Next Article in Journal
Interprofessional Collaboration and Diabetes Management in Primary Care: A Systematic Review and Meta-Analysis of Patient-Reported Outcomes
Next Article in Special Issue
Fasting Proinsulin Independently Predicts Incident Type 2 Diabetes in the General Population
Previous Article in Journal
Genetic and Clinical Factors Associated with Olokizumab Treatment in Russian Patients with Rheumatoid Arthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prioritization of Candidate Biomarkers for Degenerative Aortic Stenosis through a Systems Biology-Based In-Silico Approach

by
Nerea Corbacho-Alonso
1,†,
Tamara Sastre-Oliva
1,†,
Cecilia Corros
2,
Teresa Tejerina
3,
Jorge Solis
2,4,5,
Luis F. López-Almodovar
6,
Luis R. Padial
7,
Laura Mourino-Alvarez
1,‡ and
Maria G. Barderas
1,*,‡
1
Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos, SESCAM, 45071 Toledo, Spain
2
Department of Cardiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
3
Department of Pharmacology, School of Medicine, Universidad Complutense, 28040 Madrid, Spain
4
AtriaClinic, 28047 Madrid, Spain
5
Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
6
Cardiac Surgery, Hospital Virgen de la Salud, SESCAM, 45004 Toledo, Spain
7
Department of cardiology, Hospital Virgen de la Salud, SESCAM, 45004 Toledo, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Submission received: 4 March 2022 / Revised: 4 April 2022 / Accepted: 13 April 2022 / Published: 15 April 2022
(This article belongs to the Special Issue Novel Biomarkers and Diagnostic Methods of Cardiovascular Disease)

Abstract

:
Degenerative aortic stenosis is the most common valve disease in the elderly and is usually confirmed at an advanced stage when the only treatment is surgery. This work is focused on the study of previously defined biomarkers through systems biology and artificial neuronal networks to understand their potential role within aortic stenosis. The goal was generating a molecular panel of biomarkers to ensure an accurate diagnosis, risk stratification, and follow-up of aortic stenosis patients. We used in silico studies to combine and re-analyze the results of our previous studies and, with information from multiple databases, established a mathematical model. After this, we prioritized two proteins related to endoplasmic reticulum stress, thrombospondin-1 and endoplasmin, which have not been previously validated as markers for aortic stenosis, and analyzed them in a cell model and in plasma from human subjects. Large-scale bioinformatics tools allow us to extract the most significant results after using high throughput analytical techniques. Our results could help to prevent the development of aortic stenosis and open the possibility of a future strategy based on more specific therapies.

1. Introduction

Aortic stenosis (AS) is defined as an abnormal narrowing of the aortic valve (AV) opening, which blocks blood flow from the left ventricle into the aorta and, consequently, to the rest of the organism. The most common valve disease in the elderly is calcific or degenerative AS, which remains the main cause of AV replacement in developed countries [1,2,3].
AS progresses from an initial stage of aortic sclerosis, with a thickening and stiffening of the AV, to severe calcific stenosis. Unfortunately, the disease is usually diagnosed at an advanced stage since the symptoms are usually insidious at the onset. The appearance of its most common symptoms, such as dyspnea, angina, and syncope, predict a rapid deterioration of left ventricular function and the development of heart failure, potentially provoking the death of the patient if the pathology progresses. The only effective treatment to avoid this and improve survival is AV replacement, either surgically or via a transcatheter, which makes the management of these patients difficult [4,5]. As surgery should only be performed when the risks of AS outweigh those of the intervention, it is important to define different indicators to stratify the risk and timing of such interventions [6]. Early interventions may expose the patient to an unnecessary risk of complications, including living with a prosthetic valve and lifetime anticoagulation therapy, whereas an excessive delay may produce irreversible damage to the myocardium [7].
Ideally, the assessment of the global risk requires the integration of multiple biomarkers (including clinical factors) and an evaluation of molecular indicators belonging to independent pathways [8,9]. In an effort to identify suitable markers, large-scale analysis or -omics studies are powerful tools that enable panels of biomarkers to be defined that may later be assessed in patient cohorts. Combining and re-analyzing the results of multiple -omics studies through a systems biology approach allow AS treatment to be considered as a holistic process without applying a targeted hypothesis. As such, here, we used in silico studies that enabled us to combine results from our previous proteomics studies [10,11,12,13,14] with information from multiple databases, establishing a mathematical model thanks to the use of complex systems biology algorithms. Through this roadmap, we prioritized two proteins related to endoplasmic reticulum (ER) stress that have not been previously validated as markers for AS and analyzed them in a cell model as well as in plasma samples from human subjects.

2. Materials and Methods

2.1. Molecular Characterization of AS

For the molecular characterization of AS, as well as the generation of mathematical models and candidate prioritization, an exhaustive bibliographic search of the molecular and cellular processes involved in the disease allowed the main pathophysiological events in AS (motives) to be identified and novel candidates to be defined (Figure 1). In this workflow, a search for reviews on the molecular pathogenesis and pathophysiology of the condition was performed in the PubMed database on 8 April 2019. The specific search was: (“degenerative aortic stenosis” [Title] OR “aortic stenosis” [Title] OR “calcific aortic valve disease”[Title] OR “calcific aortic stenosis” [Title]) AND (pathogenesis [Title/Abstract] OR pathophysiology [Title/Abstract] OR molecular [Title/Abstract]) and Review [ptyp]. Additionally, if the evidence of the implication of a candidate in the condition was judged not consistent enough to be assigned as an effector, an additional PubMed search was performed specifically for the candidate, including all the protein names according to UniProtKB.

2.2. Generation of the Mathematical Models

To generate systems biology-based mathematical models, a biological map was built around the molecular processes and key proteins defined during the characterization of AS. The map was extended by adding knowledge-oriented connectivity layers (i.e., protein-to-protein interactions), including physical interactions and modulations, signaling and metabolic relationships, and the regulation of gene expression. Data were obtained from public and private databases (KEGG [15], BioGRID [16], IntAct [17], REACTOME [18], TRRUST [19], and HPRD [20]) and from manual curation of the relevant scientific literature. The models were then trained with a proprietary “Truth Table” containing publicly available data. The models must be able to weight the relative value of each protein (nodes), and since the number of links is very high, the number of parameters that must be resolved increases exponentially. The use of artificial intelligence technologies to model complex network behavior, including: graph theory and statistical pattern recognition technologies; genetic algorithms; artificial neural networks; dimensionality reduction techniques; and stochastic methods such assimulated annealing, Monte Carlo, etc.

2.3. Candidate Prioritization

The first step in candidate prioritization was the confection of a list of 126 proteins based on our previous studies (Table 1). Once the mathematical models had been generated, their predictive power can be exploited through an artificial neural network (ANN) strategy [21] in order to prioritize the different proteins and protein combinations based on their potential relationships with defined AS related processes (motives). Specifically, the potential relationship between each differentially expressed protein and the protein sets defining each AS motive (process) of interest was predicted through ANNs. This approach attempts to find the shortest distance between the protein sets, thereby generating a list of differentially expressed proteins ordered according to their association with the selected disease or pathway.
The ANNs evaluate the relationships among the protein sets or regions within the network, providing a predictive score that quantifies the probability a functional relationship exists between the network regions evaluated. Each score is associated with a p-value that describes the probability of the result being a true positive result. Three categories were used to group the proteins analyzed according to the predicted relationship value (Table 2): strongly related proteins (including the “Very high” group with a predicted ANN value ≥92% (p < 0.01), the “High” group with a predicted ANN value <92–≥78% (p values between 0.01 and 0.05), and the “Medium-high” group with a predicted ANN value <78–≥63% (p values between 0.05 and 0.15)); moderately related proteins (the “Medium” group with a predicted ANN value <63–≥38 (p values between 0.15 and 0.25)); and proteins with a low or no relationship (the “Low” group predicted ANN value <38% (p > 0.25)).
This classification defined those proteins predicted to have a:
  • “strong relationship” with the processes under study, with very high, high or, medium-high predicted relationships with any of the sub-processes used in the characterization, and considered to be good candidates;
  • “medium relationship” with the processes under study, with at least a medium predicted relationship with any of the sub-processes used in the characterization;
  • “low or no relationship” with the processes under study and with a weak predicted relationship with all the sub-processes used in the characterization.

2.4. Cell Culture and Differentiation

Human cardiac valvular interstitial cells (HAVICs: Innoprot, P10462) were used in this study, cells isolated from heart valves, cryopreserved in primary cultures, and guaranteed to further expand for 10 population doublings under the conditions indicated in the data sheet. HAVICs were cultured in Fibroblast Medium-2 (FM-2: Innoprot), designed for optimal growth of normal human cardiac fibroblasts invitro, and containing essential and non-essential amino acids, vitamins, organic and inorganic compounds, hormones, growth factors, trace minerals, and a low concentration of fetal bovine serum (FBS, 5%). For the experiments, HAVICs were used at passage 5, and during the previous passage 4, the medium was replaced by a special medium for fibroblasts (FIBm) that favors a quiescent phenotype: Dulbecco’s Modified Eagle Medium (DMEM: Hyclone) supplemented with 2% heat-inactivated FBS, 150 U/mL penicillin-streptomycin, 2 mM L-glutamine, 10 ng/mL fibroblast growth factor (FGF-2), and 50 ng/mL insulin [22]. In the experiments, the cells were cultured for up 14 days in two different media, FIBm and osteogenic medium, to induce the osteogenic differentiation of the HAVICs (OSTm—FIBm supplemented with 50 µg/mL ascorbic acid, 10 mM β-glycerophosphate, and 100 nM dexamethasone) [23].

2.5. Alizarin Red Staining

The cells were washed with PBS, fixed with 4% paraformaldehyde for 15 min, and then incubated for 10 min with alizarin red S (Sigma Aldrich, St. Louis, MO, USA) [24]. After washing with deionized water, calcium deposition was visualized under an Olympus IX83 inverted microscope, capturing 49 images per well, and analyzing this with Scan^R software. These experiments were performed in triplicate.

2.6. Patient Selection and Plasma Extraction

Peripheral blood samples were collected from control subjects (n = 18) and from patients with severe AS (n = 18) who underwent follow-up at the Hospital 12 de Octubre (Madrid, Spain) and/or Hospital Virgen de la Salud (Toledo, Spain) from November 2018 to December 2019. All patients had severe AS diagnosed with two-dimensional echocardiography/doppler and were at least 50 years old. Control subjects were also subjected to echocardiographic control to avoid the presence of valve disease. Samples from patients with a severe morbidity (ischemic heart disease with ventricular dysfunction, end-stage chronic kidney disease), bicuspid AV, a family or personal history of aortopathy, rheumatic valve disease, and ≥moderate mitral valve disease were excluded from the study. Importantly, subjects were selected to avoid significant differences between the groups in terms of the main cardiovascular risk factors: gender, obesity, hypertension, dyslipidemia, and diabetes. Clinical characteristics of both groups are shown in Table 3.Blood samples (28 mL) were collected in tubes containing EDTA and centrifuged at 1125× g for 15 min, immediately freezing the resulting supernatant at −80 °C until analysis.
This study was carried out in accordance with the recommendations of the Helsinki Declaration, and it was approved by the Ethics Committee at the participant hospitals (approval reference numbers: 18/315 and 07/036). Signed informed consent was obtained from all subjects prior to their inclusion on the study.

2.7. Western Blotting

HAVICs were trypsinized and homogenized in lysis buffer containing protease inhibitors on day 7 or 14 of treatment [25]. The protein concentration of both the cell extracts and plasma samples was determined by the Bradford–Lowry method (Bio-Rad protein assay) [26]. Equal amounts of protein from the samples (10 µg for cell extracts and 25 µg for plasma) were resolved by SDS–PAGE in a Bio-Rad Miniprotean II electrophoresis cell run at a constant current of 25 mA/gel. After electrophoresis, the proteins were transferred to a nitrocellulose membrane under a constant voltage of 20V for 30 min, and the membranes were stained with Ponceau S to guarantee an equal amount of protein was loaded for each patient. Subsequently, the membranes were blocked for 1 h with PBS-Tween 20 (PBS-T) containing 7.5% non-fat dry milk and incubated overnight with the primary antibody in PBS-T with 5% non-fat dry milk. The primary antibodies used were antisera against thrombospondin-1 (THBS, 1/100, Abcam ab85762, Cambridge, UK), endoplasmin (GRP94, 1/100, Abcam ab3674, Cambridge, UK), and α-smooth muscle actin (SMA, 1/100, Abcam ab7817, Cambridge, UK). After washing, the membranes were incubated with a specific HRP-conjugated secondary antibody in PBS-T containing 5% non-fat dry milk, and antibody binding was detected by enhanced chemiluminescence (ECL: GE Healthcare), according to the manufacturers’ instructions. Densitometry was performed with the ImageQuantTL software (GE Healthcare). We used Ponceau S stain images to normalize Western blot data from cell cultures, a more consistent way of normalizing data than using a single house-keeping protein [27].

2.8. Statistics

Dichotomous variables are expressed as prevalence in number and percent, and continuous variables, such as age, are expressed as mean ± s.d. The normality of the data was assessed with the Kolmogorov–Smirnov test. Two-tailed Student t-tests were employed to calculate the differences between the groups and a general linear model adjusted for age was used to avoid the effect of age as confounder. All statistical analyses were performed using SPSS 15.0 for Windows software (SPSS Inc., Chicago, IL, USA). Statistical significance was accepted at p < 0.05.

3. Results

3.1. Molecular Motives of AS

After the bibliographic review of AS, eight pathophysiological processes or ‘motives’ were identified as being associated with this condition. These motives can be classified at two levels depending on their involvement in the pathology: causative, motives that are directly related to the onset or pathophysiology of the condition characterized; and symptomatic, motives that are a consequence of the pathology. Lipoprotein accumulation, inflammation, oxidative stress, endothelial dysfunction, oxidative stress, and the renin–angiotensin–aldosterone (RAA) system are all causatives motives in AS, whereashypertrophy and myocardial fibrosis are symptomatic. Calcification is included at both levels, as a cause and manifestation of the disease. The results of this search were thoroughly reviewed to identify protein/gene candidates that might be condition effectors, i.e., proteins whose activity (or lack thereof) is functionally associated witheach motive. A total of 168 proteins were defined as effectors of particular processes in AS or to AS in general (Table S2).

3.2. Candidate Prioritization

The mechanistic ANN ranking enabled the list of 126 proteins to be classified based on their predicted functional or mechanistic relationship. The ANN analysis indicated that, of the 126 candidate proteins, 61 (48.41%) were predicted to have a strong relationship with at least one process involved in degenerative AS or with degenerative AS in general (Table 4). Of these, 20 proteins are degenerative AS effectors already described in the molecular characterization of the disease, whereasthe remaining 41 proteins were not included in this characterization. Moreover, 32 of the 61 proteins are associated with more than one of the processes. The list of all proteins analyzed and the ANN score or relationship predicted values to the entire disease are presented in Supplementary Table S3. Whether the proteins are effectors of the disease is also displayed.
Moreover, there were 22 proteins strongly related to three or more of the processes evaluated, including general AS characterization (Table 5). Among these, eight proteins were not present in the molecular characterization: endoplasmin, decorin, alpha-2-macroglobulin, serum albumin, transthyretin, clusterin, and Thbs1.

3.3. Confirmation of the Prioritized Candidates in a Cell Model and Plasma

Protein extracts from HAVICs were analyzed at 7 and 14 days of treatment, when higher levels of alizarin red and α-SMA were evident in the treated cells, confirming their osteoblastic differentiation (Figure 2a,b). There was also more total Thbs1 (day 7 p-value= 0.002; day 14 p-value = 0.045) and endoplasmin (day 7 p-value = 0.014; day 14 p-value = 0.038) in these HAVICs maintained in OSTm (Figure 2c,d). We found two different bands in the Western blot of Thbs1, one higher than 250 KDa (day 7, FIB medium = 0.681 ± 0.088, osteogenic medium = 1.268 ± 0.192, p-value = 0.009; and day 14, FIB medium = 0.862 ± 0.048, osteogenic medium = 2.527 ± 0.241, p-value = 0.005) and one at 200 KDa (day 7, FIB medium = 3.451 ± 0.458, osteogenic medium = 6.828 ± 0.968, p-value = 0.005; and day 14, FIB medium = 5.679 ± 0.467, osteogenic medium = 7.268 ± 1.708, p-value = 0.195), whichwere also analyzed separately.
The alterations to Thbs1 and endoplasmin were confirmed in Western blots of plasma from control subjects and severe AS patients. Consequently, we found lower levels of total Thbs1 (p-value = 0.007; age-adjusted p-value = 0.017) and endoplasmin (p-value = 0.024; age-adjusted p-value = 0.021) in the AS patients in both non-adjusted and age-adjusted model (Figure 3).

4. Discussion

Currently, there area large amount of data generated by high-throughput techniques such asproteomics, such that the interpretation and analysis of these data is becoming a complicated task. To overcome this challenge, systems biology approaches are essential, as they bring together all this information along with newly generated data. Systems biology uses a network-based approach to model complex biological systems and processes, employing mathematical models and computational approaches. These strategies allow new properties or mechanisms involved in a disease to be discovered that were not previously evident with traditional reductionist approaches [28].
In this work, systems biology approaches were used to evaluate and prioritize potential AS candidate biomarkers based on their association with the disease and their mechanistic implications. This ANN strategy provides a specific predictive value to the candidate markers identified, giving an idea of the probability that a relationship exists between each differentially expressed protein and the processes studied. This value is based on validating the predictive capacity of these models through the information available in the databases.
Our initial general characterization of AS identified six causative (calcification, lipoprotein accumulation, inflammation, oxidative stress, endothelial dysfunction, and RAA system) and two manifestation motives (hypertrophy and myocardial fibrosis). During AV degeneration, the causative motives are tightly related. In the initial phase, endothelial dysfunction occurs due to classic cardiovascular risk factors, such as advanced age, hypertension, smoking, diabetes mellitus, and the presence of high concentrations of cholesterol in the blood [29]. As a consequence, the permeability of the area increases, allowing the passage of molecules that leads to lipoprotein accumulation and inflammatory cytokine release. These lipids and cytokines further contribute to endothelial damage, amplifying the inflammatory process. In addition, this chronic inflammation causes oxidative stress, which, in turn, drives gene expression involved in the inflammatory process, thereby establishing a noxious vicious circle whereby inflammation causes oxidative stress and vice versa [30]. This activation of the immune system will provoke the differentiation of valvular interstitial cells from fibroblast to myofibroblasts, which will, in turn, develop angiogenic activity and produce a matrix of metalloproteins. The pro-inflammatory cytokines will induce the differentiation of a subgroup of myofibroblasts to osteoblasts, which leads to severe calcification and valve dysfunction [3,31]. Likewise, the RAA system plays an important role in the pathogenesis of AS. Its activation enhances collagen I and III mRNA expression, leading to myocardial fibrosis [32], and it is associated with left ventricle pressure overload. The combination of valve obstruction and elevated blood pressure imposes a high hemodynamic load on the left ventricle that leads to both left ventricle hypertrophy and myocardial fibrosis, two motives manifested in the general characterization of AS [33,34,35].
After the molecular characterization, and according to the mechanistic ANN ranking analysis, 22 proteins were found to be strongly related tothree or more of the processes evaluated. Of those 22 proteins, we highlight 8 of these that were not defined as effectors during the molecular characterization: decorin, alpha-2-macroglobulin, serum albumin, transthyretin, clusterin, endoplasmin and Thbs1. This study focused specifically on endoplasmin and Thbs1, as they are located in the ER. The ER is a major site for the regulation of calcium and lipid homeostasis, and it is essential for protein synthesis, folding, and transportation. When the influx of unfolded proteins to the ER exceeds its capacity to fold them correctly, unfolded and misfolded proteins accumulate in the ER lumen. This build-up creates a state defined as ER stress, and it activates a signaling pathway known as the unfolded protein response (UPR). In the context of AS, several studies indicate that oxidized low-density lipoprotein (oxLDL) causes ER stress in valvular interstitial cells by increasing cytosolic calcium levels [36,37]. Furthermore, oxLDL induces osteoblastic differentiation and promotes inflammatory responses via different ER stress-mediated pathways [38,39].
Endoplasmin, also known as glucose-regulated protein 94 (GRP94), HSP90b1, and gp96, is the most abundant glycoprotein in the ER and one of the major chaperones. Activation of the UPR results in the expression of genes encoding endoplasmin and other chaperones that mitigate the effects of increased load of unfolded proteins [40,41]. As all three branches of the UPR, the protein kinase-like ER kinase (PERK), inositol-requiring transmembrane kinase and endonuclease-1α (IRE1α), and activating transcription factor (ATF), are activated during bone formation to regulate expression of osteogenic genes, it is crucial to elucidate the role of endoplasmin in valve calcification [42,43,44,45,46]. Importantly, elevated levels of endoplasmin have been found in calcified vascular smooth muscle cells [47] and in the calcified aorta [48,49], consistent with our results.
Another protective mechanism in the calcified valve may be the increase in the levels of Thbs1, a multimeric Ca2+-binding glycoprotein that resides within the ER and that can be secreted by cells depending on the Ca2+ levels or the cell type examined [50]. As it matures in the ER, this protein also forms a complex with endoplasmin and other chaperones, such as PDI, BiP, and ERp72 [51], and it has the ability to mediate an ATF6α-dependent ER-stress response [52]. It has been suggested that Thbs1 is induced in the pressure-overloaded myocardium given that Thbs1−/− mice have greater cardiac hypertrophy than wild-type mice when submitted to pressure overload stimulation [53,54]. Our results are consistent with that phenomenon, and it seems that Thbs1 may act as a protective signal that prevents cardiac remodeling by altering fibroblast function and matrix metabolism. The appearance of two different protein isoforms of Thbs1 should also be further studied. It is known that this protein has a complex structure that includes a heparin-binding domain along with a procollagen homology domain at the amino terminus, and type I, II, and III repeats at the carboxyl-terminal end [55,56]. Thbs1 is implicated in several activities, such as homeostasis, apoptosis, or cell adhesion, as its domains can bind to receptors and specific proteins anchored in or secreted into the extracellular matrix [57,58,59,60]. As such, its synthesis and degradation are carefully regulated. Once secreted, the exposure of Thbs1 to specific microenvironmental milieus alters its structure and activity in a tissue and pathophysiological specific manner [61]. Several studies have found Thbs1 species of different molecular weights, and it has been suggested that this protein is rapidly cleared from circulation once secreted [62,63]. The influence of Thbs1 on cardiovascular diseases is complex and multifactorial, since its activity depends on the vessel type, the stage of the lesions, and associations with obesity, diabetes, or other metabolic diseases [64,65]. Thus, this protein should undoubtedly be further studied in the context of AS.
Confirmation of these proteins in both the cellular model and human plasma sample has different targets. Firstly, we used protein extracts from HAVICs submitted to osteogenic treatment. Although these proteins have previously been described in a small number of human samples, AS is a multifactorial disease and so it is difficult to discriminate if the alterations are due to the cardiocirculatory alterations caused by AV dysfunction or due to calcification itself. Moreover, AS patients are most often elderly and present different co-morbidities. These are the main limitations of this work: we have a small cohort and with different co-morbidities (although all related to cardiovascular disease). We have used a cohort of controls matched for risk factors, and we have excluded subjects with serious co-morbidities from the study, but we are aware that this may not be enough. All these drawbacks are partially avoided by the use of the cell model; this is not as complex a system as the organism, and thus the information obtained is not so complete. For this reason, in this work, we combined the insilico study and the cell model with an analysis of a larger cohort of patients to confirm the results. We searched for these proteins directly in plasma from healthy individuals and patients with severe AS. This step is important as it provides information about the usefulness of these proteins as diagnostic markers and may help translate the results to the clinical field, particularly as blood samples are easy to obtain and not too invasive compared to biopsies and surgical procedures. In the future, it would be interesting to quantify these proteins in a larger cohort, which will ideally allow the stratification of the subjects by age and co-morbidities. This will be an important step to improve precision medicine, as it will enable different thresholds to be established according to the specific characteristics of each patient, facilitating their management by clinicians.

5. Conclusions

In this work, we set out to demonstrate the importance of using largescale bioinformatics tools that allow us to consider all the data obtained through high-throughput analytical techniques to select the most significant results. Consequently, we will be able to select more specific targets and design future studies in a much more efficient way, better direct financial and social resources, and obtain higher quality results with a better chance of making advances and breakthroughs in our understanding and treatment of AS.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/jpm12040642/s1, Figure S1: Ponceau S stain image of the nitrocellulose membrane used for Western blots from cell cultures (a) and analysis of Thsb-1 (b) and endoplasmin (c) in plasma samples; Table S1: Functional analysis of the 126 proteins of interest selected from our previous studies. The proteins are represented in clusters according to their function. The enrichment score, number of terms and proteins included in each cluster are shown; Table S2: Effectors defined during the molecular characterization of the “Motives”: (1) calcification; (2) lipoprotein accumulation; (3) inflammation; (4) oxidative stress; (5) endothelial dysfunction; (6) RAA system; (7) hypertrophy; (8) myocardial fibrosis; Table S3: ANN score of each protein for each specific motive. The column effector indicates whether the protein was described in the molecular characterization, specifically in that motive [2,11,33,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103].

Author Contributions

Conceptualization, M.G.B., L.M.-A.; methodology, N.C.-A., T.S.-O., C.C.; validation, T.T., J.S., L.R.P., L.M.-A. and M.G.B.; formal analysis, N.C.-A., T.S.-O.; investigation, N.C.-A., T.S.-O., L.M.-A. and M.G.B.; resources, C.C., J.S., L.F.L.-A., L.R.P.; data curation, T.T., J.S., L.R.P., L.M.-A. and M.G.B.; writing—original draft preparation, N.C.-A., T.S.-O., L.M.-A. and M.G.B.; writing—review and editing, N.C.-A., T.S.-O., C.C., T.T., J.S., L.F.L.-A., L.R.P., L.M.-A., M.G.B.; visualization, N.C.-A. and T.S.-O.; supervision, L.M.-A. and M.G.B.; project administration, M.G.B.; funding acquisition, M.G.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Instituto de Salud Carlos III through the project PI18/00995, PI21/00384 (co-funded by European Regional Development Fund/European Social Fund—“Investing in your future”) Sociedad Española de Cardiología, 2020, Grant PRB3 (IPT17/0019—ISCIII-SGEFI/ERDF), and Junta de Comunidades de Castilla-La Mancha (JCCM, co-funded by the European Regional Development Fund, SBPLY/19/180501/000226). These results are aligned with the Spanish initiative on the Human Proteome Project (SpHPP).

Institutional Review Board Statement

This study was carried out in accordance with the recommendations of the Helsinki Declaration, and it was approved by the Ethics Committee at the participant hospitals (approval reference numbers: 18/315 and 07/036).

Informed Consent Statement

Signed informed consent was obtained from all subjects prior to their inclusion on the study.

Data Availability Statement

Not applicable.

Acknowledgments

Authors are thankful to Anaxomics Biotech (Barcelona, Spain) and the Service of Microscopy and Image Analysis at the HNP.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goldbarg, S.H.; Elmariah, S.; Miller, M.A.; Fuster, V. Insights into degenerative aortic valve disease. J. Am. Coll. Cardiol. 2007, 50, 1205–1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Helske, S.; Kupari, M.; Lindstedt, K.A.; Kovanen, P.T. Aortic valve stenosis: An active atheroinflammatory process. Curr. Opin. Lipidol. 2007, 18, 483–491. [Google Scholar] [CrossRef] [PubMed]
  3. Freeman, R.V.; Otto, C.M. Spectrum of calcific aortic valve disease: Pathogenesis, disease progression, and treatment strategies. Circulation 2005, 111, 3316–3326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Leon, M.B.; Smith, C.R.; Mack, M.J.; Makkar, R.R.; Svensson, L.G.; Kodali, S.K.; Thourani, V.H.; Tuzcu, E.M.; Miller, D.C.; Herrmann, H.C.; et al. Transcatheter or surgical aortic-valve replacement in intermediate-risk patients. N. Engl. J. Med. 2016, 374, 1609–1620. [Google Scholar] [CrossRef]
  5. Mourino-Alvarez, L.; Martin-Rojas, T.; Corros-Vicente, C.; Corbacho-Alonso, N.; Padial, L.R.; Solis, J.; Barderas, M.G. Patient management in aortic stenosis: Towards precision medicine through protein analysis, imaging and diagnostic tests. J. Clin. Med. 2020, 9, 2421. [Google Scholar] [CrossRef]
  6. Small, A.; Kiss, D.; Giri, J.; Anwaruddin, S.; Siddiqi, H.; Guerraty, M.; Chirinos, J.A.; Ferrari, G.; Rader, D.J. Biomarkers of calcific aortic valve disease. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 623–632. [Google Scholar] [CrossRef] [Green Version]
  7. Everett, R.J.; Clavel, M.-A.; Pibarot, P.; Dweck, M.R. Timing of intervention in aortic stenosis: A review of current and future strategies. Heart 2018, 104, 2067–2076. [Google Scholar] [CrossRef] [Green Version]
  8. Ky, B.; French, B.; Levy, W.C.; Sweitzer, N.K.; Fang, J.C.; Wu, A.H.B.; Goldberg, L.R.; Jessup, M.; Cappola, T.P. Multiple biomarkers for risk prediction in chronic heart failure. Circ. Heart Fail. 2012, 5, 183–190. [Google Scholar] [CrossRef] [Green Version]
  9. Wang, T.J.; Wollert, K.C.; Larson, M.G.; Coglianese, E.; McCabe, E.L.; Cheng, S.; Ho, J.E.; Fradley, M.G.; Ghorbani, A.; Xanthakis, V.; et al. Prognostic utility of novel biomarkers of cardiovascular stress: The Framingham Heart Study. Circulation 2012, 126, 1596–1604. [Google Scholar] [CrossRef] [Green Version]
  10. Alvarez-Llamas, G.; Martin-Rojas, T.; de la Cuesta, F.; Calvo, E.; Gil-Dones, F.; Darde, V.M.; Lopez-Almodovar, L.F.; Padial, L.R.; Lopez, J.-A.; Vivanco, F.; et al. Modification of the secretion pattern of proteases, inflammatory mediators, and extracellular matrix proteins by human aortic valve is key in severe aortic stenosis. Mol. Cell. Proteom. 2013, 12, 2426–2439. [Google Scholar] [CrossRef] [Green Version]
  11. Martin-Rojas, T.; Mourino-Alvarez, L.; Alonso-Orgaz, S.; Rosello-Lleti, E.; Calvo, E.; Lopez-Almodovar, L.F.; Rivera, M.; Padial, L.R.; Lopez, J.A.; de la Cuesta, F.; et al. iTRAQ proteomic analysis of extracellular matrix remodeling in aortic valve disease. Sci. Rep. 2015, 5, 17290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Gil-Dones, F.; Darde, V.; Orgaz, S.A.; Lopez-Almodovar, L.F.; Mourino-Alvarez, L.; Padial, L.R.; Vivanco, F.; Barderas, M.G. Inside human aortic stenosis: A proteomic analysis of plasma. J. Proteom. 2012, 75, 1639–1653. [Google Scholar] [CrossRef] [PubMed]
  13. Martin-Rojas, T.; Gil-Dones, F.; Lopez-Almodovar, L.F.; Padial, L.R.; Vivanco, F.; Barderas, M.G. Proteomic profile of human aortic stenosis: Insights into the degenerative process. J. Proteome Res. 2012, 11, 1537–1550. [Google Scholar] [CrossRef] [PubMed]
  14. Mourino-Alvarez, L.; Iloro, I.; de la Cuesta, F.; Azkargorta, M.; Sastre-Oliva, T.; Escobes, I.; Lopez-Almodovar, L.F.; Sanchez, P.L.; Urreta, H.; Fernandez-Aviles, F.; et al. MALDI-Imaging Mass Spectrometry: A step forward in the anatomopathological characterization of stenotic aortic valve tissue. Sci. Rep. 2016, 6, 27106. [Google Scholar] [CrossRef] [PubMed]
  15. Kanehisa, M.; Furumichi, M.; Tanabe, M.; Sato, Y.; Morishima, K. KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017, 45, D353–D361. [Google Scholar] [CrossRef] [Green Version]
  16. Chatr-Aryamontri, A.; Oughtred, R.; Boucher, L.; Rust, J.; Chang, C.; Kolas, N.K.; O’Donnell, L.; Oster, S.; Theesfeld, C.; Sellam, A.; et al. The BioGRID interaction database: 2017 update. Nucleic Acids Res. 2017, 45, D369–D379. [Google Scholar] [CrossRef]
  17. Orchard, S.; Ammari, M.; Aranda, B.; Breuza, L.; Briganti, L.; Broackes-Carter, F.; Campbell, N.H.; Chavali, G.; Chen, C.; del-Toro, N.; et al. The MIntAct project—IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Res. 2014, 42, D358–D363. [Google Scholar] [CrossRef] [Green Version]
  18. Fabregat, A.; Sidiropoulos, K.; Garapati, P.; Gillespie, M.; Hausmann, K.; Haw, R.; Jassal, B.; Jupe, S.; Korninger, F.; McKay, S.; et al. The Reactome pathway Knowledgebase. Nucleic Acids Res. 2016, 44, D481–D487. [Google Scholar] [CrossRef] [Green Version]
  19. Han, H.; Cho, J.W.; Lee, S.; Yun, A.; Kim, H.; Bae, D.; Yang, S.; Kim, C.Y.; Lee, M.; Kim, E.; et al. TRRUST v2: An expanded reference database of human and mouse transcriptional regulatory interactions. Nucleic Acids Res. 2018, 46, D380–D386. [Google Scholar] [CrossRef]
  20. Keshava Prasad, T.S.; Goel, R.; Kandasamy, K.; Keerthikumar, S.; Kumar, S.; Mathivanan, S.; Telikicherla, D.; Raju, R.; Shafreen, B.; Venugopal, A.; et al. Human Protein Reference Database—2009 update. Nucleic Acids Res. 2009, 37, D767–D772. [Google Scholar] [CrossRef] [Green Version]
  21. Bishop, C.M. Pattern Recognition and Machine Learning; Information Science and Statistics; Springer: New York, NY, USA, 2006; p. 738. [Google Scholar]
  22. Latif, N.; Quillon, A.; Sarathchandra, P.; McCormack, A.; Lozanoski, A.; Yacoub, M.H.; Chester, A.H. Modulation of human valve interstitial cell phenotype and function using a Fibroblast Growth Factor 2 formulation. PLoS ONE 2015, 10, e0127844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. van Engeland, N.C.A.; Bertazzo, S.; Sarathchandra, P.; McCormack, A.; Bouten, C.V.C.; Yacoub, M.H.; Chester, A.H.; Latif, N. Aortic calcified particles modulate valvular endothelial and interstitial cells. Cardiovasc. Pathol. 2017, 28, 36–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Gregory, C.A.; Gunn, W.G.; Peister, A.; Prockop, D.J. An Alizarin red-based assay of mineralization by adherent cells in culture: Comparison with cetylpyridinium chloride extraction. Anal. Biochem. 2004, 329, 77–84. [Google Scholar] [CrossRef] [PubMed]
  25. Gil-Dones, F.; Martin-Rojas, T.; Lopez-Almodovar, L.; De la Cuesta, F.; Darde, V.; Alvarez-Llamas, G.; Juarez-Tosina, T.; Barroso, G.; Vivanco, F.; Padial, L.; et al. Valvular aortic stenosis: A proteomic insight. Clin. Med. Insights Cardiol. 2010, 4, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bradford, M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 1976, 72, 248–254. [Google Scholar] [CrossRef]
  27. Gilda, J.E.; Gomes, A.V. Western blotting using in-gel protein labeling as a normalization control: Stain-free technology. Methods Mol. Biol. 2015, 1295, 381–391. [Google Scholar] [CrossRef]
  28. Chung, K.F.; Adcock, I.M. Precision medicine for the discovery of treatable mechanisms in severe asthma. Allergy 2019, 74, 1649–1659. [Google Scholar] [CrossRef] [Green Version]
  29. Sathyamurthy, I.; Alex, S.; Kirubakaran, K.; Sengottuvelu, G.; Srinivasan, K.N. Risk factor profile of calcific aortic stenosis. Indian Heart J. 2016, 68, 828–831. [Google Scholar] [CrossRef] [Green Version]
  30. Hofmanis, J.; Hofmane, D.; Svirskis, S.; Mackevics, V.; Tretjakovs, P.; Lejnieks, A.; Signorelli, S.S. HDL-C role in acquired aortic valve stenosis patients and its relationship with oxidative stress. Medicina 2019, 55, 416. [Google Scholar] [CrossRef] [Green Version]
  31. Lerman, D.A.; Prasad, S.; Alotti, N. Calcific aortic valve disease: Molecular mechanisms and therapeutic approaches. Eur. Cardiol. 2015, 10, 108–112. [Google Scholar] [CrossRef] [Green Version]
  32. Fielitz, J.; Hein, S.; Mitrovic, V.; Pregla, R.; Zurbrügg, H.R.; Warnecke, C.; Schaper, J.; Fleck, E.; Regitz-Zagrosek, V. Activation of the cardiac renin-angiotensin system and increased myocardial collagen expression in human aortic valve disease. J. Am. Coll. Cardiol. 2001, 37, 1443–1449. [Google Scholar] [CrossRef] [Green Version]
  33. Lindman, B.R.; Clavel, M.A.; Mathieu, P.; Iung, B.; Lancellotti, P.; Otto, C.M.; Pibarot, P. Calcific aortic stenosis. Nat. Rev.Dis. Primers 2016, 2, 16006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Pibarot, P.; Dumesnil, J.G. Improving assessment of aortic stenosis. J. Am. Coll. Cardiol. 2012, 60, 169–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Antonini-Canterin, F.; Huang, G.; Cervesato, E.; Faggiano, P.; Pavan, D.; Piazza, R.; Nicolosi, G.L. Symptomatic aortic stenosis: Does systemic hypertension play an additional role? Hypertension 2003, 41, 1268–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sanson, M.; Augé, N.; Vindis, C.; Muller, C.; Bando, Y.; Thiers, J.C.; Marachet, M.A.; Zarkovic, K.; Sawa, Y.; Salvayre, R.; et al. Oxidized low-density lipoproteins trigger endoplasmic reticulum stress in vascular cells: Prevention by oxygen-regulated protein 150 expression. Circ. Res. 2009, 104, 328–336. [Google Scholar] [CrossRef] [Green Version]
  37. Dong, Y.; Zhang, M.; Wang, S.; Liang, B.; Zhao, Z.; Liu, C.; Wu, M.; Choi, H.C.; Lyons, T.J.; Zou, M.H. Activation of AMP-activated protein kinase inhibits oxidized LDL-triggered endoplasmic reticulum stress in vivo. Diabetes 2010, 59, 1386–1396. [Google Scholar] [CrossRef] [Green Version]
  38. Deng, J.; Lu, P.D.; Zhang, Y.; Scheuner, D.; Kaufman, R.J.; Sonenberg, N.; Harding, H.P.; Ron, D. Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2. Mol. Cell. Biol. 2004, 24, 10161–10168. [Google Scholar] [CrossRef] [Green Version]
  39. Cai, Z.; Li, F.; Gong, W.; Liu, W.; Duan, Q.; Chen, C.; Ni, L.; Xia, Y.; Cianflone, K.; Dong, N.; et al. Endoplasmic reticulum stress participates in aortic valve calcification in hypercholesterolemic animals. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2345–2354. [Google Scholar] [CrossRef] [Green Version]
  40. Ron, D.; Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 2007, 8, 519–529. [Google Scholar] [CrossRef]
  41. Luo, S.; Baumeister, P.; Yang, S.; Abcouwer, S.F.; Lee, A.S. Induction of Grp78/BiP by translational block: Activation of the Grp78 promoter by ATF4 through and upstream ATF/CRE site independent of the endoplasmic reticulum stress elements. J. Biol. Chem. 2003, 278, 37375–37385. [Google Scholar] [CrossRef] [Green Version]
  42. Clauss, I.M.; Gravallese, E.M.; Darling, J.M.; Shapiro, F.; Glimcher, M.J.; Glimcher, L.H. In situ hybridization studies suggest a role for the basic region-leucine zipper protein hXBP-1 in exocrine gland and skeletal development during mouse embryogenesis. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 1993, 197, 146–156. [Google Scholar] [CrossRef] [PubMed]
  43. Tohmonda, T.; Miyauchi, Y.; Ghosh, R.; Yoda, M.; Uchikawa, S.; Takito, J.; Morioka, H.; Nakamura, M.; Iwawaki, T.; Chiba, K.; et al. The IRE1α-XBP1 pathway is essential for osteoblast differentiation through promoting transcription of Osterix. EMBO Rep. 2011, 12, 451–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Han, X.; Zhou, J.; Zhang, P.; Song, F.; Jiang, R.; Li, M.; Xia, F.; Guo, F.J. IRE1α dissociates with BiP and inhibits ER stress-mediated apoptosis in cartilage development. Cell Signal. 2013, 25, 2136–2146. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, P.; McGrath, B.; Li, S.; Frank, A.; Zambito, F.; Reinert, J.; Gannon, M.; Ma, K.; McNaughton, K.; Cavener, D.R. The PERK eukaryotic initiation factor 2 alpha kinase is required for the development of the skeletal system, postnatal growth, and the function and viability of the pancreas. Mol. Cell. Biol. 2002, 22, 3864–3874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Yang, X.; Matsuda, K.; Bialek, P.; Jacquot, S.; Masuoka, H.C.; Schinke, T.; Li, L.; Brancorsini, S.; Sassone-Corsi, P.; Townes, T.M.; et al. ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology; implication for Coffin-Lowry Syndrome. Cell 2004, 117, 387–398. [Google Scholar] [CrossRef] [Green Version]
  47. Furmanik, M.; van Gorp, R.; Whitehead, M.; Ahmad, S.; Bordoloi, J.; Kapustin, A.; Schurgers, L.J.; Shanahan, C.M. Endoplasmic reticulum stress mediates vascular smooth muscle cell calcification via increased release of Grp78 (Glucose-Regulated Protein, 78 kDa)-loaded extracellular vesicles. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 898–914. [Google Scholar] [CrossRef]
  48. Wang, Q.; Lin, P.; Feng, L.; Ren, Q.; Xie, X.; Zhang, B. Ameliorative effect of allicin on vascular calcification via inhibiting endoplasmic reticulum stress. Vascular 2021, 17085381211035291. [Google Scholar] [CrossRef]
  49. Song, X.; Li, J.; Jiao, M.; Chen, Y.; Pan, K. Effect of endoplasmic reticulum stress-induced apoptosis in the role of periodontitis on vascular calcification in a rat model. J. Mol. Histol. 2021, 52, 1097–1104. [Google Scholar] [CrossRef]
  50. Veliceasa, D.; Ivanovic, M.; Hoepfner, F.T.; Thumbikat, P.; Volpert, O.V.; Smith, N.D. Transient potential receptor channel 4 controls thrombospondin-1 secretion and angiogenesis in renal cell carcinoma. FEBS J. 2007, 274, 6365–6377. [Google Scholar] [CrossRef]
  51. Kuznetsov, G.; Chen, L.B.; Nigam, S.K. Multiple molecular chaperones complex with misfolded large oligomeric glycoproteins in the endoplasmic reticulum. J. Biol. Chem. 1997, 272, 3057–3063. [Google Scholar] [CrossRef] [Green Version]
  52. Lynch, J.M.; Maillet, M.; Vanhoutte, D.; Schloemer, A.; Sargent, M.A.; Blair, N.S.; Lynch, K.A.; Okada, T.; Aronow, B.J.; Osinska, H.; et al. A thrombospondin-dependent pathway for a protective ER stress response. Cell 2012, 149, 1257–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xia, Y.; Dobaczewski, M.; Gonzalez-Quesada, C.; Chen, W.; Biernacka, A.; Li, N.; Lee, D.W.; Frangogiannis, N.G. Endogenous thrombospondin 1 protects the pressure-overloaded myocardium by modulating fibroblast phenotype and matrix metabolism. Hypertension 2011, 58, 902–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Vanhoutte, D.; Schips, T.G.; Vo, A.; Grimes, K.M.; Baldwin, T.A.; Brody, M.J.; Accornero, F.; Sargent, M.A.; Molkentin, J.D. Thbs1 induces lethal cardiac atrophy through PERK-ATF4 regulated autophagy. Nat. Commun. 2021, 12, 3928. [Google Scholar] [CrossRef] [PubMed]
  55. Calzada, M.J.; Sipes, J.M.; Krutzsch, H.C.; Yurchenco, P.D.; Annis, D.S.; Mosher, D.F.; Roberts, D.D. Recognition of the N-terminal modules of thrombospondin-1 and thrombospondin-2 by alpha6beta1 integrin. J. Biol. Chem. 2003, 278, 40679–40687. [Google Scholar] [CrossRef] [Green Version]
  56. Lopez-Dee, Z.; Pidcock, K.; Gutierrez, L.S. Thrombospondin-1: Multiple paths to inflammation. Mediat. Inflamm. 2011, 2011, 296069. [Google Scholar] [CrossRef] [Green Version]
  57. Lawler, J.; Hynes, R.O. The structure of human thrombospondin, an adhesive glycoprotein with multiple calcium-binding sites and homologies with several different proteins. J. Cell Biol. 1986, 103, 1635–1648. [Google Scholar] [CrossRef] [Green Version]
  58. Good, D.J.; Polverini, P.J.; Rastinejad, F.; Le Beau, M.M.; Lemons, R.S.; Frazier, W.A.; Bouck, N.P. A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin. Proc. Natl. Acad. Sci. USA 1990, 87, 6624–6628. [Google Scholar] [CrossRef] [Green Version]
  59. Mumby, S.M.; Raugi, G.J.; Bornstein, P. Interactions of thrombospondin with extracellular matrix proteins: Selective binding to type V collagen. J. Cell Biol. 1984, 98, 646–652. [Google Scholar] [CrossRef] [Green Version]
  60. Li, Z.; He, L.; Wilson, K.; Roberts, D. Thrombospondin-1 inhibits TCR-mediated T lymphocyte early activation. J. Immunol. 2001, 166, 2427–2436. [Google Scholar] [CrossRef] [Green Version]
  61. Iruela-Arispe, M.L. Regulation of thrombospondin1 by extracellular proteases. Curr. Drug Targets 2008, 9, 863–868. [Google Scholar] [CrossRef] [Green Version]
  62. Starlinger, P.; Moll, H.P.; Assinger, A.; Nemeth, C.; Hoetzenecker, K.; Gruenberger, B.; Gruenberger, T.; Kuehrer, I.; Schoppmann, S.F.; Gnant, M.; et al. Thrombospondin-1: A unique marker to identify in vitro platelet activation when monitoring in vivo processes. J. Thromb. Haemost. 2010, 8, 1809–1819. [Google Scholar] [CrossRef] [PubMed]
  63. Mikhailenko, I.; Krylov, D.; Argraves, K.M.; Roberts, D.D.; Liau, G.; Strickland, D.K. Cellular internalization and degradation of thrombospondin-1 is mediated by the amino-terminal heparin binding domain (HBD). High affinity interaction of dimeric HBD with the low density lipoprotein receptor-related protein. J. Biol. Chem. 1997, 272, 6784–6791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Gutierrez, L.S.; Gutierrez, J. Thrombospondin 1 in metabolic diseases. Front. Endocrinol. 2021, 12, 638536. [Google Scholar] [CrossRef] [PubMed]
  65. Stenina, O.I.; Plow, E.F. Counterbalancing forces: What is thrombospondin-1 doing in atherosclerotic lesions? Circ. Res. 2008, 103, 1053–1055. [Google Scholar] [CrossRef] [PubMed]
  66. Adil, S.O.; Ibran, E.A.; Nisar, N.; Shafique, K. Pattern of unintentional burns: A hospital based study from Pakistan. Burns 2016, 42, 1345–1349. [Google Scholar] [CrossRef]
  67. Capoulade, R.; Mahmut, A.; Tastet, L.; Arsenault, M.; Bédard, É.; Dumesnil, J.G.; Després, J.P.; Larose, É.; Arsenault, B.J.; Bossé, Y.; et al. Impact of plasma Lp-PLA2 activity on the progression of aortic stenosis: The PROGRESSA study. JACC Cardiovasc. Imaging 2015, 8, 26–33. [Google Scholar] [CrossRef] [Green Version]
  68. Cho, K.I.; Sakuma, I.; Sohn, I.S.; Jo, S.H.; Koh, K.K. Inflammatory and metabolic mechanisms underlying the calcific aortic valve disease. Atherosclerosis 2018, 277, 60–65. [Google Scholar] [CrossRef]
  69. Cowell, S.J.; Newby, D.E.; Boon, N.A.; Elder, A.T. Calcific aortic stenosis: Same old story? Age Ageing 2004, 33, 538–544. [Google Scholar] [CrossRef] [Green Version]
  70. Doris, M.K.; Everett, R.J.; Shun-Shin, M.; Clavel, M.A.; Dweck, M.R. The Role of imaging in measuring disease progression and assessing novel therapies in aortic stenosis. JACC Cardiovasc. Imaging 2019, 12, 185–197. [Google Scholar] [CrossRef]
  71. Elmariah, S.; Mohler, E.R., 3rd. The Pathogenesis and treatment of the valvulopathy of aortic stenosis: Beyond the SEAS. Curr. Cardiol. Rep. 2010, 12, 125–132. [Google Scholar] [CrossRef] [Green Version]
  72. Gallo, G.; Presta, V.; Volpe, M.; Rubattu, S. Molecular and clinical implications of natriuretic peptides in aortic valve stenosis. J. Mol. Cell. Cardiol. 2019, 129, 266–271. [Google Scholar] [CrossRef] [PubMed]
  73. García-Rodríguez, C.; Parra-Izquierdo, I.; Castaños-Mollor, I.; López, J.; San Román, J.A.; Sánchez Crespo, M. Toll-Like receptors, inflammation, and calcific aortic valve disease. Front. Physiol. 2018, 9, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Izquierdo-Gómez, M.M.; Hernández-Betancor, I.; García-Niebla, J.; Marí-López, B.; Laynez-Cerdeña, I.; Lacalzada-Almeida, J. Valve calcification in aortic stenosis: Etiology and diagnostic imaging techniques. BioMed Res. Int. 2017, 2017, 5178631. [Google Scholar] [CrossRef] [PubMed]
  75. Kaden, J.J.; Bickelhaupt, S.; Grobholz, R.; Vahl, C.F.; Hagl, S.; Brueckmann, M.; Haase, K.K.; Dempfle, C.E.; Borggrefe, M. Expression of bone sialoprotein and bone morphogenetic protein-2 in calcific aortic stenosis. J. Heart Valve Dis. 2004, 13, 560–566. [Google Scholar]
  76. Kapelouzou, A.; Tsourelis, L.; Kaklamanis, L.; Degiannis, D.; Kogerakis, N.; Cokkinos, D.V. Serum and tissue biomarkers in aortic stenosis. Glob. Cardiol. Sci. Pract. 2015, 2015, 49. [Google Scholar] [CrossRef]
  77. Katholi, R.E.; Couri, D.M. Left ventricular hypertrophy: Major risk factor in patients with hypertension: Update and practical clinical applications. Int. J. Hypertens. 2011, 2011, 495349. [Google Scholar] [CrossRef] [Green Version]
  78. Kleinauskienė, R.; Jonkaitienė, R. Degenerative aortic stenosis, dyslipidemia and possibilities of medical treatment. Medicina 2018, 54, 24. [Google Scholar] [CrossRef] [Green Version]
  79. Kolasa-Trela, R.; Konieczynska, M.; Bazanek, M.; Undas, A. Specific changes in circulating cytokines and growth factors induced by exercise stress testing in asymptomatic aortic valve stenosis. PLoS ONE 2017, 12, e0173787. [Google Scholar] [CrossRef] [Green Version]
  80. Lee, S.H.; Choi, J.H. Involvement of inflammatory responses in the early development of calcific aortic valve disease: Lessons from statin therapy. Anim. Cells Syst. 2018, 22, 390–399. [Google Scholar] [CrossRef]
  81. Legere, S.A.; Haidl, I.D.; Légaré, J.F.; Marshall, J.S. Mast cells in cardiac fibrosis: New insights suggest opportunities for intervention. Front. Immunol. 2019, 10, 580. [Google Scholar] [CrossRef]
  82. Liu, T.; Song, D.; Dong, J.; Zhu, P.; Liu, J.; Liu, W.; Ma, X.; Zhao, L.; Ling, S. Current understanding of the pathophysiology of myocardial fibrosis and its quantitative assessment in heart failure. Front. Physiol. 2017, 8, 238. [Google Scholar] [CrossRef] [PubMed]
  83. Ma, Z.G.; Yuan, Y.P.; Wu, H.M.; Zhang, X.; Tang, Q.Z. Cardiac fibrosis: New insights into the pathogenesis. Int. J. Biol. Sci. 2018, 14, 1645–1657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Mathieu, P.; Bouchareb, R.; Boulanger, M.C. Innate and adaptive immunity in calcific aortic valve disease. J. Immunol. Res. 2015, 2015, 851945. [Google Scholar] [CrossRef] [PubMed]
  85. Mathieu, P.; Boulanger, M.C. Basic mechanisms of calcific aortic valve disease. Can. J. Cardiol. 2014, 30, 982–993. [Google Scholar] [CrossRef]
  86. Mathieu, P.; Boulanger, M.C.; Bouchareb, R. Molecular biology of calcific aortic valve disease: Towards new pharmacological therapies. Expert Rev. Cardiovasc. Ther. 2014, 12, 851–862. [Google Scholar] [CrossRef]
  87. Miller, J.D.; Chu, Y.; Brooks, R.M.; Richenbacher, W.E.; Peña-Silva, R.; Heistad, D.D. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic valvular stenosis in humans. J. Am. Coll. Cardiol. 2008, 52, 843–850. [Google Scholar] [CrossRef] [Green Version]
  88. Musa, T.A.; Treibel, T.A.; Vassiliou, V.S.; Captur, G.; Singh, A.; Chin, C.; Dobson, L.E.; Pica, S.; Loudon, M.; Malley, T.; et al. Myocardial Scar and Mortality in Severe Aortic Stenosis. Circulation 2018, 138, 1935–1947. [Google Scholar] [CrossRef]
  89. Myasoedova, V.A.; Ravani, A.L.; Frigerio, B.; Valerio, V.; Moschetta, D.; Songia, P.; Poggio, P. Novel pharmacological targets for calcific aortic valve disease: Prevention and treatments. Pharmacol. Res. 2018, 136, 74–82. [Google Scholar] [CrossRef]
  90. Nakamura, M.; Sadoshima, J. Mechanisms of physiological and pathological cardiac hypertrophy. Nat. Rev. Cardiol. 2018, 15, 387–407. [Google Scholar] [CrossRef]
  91. O’Brien, K.D.; Reichenbach, D.D.; Marcovina, S.M.; Kuusisto, J.; Alpers, C.E.; Otto, C.M. Apolipoproteins B, (a), and E accumulate in the morphologically early lesion of ’degenerative’ valvular aortic stenosis. Arterioscler. Thromb. Vasc. Biol. 1996, 16, 523–532. [Google Scholar] [CrossRef]
  92. Osman, L.; Yacoub, M.H.; Latif, N.; Amrani, M.; Chester, A.H. Role of human valve interstitial cells in valve calcification and their response to atorvastatin. Circulation 2006, 114, I547–I552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Pasipoularides, A. Calcific aortic valve disease: Part 2-morphomechanical abnormalities, gene reexpression, and gender effects on ventricular hypertrophy and its reversibility. J. Cardiovasc. Transl. Res. 2016, 9, 374–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Peltonen, T.; Taskinen, P.; Näpänkangas, J.; Leskinen, H.; Ohtonen, P.; Soini, Y.; Juvonen, T.; Satta, J.; Vuolteenaho, O.; Ruskoaho, H. Increase in tissue endothelin-1 and ETA receptor levels in human aortic valve stenosis. Eur. Heart J. 2009, 30, 242–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Perrucci, G.L.; Zanobini, M.; Gripari, P.; Songia, P.; Alshaikh, B.; Tremoli, E.; Poggio, P. Pathophysiology of aortic stenosis and mitral regurgitation. Compr. Physiol. 2017, 7, 799–818. [Google Scholar] [CrossRef] [PubMed]
  96. Sverdlov, A.L.; Ngo, D.T.; Chapman, M.J.; Ali, O.A.; Chirkov, Y.Y.; Horowitz, J.D. Pathogenesis of aortic stenosis: Not just a matter of wear and tear. Am. J. Cardiovasc. Dis. 2011, 1, 185–199. [Google Scholar]
  97. Syväranta, S.; Alanne-Kinnunen, M.; Oörni, K.; Oksjoki, R.; Kupari, M.; Kovanen, P.T.; Helske-Suihko, S. Potential pathological roles for oxidized low-density lipoprotein and scavenger receptors SR-AI, CD36, and LOX-1 in aortic valve stenosis. Atherosclerosis 2014, 235, 398–407. [Google Scholar] [CrossRef] [PubMed]
  98. Towler, D.A. Oxidation, inflammation, and aortic valve calcification peroxide paves an osteogenic path. J. Am. Coll. Cardiol. 2008, 52, 851–854. [Google Scholar] [CrossRef] [Green Version]
  99. Venardos, N.; Nadlonek, N.A.; Zhan, Q.; Weyant, M.J.; Reece, T.B.; Meng, X.; Fullerton, D.A. Aortic valve calcification is mediated by a differential response of aortic valve interstitial cells to inflammation. J. Surg. Res. 2014, 190, 1–8. [Google Scholar] [CrossRef] [Green Version]
  100. Weber, C.; Noels, H. Atherosclerosis: Current pathogenesis and therapeutic options. Nat. Med. 2011, 17, 1410–1422. [Google Scholar] [CrossRef]
  101. Yetkin, E.; Waltenberger, J. Molecular and cellular mechanisms of aortic stenosis. Int. J. Cardiol. 2009, 135, 4–13. [Google Scholar] [CrossRef]
  102. Yip, C.Y.; Simmons, C.A. The aortic valve microenvironment and its role in calcific aortic valve disease. Cardiovasc. Pathol. 2011, 20, 177–182. [Google Scholar] [CrossRef] [PubMed]
  103. Zhan, Q.; Zeng, Q.; Song, R.; Zhai, Y.; Xu, D.; Fullerton, D.A.; Dinarello, C.A.; Meng, X. IL-37 suppresses MyD88-mediated inflammatory responses in human aortic valve interstitial cells. Mol. Med. 2017, 23, 83–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Mathematical model pipeline. Aortic stenosis was defined at molecular level through bibliography and database revision, a biological map was built, and mathematical models were trained. Then, candidate proteins were prioritized according to the functional relationship with the disease.
Figure 1. Mathematical model pipeline. Aortic stenosis was defined at molecular level through bibliography and database revision, a biological map was built, and mathematical models were trained. Then, candidate proteins were prioritized according to the functional relationship with the disease.
Jpm 12 00642 g001
Figure 2. Verification of the osteoblastic differentiation through alizarin red staining (a) and α-SMA (b) and Western blot confirmation of thrombospondin-1 (c) and endoplasmin (d) levels in HAVICs treated with FIB medium (C) and osteogenic medium (Ost) after 7 and 14 days of culture. Data from western blots were normalized to total protein level (Ponceau S stain, Figure S1).*= p < 0.05.
Figure 2. Verification of the osteoblastic differentiation through alizarin red staining (a) and α-SMA (b) and Western blot confirmation of thrombospondin-1 (c) and endoplasmin (d) levels in HAVICs treated with FIB medium (C) and osteogenic medium (Ost) after 7 and 14 days of culture. Data from western blots were normalized to total protein level (Ponceau S stain, Figure S1).*= p < 0.05.
Jpm 12 00642 g002
Figure 3. Western blot confirmation of thrombospondin-1 (a) and endoplasmin (b) levels in the plasma samples (control and severe AS subjects), with the corresponding p-values (Student’s t-test) for each protein analyzed: *= p < 0.05; RI, relative intensity.
Figure 3. Western blot confirmation of thrombospondin-1 (a) and endoplasmin (b) levels in the plasma samples (control and severe AS subjects), with the corresponding p-values (Student’s t-test) for each protein analyzed: *= p < 0.05; RI, relative intensity.
Jpm 12 00642 g003
Table 1. List of 126 proteins of interest based on our previous studies, showing the original work used for the selection of each protein. Additional information about their biological functions is shown in Table S1. These proteins were subsequently evaluated using the ANN strategy.
Table 1. List of 126 proteins of interest based on our previous studies, showing the original work used for the selection of each protein. Additional information about their biological functions is shown in Table S1. These proteins were subsequently evaluated using the ANN strategy.
Protein NameUniprot IDReferenceProtein NameUniprot IDReference
72 kDa type IV collagenaseP08253Alvarez-Llamas G et al., 2013 [10]Glutathione S-transferase PP09211Martin-Rojas T et al., 2012 [13]
Alcohol dehydrogenase 1BP00325Martin-Rojas T et al., 2015 [11]Glycogen phosphorylase, liver formP06737Alvarez-Llamas G et al., 2013 [10]
Alpha-1-acid glycoprotein 1P02763Martin-Rojas T et al., 2015 [11]HaptoglobinP00738Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]
Alpha-1-antichymotrypsinP01011Gil-Dones F et al., 2012 [12]; Alvarez-Llamas G et al., 2013 [10]Hemoglobin subunit betaP68871Gil-Dones F et al., 2012 [12]
Alpha-1-antitrypsinP01009Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]; Gil-Dones F et al., 2012 [12]HemopexinP02790Martin-Rojas T et al., 2015 [11]; Gil-Dones F et al., 2012 [12]
Alpha-1B-glycoproteinP04217Martin-Rojas T et al., 2012 [13]Histone H2A type 1-HQ96KK5Martin-Rojas T et al., 2015 [11]
Alpha-2-HS-glycoproteinP02765Martin-Rojas T et al., 2015 [11]; Gil-Dones F et al., 2012 [12]Ig gamma-1 chain C regionP01857Martin-Rojas T et al., 2015 [11]; Alvarez-Llamas G et al., 2013 [10]
Alpha-2-macroglobulinP01023Alvarez-Llamas G et al., 2013 [10]Ig kappa chain C regionP01834Gil-Dones F et al., 2012 [12]
Alpha-enolaseP06733Martin-Rojas T et al., 2015 [11]Ig lambda-1 chain C regionsP0CG04Gil-Dones F et al., 2012 [12]
AngiotensinogenP01019Alvarez-Llamas G et al., 2013 [10]Ig mu chain C regionP01871Gil-Dones F et al., 2012 [12]
Annexin A1P04083Martin-Rojas T et al., 2015 [11]Insulin-like growth factor-binding protein 5P24593Alvarez-Llamas G et al., 2013 [10]
Annexin A2P07355Martin-Rojas T et al., 2015 [11]Insulin-like growth factor-binding protein 7Q16270Alvarez-Llamas G et al., 2013 [10]
Antithrombin-IIIP01008Gil-Dones F et al., 2012 [12]Inter-alpha-trypsin inhibitor heavy chain H4Q14624Gil-Dones F et al., 2012 [12]
Apolipoprotein A-IP02647Martin-Rojas T et al., 2012 [13]; Gil-Dones F et al., 2012 [12]Interleukin-6P05231Alvarez-Llamas G et al., 2013 [10]
Apolipoprotein A-IVP06727Gil-Dones F et al., 2012 [12]Killer cell immunoglobulin-like receptor 3DL3Q8N743Alvarez-Llamas G et al., 2013 [10]
Apolipoprotein B-100P04114Alvarez-Llamas G et al., 2013 [10]Kininogen-1P01042Gil-Dones F et al., 2012 [12]
Apolipoprotein C-IIP02655Martin-Rojas T et al., 2015 [11]Leucine-rich alpha-2-glycoproteinP02750Gil-Dones F et al., 2012 [12]
Apolipoprotein C-IIIP02656Gil-Dones F et al., 2012 [12]Leukocyte receptor cluster member 9Q96B70Alvarez-Llamas G et al., 2013 [10]
Apolipoprotein EP02649Gil-Dones F et al., 2012 [12]L-lactate dehydrogenase A chainP00338Martin-Rojas T et al., 2015 [11]
Beta-1,4-galactosyl- transferase 2O60909Alvarez-Llamas G et al., 2013 [10]LumicanP51884Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]; Alvarez-Llamas G et al., 2013 [10]
BiglycanP21810Martin-Rojas T et al., 2015 [11]; Alvarez-Llamas G et al., 2013 [10]Mannose-binding protein CP11226Gil-Dones F et al., 2012 [12]
Biogenesis of lysosome-related organelles complex 1 subunit 5Q8TDH9Martin-Rojas T et al., 2015 [11]Metalloproteinase inhibitor 1P01033Alvarez-Llamas G et al., 2013 [10]
Calcineurin-binding protein cabin-1Q9Y6J0Martin-Rojas T et al., 2015 [11]Metalloproteinase inhibitor 3P35625Alvarez-Llamas G et al., 2013 [10]
CalreticulinP27797Martin-Rojas T et al., 2012 [13]MoesinP26038Martin-Rojas T et al., 2015 [11]
Cartilage oligomeric matrix proteinP49747Alvarez-Llamas G et al., 2013 [10]Nuclear factor NF-kappa-B p100 subunitQ00653Alvarez-Llamas G et al., 2013 [10]
Cathepsin BP07858Alvarez-Llamas G et al., 2013 [10]Pentraxin-related protein PTX3P26022Alvarez-Llamas G et al., 2013 [10]
Cathepsin DP07339Alvarez-Llamas G et al., 2013 [10]Peptidyl-prolyl cis-trans isomerase AP62937Martin-Rojas T et al., 2015 [11]
CD5 antigen-likeO43866Gil-Dones F et al., 2012 [12]PeriostinQ15063Martin-Rojas T et al., 2015 [11]
CD9 antigenP21926Alvarez-Llamas G et al., 2013 [10]Peroxiredoxin-1Q06830Martin-Rojas T et al., 2015 [11]
CeruloplasminP00450Gil-Dones F et al., 2012 [12]; Alvarez-Llamas G et al., 2013 [10]Phosphoglycerate kinase 1P00558Martin-Rojas T et al., 2015 [11]
Chitinase-3-like protein 1P36222Alvarez-Llamas G et al., 2013 [10]Phospholipid transfer proteinP55058Alvarez-Llamas G et al., 2013 [10]
Chitinase-3-like protein 2Q15782Alvarez-Llamas G et al., 2013 [10]Pigment epithelium-derived factorP36955Alvarez-Llamas G et al., 2013 [10]
ClusterinP10909Gil-Dones F et al., 2012 [12]; Alvarez-Llamas G et al., 2013 [10]Plasma protease C1 inhibitorP05155Gil-Dones F et al., 2012 [12]; Alvarez-Llamas G et al., 2013 [10]
Coagulation factor XIIP00748Gil-Dones F et al., 2012 [12]Plasminogen activator inhibitor 1P05121Alvarez-Llamas G et al., 2013 [10]
Collagen alpha-1(III) chainP02461Alvarez-Llamas G et al., 2013 [10]Pre-B-cell leukemia transcription factor-interacting protein 1Q96AQ6Alvarez-Llamas G et al., 2013 [10]
Collagen alpha-1(VI) chainP12109Martin-Rojas T et al., 2015 [11]; Alvarez-Llamas G et al., 2013 [10]Procollagen C-endopeptidase enhancer 2Q9UKZ9Alvarez-Llamas G et al., 2013 [10]
Collagen alpha-1(XIV) chainQ05707Alvarez-Llamas G et al., 2013 [10]ProlarginP51888Martin-Rojas T et al., 2015 [11]
Collagen alpha-2(I) chainP08123Alvarez-Llamas G et al., 2013 [10]ProsaposinP07602Alvarez-Llamas G et al., 2013 [10]
Collagen alpha-3(VI) chainP12111Mourino-Alvarez L et al., 2016 [14]Prostaglandin-H2 D-isomeraseP41222Alvarez-Llamas G et al., 2013 [10]
Complement C1s subcomponentP09871Alvarez-Llamas G et al., 2013 [10]Protein AMBPP02760Gil-Dones F et al., 2012 [12]
Complement C3P01024Gil-Dones F et al., 2012 [12]; Alvarez-Llamas G et al., 2013 [10]Protein NDRG2Q9UN36Mourino-Alvarez L et al., 2016 [14]
Complement C4-AP0C0L4Gil-Dones F et al., 2012 [12]Protein phosphatase 1 regulatory subunit 3EQ9H7J1Alvarez-Llamas G et al., 2013 [10]
Complement C4-BP0C0L5Gil-Dones F et al., 2012 [12]Protein S100-A6P06703Martin-Rojas T et al., 2015 [11]
Complement component C9P02748Gil-Dones F et al., 2012 [12]ProthrombinP00734Gil-Dones F et al., 2012 [12]
Complement factor HP08603Gil-Dones F et al., 2012 [12]Serine protease HTRA1Q92743Alvarez-Llamas G et al., 2013 [10]
Complement factor H-related protein 1Q03591Gil-Dones F et al., 2012 [12]SerotransferrinP02787Martin-Rojas T et al., 2015 [11]
Complement factor IP05156Gil-Dones F et al., 2012 [12]Serum albuminP02768Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]
Cystatin-CP01034Alvarez-Llamas G et al., 2013 [10]Serum amyloid P-componentP02743Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]
DecorinP07585Martin-Rojas T et al., 2015 [11]Serum paraoxonase/arylesterase 1P27169Gil-Dones F et al., 2012 [12]
EGF-containing fibulin-like extracellular matrix protein 1Q12805Alvarez-Llamas G et al., 2013 [10]Serum paraoxonase/lactonase 3Q15166Alvarez-Llamas G et al., 2013 [10]
EndoplasminP14625Martin-Rojas T et al., 2015 [11]Spondin-1Q9HCB6Alvarez-Llamas G et al., 2013 [10]
Extracellular superoxide dismutase [Cu-Zn]P08294Martin-Rojas T et al., 2012 [13]Superoxide dismutase [Cu-Zn]P00441Martin-Rojas T et al., 2015 [11]
Fatty acid-binding protein, adipocyteP15090Martin-Rojas T et al., 2012 [13]Superoxide dismutase [Mn], mitochondrialP04179Martin-Rojas T et al., 2015 [11]
Fibrinogen alpha chainP02671Gil-Dones F et al., 2012 [12]SWI/SNF complex subunit SMARCC1Q92922Alvarez-Llamas G et al., 2013 [10]
Fibrinogen beta chainP02675Gil-Dones F et al., 2012 [12]Tenascin-XP22105Alvarez-Llamas G et al., 2013 [10]
Fibrinogen gamma chainP02679Martin-Rojas T et al., 2012 [13]; Gil-Dones F et al., 2012 [12]Thrombospondin-1P07996Alvarez-Llamas G et al., 2013 [10]
FibronectinP02751Alvarez-Llamas G et al., 2013 [10]Transforming growth factor-beta-induced protein ig-h3Q15582Martin-Rojas T et al., 2015 [11]
Ficolin-2Q15485Gil-Dones F et al., 2012 [12]TransgelinQ01995Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]
Follistatin-related protein 3O95633Alvarez-Llamas G et al., 2013 [10]TransthyretinP02766Martin-Rojas T et al., 2012 [13]
FRAS1-related extracellular matrix protein 2Q5SZK8Alvarez-Llamas G et al., 2013 [10]Triosephosphate isomeraseP60174Martin-Rojas T et al., 2015 [11]
Galectin-1P09382Martin-Rojas T et al., 2015 [11]Tubulin beta chainP07437Martin-Rojas T et al., 2015 [11]
GelsolinP06396Alvarez-Llamas G et al., 2013 [10]VimentinP08670Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]
Glutathione peroxidase 3P22352Martin-Rojas T et al., 2012 [13]; Martin-Rojas T et al., 2015 [11]VitronectinP04004Gil-Dones F et al., 2012 [12]
Table 2. Category division of ANN score, in decreasing order, according to probability of being a true positive result.
Table 2. Category division of ANN score, in decreasing order, according to probability of being a true positive result.
ANN CategoryANN ScoreAssociated p-Value
Strong relationshipVery high>92<0.01
High78–920.01–0.05
Medium-High63–780.05–0.15
Medium relationshipMedium38–630.15–0.25
Low or no relationshipLow<38>0.25
Table 3. Clinical characteristics of the subjects in the study: M/F, male/female; AHT, arterial hypertension; IHD, ischemic heart disease; BMI, body mass index.
Table 3. Clinical characteristics of the subjects in the study: M/F, male/female; AHT, arterial hypertension; IHD, ischemic heart disease; BMI, body mass index.
ControlsSevere ASp-Value
Age67.76 ± 10.0379.94 ± 7.210.000
Gender (M/F)11/79/90.584
BMI28.48 ± 4.6226.92 ± 3.670.282
AHT (Yes,%)9 (50%)12 (67%)0.406
Dyslipidemia (Yes, %)5 (28%)9 (50%)0.265
Diabetes (Yes, %)0 (0%)0 (0%)1.000
Smokers (Yes, %)2 (11%)0 (0%)0.584
Pneumopathy (Yes, %)0 (0%)1 (5%)0.791
Chronic IHD (Yes, %)0 (0%)0 (0%)1.000
Table 4. Categorization of the ANN score according to the probability of being a true positive result, showing the number of proteins with a strong relationship in each category. DAS, degenerative AS.
Table 4. Categorization of the ANN score according to the probability of being a true positive result, showing the number of proteins with a strong relationship in each category. DAS, degenerative AS.
Very HighHighMedium-High
EffectorsNo EffectorsEffectorsNo EffectorsEffectorsNo EffectorsTOTAL
DAS general characterization--7-131535
1. Calcification--411410
2. Lipoprotein accumulation----21012
3. Inflammation--3511120
4. Oxidative stress--01337
5. Endothelial dysfunction--5221322
6. RAA system----145
7. Hypertrophy--11-810
8. Myocardial fibrosis1-311410
Table 5. Protein candidates that display a strong relationship with at least 3 degenerative AS motives, arranged in decreasing order of the highest ANN score. Proteins that were not described in the molecular characterization and are not considered effector proteins are in bold and highlighted.
Table 5. Protein candidates that display a strong relationship with at least 3 degenerative AS motives, arranged in decreasing order of the highest ANN score. Proteins that were not described in the molecular characterization and are not considered effector proteins are in bold and highlighted.
Uniprot IDGene NameProtein NameMotive EffectorANN ScoreRelated Motive
P08123COL1A2Collagen alpha-2(I) chainYes92.52Myocardial fibrosis
No90.86Inflammation
Yes87.50Endothelial dysfunction
Yes69.82DAS General
P35625TIMP3Metalloproteinase inhibitor 3Yes91.85Endothelial dysfunction
Yes82.53DAS General
No69.20Calcification
No63.61RAA system
P02461COL3A1Collagen alpha-1(III) chainYes91.69Myocardial fibrosis
Yes87.29Endothelial dysfunction
Yes76.47DAS General
P01033TIMP1Metalloproteinase inhibitor 1Yes91.45Endothelial dysfunction
Yes84.60DAS General
No68.80Calcification
P05231IL6Interleukin-6Yes90.45Myocardial fibrosis
Yes88.05Calcification
Yes87.64Inflammation
Yes73.24DAS General
P01042KNG1Kininogen-1Yes90.45DAS General
Yes87.34Inflammation
No70.10RAA system
No63.40Calcification
P07339CTSDCathepsin DYes87.00DAS General
Yes84.46Endothelial dysfunction
No71.66Hypertrophy
P21810BGNBiglycanYes86.97Inflammation
No84.26Calcification
No83.08Myocardial fibrosis
Yes80.74DAS General
No78.98Endothelial dysfunction
No70.50Lipoprotein accumulation
P14625HSP90B1EndoplasminNo86.48Inflammation
No74.62Endothelial dysfunction
No70.58DAS General
No65.44Lipoprotein accumulation
P01019AGTAngiotensinogenYes84.81DAS General
Yes82.54Myocardial fibrosis
Yes78.60Hypertrophy
Yes65.54RAA system
P08253MMP272 kDa type IV collagenaseYes84.40Calcification
Yes75.40Endothelial dysfunction
Yes74.10Myocardial fibrosis
Yes73.98DAS General
No71.88Hypertrophy
No64.52Oxidative stress
P02766TTRTransthyretinNo79.18Oxidative stress
No74.11Inflammation
No71.21DAS General
P00441SOD1Superoxide dismutase [Cu-Zn]No78.56Inflammation
Yes74.41DAS General
Yes69.12Oxidative stress
P10909CLUClusterinNo76.50Inflammation
No67.68Endothelial dysfunction
No64.01Lipoprotein accumulation
P07996THBS1Thrombospondin-1No76.17Endothelial dysfunction
No71.70Myocardial fibrosis
No63.52RAA system
P02768ALBSerum albuminNo76.03Inflammation
No75.65Endothelial dysfunction
No68.55Hypertrophy
No68.34Lipoprotein accumulation
Q00653NFKB2Nuclear factor NF-kappa-B p100 subunitYes75.62Inflammation
Yes74.83Calcification
Yes72.15DAS General
P04114APOBApolipoprotein B-100No75.04Endothelial dysfunction
No74.02Inflammation
Yes71.61DAS General
Yes64.60Lipoprotein accumulation
P02647APOA1Apolipoprotein A-IYes74.73DAS General
No72.41Endothelial dysfunction
Yes64.68Lipoprotein accumulation
P07585DCNDecorinNo74.42Calcification
No73.11Endothelial dysfunction
No71.71Hypertrophy
No68.53Myocardial fibrosis
Q92743HTRA1Serine protease HTRA1No72.82Myocardial fibrosis
No72.73Endothelial dysfunction
No70.23DAS General
P01023A2MAlpha-2-macroglobulinNo72.55Oxidative stress
No70.52Myocardial fibrosis
No68.56Inflammation
No68.30Endothelial dysfunction
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Corbacho-Alonso, N.; Sastre-Oliva, T.; Corros, C.; Tejerina, T.; Solis, J.; López-Almodovar, L.F.; Padial, L.R.; Mourino-Alvarez, L.; Barderas, M.G. Prioritization of Candidate Biomarkers for Degenerative Aortic Stenosis through a Systems Biology-Based In-Silico Approach. J. Pers. Med. 2022, 12, 642. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm12040642

AMA Style

Corbacho-Alonso N, Sastre-Oliva T, Corros C, Tejerina T, Solis J, López-Almodovar LF, Padial LR, Mourino-Alvarez L, Barderas MG. Prioritization of Candidate Biomarkers for Degenerative Aortic Stenosis through a Systems Biology-Based In-Silico Approach. Journal of Personalized Medicine. 2022; 12(4):642. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm12040642

Chicago/Turabian Style

Corbacho-Alonso, Nerea, Tamara Sastre-Oliva, Cecilia Corros, Teresa Tejerina, Jorge Solis, Luis F. López-Almodovar, Luis R. Padial, Laura Mourino-Alvarez, and Maria G. Barderas. 2022. "Prioritization of Candidate Biomarkers for Degenerative Aortic Stenosis through a Systems Biology-Based In-Silico Approach" Journal of Personalized Medicine 12, no. 4: 642. https://0-doi-org.brum.beds.ac.uk/10.3390/jpm12040642

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop