human aortic valve
Recently Published Documents


TOTAL DOCUMENTS

220
(FIVE YEARS 60)

H-INDEX

31
(FIVE YEARS 6)

2021 ◽  
Vol 12 ◽  
Author(s):  
Fan Zhang ◽  
Naixuan Cheng ◽  
Yingchun Han ◽  
Congcong Zhang ◽  
Haibo Zhang

Calcific aortic valve disease (CAVD) is the most common structural heart disease, and the morbidity is increased with elderly population. Several microRNAs (miRNAs) have been identified to play crucial roles in CAVD, and numerous miRNAs are still waiting to be explored. In this study, the miRNA expression signature in CAVD was analyzed unbiasedly by miRNA-sequencing, and we found that, compared with the normal control valves, 152 miRNAs were upregulated and 186 miRNAs were downregulated in calcified aortic valves. The functions of these differentially expressed miRNAs were associated with cell differentiation, apoptosis, adhesion and immune response processes. Among downregulated miRNAs, the expression level of miR-139-5p was negatively correlated with the osteogenic gene RUNX2, and miR-139-5p was also downregulated during the osteogenic differentiation of primary human aortic valve interstitial cells (VICs). Subsequent functional studies revealed that miR-139-5p overexpression inhibited the osteogenic differentiation of VICs by negatively modulating the expression of pro-osteogenic gene FZD4 and CTNNB1. In conclusion, these results suggest that miR-139-5p plays an important role in osteogenic differentiation of VICs via the Wnt/β-Catenin pathway, which may further provide a new therapeutic target for CAVD.


2021 ◽  
Vol 12 ◽  
Author(s):  
Maha Alaqeeli ◽  
Dominique Mayaki ◽  
Sabah N. A. Hussain

Background: Long non-coding RNAs (lncRNAs) are non-coding RNAs that have more than 200 nucleotides. They have recently emerged as important regulators of angiogenesis. To identify novel lncRNAs that may be involved in the regulation of angiogenesis, we detected the mRNA of 84 lncRNAs in human umbilical vein endothelial cells (HUVECs) exposed to hypoxia for 24h. One of these, rhabdomyosarcoma 2-associated transcript (RMST), is significantly upregulated by hypoxia. Little is known about the presence and roles of RMST in EC function.Objective: The main objective of the study was to investigate the regulation of RMST in ECs and to determine its role in EC survival, proliferation, migration, and differentiation.Methods: Using qPCR, basal mRNA levels of 10 RMST isoforms in HUVECs were measured. Levels were then measured in response to 24h of hypoxia, 7days of differentiation in a co-culture assay, and exposure to four different angiogenesis factors. Functional roles of RMST in EC survival, migration, and differentiation were quantified by using a loss-of-function approach (transfection with single-stranded antisense LNA GapmeRs). EC survival was measured using cell counts and crystal violet assays. Cell migration and differentiation were measured using scratch wound healing and Matrigel® differentiation assays, respectively.Results: Five RMST isoforms (RMST-202, -203, -204, -206, and -207) were detected in HUVECs and human microvascular endothelial cells (HMEC-1s). Other types of vascular cells, including human aortic valve interstitial cells and human aortic smooth muscle cells, did not display this expression profile. RMST was significantly upregulated in response to 24h of hypoxia and in response to 7days of HUVEC co-culture with human lung fibroblasts. RMST was significantly downregulated by angiopoietin-2 (Ang-2), but not by VEGF, FGF-2, or angiopoietin-1 (Ang-1). Selective knockdown of RMST demonstrated that it promotes EC survival in response to serum deprivation. It is also required for VEGF- and Ang-1-induced EC survival and migration, but not for differentiation.Conclusion: We conclude that RMST is expressed in human ECs and that this expression is upregulated in response to hypoxia and during differentiation into capillary-like structures. We also conclude that RMST plays important roles in EC survival and migration.


Author(s):  
Miguel Sáinz-Jaspeado ◽  
Ross O. Smith ◽  
Oscar Plunde ◽  
Sven-Christian Pawelzik ◽  
Yi Jin ◽  
...  

Background: Palmdelphin (PALMD) belongs to the family of Paralemmin proteins implicated in cytoskeletal regulation. Single nucleotide polymorphisms (SNPs) in the PALMD locus that result in reduced expression are strong risk factors for development of calcific aortic valve stenosis (CAVS) and predict severity of the disease. Methods: Immunodetection and public database screening showed dominant expression of PALMD in endothelial cells (ECs) in brain and cardiovascular tissues including aortic valves. Mass spectrometry, co-immunoprecipitation and immunofluorescent staining allowed identification of PALMD partners. The consequence of loss of PALMD expression was assessed in siRNA-treated EC cultures, in knockout mice, and human valve samples. RNA sequencing of ECs and transcript arrays on valve samples from an aortic valve study cohort including patients with the SNP rs7543130, informed about gene regulatory changes. Results: ECs express the cytosolic PALMD-KKVI splice variant, which associated with RAN GTPase activating protein1 (RANGAP1). RANGAP1 regulates the activity of the GTPase RAN and thereby, nucleocytoplasmic shuttling via Exportin1 (XPO1). Reduced PALMD expression resulted in subcellular relocalization of RANGAP1 and XPO1, and nuclear arrest of the XPO1 cargoes p53 and p21. This indicates an important role for PALMD in nucleocytoplasmic transport and consequently, in gene regulation due to the impact on localization of transcriptional regulators. Changes in EC responsiveness upon loss of PALMD expression included failure to form a perinuclear actin cap when exposed to flow, indicating lack of protection against mechanical stress. Loss of the actin cap correlated with misalignment of the nuclear long axis relative to the cell body, observed in PALMD -deficient ECs, Palmd −/− mouse aorta and human aortic valve samples derived from CAVS patients. In agreement with these changes in EC behavior, gene ontology analysis showed enrichment of nuclear- and cytoskeleton-related terms in PALMD -silenced ECs. Conclusions: We identify RANGAP1 as a PALMD partner in ECs. Disrupting the PALMD/RANGAP1 complex alters the subcellular localization of RANGAP1 and XPO1, and leads to nuclear arrest of the XPO1 cargoes p53 and p21, accompanied by gene regulatory changes and loss of actin-dependent nuclear resilience. Combined, these consequences of reduced PALMD expression provide a mechanistic underpinning for PALMD's contribution to CAVS pathology.


2021 ◽  
Vol 7 (2) ◽  
pp. 45-48
Author(s):  
Maximilian Winkelkotte ◽  
Florian Schmieder ◽  
Stephan Behrens ◽  
Dominic Salminger ◽  
Anett Jannasch ◽  
...  

Abstract Aortic valve (AV) stenosis is characterized by tissue fibrosis and calcification. Fibrous thickening can result in reduced tissue oxygen supply leading to pathological valvular interstitial cell (VIC) differentiation and calcification. Static 2D VIC cultures and animal models are limited in the ability to reflect human AV calcification. Culturing of VICs in micro-physiological-systems (MPS) in a pulsatile flow and the establishment of a modular AV tissue incubation chamber (TIC) are new approaches to evaluate pathophysiological processes of AV disease. Therefore, a MPS able to adjust hypoxic conditions was applied for VIC culture. A significant increase of mRNA-expression of EGLN1 and HIF1α- regulated LDHA and HIF1α nuclear localisation were proven under hypoxia. AV tissue culture was established within a TIC and viability was monitored by Resazurin-reduction in the incubation medium and visualized by LDH-activity in tissue cryosections. Viability was compared between fluid and static incubated tissues revealing an advantageous effect of the fluidic assay condition. Consecutively, the application of MPS in AV research allows i) the investigation of VIC cultures with efficient oxygen regulation and ii) the culture of porcine or human AV tissues preserving viability and specifically reflecting in vivo parameters. These methods open up new possibilities beyond static 2D culture and facilitate a reduction of animal experiments in AV research.


2021 ◽  
Vol 42 (Supplement_1) ◽  
Author(s):  
K Dhayni ◽  
Y Chabry ◽  
L Henaut ◽  
H Ouled-Haddou ◽  
C Avondo ◽  
...  

Abstract Background/Introduction Inflammation is a key feature of calcific aortic valve stenosis (CAVS) against which there is currently no pharmacological treatment. Purpose To verify the hypothesis that interleukin-8 (IL-8), a pro-inflammatory factor involved in arterial calcifications, also promotes the calcification of human aortic valve interstitial cells (hVICs). Methods Primary hVICs were isolated from healthy pieces of aortic valves harvested from patients undergoing surgical valve replacement. They were cultured in a pro-calcifying condition (Pi-3.8mM) with or without IL-8 (5 to 50 pg/ml) for up to 21 days. Calcification was analysed by alizarin red staining and calcium content was measured with the o-cresolphthalein complexone method. The viability of hVICs was verified by the MTT assay. The expression of osteogenic (BMP2, OPN, osterix and ALP) and myofibrotic (alpha-SMA, collagen-1, collagen-3 and elastin) markers as well as that of metalloproteases (MMP-2, -9 and -12) was analysed by RT-qPCR. The expression of IL-8 receptors, CXCR-1 and CXCR-2 was evaluated by Western blot and flow cytometry, and the effects of IL-8 were tested in the presence or absence of SCH527123, an antagonist of CXCR-1 and CXCR-2. Finally, the expression of CXCR-1 and -2 and elastin was analysed by immunohistochemistry in the calcified and non-calcified areas of human aortic valve samples. All of these experiments were carried out from valves of at least 5 different donors and a P<0.05 was considered statistically significant. Results IL-8 (15 pg/mL) caused a significant ∼2-fold increase in the calcification of hVICs in the Pi condition, compared to the Pi-only condition, without modulation of cell viability. In the presence of Pi, IL-8 exposure significantly stimulated the expression of the transcripts of elastin and MMP-12, an elastase, and reduced that of OPN, a well-known inhibitor of calcification. The effects of IL-8 on hVICs calcification and on the expression of MMP-12, elastin and OPN transcripts were significantly prevented by the addition of SCH527123. In addition, the expression of CXCR-1 and -2 was confirmed in histological samples of human aortic valves. This expression was more pronounced in calcified areas compared to non-calcified areas and co-localized with degraded elastin. Conclusion IL-8 promoted the calcification of hVICs in culture. This effect was significantly prevented by antagonizing CXCR-1 and CXCR-2 IL-8 receptors, which we showed for the first time to be expressed by human VICs and aortic valves of patients with CAVS. Further studies are underway to clarify the cellular mechanisms involved. FUNDunding Acknowledgement Type of funding sources: Public Institution(s). Main funding source(s): Fédération Hospitalo-Universitaire REMOD-VHF


2021 ◽  
Vol 22 (19) ◽  
pp. 10605
Author(s):  
Iván Parra-Izquierdo ◽  
Tania Sánchez-Bayuela ◽  
Javier López ◽  
Cristina Gómez ◽  
Enrique Pérez-Riesgo ◽  
...  

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.


2021 ◽  
Vol 8 ◽  
Author(s):  
Joshua D. Hutcheson ◽  
Florian Schlotter ◽  
Michael D. Creager ◽  
Xiaoshuang Li ◽  
Tan Pham ◽  
...  

Objective: Aortic valve (AV) leaflets rely on a precise extracellular matrix (ECM) microarchitecture for appropriate biomechanical performance. The ECM structure is maintained by valvular interstitial cells (VICs), which reside within the leaflets. The presence of pigment produced by a melanocytic population of VICs in mice with dark coats has been generally regarded as a nuisance, as it interferes with histological analysis of the AV leaflets. However, our previous studies have shown that the presence of pigment correlates with increased mechanical stiffness within the leaflets as measured by nanoindentation analyses. In the current study, we seek to better characterize the phenotype of understudied melanocytic VICs, explore the role of these VICs in ECM patterning, and assess the presence of these VICs in human aortic valve tissues.Approach and Results: Immunofluorescence and immunohistochemistry revealed that melanocytes within murine AV leaflets express phenotypic markers of either neuronal or glial cells. These VIC subpopulations exhibited regional patterns that corresponded to the distribution of elastin and glycosaminoglycan ECM proteins, respectively. VICs with neuronal and glial phenotypes were also found in human AV leaflets and showed ECM associations similar to those observed in murine leaflets. A subset of VICs within human AV leaflets also expressed dopachrome tautomerase, a common melanocyte marker. A spontaneous mouse mutant with no aortic valve pigmentation lacked elastic fibers and had reduced elastin gene expression within AV leaflets. A hyperpigmented transgenic mouse exhibited increased AV leaflet elastic fibers and elastin gene expression.Conclusions: Melanocytic VIC subpopulations appear critical for appropriate elastogenesis in mouse AVs, providing new insight into the regulation of AV ECM homeostasis. The identification of a similar VIC population in human AVs suggests conservation across species.


2021 ◽  
Vol 10 (2) ◽  
pp. 16-24
Author(s):  
T. V. Glushkova ◽  
A. E. Kostyunin

Highlights. The morphology and elemental composition of calcium deposits formed in the tissues of epoxytreated aortic and mitral bioprostheses do not differ from those in the mineralized matrix of stenotic human aortic valve leaflets. Despite similar elemental composition of mineral deposits in the KemCor and UniLine bioprostheses, the morphology of these calcifications differs between bioprosthetic heart valve substitutes and, apparently, is associated with the specific structure of the fibrous matrix of the biological tissues that are used for their manufacturing.Aim. To analyze the morphology and elemental composition of mineral deposits formed in epoxy-treated aortic and mitral bioprosthetic heart valves made from xenoaortic or xenopericardial material and to compare the obtained findings with the data on calcified human aortic valve.Methods. Leaflets of the mitral and aortic bioprosthetic heart valves KemCor and UniLine (NeoKor, L Russia, Kemerovo) that were explanted due to their failure, as well as leaflets of the calcified native aortic valve were evaluated. The morphology of calcifications was studied by scanning electron microscopy using an S-3400N microscope (Hitachi, Japan). The elemental composition of calcium deposits was studied by electron probe microanalysis using Hitachi S-3400N microscope with energy dispersive spectrometer Bruker XFlash 4010 (Bruker, Germany).Results. Large calcifications located at the internal layers of samples were surrounded by collagen fibers commonly with evident signs of the onset of mineralization. Calcium deposits in the native aortic valve and xenoartic bioprostheses KemCor were located mainly at the spongy layer and had a loose structure, while dense lamellar deposits were found at the leaflets of pericardial bioprostheses UniLine. The elemental composition of calcium deposits showed the presence of Ca, P, O, Mg, and Na in the mineralized regions and the presence of S in the regions of low electron density. The calcium to phosphorus ratio (Ca:P) in the calcifications of the aortic valve leaflets was 1.81 (1.79-1.84; min - 1.48; max - 2.05), whereas the Ca:P ratios in the UniLine and KemCor bioprostheses were 1.78 (1.75-1.86; min - 1.52; max - 2.03) and 1.82 (1.81-1.88; min - 1.71; max - 2.06), respectively. There were no significant differences in the Ca:P ratios between calcifications in the study groups (p>0.05).Conclusion. Calcium deposits detected in epoxy-treated bioprostheses and human aortic valve appeared to be formed under dystrophic calcification. The morphology of calcifications in bioprostheses depended on the type of biological tissue. None correlations between the morphological structure of calcifications and the implantation position were found in bioprosthetic leaflets. The elemental composition of mineral deposits was similar in all study samples.


Sign in / Sign up

Export Citation Format

Share Document