Skip to main content
Erschienen in: BMC Cardiovascular Disorders 1/2019

Open Access 01.12.2019 | Research article

Heat shock protein 90 is downregulated in calcific aortic valve disease

verfasst von: Jonna Weisell, Pauli Ohukainen, Juha Näpänkangas, Steffen Ohlmeier, Ulrich Bergmann, Tuomas Peltonen, Panu Taskinen, Heikki Ruskoaho, Jaana Rysä

Erschienen in: BMC Cardiovascular Disorders | Ausgabe 1/2019

Abstract

Background

Calcific aortic valve disease (CAVD) is an atheroinflammatory process; finally it leads to progressive calcification of the valve. There is no effective pharmacological treatment for CAVD and many of the underlying molecular mechanisms remain unknown. We conducted a proteomic study to reveal novel factors associated with CAVD.

Methods

We compared aortic valves from patients undergoing valvular replacement surgery due to non-calcified aortic insufficiency (control group, n = 5) to a stenotic group (n = 7) using two-dimensional difference gel electrophoresis (2D-DIGE). Protein spots were identified with mass spectrometry. Western blot and immunohistochemistry were used to validate the results in a separate patient cohort and Ingenuity Pathway Analysis (IPA) was exploited to predict the regulatory network of CAVD.

Results

We detected an upregulation of complement 9 (C9), serum amyloid P-component (APCS) and transgelin as well as downregulation of heat shock protein (HSP90), protein disulfide isomerase A3 (PDIA3), annexin A2 (ANXA2) and galectin-1 in patients with aortic valve stenosis. The decreased protein expression of HSP90 was confirmed with Western blot.

Conclusions

We describe here a novel data set of proteomic changes associated with CAVD, including downregulation of the pro-inflammatory cytosolic protein, HSP90.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12872-019-01294-2.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
2D-DIGE
Two-dimensional difference gel electrophoresis
Akt
Protein kinase B
ANXA2
Annexin A2
APCS
Serum amyloid P-component
AS
Aortic stenosis
BMP2
Bone morphogenic protein 2
C9
Complement 9
CAVD
Calcific aortic valve disease
eNOS
Endothelial nitric oxide synthase
ERK
Extracellular signal regulated kinase
HSP90
Heat shock protein 90
MAPK
Mitogen activated protein kinase
PDIA3
Protein disulfide isomerase A3
ROS
Reactive oxygen species
RUNX2
Runt-related transcription factor 2
VICs
Valve interstitial cells

Background

Calcific aortic valve disease (CAVD) is a progressive disease, which originates from endothelial cell damage on the aortic surface of aortic valve followed by an accumulation of oxidized lipids and the infiltration of inflammatory cells into the valve [1]. This promotes active remodeling of the extracellular matrix with the disorganization of collagen fibers, resulting in a thickening of the aortic valve leaflets. Furthermore, osteogenic programming of valve interstitial cells (VICs), causes progressive calcification and ultimately a severe obstruction of cardiac outflow. Several factors and signaling pathways have been linked to CAVD, e.g. interleukins, tumor necrosis factor (TNF), matrix metalloproteinases, bone morphogenic protein 2 (BMP2) and osteogenic regulator runt-related transcription factor 2 (RUNX2) [1, 2]. However, despite recent progress in understanding the molecular pathogenesis of CAVD, the factors driving the progression of this disease are not fully understood.
Various omics-analyses have been performed to gain a better understanding of the molecular mechanism underpinning CAVD. Transcriptomic studies of human CAVD have been undertaken to identify differentially expressed genes [36] and microRNAs [4, 7] in different stages of aortic valve calcification. In addition, there have been multiple proteomics studies of CAVD performed [816]. However, only three of them compared stenotic valves to control valves [9, 12, 16], and of these, only Schlotter et al. [12] reported the use of tricuspid valves in their study. Furthermore, Schlotter et al. [12] combined the results from proteomics and transcriptomics to describe the integrated molecular dataset of human CAVD [12].
In this study, we conducted a proteomic analysis of aortic valve calcification by comparing control and stenotic human aortic valves using two-dimensional difference gel electrophoresis (2D-DIGE). Selected proteomic changes were confirmed with Western blotting and immunohistochemistry. In addition, Ingenuity pathway analysis (IPA) was used to clarify the potential signaling pathways associated with identified proteins.

Methods

Patients

The aortic valves examined in this study were obtained from 50 patients at the time of aortic valve or aortic root surgery. All operations were made following normal surgical procedures. The study protocol was approved by the Research Ethics Committee of Oulu University Hospital and it conformed to the principles outlined in the Declaration of Helsinki. The aortic valve cusps were immersed immediately after removal into liquid nitrogen and stored at − 70 °C until analyzed.
For proteomics study, patients were divided into two groups: the control group (C, n = 5) consisted of patients with normal, non-calcified, smooth and pliable aortic valve cusps, operated due to ascending aortic pathology (aneurysm or dissection) or aortic regurgitation. The aortic stenosis group (AS, n = 7) consisted of patients who had non-rheumatic, severe aortic valve sclerosis with an increased degree of calcification. Patients who were identified as exhibiting macroscopic thickenings of aortic valve cusps, which were microscopically identified mainly as fibrotic and mild sclerotic lesions, were excluded from the study.
The patients’ demographics are presented in Table 1. There were no significant differences in gender, left ventricular ejection fraction or comorbidities between the study groups, and valve anatomy. However, the average age of the aortic stenosis (AS) patients was significantly higher than the patients in the control group. Histologically, the stenotic valves had a significantly elevated amount of calcium and more neovessels in comparison with control valves [4, 17, 18]. For validation of proteomics results, a separate matching group of patients (n = 39) was selected.
Table 1
Demographics of the patients examined in the proteomic analysis
 
Control
Aortic Stenosis
P-value
Patients, n
5
7
 
Male, n (%)
5 (100%)
5 (71.4%)
0.46
Bicuspid valve
0
2 (28.6%)
0.47
Age (years), range
43.4 (33.2–53.6)
66.6 ± (53.8–79.3)
0.007
LVEF (SD)
58.0 ± 11.2
56.7 ± 8.8
0.53
DM, n (%)
0
1 (14.3%)
≥0.9
CHD, n (%)
0
3 (42.9%)
0.21
ASO, n (%)
0
1 (14.3%)
≥0.9
Statin use, n (%)
1 (20%)
4 (66.7%)
0.24
ASO Peripheral atherosclerosis, CHD Coronary heart disease, DM Diabetes mellitus, LVEF Left ventricle ejection fraction; SD, standard deviation

Two-dimensional difference gel electrophoresis (2D-DIGE)

The proteins extracted from control (C, n = 5) and calcified (AS, n = 7) aortic valves were further purified by buffer exchange using an Amicon Ultra ultrafiltration unit with a 10 kDa cutoff (Millipore) and urea buffer (7 M urea, 2 M thiourea, 4% [w/v] CHAPS, 30 mM Tris, pH 8.5) and then the protein samples were sonicated and centrifuged. Protein amounts in the supernatants were determined with a Bradford-based assay according to the manufacturer’s instructions (Roti®-Nanoquant) and the aliquots were stored at − 70 °C. Protein labeling was performed with CyDye DIGE Fluor minimal dyes (GE Healthcare) according to the manufacturer’s protocol using 400 pmol Cy3 (pooled standard) and Cy5 (control, AS, respectively) for 50 μg protein. Proteins were separated as described earlier [19]. In brief, immobilized pH gradient (IPG) strips (pH 3–10 nonlinear, 24 cm, GE Healthcare) were incubated overnight in 650 μl rehydration buffer (7 M urea, 2 M thiourea, 4% [w/v] CHAPS, 130 mM [w/v] DTT, 2%[v/v] carrier ampholytes 3–10, Complete Mini protease inhibitor cocktail [Roche Life Science]). Isoelectric focusing (IEF) after anodic sample cup-loading was carried out with the Multiphor II system (GE Healthcare) under paraffin oil with 67 kVh. SDS-PAGE was performed overnight in polyacrylamide gels (12.5%) with the Ettan DALT II system (GE Healthcare) at 1–2 W per gel in 12 °C. Fluorescence signals were detected with a Typhoon 9400 (GE Healthcare) and 2-D gels analyzed with Delta2D 4.0 (Decodon). Theoretical spot positions were calculated with the Compute pI/Mw tool (http://​ca.​expasy.​org/​tools/​pi_​tool.​html). Principal Component Analysis was performed with the Delta2D v4.0 software (Decodon) according to the spot intensities on every gel image.

Mass spectrometry

For protein identification, additional 2-D gels were run with a higher amount of unlabelled protein (400–600 μg) combined with 50 μg Cy3-labelled internal standard. After detection of the fluorescence signals (see above) and silver staining, labelled and unlabelled protein patterns were matched with the 2-D PAGE image analysis software Melanie 3.0 (GeneBio). Spots with correctly matched centers were excised, digested with trypsin (recombinant; Roche) and prepared for MALDI-TOF mass spectrometry as described previously [19]. The extracted and dried peptides were dissolved in 5 μl alpha-Cyano-3-hydroxycinnamic acid (98%, recrystallized from ethanol-water, 5 mg/ ml in 50% acetonitrile and 0.1% TFA) and 0.5 μl applied onto the sample plate using the dried-droplet method. Proteins were identified from PMF obtained with a VOYAGER-DE™ STR (Applied Biosystems) as described earlier [19]. In general, the clearest peaks (up to 50) visible in the mass spectrum were used to identify proteins with Mascot (http://​www.​matrixscience.​com/​) using Swiss-Prot as the corresponding protein database. Search parameters were enzyme: trypsin; modifications: oxidation of Met; missed cleavage: 1; resolution: monoisotopic; ion mode: [M + H]; threshold: 50 ppm. The protein identification was accepted if at least 4 major peaks matched to the protein with the highest Mascot score. In addition, the identification was confirmed by analyzing the induced spot from different gels. During later stages of the project, mass spectra of the tryptic digests were obtained with a UltrafleXtreme MALDI TOF/TOF instrument (Bruker Daltonics) where up to 10 ions from each peptide fingerprint were subjected to the MS/MS measurement. Data were processed with Flexanalyis and Biotools (Bruker) and combined PMF/MS/MS spectra were searched against the NCBI or Swiss-Prot non-redundant protein database using Mascot (Matrix science) with standard search parameters (MS tolerance: 30 ppm, MS/MS tolerance: 0.7 Da, modifications: Carbamidomethyl (Cys) and optional oxidation of Met, up to 1 missed cleavage).

Protein extraction and Western blot

In the western blot experiments, aortic valve samples were obtained from a separate matching cohort (C, n = 19 AS, n = 20). The samples were ground in liquid nitrogen, and then homogenized for 10 min in a lysis buffer containing inhibitors. The lysis buffer itself contained 1 M Tris (pH 7.5), 3 M NaCl, 0.25 M EDTA (pH 8.0), 0.1 M EGTA (pH 7.9), 1 mmol/l β-glycerophosphate, 1 mmol/l Na3VO4, 2 mmol/l benzamidine, 1 mmol/l phenylmethylsulfoxide, 50 mmol/l NaF, 1 mmol/l dithiothreitol and 10 μg/ml each of leupeptin, pepstatin, aprotininand and distilled water. The valve tissue samples were homogenized using a MagnaLyser instrument (Roche). After homogenization, the samples were centrifuged for 20 min in 12,500 rpm and + 4 °C and then the supernatant was collected for protein isolation. 5x NEB lysis buffer (100 mM Tris-HCl [pH 7.5], 750 mM.
NaCl, 5 mM EDTA, 5 mM EGTA, 5% Triton X 100, 12 mM sodium pyrophosphate, 5 mM β-glycerophosphate, 5 mM Na3VO4) was added and mixed following centrifugation for 20 min in 12,500 rpm in + 4 °C. Supernatant containing the total fraction was collected. Western blot was performed using a 1.0 mm, 12% gel with 40 μg of protein/well. The following primary antibodies were used: HSP90α (ADI-SPS-771) and HSP90β (ADI-SPA-844) from Enzo Life Sciences, Protein kinase B (Akt) (#9272), Phospho- Akt (#4056), p38 mitogen activated protein kinase (MAPK) (#9212), phospho-p38 MAPK (#9211), extracellular signal regulated kinase p44/42 MAPK (Erk1/2) (#9102) and Phospho-p44/42 MAPK (pErk1/2) (#9106) from Cell Signaling Technology, Inc., Anti-Annexin II (610,068, BD Transduction Laboratories), and Anti-Galectin 1 (ab25138, Abcam). Anti-mouse-IgG HRP-labeled (GE Healthcare), Anti-rabbit-IgG Peroxidase conjugate (Calbiochem), anti-IgG HRP-linked rabbit (#7074, Cell Signaling Technology, Inc.), and anti-IgG HRP-linked mouse (#7076, Cell Signaling Technology, Inc.) secondary antibodies were used. Data was quantified using the QuantityOne Software (Bio-Rad).

Histological stainings

The localization of HSP90α and HSP90β in the aortic valve cusps was studied by using immunohistochemical stainings. The aortic valve samples, sent for routine diagnistics, were fixed in buffered formalin solution and embedded in paraffin. Decalcification with EDTA was done if needed. For total valve area and calcified valve area slides were photographed with a Leica DFC420 camera (Wetzlar) and areas were quantified with Image J analysis software. Calcified area to total area was calculated with the following formula: (calcified valve area/ total valve area)*100. Before application of the primary antibodies, the 5-μm-thick sections of valves samples were heated in a microwave oven in citrate buffer, pH 6.0, for 30 min. Rabbit monoclonal antibodies ab133492 at a dilution of 1:2000 (Abcam) for HSP90α and ab32568 at a dilution of 1:300 (Abcam) for HSP90β were used to stain. 3,3′Diaminobenzidine (DAP) was used as the chromogen in immunostaining process. Negative control stainings were carried out by substituting nonimmune rabbit serum for the primary antibodies.

Molecular network analysis

The up- or down-regulated proteins with their respective expression values were uploaded for processing by the Ingenuity Pathway Analysis (IPA) software (Qiagen). A core analysis was performed with the following parameters: core analysis, reference set user-defined (i.e., only the set of differentially expressed genes by GeneSpring-software mapped to the IPA database), direct and indirect relationships included, confidence = experimentally observed. Then, the IPA-software was used to generate a molecular network showing the interrelationships between up- or down-regulated proteins as previously described [20], based on the information contained in the Ingenuity Pathways Knowledge database.

Statistical analysis

The results are expressed as mean with standard deviation (SD) unless otherwise stated. Continuous variables were analyzed by using Student’s t-test, semi-continuous variables using Mann-Whitney U-test and Fisher’s exact test for categorical variables. Analyses were performed using SPSS for Windows (IBM Corp. Released 2018. IBM SPSS Statistics for Windows, Version 25.0). Correlations were detected with linear regression model in Graphpad Prism 5. P < 0.05 was considered statistically significant.

Results

Proteomic analysis of aortic valve calcification

The proteomic analysis, based on the minimal DIGE, identified a total of 15 differentially abundant protein spots in stenotic valves as compared to control valves according to the selection criteria (fold change ≥1.5 and P ≤ 0.05). A typical 2D gel representing calcified aortic valve proteins is shown in Fig. 1. Further, mass spectrometry analyses identified seven proteins within 12 spots (Table 2) which had undergone significant upregulation i.e. complement 9, serum amyloid P-component (APCS) and transgelin (1.7-, 2.3- and 3.5-fold, respectively, P < 0.05) as well as downregulation of heat shock protein HSP90 (α/β; genes HSP90AA1/AB1), protein disulfide isomerase A3 (PDIA3), annexin A2 (ANXA2) and galectin-1 (2.1-, 3.5-, 2.2- and 2.2-fold, respectively, P < 0.05) in stenotic valves (Fig. 1, Table 2).
Table 2
Up- and down-regulated proteins as determined by 2D-DIGE in calcified valves as compared to control valves
 
Comparison Control vs. calcified valvesa
 
Protein identificationc
Spot
Protein
UniProt
Description
Ratio
P-value
Theoretical pI/MW (kDa)b
Score
(MS, MSMS)
SC (P)
(MS, MSMS)
1
HSP90α/β
P07900/ P08238
Heat shock protein HSP90AA1/AB1
− 2.05
0.022
AA1: 1: 4.94/84.7
113.0, −
39(18),-
2: 5.07/98.2
  
AB1: 4.96/83.3
  
2
C9
P02748
Complement 9
1.74
0.014
5.43/63.2 (5.42/60.9)
83.2, −
21(14),-
3
   
1.68
0.016
(C9a: 4.59/27.8)
88.5, −
18(13),-
4
   
1.55
0.018
(C9b: 8.63/33.2)
-, 72.9
-, 5(2)
6
PDIA3
P30101
Protein disulfide-isomerase A3
−3.35
0.016
5.98/56.8 (5.61/54.3)
162.0,-
46.8(21),-
8
ANXA2
P07355
Annexin A2
−2.22
0.012
1: 7.57/38.6 (7.56/38.5)
71.1,-
21(5),-
9
   
− 2.95
0.019
2: 8.53/40.4 (8.54/40.3)
199*,-
33(13),-
10
APCS
P02743
Serum amyloid P-component
2.26
0.014
6.10/25.4 (6.12/23.3)
75*, −
20(4),-
11
   
1.59
0.039
 
80*,-
20(5),-
12
   
1.51
0.046
 
82*,-
25(5),-
13
TAGLN
Q01995
Transgelin
3.45
0.032
8.87/22.6 (8.88/22.5)
176.00,-
76(27),-
15
LGALS1
P09382
Galectin-1
−2.19
0.012
5.30/14.7 (5.30/14.6)
92.4, 64.8
57(7), 44(2)
aThe ratio represents the change of the mean normalized volumes. Statistical significance is shown with the t-test (P < 0.05). Ratio and t-test values of the unidentified spots were as follows: spot 5 (−2.60, 0.0200), spot 7 (− 2.91, 0.0311) and spot 14 (− 2.12, 0.0159)
bThe theoretical expected spot position in the gel according to the full or matured (in brackets) protein sequence is shown. If the protein exists in different isoforms, then the specific number of the isoform is likewise indicated
cThe protein identification shows the MS and/or MSMS scores based on analysis with the UltrafleXtreme MALDI TOF/TOF instrument or measurements with the VOYAGER-DE™ STR (*). In addition, the sequence coverage (SC) and number of matched peptides (P) are shown
Table 3
Full annotation of genes illustrated in Fig. 4
Symbol
Entrez Gene Name
Location
Family
Entrez Gene
Akt
AKT Serine/Threonine Kinase
Cytoplasm
group
 
ALPK1
Alpha kinase 1
Other
kinase
80,216
ANXA2
Annexin A2
Plasma Membrane
other
302
APCS
Amyloid P component, serum
Extracellular Space
other
325
ARMC5
Armadillo repeat containing 5
Cytoplasm
other
79,798
C20orf194
Chromosome 20 open reading frame 194
Nucleus
other
25,943
C9
Complement C9
Extracellular Space
other
735
Ck2
Casein Kinase II
Cytoplasm
complex
 
DCLK2
Doublecortin like kinase 2
Cytoplasm
kinase
166,614
DDX59
DEAD-box helicase 59
Other
enzyme
83,479
DMRTA1
DMRT like family A1
Nucleus
transcription regulator
63,951
ERK
Extracellular Signal-Regulated Kinase 1/2
Other
group
 
G2E3
G2/M-phase specific E3 ubiquitin protein ligase
Cytoplasm
enzyme
55,632
GRK7
G protein-coupled receptor kinase 7
Cytoplasm
kinase
131,890
HSP90
Heat shock protein 90
Cytoplasm
group
 
HSP90AA1
Heat shock protein 90 alpha family class A member 1
Cytoplasm
enzyme
3320
HSP90AB1
Heat shock protein 90 alpha family class B member 1
Cytoplasm
enzyme
3326
IL6
Interleukin 6
Extracellular Space
cytokine
3569
INSRR
Insulin receptor related receptor
Plasma Membrane
kinase
3645
KCTD8
Potassium channel tetramerization domain containing 8
Other
other
386,617
LGALS1
Galectin 1
Extracellular Space
other
3956
MAP 3 K15
Mitogen-activated protein kinase kinase kinase 15
Other
other
389,840
MYLK4
Myosin light chain kinase family member 4
Cytoplasm
kinase
340,156
MYO3B
Myosin IIIB
Plasma Membrane
kinase
140,469
P38 MAPK
Mitogen-Activated Protein Kinase P38 Alpha
Cytoplasm
group
1432
PDIA3
Protein disulfide isomerase family A member 3
Cytoplasm
peptidase
2923
PSKH1
Protein serine kinase H1
Nucleus
kinase
5681
RPS6KL1
Ribosomal protein S6 kinase like 1
Other
kinase
83,694
STK32B
Serine/threonine kinase 32B
Other
kinase
55,351
TAGLN
Transgelin
Cytoplasm
other
6876
TNF
Tumor necrosis factor
Extracellular Space
cytokine
7124
TSSK2
Testis specific serine kinase 2
Cytoplasm
kinase
23,617
ZBED4
Zinc finger BED-type containing 4
Nucleus
transcription regulator
9889
ZNF215
Zinc finger protein 215
Nucleus
transcription regulator
7762

Confirmation of proteomics results

Western blot was used to validate the the proteomic results of HSP90, ANXA2 and galectin-1. The candidate proteins were selected based on their unknown role in CAVD. A marked downregulation of HSP90β protein levels was detected in stenotic valves compared to controls (Fig. 2a-b), whereas no change in HSP90α protein levels was observed (data not shown). Considerable interindividual variability was seen in the levels of ANXA2 protein, since it was highly expressed in only two out of three control samples (Fig. 3c). There was no significant difference in galectin-1 protein levels between stenotic and control levels (Fig. 3a-b).
To test if valvular anatomy impact on our data, we performed an unsupervised clustering analysis (principal component analysis, PCA) on the raw proteomic data (Additional file 1: Figure S1). We hypothesized that if valvular anatomy would affect protein expression profile, bicuspid valves should cluster together and distinctly from tricuspid valves. However, we find no evidence of such clustering and in fact, the clearest distinction emerges between control, and AS.
We correlated HSP90 expression levels both with age and calcification of the valves (expressed as proportion of calcified area in aortic valve cusps to total aortic valve). As shown in Additional file 2: Figure S2A-B, the HSP90β protein levels correlated with age of the patients (P < 0.01) and calcification of the valves (P < 0.05). In addition, the valvular calcification correlated with the age (P < 0.01) (Additional file 2: Figure S2C).
In the immunohistochemical stainings, the localization of HSP90α and HSP90β was virtually identical (Fig. 2b-e). VICs in both normal and calcified valves were positive. In addition, the endothelium of neovasculature was widely positively stained, whereas in the surface endothelium, the positive reaction was more patchy. Furthermore, most of the inflammatory cells, mainly lymphocytes, were also positively stained.

Identification of the molecular network between up- and down-regulated proteins

An IPA-analysis was used to determine the biological relationships among the differentially expressed proteins. The main molecular network exhibiting expression changes based on Fisher’s exact test is shown in Fig. 4.
Among the novel putative interactions suggested by IPA, HSP90 was linked to Akt and ERK, and further to p38 MAPK (Fig. 4). Therefore, we conducted Western blot analyses to evaluate the activation of Akt, ERK and p38 MAPK kinases in control and stenotic valves. The ratio of phosphorylated ERK to total ERK was increased (1.5-fold, P < 0.05) whereas the ratio of phosphorylated Akt to total Akt was reduced (0.7-fold, P < 0.05) suggesting that the Akt and ERK pathways were disturbed in the stenotic valves (Fig. 5a,c). Instead, there was no change in the phosphorylation pattern of p38 MAPK in stenotic valves (Fig. 5b).

Discussion

Here we describe a 2D-DIGE-determined proteomic profile associated with CAVD. We detected a distinct upregulation of APCS, C9 and transgelin as well as a downregulation of HSP90, PDIA3, ANXA2 and galectin-1 proteins in calcified valves in comparison to control valves. We confirmed this decrease in HSP90β protein levels in calcified valves by performing a Western blot analysis and then conducted an IPA analysis to predict HSP90 interactions in CAVD.
This is the first study revealing a decreased expression of HSP90 in calcified aortic valves. HSP90 is a molecular chaperone and a member of a large HSP family. Previously, a decreased expression of HSP27 in calcified valves has been reported in the proteomic study conducted by Martin-Rojas et al. [16]. In addition, several HSP family members are linked to the pathophysiology of atherosclerosis in which they have pro-inflammatory effects and regulate endothelial function (reviewed in [21]). In the endothelium, HSP90 is part of a complex with endothelial nitric oxide synthase ((eNOS)/HSP90) [2224]. The dissociation of HSP90 causes uncoupling of eNOS, leading to the production of reactive oxygen species (ROS) and endothelial dysfunction [23]. This process might be initiated by pro-inflammatory lipids [23, 24]. Interestingly, uncoupling of NOS and the increased level of oxidative stress have also been reported in calcified stenotic aortic valves [25]. In our study, HSP90 positivity was seen in the surface endothelium of the valves, and in the endothelium of the neovasculature of the calcified valves. Our findings are similar to those of Martín-Rojas et al. [16], who reported decreased HSP27 protein levels in the endothelium layer of stenotic valves [16]. In summary, the expression of HSP90 in the endothelium supports the idea that HSP90 has a role in regulating endothelial function in the progression of aortic valve calcification.
HSP90 was expressed in inflammatory cells that were mainly lymphocytes. In atherosclerosis, HSP90 is overexpressed in inflammatory sites of human atherosclerotic plaques [26, 27]. Furthermore, several studies have reported that inhibition of HSP90 can exert atheroprotective effects (decreased plaque size and reduced inflammatory responses) [27, 28] and less oxidative stress [29]. In CAVD, the amount of inflammation decreases when the disease progresses and acquires its more calcific phenotype (reviewed in [2]). If the role of HSP90 is related to the propagation of inflammation, it may have a different function in a heavily calcific environment as was present in these stenotic valves. However, further studies should focus on HSP90’s role in calcific VICs.
HSP90 mediates an ATP-dependent folding of its target proteins that are involved in many diverse cellular processes ranging from intracellular transportation to signal transduction (for general reviews on HSP90 biology, see [30]. The IPA analysis predicted that downregulation of HSP90 would be linked with Akt, p38 MAPK and ERK signalling pathways. We observed an increased activation of ERK1/2 and reduced activation of Akt-kinase in calcified valves whereas there was no change in the phosphorylation of p38 MAPK. Previously, Akt and ERK have been shown to mediate leptin induced osteoblast differentiation [31]. In addition, activation of Akt has been demonstrated to regulate calcium deposition [32] and ROS-induced expression of RUNX2 in human VICs [33]. Furthermore, a study with cultured porcine VICs revealed that ERK inhibition reduced gene expression of myofibroblastic and osteoblastic markers [34]. In our study, Akt and ERK exhibited distinctive phosphorylation patterns suggesting differential regulation of these signalling pathways in the heavily calcified valves.
In agreement with the previous proteomics studies of CAVD [10], [12], [16], we identified increased protein expression of the C9 component of the complement system and APCS. Upregulation of C9 is supported by the data by Helske et al. [35] who were the first investigators to show an activation of the complement system in stenotic valves. APCS, also known as pentraxin-2, is involved in amyloidosis but it is also present in human atherosclerotic lesions [36]. It exerts anti-inflammatory and antifibrotic properties e.g. inhibiting monocyte differentiation into proinflammatory macrophages [37]. APCS has also been implicated in several cardiovascular pathologies (reviewed in [3840]), and the role of this protein in CAVD should be investigated in detail. Our finding of decreased ANXA2 protein levels in calcified valves confirms the similar proteomic profiling findings of Matsumoto et al. [10]. In addition, ANXA1 and ANXA3 have been previously detected in calcified regions of aortic valves [10, 12]. Furthermore, Cui et al. [41] detected an up-regulation of annexins I, II, III, IV, V, VI, VII, and XI in calcifying VIC-derived matrix vesicles, highlighting the significance of the annexins in the calcification process. However, in contrast to a previous proteomic study [16], we observed increased transgelin (SM22) levels in stenotic valves. Since in both studies, transgelin was detected at different positions in the 2D gel, this discrepancy might be explained by the presence of different transgelin variants.
A major limitation of our work is small number of valves in our proteomic profiling study. This limited sample size raises issue that results do not represent heterogeneity of aortic stenosis patients. This might be the reason why we were not able to confirm with the Western blots the results of the proteomic data on Annexin II and galectin-1. Consequently, the generalizability of these results have to be confirmed in larger sample population. However, it is noteworthy that despite limited number patients, we also noticed increased expressions of CP9 component, APCS and ANXA2, in agreement with previous proteomic studies.
In our analysis, HSP90 expression correlated both with age and calcification of the valves. Since aortic valve calcification correlated with age of the patients, these correlations can be explained to be due the fact that aortic valve calcification is most prevalent in the elderly [42]. Thus, we cannot rule out that the changes in HSP90 protein expression are due to the aging and not aortic valve calcification. Furthermore, we did not confirm all the proteomic profiling results with another method. Finally, all the HSP90 associations are only predicted; the experimental validation of HSP90 interaction with Akt and ERK signaling pathways in aortic valve calcification will have to be the subject of some future work.

Conclusion

Our proteomic analysis identified seven dysregulated proteins in calcified valves when compared to control valves. These proteins may have roles in regulating processes associated with the pathogenesis of CAVD such as the immune response and calcification. Overall, our findings suggest novel insights into the mechanisms of aortic valve calcification and HSP90 may be a central signaling molecule in aortic valve calcification.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12872-019-01294-2.

Acknowledgments

We thank Dr. Ewen MacDonald for language revision and Dr. Pasi Ohtonen for his help with statistics.
This study was approved by the Research Ethics Committee of Oulu University Hospital (decision number 23/2008) and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Informed consent was obtained written from all individual participants included in the study.
Not applicable.

Competing interests

TP is currently an employee of MSD Finland.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Dweck MR, Boon NA, Newby DE. Calcific aortic stenosis: a disease of the valve and the myocardium. J Am Coll Cardiol. 2012;60(19):1854–63.PubMedCrossRef Dweck MR, Boon NA, Newby DE. Calcific aortic stenosis: a disease of the valve and the myocardium. J Am Coll Cardiol. 2012;60(19):1854–63.PubMedCrossRef
3.
Zurück zum Zitat Bosse Y, Miqdad A, Fournier D, Pibarot P, Mathieu P. Refining molecular pathways leading to calcific aortic valve stenosis by studying gene expression profile of Normal and calcified Stenotic human aortic valves. Circ Genet. 2009;2:489–98.CrossRef Bosse Y, Miqdad A, Fournier D, Pibarot P, Mathieu P. Refining molecular pathways leading to calcific aortic valve stenosis by studying gene expression profile of Normal and calcified Stenotic human aortic valves. Circ Genet. 2009;2:489–98.CrossRef
4.
Zurück zum Zitat Ohukainen P, Syvaranta S, Napankangas J, Rajamaki K, Taskinen P, Peltonen T, et al. MicroRNA-125b and chemokine CCL4 expression are associated with calcific aortic valve disease. Ann Med. 2015;47(5):423–9.PubMedCrossRef Ohukainen P, Syvaranta S, Napankangas J, Rajamaki K, Taskinen P, Peltonen T, et al. MicroRNA-125b and chemokine CCL4 expression are associated with calcific aortic valve disease. Ann Med. 2015;47(5):423–9.PubMedCrossRef
5.
Zurück zum Zitat Guauque-Olarte S, Messika-Zeitoun D, Droit A, Lamontagne M, Tremblay-Marchand J, Lavoie-Charland E, et al. Calcium signaling pathway genes RUNX2 and CACNA1C are associated with calcific aortic valve disease. Circ Genet. 2015;8(6):812–22.CrossRef Guauque-Olarte S, Messika-Zeitoun D, Droit A, Lamontagne M, Tremblay-Marchand J, Lavoie-Charland E, et al. Calcium signaling pathway genes RUNX2 and CACNA1C are associated with calcific aortic valve disease. Circ Genet. 2015;8(6):812–22.CrossRef
6.
Zurück zum Zitat Guauque-olarte XS, Droit A, Tremblay-marchand J, Gaudreault N, Kalavrouziotis D, Dagenais F, et al. RNA expression profile of calcified bicuspid , tricuspid , and normal human aortic valves by RNA sequencing. Physiol Genomics. 2016;48:749–61.PubMedPubMedCentralCrossRef Guauque-olarte XS, Droit A, Tremblay-marchand J, Gaudreault N, Kalavrouziotis D, Dagenais F, et al. RNA expression profile of calcified bicuspid , tricuspid , and normal human aortic valves by RNA sequencing. Physiol Genomics. 2016;48:749–61.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Coffey S, Williams MJA, Phillips LV, Galvin IF, Bunton RW, Jones GT. Integrated microRNA and messenger RNA analysis in aortic stenosis. Sci Rep. 2016;6(1):36904.PubMedPubMedCentralCrossRef Coffey S, Williams MJA, Phillips LV, Galvin IF, Bunton RW, Jones GT. Integrated microRNA and messenger RNA analysis in aortic stenosis. Sci Rep. 2016;6(1):36904.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Gil-Dones F, Martin-Rojas T, Lopez-Almodovar LF, De La Cuesta F, Darde VM, Alvarez-Llamas G, et al. Valvular Aortic Stenosis: A Proteomic Insight. Clin Med Insights Cardiol. 2010;4:CMC.S3884.CrossRef Gil-Dones F, Martin-Rojas T, Lopez-Almodovar LF, De La Cuesta F, Darde VM, Alvarez-Llamas G, et al. Valvular Aortic Stenosis: A Proteomic Insight. Clin Med Insights Cardiol. 2010;4:CMC.S3884.CrossRef
9.
Zurück zum Zitat Martin-Rojas T, Mourino-Alvarez L, Alonso-Orgaz S, Rosello-Lleti E, Calvo E, Lopez-Almodovar LF, et al. ITRAQ proteomic analysis of extracellular matrix remodeling in aortic valve disease. Sci Rep. 2015;5(1):1–12.CrossRef Martin-Rojas T, Mourino-Alvarez L, Alonso-Orgaz S, Rosello-Lleti E, Calvo E, Lopez-Almodovar LF, et al. ITRAQ proteomic analysis of extracellular matrix remodeling in aortic valve disease. Sci Rep. 2015;5(1):1–12.CrossRef
10.
Zurück zum Zitat Matsumoto K-II, Satoh K, Maniwa T, Araki A, Maruyama R, Oda T. Noticeable decreased expression of Tenascin-X in calcific aortic valves. Connect Tissue Res. 2012;53(6):460–8.PubMedCrossRef Matsumoto K-II, Satoh K, Maniwa T, Araki A, Maruyama R, Oda T. Noticeable decreased expression of Tenascin-X in calcific aortic valves. Connect Tissue Res. 2012;53(6):460–8.PubMedCrossRef
11.
Zurück zum Zitat Mourino-Alvarez L, Iloro I, De La Cuesta F, Azkargorta M, Sastre-Oliva T, Escobes I, et al. MALDI-imaging mass spectrometry: a step forward in the anatomopathological characterization of stenotic aortic valve tissue. Sci Rep. 2016;6:1–12.CrossRef Mourino-Alvarez L, Iloro I, De La Cuesta F, Azkargorta M, Sastre-Oliva T, Escobes I, et al. MALDI-imaging mass spectrometry: a step forward in the anatomopathological characterization of stenotic aortic valve tissue. Sci Rep. 2016;6:1–12.CrossRef
12.
Zurück zum Zitat Schlotter F, Halu A, Goto S, Blaser MC, Body SC, Lee LH, et al. Spatiotemporal multi-Omics mapping generates a molecular atlas of the aortic valve and reveals networks driving disease. Circulation. 2018;138(4):377–93.PubMedPubMedCentralCrossRef Schlotter F, Halu A, Goto S, Blaser MC, Body SC, Lee LH, et al. Spatiotemporal multi-Omics mapping generates a molecular atlas of the aortic valve and reveals networks driving disease. Circulation. 2018;138(4):377–93.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Kokudo T, Suzuki Y, Yoshimatsu Y, Yamazaki T, Watabe T, Miyazono K. Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells. J Cell Sci. 2008;121(Pt 20):3317–24.PubMedCrossRef Kokudo T, Suzuki Y, Yoshimatsu Y, Yamazaki T, Watabe T, Miyazono K. Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells. J Cell Sci. 2008;121(Pt 20):3317–24.PubMedCrossRef
14.
Zurück zum Zitat Suzuki H, Chikada M, Yokoyama MK, Kurokawa MS, Ando T, Furukawa H, et al. Aberrant glycosylation of Lumican in aortic valve stenosis revealed by proteomic analysis. Int Heart J. 2016;57(1):104–11.PubMedCrossRef Suzuki H, Chikada M, Yokoyama MK, Kurokawa MS, Ando T, Furukawa H, et al. Aberrant glycosylation of Lumican in aortic valve stenosis revealed by proteomic analysis. Int Heart J. 2016;57(1):104–11.PubMedCrossRef
15.
Zurück zum Zitat Trindade F, Ferreira R, Magalhães B, Leite-Moreira A, Falcão-Pires I, Vitorino R. How to use and integrate bioinformatics tools to compare proteomic data from distinct conditions? A tutorial using the pathological similarities between aortic valve stenosis and coronary artery disease as a case-study. J Proteome. 2018;171:37–52.CrossRef Trindade F, Ferreira R, Magalhães B, Leite-Moreira A, Falcão-Pires I, Vitorino R. How to use and integrate bioinformatics tools to compare proteomic data from distinct conditions? A tutorial using the pathological similarities between aortic valve stenosis and coronary artery disease as a case-study. J Proteome. 2018;171:37–52.CrossRef
16.
Zurück zum Zitat Martín-Rojas T, Gil-Dones F, Lopez-Almodovar LF, Padial LR, Vivanco F, Barderas MG, et al. Proteomic profile of human aortic stenosis: insights into the degenerative process. J Proteome Res. 2012;11(3):1537–50.PubMedCrossRef Martín-Rojas T, Gil-Dones F, Lopez-Almodovar LF, Padial LR, Vivanco F, Barderas MG, et al. Proteomic profile of human aortic stenosis: insights into the degenerative process. J Proteome Res. 2012;11(3):1537–50.PubMedCrossRef
17.
Zurück zum Zitat Pohjolainen V, Mustonen E, Taskinen P, Näpänkangas J, Leskinen H, Ohukainen P, et al. Increased thrombospondin-2 in human fibrosclerotic and stenotic aortic valves. Atherosclerosis. 2012;220(1):66–71.PubMedCrossRef Pohjolainen V, Mustonen E, Taskinen P, Näpänkangas J, Leskinen H, Ohukainen P, et al. Increased thrombospondin-2 in human fibrosclerotic and stenotic aortic valves. Atherosclerosis. 2012;220(1):66–71.PubMedCrossRef
18.
Zurück zum Zitat Näpänkangas J, Ohtonen P, Ohukainen P, Weisell J, Väisänen T, Peltonen T, et al. Increased mesenchymal podoplanin expression is associated with calcification in aortic valves. Cardiovasc Pathol. 2019;39:30–7.PubMedCrossRef Näpänkangas J, Ohtonen P, Ohukainen P, Weisell J, Väisänen T, Peltonen T, et al. Increased mesenchymal podoplanin expression is associated with calcification in aortic valves. Cardiovasc Pathol. 2019;39:30–7.PubMedCrossRef
19.
Zurück zum Zitat Ohlmeier S, Mazur W, Salmenkivi K, Myllärniemi M, Bergmann U, Kinnula VL. Proteomic studies on receptor for advanced glycation end product variants in idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease. Proteomics Clin Appl. 2010;4(1):97–105.PubMedCrossRef Ohlmeier S, Mazur W, Salmenkivi K, Myllärniemi M, Bergmann U, Kinnula VL. Proteomic studies on receptor for advanced glycation end product variants in idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease. Proteomics Clin Appl. 2010;4(1):97–105.PubMedCrossRef
21.
Zurück zum Zitat Lu X, Kakkar V. The role of heat shock protein (HSP) in atherosclerosis: pathophysiology and clinical opportunities. Curr Med Chem. 2010;17(10):957–73.PubMedCrossRef Lu X, Kakkar V. The role of heat shock protein (HSP) in atherosclerosis: pathophysiology and clinical opportunities. Curr Med Chem. 2010;17(10):957–73.PubMedCrossRef
22.
Zurück zum Zitat Reventun P, Alique M, Cuadrado I, Márquez S, Toro R, Zaragoza C, et al. INOS-derived nitric oxide induces integrin-linked kinase Endocytic lysosome-mediated degradation in the vascular endothelium. Arterioscler Thromb Vasc Biol. 2017;37(7):1272–81.PubMedCrossRef Reventun P, Alique M, Cuadrado I, Márquez S, Toro R, Zaragoza C, et al. INOS-derived nitric oxide induces integrin-linked kinase Endocytic lysosome-mediated degradation in the vascular endothelium. Arterioscler Thromb Vasc Biol. 2017;37(7):1272–81.PubMedCrossRef
23.
Zurück zum Zitat Ou Z-J, Chen J, Dai W-P, Liu X, Yang Y-K, Li Y, et al. 25-Hydroxycholesterol impairs endothelial function and vasodilation by uncoupling and inhibiting endothelial nitric oxide synthase. Am J Physiol Metab. 2016;311(4):E781–90. Ou Z-J, Chen J, Dai W-P, Liu X, Yang Y-K, Li Y, et al. 25-Hydroxycholesterol impairs endothelial function and vasodilation by uncoupling and inhibiting endothelial nitric oxide synthase. Am J Physiol Metab. 2016;311(4):E781–90.
24.
Zurück zum Zitat Yan FX, Li HM, Li SX, He SH, Dai WP, Li Y, et al. The oxidized phospholipid POVPC impairs endothelial function and vasodilation via uncoupling endothelial nitric oxide synthase. J Mol Cell Cardiol. 2017;112:40–8.PubMedCrossRef Yan FX, Li HM, Li SX, He SH, Dai WP, Li Y, et al. The oxidized phospholipid POVPC impairs endothelial function and vasodilation via uncoupling endothelial nitric oxide synthase. J Mol Cell Cardiol. 2017;112:40–8.PubMedCrossRef
25.
Zurück zum Zitat Miller JD, Chu Y, Brooks RM, Richenbacher WE, Peña-Silva R, Heistad DD. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic Valvular stenosis in humans. J Am Coll Cardiol. 2008;52(10):843–50.PubMedPubMedCentralCrossRef Miller JD, Chu Y, Brooks RM, Richenbacher WE, Peña-Silva R, Heistad DD. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic Valvular stenosis in humans. J Am Coll Cardiol. 2008;52(10):843–50.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Businaro R, Profumo E, Tagliani A, Buttari B, Leone S, Amati GD, et al. Heat-shock protein 90 : A novel autoantigen in human carotid atherosclerosis. Atherosclerosis. 2009;207:74–83.PubMedCrossRef Businaro R, Profumo E, Tagliani A, Buttari B, Leone S, Amati GD, et al. Heat-shock protein 90 : A novel autoantigen in human carotid atherosclerosis. Atherosclerosis. 2009;207:74–83.PubMedCrossRef
27.
Zurück zum Zitat Madrigal-Matute J, López-Franco O, Blanco-colio LM, Muñoz-García B, Ramos-Mozo P, Ortega L, et al. Heat shock protein 90 inhibitors attenuate inflammatory responses in atherosclerosis. Cardiovasc Res. 2010;86(2):330–7.PubMedCrossRef Madrigal-Matute J, López-Franco O, Blanco-colio LM, Muñoz-García B, Ramos-Mozo P, Ortega L, et al. Heat shock protein 90 inhibitors attenuate inflammatory responses in atherosclerosis. Cardiovasc Res. 2010;86(2):330–7.PubMedCrossRef
28.
Zurück zum Zitat Lazaro I, Oguiza A, Recio C, Lopez-Sanz L, Bernal S, Egido J, et al. Interplay between HSP90 and Nrf2 pathways in diabetes-associated atherosclerosis. Clin Investig Arterioscler. 2017;29(2):51–9.PubMed Lazaro I, Oguiza A, Recio C, Lopez-Sanz L, Bernal S, Egido J, et al. Interplay between HSP90 and Nrf2 pathways in diabetes-associated atherosclerosis. Clin Investig Arterioscler. 2017;29(2):51–9.PubMed
29.
Zurück zum Zitat Madrigal-Matute J, Fernandez-Garcia CE, Gomez-Guerrero C, Lopez-Franco O, Muñoz-Garcia B, Egido J, et al. HSP90 inhibition by 17-DMAG attenuates oxidative stress in experimental atherosclerosis. Cardiovasc Res. 2012;95(1):116–23.PubMedCrossRef Madrigal-Matute J, Fernandez-Garcia CE, Gomez-Guerrero C, Lopez-Franco O, Muñoz-Garcia B, Egido J, et al. HSP90 inhibition by 17-DMAG attenuates oxidative stress in experimental atherosclerosis. Cardiovasc Res. 2012;95(1):116–23.PubMedCrossRef
30.
Zurück zum Zitat Schopf FH, Biebl MM, Buchner J. The HSP90 chaperone machinery. Nat Rev Mol Cell Biol. 2017;18(6):345–60.PubMedCrossRef Schopf FH, Biebl MM, Buchner J. The HSP90 chaperone machinery. Nat Rev Mol Cell Biol. 2017;18(6):345–60.PubMedCrossRef
31.
Zurück zum Zitat Rosa M, Paris C, Sottejeau Y, Corseaux D, Robin E, Tagzirt M, et al. Leptin induces osteoblast differentiation of human valvular interstitial cells via the Akt and ERK pathways. Acta Diabetol. 2017;54(6):551–60.PubMedCrossRef Rosa M, Paris C, Sottejeau Y, Corseaux D, Robin E, Tagzirt M, et al. Leptin induces osteoblast differentiation of human valvular interstitial cells via the Akt and ERK pathways. Acta Diabetol. 2017;54(6):551–60.PubMedCrossRef
32.
Zurück zum Zitat Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, et al. Osteopontin-CD44v6 interaction mediates calcium deposition via phospho-akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arterioscler Thromb Vasc Biol. 2014;34(9):2086–94.PubMedPubMedCentralCrossRef Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, et al. Osteopontin-CD44v6 interaction mediates calcium deposition via phospho-akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arterioscler Thromb Vasc Biol. 2014;34(9):2086–94.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Branchetti E, Sainger R, Poggio P, Grau JB, Patterson-Fortin J, Bavaria JE, et al. Antioxidant enzymes reduce DNA damage and early activation of valvular interstitial cells in aortic valve sclerosis. Arterioscler Thromb Vasc Biol. 2013;33(2):66–74.CrossRef Branchetti E, Sainger R, Poggio P, Grau JB, Patterson-Fortin J, Bavaria JE, et al. Antioxidant enzymes reduce DNA damage and early activation of valvular interstitial cells in aortic valve sclerosis. Arterioscler Thromb Vasc Biol. 2013;33(2):66–74.CrossRef
34.
Zurück zum Zitat Gu X, Masters KS. Role of the MAPK/ERK pathway in valvular interstitial cell calcification. Am J Physiol Circ Physiol. 2009;296(6):H1748–57.CrossRef Gu X, Masters KS. Role of the MAPK/ERK pathway in valvular interstitial cell calcification. Am J Physiol Circ Physiol. 2009;296(6):H1748–57.CrossRef
35.
Zurück zum Zitat Helske S, Oksjoki R, Lindstedt KA, Lommi J, Turto H, Werkkala K, et al. Complement system is activated in stenotic aortic valves. Atherosclerosis. 2008;196(1):190–200.PubMedCrossRef Helske S, Oksjoki R, Lindstedt KA, Lommi J, Turto H, Werkkala K, et al. Complement system is activated in stenotic aortic valves. Atherosclerosis. 2008;196(1):190–200.PubMedCrossRef
36.
Zurück zum Zitat Li XA, Hatanaka K, Ishibashi-Ueda H, Yutani C, Yamamoto A. Characterization of serum amyloid P component from human aortic atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 1995;15(2):252–7.PubMedCrossRef Li XA, Hatanaka K, Ishibashi-Ueda H, Yutani C, Yamamoto A. Characterization of serum amyloid P component from human aortic atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 1995;15(2):252–7.PubMedCrossRef
37.
Zurück zum Zitat Moreira AP, Cavassani KA, Hullinger R, Rosada RS, Fong DJ, Murray L, et al. Serum amyloid P attenuates M2 macrophage activation and protects against fungal spore–induced allergic airway disease. J Allergy Clin Immunol. 2010;126(4):712–21 e7.PubMedCrossRef Moreira AP, Cavassani KA, Hullinger R, Rosada RS, Fong DJ, Murray L, et al. Serum amyloid P attenuates M2 macrophage activation and protects against fungal spore–induced allergic airway disease. J Allergy Clin Immunol. 2010;126(4):712–21 e7.PubMedCrossRef
38.
Zurück zum Zitat Norata GD, Garlanda C, Catapano AL. The long pentraxin PTX3: a modulator of the immunoinflammatory response in atherosclerosis and cardiovascular diseases. Trends Cardiovasc Med. 2010;20:35–40.PubMedCrossRef Norata GD, Garlanda C, Catapano AL. The long pentraxin PTX3: a modulator of the immunoinflammatory response in atherosclerosis and cardiovascular diseases. Trends Cardiovasc Med. 2010;20:35–40.PubMedCrossRef
39.
Zurück zum Zitat Vilahur G, Badimon L. Biological actions of pentraxins. Vasc Pharmacol. 2015;73:38–44.CrossRef Vilahur G, Badimon L. Biological actions of pentraxins. Vasc Pharmacol. 2015;73:38–44.CrossRef
40.
Zurück zum Zitat Xi D, Luo T, Xiong H, Liu J, Lu H, Li M, et al. SAP: structure, function, and its roles in immune-related diseases. Int J Cardiol. 2015;187:20–6.PubMedCrossRef Xi D, Luo T, Xiong H, Liu J, Lu H, Li M, et al. SAP: structure, function, and its roles in immune-related diseases. Int J Cardiol. 2015;187:20–6.PubMedCrossRef
41.
Zurück zum Zitat Cui L, Rashdan NA, Zhu D, Milne EM, Ajuh P, Milne G, et al. End stage renal disease-induced hypercalcemia may promote aortic valve calcification via Annexin VI enrichment of valve interstitial cell derived-matrix vesicles. J Cell Physiol. 2017;232(11):2985–95.PubMedPubMedCentralCrossRef Cui L, Rashdan NA, Zhu D, Milne EM, Ajuh P, Milne G, et al. End stage renal disease-induced hypercalcemia may promote aortic valve calcification via Annexin VI enrichment of valve interstitial cell derived-matrix vesicles. J Cell Physiol. 2017;232(11):2985–95.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Coffey S, Cox B, Williams MJA. The prevalence, incidence, progression, and risks of aortic valve sclerosis: a systematic review and meta-analysis. J Am Coll Cardiol. 2014;63(25):2852–61.PubMedCrossRef Coffey S, Cox B, Williams MJA. The prevalence, incidence, progression, and risks of aortic valve sclerosis: a systematic review and meta-analysis. J Am Coll Cardiol. 2014;63(25):2852–61.PubMedCrossRef
Metadaten
Titel
Heat shock protein 90 is downregulated in calcific aortic valve disease
verfasst von
Jonna Weisell
Pauli Ohukainen
Juha Näpänkangas
Steffen Ohlmeier
Ulrich Bergmann
Tuomas Peltonen
Panu Taskinen
Heikki Ruskoaho
Jaana Rysä
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Cardiovascular Disorders / Ausgabe 1/2019
Elektronische ISSN: 1471-2261
DOI
https://doi.org/10.1186/s12872-019-01294-2

Weitere Artikel der Ausgabe 1/2019

BMC Cardiovascular Disorders 1/2019 Zur Ausgabe

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

TAVI versus Klappenchirurgie: Neue Vergleichsstudie sorgt für Erstaunen

21.05.2024 TAVI Nachrichten

Bei schwerer Aortenstenose und obstruktiver KHK empfehlen die Leitlinien derzeit eine chirurgische Kombi-Behandlung aus Klappenersatz plus Bypass-OP. Diese Empfehlung wird allerdings jetzt durch eine aktuelle Studie infrage gestellt – mit überraschender Deutlichkeit.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.