Next Article in Journal
Conventional Laparoscopy versus Robotic-Assisted Aortic Lymph-Nodal Staging for Locally Advanced Cervical Cancer: A Systematic Review and Meta-Analysis
Next Article in Special Issue
N-Terminal of the Prohormone Brain Natriuretic Peptide Predicts Postoperative Cardiogenic Shock Requiring Extracorporeal Membrane Oxygenation
Previous Article in Journal
Correlation of Tumor Pathology with Fluorescein Uptake and MRI Contrast-Enhancement in Stereotactic Biopsies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Implication of Lipids in Calcified Aortic Valve Pathogenesis: Why Did Statins Fail?

1
Department of Cell Biology and Molecular Medicine, Rutgers University, Newark, NJ 07103, USA
2
Department of Medicine, Cardiovascular Research Institute, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
3
Thomas Jefferson University East Falls, Philadelphia, PA 19144, USA
4
Department of Pharmaceutical Sciences, Division of Molecular and Systems Toxicology, University of Basel, 4056 Basel, Switzerland
5
L’institut Du Thorax, Nantes Université, CNRS, INSERM, F-44000 Nantes, France
6
Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(12), 3331; https://doi.org/10.3390/jcm11123331
Submission received: 29 March 2022 / Revised: 1 June 2022 / Accepted: 6 June 2022 / Published: 10 June 2022
(This article belongs to the Special Issue Valvular Heart Disease: From Basic to Clinical Advances)

Abstract

:
Calcific Aortic Valve Disease (CAVD) is a fibrocalcific disease. Lipoproteins and oxidized phospholipids play a substantial role in CAVD; the level of Lp(a) has been shown to accelerate the progression of valve calcification. Indeed, oxidized phospholipids carried by Lp(a) into the aortic valve stimulate endothelial dysfunction and promote inflammation. Inflammation and growth factors actively promote the synthesis of the extracellular matrix (ECM) and trigger an osteogenic program. The accumulation of ECM proteins promotes lipid adhesion to valve tissue, which could initiate the osteogenic program in interstitial valve cells. Statin treatment has been shown to have the ability to diminish the death rate in subjects with atherosclerotic impediments by decreasing the serum LDL cholesterol levels. However, the use of HMG-CoA inhibitors (statins) as cholesterol-lowering therapy did not significantly reduce the progression or the severity of aortic valve calcification. However, new clinical trials targeting Lp(a) or PCSK9 are showing promising results in reducing the severity of aortic stenosis. In this review, we discuss the implication of lipids in aortic valve calcification and the current findings on the effect of lipid-lowering therapy in aortic stenosis.

1. Introduction—Calcific Aortic Valve Disease (CAVD)

Calcific Aortic Valve Disease (CAVD) is the most prevalent heart valve disorder in developed countries; it is a progressive fibrocalcific disease [1,2]. Different risk factors, such as age, male gender, hypertension, metabolic syndrome, diabetes, and bicuspid aortic valve (BAV), have been associated with CAVD [1]. The pathophysiology of CAVD is complex, involving several pathological processes including lipid retention, oxidation, chronic inflammation, fibrotic remodeling, and calcification. Histological analysis of explanted calcified aortic valves revealed the presence of osteoblast-like cells, chondrocytes [3], and inflammatory cells [4,5]. Inflammation and growth factors play an important role in promoting extracellular matrix (ECM) synthesis and in triggering the activation of the osteogenic program in valvular interstitial cells (VICs) [6,7,8,9]. Indeed, proteomics analysis of explanted human pathological aortic valves showed significant disruption of ECM components [7]. Proteoglycans, such as biglycan and decorin, play a major role in lipid retention and modification in CAVD [8,10,11]. In this regard, biglycan, which is an endogenous agonist of Toll-like receptor 2 (TLR2), has been shown to promote inflammation and the osteogenic transdifferentiation of VICs [11].
Moreover, genome-wide association and Mendelian randomization studies have pointed out the association of low-density lipoproteins (LDLs) and lipoprotein(a) (Lp(a)) with the development of CAVD [12,13]. In this regard, several mechanistic studies have confirmed the implication of Lp(a) and its lipid content in the activation of the osteogenic program of VICs [14,15,16].
Although lipids seem to be a key factor in the pathogenesis of CAVD, three randomized clinical trials (RCTs) failed to demonstrate any significant benefit of LDL lowering with statins on the progression of aortic stenosis [17,18,19]. Furthermore, plasma levels of Lp(a) are not significantly modified by statin therapy [20]. Therefore, there is significant interest in targeting Lp(a) levels with novel therapeutic agents, such as antisense oligonucleotides, to slow the progression of CAVD [21,22,23]. Furthermore, Langsted et al. (2016) found that patients with a PCSK9 loss-of-function mutation (PCSK9 R46L) have lower serum LDL and Lp(a) levels and a lower risk of calcific aortic stenosis based on data from 103,083 people in the Copenhagen General Population Study [24]. Herein, we examine the implication of lipids in CAVD. In addition, we have placed special emphasis on statins’ failure to reduce the progression of aortic stenosis.

2. Role of Lipoprotein(a) and Oxidized Phospholipids in CAVD

2.1. The Implication of Nitric Oxide (NO) Activity in CAVD

Endothelial nitric oxide synthase (eNOS) uncoupling appears to be one central mechanism during early-stage disease and contributes to CAVD progression [25]. Indeed, NO synthesis and signaling are markedly affected by the oscillatory shear stress in the endothelium lining the calcification-prone fibrosa compared with the disease-resilient ventricularis [25]. In vivo studies have shown that a lack of eNOS in mice promotes a CAVD-like phenotype [26], whereas restoring paracrine NO signaling blunts VIC-driven calcification in different experimental models [27]. Furthermore, studies have also reported that NO maintains valvular homeostasis through guanylyl cyclase/cGMP- and NOTCH1-dependent mechanisms [28,29]. More recently, Majumdar et al. (2021) deepened mechanistic insights by showing that valvular endothelial cell (VEC)-derived NO rescues calcification by an S-nitrosylation-mediated mechanism in porcine aortic valve interstitial cells. The alteration of nitric oxide (NO) signaling and activity in CAVD induces reactive oxygen species generation, which may promote the production of oxidative lipid species, triggering inflammation signaling activation in the aortic valve [27]. Targeting eNOS may open new therapeutic avenues to restore the paracrine homeostasis of endothelial/valve interstitial cells [29].

2.2. The LPA Gene Locus and CAVD

The LPA gene locus determines circulating Lp(a) levels primarily, with no significant dietary or environmental influences (Thanassoulis et al., 2013). The LPA gene is located on chromosome 6 (6q25.3-q26) and has a high degree of homology with the plasminogen gene (PLG). The LPA locus is complex, with copy number variants (CNVs) in the region encoding the kringle IV type 2 (KIV2) domain, which is inversely related to Lp(a) levels in the blood [30]. Genome-wide association (GWA) studies and Mendelian randomization (MR) analyses have underlined a causal association between a common gene variant, rs10455872, located in the LPA gene locus and CAVD [12,31].
However, a recent GWA study did not find an association between LPA and CAVD in patients with a congenital bicuspid aortic valve [31]. A large meta-analysis conducted on 1797 CAVD cases and 131,932 controls revealed that carriers of rs10455872 have a 1.66-fold higher risk of developing CAVD [32]. Furthermore, studies showed that genetically determined lower levels of Lp(a) were associated with a 37% reduced risk of CAVD [33]. These findings suggest that lowering Lp(a) levels and/or blocking the specific pathways by which Lp(a) promotes CAVD could lead to therapies that slow the progression of CAVD.

2.3. Lp(a), a Major Carrier of Oxidized Phospholipids (Ox-PL), Is a Risk Factor for Aortic Stenosis

Lp(a) is composed of low-density lipoprotein (LDL)-like particles in which apolipoproteinB-100 (apoB) is covalently bound by a single disulfide bond to the glycoprotein apolipoprotein(a) (apo(a)) [30,34]. Apolipoprotein(a) is highly polymorphic, with a variable number of (KIV2) domains. The copy number variant of KIV2 domains determines the length of the lipoprotein, and it is inversely related to the level of Lp(a) in circulation. Studies have underlined the high-content oxidized phospholipids (Ox-PL) in Lp(a) particles [35]. Ox-PL binds to the KIV type 10 domain and is thus ferried, as a cargo, by Lp(a). Hence, Ox-PLs carried by Lp(a) may contribute to endothelial dysfunction, inflammation, and the expression of genes with pro-calcifying properties [35].
Ox-PL has been established as a causal risk factor for AS in several genetic and population studies [12,31,36]. The genetically determined level of Ox-PL linked to apo(a) (Ox-PL-apo(a)) increases the risk of CAVD 1.09-fold [37]. Interestingly, clinical imaging with 18F sodium fluoride positron emission tomography or computed tomography has revealed the presence of aortic valve micro-calcification in individuals with elevated Lp(a) prior to the development of clinical manifestations of CAVD (Despres et al., 2019). Together, these studies thus highlight that Lp(a) and its cargo, Ox-PL, are involved in the development of CAVD.

2.4. Lipid Oxidation Promotes Calcific Aortic Stenosis

Increasing evidence suggests that the infiltration of lipoproteins into the aortic valve plays a central role in promoting inflammation, which, in turn, might induce the activation of the osteogenic program in VICs [14,38]. Histological analysis of explanted calcified aortic valves has revealed the presence of several apolipoproteins (apo), such as apoB, apoE, apoA1, apolipoprotein E, and apo(a) [39,40,41]. Furthermore, there is an association between the level of Ox-LDL, the degree of inflammation, and fibrocalcific remodeling [42,43].
In vitro studies showed that Ox-LDL and several oxidized phospholipid (Ox-PL) species carried by the Lp(a) fraction promote the calcification of VICs [44]. In turn, lipoprotein-associated phospholipase A2 (Lp-PLA2) transforms Ox-PLs into lysophosphatidylcholine (LysoPC), which acts as a reactive metabolite that promotes the mineralization of VICs [45]. Immunohistochemical studies have highlighted the co-localization of Lp-PLA2 with Ox-LDL, suggesting that Lp-PLA2 could be transported by lipoproteins into the aortic valve [45,46,47]. Together, these studies suggest that the accumulation of oxidized lipids triggers osteogenic response activation in VICs [35] (Figure 1).

3. Autotaxin (ATX)–Lysophosphatidic Acid (LysoPA) Axis Mediates Mineralization of the Aortic Valve

Autotaxin (ATX) is a member of the ecto-nucleotidase family of enzymes encoded by the ENPP2 gene [48]. It was initially isolated from melanoma cell lines and was identified as a motility factor [49]. ATX is a secreted glycoprotein that hydrolyzes lysophosphatidylcholine (LysoPC) into lysophosphatidic acid (LysoPA). LysoPA is an active metabolite with potent and diverse biological properties. It promotes cell motility, inflammation, calcification, and fibrosis [49]. It is believed that the majority of circulating LysoPA is derived from ATX [50]. According to Bouchareb et al. (2015), ATX is likely transported into the aortic valve by Lp(a), and it is also secreted by VICs in response to inflammatory stimuli [15]. Indeed, ATX activity is enriched in an isolated fraction of Lp(a). Moreover, binding assay analysis, using human purified Lp(a), confirmed the physical association between Lp(a) and ATX [51]. In vitro inhibition of ATX prevented the mineralization of VICs induced by LysoPC, suggesting that LysoPA is the mediator promoting the activation of the osteogenic program in VICs [15]. Of interest, ATX expression and activity were increased in human explanted pathological aortic valves [15]. To this effect, stimulation of VICs with LysoPC and Ox-PLs treatment induces the expression of ATX [15,38]. Moreover, the administration of LysoPA to LDR-/- apoB100/100 IGFII mice increased the osteogenic activity in the aortic valve and accelerated the development of CAVD [15,38]. Following a series of in-depth investigations, it has been shown that ATX and LysoPA promote aortic valve inflammation and mineralization through the activation of the NF-κB/bone morphogenetic 2 (BMP2) pathway [15,38]. In this regard, a significant interaction term was found between ATX activity and Lp(a) level [16]. Together, these studies indicate that ATX is carried by Lp(a) and is also secreted by VICs, increasing LysoPA levels and, therefore, stimulating inflammation.
More recently, Bouchareb et al. (2019) also showed that activated platelets promote VIC mineralization in vitro through the activation of ATX. In addition, ATX activity was higher in platelets from patients with CAVD compared to control patients (Bouchareb et al., 2019).
ATX promotes inflammation and the osteogenic transdifferentiation of VICs through the production of LysoPA, which is a small lipid derivative acting on G-protein coupled receptors with various biological functions [38]. In vitro studies have shown that oxidized LDL (Ox-LDL) induces the mineralization of VIC cultures, whereas treatment with an antagonist of LPAR1 prevents this effect [38]. The same study has also highlighted the overexpression of LPAR1 in human calcified aortic valves. In vitro studies using human VICs showed that lysoPA treatment stimulates the expression of the bone morphogenetic protein (BMP2) via the activation of the NFkB pathway. The promoter region of BMP2 contains NFkB-responsive elements and LysoPA promotes the phosphorylation of p65 on serine 536 (p65 S536). Of particular interest, phosphorylated p65 S536 was recruited to the promoter of BMP2 to activate BMP2 gene expression [38]. The pharmacological inhibition of LPAR1 with Ki16425 in LDLR-/- apoB 100/100 IGFII mice reduced the progression of CAVD and downregulated the expression of BMP2 in aortic valve cusps [38].
Lastly, Mkannez et al. (2018) [52] have recently underlined that the expression and the enzymatic activity of PLPP3 (also known as PPAP2B), a phospholipid phosphatase that inactivates LysoPA, were decreased in human calcified aortic valves compared to controls. Consistently, aortic valves with lower expression of PLPP3 had an increased level of LysoPA [52]. Furthermore, the knockdown of PLPP3 exacerbated the LysoPA-induced expression of BMP2 and consequently simulated in vitro VICs’ mineralization [52].

4. The Implication of apoC-III in the Calcification of the Aortic Valve

Metabolic syndrome is described as a dysmetabolism related to insulin resistance and visceral obesity, which leads to a pro-inflammatory and pro-thrombotic state [53]. This syndrome has been associated with the increased incidence and progression of aortic valve calcification [54] and hemodynamic progression of aortic valve stenosis [55], making the visceral obesity-related perturbations a potential target to reduce the development and progression of CAVD.
One of the main features of metabolic syndrome is hypertriglyceridemia. The apolipoprotein C-III (apoC-III) is associated with elevated triglyceride levels. As opposed to other apolipoproteins, such as apo(a) or apoB, multiple particles of ApoC-III are carried by all lipoproteins [56,57]. This finding supports the use of ApoC-III as a potential biomarker, independent of the other lipid factors, and a potential therapeutic target. Indeed, ApoC-III’s circulating levels have been linked to an increased risk of cardiovascular events, and targeting this apolipoprotein is a promising way to lower the risk for these patients [58,59,60,61].
ApoC-III is described as a multifunctional protein, playing a role in the metabolism of several lipoproteins, glucose homeostasis, endothelial cell dysfunction, inflammation, and the coagulation cascade, and in increasing lipoprotein affinity to the extracellular matrix (especially proteoglycans) [57]. Interestingly, all these features have also been associated with the development and/or progression of CAVD and support a potential detrimental action of this apolipoprotein in the pathogenesis of CAVD. This was further studied in a post-hoc analysis of the ASTRONOMER trial, where the Lp(a) content in ApoC-III has been related to faster CAVD progression [61]. As previously stated, this study provides evidence that, in addition to Lp(a) plasma levels, the content of this lipoprotein would be of great interest in understanding the mechanisms leading to aortic valve calcification and the development of aortic stenosis. Further studies focused on these aspects are needed and, by compiling data, would provide mechanistic evidence to target these lipoproteins in the context of CAVD.

5. The Effect of Lipid-Lowering Therapy in Aortic Stenosis

5.1. Rationale of Statins

Statins or the hydroxymethylglutaryl-CoA reductase (HMG-CoA) inhibitors are considered a potent therapeutic strategy in patients with atherosclerotic plaques. Statins are unquestionably well documented for their lipid-lowering effects [62,63]. Indeed, statins have demonstrated an improved survival rate in subjects with atherosclerotic coronary heart disease (CHD) [23]. They target the HMG-CoA reductase that catalyzes the switching of HMG-CoA to mevalonic acid, which abolishes the production of cholesterol [64]. The inhibition of HMG-CoA reduces cholesterol levels through the up-regulation of hepatocyte-LDL receptors, thus increasing the uptake of circulating LDL-cholesterol into the hepatocytes and subsequently decreasing LDL levels [65].

5.2. Statins’ Mechanism of Action

Statins obstruct HMG-CoA reductases, the pivotal catalytic enzyme in the cholesterol biosynthesis pathway (Figure 2 and Figure 3). This enzyme predominantly regulates the conversion of HMG-CoA synthases to mevalonic acid and, hence, in the course of action, manifests a decline in the plasma LDL levels [62]. Primarily, the lipid-lowering pleiotropic effects of statins are supposedly based on hampering the production of significant isoprenoid intermediates such as farnesyl pyrophosphate and geranylgeranyl pyrophosphate in the cholesterol biosynthesis pathway [66,67,68,69]. In addition, they were proven to block the destabilizing effects of mevalonate on nitric oxide synthase (NOS)-mRNA in human endothelial cells (ECs), thereby resulting in the increased synthesis and function of the NOS enzyme [23].

5.3. Statin-Mediated Lipid-Lowering Therapeutic Approaches to Target CAVD: Past, Present, and Future Prospects

The existence of similarities in the pathophysiology of atherosclerosis and aortic stenosis has led the clinical and research community to consider statins as a treatment to decelerate the progression or to reduce the severity of aortic stenosis. Indeed, studies are showing the implication of Ox-LDL in the activation of osteogenic transition in calcified aortic valves [5,16,38,45]. In light of the facts presented, these results elicited considerable interest among the scientific community that eventually paved the way to the initiation of numerous randomized controlled clinical trials to elucidate the effect of a lipid-lowering therapeutic regimen in aortic stenosis. Clinical studies, however, found no benefit of statins in terms of the hemodynamic progression or disease severity of aortic stenosis [17,18]. In the Simvastatin and Ezetimibe in Aortic Stenosis (SEAS) trial, patients were randomized to receive simvastatin 40 mg plus ezetimibe 10 mg, or a placebo [18,70]. The drug combination significantly lowered LDL levels compared to the placebo. However, no effect on the progression of aortic stenosis was observed [70]. To further complicate the situation, another meta-analysis study by Teo et al. [71] using randomized placebo-controlled clinical trials on 2344 patients reported no differences in clinical outcomes between the placebo and the treatment group. As a result of these negative outcomes of the conducted trials, the American Heart Association/American College of Cardiology and the European Society of Cardiology guidelines together do not endorse the use of statins for the treatment of CAVD [72,73].
Nonetheless, the Aortic Stenosis Progression Observation: Measuring Effects of Rosuvastatin (ASTRONOMER) study showed an association between elevated levels of oxidized phospholipids and lipoprotein(a) (Lp(a)) in patients with accelerated hemodynamic progression of CAVD [74]. These findings support the hypothesis that Lp(a) mediates AS progression through its binding to Ox-PL [15,37,74]. These findings paved the way to the implementation of randomized clinical trials focusing on Lp(a)-lowering therapy in mild-to-moderate CAVS patients with elevated Lp(a) plasma levels (Figure 4).
Two randomized, double-blind, placebo-controlled trials are ongoing. In patients with elevated Lp(a), phase 1 and 2 trials demonstrated the tolerability, safety, and beneficial effect of lowering Lp(a) concentrations with IONIS-APO(a)Rx, an oligonucleotide targeting apolipoprotein(a). This therapeutic agent was found to reduce circulating Lp(a) by 80% (Koren et al., 2022; Tsimikas et al., 2015; Viney et al., 2016). A phase 3 clinical trial (Lp(a) HORIZON trial; NCT04023552) is now underway to assess the impact of this treatment on clinical outcomes in patients with established cardiovascular disease and elevated Lp(a) plasma levels.
Proprotein convertase subtilisin/Kexin type 9 (PCSK9), an enzyme that is formed in the liver, has been described as a central player in cholesterol metabolism, particularly because PCSK9 stimulates the degradation of the LDL receptor and then leads to an increase in circulating LDL [75]. Inhibition of PCSK9 and its associated drastic reduction in circulating LDL suggests a promising treatment for patients with cardiovascular diseases [76,77]. In addition to lowering LDL, PCSK9 inhibitors were associated with a 15 to 30% reduction in Lp(a) plasma levels, which may benefit CAVD patients. Indeed, Langsted et al. [24] have shown, in 103,083 individuals from the Copenhagen General Population Study, that patients with loss-of-function mutation of PCSK9 (PCSK9 R46L) are associated with decreased serum LDL and Lp(a) levels and a reduced risk of calcific aortic stenosis (CAVD). With this underlying substantial evidence, the PCSK9 inhibitors (deliberated as monoclonal antibodies: alirocumab and evolocumab) are currently being examined as pharmacological routes to delay the progression of aortic stenosis [78]. The exploratory investigations from the Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk (FOURIER) randomized clinical trial support this approach. In this post-hoc analysis, patients randomized to evolocumab had a 50 % decrease in the incidence of CAVS over a median follow-up of 2.2 (1.8–2.5) years [79]. Furthermore, recent experimental studies [75,80] have shown that PCSK9 could be involved in the process leading to aortic valve calcification and that the in vitro inhibition of PCSK9 could decrease VIC calcification.

5.4. Statins’ Off-Target Effects

Increased evidence points out several off-target effects of statins (Ward et al., 2019), including statin-associated muscle symptoms (SAMS), diabetes mellitus (DM), and effects on the central nervous system (Thompson et al., 2016) (Figure 5).
One of the main overlooked aspects in the context of statins’ myotoxicity is calcium signaling. Studies showed that only lipophilic statins stimulate calcium release from the sarcoplasmic reticulum in rat and human skeletal muscle [81,82]. Acute applications of simvastatin on skeletal muscle fibers increased the cytosolic Ca2+ concentrations released from the sarcoplasmic reticulum [83]. Moreover, a previous study showed that simvastatin impaired ryanodine receptor 1 (RyR1) Ca2+ function, causing aberrant Ca2+ handling, which led ultimately to cell apoptosis [84].
The mevalonate pathway (Figure 2) produces ubiquinone or coenzyme Q10, which is an important player in the mitochondrial electron transport chain (ETC). Studies by Bouitbir and others showed clearly that statins impair mitochondrial function at different levels (Figure 5) [85,86,87,88]. For instance, simvastatin inhibits complex I activity, as observed in rat and human skeletal muscles treated with simvastatin [82]. The inhibition of mitochondrial function was only observed with lipophilic statins and was not rescued by the addition of cholesterol intermediate mevalonate, which would indicate that statins directly impair mitochondrial function, independently of the inhibition of HMG-CoA reductase [88]. Furthermore, patients treated with statins and experiencing muscle-related side effects presented impaired mitochondrial function, lower mitochondrial content, and increased mitochondrial reactive oxygen (ROS) production in deltoid biopsies [86]. However, the same study showed that statins increased mitochondrial function in heart biopsies of the same patients [86]. These findings showed clearly that oxidative muscles such as the heart are more resistant to statins than glycolytic muscles, most likely due to the higher mitochondrial content and higher anti-oxidative capacities (Figure 5).
Akt represents an important kinase governing the homeostasis between cell growth, survival, and metabolism. Several in vitro studies documented an impaired Akt function and its downstream signaling pathway, which lead to cell apoptosis and protein degradation [85,89]. Moreover, it was shown that decreased Akt phosphorylation and impaired mitochondria respiration were responsible for simvastatin-induced myopathy in C2C12 myo-tubes [90].
Recently, new-onset diabetes mellitus has been reported as a new adverse event in patients treated with statins [91]. The incidence of diabetes associated with statin therapy is estimated at up to 30% [92]. The JUPITER trial reported a significant increase in type 2 diabetes in patients treated with rosuvastatin [91]. Moreover, decreased insulin sensitivity and hyperglycemia in hypercholesteremic patients have been observed with atorvastatin [21]. However, the impairment of insulin sensitivity was observed only with lipophilic statins [21]. The impaired insulin signaling and the disturbed GLUT4 synthesis or translocation seem to establish the link between statins and insulin resistance [93]. Moreover, impaired translocation of the GLUT4 vesicles to the cell membrane was observed when cells were exposed to atorvastatin [94]. Statins impair the isoprenylation of several proteins and GTPases by inhibiting cholesterol biosynthesis. Interestingly, this effect was not observed with pravastatin, highlighting a class effect. Because of these various off-target effects and the complex physiopathology of aortic stenosis, statins did not show any beneficial effect on stopping the progression of aortic stenosis.
Finally, the concept of a lipid-lowering therapeutic regimen in CAVS is reassuring. The heterogeneity of lipid species in the aortic valve might explain the failure of statins to reduce the progression of aortic stenosis. However, the clinical trials using antisense oligonucleotides to inhibit the expression of Lp(a) (Figure 4) or the use of monoclonal antibodies to inhibit the expression of PCSK9 still support the implication of lipids in Calcified Aortic Valve Disease. The use of lipidomic studies to explore the circulating and valvular lipid species in patients with CAVS might provide molecular cues for more efficient and compelling therapeutic targets to reduce the progression of aortic stenosis.
Figure 5. Off-target effects of statins. Statins act at different levels in skeletal muscle cells. First, they provoke the increased release of calcium from the sarcoplasmic reticulum to the cytoplasm and perturb the contractility of muscle fibers. In parallel, statins impair mitochondrial function, leading to the accumulation of ROS and to the activation of apoptosis. Finally, statins impair the function of Akt due to the impaired insulin signaling pathway and due to the impaired function of mTORC2. As a result, statins induce increased protein degradation and impaired protein synthesis, promoting skeletal muscle atrophy [95].
Figure 5. Off-target effects of statins. Statins act at different levels in skeletal muscle cells. First, they provoke the increased release of calcium from the sarcoplasmic reticulum to the cytoplasm and perturb the contractility of muscle fibers. In parallel, statins impair mitochondrial function, leading to the accumulation of ROS and to the activation of apoptosis. Finally, statins impair the function of Akt due to the impaired insulin signaling pathway and due to the impaired function of mTORC2. As a result, statins induce increased protein degradation and impaired protein synthesis, promoting skeletal muscle atrophy [95].
Jcm 11 03331 g005

Author Contributions

M.J.N. writing the section about aortic valve calcification and lipids; A.D. and J.B. writing section about statins; J.B. and R.B. writing a section on statins off-target; R.C. writing the section on apoCIII and writing the abstract and over read the sections; E.G. designing, supervision and editing of all the sections. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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]
  2. Mathieu, P.; Boulanger, M.C. Basic Mechanisms of Calcific Aortic Valve Disease. Can. J. Cardiol. 2014, 30, 982–993. [Google Scholar] [CrossRef] [PubMed]
  3. Matsui, M.; Bouchareb, R.; Storto, M.; Hussain, Y.; Gregg, A.; Marx, S.O.; Pitt, G.S. Increased Ca2+ influx through CaV1.2 drives aortic valve calcification. JCI Insight 2022, 7, e155569. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, J.H.; Chen, W.L.; Sider, K.L.; Yip, C.Y.; Simmons, C.A. beta-catenin mediates mechanically regulated, transforming growth factor-beta1-induced myofibroblast differentiation of aortic valve interstitial cells. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 590–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bouchareb, R.; Boulanger, M.C.; Fournier, D.; Pibarot, P.; Messaddeq, Y.; Mathieu, P. Mechanical strain induces the production of spheroid mineralized microparticles in the aortic valve through a RhoA/ROCK-dependent mechanism. J. Mol. Cell Cardiol. 2014, 67, 49–59. [Google Scholar] [CrossRef] [PubMed]
  6. Yu, Z.; Seya, K.; Daitoku, K.; Motomura, S.; Fukuda, I.; Furukawa, K. Tumor necrosis factor-alpha accelerates the calcification of human aortic valve interstitial cells obtained from patients with calcific aortic valve stenosis via the BMP2-Dlx5 pathway. J. Pharmacol. Exp. Ther. 2011, 337, 16–23. [Google Scholar] [CrossRef]
  7. Bouchareb, R.; Guauque-Olarte, S.; Snider, J.; Zaminski, D.; Anyanwu, A.; Stelzer, P.; Lebeche, D. Proteomic Architecture of Valvular Extracellular Matrix: FNDC1 and MXRA5 Are New Biomarkers of Aortic Stenosis. JACC Basic Transl. Sci. 2021, 6, 25–39. [Google Scholar] [CrossRef]
  8. Katsi, V.; Magkas, N.; Antonopoulos, A.; Trantalis, G.; Toutouzas, K.; Tousoulis, D. Aortic valve: Anatomy and structure and the role of vasculature in the degenerative process. Acta Cardiol. 2021, 76, 335–348. [Google Scholar] [CrossRef]
  9. Bouchareb, R.; Lebeche, D. Isolation of Mouse Interstitial Valve Cells to Study the Calcification of the Aortic Valve In Vitro. J. Vis. Exp. 2021, 171, e62419. [Google Scholar] [CrossRef]
  10. Dupuis, L.E.; McCulloch, D.R.; McGarity, J.D.; Bahan, A.; Wessels, A.; Weber, D.; Diminich, A.M.; Nelson, C.M.; Apte, S.S.; Kern, C.B. Altered versican cleavage in ADAMTS5 deficient mice; a novel etiology of myxomatous valve disease. Dev. Biol. 2011, 357, 152–164. [Google Scholar] [CrossRef] [Green Version]
  11. Derbali, H.; Bosse, Y.; Cote, N.; Pibarot, P.; Audet, A.; Pepin, A.; Arsenault, B.; Couture, C.; Despres, J.P.; Mathieu, P. Increased biglycan in aortic valve stenosis leads to the overexpression of phospholipid transfer protein via Toll-like receptor 2. Am. J. Pathol. 2010, 176, 2638–2645. [Google Scholar] [CrossRef] [PubMed]
  12. Thanassoulis, G.; Campbell, C.Y.; Owens, D.S.; Smith, J.G.; Smith, A.V.; Peloso, G.M.; Kerr, K.F.; Pechlivanis, S.; Budoff, M.J.; Harris, T.B.; et al. Genetic associations with valvular calcification and aortic stenosis. N. Engl. J. Med. 2013, 368, 503–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Nordestgaard, B.G.; Chapman, M.J.; Ray, K.; Boren, J.; Andreotti, F.; Watts, G.F.; Ginsberg, H.; Amarenco, P.; Catapano, A.; Descamps, O.S.; et al. Lipoprotein(a) as a cardiovascular risk factor: Current status. Eur. Heart J. 2010, 31, 2844–2853. [Google Scholar] [CrossRef]
  14. Yu, B.; Hafiane, A.; Thanassoulis, G.; Ott, L.; Filwood, N.; Cerruti, M.; Gourgas, O.; Shum-Tim, D.; Al Kindi, H.; de Varennes, B.; et al. Lipoprotein(a) Induces Human Aortic Valve Interstitial Cell Calcification. JACC Basic Transl. Sci. 2017, 2, 358–371. [Google Scholar] [CrossRef] [PubMed]
  15. Bouchareb, R.; Mahmut, A.; Nsaibia, M.J.; Boulanger, M.C.; Dahou, A.; Lepine, J.L.; Laflamme, M.H.; Hadji, F.; Couture, C.; Trahan, S.; et al. Autotaxin Derived from Lipoprotein(a) and Valve Interstitial Cells Promotes Inflammation and Mineralization of the Aortic Valve. Circulation 2015, 132, 677–690. [Google Scholar] [CrossRef]
  16. Nsaibia, M.J.; Mahmut, A.; Boulanger, M.C.; Arsenault, B.J.; Bouchareb, R.; Simard, S.; Witztum, J.L.; Clavel, M.A.; Pibarot, P.; Bosse, Y.; et al. Autotaxin interacts with lipoprotein(a) and oxidized phospholipids in predicting the risk of calcific aortic valve stenosis in patients with coronary artery disease. J. Intern. Med. 2016, 280, 509–517. [Google Scholar] [CrossRef] [PubMed]
  17. Cowell, S.J.; Newby, D.E.; Prescott, R.J.; Bloomfield, P.; Reid, J.; Northridge, D.B.; Boon, N.A.; Scottish Aortic, S.; Lipid Lowering Trial, I.o.R.I. A randomized trial of intensive lipid-lowering therapy in calcific aortic stenosis. N. Engl. J. Med. 2005, 352, 2389–2397. [Google Scholar] [CrossRef]
  18. Rossebo, A.B.; Pedersen, T.R.; Boman, K.; Brudi, P.; Chambers, J.B.; Egstrup, K.; Gerdts, E.; Gohlke-Barwolf, C.; Holme, I.; Kesaniemi, Y.A.; et al. Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. N. Engl. J. Med. 2008, 359, 1343–1356. [Google Scholar] [CrossRef] [Green Version]
  19. Chan, K.L.; Teo, K.; Dumesnil, J.G.; Ni, A.; Tam, J.; Investigators, A. Effect of Lipid lowering with rosuvastatin on progression of aortic stenosis: Results of the aortic stenosis progression observation: Measuring effects of rosuvastatin (ASTRONOMER) trial. Circulation 2010, 121, 306–314. [Google Scholar] [CrossRef] [Green Version]
  20. Tsimikas, S.; Gordts, P.; Nora, C.; Yeang, C.; Witztum, J.L. Statin therapy increases lipoprotein(a) levels. Eur. Heart J. 2020, 41, 2275–2284. [Google Scholar] [CrossRef]
  21. Koh, K.K.; Quon, M.J.; Han, S.H.; Lee, Y.; Kim, S.J.; Shin, E.K. Atorvastatin causes insulin resistance and increases ambient glycemia in hypercholesterolemic patients. J. Am. Coll. Cardiol. 2010, 55, 1209–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Greco, M.F.; Sirtori, C.R.; Corsini, A.; Ezhov, M.; Sampietro, T.; Ruscica, M. Lipoprotein(a) Lowering-From Lipoprotein Apheresis to Antisense Oligonucleotide Approach. J. Clin. Med. 2020, 9, 2103. [Google Scholar] [CrossRef] [PubMed]
  23. Laufs, U.; Fata, V.L.; Liao, J.K. Inhibition of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase blocks hypoxia-mediated down-regulation of endothelial nitric oxide synthase. J. Biol. Chem. 1997, 272, 31725–31729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Langsted, A.; Nordestgaard, B.G.; Benn, M.; Tybjaerg-Hansen, A.; Kamstrup, P.R. PCSK9 R46L Loss-of-Function Mutation Reduces Lipoprotein(a), LDL Cholesterol, and Risk of Aortic Valve Stenosis. J. Clin. Endocrinol. Metab. 2016, 101, 3281–3287. [Google Scholar] [CrossRef]
  25. Kraler, S.; Garg, V.; Akhmedov, A. Calcific aortic valve disease: Novel insights into nitric oxide signalling. Eur. Heart J. 2022, 43, 1665–1667. [Google Scholar] [CrossRef]
  26. El Accaoui, R.N.; Gould, S.T.; Hajj, G.P.; Chu, Y.; Davis, M.K.; Kraft, D.C.; Lund, D.D.; Brooks, R.M.; Doshi, H.; Zimmerman, K.A.; et al. Aortic valve sclerosis in mice deficient in endothelial nitric oxide synthase. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H1302–H1313. [Google Scholar] [CrossRef] [Green Version]
  27. Majumdar, U.; Manivannan, S.; Basu, M.; Ueyama, Y.; Blaser, M.C.; Cameron, E.; McDermott, M.R.; Lincoln, J.; Cole, S.E.; Wood, S.; et al. Nitric oxide prevents aortic valve calcification by S-nitrosylation of USP9X to activate NOTCH signaling. Sci. Adv. 2021, 7, eabe3706. [Google Scholar] [CrossRef]
  28. Gould, S.T.; Matherly, E.E.; Smith, J.N.; Heistad, D.D.; Anseth, K.S. The role of valvular endothelial cell paracrine signaling and matrix elasticity on valvular interstitial cell activation. Biomaterials 2014, 35, 3596–3606. [Google Scholar] [CrossRef] [Green Version]
  29. Kraler, S.; Blaser, M.C.; Aikawa, E.; Camici, G.G.; Luscher, T.F. Calcific aortic valve disease: From molecular and cellular mechanisms to medical therapy. Eur. Heart J. 2022, 43, 683–697. [Google Scholar] [CrossRef]
  30. Koschinsky, M.L.; Beisiegel, U.; Henne-Bruns, D.; Eaton, D.L.; Lawn, R.M. Apolipoprotein(a) size heterogeneity is related to variable number of repeat sequences in its mRNA. Biochemistry 1990, 29, 640–644. [Google Scholar] [CrossRef]
  31. Arsenault, B.J.; Boekholdt, S.M.; Dube, M.P.; Rheaume, E.; Wareham, N.J.; Khaw, K.T.; Sandhu, M.S.; Tardif, J.C. Lipoprotein(a) levels, genotype, and incident aortic valve stenosis: A prospective Mendelian randomization study and replication in a case-control cohort. Circ. Cardiovasc. Genet. 2014, 7, 304–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Cairns, B.J.; Coffey, S.; Travis, R.C.; Prendergast, B.; Green, J.; Engert, J.C.; Lathrop, M.; Thanassoulis, G.; Clarke, R. A Replicated, Genome-Wide Significant Association of Aortic Stenosis with a Genetic Variant for Lipoprotein(a): Meta-Analysis of Published and Novel Data. Circulation 2017, 135, 1181–1183. [Google Scholar] [CrossRef] [PubMed]
  33. Enas, E.A.; Varkey, B.; Dharmarajan, T.S.; Pare, G.; Bahl, V.K. Lipoprotein(a): An independent, genetic, and causal factor for cardiovascular disease and acute myocardial infarction. Indian Heart J. 2019, 71, 99–112. [Google Scholar] [CrossRef] [PubMed]
  34. Cesaro, A.; Schiavo, A.; Moscarella, E.; Coletta, S.; Conte, M.; Gragnano, F.; Fimiani, F.; Monda, E.; Caiazza, M.; Limongelli, G.; et al. Lipoprotein(a): A genetic marker for cardiovascular disease and target for emerging therapies. J. Cardiovasc. Med. 2021, 22, 151–161. [Google Scholar] [CrossRef] [PubMed]
  35. Chignon, A.; Bon-Baret, V.; Boulanger, M.C.; Bossé, Y.; Mathieu, P. Oxyphospholipids in Cardiovascular Calcification. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 11–19. [Google Scholar] [CrossRef] [PubMed]
  36. Kamstrup, P.R.; Tybjaerg-Hansen, A.; Nordestgaard, B.G. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general population. J. Am. Coll. Cardiol. 2014, 63, 470–477. [Google Scholar] [CrossRef] [Green Version]
  37. Kamstrup, P.R.; Hung, M.Y.; Witztum, J.L.; Tsimikas, S.; Nordestgaard, B.G. Oxidized Phospholipids and Risk of Calcific Aortic Valve Disease: The Copenhagen General Population Study. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1570–1578. [Google Scholar] [CrossRef] [Green Version]
  38. Nsaibia, M.J.; Boulanger, M.C.; Bouchareb, R.; Mkannez, G.; Le Quang, K.; Hadji, F.; Argaud, D.; Dahou, A.; Bosse, Y.; Koschinsky, M.L.; et al. OxLDL-derived lysophosphatidic acid promotes the progression of aortic valve stenosis through a LPAR1-RhoA-NF-kappaB pathway. Cardiovasc. Res. 2017, 113, 1351–1363. [Google Scholar] [CrossRef] [Green Version]
  39. 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]
  40. Otto, C.M.; Kuusisto, J.; Reichenbach, D.D.; Gown, A.M.; O’Brien, K.D. Characterization of the early lesion of ‘degenerative’ valvular aortic stenosis. Histological and immunohistochemical studies. Circulation 1994, 90, 844–853. [Google Scholar] [CrossRef] [Green Version]
  41. Gebhard, C.; Maafi, F.; Stahli, B.E.; Dang, J.; Nachar, W.; de Oliveira Moraes, A.B.; Kernaleguen, A.E.; Lavoie, V.; Mecteau, M.; Mihalache-Avram, T.; et al. Apolipoprotein A-I proteolysis in aortic valve stenosis: Role of cathepsin S. Basic Res. Cardiol. 2018, 113, 30. [Google Scholar] [CrossRef] [PubMed]
  42. Cote, C.; Pibarot, P.; Despres, J.P.; Mohty, D.; Cartier, A.; Arsenault, B.J.; Couture, C.; Mathieu, P. Association between circulating oxidised low-density lipoprotein and fibrocalcific remodelling of the aortic valve in aortic stenosis. Heart 2008, 94, 1175–1180. [Google Scholar] [CrossRef] [PubMed]
  43. Que, X.; Hung, M.Y.; Yeang, C.; Gonen, A.; Prohaska, T.A.; Sun, X.; Diehl, C.; Maatta, A.; Gaddis, D.E.; Bowden, K.; et al. Oxidized phospholipids are proinflammatory and proatherogenic in hypercholesterolaemic mice. Nature 2018, 558, 301–306. [Google Scholar] [CrossRef] [PubMed]
  44. Tsimikas, S.; Witztum, J.L. The role of oxidized phospholipids in mediating lipoprotein(a) atherogenicity. Curr. Opin. Lipidol. 2008, 19, 369–377. [Google Scholar] [CrossRef]
  45. Mahmut, A.; Boulanger, M.C.; El Husseini, D.; Fournier, D.; Bouchareb, R.; Despres, J.P.; Pibarot, P.; Bosse, Y.; Mathieu, P. Elevated expression of lipoprotein-associated phospholipase A2 in calcific aortic valve disease: Implications for valve mineralization. J. Am. Coll. Cardiol. 2014, 63, 460–469. [Google Scholar] [CrossRef] [Green Version]
  46. Mahmut, A.; Mahjoub, H.; Boulanger, M.C.; Fournier, D.; Despres, J.P.; Pibarot, P.; Mathieu, P. Lp-PLA2 is associated with structural valve degeneration of bioprostheses. Eur. J. Clin. Investig. 2014, 44, 136–145. [Google Scholar] [CrossRef]
  47. Capoulade, R.; Mahmut, A.; Tastet, L.; Arsenault, M.; Bedard, E.; Dumesnil, J.G.; Despres, J.P.; Larose, E.; Arsenault, B.J.; Bosse, 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]
  48. Panagopoulou, M.; Fanidis, D.; Aidinis, V.; Chatzaki, E. ENPP2 Methylation in Health and Cancer. Int. J. Mol. Sci. 2021, 22, 11958. [Google Scholar] [CrossRef]
  49. Umezu-Goto, M.; Kishi, Y.; Taira, A.; Hama, K.; Dohmae, N.; Takio, K.; Yamori, T.; Mills, G.B.; Inoue, K.; Aoki, J.; et al. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J. Cell Biol. 2002, 158, 227–233. [Google Scholar] [CrossRef]
  50. Aoki, J.; Inoue, A.; Okudaira, S. Two pathways for lysophosphatidic acid production. Biochim. Biophys. Acta 2008, 1781, 513–518. [Google Scholar] [CrossRef]
  51. Bourgeois, R.; Devillers, R.; Perrot, N.; Despres, A.A.; Boulanger, M.C.; Mitchell, P.L.; Guertin, J.; Couture, P.; Boffa, M.B.; Scipione, C.A.; et al. Interaction of Autotaxin with Lipoprotein(a) in Patients with Calcific Aortic Valve Stenosis. JACC Basic Transl. Sci. 2020, 5, 888–897. [Google Scholar] [CrossRef] [PubMed]
  52. Mkannez, G.; Gagne-Ouellet, V.; Jalloul Nsaibia, M.; Boulanger, M.C.; Rosa, M.; Argaud, D.; Hadji, F.; Gaudreault, N.; Rheaume, G.; Bouchard, L.; et al. DNA methylation of a PLPP3 MIR transposon-based enhancer promotes an osteogenic programme in calcific aortic valve disease. Cardiovasc. Res. 2018, 114, 1525–1535. [Google Scholar] [CrossRef] [PubMed]
  53. Huang, P.L. A comprehensive definition for metabolic syndrome. Dis. Models Mech. 2009, 2, 231–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Katz, R.; Wong, N.D.; Kronmal, R.; Takasu, J.; Shavelle, D.M.; Probstfield, J.L.; Bertoni, A.G.; Budoff, M.J.; O’Brien, K.D. Features of the metabolic syndrome and diabetes mellitus as predictors of aortic valve calcification in the Multi-Ethnic Study of Atherosclerosis. Circulation 2006, 113, 2113–2119. [Google Scholar] [CrossRef] [Green Version]
  55. Capoulade, R.; Clavel, M.A.; Dumesnil, J.G.; Chan, K.L.; Teo, K.K.; Tam, J.W.; Cote, N.; Mathieu, P.; Despres, J.P.; Pibarot, P.; et al. Impact of metabolic syndrome on progression of aortic stenosis: Influence of age and statin therapy. J. Am. Coll. Cardiol. 2012, 60, 216–223. [Google Scholar] [CrossRef] [Green Version]
  56. Norata, G.D.; Tsimikas, S.; Pirillo, A.; Catapano, A.L. Apolipoprotein C-III: From Pathophysiology to Pharmacology. Trends Pharmacol. Sci. 2015, 36, 675–687. [Google Scholar] [CrossRef]
  57. D’Erasmo, L.; Di Costanzo, A.; Gallo, A.; Bruckert, E.; Arca, M. ApoCIII: A multifaceted protein in cardiometabolic disease. Metabolism 2020, 113, 154395. [Google Scholar] [CrossRef]
  58. Pechlaner, R.; Tsimikas, S.; Yin, X.; Willeit, P.; Baig, F.; Santer, P.; Oberhollenzer, F.; Egger, G.; Witztum, J.L.; Alexander, V.J.; et al. Very-Low-Density Lipoprotein-Associated Apolipoproteins Predict Cardiovascular Events and Are Lowered by Inhibition of APOC-III. J. Am. Coll. Cardiol. 2017, 69, 789–800. [Google Scholar] [CrossRef] [Green Version]
  59. Jorgensen, A.B.; Frikke-Schmidt, R.; Nordestgaard, B.G.; Tybjaerg-Hansen, A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N. Engl. J. Med. 2014, 371, 32–41. [Google Scholar] [CrossRef] [Green Version]
  60. TG and HDL Working Group of the Exome Sequencing Project; National Heart, Lung, and Blood Institute; Crosby, J.; Peloso, G.M.; Auer, P.L.; Crosslin, D.R.; Stitziel, N.O.; Lange, L.A.; Lu, Y.; Tang, Z.Z.; et al. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N. Engl. J. Med. 2014, 371, 22–31. [Google Scholar] [CrossRef] [Green Version]
  61. Capoulade, R.; Torzewski, M.; Mayr, M.; Chan, K.L.; Mathieu, P.; Bosse, Y.; Dumesnil, J.G.; Tam, J.; Teo, K.K.; Burnap, S.A.; et al. ApoCIII-Lp(a) complexes in conjunction with Lp(a)-OxPL predict rapid progression of aortic stenosis. Heart 2020, 106, 738–745. [Google Scholar] [CrossRef] [PubMed]
  62. Corsini, A.; Maggi, F.M.; Catapano, A.L. Pharmacology of competitive inhibitors of HMG-CoA reductase. Pharmacol. Res. 1995, 31, 9–27. [Google Scholar] [CrossRef]
  63. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: The Scandinavian Simvastatin Survival Study (4S). Lancet 1994, 344, 1383–1389.
  64. Rosenson, R.S. Statins in atherosclerosis: Lipid-lowering agents with antioxidant capabilities. Atherosclerosis 2004, 173, 1–12. [Google Scholar] [CrossRef]
  65. Halcox, J.P.; Deanfield, J.E. Beyond the laboratory: Clinical implications for statin pleiotropy. Circulation 2004, 109, II42–II48. [Google Scholar] [CrossRef] [Green Version]
  66. Takemoto, M.; Liao, J.K. Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 1712–1719. [Google Scholar] [CrossRef] [Green Version]
  67. Goldstein, J.L.; Brown, M.S. Regulation of the mevalonate pathway. Nature 1990, 343, 425–430. [Google Scholar] [CrossRef]
  68. Liao, J.K. Isoprenoids as mediators of the biological effects of statins. J. Clin. Investig. 2002, 110, 285–288. [Google Scholar] [CrossRef]
  69. Van Aelst, L.; D’Souza-Schorey, C. Rho GTPases and signaling networks. Genes Dev. 1997, 11, 2295–2322. [Google Scholar] [CrossRef] [Green Version]
  70. Carabello, B.A. The SEAS Trial. Curr. Cardiol. Rep. 2010, 12, 122–124. [Google Scholar] [CrossRef]
  71. Teo, K.K.; Corsi, D.J.; Tam, J.W.; Dumesnil, J.G.; Chan, K.L. Lipid lowering on progression of mild to moderate aortic stenosis: Meta-analysis of the randomized placebo-controlled clinical trials on 2344 patients. Can. J. Cardiol. 2011, 27, 800–808. [Google Scholar] [CrossRef] [PubMed]
  72. Nishimura, R.A.; Otto, C.M.; Bonow, R.O.; Carabello, B.A.; Erwin, J.P., 3rd; Guyton, R.A.; O’Gara, P.T.; Ruiz, C.E.; Skubas, N.J.; Sorajja, P.; et al. 2014 AHA/ACC guideline for the management of patients with valvular heart disease: Executive summary: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2014, 63, 2438–2488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vahanian, A.; Alfieri, O.; Andreotti, F.; Antunes, M.J.; Barón-Esquivias, G.; Baumgartner, H.; Borger, M.A.; Carrel, T.P.; De Bonis, M.; Evangelista, A.; et al. Joint task force on the management of valvular heart disease of the European society of cardiology (ESC); European association for cardio-thoracic surgery (EACTS). guidelines on the management of valvular heart disease (version 2012). Eur. Heart J. 2012, 33, 2451–2496. [Google Scholar] [PubMed] [Green Version]
  74. Capoulade, R.; Yeang, C.; Chan, K.L.; Pibarot, P.; Tsimikas, S. Association of Mild to Moderate Aortic Valve Stenosis Progression with Higher Lipoprotein(a) and Oxidized Phospholipid Levels: Secondary Analysis of a Randomized Clinical Trial. JAMA Cardiol. 2018, 3, 1212–1217. [Google Scholar] [CrossRef]
  75. Poggio, P.; Songia, P.; Cavallotti, L.; Barbieri, S.S.; Zanotti, I.; Arsenault, B.J.; Valerio, V.; Ferri, N.; Capoulade, R.; Camera, M. PCSK9 Involvement in Aortic Valve Calcification. J. Am. Coll. Cardiol. 2018, 72, 3225–3227. [Google Scholar] [CrossRef]
  76. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef]
  77. Ruscica, M.; Greco, M.F.; Ferri, N.; Corsini, A. Lipoprotein(a) and PCSK9 inhibition: Clinical evidence. Eur. Heart J. Suppl. 2020, 22, L53–L56. [Google Scholar] [CrossRef]
  78. De Luca, L.; Corsini, A.; Uguccioni, M.; Colivicchi, F. Statins plus ezetimibe in the era of proprotein convertase subtilisin/kexin type 9 inhibitors. Kardiol. Pol. 2020, 78, 850–860. [Google Scholar] [CrossRef]
  79. Bergmark, B.A.; O’Donoghue, M.L.; Murphy, S.A.; Kuder, J.F.; Ezhov, M.V.; Ceška, R.; Gouni-Berthold, I.; Jensen, H.K.; Tokgozoglu, S.L.; Mach, F.; et al. An Exploratory Analysis of Proprotein Convertase Subtilisin/Kexin Type 9 Inhibition and Aortic Stenosis in the FOURIER Trial. JAMA Cardiol. 2020, 5, 709–713. [Google Scholar] [CrossRef]
  80. Perrot, N.; Valerio, V.; Moschetta, D.; Boekholdt, S.M.; Dina, C.; Chen, H.Y.; Abner, E.; Martinsson, A.; Manikpurage, H.D.; Rigade, S.; et al. Genetic and In Vitro Inhibition of PCSK9 and Calcific Aortic Valve Stenosis. JACC Basic Transl. Sci. 2020, 5, 649–661. [Google Scholar] [CrossRef]
  81. Galtier, F.; Mura, T.; Raynaud de Mauverger, E.; Chevassus, H.; Farret, A.; Gagnol, J.P.; Costa, F.; Dupuy, A.; Petit, P.; Cristol, J.P.; et al. Effect of a high dose of simvastatin on muscle mitochondrial metabolism and calcium signaling in healthy volunteers. Toxicol. Appl. Pharmacol. 2012, 263, 281–286. [Google Scholar] [CrossRef] [PubMed]
  82. Sirvent, P.; Bordenave, S.; Vermaelen, M.; Roels, B.; Vassort, G.; Mercier, J.; Raynaud, E.; Lacampagne, A. Simvastatin induces impairment in skeletal muscle while heart is protected. Biochem. Biophys. Res. Commun. 2005, 338, 1426–1434. [Google Scholar] [CrossRef] [PubMed]
  83. Sirvent, P.; Mercier, J.; Vassort, G.; Lacampagne, A. Simvastatin triggers mitochondria-induced Ca2+ signaling alteration in skeletal muscle. Biochem. Biophys. Res. Commun. 2005, 329, 1067–1075. [Google Scholar] [CrossRef] [PubMed]
  84. Lotteau, S.; Ivarsson, N.; Yang, Z.; Restagno, D.; Colyer, J.; Hopkins, P.; Weightman, A.; Himori, K.; Yamada, T.; Bruton, J.; et al. A Mechanism for Statin-Induced Susceptibility to Myopathy. JACC Basic Transl. Sci. 2019, 4, 509–523. [Google Scholar] [CrossRef] [PubMed]
  85. Bonifacio, A.; Sanvee, G.M.; Bouitbir, J.; Krahenbuhl, S. The AKT/mTOR signaling pathway plays a key role in statin-induced myotoxicity. Biochim. Biophys. Acta 2015, 1853, 1841–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Bouitbir, J.; Charles, A.L.; Echaniz-Laguna, A.; Kindo, M.; Daussin, F.; Auwerx, J.; Piquard, F.; Geny, B.; Zoll, J. Opposite effects of statins on mitochondria of cardiac and skeletal muscles: A ‘mitohormesis’ mechanism involving reactive oxygen species and PGC-1. Eur. Heart J. 2012, 33, 1397–1407. [Google Scholar] [CrossRef]
  87. Bonifacio, A.; Mullen, P.J.; Mityko, I.S.; Navegantes, L.C.; Bouitbir, J.; Krahenbuhl, S. Simvastatin induces mitochondrial dysfunction and increased atrogin-1 expression in H9c2 cardiomyocytes and mice in vivo. Arch. Toxicol. 2016, 90, 203–215. [Google Scholar] [CrossRef]
  88. Bouitbir, J.; Singh, F.; Charles, A.L.; Schlagowski, A.I.; Bonifacio, A.; Echaniz-Laguna, A.; Geny, B.; Krahenbuhl, S.; Zoll, J. Statins Trigger Mitochondrial Reactive Oxygen Species-Induced Apoptosis in Glycolytic Skeletal Muscle. Antioxid. Redox Signal. 2016, 24, 84–98. [Google Scholar] [CrossRef]
  89. Sanvee, G.M.; Hitzfeld, L.; Bouitbir, J.; Krahenbuhl, S. mTORC2 is an important target for simvastatin-associated toxicity in C2C12 cells and mouse skeletal muscle—Roles of Rap1 geranylgeranylation and mitochondrial dysfunction. Biochem. Pharmacol. 2021, 192, 114750. [Google Scholar] [CrossRef]
  90. Mullen, P.J.; Zahno, A.; Lindinger, P.; Maseneni, S.; Felser, A.; Krahenbuhl, S.; Brecht, K. Susceptibility to simvastatin-induced toxicity is partly determined by mitochondrial respiration and phosphorylation state of Akt. Biochim. Biophys. Acta 2011, 1813, 2079–2087. [Google Scholar] [CrossRef] [Green Version]
  91. Ridker, P.M.; Pradhan, A.; MacFadyen, J.G.; Libby, P.; Glynn, R.J. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: An analysis from the JUPITER trial. Lancet 2012, 380, 565–571. [Google Scholar] [CrossRef] [Green Version]
  92. Crandall, J.P.; Mather, K.; Rajpathak, S.N.; Goldberg, R.B.; Watson, K.; Foo, S.; Ratner, R.; Barrett-Connor, E.; Temprosa, M. Statin use and risk of developing diabetes: Results from the Diabetes Prevention Program. BMJ Open Diabetes Res. Care 2017, 5, e000438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yaluri, N.; Modi, S.; Kokkola, T. Simvastatin induces insulin resistance in L6 skeletal muscle myotubes by suppressing insulin signaling, GLUT4 expression and GSK-3beta phosphorylation. Biochem. Biophys. Res. Commun. 2016, 480, 194–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Sun, B.; Zhong, Z.; Wang, F.; Xu, J.; Xu, F.; Kong, W.; Ling, Z.; Shu, N.; Li, Y.; Wu, T.; et al. Atorvastatin impaired glucose metabolism in C2C12 cells partly via inhibiting cholesterol-dependent glucose transporter 4 translocation. Biochem. Pharmacol. 2018, 150, 108–119. [Google Scholar] [CrossRef] [PubMed]
  95. Bouitbir, J.; Sanvee, G.M.; Panajatovic, M.V.; Singh, F.; Krahenbuhl, S. Mechanisms of statin-associated skeletal muscle-associated symptoms. Pharmacol. Res. 2020, 154, 104201. [Google Scholar] [CrossRef]
Figure 1. The implication of Ox-LDL in the calcification of the aortic valve. The infiltration of Ox-LDL into the aortic valve activates inflammation and, consequently, the release of PLA2G7 from macrophages, leading to the production of LysoPC. The activation of autotaxin leads to the preproduction of LPA, which amplifies the inflammation and the activation of the osteoblastic-like phenotype switch.
Figure 1. The implication of Ox-LDL in the calcification of the aortic valve. The infiltration of Ox-LDL into the aortic valve activates inflammation and, consequently, the release of PLA2G7 from macrophages, leading to the production of LysoPC. The activation of autotaxin leads to the preproduction of LPA, which amplifies the inflammation and the activation of the osteoblastic-like phenotype switch.
Jcm 11 03331 g001
Figure 2. Statins’ mechanism of action. Statins inhibit the HMG-CoA reductase to block the synthesis of mevalonic acid and, consequently, the production of cholesterol.
Figure 2. Statins’ mechanism of action. Statins inhibit the HMG-CoA reductase to block the synthesis of mevalonic acid and, consequently, the production of cholesterol.
Jcm 11 03331 g002
Figure 3. The effects of statins at systemic levels. Statins increase the expression of LDL receptors on hepatocytes to increase LDL uptake and catabolism, leading to a systemic decrease in LDLs, vLDLs, and the production of HDL.
Figure 3. The effects of statins at systemic levels. Statins increase the expression of LDL receptors on hepatocytes to increase LDL uptake and catabolism, leading to a systemic decrease in LDLs, vLDLs, and the production of HDL.
Jcm 11 03331 g003
Figure 4. The effect of Lp(a) on aortic valve calcification. Lp(a) particles carry oxidized LDL, which stimulate aortic valve cells’ calcification. Targeting Lp(a) might inhibit valve inflammation and, consequently, reduce valve cell calcification.
Figure 4. The effect of Lp(a) on aortic valve calcification. Lp(a) particles carry oxidized LDL, which stimulate aortic valve cells’ calcification. Targeting Lp(a) might inhibit valve inflammation and, consequently, reduce valve cell calcification.
Jcm 11 03331 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nsaibia, M.J.; Devendran, A.; Goubaa, E.; Bouitbir, J.; Capoulade, R.; Bouchareb, R. Implication of Lipids in Calcified Aortic Valve Pathogenesis: Why Did Statins Fail? J. Clin. Med. 2022, 11, 3331. https://doi.org/10.3390/jcm11123331

AMA Style

Nsaibia MJ, Devendran A, Goubaa E, Bouitbir J, Capoulade R, Bouchareb R. Implication of Lipids in Calcified Aortic Valve Pathogenesis: Why Did Statins Fail? Journal of Clinical Medicine. 2022; 11(12):3331. https://doi.org/10.3390/jcm11123331

Chicago/Turabian Style

Nsaibia, Mohamed J., Anichavezhi Devendran, Eshak Goubaa, Jamal Bouitbir, Romain Capoulade, and Rihab Bouchareb. 2022. "Implication of Lipids in Calcified Aortic Valve Pathogenesis: Why Did Statins Fail?" Journal of Clinical Medicine 11, no. 12: 3331. https://doi.org/10.3390/jcm11123331

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