medium from untreated monocytes (control CM) had no effect. Inhibition or knockdown of TLR2 in AVICs markedly reduced ICAM-1 and VCAM-1 expression induced by Pam3 CM. Further, Pam3 CM increased TLR2 levels in AVICs. Multiplex-ELISA analysis of Pam3 CM identified greater levels of TNF-α. Neutralization of TNF-α abolished the effect of Pam3 CM on AVIC TLR2 levels, resulting in marked attenuation of its potency in the induction of adhesion molecule expression. Conclusions: This study demonstrates that activated monocytes use paracrine signaling to sensitize AVICs for inflammatory responses to a low level of TLR2 activator. The mechanism of sensitization involves up-regulation of AVIC TLR2 levels by TNF-α from monocytes. Infiltrated monocytes in aortic valve tissue may exacerbate valvular inflammation by rendering AVICs hypersensitive to TLR2 activators.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Monocytes , Toll-Like Receptor 2 , Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Cells, Cultured , Humans , Monocytes/cytology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Up-Regulation
15.
PLoS One ; 15(10): e0240532, 2020.
Article in English | MEDLINE | ID: mdl-33057457

ABSTRACT

BACKGROUND: The ability of heart valve cells to respond to their mechanical environment represents a key mechanism by which the integrity and function of valve cusps is maintained. A number of different mechanotransduction pathways have been implicated in the response of valve cells to mechanical stimulation. In this study, we explore the expression pattern of several mechanosensitive ion channels (MSC) and their potential to mediate mechanosensitive responses of human valve interstitial cells (VIC). METHODS: MSC presence and function were probed using the patch clamp technique. Protein abundance of key MSC was evaluated by Western blotting in isolated fibroblastic VIC (VICFB) and in VIC differentiated towards myofibroblastic (VICMB) or osteoblastic (VICOB) phenotypes. Expression was compared in non-calcified and calcified human aortic valves. MSC contributions to stretch-induced collagen gene expression and to VIC migration were assessed by pharmacological inhibition of specific channels. RESULTS: Two MSC types were recorded in VICFB: potassium selective and cation non-selective channels. In keeping with functional data, the presence of both TREK-1 and Kir6.1 (potassium selective), as well as TRPM4, TRPV4 and TRPC6 (cationic non-selective) channels was confirmed in VIC at the protein level. Differentiation of VICFB into VICMB or VICOB phenotypes was associated with a lower expression of TREK-1 and Kir6.1, and a higher expression of TRPV4 and TRPC6. Differences in MSC expression were also seen in non-calcified vs calcified aortic valves where TREK-1, TRPM4 and TRPV4 expression were higher in calcified compared to control tissues. Cyclic stretch-induced expression of COL I mRNA in cultured VICFB was blocked by RN-9893, a selective inhibitor of TRPV4 channels while having no effect on the stretch-induced expression of COL III. VICFB migration was blocked with the non-specific MSC blocker streptomycin and by GSK417651A an inhibitor of TRPC6/3. CONCLUSION: Aortic VIC express a range of MSC that play a role in functional responses of these cells to mechanical stimulation. MSC expression levels differ in calcified and non-calcified valves in ways that are in part compatible with the change in expression seen between VIC phenotypes. These changes in MSC expression, and associated alterations in the ability of VIC to respond to their mechanical environment, may form novel targets for intervention during aortic valvulopathies.


Subject(s)
Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Calcinosis/pathology , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Myofibroblasts/metabolism , Osteoblasts/metabolism , Aortic Valve/cytology , Aortic Valve Stenosis/drug therapy , Calcinosis/drug therapy , Cell Differentiation , Cells, Cultured , Humans , Ion Channels/antagonists & inhibitors , Mechanotransduction, Cellular/drug effects , Myofibroblasts/drug effects , Osteoblasts/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Primary Cell Culture , Streptomycin/pharmacology , Streptomycin/therapeutic use
16.
Int J Mol Sci ; 21(17)2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32878299

ABSTRACT

Aortic valve interstitial cells (VICs) constitute a heterogeneous population involved in the maintenance of unique valvular architecture, ensuring proper hemodynamic function but also engaged in valve degeneration. Recently, cells similar to telocytes/interstitial Cajal-like cells described in various organs were found in heart valves. The aim of this study was to examine the density, distribution, and spatial organization of a VIC subset co-expressing CD34 and PDGFRα in normal aortic valves and to investigate if these cells are associated with the occurrence of early signs of valve calcific remodeling. We examined 28 human aortic valves obtained upon autopsy. General valve morphology and the early signs of degeneration were assessed histochemically. The studied VICs were identified by immunofluorescence (CD34, PDGFRα, vimentin), and their number in standardized parts and layers of the valves was evaluated. In order to show the complex three-dimensional structure of CD34+/PDGFRα+ VICs, whole-mount specimens were imaged by confocal microscopy, and subsequently rendered using the Imaris (Bitplane AG, Zürich, Switzerland) software. CD34+/PDGFRα+ VICs were found in all examined valves, showing significant differences in the number, distribution within valve tissue, spatial organization, and morphology (spherical/oval without projections; numerous short projections; long, branching, occasionally moniliform projections). Such a complex morphology was associated with the younger age of the subjects, and these VICs were more frequent in the spongiosa layer of the valve. Both the number and percentage of CD34+/PDGFRα+ VICs were inversely correlated with the age of the subjects. Valves with histochemical signs of early calcification contained a lower number of CD34+/PDGFRα+ cells. They were less numerous in proximal parts of the cusps, i.e., areas prone to calcification. The results suggest that normal aortic valves contain a subpopulation of CD34+/PDGFRα+ VICs, which might be involved in the maintenance of local microenvironment resisting to pathologic remodeling. Their reduced number in older age could limit the self-regenerative properties of the valve stroma.


Subject(s)
Antigens, CD34/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve/cytology , Calcinosis/pathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Calcinosis/metabolism , Cells, Cultured , Female , Humans , Male , Middle Aged
17.
Sci Rep ; 10(1): 10317, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32587322

ABSTRACT

Calcific aortic valve disease (CAVD) is the most common form of valve disease where the only available treatment strategy is surgical valve replacement. Technologies for the early detection of CAVD would benefit the development of prevention, mitigation and alternate therapeutic strategies. Two-photon excited fluorescence (TPEF) microscopy is a label-free, non-destructive imaging technique that has been shown to correlate with multiple markers for cellular differentiation and phenotypic changes in cancer and wound healing. Here we show how specific TPEF markers, namely, the optical redox ratio and mitochondrial fractal dimension, correlate with structural, functional and phenotypic changes occurring in the aortic valve interstitial cells (VICs) during osteogenic differentiation. The optical redox ratio, and fractal dimension of mitochondria were assessed and correlated with gene expression and nuclear morphology of VICs. The optical redox ratio decreased for VICs during early osteogenic differentiation and correlated with biological markers for CAVD progression. Fractal dimension correlated with structural and osteogenic markers as well as measures of nuclear morphology. Our study suggests that TPEF imaging markers, specifically the optical redox ratio and mitochondrial fractal dimension, can be potentially used as a tool for assessing early CAVD progression in vitro.


Subject(s)
Aortic Valve Stenosis/diagnosis , Aortic Valve/pathology , Calcinosis/diagnosis , Intravital Microscopy/methods , Osteoblasts/pathology , Animals , Aortic Valve/cytology , Aortic Valve Stenosis/pathology , Calcinosis/pathology , Cell Differentiation , Cell Nucleus/pathology , Cells, Cultured , Disease Progression , Feasibility Studies , Humans , Male , Microscopy, Fluorescence, Multiphoton/methods , Mitochondria/pathology , Osteoblasts/cytology , Oxidation-Reduction , Primary Cell Culture , Swine
18.
Atherosclerosis ; 298: 27-35, 2020 04.
Article in English | MEDLINE | ID: mdl-32169720

ABSTRACT

BACKGROUND AND AIMS: Reduced bioavailability of nitric oxide (NO) has been implicated in the pathogenesis of calcific aortic stenosis. Herein, we investigated the effects of l-Arginine, the main precursor of NO, on the osteogenic differentiation of aortic interstitial valve cells (VICs). METHODS: We isolated a clonal population of bovine VICs that expresses osteogenic markers and induces calcification of collagen matrix after stimulation with endotoxin (LPS 500 ng/mL). VICs were treated in vitro with different combinations of LPS ± l-Arginine (50 or 100 mM) and cell extracts were collected to perform proteomic (iTRAQ) and gene expression (RT-PCR) analysis. RESULTS: l-Arginine prevents the over-expression of alkaline phosphatase (ALP, p < 0.001) and reduces matrix calcification (p < 0.05) in VICs treated with LPS. l-Arginine also reduces the over-expression of inflammatory molecules induced by LPS (TNF-alpha, IL-6 and IL-1beta, p < 0.001). The proteomic analysis allowed to identify 49 proteins with an altered expression profile after stimulation with LPS and significantly modified by l-Arginine. These include proteins involved in the redox homeostasis of the cells (i.e. Xanthine Oxidase, Catalase, Aldehyde Oxidase), remodeling of the extracellular matrix (i.e. ADAMTSL4, Basigin, COL3A1) and cellular signaling (i.e. Fibrillin-1, Legumain, S100A13). The RT-PCR analysis confirmed the modifications of Fibrillin-1, ADAMTSL4, Basigin and Xanthine Oxidase, whose expression levels increase after stimulation with LPS and are reduced by l-Arginine (p < 0.05). CONCLUSIONS: l-Arginine prevents osteogenic differentiation of VICs and reduces matrix calcification. This effect is achieved through the modulation of proteins involved in the cellular redox system, remodeling of extracellular matrix and inflammatory activation of VICs.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/drug effects , Aortic Valve/pathology , Arginine/metabolism , Arginine/pharmacology , Arteritis/metabolism , Calcinosis/metabolism , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Animals , Aortic Valve/cytology , Aortic Valve/metabolism , Cattle , Cell Differentiation/drug effects , Cells, Cultured , Osteogenesis/drug effects , Proteomics
19.
Int J Mol Sci ; 21(4)2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32074942

ABSTRACT

BACKGROUND: Calcific aortic valve disease (CAVD) is a chronic inflammatory disease that manifests as progressive valvular fibrosis and calcification. An inflammatory milieu in valvular tissue promotes fibrosis and calcification. Aortic valve interstitial cell (AVIC) proliferation and the over-production of the extracellular matrix (ECM) proteins contribute to valvular thickening. However, the mechanism underlying elevated AVIC fibrogenic activity remains unclear. Recently, we observed that AVICs from diseased aortic valves express higher levels of neurotrophin 3 (NT3) and that NT3 exerts pro-osteogenic and pro-fibrogenic effects on human AVICs. HYPOTHESIS: Pro-inflammatory stimuli upregulate NT3 production in AVICs to promote fibrogenic activity in human aortic valves. METHODS AND RESULTS: AVICs were isolated from normal human aortic valves and were treated with lipopolysaccharide (LPS, 0.20 µg/mL). LPS induced TLR4-dependent NT3 production. This effect of LPS was abolished by inhibition of the Akt and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) pathways. The stimulation of TLR4 in human AVICs with LPS resulted in a greater proliferation rate and an upregulated production of matrix metallopeptidases-9 (MMP-9) and collagen III, as well as augmented collagen deposition. Recombinant NT3 promoted AVIC proliferation in a tropomyosin receptor kinase (Trk)-dependent fashion. The neutralization of NT3 or the inhibition of Trk suppressed LPS-induced AVIC fibrogenic activity. CONCLUSIONS: The stimulation of TLR4 in human AVICs upregulates NT3 expression and promotes cell proliferation and collagen deposition. The NT3-Trk cascade plays a critical role in the TLR4-mediated elevation of fibrogenic activity in human AVICs. Upregulated NT3 production by endogenous TLR4 activators may contribute to aortic valve fibrosis associated with CAVD progression.


Subject(s)
Heart Defects, Congenital/pathology , Heart Valve Diseases/pathology , Neurotrophin 3/metabolism , Toll-Like Receptor 4/metabolism , Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve/pathology , Bicuspid Aortic Valve Disease , Carbazoles/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Female , Heart Defects, Congenital/metabolism , Heart Valve Diseases/metabolism , Humans , Indole Alkaloids/pharmacology , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinase 9/metabolism , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Up-Regulation/drug effects
20.
Mol Cell Probes ; 50: 101509, 2020 04.
Article in English | MEDLINE | ID: mdl-31945413

ABSTRACT

AIM: Degenerative calcific aortic valve disease (DCAVD) is a common valve disease characterized by massive calcium deposits in the aortic valve. Osteoblast differentiation of valve interstitial cells (VICs) is responsible for the formation of calcific nodules. This study aims to explore the function and underlying mechanism of long non-coding RNA (lncRNA) AFAP1-AS1 (actin filament-associated protein 1 antisense RNA 1) in the pathogenesis of DCAVD. METHODS: AFAP1-AS1, miR-155 and mRNA levels were detected by qRT-PCR. Protein levels were measured by Western blot. Calcification deposition was examined by Alizarin Red staining. The interaction between AFAP1-AS1 and miR-155, as well as miR-155 and SMAD5 was evaluated using luciferase reporter assay. RESULTS: AFAP1-AS1 expression was increased both in calcified aortic valves from DCAVD patients and after osteogenic induction in human VICs. Furthermore, AFAP1-AS1 overexpression promoted osteogenic differentiation of VICs, whereas AFAP1-AS1 knockdown inhibited osteogenic differentiation. Mechanistically, AFAP1-AS1 acted as a sponge for miR-155 to elevate SMAD5 expression. Further functional assays revealed that miR-155 mimic and SMAD5 silencing effectively reversed AFAP1-AS1-promoted osteogenic differentiation of VICs. CONCLUSION: Collectively, AFAP1-AS1 promotes osteogenic differentiation of VICs, at least in part, by sponging miR-155 to upregulate SMAD5. This study sheds new light on lncRNA-directed therapeutics in DCAVD.


Subject(s)
Aortic Valve/cytology , Cell Differentiation/genetics , MicroRNAs/metabolism , Osteoblasts/cytology , RNA, Long Noncoding/metabolism , Signal Transduction , Smad5 Protein/metabolism , Aortic Valve/pathology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Base Sequence , Calcinosis/genetics , Calcinosis/pathology , Female , Humans , Male , MicroRNAs/genetics , Middle Aged , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteopontin/genetics , Osteopontin/metabolism , RNA, Long Noncoding/genetics , Smad5 Protein/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...