You are on page 1of 16

Arteriosclerosis, Thrombosis, and Vascular Biology

BRIEF REVIEW

Aortic Valve Stenosis


From Basic Mechanisms to Novel Therapeutic Targets
Philip Roger Goody,* Mohammed Rabiul Hosen, Dominik Christmann, Sven Thomas Niepmann, Andreas Zietzer, Matti Adam,
Florian Bönner, Sebastian Zimmer, Georg Nickenig, Felix Jansen*

ABSTRACT: Aortic valve stenosis is the most prevalent heart valve disease worldwide. Although interventional treatment options
have rapidly improved in recent years, symptomatic aortic valve stenosis is still associated with high morbidity and mortality.
Calcific aortic valve stenosis is characterized by a progressive fibro-calcific remodeling and thickening of the aortic valve
cusps, which subsequently leads to valve obstruction. The underlying pathophysiology is complex and involves endothelial
dysfunction, immune cell infiltration, myofibroblastic and osteoblastic differentiation, and, subsequently, calcification. To date,
no pharmacotherapy has been established to prevent aortic valve calcification. However, novel promising therapeutic targets
have been recently identified. This review summarizes the current knowledge of pathomechanisms involved in aortic valve
calcification and points out novel treatment strategies.

Key Words:  aortic valve ◼ aortic valve stenosis ◼ calcification ◼ heart valve disease ◼ inflammation ◼ valvular endothelial cells ◼ valvular interstitial cells

A
ortic valve stenosis (AVS) is the most common which is comprised of valvular endothelial cells (VECs)
acquired heart valve disease in the western world.1,2 on both sides (ventricularis and fibrosa). The interstitium
Downloaded from http://ahajournals.org by on April 8, 2022

Its prevalence is >2% in patients >60 years of age.3 of the valve consists of 3 distinct layers: the laminae
When symptomatic, the 2-year mortality rate of severe fibrosa, spongiosa, and ventricularis. The main cell popu-
AVS is around 50%.4,5 lation found here is composed of valvular interstitial cells
In most humans, the aortic valve consists of 3 semilunar (VICs).12 To meet the different physicomechanical needs
leaflets, also called cusps (tricuspid aortic valve). There is a of the valve, these layers have a complex ECM. The lam-
congenital variant of bicuspid aortic valves (BAV) with a prev- ina fibrosa, representing the largest part of the valve and
alence of 0.5% to 2 %.6 These patients develop severe AVS the main load-bearing structure, has abundant type I col-
early in life, in contrast to humans with tricuspid valves, and lagen fibrils. The lamina ventricularis consists of collagen
show pronounced calcification owing to higher mechanical and radially aligned elastin, which help to distribute radial
strain on the valve cusps.7,8 Furthermore, genetic variants in forces during valve opening. In between lies the lamina
patients with BAV were identified. These variants include spongiosa that is largely made up of glycosaminoglycans
mutations in NOTCH1 and in the GATA family of zinc finger (GAGs), which are believed to have a lubricating function
proteins, leading to accelerated calcification and changes for the outer layers.13,14
in the ECM (extracellular matrix) through dysregulation of AVS develops through an initial endothelial injury and
matrix metalloproteinases, respectively.9–11 The underlying dysfunction, immune cell infiltration, myofibroblastic/
molecular mechanisms discussed in this review are likely to osteoblastic differentiation of VICs, and subsequent cal-
apply to both bicuspid and tricuspid aortic valves, although cification.15 This inhibits separation due to stiffening or
there is a great need to study differences in calcification fusion of the cusps and consequently causes a reduction
processes of these 2 variants. in valve opening with increased blood flow velocity.16 Once
In healthy humans, the valve cusps are less than a fibrosis and calcification of the valve have commenced,
millimeter thick and are covered by an endothelial layer, the majority of patients develop severe AVS.17,18 The only


Correspondence to: Philip Roger Goody and Felix Jansen, Heart Center Bonn, Department of Medicine II, University Hospital Bonn, Venusberg-Campus 1, 53127,
Bonn, Germany, Emails philip.goody@ukbonn.de, felix.jansen@ukbonn.de
*These authors contributed equally to this article.
For Sources of Funding and Disclosures, see page 896.
© 2020 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol is available at www.ahajournals.org/journal/atvb

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   885
Goody et al Aortic Valve Stenosis

Nonstandard Abbreviations and Acronyms Highlights


Brief Review - AL

α-SMA α-smooth muscle actin • Calcific aortic valve stenosis is a disease with high
ALP alkaline phosphatase prevalence and mortality.
AVS aortic valve stenosis • Currently, no medical treatment is available to halt
disease progression or reverse calcification.
BAV bicuspid aortic valve
• Lack of mechanistic understanding hampers devel-
BMP bone morphogenic protein opment of specific anti-inflammatory and anticalcific
CAVD calcific aortic valve disease drugs.
CDH11 cadherin-11 • This review summarizes our current knowledge of
DPP-4 dipeptidyl peptidase-4 disease pathology and highlights potential drug-
gable pathways.
ECM extracellular matrix
EndMT endothelial to mesenchymal transition
eNOS endothelial nitric oxide synthase replacement are on the rise. In fact, recently, 2 large pro-
ENPP1 ectonucleotide pyrophosphatase/phos- spective, multicenter studies showed that transcatheter
phodiesterase 1 aortic valve replacement can outperform surgical aortic
EVs extracellular vesicles valve replacement, even in low-risk patients.19,20 However,
FGF-2 fibroblast growth factor 2 although these interventions yield very good results, there
HOTAIR HOX transcript antisense RNA are still peri-procedural risks, including left bundle branch
ICAM-1 intercellular adhesion molecule 1 block, acute kidney injury, disabling stroke, bleeding com-
IGF-1 insulin-like growth factor 1 plications, and death.19,20 At this time, no therapeutic or
IL interleukin preventative medical treatment is available for AVS. It
lncRNA long noncoding RNA is, therefore, fundamentally important to understand the
mechanisms of disease initiation and progression in order
LRP5 LDL receptor–related protein 5
to identify possible therapeutic targets and develop new
MALAT1 metastasis-associated lung adenocarci-
strategies to ameliorate this frequent and severe illness.
noma transcript 1
Here, we summarize current knowledge about the
MAPK mitogen-activated protein kinase
pathophysiological mechanisms that lead to calcific AVS.
Downloaded from http://ahajournals.org by on April 8, 2022

MGP matrix-γ-carboxyglutamic acid protein


Additionally, we will highlight some innovative treatment
miRNA microRNA options with the potential to limit aortic valve calcification
ncRNA noncoding RNA and reduce the burden of this disease.
NF-κB nuclear factor ‘kappa-light-chain-
enhancer’ of activated B-cells
OPG osteoprotegerin Phases of Calcific Aortic Valve Disease
ox-LDL oxidized low-density lipoprotein The initiation and progression of calcific aortic valve dis-
P2Y purinoceptor ease (CAVD) can be seen as 2 distinct phases, with dif-
RANK receptor activator of nuclear factor ferent immunologic, metabolic, physicomechanical, and
kappa B cellular responses leading to end-stage calcification as
RANKL receptor activator of nuclear factor first described by Aikawa et al in 2007.21–23 In the first of
kappa B ligand these phases (initiation), there is lipid deposition, accom-
Runx2 Runt-related transcription factor 2 panied by injury and inflammation. In the second phase
SMAD small mothers against decapentaplegic (propagation), osteogenic differentiation and calcifica-
TGF transforming growth factor tion are responsible for disease progression (Figure 1).21
TNF tumor necrosis factor
TUG1 taurine upregulated gene 1 Initiation Phase
VCAM-1 vascular cell adhesion protein 1
Endothelial Dysfunction
VEC valvular endothelial cell
Mechanical and (oscillatory) shear stress can induce
VIC valvular interstitial cell endothelial dysfunction of the VEC layer, which leads
Wnt wingless and Int-1 to the deposition of lipoproteins and the infiltration of
immune cells.24,25 The integrity of the endothelial layer is
possible treatment for severe aortic valve stenosis is an important factor of valvular homeostasis (Figure 1).26
either surgical aortic valve replacement or transcatheter During embryogenesis and valve formation, cardiac cush-
aortic valve replacement. Currently, interventional implan- ions develop, which are a local expansion of the ECM.27
tation methods such as transfemoral or transapical valve These cushions are invaded by endocardial endothelial

886   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

Brief Review - AL
Downloaded from http://ahajournals.org by on April 8, 2022

Figure 1. Phases of calcific aortic valve disease (CAVD).


Pathomechanisms of initiation and progression of CAVD can be seen as distinct disease phases: endothelial dysfunction and lipid deposition,
inflammation, fibrosis, and calcification. Different stimuli, for example, mechanical/shear stress, induce endothelial dysfunction of valvular
endothelial cells (VECs), which allows lipoprotein infiltration, specifically LDL (low-density lipoprotein) and Lp(a) (lipoprotein[a]), and
consequently extravasation of immune cells into the interstitium of valve leaflets. A dysregulation of the eNOS (endothelial nitric oxide synthase)
pathway induces the production of reactive oxygen species (ROS), which stimulates the oxidation of infiltrated lipids into ox-LDL (oxidized LDL)
and oxidized phospholipids (ox-PLs). These molecules are further transformed into lysophosphatidylcholine (lysoPC), promoting apoptosis
of valvular interstitial cells (VICs) and release of apoptotic bodies leading to diffuse calcification. Following the oxidation of lipids in the valve,
immune cells infiltrate the tissue and are activated. Osteogenic differentiation of VICs is stimulated by macrophages and T cells via secretion
of TNF (tumor necrosis factor), IL (interleukin)-1β, IL-6, and RANKL (receptor activator of nuclear factor kappa B). Calcification is further
promoted by the release of so-called matrix vesicles /calcifying microvesicles from macrophages and VICs. BMP-2 (bone morphogenic protein
2) and Runx2 (runt-related transcription factor 2) induce osteogenic differentiation. Notch1 protects against osteoblastic differentiation via
inhibition of BMP-2 expression. ENPP1 (ectonucleotide pyrophosphatase/phosphodiesterase 1) catalyze the hydrolysis of ATP to AMP and
pyrophosphate (PPi). Wnt (wingless and Int-1) induces osteoblastic differentiation. TGF indicates transforming growth factor.

cells, which can differentiate into mesenchymal valve a weakening of the endothelial barrier function, owing to
progenitor cells—a precursor of VICs—in a process called the loss of adherens junctions.29 This process of trans-
endothelial to mesenchymal transition (EndMT).7,27,28 differentiation is also believed to occur during CAVD in
Notch signaling increases the levels of TGF (transform- adults.30 VECs differentiate into endothelial-derived VICs,
ing growth factor)-β in these cells, which, in turn, activates which in turn can also differentiate into osteoblastic VICs,
the transcription factors Zinc finger protein SNAI1 and 2 promoting calcification. Interestingly, VICs can inhibit this
(Snail/Slug) and results in a decrease of VE-cadherin transdifferentiation, which was shown through cocul-
(vascular endothelial cadherin; Figure 1).29 This leads to ture experiments in vitro.30 It is thought that a low rate

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   887
Goody et al Aortic Valve Stenosis

of EndMT functions to replenish defunct VICs but that a isolated VICs.47 Furthermore, activation of TLR3 in human
higher rate of EndMT contributes to the progression of VICs with Poly(I:C) (polyinosinic:polycytidylic acid) can
Brief Review - AL

disease.30,31 The role of EndMT in vivo is not yet entirely induce an upregulation of BMP (bone morphogenic pro-
clear but warrants further investigation. The endothelial tein)-2, TGF-β1 and ALP (alkaline phosphatase), thus lead-
cells on the fibrosa (=aortic) and ventricularis sides of ing to calcium deposition.48 Monocytes and macrophages
the valve have different phenotypes, as demonstrated in stimulate the osteogenic differentiation of VICs as well as
porcine aortic valve specimens.32 Furthermore, a differ- calcification via secretion of TNF (tumor necrosis factor),
ence in the expression of eNOS (endothelial nitric oxide followed by activation of NF-κB and IL (interleukin)-1β and
synthase) was observed in calcified versus noncalcified IL-6, possibly due to an induction of the homeobox protein
human aortic valve samples.33 The expression level was MSX2 and DR4 (death receptor 4).49–52
reduced overall in calcified valves, with higher expres- It was recently shown that another pattern recognition
sion found on the ventricularis side. Additionally, eNOS receptor, namely CLEC4E (C-type lectin domain family 4
expression levels are higher in the ventricular endothe- member E), is highly expressed in human stenotic aortic
lium of porcine aortic valves.33 This could be one reason valve tissue.53 This receptor is found on phagocytic cells,
why more pronounced calcification is frequently observed such as monocytes and macrophages and can recognize
on the aortic side of the cusps. Thus, endothelial cell het- cell debris and cholesterol crystals, promote their incor-
erogeneity could play a major role in the initiation of dis- poration, intracellular degradation, and consequent vas-
ease, and local differences in gene and noncoding RNA cular inflammation and atherogenesis.54
(ncRNA) expression could provide further insight into the Localized cell death, as a consequence of endothe-
disease mechanisms. lial dysfunction and inflammation, results in the release
of apoptotic bodies that cause microcalcification, which
Lipid Deposition
is further promoted by the release of extracellular vesi-
There is growing evidence that the deposition of lipopro-
cles (EV) from macrophages and VICs.55–58 This process
teins strongly induces chronic inflammation, which is a
shows similarities to bone formation, where EVs filled
hallmark of the early stages of CAVD and can be visualized
with calcium and inorganic phosphate are shed. These
long before calcification occurs by fluorodeoxyglucose
calcium-phosphate complexes within the EV can form
positron emission tomography/computed tomography
hydroxyapatite crystals, which grow in size and ultimately
(FDG-PET/CT).34 Different apolipoproteins, such as apoB,
destroy the EV. The hydroxyapatite crystals come into
apoA1, and apo(a), as well as a dysregulation of the eNOS
contact with the ECM, where they function as nucleation
Downloaded from http://ahajournals.org by on April 8, 2022

pathway and increased nicotinamide adenine dinucleo-


sites for further macrocalcification.55,56 Such calcification
tide phosphate (NADPH) oxidase levels, were observed
triggers an even more pronounced immune response,
in explanted human aortic valves (Figure 1).35–37 The for-
creating a vicious circle and consequently leading to the
mation of reactive oxygen species may act as a stimulus
propagation phase of the disease.
of calcification, as shown in a vascular smooth muscle
cell (VSMC) calcification model.38 This increased oxida-
tive stress also promotes the formation of ox-LDLs (oxi- Propagation Phase
dized low-density lipoproteins) and oxidized phospholipids,
Myofibroblastic and Osteoblastic Differentiation
which triggers calcification in vascular cells.38 Oxidized
After the initial inflammatory phase, where lipid deposi-
phospholipids can be transformed into lysophosphatidyl-
tion and chronic inflammation initiate CAVD, the face of
choline by Lp-PLA2 (lipoprotein-phospholipase A2), lead-
the disease changes, and calcification becomes the driv-
ing to apoptosis in VICs.39,40 Lp-PLA2 levels are higher in
ing force. Differentiation of VICs into myofibroblastic and
stenotic aortic valve tissue.40,41 Ox-LDL induces upregula-
osteoblastic phenotypes is regarded as a fundamental
tion of cell adhesion molecules, including ICAM-1 (inter-
step during the propagation phase. The initiation of this
cellular adhesion molecule 1) and VCAM-1 (vascular cell
differentiation seems to be regulated by cytokines that
adhesion protein 1), and consequently, there is greater
are secreted by immune cells.59 During progression of
adhesion and extravasation of immune cells.35,42–44
the disease into the propagation phase, calcific stimuli
Inflammation become more independent of immune cells, and the cal-
Oxidized lipids also trigger an inflammatory response in val- cification process is regulated by a number of calcific
vular tissue (Figure 1). Following the uptake of oxidized lip- pathways. In an elegant study by the Aikawa lab, pro-
ids, local macrophages, CD4+ and CD8+ T lymphocytes, teomic analyses confirmed differential protein expression
and mast cells are activated.24,45 This is in part a result of amongst the 3 layers of the aortic valve cusps.60 VICs had
the activation of PRRs (pattern recognition receptors), such a greater potential for calcification on the fibrosa side of
as TLRs (toll-like receptors) and the NF-κB (nuclear fac- the valve, possibly owing to the activation of pathogenic
tor κB) pathway.46 TLR2 and TLR4 are expressed in VICs pathways, such as collagen biosynthesis and modifica-
and treatment of the cells with lipopolysaccharide, a proin- tion, ECM degradation, GAG metabolism, and lipid metab-
flammatory TLR ligand, induces an osteogenic response in olism.52 These results demonstrate a large spatiotemporal

888   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

heterogeneity within the valvular tissue contributing to downregulation of the long ncRNA (lncRNA) HOTAIR
regional differences in calcification patterns and dem- resulting in an increase in ALP and BMP-2 levels, pro-

Brief Review - AL
onstrates the great plasticity of this cell type. VICs can moting osteoblastic differentiation of VICs.74,75 The exact
differentiate into myofibroblast- and osteoblast-like cells. mechanisms of dystrophic calcification are still poorly
Calcification is a result of both of these differentiation understood and warrant extensive research to unravel
pathways, although the relative contribution of each of the underlying pathways.
these pathways in vivo is currently unclear.12 Myofibroblas-
tic differentiation can be achieved in vitro by stimulating Molecular Calcific Pathways
VICs with TGF-β1 and can be inhibited by FGF (fibro- In the initiation phase of CAVD, osteoblastic differen-
blast growth factor)-2.61 This differentiation leads to an tiation seems to be coordinated by infiltrating macro-
increased expression of α-SMA (α-smooth muscle actin), phages, which has been demonstrated by in vivo imaging,
vimentin, and smooth muscle myosin.62 Downstream as mentioned above.23 During the later stages of disease,
effects include activation of the canonical MAPK (mito- other pathways become more important. These include
gen-activated protein kinase) pathway via SMAD (small the RANK (receptor activator of nuclear factor kappa
mothers against decapentaplegic) and noncanonical p38 B)/RANKL (RANK ligand)/OPG (osteoprotegerin),
activation.63 This leads to increased binding of LDL to Wnt (wingless and Int-1)/β-catenin, and Notch signal-
GAGs and nodule formation as well as increased ALP ing pathways (Figures 1 and 2).16 Notch1 gained interest
activity, respectively.63,64 A further effect of TGF-β1 treat- when 2 families with frame-shift mutations in NOTCH1,
ment is enhanced expression of CDH11 (cadherin-11), resulting in a premature stop codon, were reported.76 In
leading to its binding with α-SMA and consequent nodule these families, a high rate of BAV and severe aortic valve
formation.65 CDH11 was found to be highly expressed in calcification was observed. Further studies in mice and
diseased aortic valves of humans and in a mouse-model VICs demonstrated that functional Notch1 represses
of AVS.66 CDH11 participates in mesenchymal to osteo- BMP-2 expression and Runx2 transcriptional activity,
chondral differentiation and was found to be expressed thus protecting VICs from osteoblastic differentiation
on 91% of myofibroblastic and 96% of osteoblastic and calcification (Figure 1).77 The Wnt/β-catenin path-
cells.66 Increased ALP activity and osteocalcin expression way appears to be a positive regulator of osteoblastic
with increased calcification can also occur upon treat- differentiation, in which binding of Wnt to the LRP5 (LDL
ment with TGF-β3 and BMP-2, BMP-4, BMP-7 or an receptor–related protein 5) leads to activation of the
osteogenic medium, containing ascorbate-2-phosphate, canonical Wnt/β-catenin pathway.78 Calcium deposition
Downloaded from http://ahajournals.org by on April 8, 2022

dexamethasone, and β-glycerol phosphate.67 The latter is leads to stiffening of the cusps, which increases mechan-
commonly used to induce osteoblastic differentiation in ical strain and injury and further induces activation of
vitro. These treatments lead to an increase in ALP activ- the Wnt/β-catenin pathway, eventually leading to even
ity and expression of osteoblast markers, such as Runx2 more osteoblastic differentiation of VICs through ENPP
(runt-related transcription factor 2) and BMP-2. BMP-2 (ectonucleotide pyrophosphatase/phosphodiesterase
is a potent initiator of osteoblastic differentiation, whereas 1; Figures 1 and 2). ENPP1 hydrolyzes extracellular
Runx2 is seen as the master transcriptional regulator for ATP generating inorganic phosphate and thus promot-
osteogenesis.68,69 This process seems to be passage- ing calcification.79 A single nucleotide polymorphism
dependent, in that cells from higher numbers of passages in intron 9 of ENPP1 was associated with CAVD, and
show less potential for calcification.70 Cells treated with a higher expression levels of ENPP1 have been detected
so-called procalcifying medium, containing NaH2PO4 and in stenotic valves by quantitative polymerase chain reac-
L-ascorbic acid, showed ALP- and passage-independent tion.79 Loss of extracellular ATP leads to more apopto-
calcification.70 sis of VICs because ATP can act as a survival signal by
Calcification of valve tissue can arise as a result of binding to purinergic P2Y2 (purinoceptor) receptors on
osteoblastic differentiation of VICs and lead to osteo- VICs.79 Diminished P2Y2 signaling increases levels of
genic calcification with formation of bone-like struc- IL-6, which in turn promotes the expression of BMP-2
tures.71,72 This pathway only accounts for around 13% of and leads to greater calcification of VICs.80
calcific deposits in tricuspid aortic valves, whereas BAV Another pathway involved in cardiovascular calci-
have a >2-fold higher incidence of osteogenic calcifica- fication is mediated by fetuin-A and MGP (matrix-γ-
tion.72 The majority of mineral deposition is acquired via carboxyglutamic acid protein).81 MGP is activated by
dystrophic calcification (ca. 83%), which is speculated carboxylation and phosphorylation in a vitamin K–depen-
to be the result of diffuse calcium precipitation on cel- dent manner, and low levels of vitamin K lead to inefficient
lular debris after apoptosis of VICs and is suggested to carboxylation of MGP and thus more coronary calcifica-
be regulated in part by TGF-β.71,73 A possible explana- tion.81 There is evidence that patients taking vitamin K
tion for increased osteogenic calcification in BAVs could antagonists show more vascular calcification and higher
be higher biomechanical strain on the valve cusps when rates of aortic valve degeneration.82,83 Dietary intake of
compared with tricuspid aortic valves. This can lead to menaquinone (vitamin K2) was found to reduce vascular

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   889
Goody et al Aortic Valve Stenosis
Brief Review - AL
Downloaded from http://ahajournals.org by on April 8, 2022

Figure 2. Mechanisms of calcification in calcific aortic valve disease.


Mechanical and shear stress induce endothelial dysfunction of the valvular endothelial cell (VEC) layer leading to endothelial to mesenchymal
transition (EndMT) of VECs to myofibroblastic cells and endothelial-derived valvular interstitial cells (VICs). VICs can inhibit this transdifferentiation.
TGF (transforming growth factor)-β inhibits expression of CD31 and activates the transcription factors Snail/Slug, resulting in a decrease of
VE-Cadherin (vascular endothelial cadherin) and loss of adherens junctions. TNF (tumor necrosis factor) α inhibits eNOS (endothelial NOS)
and CD31 expression and stimulates expression of α-SMA (α smooth muscle actin), necessary for myofibroblastic differentiation. VICs can also
undergo myofibroblastic or osteoblastic differentiation. Myofibroblastic differentiation is stimulated by cholesterol, CDH11 (cadherin-11), and
TGF-β1 and inhibited by FGF-2 (fibroblast growth factor 2). Myofibroblastic VICs undergo Casp (caspase) mediated apoptosis, which leads to the
release of apoptotic bodies and diffuse calcification. Osteoblastic differentiation can be induced by TGF-β 1 and 3, RANK (receptor activator of
nuclear factor kappa B), BMP (bone morphogenic proteins; BMP-2, BMP-4, BMP-7), Wnt (wingless and Int-1)/β-catenin, SMADs (small mothers
against decapentaplegic), ENPP1 (ectonucleotide pyrophosphatase/phosphodiesterase 1), TLRs (toll-like receptors) and others. Noncoding RNA
(NcRNAs) can either stimulate or inhibit differentiation, while Notch1 signaling seems to inhibit differentiation. Calcification is further promoted by
the release of calcifying extracellular vesicles from VICs. Myofibroblastic differentiation leads to increased ALP (alkaline phosphatase) expression/
activity and expression of α-SMA and vimentin. Osteoblastic differentiation leads to an increase of Runx2, BMPs, ALP, and osteocalcin levels.
Cardiovascular osteoclast-like cells (green) have impaired mineral resorption capacity, thus also aiding calcification and show low levels of TRAP
(tartrate-resistant acid phosphatase) and CTSK (cathepsin K).

890   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

calcification and mortality.84 The effects of vitamin K sup- regions of the valve display high expression of CD8+
plementation are currently being investigated in multiple T-cell gene transcripts when compared with noncalci-

Brief Review - AL
clinical trials (VitaVask [Vitamin K1 to Slow Vascular Cal- fied valve parts.100–102 IFN-γ is a well-known inductor
cification in Hemodialysis Patients], iPACK-HD [Inhibiting of M1 polarization and is widely used in vitro to achieve
the Progression of Arterial Calcification With Vitamin K M1 polarization. Stimulation of CD14+ monocytes in vitro
in HemoDialysis Patients], BASIK2 [Bicuspid Aortic Valve with IFN-γ leads to impaired osteoclastic activity with low
Stenosis and the Effect of Vitamin K2 on Calciummetab- expression of cathepsin K, TRAP, and RANKL.
olism on 18F-NaF PET/MRI], VitaK-CAC [The Effects of Impaired resorption capacity of cardiovascular osteo-
Vitamin K2 Supplementation on the Progression of Coro- clast-like cells seems to play a major role during cardio-
nary Artery Calcification]). vascular calcification by tipping the balance towards a
procalcifying milieu and thus contributing considerably to
Osteoclastic Differentiation
cardiovascular disease burden. Activating cardiovascular
Although there is an absence of real osteoclasts in the cardio-
osteoclast-like cells could be a promising route of halt-
vascular system, monocytes/macrophages can differentiate
ing or even reversing cardiovascular calcification through
into cardiovascular osteoclast-like cells, displaying mineral
resorption capacity.85,86 Indeed, during bone formation and improved mineral uptake by these cells.
homeostasis, bone preosteoclasts, which further mature into
multinucleated bone-resorbing osteoclasts, also arise from Intercellular Communication via EVs During
the monocyte cell lineage.87,88 In bone, osteoblasts are the Aortic Valve Calcification
natural counterplayer to osteoclasts, promoting bone forma-
tion.89 RANKL and MCSF (macrophage colony-stimulating Various cell types play roles during CAVD. These cells
factor), which are produced by osteoblastic stromal cells in communicate with each other and can influence pheno-
bone tissue, are necessary for osteoclast differentiation and typic switching, as shown by Hjortnaes et al.30 They dem-
maturation.90 Binding of RANKL to RANK on marrow stro- onstrated in coculture experiments that EndMT of VECs is
mal cells and preosteoclasts leads to osteoclast differentia- inhibited by the presence of VICs, and VECs significantly
tion and demineralization of the bone matrix. This effect is decreased the activation of VICs in vitro. Interestingly, the
counteracted by OPG, which acts as a soluble decoy recep- presence of elevated extracellular calcium and phosphate
tor.22 Conversely, in human VICs, RANKL binding can lead levels induces the release of calcifying EVs by VICs, and
to osteoblastic differentiation.91 This difference could be due spheroid mineralized EVs are produced from VICs under
Downloaded from http://ahajournals.org by on April 8, 2022

to the fact that although there is a high density of preosteo- mechanical strain (Figures 1 and 2).103,104 Patients under-
clasts in bone tissue, thus favoring RANKL’s ability to induce going a transcatheter aortic valve replacement proce-
osteoclastic differentiation, this cell type is absent in the car- dure showed an increase of circulating platelet-derived
diovascular system.92 The calcific effect is further enhanced EVs and a decrease in endothelial-derived EVs.105 These
by high levels of RANKL and extremely low levels of OPG in findings demonstrate that intercellular communication,
calcific aortic valves compared with healthy valves.91 including through EVs, can potentially regulate cellular
In the cardiovascular system, osteoclast-like cells can functions and phenotypes during aortic valve calcification.
arise from proinflammatory (M1) macrophages or anti- EVs are known to be actively involved during car-
inflammatory (M2) macrophages, although there is growing diovascular calcification processes and can promote
evidence of the existence of a greater variety of macrophage or inhibit calcification through differences in their lipid
subsets.85,86,93 The action of cardiovascular osteoclast-like composition and cargo.106 This cargo includes surface
cells is counterbalanced by VSMCs and VICs, which can molecules, proteins, and nucleic acids, such as DNA,
both differentiate into an osteoblastic cell type.94,95 Osteo- mRNA, and ncRNAs, which can be transferred between
blastic VSMCs are able to secrete RANKL and MCSF cells.106 By doing so, EVs are able to modify the function
required for osteoclastic differentiation of macrophages, or fate of the target/recipient cells.107,108 Within this con-
which can also be used in vitro to induce osteoclastic differ- text, ncRNAs are considered essential effectors of EVs
entiation.94,96 These osteoclast-like cells express TRAP (tar- because they regulate gene expression.109,110 Studies
trate-resistant acid phosphatase) and CTSK (cathepsin K), have shown that various stress signals in cardiovascu-
which are involved in bone resorption and mineral uptake97,98 lar disorders, including endothelial cell injury, trigger the
In atherosclerotic plaques, M2-derived cardiovascu- release of EVs and transport of ncRNAs.111,112
lar osteoclast-like cells cluster around calcific deposits The role of EVs during CAVD is currently unclear, but
and show a diminished mineral uptake capacity due to their role during atherosclerosis, cardiovascular calcifica-
decreased expression of TRAP and CTSK.97 Calcific tion, and endothelial regeneration has already been well
aortic valves contain osteoclast-like cells, which appear studied. Due to similarities of these diseases, EVs are
to arise from M1 polarized macrophages.99,100 Calci- likely to be of importance also in CAVD initiation and pro-
fied aortic valves also show a pronounced infiltration of gression. Further investigation is warranted to elucidate
IFN (interferon)-γ–secreting CD8+ T cells, and calcific the mechanisms involved.

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   891
Goody et al Aortic Valve Stenosis

Role of ncRNAs in Aortic Stenosis acts as a negative regulator of osteogenic differentiation


by repressing its direct target, Runx2, resulting in impaired
Brief Review - AL

Growing evidence suggests that ncRNAs are key regula-


expression of ALP and osteocalcin.
tors of osteogenesis in atherosclerosis, but knowledge
In addition to miRNAs, lncRNAs have also been found
about ncRNAs in CAVD remains very limited.113,114 Since
to be involved in CAVD. The lncRNA H19 was the first
their characterization during the human genome project as
nonprotein-coding sequences, ncRNAs have gained inter- lncRNA to be described in CAVD.121 Gain-of-function and
est as regulators of many biological as well as pathological loss-of-function experiments showed that NOTCH1 gene
processes, leading to the rise of a new discipline in biologi- expression is inhibited by H19, through interference with
cal research.115,116 Although the involvement of ncRNA in the binding of p53 to the NOTCH1 promoter.122 This, in
CAVD is still not completely understood, many studies have turn, induces expression of the pro-osteogenic genes
identified microRNAs (miRNAs, miRs) and lncRNAs (long RUNX2 and BMP-2.122 Xiao et al123 showed an increase
non-coding RNAs) that have specific functions within the in MALAT1 (metastasis-associated lung adenocarci-
cells of the aortic valve. The first functional study of miRNAs noma transcript 1) expression in CAVD patients. MALAT1
in AVS was carried out on miR-141. Ectopic expression of acts as a sponge for the protective miRNA, miR-204,
miRNA-141 in porcine VICs repressed TGF-β stimulated thereby reversing the inhibition of SMAD4 by miR-204
myofibroblast transformation in a BMP-2-dependent man- and promoting osteogenesis, as shown by an increase in
ner.118 Zhang et al119 revealed that miR-30b is significantly osteoblast markers, such as ALP.123 Similarly to MALAT1,
downregulated in CAVD samples and regulates human aor- taurine upregulated gene 1 (TUG1), was found to be
tic valvular calcification and apoptosis via Runx2, Smad1, significantly upregulated in calcified aortic valves and to
and caspase-3. In an attempt to further understand the func- directly interact with miR-204-5p.124 In a recent study
tional role of miRNAs in the osteoblastic differentiation of by Carrion et al,75 the lncRNA HOX transcript antisense
VICs, Wang et al120 analyzed miRNA levels in human calci- RNA (HOTAIR) was shown to be downregulated human
fied aortic valves and BMP-2-stimulated human VICs. They BAV cells after cyclic stretch and Wnt/β-catenin signal-
showed downregulation of miR-204 in both samples and ing. Furthermore, silencing HOTAIR resulted in increased
demonstrated through in vitro experiments that miR-204 expression of calcification inducing genes, including
Downloaded from http://ahajournals.org by on April 8, 2022

Figure 3. Graded wire injury model of aortic valve stenosis in mice.


Induction of aortic valve stenosis in mice is achieved by inserting a coronary wire into the left ventricle (LV) via the carotid artery. For mild and
moderate injury, a straight guidewire is pushed through the aortic valve 20 or 50× and rotated 50 or 100× (C and A). To achieve severe injury,
a spring wire with a 15° angled tip is pushed through the valve 20× and then rotated 200× (D and A). A and B, Parasternal long-axis views of
using ultra high-frequency echocardiography with straight or angled wire. E, Incidence of acute aortic insufficiency (AI) after wire injury. F, Peak
velocities over the aortic valve after sham, mild, moderate, or severe injury, compared with baseline. n.d. indicates not determined; and ns, not
significant. Adapted from Niepmann et al117 with permission. Copyright ©2019, Springer-Verlag GmbH Germany, part of Springer Nature.

892   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

ALP, thereby confirming its involvement in aortic valve valve material, especially for cell isolation or RNA analysis
calcification. because of the variable quality of the source material.

Brief Review - AL
In Vitro Calcification
Current Challenges—Model Systems The protocols for inducing calcification in vitro vary
immensely between labs. Both osteogenic media and
Although a number of interesting pathways leading to
procalcifying media have been used. The incubation of
CAVD have already been identified, the in vivo and in
isolated VICs with these media leads to an in vitro cal-
vitro data obtained can sometimes be unreliable, and
cification of cells, which can be visualized by alizarin
conflicting results have been observed from different
red staining and further quantified by quantitative poly-
laboratories. One explanation for this is the variety of dif- merase chain reaction or protein analysis of osteogenic
ferent cellular and in vivo systems and protocols used markers, for example, ALP. Osteogenic medium shows a
to examine CAVD. The following section summarizes the passage-dependent effect on VICs, where cells from a
current problems, challenges, and opportunities of exist- higher passage show diminished calcification potential.
ing model systems. This passage dependency could not be seen when using
In Vitro Models of Aortic Valve Calcification procalcifying medium.70 In addition, different growth fac-
Multiple studies have been undertaken on aortic valve– tors such as TGF-β1, TGF-β3, BMP-2, BMP-4, BMP-7,
derived cells that were isolated from humans or animals. and others have been used to induce myofibroblastic or
VECs and VICs are usually obtained from large animals, osteoblastic differentiation.12,67 Cell culture materials vary
between glass and plastic, or cells are grown on a coat-
such as pigs, sheep, and cows.125–127 It is also possible to
ing of collagen I, II, or III, or fibronectin.131 VICs cultured
isolate these cells from murine aortic valves.80 These ani-
on hard surfaces, such as plastic, show the tendency to
mal cells have a major drawback because there might be
spontaneously differentiate into a myofibroblastic phe-
large differences in the myofibroblastic and osteoblastic
notype.132 Therefore, studies have been undertaken
pathways between animals and humans.12
using 3-dimensional (3D) collagen hydrogels with differ-
One major problem for using human cells lies in the ent densities.133,134 A hybrid of methacrylated gelatin and
availability of aortic valve material, especially healthy valve methacrylated hyaluronic acid can also be used and has
tissue to be used as control samples. Diseased human shown promising results. Using this material, seeded VICs
valve tissue can be acquired from patients undergoing remained quiescent without treatment but could still dif-
Downloaded from http://ahajournals.org by on April 8, 2022

surgical aortic valve replacement due to CAVD. From this ferentiate into either myofibroblast-like cells upon TGF-
material, VECs and VICs can be isolated for in vitro exper- β stimulation or osteoblast-like cells upon treatment with
iments via collagenase treatment and magnetic-assisted osteogenic medium.30,135 Van der Valk et al105 even went
cell sorting.12 A good source of control samples is donor one step further. They analyzed the layer-specific mechan-
hearts that could not be transplanted.68 This poses a prob- ical properties of human AV tissue and created a 3D bio-
lem for most researchers since there are only few trans- printed CAVD model using UV-crosslinked methacrylated
plantation centers where these samples can be obtained. gelatin/methacrylated hyaluronic acid hydrogels. It is not
A second source can be the explanted hearts of trans- surprising that applying these different culture and calcifi-
plant recipients, if the aortic valve is not affected by the cation techniques can lead to immensely different results.
underlying disease.67 Another, more widely used control
In Vivo Models of AVS
sample are noncalcified aortic valves, which have been
Most animals do not naturally develop CAVD, but
explanted because of aortic valve insufficiency. However,
model systems have been developed in pigs and mice
these patients often present with aortic root aneurysms,
that induce AVS through a high-cholesterol diet.136,137
which has to be taken into account when analyzing the Another strategy has been to create genetic variants
acquired data.128 Pediatric valves that were removed that acquire stenosis.138 The reproducibility of these
because of an underlying congenital heart defect have systems is a problem because not all of the genetically
also been used as a source material.129 Indeed, the most modified or high-fat diet animals develop AVS. In addition
ideal control samples maybe those from patients present- to diet-induced and genetically induced AVS, Honda et
ing with aortic dissection,130 but an inherent problem is al139 developed a wire injury model, where a guidewire is
that these patients present as acute emergencies and inserted into the left ventricle of the heart under echocar-
have to be operated on quickly, making the acquisition diographic guidance via the right common carotid artery
of informed consent difficult. It is also possible to divide of C57BL/6 mice. The wire is then rotated to create an
an aortic valve cusp into calcific, fibrotic, and healthy tis- endothelial injury. Induction of AVS can be detected by
sue by using a near-infrared fluorescent calcium tracer.60 measuring the velocity of blood flow by echocardiogra-
A problem here lies in underlying genetic variants that phy.139 This method has recently been improved upon
can affect the whole valve and might distort the results to yield more consistent results.117 It is now possible to
obtained. Cadavers are the least desirable source of induce mild, moderate, or severe stenosis by changing

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   893
Goody et al Aortic Valve Stenosis

the wire type, tip angle, and the number of rotations (Fig- VICs, demonstrating sex-related differences in prolifera-
ure 3).117 Mouse aortic anatomy differs from humans in tion and apoptosis.146 Sex hormones may be responsible
Brief Review - AL

that they do not have a trilayer architecture.140 It is, there- for many of the observed effects since it is known that
fore, fundamentally necessary to reproduce any findings androgens play an important role during bone formation
from mice in human valves or human cells. as well as in the regulation of osteoblast and osteoclast
activity.147 Cardiovascular calcification was also shown to
In Vivo Imaging
occur at a higher rate in apolipoprotein E knockout mice
In vivo imaging is highly attractive because it can monitor
treated with androgens.148 Reinforcing these findings,
disease development over time in serial exams. Recent
treatment of murine VSMCs with testosterone can induce
advances in ultrahigh-frequency ultrasound make it
calcium deposition of VSMCs.149
possible to assess the cardiovasculature in small ani-
Sex-related differences in molecular pathways are
mals with high accuracy. AVS is evaluated by continu-
most likely highly relevant during initiation and progres-
ity equation, peak velocity, and mean pressure gradients
sion of CAVD and should be investigated more thor-
in suprasternal and parasternal long-axis views. Left
oughly to identify differences in disease pathology in
ventricular systolic and diastolic function can be char-
acterized by 2-dimensional echocardiography, M-mode, males and females, potentially leading to a sex-depen-
pulsed-wave–Doppler, and tissue-Doppler. Image acqui- dent therapeutic approach in the future.
sition and interpretation is quick and noninvasive, making Standardization
this imaging modality ideal for routine follow-ups.117 Many different in vivo and in vitro systems have been
Even more robust in serial measurements, magnetic established in laboratories all over the world. This het-
resonance imaging can provide additional information on erogeneity makes the reproducibility and comparabil-
3D flow and 3D structures. A comprehensive magnetic ity of studies difficult. A more standardized and uniform
resonance imaging approach, simultaneously assess- approach would be desirable for the future and could help
ing changes in valvular, left ventricular, and aortic function with the interpretation of data. A Collaborative Research
and morphology was recently proposed.141 High-resolution Initiative on Aortic Disease (TRR259, Deutsche Forschun-
magnetic resonance imaging (9,4 T) can visualize valve ori- gsgemeinschaft), consisting of the Universities of Bonn,
fice area, thickness of the cusps, 3D transvalvular flow, and Cologne, and Düsseldorf (Germany) has been established
aortic regurgitation. Furthermore, left ventricular parameters in the last year and aims to standardize in vivo and in vitro
can be calculated. Mice with AVS demonstrate a turbulent models and use these to elucidate disease pathways and
Downloaded from http://ahajournals.org by on April 8, 2022

flow pattern over the stenotic valve with increased aortic therapeutic targets to develop potential therapies.
strain and increased aortic and left ventricular wall thick-
ness.141 These noninvasive techniques and protocols are
already used in clinical scanners and might, therefore, serve Future Directions and Novel Therapeutic
as important tools for translational research approaches.142 Strategies
Sex Differences Much has been learned in the last years about aortic
The influence of sex on the development of CAVD has valve calcification, but the lack of ideal in vivo model
been described, with males harboring a 2-fold risk increase systems and standardized in vitro approaches have left
compared with females of acquiring AVS and higher rates many questions unanswered. New model systems, such
of aortic valve calcium density progression.18,143 These as the wire injury mouse model or 3D cell culture models,
findings stimulated studies investigating the calcium load could lead to a better understanding of disease initia-
of stenotic aortic valves in a sex-dependent manner. Mul- tion and progression. There are ongoing clinical studies
tidetector computed tomography unveiled higher calcium that are testing treatments targeting the pathways identi-
content in male patients with AVS while showing similar fied in the last few years. Denosumab and bisphospho-
AVS severity as females.144 Following up on this study, nates are currently being investigated in the SALTIRE II
Simard et al145 were able to show that women with com- trial (Scottish Aortic Stenosis and Lipid Lowering Trial,
parable AVS severity and valve weight density presented Impact on Regression). Administration of denosumab, a
with a higher grade of valvular fibrosis and significantly human monoclonal antibody against RANKL that inhibits
less calcium density. However, the majority of in vitro and its binding to RANK, mimics the role of OPG, thus pos-
in vivo studies have been carried out on valvular cells or sibly inhibiting osteoblastic differentiation in aortic valve
animals without addressing potential sex differences. One tissue.22 Bisphosphonates reduce bone resorption and
of the few studies looking into sex-related differences in have been shown to inhibit vascular and aortic valve cal-
gene expression of porcine aortic valve cusps revealed cification.150 They can also directly interfere with the pro-
183 significantly differentially expressed genes involved duction of inflammatory cytokines involved in CAVD.151
in disease-relevant pathways, such as apoptosis, ossifica- In 2013, Thanassoulis et al152 identified a single
tion, and inflammation. The authors further confirm their nucleotide polymorphism in the lipoprotein(a) locus
findings in sex-specific in vitro experiments with porcine (rs1045872), which was associated with aortic valve

894   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

calcification independent of coronary artery calcification possible mechanisms to selectively activate cardiovascu-
or coronary artery disease. Levels of Lp(a) (lipoprotein[a]) lar osteoclast-like cells.

Brief Review - AL
and genetic variants had already previously been shown Because the macrophage continuum seems to be
to be associated with coronary artery disease.153 Mecha- comprised of more subsets than just classical proinflam-
nistically, deposition of Lp(a), which contains apo(a), apo- matory and anti-inflammatory (M1 and M2) macrophages,
lipoprotein B100, and oxidized phospholipids in the vessel a better understanding of mineral uptake capacity of
intima, can promote atherosclerotic lesion progression and osteoclast-like cells derived from specific subsets is nec-
inflammation.154 As described earlier, oxidized lipoproteins essary. If a specific macrophage subset were to show
play a crucial role during CAVD progression. In a recent enhanced mineral uptake capacity, one could imagine
study by Zheng et al,155 it could be shown that high levels strategies to polarize cardiovascular macrophages into
of Lp(a) and oxidized phospholipid correlate with disease this subset, thus enhancing mineral uptake by osteoclast-
progression in CAVD. Furthermore, the authors could also like cells arising from this subset. This could possibly be
demonstrate their osteogenic effects on VICs in vitro. achieved by administering cytokines or synthetic peptides,
Lipid-lowering agents, such as statins and ezetimibe, were mimicking their action and thus promoting a specific mac-
shown not to be effective in halting progression of CAVD rophage subset polarization, or using existing antibodies
and even showed increased cancer rates in the SEAS trial against cytokines to block polarization into macrophage
(Simvastatin and Ezetimibe in Aortic Stenosis).156–158 One subsets with diminished mineral uptake capacity.
explanation is the lack of Lp(a)-lowering capabilities.159 Disease-modifying antirheumatic drugs, such as the
Lp(a)-lowering agents could provide a way of slowing or TNF-α antibody adalimumab, were shown to be able
halting disease progression of CAVD, and clinical trials on to influence synovial macrophage polarization to anti-
the effects of niacin, PCSK9 (proprotein convertase sub- inflammatory macrophages in vitro in an IL-10/STAT3
tilisin/kexin type 9) inhibitors, and antisense oligonucle- (Signal Transducer and Activator of Transcription 3)
otides against apo(a) mRNA are currently underway.160 dependent manner.162 Influencing macrophage polariza-
Targeting CAVD with repurposed substances also tion, therefore, seems to be a viable route and might be
appears to be a viable route. In this regard, Choi et al161 able to promote more effective calcium uptake. In the
studied the influence of DPP-4 (dipeptidyl peptidase-4) CANTOS trial (Canakinumab Antiinflammatory Throm-
inhibitors, such as sitagliptin, which is widely used for the bosis Outcome Study), treatment with Canakinumab, a
treatment of type 2 diabetes mellitus, on calcification of monoclonal antibody against IL-1,β was able to reduce
VICs. Depletion of NO in VECs, mimicking endothelial dys- cardiovascular mortality. In addition to having proinflam-
Downloaded from http://ahajournals.org by on April 8, 2022

function, resulted in activation of the NF-κB pathway in matory effects, it was shown that IL-1β produced by
VICs. NF-κB acts as a transcription factor for DPP-4, which vascular macrophages is able to directly induce calcifica-
can decrease IGF-1 (insulin-like growth factor 1) levels by tion of vascular mesenchymal cells.163 Whether observed
enzymatic degradation, leading to more osteoblastic differ- effects in the CANTOS trial were purely due to modula-
entiation of VICs. This effect was abrogated with DPP-4 tion of inflammatory pathways or also due to direct inhibi-
inhibitors. In a rabbit model of CAVD, animals fed with high- tion of osteoblastic differentiation remains unclear.
cholesterol diet and vitamin D2, and treated with sitagliptin, Key proteins during the propagation phase include
showed beneficial results on transvalvular peak velocity, RunX2 and BMP-2, whose expression is directly influ-
pressure gradients, and valve orifice area.161 These results enced by different miRNAs in CAVD. Targeting these could
could be the basis for human trials with DPP-4 inhibitors. be of special interest to break through the vicious circle
Given the prominent role of macrophages in all stages of calcification. One interesting way of lowering their con-
of CAVD, an elegant way to inhibit inflammation and val- centration in aortic valvular cells could be via the use of
vular and, in fact, also vascular calcification would be to EV- or liposome-bound miRNA sponges, small molecule
specifically target macrophages or T cells responsible inhibitors, antisense oligonucleotides, or miRNA-mimics,
for macrophage polarization. As discussed earlier in this although targeting the aortic valve can present as a prob-
review, an impaired mineral uptake capacity of cardio- lem for treatments such as these. Administering molecules
vascular osteoclast-like cells, which are derived from that also interfere with bone metabolism could lead to an
macrophages, is suggested to play an important role in increase in osteoporosis. Transcriptome analyses and next-
cardiovascular calcium deposition. Activation of osteo- generation sequencing could help identify target proteins
clast-like cells to promote mineral uptake could not only that are solely expressed on valvular cells so that EVs or
halt progression of calcific cardiovascular diseases but liposomes with homing properties could be designed to
also lead to regression in calcification. The inherent prob- administer drugs with minimal off-target effects.
lem with general activation of osteoclasts throughout the
human body would be loss of bone mass with conse-
quent osteoporosis. To our knowledge, there is no spe- CONCLUSIONS
cific activator of cardiovascular osteoclasts available at CAVD is a highly complex pathology, where an interplay
this point in time. Further studies are needed to identify between metabolic factors, immune cells, and resident

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   895
Goody et al Aortic Valve Stenosis

endothelial and interstitial cells drive progression of the 10. Michelena HI, Prakash SK, Della Corte A, Bissell MM, Anavekar N, Mathieu P,
Bossé Y, Limongelli G, Bossone E, Benson DW, et al; BAVCon Investigators.
disease. A lack of suitable in vivo model systems and
Brief Review - AL

Bicuspid aortic valve: identifying knowledge gaps and rising to the challenge
the great heterogeneity of in vitro methods used has from the international Bicuspid Aortic Valve Consortium (BAVCon). Circulation.
limited the reliability of the knowledge gained so far. 2014;129:2691–2704. doi: 10.1161/CIRCULATIONAHA.113.007851
11. Liu T, Xie M, Lv Q, Li Y, Fang L, Zhang L, Deng W, Wang J. Bicuspid
Newer methods with higher reproducibility will improve aortic valve: an update in morphology, genetics, biomarker, complica-
our understanding of this disease and undoubtedly lead tions, imaging diagnosis and treatment. Front Physiol. 2018;9:1921. doi:
to novel treatment strategies. 10.3389/fphys.2018.01921
12. Rutkovskiy A, Malashicheva A, Sullivan G, Bogdanova M, Kostareva A,
Stensløkken KO, Fiane A, Vaage J. Valve interstitial cells: the key to under-
standing the pathophysiology of heart valve calcification. J Am Heart Assoc.
ARTICLE INFORMATION 2017;6:e006339. doi:10.1161/JAHA.117.006339
Received October 16, 2019; accepted February 14, 2020. 13. Vesely I. The role of elastin in aortic valve mechanics. J Biomech.
1998;31:115–123. doi: 10.1016/s0021-9290(97)00122-x
Affiliations 14. Simionescu DT, Lovekamp JJ, Vyavahare NR. Glycosaminoglycan-degrad-
From the Heart Center Bonn, Department of Medicine II, University Hospital ing enzymes in porcine aortic heart valves: implications for bioprosthetic
Bonn, Germany (P.R.G., M.R.H., D.C., S.T.N., S.Z., G.N., F.J.); Clinic for Internal Medi- heart valve degeneration. J Heart Valve Dis. 2003;12:217–225.
cine II, University Hospital Cologne, Germany (M.A.); and Clinic for Cardiology, 15. Yutzey KE, Demer LL, Body SC, Huggins GS, Towler DA, Giachelli CM,
Pulmonology, and Angiology, University Hospital Düsseldorf, Germany (F.B.). Hofmann-Bowman MA, Mortlock DP, Rogers MB, Sadeghi MM, et al.
Calcific aortic valve disease: a consensus summary from the alliance of
Acknowledgments investigators on calcific aortic valve disease. Arterioscler Thromb Vasc Biol.
We thank Dr Deborah Goody for valuable discussions and Dr Meghan Campbell 2014;34:2387–2393. doi: 10.1161/ATVBAHA.114.302523
for critical reading of the article. 16. Lindman BR, Clavel MA, Mathieu P, Iung B, Lancellotti P, Otto CM,
Pibarot P. Calcific aortic stenosis. Nat Rev Dis Primers. 2016;2:16006. doi:
Sources of Funding 10.1038/nrdp.2016.6
P.R. Goody is funded by the Else-Kröner-Fresenius Foundation, F. Jansen is 17. Novaro GM, Katz R, Aviles RJ, Gottdiener JS, Cushman M, Psaty BM,
funded by the Deutsche Forschungsgemeinschaft (JA 2351/2-1 and SFB Otto CM, Griffin BP. Clinical factors, but not C-reactive protein, predict pro-
TRR [Sonderforschungsbereich/Transregio] 259/1) and the Corona founda- gression of calcific aortic-valve disease: the Cardiovascular Health Study. J
tion. M.R. Hosen is funded by the Corona foundation. STN is funded by the Am Coll Cardiol. 2007;50:1992–1998. doi: 10.1016/j.jacc.2007.07.064
Else-Kröner-Fresenius Foundation. A. Zietzer is funded by the BONFOR (Bonn 18. Owens DS, Katz R, Takasu J, Kronmal R, Budoff MJ, O’Brien KD. Incidence
forscht) program. F. Bönner is funded by the Deutsche Forschungsgemeinschaft and progression of aortic valve calcium in the Multi-ethnic Study of Ath-
(BO 4264/1-1 and SFB TRR 259/1), G. Nickenig, S. Zimmer, and M. Adam are erosclerosis (MESA). Am J Cardiol. 2010;105:701–708. doi: 10.1016/j.
supported by the Deutsche Forschungsgemeinschaft (SFB TRR 259/1). amjcard.2009.10.071
19. Mack MJ, Leon MB, Thourani VH, Makkar R, Kodali SK, Russo M,
Disclosures Kapadia SR, Malaisrie SC, Cohen DJ, Pibarot P, et al; PARTNER 3 Inves-
None. tigators. Transcatheter aortic-valve replacement with a balloon-expandable
valve in low-risk patients. N Engl J Med. 2019;380:1695–1705. doi:
Downloaded from http://ahajournals.org by on April 8, 2022

10.1056/NEJMoa1814052
20. Popma JJ, Deeb GM, Yakubov SJ, Mumtaz M, Gada H, O’Hair D,
REFERENCES Bajwa T, Heiser JC, Merhi W, Kleiman NS, et al; Evolut Low Risk Trial
1. Nkomo VT, Gardin JM, Skelton TN, Gottdiener JS, Scott CG, Enriquez- Investigators. Transcatheter aortic-valve replacement with a self-expand-
Sarano M. Burden of valvular heart diseases: a population-based study. ing valve in low-risk patients. N Engl J Med. 2019;380:1706–1715. doi:
Lancet. 2006;368:1005–1011. doi: 10.1016/S0140-6736(06)69208-8 10.1056/NEJMoa1816885
2. Osnabrugge RL, Mylotte D, Head SJ, Van Mieghem NM, Nkomo VT, 21. New SE, Aikawa E. Molecular imaging insights into early inflammatory
LeReun CM, Bogers AJ, Piazza N, Kappetein AP. Aortic stenosis in the stages of arterial and aortic valve calcification. Circ Res. 2011;108:1381–
elderly: disease prevalence and number of candidates for transcatheter aor- 1391. doi: 10.1161/CIRCRESAHA.110.234146
tic valve replacement: a meta-analysis and modeling study. J Am Coll Cardiol. 22. Pawade TA, Newby DE, Dweck MR. Calcification in aortic stenosis: the
2013;62:1002–1012. doi: 10.1016/j.jacc.2013.05.015 skeleton key. J Am Coll Cardiol. 2015;66:561–577. doi: 10.1016/j.jacc.
3. Nightingale AK, Horowitz JD. Aortic sclerosis: not an innocent murmur but 2015.05.066
a marker of increased cardiovascular risk. Heart. 2005;91:1389–1393. doi: 23. Aikawa E, Nahrendorf M, Figueiredo JL, Swirski FK, Shtatland T, Kohler
10.1136/hrt.2004.057117 RH, Jaffer FA, Aikawa M, Weissleder R. Osteogenesis associates with inflam-
4. Makkar RR, Fontana GP, Jilaihawi H, Kapadia S, Pichard AD, mation in early-stage atherosclerosis evaluated by molecular imaging in
Douglas PS, Thourani VH, Babaliaros VC, Webb JG, Herrmann HC, et al; vivo. Circulation. 2007;116:2841–2850. doi: 10.1161/CIRCULATIONAHA.
PARTNER Trial Investigators. Transcatheter aortic-valve replacement for 107.732867
inoperable severe aortic stenosis. N Engl J Med. 2012;366:1696–1704. 24. Otto CM, Kuusisto J, Reichenbach DD, Gown AM, O’Brien KD. Characteriza-
doi: 10.1056/NEJMoa1202277 tion of the early lesion of ‘degenerative’ valvular aortic stenosis. Histologi-
5. Otto CM, Prendergast B. Aortic-valve stenosis–from patients at risk cal and immunohistochemical studies. Circulation. 1994;90:844–853. doi:
to severe valve obstruction. N Engl J Med. 2014;371:744–756. doi: 10.1161/01.cir.90.2.844
10.1056/NEJMra1313875 25. Yearwood TL, Misbach GA, Chandran KB. Experimental fluid dynamics
6. Siu SC, Silversides CK. Bicuspid aortic valve disease. J Am Coll Cardiol. of aortic stenosis in a model of the human aorta. Clin Phys Physiol Meas.
2010;55:2789–2800. doi: 10.1016/j.jacc.2009.12.068 1989;10:11–24. doi: 10.1088/0143-0815/10/1/002
7. Wirrig EE, Yutzey KE. Conserved transcriptional regulatory mechanisms 26. Fernández Esmerats J, Heath J, Jo H. Shear-sensitive genes in aor-
in aortic valve development and disease. Arterioscler Thromb Vasc Biol. tic valve endothelium. Antioxid Redox Signal. 2016;25:401–414. doi:
2014;34:737–741. doi: 10.1161/ATVBAHA.113.302071 10.1089/ars.2015.6554
8. Yassine NM, Shahram JT, Body SC. Pathogenic mechanisms of bicuspid 27. Eisenberg LM, Markwald RR. Molecular regulation of atrioventricular valvu-
aortic valve aortopathy. Front Physiol. 2017;8:687. doi: 10.3389/fphys. loseptal morphogenesis. Circ Res. 1995;77:1–6. doi: 10.1161/01.res.77.1.1
2017.00687 28. Aikawa E, Whittaker P, Farber M, Mendelson K, Padera RF, Aikawa
9. Kerstjens-Frederikse WS, van de Laar IM, Vos YJ, Verhagen JM, M, Schoen FJ. Human semilunar cardiac valve remodeling by activated
Berger RM, Lichtenbelt KD, Klein Wassink-Ruiter JS, van der Zwaag PA, cells from fetus to adult: implications for postnatal adaptation, pathol-
du Marchie Sarvaas GJ, Bergman KA, et al. Cardiovascular malformations ogy, and tissue engineering. Circulation. 2006;113:1344–1352. doi:
caused by NOTCH1 mutations do not keep left: data on 428 probands 10.1161/CIRCULATIONAHA.105.591768
with left-sided CHD and their families. Genet Med. 2016;18:914–923. doi: 29. Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM,
10.1038/gim.2015.193 Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC, et al.

896   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

Notch promotes epithelial-mesenchymal transition during cardiac devel- of aortic valve interstitial cells to inflammation. J Surg Res. 2014;190:1–8.
opment and oncogenic transformation. Genes Dev. 2004;18:99–115. doi: doi: 10.1016/j.jss.2014.03.051

Brief Review - AL
10.1101/gad.276304 48. Zhan Q, Song R, Zeng Q, Yao Q, Ao L, Xu D, Fullerton DA, Meng X. Acti-
30. Hjortnaes J, Shapero K, Goettsch C, Hutcheson JD, Keegan J, Kluin J, vation of TLR3 induces osteogenic responses in human aortic valve
Mayer JE, Bischoff J, Aikawa E. Valvular interstitial cells suppress calcifica- interstitial cells through the NF-κB and ERK1/2 pathways. Int J Biol Sci.
tion of valvular endothelial cells. Atherosclerosis. 2015;242:251–260. doi: 2015;11:482–493. doi: 10.7150/ijbs.10905
10.1016/j.atherosclerosis.2015.07.008 49. Hsu H, Shu HB, Pan MG, Goeddel DV. TRADD-TRAF2 and TRADD-FADD
31. Paruchuri S, Yang JH, Aikawa E, Melero-Martin JM, Khan ZA, interactions define two distinct TNF receptor 1 signal transduction path-
Loukogeorgakis S, Schoen FJ, Bischoff J. Human pulmonary valve progeni- ways. Cell. 1996;84:299–308. doi: 10.1016/s0092-8674(00)80984-8
tor cells exhibit endothelial/mesenchymal plasticity in response to vascular 50. Lee HL, Woo KM, Ryoo HM, Baek JH. Tumor necrosis factor-alpha
endothelial growth factor-A and transforming growth factor-beta2. Circ Res. increases alkaline phosphatase expression in vascular smooth muscle cells
2006;99:861–869. doi: 10.1161/01.RES.0000245188.41002.2c via MSX2 induction. Biochem Biophys Res Commun. 2010;391:1087–
32. Simmons CA, Grant GR, Manduchi E, Davies PF. Spatial heterogeneity of 1092. doi: 10.1016/j.bbrc.2009.12.027
endothelial phenotypes correlates with side-specific vulnerability to calci- 51. Isoda K, Matsuki T, Kondo H, Iwakura Y, Ohsuzu F. Deficiency of interleukin-1
fication in normal porcine aortic valves. Circ Res. 2005;96:792–799. doi: receptor antagonist induces aortic valve disease in BALB/c mice. Arterioscler
10.1161/01.RES.0000161998.92009.64 Thromb Vasc Biol. 2010;30:708–715. doi: 10.1161/ATVBAHA.109.201749
33. Richards J, El-Hamamsy I, Chen S, Sarang Z, Sarathchandra P, Yacoub MH, 52. Galeone A, Brunetti G, Oranger A, Greco G, Di Benedetto A, Mori G,
Chester AH, Butcher JT. Side-specific endothelial-dependent regulation of Colucci S, Zallone A, Paparella D, Grano M. Aortic valvular interstitial cells
aortic valve calcification: interplay of hemodynamics and nitric oxide signaling. apoptosis and calcification are mediated by TNF-related apoptosis-inducing
Am J Pathol. 2013;182:1922–1931. doi: 10.1016/j.ajpath.2013.01.037 ligand. Int J Cardiol. 2013;169:296–304. doi: 10.1016/j.ijcard.2013.09.012
34. Abdelbaky A, Corsini E, Figueroa AL, Subramanian S, Fontanez S, 53. Pfeifer P, Steinmetz M, Ackerschott A, Dregger H, Jehle J, Nickenig G,
Emami H, Hoffmann U, Narula J, Tawakol A. Early aortic valve inflammation Latz E, Zimmer S. Role of CLEC4E expression in development of aortic
precedes calcification: a longitudinal FDG-PET/CT study. Atherosclerosis. valve stenosis. Eur Heart J. 2018;39 (suppl):1070. doi: 10.1093/eurheartj/
2015;238:165–172. doi: 10.1016/j.atherosclerosis.2014.11.026 ehy566.P5128
35. O’Brien KD, Reichenbach DD, Marcovina SM, Kuusisto J, Alpers CE, 54. Clément M, Basatemur G, Masters L, Baker L, Bruneval P, Iwawaki T,
Otto CM. Apolipoproteins B, (a), and E accumulate in the morphologically Kneilling M, Yamasaki S, Goodall J, Mallat Z. Necrotic cell sensor clec4e
early lesion of ‘degenerative’ valvular aortic stenosis. Arterioscler Thromb promotes a proatherogenic macrophage phenotype through activation of
Vasc Biol. 1996;16:523–532. doi: 10.1161/01.atv.16.4.523 the unfolded protein response. Circulation. 2016;134:1039–1051. doi:
36. Miller JD, Chu Y, Brooks RM, Richenbacher WE, Peña-Silva R, Heistad DD. 10.1161/CIRCULATIONAHA.116.022668
Dysregulation of antioxidant mechanisms contributes to increased oxida- 55. Kim KM. Calcification of matrix vesicles in human aortic valve and aortic
tive stress in calcific aortic valvular stenosis in humans. J Am Coll Cardiol. media. Fed Proc. 1976;35:156–162.
2008;52:843–850. doi: 10.1016/j.jacc.2008.05.043 56. Proudfoot D, Skepper JN, Hegyi L, Bennett MR, Shanahan CM, Weissberg
37. Liberman M, Bassi E, Martinatti MK, Lario FC, Wosniak J Jr, Pomerantzeff PM, PL. Apoptosis regulates human vascular calcification in vitro: evidence for
Laurindo FR. Oxidant generation predominates around calcifying foci and initiation of vascular calcification by apoptotic bodies. Circ Res. 2000;87:
enhances progression of aortic valve calcification. Arterioscler Thromb Vasc 1055–1062. doi: 10.1161/01.res.87.11.1055
Biol. 2008;28:463–470. doi: 10.1161/ATVBAHA.107.156745 57. New SE, Goettsch C, Aikawa M, Marchini JF, Shibasaki M, Yabusaki K, Libby
P, Shanahan CM, Croce K, Aikawa E. Macrophage-derived matrix vesicles: an
38. Parhami F, Morrow AD, Balucan J, Leitinger N, Watson AD, Tintut Y,
alternative novel mechanism for microcalcification in atherosclerotic plaques.
Downloaded from http://ahajournals.org by on April 8, 2022

Berliner JA, Demer LL. Lipid oxidation products have opposite effects on
Circ Res. 2013;113:72–77. doi: 10.1161/CIRCRESAHA.113.301036
calcifying vascular cell and bone cell differentiation. A possible explanation
58. Hjortnaes J, New SE, Aikawa E. Visualizing novel concepts of cardio-
for the paradox of arterial calcification in osteoporotic patients. Arterioscler
vascular calcification. Trends Cardiovasc Med. 2013;23:71–79. doi:
Thromb Vasc Biol. 1997;17:680–687. doi: 10.1161/01.atv.17.4.680
10.1016/j.tcm.2012.09.003
39. Lehti S, Käkelä R, Hörkkö S, Kummu O, Helske-Suihko S, Kupari M,
59. New SE, Aikawa E. Cardiovascular calcification: an inflammatory disease.
Werkkala K, Kovanen PT, Oörni K. Modified lipoprotein-derived lipid particles
Circ J. 2011;75:1305–1313. doi: 10.1253/circj.cj-11-0395
accumulate in human stenotic aortic valves. PLoS One. 2013;8:e65810.
60. Schlotter F, Halu A, Goto S, Blaser MC, Body SC, Lee LH, Higashi
doi: 10.1371/journal.pone.0065810
H, DeLaughter DM, Hutcheson JD, Vyas P, et al. Spatiotemporal multi-
40. Mahmut A, Boulanger MC, El Husseini D, Fournier D, Bouchareb R,
omics mapping generates a molecular atlas of the aortic valve and
Després JP, Pibarot P, Bossé Y, Mathieu P. Elevated expression of lipo-
reveals networks driving disease. Circulation. 2018;138:377–393. doi:
protein-associated phospholipase A2 in calcific aortic valve disease: impli-
10.1161/CIRCULATIONAHA.117.032291
cations for valve mineralization. J Am Coll Cardiol. 2014;63:460–469. doi:
61. Latif N, Quillon A, Sarathchandra P, McCormack A, Lozanoski A, Yacoub
10.1016/j.jacc.2013.05.105
MH, Chester AH. Modulation of human valve interstitial cell phenotype
41. Mahmut A, Mahjoub H, Boulanger MC, Fournier D, Després JP, Pibarot P, and function using a fibroblast growth factor 2 formulation. PLoS One.
Mathieu P. Lp-PLA2 is associated with structural valve degeneration of bio- 2015;10:e0127844. doi: 10.1371/journal.pone.0127844
prostheses. Eur J Clin Invest. 2014;44:136–145. doi: 10.1111/eci.12199 62. Witt W, Jannasch A, Burkhard D, Christ T, Ravens U, Brunssen C, Leuner A,
42. Olsson M, Thyberg J, Nilsson J. Presence of oxidized low density lipopro- Morawietz H, Matschke K, Waldow T. Sphingosine-1-phosphate induces
tein in nonrheumatic stenotic aortic valves. Arterioscler Thromb Vasc Biol. contraction of valvular interstitial cells from porcine aortic valves. Cardiovasc
1999;19:1218–1222. doi: 10.1161/01.atv.19.5.1218 Res. 2012;93:490–497. doi: 10.1093/cvr/cvs002
43. Mohty D, Pibarot P, Després JP, Côté C, Arsenault B, Cartier A, 63. Osman N, Grande-Allen KJ, Ballinger ML, Getachew R, Marasco S,
Cosnay P, Couture C, Mathieu P. Association between plasma LDL particle O’Brien KD, Little PJ. Smad2-dependent glycosaminoglycan elongation in
size, valvular accumulation of oxidized LDL, and inflammation in patients aortic valve interstitial cells enhances binding of LDL to proteoglycans. Car-
with aortic stenosis. Arterioscler Thromb Vasc Biol. 2008;28:187–193. doi: diovasc Pathol. 2013;22:146–155. doi: 10.1016/j.carpath.2012.07.002
10.1161/ATVBAHA.107.154989 64. Hutcheson JD, Ryzhova LM, Setola V, Merryman WD. 5-HT(2B)
44. Sucosky P, Balachandran K, Elhammali A, Jo H, Yoganathan AP. Altered antagonism arrests non-canonical TGF-β1-induced valvular myofi-
shear stress stimulates upregulation of endothelial VCAM-1 and ICAM-1 broblast differentiation. J Mol Cell Cardiol. 2012;53:707–714. doi:
in a BMP-4- and TGF-beta1-dependent pathway. Arterioscler Thromb Vasc 10.1016/j.yjmcc.2012.08.012
Biol. 2009;29:254–260. doi: 10.1161/ATVBAHA.108.176347 65. Hutcheson JD, Chen J, Sewell-Loftin MK, Ryzhova LM, Fisher CI, Su YR,
45. Mathieu P, Bouchareb R, Boulanger MC. Innate and adaptive immunity Merryman WD. Cadherin-11 regulates cell-cell tension necessary for cal-
in calcific aortic valve disease. J Immunol Res. 2015;2015:851945. doi: cific nodule formation by valvular myofibroblasts. Arterioscler Thromb Vasc
10.1155/2015/851945 Biol. 2013;33:114–120. doi: 10.1161/ATVBAHA.112.300278
46. West XZ, Malinin NL, Merkulova AA, Tischenko M, Kerr BA, Borden EC, 66. Zhou J, Bowen C, Lu G, Knapp Iii C, Recknagel A, Norris RA, Butcher JT.
Podrez EA, Salomon RG, Byzova TV. Oxidative stress induces angiogenesis Cadherin-11 expression patterns in heart valves associate with key func-
by activating TLR2 with novel endogenous ligands. Nature. 2010;467:972– tions during embryonic cushion formation, valve maturation and calcification.
976. doi: 10.1038/nature09421 Cells Tissues Organs. 2013;198:300–310. doi: 10.1159/000356762
47. Venardos N, Nadlonek NA, Zhan Q, Weyant MJ, Reece TB, Meng X, 67. Osman L, Yacoub MH, Latif N, Amrani M, Chester AH. Role
Fullerton DA. Aortic valve calcification is mediated by a differential response of human valve interstitial cells in valve calcification and their

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   897
Goody et al Aortic Valve Stenosis

response to atorvastatin. Circulation. 2006;114(suppl 1):I547–I552. doi: 87. Feng X, Teitelbaum SL. Osteoclasts: new insights. Bone Res. 2013;1:11–
10.1161/CIRCULATIONAHA.105.001115 26. doi: 10.4248/BR201301003
Brief Review - AL

68. Yang X, Fullerton DA, Su X, Ao L, Cleveland JC Jr, Meng X. Pro-osteo- 88. Teitelbaum SL. Bone resorption by osteoclasts. Science. 2000;289:1504–
genic phenotype of human aortic valve interstitial cells is associated with 1508. doi: 10.1126/science.289.5484.1504
higher levels of Toll-like receptors 2 and 4 and enhanced expression of 89. Komori T. Regulation of proliferation, differentiation and func-
bone morphogenetic protein 2. J Am Coll Cardiol. 2009;53:491–500. doi: tions of osteoblasts by Runx2. Int J Mol Sci. 2019;20:E1694. doi:
10.1016/j.jacc.2008.09.052 10.3390/ijms20071694
69. Ducy P. Cbfa1: a molecular switch in osteoblast biology. Dev Dyn. 90. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin TJ. Modulation
2000;219:461–471. doi: 10.1002/1097-0177(2000)9999:9999<:: of osteoclast differentiation and function by the new members of the tumor
AID-DVDY1074>3.0.CO;2-C necrosis factor receptor and ligand families. Endocr Rev. 1999;20:345–
70. Goto S, Rogers MA, Blaser MC, Higashi H, Lee LH, Schlotter F, Body SC, 357. doi: 10.1210/edrv.20.3.0367
Aikawa M, Singh SA, Aikawa E. Standardization of human calcific aortic 91. Kaden JJ, Bickelhaupt S, Grobholz R, Haase KK, Sarikoç A, Kiliç R,
valve disease in vitro modeling reveals passage-dependent calcification. Brueckmann M, Lang S, Zahn I, Vahl C, et al. Receptor activator of nuclear
Front Cardiovasc Med. 2019;6:49. doi: 10.3389/fcvm.2019.00049 factor kappaB ligand and osteoprotegerin regulate aortic valve calcifica-
71. Hutcheson JD, Aikawa E, Merryman WD. Potential drug targets for tion. J Mol Cell Cardiol. 2004;36:57–66. doi: 10.1016/j.yjmcc.2003.09.015
calcific aortic valve disease. Nat Rev Cardiol. 2014;11:218–231. doi: 92. Tintut Y, Demer L. Role of osteoprotegerin and its ligands and competing
10.1038/nrcardio.2014.1 receptors in atherosclerotic calcification. J Investig Med. 2006;54:395–
72. Torre M, Hwang DH, Padera RF, Mitchell RN, VanderLaan PA. Osseous 401. doi: 10.2310/6650.2006.06019
and chondromatous metaplasia in calcific aortic valve stenosis. Cardiovasc 93. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S,
Pathol. 2016;25:18–24. doi: 10.1016/j.carpath.2015.08.008 Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al. Macrophage activa-
73. Mohler ER 3rd, Gannon F, Reynolds C, Zimmerman R, Keane MG, tion and polarization: nomenclature and experimental guidelines. Immunity.
Kaplan FS. Bone formation and inflammation in cardiac valves. Circulation. 2014;41:14–20. doi: 10.1016/j.immuni.2014.06.008
2001;103:1522–1528. doi: 10.1161/01.cir.103.11.1522 94. Massy ZA, Mentaverri R, Mozar A, Brazier M, Kamel S. The pathophysi-
74. Conti CA, Della Corte A, Votta E, Del Viscovo L, Bancone C, De Santo LS, ology of vascular calcification: are osteoclast-like cells the missing link?
Redaelli A. Biomechanical implications of the congenital bicuspid aortic valve: Diabetes Metab. 2008;34(suppl 1):S16–S20. doi: 10.1016/S1262-
a finite element study of aortic root function from in vivo data. J Thorac Cardio- 3636(08)70098-3
vasc Surg. 2010;140:890–896, 896.e1. doi: 10.1016/j.jtcvs.2010.01.016 95. Rajamannan NM, Subramaniam M, Rickard D, Stock SR, Donovan J,
75. Carrion K, Dyo J, Patel V, Sasik R, Mohamed SA, Hardiman G, Nigam V. Springett M, Orszulak T, Fullerton DA, Tajik AJ, Bonow RO, et al. Human aor-
The long non-coding HOTAIR is modulated by cyclic stretch and WNT/β- tic valve calcification is associated with an osteoblast phenotype. Circulation.
CATENIN in human aortic valve cells and is a novel repressor of calcification 2003;107:2181–2184. doi: 10.1161/01.CIR.0000070591.21548.69
genes. PLoS One. 2014;9:e96577. doi: 10.1371/journal.pone.0096577 96. Doherty TM, Asotra K, Fitzpatrick LA, Qiao JH, Wilkin DJ, Detrano RC,
76. Nigam V, Srivastava D. Notch1 represses osteogenic pathways in aor- Dunstan CR, Shah PK, Rajavashisth TB. Calcification in atherosclerosis: bone
tic valve cells. J Mol Cell Cardiol. 2009;47:828–834. doi: 10.1016/j. biology and chronic inflammation at the arterial crossroads. Proc Natl Acad
yjmcc.2009.08.008 Sci U S A. 2003;100:11201–11206. doi: 10.1073/pnas.1932554100
77. Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, 97. Chinetti-Gbaguidi G, Daoudi M, Rosa M, Vinod M, Louvet L, Copin C,
Grossfeld PD, Srivastava D. Mutations in NOTCH1 cause aortic valve dis- Fanchon M, Vanhoutte J, Derudas B, Belloy L, et al. Human alternative
ease. Nature. 2005;437:270–274. doi: 10.1038/nature03940 macrophages populate calcified areas of atherosclerotic lesions and dis-
Downloaded from http://ahajournals.org by on April 8, 2022

78. Caira FC, Stock SR, Gleason TG, McGee EC, Huang J, Bonow RO, play impaired RANKL-induced osteoclastic bone resorption activity. Circ
Spelsberg TC, McCarthy PM, Rahimtoola SH, Rajamannan NM. Human Res. 2017;121:19–30. doi: 10.1161/CIRCRESAHA.116.310262
degenerative valve disease is associated with up-regulation of low-density 98. Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation.
lipoprotein receptor-related protein 5 receptor-mediated bone formation. J Nature. 2003;423:337–342. doi: 10.1038/nature01658
Am Coll Cardiol. 2006;47:1707–1712. doi: 10.1016/j.jacc.2006.02.040 99. Nagy E, Eriksson P, Yousry M, Caidahl K, Ingelsson E, Hansson GK,
79. Côté N, El Husseini D, Pépin A, Guauque-Olarte S, Ducharme V, Franco-Cereceda A, Bäck M. Valvular osteoclasts in calcification and
Bouchard-Cannon P, Audet A, Fournier D, Gaudreault N, Derbali H, et al. aortic valve stenosis severity. Int J Cardiol. 2013;168:2264–2271. doi:
ATP acts as a survival signal and prevents the mineralization of aortic valve. 10.1016/j.ijcard.2013.01.207
J Mol Cell Cardiol. 2012;52:1191–1202. doi: 10.1016/j.yjmcc.2012.02.003 100. Nagy E, Lei Y, Martínez-Martínez E, Body SC, Schlotter F, Creager M,
80. El Husseini D, Boulanger MC, Mahmut A, Bouchareb R, Laflamme MH, Assmann A, Khabbaz K, Libby P, Hansson GK, et al. Interferon-γ released by
Fournier D, Pibarot P, Bossé Y, Mathieu P. P2Y2 receptor represses activated CD8+ T lymphocytes impairs the calcium resorption potential of
IL-6 expression by valve interstitial cells through Akt: implication for osteoclasts in calcified human aortic valves. Am J Pathol. 2017;187:1413–
calcific aortic valve disease. J Mol Cell Cardiol. 2014;72:146–156. doi: 1425. doi: 10.1016/j.ajpath.2017.02.012
10.1016/j.yjmcc.2014.02.014 101. Mazzone A, Epistolato MC, De Caterina R, Storti S, Vittorini S, Sbrana S,
81. Schurgers LJ, Uitto J, Reutelingsperger CP. Vitamin K-dependent carboxyl- Gianetti J, Bevilacqua S, Glauber M, Biagini A, et al. Neoangiogenesis,
ation of matrix Gla-protein: a crucial switch to control ectopic mineralization. T-lymphocyte infiltration, and heat shock protein-60 are biological hall-
Trends Mol Med. 2013;19:217–226. doi: 10.1016/j.molmed.2012.12.008 marks of an immunomediated inflammatory process in end-stage calci-
82. Tantisattamo E, Han KH, O’Neill WC. Increased vascular calcification in fied aortic valve stenosis. J Am Coll Cardiol. 2004;43:1670–1676. doi:
patients receiving warfarin. Arterioscler Thromb Vasc Biol. 2015;35:237– 10.1016/j.jacc.2003.12.041
242. doi: 10.1161/ATVBAHA.114.304392 102. Winchester R, Wiesendanger M, O’Brien W, Zhang HZ, Maurer MS,
83. Yamamoto K, Koretsune Y, Akasaka T, Kisanuki A, Ohte N, Takenaka T, Gillam LD, Schwartz A, Marboe C, Stewart AS. Circulating activated and
Takeuchi M, Yoshida K, Iwade K, Okuyama Y, et al; Japanese Aortic Ste- effector memory T cells are associated with calcification and clonal expan-
nosis Study-2 (JASS-2) Investigators. Effects of vitamin K antagonist on sions in bicuspid and tricuspid valves of calcific aortic stenosis. J Immunol.
aortic valve degeneration in non-valvular atrial fibrillation patients: Pro- 2011;187:1006–1014. doi: 10.4049/jimmunol.1003521
spective 4-year observational study. Thromb Res. 2017;160:69–75. doi: 103. Cui L, Rashdan NA, Zhu D, Milne EM, Ajuh P, Milne G, Helfrich MH, Lim K,
10.1016/j.thromres.2017.10.027 Prasad S, Lerman DA, et al. End stage renal disease-induced hypercalcemia
84. Geleijnse JM, Vermeer C, Grobbee DE, Schurgers LJ, Knapen MH, may promote aortic valve calcification via annexin VI enrichment of valve in-
van der Meer IM, Hofman A, Witteman JC. Dietary intake of menaquinone terstitial cell derived-matrix vesicles. J Cell Physiol. 2017;232:2985–2995.
is associated with a reduced risk of coronary heart disease: the Rotterdam doi: 10.1002/jcp.25935
Study. J Nutr. 2004;134:3100–3105. doi: 10.1093/jn/134.11.3100 104. Bouchareb R, Boulanger MC, Fournier D, Pibarot P, Messaddeq Y,
85. Rogers MA, Aikawa M, Aikawa E. Macrophage heterogeneity complicates Mathieu P. Mechanical strain induces the production of spheroid mineral-
reversal of calcification in cardiovascular tissues. Circ Res. 2017;121:5–7. ized microparticles in the aortic valve through a RhoA/ROCK-dependent
doi: 10.1161/CIRCRESAHA.117.311219 mechanism. J Mol Cell Cardiol. 2014;67:49–59. doi: 10.1016/j.
86. Hénaut L, Candellier A, Boudot C, Grissi M, Mentaverri R, Choukroun G, Brazier M, yjmcc.2013.12.009
Kame S, Massy ZA. New insights into the roles of monocytes/macrophages 105. van der Valk DC, van der Ven CFT, Blaser MC, Grolman JM, Wu PJ,
in cardiovascular calcification associated with chronic kidney disease. Toxins Fenton OS, Lee LH, Tibbitt MW, Andresen JL, Wen JR, et al.
(Basel). 2019;11:529. doi:10.3390/toxins11090529 Engineering a 3D-bioprinted model of human heart valve disease using

898   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067
Goody et al Aortic Valve Stenosis

nanoindentation-based biomechanics. Nanomaterials (Basel). 2018;8:E296. 125. Butcher JT, Nerem RM. Valvular endothelial cells regulate the phenotype
doi:10.3390/nano8050296 of interstitial cells in co-culture: effects of steady shear stress. Tissue Eng.

Brief Review - AL
106. Blaser MC, Aikawa E. Roles and regulation of extracellular vesicles in car- 2006;12:905–915. doi: 10.1089/ten.2006.12.905
diovascular mineral metabolism. Front Cardiovasc Med. 2018;5:187. doi: 126. Gwanmesia P, Ziegler H, Eurich R, Barth M, Kamiya H, Karck M,
10.3389/fcvm.2018.00187 Lichtenberg A, Akhyari P. Opposite effects of transforming growth factor-
107. Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: cur- β1 and vascular endothelial growth factor on the degeneration of aortic
rent perspectives. Proteomics. 2008;8:4083–4099. doi: 10.1002/pmic. valvular interstitial cell are modified by the extracellular matrix protein
200800109 fibronectin: implications for heart valve engineering. Tissue Eng Part A.
108. Camussi G, Deregibus MC, Cantaluppi V. Role of stem-cell-derived mi- 2010;16:3737–3746. doi: 10.1089/ten.TEA.2010.0304
crovesicles in the paracrine action of stem cells. Biochem Soc Trans. 127. Renato M, Bertacco E, Franchin C, Arrigoni G, Rattazzi M. Proteomic analy-
2013;41:283–287. doi: 10.1042/BST20120192 sis of interstitial aortic valve cells acquiring a pro-calcific profile. Methods
109. Zhang J, Li S, Li L, Li M, Guo C, Yao J, Mi S. Exosome and exosomal Mol Biol. 2013;1005:95–107. doi: 10.1007/978-1-62703-386-2_8
microRNA: trafficking, sorting, and function. Genomics Proteomics 128. Yu Z, Seya K, Daitoku K, Motomura S, Fukuda I, Furukawa K. Tumor ne-
Bioinformatics. 2015;13:17–24. doi:10.1016/j.gpb.2015.02.001 crosis factor-α accelerates the calcification of human aortic valve inter-
110. Kogure T, Yan IK, Lin WL, Patel T. Extracellular vesicle-mediated transfer of stitial cells obtained from patients with calcific aortic valve stenosis via
a novel long noncoding RNA TUC339: a mechanism of intercellular signal- the BMP2-Dlx5 pathway. J Pharmacol Exp Ther. 2011;337:16–23. doi:
ing in human hepatocellular cancer. Genes Cancer. 2013;4:261–272. doi: 10.1124/jpet.110.177915
10.1177/1947601913499020 129. Jian B, Jones PL, Li Q, Mohler ER 3rd, Schoen FJ, Levy RJ. Matrix metal-
111. Jansen F, Yang X, Baumann K, Przybilla D, Schmitz T, Flender A, Paul K, loproteinase-2 is associated with tenascin-C in calcific aortic stenosis. Am
Alhusseiny A, Nickenig G, Werner N. Endothelial microparticles reduce J Pathol. 2001;159:321–327. doi: 10.1016/S0002-9440(10)61698-7
ICAM-1 expression in a microRNA-222-dependent mechanism. J Cell Mol 130. Wang B, Li F, Zhang C, Wei G, Liao P, Dong N. High-mobility group box-
Med. 2015;19:2202–2214. doi: 10.1111/jcmm.12607 1 protein induces osteogenic phenotype changes in aortic valve intersti-
112. Jansen F, Stumpf T, Proebsting S, Franklin BS, Wenzel D, Pfeifer P, tial cells. J Thorac Cardiovasc Surg. 2016;151:255–262. doi: 10.1016/j.
Flender A, Schmitz T, Yang X, Fleischmann BK, et al. Intercellular trans- jtcvs.2015.09.077
fer of miR-126-3p by endothelial microparticles reduces vascular smooth 131. Moraes C, Likhitpanichkul M, Lam CJ, Beca BM, Sun Y, Simmons CA.
muscle cell proliferation and limits neointima formation by inhibiting LRP6. Microdevice array-based identification of distinct mechanobiological re-
J Mol Cell Cardiol. 2017;104:43–52. doi: 10.1016/j.yjmcc.2016.12.005 sponse profiles in layer-specific valve interstitial cells. Integr Biol (Camb).
113. Ni WJ, Wu YZ, Ma DH, Leng XM. Noncoding RNAs in calcific aortic valve 2013;5:673–680. doi: 10.1039/c3ib20254b
disease. J Cardiovasc Pharmacol. 2018;77:317–323. doi:10.1097/FJC. 132. van der Ven CF, Wu PJ, Tibbitt MW, van Mil A, Sluijter JP, Langer R, Aikawa E.
0000000000000569 In vitro 3D model and miRNA drug delivery to target calcific aortic valve
114. Coffey S, Jones GT. MicroRNAs are central to osteogenesis: a review disease. Clin Sci (Lond). 2017;131:181–195. doi: 10.1042/CS20160378
with a focus on cardiovascular calcification. microRNA Diagnostics and 133. Bond WS, Roberts EL, Warnock JN. Evaluation of porcine aortic valve inter-
Therapeutics. 2015;1:113–125. doi:10.2478/micrnat-2014-0005 stitial cell activity using different serum types in two- and three-dimensional
115. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, culture. Tissue Eng. 2007;13:343–349. doi: 10.1089/ten.2006.0166
Devon K, Dewar K, Doyle M, FitzHugh W, et al; International Human Genome 134. Dreger SA, Thomas P, Sachlos E, Chester AH, Czernuszka JT, Taylor PM,
Sequencing Consortium. Initial sequencing and analysis of the human ge- Yacoub MH. Potential for synthesis and degradation of extracellular matrix
nome. Nature. 2001;409:860–921. doi: 10.1038/35057062 proteins by valve interstitial cells seeded onto collagen scaffolds. Tissue
Eng. 2006;12:2533–2540. doi: 10.1089/ten.2006.12.2533
Downloaded from http://ahajournals.org by on April 8, 2022

116. Fu XD. Non-coding RNA: a new frontier in regulatory biology. Natl Sci Rev.
2014;1:190–204. doi: 10.1093/nsr/nwu008 135. Hjortnaes J, Goettsch C, Hutcheson JD, Camci-Unal G, Lax L, Scherer K,
117. Niepmann ST, Steffen E, Zietzer A, Adam M, Nordsiek J, Gyamfi-Poku I, Body S, Schoen FJ, Kluin J, Khademhosseini A, et al. Simulation of early cal-
Piayda K, Sinning JM, Baldus S, Kelm M, et al. Graded murine wire-in- cific aortic valve disease in a 3D platform: a role for myofibroblast differentia-
duced aortic valve stenosis model mimics human functional and morpho- tion. J Mol Cell Cardiol. 2016;94:13–20. doi: 10.1016/j.yjmcc.2016.03.004
logical disease phenotype. Clin Res Cardiol. 2019;108:847–856. doi: 136. Yip CY, Blaser MC, Mirzaei Z, Zhong X, Simmons CA. Inhibition of
10.1007/s00392-019-01413-1 pathological differentiation of valvular interstitial cells by C-type natri-
118. Yanagawa B, Lovren F, Pan Y, Garg V, Quan A, Tang G, Singh KK, uretic peptide. Arterioscler Thromb Vasc Biol. 2011;31:1881–1889. doi:
Shukla PC, Kalra NP, Peterson MD, et al. miRNA-141 is a novel regulator 10.1161/ATVBAHA.111.223974
of BMP-2-mediated calcification in aortic stenosis. J Thorac Cardiovasc 137. Miller JD, Weiss RM, Heistad DD. Calcific aortic valve stenosis: meth-
Surg. 2012;144:256–262. doi: 10.1016/j.jtcvs.2011.10.097 ods, models, and mechanisms. Circ Res. 2011;108:1392–1412. doi:
119. Zhang M, Liu X, Zhang X, Song Z, Han L, He Y, Xu Z. MicroRNA-30b 10.1161/CIRCRESAHA.110.234138
is a multifunctional regulator of aortic valve interstitial cells. J Thorac 138. Sider KL, Blaser MC, Simmons CA. Animal models of calcific aortic valve
Cardiovasc Surg. 2014;147:1073–1080.e2. doi: 10.1016/j.jtcvs. disease. Int J Inflam. 2011;2011:364310. doi: 10.4061/2011/364310
2013.05.011 139. Honda S, Miyamoto T, Watanabe T, Narumi T, Kadowaki S, Honda Y,
120. Wang Y, Chen S, Deng C, Li F, Wang Y, Hu X, Shi F, Dong N. MicroRNA-204 Otaki Y, Hasegawa H, Netsu S, Funayama A, et al. A novel mouse model of
targets runx2 to attenuate BMP-2-induced osteoblast differentiation of aortic valve stenosis induced by direct wire injury. Arterioscler Thromb Vasc
human aortic valve interstitial cells. J Cardiovasc Pharmacol. 2015;66:63– Biol. 2014;34:270–278. doi: 10.1161/ATVBAHA.113.302610
71. doi: 10.1097/FJC.0000000000000244 140. Hinton RB Jr, Alfieri CM, Witt SA, Glascock BJ, Khoury PR, Benson DW,
121. Matouk I, Raveh E, Ohana P, Lail RA, Gershtain E, Gilon M, De Groot N, Yutzey KE. Mouse heart valve structure and function: echocardiographic and
Czerniak A, Hochberg A. The increasing complexity of the oncofetal h19 morphometric analyses from the fetus through the aged adult. Am J Physiol
gene locus: functional dissection and therapeutic intervention. Int J Mol Sci. Heart Circ Physiol. 2008;294:H2480–H2488. doi: 10.1152/ajpheart.
2013;14:4298–4316. doi: 10.3390/ijms14024298 91431.2007
122. Hadji F, Boulanger MC, Guay SP, Gaudreault N, Amellah S, 141. Quast C, Zimmer S, Boenner F, Jacoby C, Gyamfi-Poku I, Piayda K, Erkens
Mkannez G, Bouchareb R, Marchand JT, Nsaibia MJ, Guauque-Olarte S, R, Niepmann ST, Adam M, Baldus S, et al. P5993: comprehensive char-
et al. Altered DNA methylation of long noncoding RNA H19 in calcific acterization of experimental aortic valve stenosis by multiparametric MRI.
aortic valve disease promotes mineralization by silencing NOTCH1. Eur Heart J. 2019;40, Issue Supplement_1:ehz746.0714. doi: 10.1093/
Circulation. 2016;134:1848–1862. doi: 10.1161/CIRCULATIONAHA. eurheartj/ehz746.0714
116.023116 142. Garcia J, Barker AJ, Markl M. The role of imaging of flow patterns by 4D
123. Xiao X, Zhou T, Guo S, Guo C, Zhang Q, Dong N, Wang Y. LncRNA flow MRI in aortic stenosis. JACC Cardiovasc Imaging. 2019;12:252–266.
MALAT1 sponges miR-204 to promote osteoblast differentiation of hu- doi: 10.1016/j.jcmg.2018.10.034
man aortic valve interstitial cells through up-regulating Smad4. Int J Cardiol. 143. Stewart BF, Siscovick D, Lind BK, Gardin JM, Gottdiener JS, Smith VE,
2017;243:404–412. doi: 10.1016/j.ijcard.2017.05.037 Kitzman DW, Otto CM. Clinical factors associated with calcific aortic valve
124. Yu C, Li L, Xie F, Guo S, Liu F, Dong N, Wang Y. LncRNA TUG1 sponges disease. Cardiovascular Health Study. J Am Coll Cardiol. 1997;29:630–
miR-204-5p to promote osteoblast differentiation through upregulating 634. doi: 10.1016/s0735-1097(96)00563-3
Runx2 in aortic valve calcification. Cardiovasc Res. 2018;114:168–179. 144. Aggarwal SR, Clavel MA, Messika-Zeitoun D, Cueff C, Malouf J, Araoz PA,
doi: 10.1093/cvr/cvx180 Mankad R, Michelena H, Vahanian A, Enriquez-Sarano M. Sex differences

Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067 April 2020   899
Goody et al Aortic Valve Stenosis

in aortic valve calcification measured by multidetector computed tomog- 155. Zheng KH, Tsimikas S, Pawade T, Kroon J, Jenkins WSA, Doris MK,
raphy in aortic stenosis. Circ Cardiovasc Imaging. 2013;6:40–47. doi: White AC, Timmers NKLM, Hjortnaes J, Rogers MA, et al. Lipoprotein(a)
Brief Review - AL

10.1161/CIRCIMAGING.112.980052 and oxidized phospholipids promote valve calcification in patients with


145. Simard L, Côté N, Dagenais F, Mathieu P, Couture C, Trahan S, Bossé Y, aortic stenosis. J Am Coll Cardiol. 2019;73:2150–2162. doi: 10.1016/j.
Mohammadi S, Pagé S, Joubert P, et al. Sex-related discordance between jacc.2019.01.070
aortic valve calcification and hemodynamic severity of aortic steno- 156. Cowell SJ, Newby DE, Prescott RJ, Bloomfield P, Reid J, Northridge DB,
sis: is valvular fibrosis the explanation? Circ Res. 2017;120:681–691. Boon NA; Scottish Aortic Stenosis and Lipid Lowering Trial, Impact on
doi:10.1161/CIRCRESAHA.116.309306 Regression (SALTIRE) Investigators. A randomized trial of intensive lipid-
146. McCoy CM, Nicholas DQ, Masters KS. Sex-related differences in gene
lowering therapy in calcific aortic stenosis. N Engl J Med. 2005;352:2389–
expression by porcine aortic valvular interstitial cells. PLoS One. 2012;7:
2397. doi: 10.1056/NEJMoa043876
e39980. doi: 10.1371/journal.pone.0039980
157. Teo KK, Corsi DJ, Tam JW, Dumesnil JG, Chan KL. Lipid lowering on pro-
147. Vanderschueren D, Vandenput L, Boonen S, Lindberg MK, Bouillon R,
gression of mild to moderate aortic stenosis: meta-analysis of the ran-
Ohlsson C. Androgens and bone. Endocr Rev. 2004;25:389–425. doi:
10.1210/er.2003-0003 domized placebo-controlled clinical trials on 2344 patients. Can J Cardiol.
148. McRobb L, Handelsman DJ, Heather AK. Androgen-induced progression 2011;27:800–808. doi: 10.1016/j.cjca.2011.03.012
of arterial calcification in apolipoprotein E-null mice is uncoupled from 158. Rossebø AB, Pedersen TR, Boman K, Brudi P, Chambers JB,
plaque growth and lipid levels. Endocrinology. 2009;150:841–848. doi: Egstrup K, Gerdts E, Gohlke-Bärwolf C, Holme I, Kesäniemi YA, et al;
10.1210/en.2008-0760 SEAS Investigators. Intensive lipid lowering with simvastatin and ezeti-
149. Zhu D, Hadoke PWF, Wu J, Vesey AT, Lerman DA, Dweck MR, Newby DE, mibe in aortic stenosis. N Engl J Med. 2008;359:1343–1356. doi:
Smith LB, MacRae VE. Ablation of the androgen receptor from vascular 10.1056/NEJMoa0804602
smooth muscle cells demonstrates a role for testosterone in vascular cal- 159. Thanassoulis G. Lipoprotein (a) in calcific aortic valve disease: from ge-
cification. Sci Rep. 2016;6:24807. doi:10.1038/srep24807 nomics to novel drug target for aortic stenosis. J Lipid Res. 2016;57:917–
150. Price PA, Faus SA, Williamson MK. Bisphosphonates alendronate and 924. doi: 10.1194/jlr.R051870
ibandronate inhibit artery calcification at doses comparable to those that 160. Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, Yu RZ,
inhibit bone resorption. Arterioscler Thromb Vasc Biol. 2001;21:817–824. Marcovina SM, Hughes SG, Graham MJ, Crooke RM, et al. Antisense oligo-
doi:10.1161/01.ATV.21.5.817 nucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a):
151. Sansoni P, Passeri G, Fagnoni F, Mohagheghpour N, Snelli G, Brianti V, two randomised, double-blind, placebo-controlled, dose-ranging trials.
Engleman EG. Inhibition of antigen-presenting cell function by alendro- Lancet. 2016;388:2239–2253. doi: 10.1016/S0140-6736(16)31009-1
nate in vitro. J Bone Miner Res. 1995;10:1719–1725. doi: 10.1002/jbmr.
161. Choi B, Lee S, Kim SM, Lee EJ, Lee SR, Kim DH, Jang JY,
5650101115
Kang SW, Lee KU, Chang EJ, et al. Dipeptidyl peptidase-4 induces
152. Thanassoulis G, Campbell CY, Owens DS, Smith JG, Smith AV,
aortic valve calcification by inhibiting insulin-like growth factor-1 signal-
Peloso GM, Kerr KF, Pechlivanis S, Budoff MJ, Harris TB, et al; CHARGE
ing in valvular interstitial cells. Circulation. 2017;135:1935–1950. doi:
Extracoronary Calcium Working Group. Genetic associations with valvular
10.1161/CIRCULATIONAHA.116.024270
calcification and aortic stenosis. N Engl J Med. 2013;368:503–512. doi:
10.1056/NEJMoa1109034 162. Degboé Y, Rauwel B, Baron M, Boyer JF, Ruyssen-Witrand A, Constantin
153. Clarke R, Peden JF, Hopewell JC, Kyriakou T, Goel A, Heath SC, Parish S, A, Davignon JL. Polarization of rheumatoid macrophages by TNF target-
Barlera S, Franzosi MG, Rust S, et al; PROCARDIS Consortium. Genetic ing through an IL-10/STAT3 mechanism. Front Immunol. 2019;10:3. doi:
variants associated with Lp(a) lipoprotein level and coronary disease. N 10.3389/fimmu.2019.00003
Downloaded from http://ahajournals.org by on April 8, 2022

Engl J Med. 2009;361:2518–2528. doi: 10.1056/NEJMoa0902604 163. Ceneri N, Zhao L, Young BD, Healy A, Coskun S, Vasavada H, Yarovinsky TO,
154. Orsó E, Schmitz G. Lipoprotein(a) and its role in inflammation, atheroscle- Ike K, Pardi R, Qin L, et al. Rac2 modulates atherosclerotic calcification by
rosis and malignancies. Clin Res Cardiol Suppl. 2017;12(suppl 1):31–37. regulating macrophage interleukin-1β production. Arterioscler Thromb Vasc
doi:10.1007/s11789-017-0084-1 Biol. 2017;37:328–340. doi: 10.1161/ATVBAHA.116.308507

900   April 2020 Arterioscler Thromb Vasc Biol. 2020;40:885–900. DOI: 10.1161/ATVBAHA.119.313067

You might also like