ArticlePDF Available

The pathogenesis of aortic stenosis: not just a matter of wear and tear

Authors:

Abstract and Figures

Aortic valve stenosis (AS) is the commonest form of valvular heart disease in the Western world. Its prevalence increases exponentially with age and it is present in 2-7% of all patients over 65 years of age. In view of the considerable cardiovascular morbidity and mortality associated not only with AS, but even its earlier stage, aortic sclerosis, many investigations have been directed towards better understanding of its pathogenesis, with the ultimate objective of developing strategies to retard its progression. Although risk factors and downstream mediators appear similar for AS and atherosclerosis (older age, male sex, hypertension, smoking, hypercholesterolemia, and diabetes, as many as 50% of patients with AS do not have clinically significant atherosclerosis. On the basis both of recent experimental evidence and clinical trials, it appears that atherogenesis is not pivotal to the pathogenesis of AS. On the other hand, there is increasing evidence of active involvement of aortic valve fibroblasts with resultant increased production of reactive oxygen species, active pro-inflammatory and pro-fibrotic processes culminating in calcification. We also discuss the evidence of involvement of the nitric oxide system in the pathogenesis of AS. The renin-angiotensin system has also emerged as a major player in the pathogenesis of AS. Histologically, there is increased ACE expression and elevated angiotensin II levels in stenotic valves, while we have just demonstrated amelioration of AS with the use of ACE inhibitors in an animal model. We further discuss intervention studies aimed at retarding AS progression, including recent failures of statins to retard progression of AS in large randomized clinical studies. Finally, we discuss the special case of bicuspid aortic valve, including its genetics and unique associated features.
Content may be subject to copyright.
Introduction
Aortic stenosis (AS) may be defined as narrow-
ing of the aortic valve, due primarily to a combi-
nation of progressive fibrosis and calcification
of the matrix, with consequent increase in valve
stiffness, progressive reductions in valve area
and concomitant increases in left ventricular
afterload and work. The earliest stages of AS
have been designated aortic valve sclerosis
(ASc), implying disordered valve morphology
(including potential calcification as well as fibro-
sis) in the absence of marked obstruction to left
ventricular outflow.
AS is currently the most common form of valvu-
lar heart disease in the Western world [1], in
large part because the most frequently occur-
ring form of AS develops predominantly in indi-
viduals of advancing age. For example, in the
Helsinki Ageing Study [2], the proportion of indi-
viduals with detectable valve calcification in-
creased from approximately 40% to 75% be-
tween ages of 65 and 85 years, while approxi-
mately 3% of subjects over 75 years of age had
severe AS.
Given that the only proven therapy for severe AS
is aortic valve replacement, and that there is
currently no definitive evidence that any treat-
ment can retard progression of the disease, the
development of severe symptomatic AS in eld-
erly individuals presents an increasing medical
and health economic dilemma. On the one
hand, severe AS rarely remains asymptomatic
for any great length of time: patients classically
develop variable components of exertional dysp-
noea, angina (irrespective of the presence or
Am J Cardiovasc Dis 2011;1(2):185-199
www.AJCD.us /ISSN: 2160-200X/AJCD1107002
Review Article
Pathogenesis of aortic stenosis: not just a matter of wear
and tear
Aaron L Sverdlov, Doan TM Ngo, Matthew J Chapman, Onn Akbar Ali, Yuliy Y Chirkov, John D Horowitz
The Queen Elizabeth Hospital, University of Adelaide, South Australia, Australia
Received July 3, 2011; accepted July 20, 2011; Epub July 28, 2011; published August 15, 2011
Abstract: Aortic valve stenosis (AS) is the commonest form of valvular heart disease in the Western world. Its preva-
lence increases exponentially with age and it is present in 2-7% of all patients over 65 years of age. In view of the
considerable cardiovascular morbidity and mortality associated not only with AS, but even its earlier stage, aortic
sclerosis, many investigations have been directed towards better understanding of its pathogenesis, with the ultimate
objective of developing strategies to retard its progression. Although risk factors and downstream mediators appear
similar for AS and atherosclerosis (older age, male sex, hypertension, smoking, hypercholesterolemia, and diabetes,
as many as 50% of patients with AS do not have clinically significant atherosclerosis. On the basis both of recent ex-
perimental evidence and clinical trials, it appears that atherogenesis is not pivotal to the pathogenesis of AS. On the
other hand, there is increasing evidence of active involvement of aortic valve fibroblasts with resultant increased
production of reactive oxygen species, active pro-inflammatory and pro-fibrotic processes culminating in calcification.
We also discuss the evidence of involvement of the nitric oxide system in the pathogenesis of AS. The renin-
angiotensin system has also emerged as a major player in the pathogenesis of AS. Histologically, there is increased
ACE expression and elevated angiotensin II levels in stenotic valves, while we have just demonstrated amelioration of
AS with the use of ACE inhibitors in an animal model. We further discuss intervention studies aimed at retarding AS
progression, including recent failures of statins to retard progression of AS in large randomized clinical studies. Fi-
nally, we discuss the special case of bicuspid aortic valve, including its genetics and unique associated features.
Keywords: Aortic valve stenosis, bicuspid aortic valve, nitric oxide, oxidative stress, inflammation, fibrosis, calcifica-
tion, intervention studies
Pathogenesis of aortic stenosis
186 Am J Cardiovasc Dis 2011;1(2):185-199
absence of epicardial coronary artery disease)
and atrial or ventricular arrhythmias, resulting in
poor quality of life, increased rates of hospitali-
zation and increased mortality rates [3]. Al-
though the precise natural history of severe AS
in the elderly is uncertain, a number of studies
suggested that mortality is high in the some-
what selected subgroup of individuals who do
not undergo aortic valve replacement [4-7]. Ad-
ditionally, both ASc and AS may be associated
with cardiovascular problems which are appar-
ently remote from the valve itself. For example,
there is considerable evidence that ASc is an
independent marker of increased risk of cardio-
vascular events [8, 9], while severe AS is associ-
ated with increased risk of haemorrhage, espe-
cially into the gastrointestinal tract, primarily
because of the development of acquired von
Willebrand's factor deficiency [10, 11].
It should also be stated at the outset that AS is
a substantially heterogeneous disease, but with
only two "common causes". These are AS devel-
oping in a previously normal trileaflet valve, and
AS associated with congenitally bicuspid aortic
valves (BAV). These and other, rarer, causes of
AS are summarized in Table 1.
Given that AS frequently develops in otherwise
normal valves in aged individuals, this has been
regarded as degenerative AS, implying a rela-
tionship to the normal process of "wear and
tear" within the valve [12]. The purpose of this
review is to demonstrate that AS is not primarily
a "degenerative" process, but rather the result
of a progressive inflammatory process within
the valve matrix. Furthermore, we will present
evidence that the development and progression
of AS, rather than occurring with the implied
inevitability of a purely degenerative disease,
should theoretically be amenable to pharmaco-
therapy.
Aortic valve anatomy and physiology: what are
the homeostatic determinants?
The normal aortic valve consists of several lay-
ers of fibroblast-rich tissue, containing both col-
lagen and elastin fibres, covered by a
monolayer of endothelial cells [13]. There are
also minute intravalvular blood vessels [14],
consistent with a substantial oxygen demand by
the other matrix cells.
The normal aortic valve interstitial cells are
probably mainly fibroblasts, but some smooth
muscle cells have been identified [15], raising
the issue of potential variability in tone of the
valve, an area which has been pursued in a
number of experimental studies. For example,
Pompilio et al [16] showed that intact porcine
aortic valve contracted in response to
phenylephrine, and also exhibited (endothelium-
dependent) relaxation with acetylcholine. Fur-
thermore, normal aortic valve interstitial cells
(presumably including smooth muscle cells)
have been shown to contract in response to 5-
hydroxytryptamine, endothelin-1 and norepi-
nephrine [17]. An important, but incompletely
resolved issue relates to the prevalence and
function of myofibroblasts, which exhibit some
contractile properties, within normal aortic
valves. The role of the myofibroblasts will be
discussed more extensively in the context of
valve pathology.
The physiological role of the aortic valve endo-
thelium has attracted considerable attention
over the past 15 years, although its role is still
incompletely understood. A number of studies
suggest that valve endothelium behaves in a
qualitatively similar manner to vascular endo-
thelium, for example releasing nitric oxide (NO)
in response to stimuli such as acetylcholine
[16], or 5-hydroxytryptamine [18].
Table 1. Causes of AS. Wide frequency range generally reflects the age group(s) assessed by individual
studies as well as population subgroups studied [79, 115-117].
Etiology Approximate frequency (%) Associated features
Aging/calcific 50 - 70 Increased risk of coronary events.
Bicuspid aortic valve 6 - 40 Dilatation or dissection of the aorta, involving the aortic root,
ascending aorta, or aortic arch.
Rheumatic 2 - 11 Mitral valve almost always affected as well
Unicuspid aortic valve <1 Dilatation or dissection of the aorta, involving the aortic root,
ascending aorta, or aortic arch.
Post-endocarditis <1 Extra-cardiac embolic phenomena.
Pathogenesis of aortic stenosis
187 Am J Cardiovasc Dis 2011;1(2):185-199
A critical but unresolved issue is what is the
physiological response of the valve endothelium
to shear stress, a normal stimulus for NO re-
lease, but also potentially for activation of endo-
thelial NAD(P)H oxidase [19, 20], which in turn
would lead to release of superoxide anion (O2-)
and "scavenging" of NO. Despite ongoing uncer-
tainties, the differences between normal aortic
valve and vascular endothelial cells regarding
responses to shear stress were reviewed by
Butcher et al [21], who demonstrated that the
changes in gene expression in response to
shear stress varied substantially.
Finally, little work has been done regarding
other endothelial autocoids within the valve.
Notably, little is known about the physiology of
prostacyclin release from the valve, although it
has been detected [22]. No studies have prop-
erly addressed the issue of tissue penetration of
autocoids released from valvular endothelium,
other than the obvious implications of studies
measuring valve contraction. It is therefore un-
certain to what extent endothelial NO and
prostacyclin might modulate the function of
valve interstitial cells, a physiologically impor-
tant issue given evidence that in AS valve endo-
thelium is dysfunctional or absent, as discussed
below.
Cellular histopathology of AS
Features of stenotic aortic valve lesions
Histopathologic studies have demonstrated that
development and progression of calcific AS are
based on an active process that shares some
similarities with atherosclerosis. It has been
suggested that aortic valve lesions begin with
disruption of valve endothelium predominantly
on the aortic side due to high shear stress [23-
25].
Inflammation and lipid deposition
Aortic valve lesions typically present with areas
of subendothelial thickening, which represents
the early stage of aortic stenosis. Increased
thickening of aortic valve leaflets is character-
ized by accumulation of inflammatory infiltrates
of predominantly macrophages and T-
lymphocytes, lipids, oxidized lipids, (summarized
in Figure 1) [25-28] all of which potentially acti-
vate a host of pro-fibrotic and pro-inflammatory
markers. Macrophages and T-lymphocytes have
been detected and tend to be located near the
surface of the lesion [25-28]. Immunohisto-
chemical studies have found co-localization of
apolipoproteins (apo) B, apo (a), apoE with lipid
laden foam cells and macrophages [29] as well
as oxidative modification of residential low den-
sity lipoproteins (LDLs) in early stenotic aortic
valve lesions [30].
Valvular matrix remodelling and fibrosis
The presence of macrophages and T-
lymphocytes, along with oxidized LDL and apoli-
poprotein accumulation activate several pro-
fibrotic and pro-inflammatory cytokines which
may modulate aortic valve remodelling and sub-
sequent calcification. Transforming growth fac-
tor β1 (TGF-β1) [31] and interleukin-1β [32]
have been found in valve matrix and are associ-
ated with increased local production of matrix
metalloproteinases I and II (MMP-1 and MMP-
2). All of these contribute to cell apoptosis, ex-
tracellular matrix formation, remodelling and
consequently predispose to calcification. In ad-
dition to TGF-β1 and interleukin-1β, tumour ne-
crosis factor-α (TNFα), another pro-inflammatory
cytokine commonly responsible for immune
regulation, inflammation and tissue remodel-
ling, is co-localized with MMP-1 [33]. Further-
more, tenascin C, an extracellular matrix glyco-
protein implicated in cell proliferation, migra-
tion, differentiation and apoptosis, which is in-
volved in stimulation of bone formation and
mineralization, is co-localized with MMP-2 in
calcified aortic valve leaflets [34], and is associ-
ated with progression of AS [35].
Angiotensin II (Ang II), an important mediator of
inflammation and fibrosis, could be formed by
angiotensin converting enzyme (ACE) as well as
the mast cell (MC)-derived neutral protease,
chymase [36]. ACE has been identified in
stenotic but not in normal aortic valves [37]. It
has been shown that MC-derived chymase is
also upregulated in stenotic valves, providing
further evidence for local production of Ang II
[38]. In addition, cathepsin G, another neutral
protease also capable of generating Ang II, is
present in increased concentrations throughout
human stenotic aortic valves compared to nor-
mal valves [39]. These findings provide a poten-
tial basis for a role of angiotensin II in aortic
valve remodelling along with other pro-fibrotic
and pro-inflammatory mechanisms.
As summarized in Figure 1, current concepts of
the pathogenesis of AS centre histologically on
Pathogenesis of aortic stenosis
188 Am J Cardiovasc Dis 2011;1(2):185-199
inflammation and lipid deposition, and bio-
chemically on activation of cytokines and matrix
metalloproteinases, together with generation of
angiotensin II. These processes are postulated
to induce injury of all valve components, leading
to fibrosis and calcification.
Calcification
Calcification of aortic valve leaflets tends to
occur more predominantly in the later stages of
AS, and is located deeper in the lesion [25].
Active calcification is a major factor in reducing
valvular mobility in severe AS. Early lesions of
aortic valves show fine stippled mineralization,
progressing to active bone formation resulting
in gross calcification at later stages of the dis-
ease (Figure 1).
The process of calcification (and sometimes
Figure 1. Schema of postulated mechanisms underlying aortic valve lesion formation. Inflammatory infiltrations of T-
lymphocytes and macrophages, along with lipid accumulation, is primarily responsible for early thickening of aortic
valves. Interactions between chemical stimuli and disruption of valvular homeostasis: pro- and anti- fibrotic mecha-
nisms. Later stages of aortic stenosis: - cytokine release and angiotensin II promote extracellular matrix proteins se-
cretion at early stages of mineralization which in turns begin the processes of bone formation. This process occurs
largely at the end stage of aortic stenosis where aortic valves mobility is significantly reduced due to a build up of
bone-like calcific nodules.
Pathogenesis of aortic stenosis
189 Am J Cardiovasc Dis 2011;1(2):185-199
ossification) of aortic valve leaflets resembles
that associated with atheroma formation. The
presence of inflammation, fatty streak forma-
tion from lipid depositions, cytokine release,
metalloproteinases, ACE, Ang II, all possibly con-
tribute to the production of an extracellular ma-
trix, and matrix vesicles that initiate mineraliza-
tion.
Co-localization of macrophages and oxidized
LDLs with osteopontin, a protein needed in
bone formation, has been found and thought to
be involved in extracellular matrix production in
human stenotic aortic valves [40, 41]. Further-
more, Mohler et al [42] described an active
process of calcification using immunohisto-
chemistry in an extensive study of 347 human
stenotic aortic valves. In addition to active os-
teoblasts and osteoclasts, this and other inves-
tigators [43] detected bone morphogenic pro-
teins 2- and 4- (BMP-2, BMP-4). BMPs stimulate
osteoblastic differentiation with subsequent
calcification. In agreement with previous studies
[25, 41], Mohler et al [42] also found that
macrophages and lymphocytes accumulate in
areas of calcification.
There is also evidence of angiogenesis, which is
essential for longitudinal bone growth in
stenotic valves. T-lymphocytes aggregates tend
to co-localize with sites of neoangiogenesis
within ossified valves [42]. This and the pres-
ence of heat shock protein 60 (hsp60) [44],
commonly expressed by cells under stress con-
ditions, together indicate a highly active, and
chronic immunomediated process from stress
to inflammation to calcification.
It has also been shown that the calcification
process of aortic valves may also be regulated
by receptor activator of nuclear factor κB, its
ligand (RANK, and RANKL), and the soluble re-
ceptor osteoprotegerin (OPG) [45]. The study
detected increased concentrations of RANKL in
calcified compared to control valves. Further-
more, there was a significant reduction of OPG
positive cells in aortic stenotic valves compared
to controls. It has been shown that in mice defi-
cient for OPG, vascular calcification was associ-
ated with increased expression of RANKL [46].
Additionally, in Kaden et al [45] shown that long
-term cell culture of stenotic aortic valves in the
presence of RANKL induced significant increase
in matrix calcium deposition and the formation
of cell nodules compared to controls. Thus, the
RANKL/OPG pathway may also be involved in
the calcific process of aortic valve stenosis.
A recent study [47] also revealed an association
between presence of aortic stenosis and low
serum levels of the anti-calcific protein fetuin-A.
Furthermore, there was evidence that fetuin-A
was deposited in calcific aortic valves. Never-
theless, the relative importance of these find-
ings to the overall pathogenesis of aortic steno-
sis remains uncertain.
Matrix Gla-protein (MGP), another anti-calcific
protein implicated in development of AS, is syn-
thesised in different tissues and undergoes vita-
min K dependent γ-carboxylation (reviewed by
[48]. The γ-carboxylated form of MGP is involved
in inhibition of tissue calcification possibly by
preventing differentiation of vascular smooth
muscle cells into chondrocyte-like cells or by
blocking BMP function [48]. Patients with aortic
valve calcification were found to have lower
concentrations of uncarboxylated MGP com-
pared with normal controls [49]; additionally
both renal dysfunction and oral warfarin therapy
were predictive of low MGP levels. Interestingly,
another study found elevated levels of both un-
carboxylated and carboxylated de-phosporylated
forms of MGP in patients with severe AS [50].
The exact biological significance of these find-
ings is still unclear.
Oxidative stress
A number of studies have documented in-
creased intravalvular content of a variety of pro-
oxidants in models of AS and in clinical samples
[23, 51-54]. Importantly, this evidence of in-
creased oxidative stress was not specifically
associated with extent of local atherogenesis in
any study.
Thioredoxin, thioredoxin reductase, NADPH oxi-
dase and thioredoxin binding protein (TXNIP)
are components of a ubiquitous system, which
regulates intracellular redox stress [55]. TXNIP
in particular is a fundamental mediator of in-
creased redox stress as it binds to, and inacti-
vates thioredoxin [56]. Increased expression of
TXNIP is associated with activation of apoptosis
signalling pathways [57], and is suppressed by
endothelial NO release [58]. We have recently
demonstrated in a rabbit model of mild AS with
histological features similar to that of human
disease, increased intravalvular concentration
Pathogenesis of aortic stenosis
190 Am J Cardiovasc Dis 2011;1(2):185-199
of TXNIP compared to control animals [53]. Fur-
thermore, co-treatment with ramipril retarded
the development of AS in this model, as meas-
ured by reduction both in transvalvular velocity
and valve echogenicity on echocardiography
[59]. This retardation further correlated with
reduction in valvular calcium and macrophage
infiltration and a reduction in TXNIP accumula-
tion within the valve matrix. This finding further
underscores the importance of ACE - Ang II sys-
tem in pathogenesis of AS.
Evidence of the role of nitric oxide in the patho-
genesis of aortic stenosis
AS, even in its early phases is associated with
the pathogenesis of acute coronary syndromes
(ACS) [8, 60, 61], which are paralleled by plate-
let hyper-aggregability [62]. Stenotic aortic
valves constitute a pro-aggregatory milieu, po-
tentially contributing to thromboembolism [24,
63]. It has been shown that patients with AS
exhibit increased platelet reactivity [64], and
thrombus formation has been documented on
severely stenosed valves [65]. AS has recently
been linked to the phenomenon of nitric oxide
(NO) resistance, even at its earliest stages [66,
67].
NO is a physiological modulator of both vasomo-
tor tone and platelet aggregation. These effects
of NO are predominantly mediated by cyclic
guanosine-3,'5'-monophosphate (cGMP), via
activation of soluble guanylate cyclase (sGC).
However, in patients with ischemic heart dis-
ease, platelets and coronary/peripheral arteries
respond poorly to the anti-aggregatory and vaso-
dilator effects of NO donors (e.g. nitroglycerin)
[68]. This “NO resistance” represents a multi-
faceted disorder at sites of abnormal NO-driven
physiology, and as such may contribute to the
increased risk of ischemic events. NO resis-
tance results from a combination of
“scavenging” of NO by superoxide radical (O2
)
and of inactivation of sGC. The haem moiety of
sGC is a “receptor” for NO and a mediator of NO
-dependent activation. However, reactive oxygen
species, and O2-in particular, diminish sGC sen-
sitivity to NO because of the oxidation of the
enzyme-bound haem and its subsequent loss
(haem-deficient sGC).
We have investigated [66] whether AS, either
with or without concomitant coronary artery dis-
ease, is associated with impaired NO respon-
siveness. Two major abnormalities of platelet
function were documented in patients with AS.
First, platelets manifested increased aggregabil-
ity in response to ADP. Second, the anti-
aggregating effects of NO donor SNP were sig-
nificantly reduced, thus representing NO resis-
tance at the platelet level. However, presence of
the former abnormality did not account for the
occurrence of the latter.
The severity of NO resistance was independent
of presence/absence of hemodynamically sig-
nificant coronary artery disease. These results
indicate therefore that AS represents an addi-
tional "marker" of platelet NO resistance. Inter-
estingly, there was no correlation between AVS
severity and the extent of platelet NO resis-
tance. This suggests that platelet abnormalities
and impairment of NO-related mechanisms may
appear early in the clinical course of AVS.
Indeed, in our recent cross-sectional population
study evaluating biochemical and physiological
correlates of the presence of aortic valve sclero-
sis (ASc), a precursor to AS, in aging individuals
[67] we have documented that ASc was also
associated with impaired platelet responsive-
ness to NO. In that study, the extent of ASc
manifestation was strongly associated with the
extent of platelet resistance to NO. This finding
is of potential relevance to the association be-
tween ASc and thrombotic events [8]. Further-
more, tissue resistance to NO per se might also
contribute to the calcification process [69].
Asymmetric dimethylarginine (ADMA) is an en-
dogenous competitive inhibitor of NO synthase
(eNOS) and a marker and mediator of endothe-
lial dysfunction [70-72]. We have also demon-
strated that plasma concentrations of ADMA are
elevated in patients with AS, compared with
controls [73]. Thus, this finding provides further
support to the suggestion that NO generation is
impaired in AS.
A critical question which arises is whether the
pathogenesis of AS is fundamentally related to
dysfunction of the valve endothelium. NO resis-
tance in AS may be paralleled by impairment of
aortic valve endothelial function. Previous stud-
ies [63, 74, 75] have demonstrated that even
Pathogenesis of aortic stenosis
191 Am J Cardiovasc Dis 2011;1(2):185-199
early aortic valve calcification is associated with
some decrease in endothelial function and even
loss of valvular endothelium. Narrowing of the
aortic valve orifice in AS, together with deforma-
tion of leaflets and increasingly rough surface of
the valve, contributes to local turbulence in
blood flow which creates shear stress, affecting
both valve endothelium and passing platelets
[76]. Furthermore, loss of aortic valve endothe-
lium may predispose towards calcification of the
aortic valve leaflets [69, 77, 78]. Potentially,
aortic valve endothelium plays a critical role in
maintaining normal aortic valve function.
In isolated porcine and canine aortic valves, it
was shown that the aortic valve endothelium
releases NO and prostacyclin, which both exert
local anti-thrombotic effects and reduce leaflet
tone [16, 18, 63]. The main "luminal" implica-
tion of these studies was that the stenotic aortic
valve constitutes a pro-aggregatory milieu: - the
clinical consequences of a loss of valve anti-
aggregatory function must be considered to-
gether with platelet hyperaggregability and
platelet resistance to NO occurring in AVS [66].
However, it is equally possible that disordered
valve endothelial function contributes to devel-
opment of valve matrix pathology. In our recent
studies [69] with porcine aortic valve interstitial
cell cultures, calcific nodule formation and asso-
ciated redox stress were inhibited by NO donors.
This suggests that NO has a direct protective
effect extending to the inhibition of the calcifica-
tion processes in aortic valve cells.
Bicuspid aortic valve (BAV): a special case?
BAV represents the most common congenital
cardiac abnormality, with estimates of its preva-
lence ranging from 0.5 to 2% of the general
population [79]. Structurally, BAV is heterogene-
ous, depending on the pattern of valve leaflet
fusion, and its association with extravalvular
disease, as summarized in Table 2. However,
the most common pattern, accounting for about
75% of BAV cases, is fusion of the right and left
(R-L: Type I) valve leaflets, while Type II BAV
(fusion of the right and non-coronary cusps)
accounts for virtually all other cases.
A fundamental and unique issue with BAV is the
association with dilatation of the aortic root and
arch, which is a particularly prominent feature
of Type I BAV, and which is associated with risk
of aortic dissection and rupture. On the other
hand, progression of AS appears to be more
rapid in Type II [79], where most patients re-
quire eventual aortic valve replacement.
From a pathophysiological point of view, BAV is
unique and intriguing because it presents as a
basis for a rapidly progressive form of AS and/or
aortopathy, and because of the possible insight
it provides regarding the pathogenesis of AS.
As regards the rapid progression, in a retrospec-
tive study of 156 adult patients with BAV, Tha-
nassoulis et al [80] have demonstrated a mean
increase in diameter of ascending aorta of
0.37mm/year and of aortic sinus of Valsava of
0.17mm/year. Tzemos et al [81] documented
increase in aortic valve gradient of 0.7 mmHg
per annum in a prospective cohort of 642 sub-
jects with BAV.
As regards available data on the pathogenesis
of BAV, despite a very large number of recent
investigations, both basic and clinical, no coher-
ent picture has emerged. Rather, it needs to be
emphasised that there is increasing evidence of
heterogeneity of pathogenesis, notably between
Type I and Type II BAV, with increasing evidence
of redox stress and inflammatory activation for
Type I, and of predominant endothelial dysfunc-
tion for Type II [82]. Furthermore, there appears
to be a substantial genetic component to BAV
Table 2. Subtypes of BAV by anatomical structure and associated features [118].
Subtype of BAV Valvular anatomical features Associated features
Type I (70-75%) Fusion of the right and left coronary cusp
resulting in anterior – posterior commissural
orientation
More common in males.
Aortic root dilatation and higher
prevalence of aortic coarctation.
Type II (25-30%) Fusion of the right and non-coronary cusp
result in right and left commissural orienta-
tion.
More common in females.
Higher association with progression
to aortic stenosis and regurgitation
Type III (~1%) Fusion of the non-and left coronary cusp.
Pathogenesis of aortic stenosis
192 Am J Cardiovasc Dis 2011;1(2):185-199
pathogenesis.
As regards the issue of inheritance of BAV, al-
though all series show a predominance of male
patients, the overall thrust of genetic investiga-
tions is to suggest autosomal dominant inheri-
tance in most cases. A number of studies reveal
multiple cases within kindred [83-85]. The most
extensively documented mutations underlying
BAV occur in the NOTCH1 gene [86], which
would also predispose towards valve calcifica-
tion, given that NOTCH signalling has been
linked to expression of osteogenic genes osteo-
pontin and osteocalcin [87]. However the rele-
vant signal transduction pathways for AS devel-
opment in BAV remain uncertain [88] and there
is also evidence of multiple mutations associ-
ated with BAV [85].
The issue of association of BAV with endothelial
dysfunction and possibly with impaired eNOS
signalling has attracted considerable attention
since it was observed that mice lacking the
eNOS gene frequently also had BAV (but not
other congenital cardiovascular abnormalities)
[89]. However, Fernandez et al [82] have more
recently clarified this observation: the abnormal-
ity is actually Type II BAV (right - non-coronary
cusp fusion). Interestingly, these investigators
documented that Type I BAV is present in inbred
Syrian hamsters, without any known association
with eNOS deficiency
While no other endothelial function studies
have specified BAV subtype, there is substantial
additional evidence that BAV may indeed be
associated with eNOS deficiency and/or endo-
thelial dysfunction. Aicher et al [90] quantitated
eNOS protein expression in the aortic wall (but
not the valve) at the time of surgical valve re-
placement and observed lower levels of eNOS in
BAV versus tricuspid ("normal") valves in the
presence of AS. Furthermore, eNOS expression
was negatively correlated with aortic diameter.
A further study has examined brachial flow-
mediated dilatation (FMD): - in a group of men
with BAV, there was an association between
aortic dilatation and impaired FMD [91], consis-
tent with findings of Archer el al [90]. A number
of other studies have evaluated the association
between AS and NO deficiency, but not always
in the context of BAV: these findings will be dis-
cussed later.
The concept of inflammatory change and oxida-
tive stress is a relevant component of the patho-
genesis of BAV, and is in a sense linked to that
of NO deficiency. For example, Tzemos et al [91]
observed that aortic dilatation in BAV was asso-
ciated not only with impaired FMD, but also with
increased plasma levels of matrix metallopro-
teinase 2 (MMP-2), which might have contrib-
uted to the dilatation process. Furthermore,
Phillippi et al [92] demonstrated that suscepti-
bility to oxidative stress was increased, and re-
sponsive expression of metallothionein de-
creased in BAV aortae. While these findings
implicate oxidative stress and inflammatory
activation, there is also evidence that endothe-
lial progenitor cell mobilization in response to
these changes may be abnormal [93], contribut-
ing to perturbation of aortic and valvular endo-
thelial function.
Intervention studies to retard the progression of
AS
Statins
Statins were the first commercially available
drugs studied in aortic stenosis given their es-
tablished benefit in primary and secondary pre-
vention of cardiovascular diseases, as well as
evidence of association of lipid infiltration of
aortic valve in AS [25, 29, 30]. Whilst there are
many "dyslipidemic" animal models of AS (see
animal model section), only one model so far
has shown benefit of statin therapy [94].
There were a number of very encouraging hu-
man retrospective studies of statins in AS, sug-
gesting significant reduction in the rate of pro-
gression of AS with statin use [95-99]. Yet, only
one small prospective open-label non-
randomized observational study of rosuvastatin
in patients with moderate AS showed slowing of
haemodynamic progression of AS [100]. Three
large prospective double-blind randomized pla-
cebo control trials of statins in AS failed to show
any retardation of AS progression [101-103].
ACE-inhibitors (ACEI)/Angiotensin receptor
blockers (ARB)
Numerous studies have demonstrated in-
creased ACE activity/expression and Ang II pres-
ence in stenotic valves [37-39], providing a ra-
tionale for investigation of benefits of ACEI/ARB
therapy in AS. Treatment of cholesterol-fed rab-
bits with angiotensin receptor-1 blocker olme-
Pathogenesis of aortic stenosis
193 Am J Cardiovasc Dis 2011;1(2):185-199
sartan was associated with decreased macro-
phage infiltration and reductions in osteopontin
and ACE in aortic valves [104]. Unfortunately,
no hemodynamic valvular measures of AS pro-
gression were performed. We have demon-
strated hemodynamic retardation of AS, con-
comitantly with reduction in calcification, macro-
phage infiltration, redox stress and improve-
ment in endothelial function, with ACEI ramipril
treatment in a rabbit model of AS [59].
As regards human studies, no prospective trials
of either class of agents have been published to
date. Two main retrospective evaluations of
population studies have provided conflicting
data: - Rosenhek et al [98], utilizing echocardo-
graphic parameters, found no significant effect
of ACE-I therapy on AS progression (albeit with a
trend to slower progression), while O'Brien et al
[105], in a study using CT-based calcification
assessment, found lower rates of AS calcifica-
tion, after correction for comorbidities.
Bisphosphonates
Bisphosphonates, usually used for treatment of
osteoporosis in humans, inhibit bone resorption
as they cause osteoclast apoptotosis [106,
107]. They also inhibit an enzyme in the choles-
terol synthesis pathway, which causes abnor-
malities in the cytoskeleton in the osteoclast,
thus reducing bone resorption [108]. Thus,
bisphosphonates may directly reduce valvular
calcification via their osteoblast action, as well
as indirectly via inhibition of inflammation and
resultant fibrosis. A study in a rat model of dialy-
sis suggested that etidronate, now rarely used
bisphosphanate, limited aortic calcification
[109].
Two small retrospective human studies of
bisphophonates demonstrated reduction in the
AS progression rate [110, 111]. However the
small size and observational nature of these
studies make the results hypothesis generating
at best.
Aldosterone blockade
Aldosterone has been implicated in animal stud-
ies in vascular inflammation and myocardial
fibrosis, and these were ameliorated by aldos-
terone blockade [112]. Eplerenone, an aldoster-
one-receptor antagonist, shown to improve out-
comes in heart failure patients [113], was tri-
alled in a randomized double-blind placebo-
controlled study in patients with moderate-
severe AS [114]. This small trial failed to show
reduction in rate of progression of valve steno-
sis.
Conclusion
The major objective of this review is to present
the case that pathogenesis of AS is an active
process that involves a combination of inflam-
matory activation, increased oxidative stress,
fibrosis and calcification, which should be ame-
nable to therapeutic intervention.
AS is the commonest form of valvular heart dis-
ease and its prevalence is rising due to increas-
ing longevity, especially in Western world. We
present evidence that AS, previously thought to
be "degenerative" disorder of aging, is a com-
plex active process, involving valvular endothe-
lium, fibroblasts and extracellular matrix. The
process is characterized by inflammatory activa-
tion and lipid deposition within valve lesions.
There is extensive valvular matrix remodelling
and fibrosis with increased production of MMP-
1 and -2, TGF-β1, interleukin-1β and TNFα.
There is extensive evidence for increased pro-
duction of Ang II, a major pro-inflammatory and
pro-fibrotic mediator, within stenotic valves. This
would lead to further fibrosis and calcification.
Impaired activation of anti-calcific modulators,
such as fetuin-A and MGP, is alos important in
AS. There is concurrent increased in oxidative
stress and evidence of impairment of the nitric
oxide system as well as associated systemic
endothelial dysfunction.
In BAV, representing the most common congeni-
tal cardiac abnormality, valvular inflammation is
combined with aortopathy. BAV illustrates
unique interplay of genetically driven inflamma-
tory activation and, in some cases, deficiency of
nitric oxide formation.
The most promising targets for pharmacological
interventions have been thought to be lipid infil-
tration and atheroma formation. However, re-
sults of all randomized double-blind prospective
studies of statins have been disappointing [101
-103]. ACE-I/ARB therapy, on the other hand,
shows promising results. Although post-hoc
clinical data are limited and inconclusive, we
have recently demonstrated in an animal model
that ACE-I ramipril retards progression of mild
Pathogenesis of aortic stenosis
194 Am J Cardiovasc Dis 2011;1(2):185-199
AS [59]. In a separate study, olmesartan treat-
ment was also associated with reductions in
macrophage infiltration and ACE in aortic valves
in a rabbit model [104].
Therefore AS represents the result of a pro-
longed inflammatory process leading to valve
calcification and ossification, which represents
a potential target for therapeutic interventions.
Acknowledgements
This work is supported in part by research
grants from the National Health and Medical
Research Council of Australia (NHMRC) and
Cardiovascular Lipid Research Grants
(Australia).
Address correspondence to: John D Horowitz, Cardiol-
ogy Unit, The Queen Elizabeth Hospital, University of
Adelaide, 28 Woodville Road, Woodville, SA 5011,
Australia. Tel: +61 8 82226000; Fax: +61 8
82227201, E-mail: john.horowitz@adelaide.edu.au
References
[1] Carabello BA and Paulus WJ. Aortic stenosis.
Lancet 2009; 373: 956-966.
[2] Lindroos M, Kupari M, Heikkila J and Tilvis R.
Prevalence of aortic valve abnormalities in the
elderly: an echocardiographic study of a ran-
dom population sample. J Am Coll Cardiol
1993; 21: 1220-1225.
[3] Braunwald E and Goldman L. Primary cardiol-
ogy. Philadelphia: Saunders, 2003.
[4] Ross J, Jr. and Braunwald E. Aortic stenosis.
Circulation 1968; 38: 61-67.
[5] Bakaeen FG, Chu D, Ratcliffe M, Gopaldas RR,
Blaustein AS, Venkat R, Huh J, LeMaire SA,
Coselli JS and Carabello BA. Severe aortic
stenosis in a veteran population: treatment
considerations and survival. Ann Thorac Surg
2010; 89: 453-458.
[6] Pai RG, Kapoor N, Bansal RC and Varadarajan
P. Malignant natural history of asymptomatic
severe aortic stenosis: benefit of aortic valve
replacement. Ann Thorac Surg 2006; 82:
2116-2122.
[7] Varadarajan P, Kapoor N, Bansal RC and Pai
RG. Clinical profile and natural history of 453
nonsurgically managed patients with severe
aortic stenosis. Ann Thorac Surg 2006; 82:
2111-2115.
[8] Otto CM, Lind BK, Kitzman DW, Gersh BJ and
Siscovick DS. Association of aortic-valve scle-
rosis with cardiovascular mortality and mor-
bidity in the elderly. N Engl J Med 1999; 341:
142-147.
[9] Volzke H, Haring R, Lorbeer R, Wallaschofski
H, Reffelmann T, Empen K, Rettig R, John U,
Felix SB and Dorr M. Heart valve sclerosis
predicts all-cause and cardiovascular mortal-
ity. Atherosclerosis 2010; 209: 606-610.
[10] Veyradier A, Balian A, Wolf M, Giraud V, Mon-
tembault S, Obert B, Dagher I, Chaput JC,
Meyer D and Naveau S. Abnormal von Wille-
brand factor in bleeding angiodysplasias of
the digestive tract. Gastroenterology 2001;
120: 346-353.
[11] King RM, Pluth JR and Giuliani ER. The asso-
ciation of unexplained gastrointestinal bleed-
ing with calcific aortic stenosis. Ann Thorac
Surg 1987; 44: 514-516.
[12] Freeman RV and Otto CM. Spectrum of calcific
aortic valve disease: pathogenesis, disease
progression, and treatment strategies. Circula-
tion 2005; 111: 3316-3326.
[13] Mulholland DL and Gotlieb AI. Cell biology of
valvular interstitial cells. Can J Cardiol 1996;
12: 231-236.
[14] Weind KL, Ellis CG and Boughner DR. The
aortic valve blood supply. J Heart Valve Dis
2000; 9: 1-7; discussion 7-8.
[15] Cimini M, Rogers KA and Boughner DR. Aortic
valve interstitial cells: an evaluation of cell
viability and cell phenotype over time. J Heart
Valve Dis 2002; 11: 881-887.
[16] Pompilio G, Rossoni G, Sala A, Polvani GL,
Berti F, Dainese L, Porqueddu M and Biglioli P.
Endothelial-dependent dynamic and anti-
thrombotic properties of porcine aortic and
pulmonary valves. Ann Thorac Surg 1998; 65:
986-992.
[17] Filip D, Radu A and Simionescu M. Interstitial
cells of the heart valves possess characteris-
tics similar to smooth muscle cells. Circ Res
1986; 59: 310-320.
[18] El-Hamamsy I, Balachandran K, Yacoub MH,
Stevens LM, Sarathchandra P, Taylor PM,
Yoganathan AP and Chester AH. Endothelium-
dependent regulation of the mechanical prop-
erties of aortic valve cusps. J Am Coll Cardiol
2009; 53: 1448-1455.
[19] Godbole AS, Lu X, Guo X and Kassab GS.
NADPH oxidase has a directional response to
shear stress. American Journal of Physiology -
Heart and Circulatory Physiology 2009; 296:
H152-H158.
[20] McNally JS, Davis ME, Giddens DP, Saha A,
Hwang J, Dikalov S, Jo H and Harrison DG.
Role of xanthine oxidoreductase and NAD(P)H
oxidase in endothelial superoxide production
in response to oscillatory shear stress. Ameri-
can Journal of Physiology - Heart and Circula-
tory Physiology 2003; 285: H2290-H2297.
[21] Butcher JT, Tressel S, Johnson T, Turner D,
Sorescu G, Jo H and Nerem RM. Transcrip-
tional profiles of valvular and vascular endo-
thelial cells reveal phenotypic differences:
influence of shear stress. Arterioscler Thromb
Vasc Biol 2006; 26: 69-77.
[22] Ku DD, Nelson JM, Caulfield JB and Winn MJ.
Pathogenesis of aortic stenosis
195 Am J Cardiovasc Dis 2011;1(2):185-199
Release of endothelium-derived relaxing fac-
tors from canine cardiac valves. J Cardiovasc
Pharmacol 1990; 16: 212-218.
[23] Chen K, Thomas SR and Keaney JF, Jr. Beyond
LDL oxidation: ROS in vascular signal trans-
duction. Free Radic Biol Med 2003; 35: 117-
132.
[24] Mohler Iii ER. Mechanisms of aortic valve cal-
cification. The American Journal of Cardiology
2004; 94: 1396-1402.
[25] Otto CM, Kuusisto J, Reichenbach DD, Gown
AM and O'Brien KD. Characterization of the
early lesion of 'degenerative' valvular aortic
stenosis. Histological and immunohistochemi-
cal studies. Circulation 1994; 90: 844-853.
[26] Olsson M, Dalsgaard CJ, Haegerstrand A,
Rosenqvist M, Ryden L and Nilsson J. Accumu-
lation of T lymphocytes and expression of in-
terleukin-2 receptors in nonrheumatic stenotic
aortic valves. J Am Coll Cardiol 1994; 23:
1162-1170.
[27] Wallby L, Janerot-Sjoberg B, Steffensen T and
Broqvist M. T lymphocyte infiltration in non-
rheumatic aortic stenosis: a comparative de-
scriptive study between tricuspid and bicuspid
aortic valves. Heart 2002; 88: 348-351.
[28] Warren BA and Yong JL. Calcification of the
aortic valve: its progression and grading. Pa-
thology 1997; 29: 360-368.
[29] O'Brien KD, Reichenbach DD, Marcovina SM,
Kuusisto J, Alpers CE and Otto CM. Apolipopro-
teins B, (a), and E accumulate in the morpho-
logically early lesion of 'degenerative' valvular
aortic stenosis. Arterioscler Thromb Vasc Biol
1996; 16: 523-532.
[30] Olsson M, Thyberg J and Nilsson J. Presence
of oxidized low density lipoprotein in nonrheu-
matic stenotic aortic valves. Arterioscler
Thromb Vasc Biol 1999; 19: 1218-1222.
[31] Jian B, Narula N, Li QY, Mohler ER, 3rd and
Levy RJ. Progression of aortic valve stenosis:
TGF-beta1 is present in calcified aortic valve
cusps and promotes aortic valve interstitial
cell calcification via apoptosis. Ann Thorac
Surg 2003; 75: 457-465; discussion 465-
456.
[32] Kaden JJ, Dempfle CE, Grobholz R, Tran HT,
Kilic R, Sarikoc A, Brueckmann M, Vahl C, Hagl
S, Haase KK and Borggrefe M. Interleukin-1
beta promotes matrix metalloproteinase ex-
pression and cell proliferation in calcific aortic
valve stenosis. Atherosclerosis 2003; 170:
205-211.
[33] Kaden JJ, Dempfle CE, Grobholz R, Fischer CS,
Vocke DC, Kilic R, Sarikoc A, Pinol R, Hagl S,
Lang S, Brueckmann M and Borggrefe M. In-
flammatory regulation of extracellular matrix
remodeling in calcific aortic valve stenosis.
Cardiovasc Pathol 2005; 14: 80-87.
[34] Jian B, Jones PL, Li Q, Mohler ER, 3rd, Schoen
FJ and Levy RJ. Matrix metalloproteinase-2 is
associated with tenascin-C in calcific aortic
stenosis. Am J Pathol 2001; 159: 321-327.
[35] Satta J, Melkko J, Pollanen R, Tuukkanen J,
Paakko P, Ohtonen P, Mennander A and Soini
Y. Progression of human aortic valve stenosis
is associated with tenascin-C expression. J Am
Coll Cardiol 2002; 39: 96-101.
[36] Nishimoto M, Takai S, Kim S, Jin D, Yuda A,
Sakaguchi M, Yamada M, Sawada Y, Kondo K,
Asada K, Iwao H, Sasaki S and Miyazaki M.
Significance of chymase-dependent angio-
tensin II-forming pathway in the development
of vascular proliferation. Circulation 2001;
104: 1274-1279.
[37] O'Brien KD, Shavelle DM, Caulfield MT,
McDonald TO, Olin-Lewis K, Otto CM and
Probstfield JL. Association of angiotensin-
converting enzyme with low-density lipoprotein
in aortic valvular lesions and in human
plasma. Circulation 2002; 106: 2224-2230.
[38] Helske S, Lindstedt KA, Laine M, Mayranpaa
M, Werkkala K, Lommi J, Turto H, Kupari M
and Kovanen PT. Induction of local angio-
tensin II-producing systems in stenotic aortic
valves. J Am Coll Cardiol 2004; 44: 1859-
1866.
[39] Helske S, Syvaranta S, Kupari M, Lappalainen
J, Laine M, Lommi J, Turto H, Mayranpaa M,
Werkkala K, Kovanen PT and Lindstedt KA.
Possible role for mast cell-derived cathepsin G
in the adverse remodelling of stenotic aortic
valves. Eur Heart J 2006; 27: 1495-1504.
[40] Mohler ER, 3rd, Adam LP, McClelland P, Gra-
ham L and Hathaway DR. Detection of osteo-
pontin in calcified human aortic valves. Arte-
rioscler Thromb Vasc Biol 1997; 17: 547-552.
[41] O'Brien KD, Kuusisto J, Reichenbach DD, Fer-
guson M, Giachelli C, Alpers CE and Otto CM.
Osteopontin is expressed in human aortic
valvular lesions. Circulation 1995; 92: 2163-
2168.
[42] Mohler ER, 3rd, Gannon F, Reynolds C, Zim-
merman R, Keane MG and Kaplan FS. Bone
formation and inflammation in cardiac valves.
Circulation 2001; 103: 1522-1528.
[43] Kaden JJ, Bickelhaupt S, Grobholz R, Vahl CF,
Hagl S, Brueckmann M, Haase KK, Dempfle
CE and Borggrefe M. Expression of bone sialo-
protein and bone morphogenetic protein-2 in
calcific aortic stenosis. J Heart Valve Dis
2004; 13: 560-566.
[44] Mazzone A, Epistolato MC, De Caterina R,
Storti S, Vittorini S, Sbrana S, Gianetti J, Be-
vilacqua S, Glauber M, Biagini A and Tangan-
elli P. Neoangiogenesis, T-lymphocyte infiltra-
tion, and heat shock protein-60 are biological
hallmarks of an immunomediated inflamma-
tory process in end-stage calcified aortic valve
stenosis. J Am Coll Cardiol 2004; 43: 1670-
1676.
[45] Kaden JJ, Bickelhaupt S, Grobholz R, Haase
KK, Sarikoc A, Kilic R, Brueckmann M, Lang S,
Zahn I, Vahl C, Hagl S, Dempfle CE and
Pathogenesis of aortic stenosis
196 Am J Cardiovasc Dis 2011;1(2):185-199
Borggrefe M. Receptor activator of nuclear
factor kappaB ligand and osteoprotegerin
regulate aortic valve calcification. J Mol Cell
Cardiol 2004; 36: 57-66.
[46] Bucay N, Sarosi I, Dunstan CR, Morony S, Tar-
pley J, Capparelli C, Scully S, Tan HL, Xu W,
Lacey DL, Boyle WJ and Simonet WS. osteo-
protegerin-deficient mice develop early onset
osteoporosis and arterial calcification. Genes
Dev 1998; 12: 1260-1268.
[47] Kaden JJ, Reinohl JO, Blesch B, Brueckmann
M, Haghi D, Borggrefe M, Schmitz F, Klomfass
S, Pillich M and Ortlepp JR. Systemic and local
levels of fetuin-A in calcific aortic valve steno-
sis. Int J Mol Med 2007; 20: 193-197.
[48] Schurgers LJ, Cranenburg EC and Vermeer C.
Matrix Gla-protein: the calcification inhibitor in
need of vitamin K. Thromb Haemost 2008;
100: 593-603.
[49] Koos R, Krueger T, Westenfeld R, Kuhl HP,
Brandenburg V, Mahnken AH, Stanzel S, Ver-
meer C, Cranenburg EC, Floege J, Kelm M and
Schurgers LJ. Relation of circulating Matrix Gla
-Protein and anticoagulation status in patients
with aortic valve calcification. Thromb
Haemost 2009; 101: 706-713.
[50] Ueland T, Gullestad L, Dahl CP, Aukrust P,
Aakhus S, Solberg OG, Vermeer C and
Schurgers LJ. Undercarboxylated matrix Gla
protein is associated with indices of heart
failure and mortality in symptomatic aortic
stenosis. J Intern Med 2010; 268: 483-492.
[51] Miller JD, Chu Y, Brooks RM, Richenbacher
WE, Pena-Silva R and Heistad DD. Dysregula-
tion of antioxidant mechanisms contributes to
increased oxidative stress in calcific aortic
valvular stenosis in humans. J Am Coll Cardiol
2008; 52: 843-850.
[52] Liberman M, Bassi E, Martinatti MK, Lario FC,
Wosniak J, Jr., Pomerantzeff PM and Laurindo
FR. Oxidant generation predominates around
calcifying foci and enhances progression of
aortic valve calcification. Arterioscler Thromb
Vasc Biol 2008; 28: 463-470.
[53] Ngo DT, Stafford I, Kelly DJ, Sverdlov AL, Wut-
tke RD, Weedon H, Nightingale AK, Rosenk-
ranz AC, Smith MD, Chirkov YY, Kennedy JA
and Horowitz JD. Vitamin D(2) supplementa-
tion induces the development of aortic steno-
sis in rabbits: interactions with endothelial
function and thioredoxin-interacting protein.
Eur J Pharmacol 2008; 590: 290-296.
[54] Ngo DT, Stafford I, Sverdlov AL, Qi W, Wuttke
RD, Zhang Y, Kelly DJ, Weedon H, Smith MD,
Kennedy JA and Horowitz JD. Ramipril retards
development of aortic valve stenosis in a rab-
bit model: mechanistic considerations. Br J
Pharmacol 2011;162:722-732.
[55] World CJ, Yamawaki H and Berk BC. Thiore-
doxin in the cardiovascular system. J Mol Med
2006; 84: 997-1003.
[56] Junn E, Han SH, Im JY, Yang Y, Cho EW, Um
HD, Kim DK, Lee KW, Han PL, Rhee SG and
Choi I. Vitamin D3 up-regulated protein 1 me-
diates oxidative stress via suppressing the
thioredoxin function. J Immunol 2000; 164:
6287-6295.
[57] Xiang G, Seki T, Schuster MD, Witkowski P,
Boyle AJ, See F, Martens TP, Kocher A, Son-
dermeijer H, Krum H and Itescu S. Catalytic
degradation of vitamin D up-regulated protein
1 mRNA enhances cardiomyocyte survival and
prevents left ventricular remodeling after myo-
cardial ischemia. J Biol Chem 2005; 280:
39394-39402.
[58] Schulze PC, Liu H, Choe E, Yoshioka J, Shalev
A, Bloch KD and Lee RT. Nitric oxide-
dependent suppression of thioredoxin-
interacting protein expression enhances thio-
redoxin activity. Arterioscler Thromb Vasc Biol
2006; 26: 2666-2672.
[59] Ngo DT, Stafford I, Sverdlov AL, Qi W, Wuttke
RD, Zhang Y, Kelly DJ, Weedon H, Smith MD,
Kennedy JA and Horowitz JD. Ramipril retards
development of aortic valve stenosis in a rab-
bit model: mechanistic considerations. Br J
Pharmacol 2011; 162: 722-732.
[60] Aronow WS, Ahn C, Shirani J and Kronzon I.
Comparison of frequency of new coronary
events in older persons with mild, moderate,
and severe valvular aortic stenosis with those
without aortic stenosis. Am J Cardiol 1998;
81: 647-649.
[61] Stewart BF, Siscovick D, Lind BK, Gardin JM,
Gottdiener JS, Smith VE, Kitzman DW and Otto
CM. Clinical factors associated with calcific
aortic valve disease. Cardiovascular Health
Study. J Am Coll Cardiol 1997; 29: 630-634.
[62] Theroux P and Fuster V. Acute coronary syn-
dromes: unstable angina and non-Q-wave
myocardial infarction. Circulation 1998; 97:
1195-1206.
[63] Chirkov YY, Mishra K, Chandy S, Holmes AS,
Kanna R and Horowitz JD. Loss of anti-
aggregatory effects of aortic valve tissue in
patients with aortic stenosis. J Heart Valve Dis
2006; 15: 28-33.
[64] Riddle JM, Stein PD, Magilligan DJ, Jr. and
McElroy HH. Evaluation of platelet reactivity in
patients with valvular heart disease. J Am Coll
Cardiol 1983; 1: 1381-1384.
[65] Stein PD, Sabbah HN and Pitha JV. Continuing
disease process of calcific aortic stenosis.
Role of microthrombi and turbulent flow. Am J
Cardiol 1977; 39: 159-163.
[66] Chirkov YY, Holmes AS, Willoughby SR, Stew-
art S and Horowitz JD. Association of aortic
stenosis with platelet hyperaggregability and
impaired responsiveness to nitric oxide. Am J
Cardiol 2002; 90: 551-554.
[67] Ngo DT, Sverdlov AL, Willoughby SR, Nightin-
gale AK, Chirkov YY, McNeil JJ and Horowitz
JD. Determinants of occurrence of aortic scle-
rosis in an aging population. JACC Cardiovasc
Pathogenesis of aortic stenosis
197 Am J Cardiovasc Dis 2011;1(2):185-199
Imaging 2009; 2: 919-927.
[68] Chirkov YY and Horowitz JD. Impaired tissue
responsiveness to organic nitrates and nitric
oxide: a new therapeutic frontier? Pharmacol
Ther 2007; 116: 287-305.
[69] Kennedy JA, Hua X, Mishra K, Murphy GA,
Rosenkranz AC and Horowitz JD. Inhibition of
calcifying nodule formation in cultured porcine
aortic valve cells by nitric oxide donors. Eur J
Pharmacol 2009; 602: 28-35.
[70] Sydow K, Schwedhelm E, Arakawa N, Bode-
Boger SM, Tsikas D, Hornig B, Frolich JC and
Boger RH. ADMA and oxidative stress are re-
sponsible for endothelial dysfunction in hyper-
homocyst(e)inemia: effects of L-arginine and B
vitamins. Cardiovasc Res 2003; 57: 244-252.
[71] Boger RH. The emerging role of asymmetric
dimethylarginine as a novel cardiovascular
risk factor. Cardiovasc Res 2003; 59: 824-
833.
[72] Boger RH, Bode-Boger SM, Brandes RP, Phiv-
thong-ngam L, Bohme M, Nafe R, Mugge A
and Frolich JC. Dietary L-arginine reduces the
progression of atherosclerosis in cholesterol-
fed rabbits: comparison with lovastatin. Circu-
lation 1997; 96: 1282-1290.
[73] Ngo DT, Heresztyn T, Mishra K, Marwick TH
and Horowitz JD. Aortic stenosis is associated
with elevated plasma levels of asymmetric
dimethylarginine (ADMA). Nitric Oxide 2007;
16: 197-201.
[74] Brener SJ, Duffy CI, Thomas JD and Stewart
WJ. Progression of aortic stenosis in 394 pa-
tients: relation to changes in myocardial and
mitral valve dysfunction. J Am Coll Cardiol
1995; 25: 305-310.
[75] Wu HD, Maurer MS, Friedman RA, Marboe CC,
Ruiz-Vazquez EM, Ramakrishnan R, Schwartz
A, Tilson MD, Stewart AS and Winchester R.
The lymphocytic infiltration in calcific aortic
stenosis predominantly consists of clonally
expanded T cells. J Immunol 2007; 178: 5329
-5339.
[76] Joris I, Zand T and Majno G. Hydrodynamic
injury of the endothelium in acute aortic
stenosis. Am J Pathol 1982; 106: 394-408.
[77] Dahm M, Dohmen G, Groh E, Krummenauer F,
Hafner G, Mayer E, Hake U and Oelert H. De-
calcification of the aortic valve does not pre-
vent early recalcification. J Heart Valve Dis
2000; 9: 21-26.
[78] Mirzaie M, Meyer T, Schorn B, Schwartz P,
Baryalei M, Rastan A, Lotfi S and Dalichau H.
Calcification tendency of various biological
aortic valves in an experimental animal model.
Cardiovasc Surg 1999; 7: 735-741.
[79] Siu SC and Silversides CK. Bicuspid aortic
valve disease. J Am Coll Cardiol 2010; 55:
2789-2800.
[80] Thanassoulis G, Yip JWL, Filion K, Jamorski M,
Webb G, Siu SC and Therrien J. Retrospective
study to identify predictors of the presence
and rapid progression of aortic dilatation in
patients with bicuspid aortic valves. Nat Clin
Pract Cardiovasc Med 2008; 5: 821-828.
[81] Tzemos N, Therrien J, Yip J, Thanassoulis G,
Tremblay S, Jamorski MT, Webb GD and Siu
SC. Outcomes in Adults With Bicuspid Aortic
Valves. JAMA: The Journal of the American
Medical Association 2008; 300: 1317-1325.
[82] Fernandez B, Duran AC, Fernandez-Gallego T,
Fernandez MC, Such M, Arque JM and Sans-
Coma V. Bicuspid aortic valves with different
spatial orientations of the leaflets are distinct
etiological entities. J Am Coll Cardiol 2009;
54: 2312-2318.
[83] Clementi M, Notari L, Borghi A and Tenconi R.
Familial congenital bicuspid aortic valve: a
disorder of uncertain inheritance. Am J Med
Genet 1996; 62: 336-338.
[84] Glick BN and Roberts WC. Congenitally bicus-
pid aortic valve in multiple family members.
Am J Cardiol 1994; 73: 400-404.
[85] Cripe L, Andelfinger G, Martin LJ, Shooner K
and Benson DW. Bicuspid aortic valve is heri-
table. J Am Coll Cardiol 2004; 44: 138-143.
[86] Garg V, Muth AN, Ransom JF, Schluterman
MK, Barnes R, King IN, Grossfeld PD and
Srivastava D. Mutations in NOTCH1 cause
aortic valve disease. Nature 2005; 437: 270-
274.
[87] Ducy P, Zhang R, Geoffroy V, Ridall AL and
Karsenty G. Osf2/Cbfa1: a transcriptional
activator of osteoblast differentiation. Cell
1997; 89: 747-754.
[88] Niessen K and Karsan A. Notch signaling in
cardiac development. Circ Res 2008; 102:
1169-1181.
[89] Lee TC, Zhao YD, Courtman DW and Stewart
DJ. Abnormal aortic valve development in
mice lacking endothelial nitric oxide synthase.
Circulation 2000; 101: 2345-2348.
[90] Aicher D, Urbich C, Zeiher A, Dimmeler S and
Schafers HJ. Endothelial nitric oxide synthase
in bicuspid aortic valve disease. Ann Thorac
Surg 2007; 83: 1290-1294.
[91] Tzemos N, Lyseggen E, Silversides C, Jamorski
M, Tong JH, Harvey P, Floras J and Siu S. En-
dothelial function, carotid-femoral stiffness,
and plasma matrix metalloproteinase-2 in
men with bicuspid aortic valve and dilated
aorta. J Am Coll Cardiol 2010; 55: 660-668.
[92] Phillippi JA, Klyachko EA, Kenny JPt, Eskay MA,
Gorman RC and Gleason TG. Basal and oxida-
tive stress-induced expression of metal-
lothionein is decreased in ascending aortic
aneurysms of bicuspid aortic valve patients.
Circulation 2009; 119: 2498-2506.
[93] Vaturi M, Perl L, Leshem-Lev D, Dadush O,
Bental T, Shapira Y, Yedidya I, Greenberg G,
Kornowski R, Sagie A, Battler A and Lev EI.
Circulating Endothelial Progenitor Cells in Pa-
tients With Dysfunctional Versus Normally
Functioning Congenitally Bicuspid Aortic
Pathogenesis of aortic stenosis
198 Am J Cardiovasc Dis 2011;1(2):185-199
Valves. Am J Cardiol 2011; 108:272-276.
[94] Rajamannan NM, Subramaniam M, Springett
M, Sebo TC, Niekrasz M, McConnell JP, Singh
RJ, Stone NJ, Bonow RO and Spelsberg TC.
Atorvastatin inhibits hypercholesterolemia-
induced cellular proliferation and bone matrix
production in the rabbit aortic valve. Circula-
tion 2002; 105: 2660-2665.
[95] Novaro GM, Tiong IY, Pearce GL, Lauer MS,
Sprecher DL and Griffin BP. Effect of hydroxy-
methylglutaryl coenzyme a reductase inhibi-
tors on the progression of calcific aortic steno-
sis. Circulation 2001; 104: 2205-2209.
[96] Shavelle DM, Takasu J, Budoff MJ, Mao S,
Zhao XQ and O'Brien KD. HMG CoA reductase
inhibitor (statin) and aortic valve calcium. Lan-
cet 2002; 359: 1125-1126.
[97] Bellamy MF, Pellikka PA, Klarich KW, Tajik AJ
and Enriquez-Sarano M. Association of choles-
terol levels, hydroxymethylglutaryl coenzyme-A
reductase inhibitor treatment, and progres-
sion of aortic stenosis in the community. J Am
Coll Cardiol 2002; 40: 1723-1730.
[98] Rosenhek R, Rader F, Loho N, Gabriel H,
Heger M, Klaar U, Schemper M, Binder T,
Maurer G and Baumgartner H. Statins but not
angiotensin-converting enzyme inhibitors de-
lay progression of aortic stenosis. Circulation
2004; 110: 1291-1295.
[99] Aronow WS, Ahn C, Kronzon I and Goldman
ME. Association of coronary risk factors and
use of statins with progression of mild valvular
aortic stenosis in older persons. Am J Cardiol
2001; 88: 693-695.
[100] Moura LM, Ramos SF, Zamorano JL, Barros
IM, Azevedo LF, Rocha-Goncalves F and Raja-
mannan NM. Rosuvastatin affecting aortic
valve endothelium to slow the progression of
aortic stenosis. J Am Coll Cardiol 2007; 49:
554-561.
[101] Chan KL, Teo K, Dumesnil JG, Ni A and Tam J.
Effect of Lipid lowering with rosuvastatin on
progression of aortic stenosis: results of the
aortic stenosis progression observation: meas-
uring effects of rosuvastatin (ASTRONOMER)
trial. Circulation 2010; 121: 306-314.
[102] Cowell SJ, Newby DE, Prescott RJ, Bloomfield
P, Reid J, Northridge DB and Boon NA. A ran-
domized trial of intensive lipid-lowering ther-
apy in calcific aortic stenosis. N Engl J Med
2005; 352: 2389-2397.
[103] Rossebo AB, Pedersen TR, Boman K, Brudi P,
Chambers JB, Egstrup K, Gerdts E, Gohlke-
Barwolf C, Holme I, Kesaniemi YA, Malbecq W,
Nienaber CA, Ray S, Skjaerpe T, Wachtell K
and Willenheimer R. Intensive lipid lowering
with simvastatin and ezetimibe in aortic steno-
sis. N Engl J Med 2008; 359: 1343-1356.
[104] Arishiro K, Hoshiga M, Negoro N, Jin D, Takai
S, Miyazaki M, Ishihara T and Hanafusa T.
Angiotensin receptor-1 blocker inhibits athero-
sclerotic changes and endothelial disruption
of the aortic valve in hypercholesterolemic
rabbits. J Am Coll Cardiol 2007; 49: 1482-
1489.
[105] O'Brien KD, Probstfield JL, Caulfield MT, Nasir
K, Takasu J, Shavelle DM, Wu AH, Zhao XQ
and Budoff MJ. Angiotensin-converting en-
zyme inhibitors and change in aortic valve
calcium. Arch Intern Med 2005; 165: 858-
862.
[106] Plotkin LI, Weinstein RS, Parfitt AM, Roberson
PK, Manolagas SC and Bellido T. Prevention of
osteocyte and osteoblast apoptosis by
bisphosphonates and calcitonin. J Clin Invest
1999; 104: 1363-1374.
[107] Plotkin LI, Aguirre JI, Kousteni S, Manolagas
SC and Bellido T. Bisphosphonates and estro-
gens inhibit osteocyte apoptosis via distinct
molecular mechanisms downstream of ex-
tracellular signal-regulated kinase activation. J
Biol Chem 2005; 280: 7317-7325.
[108] Rodan GA and Fleisch HA. Bisphosphonates:
mechanisms of action. J Clin Invest 1996; 97:
2692-2696.
[109] Tamura K, Suzuki Y, Matsushita M, Fujii H,
Miyaura C, Aizawa S and Kogo H. Prevention
of aortic calcification by etidronate in the renal
failure rat model. Eur J Pharmacol 2007; 558:
159-166.
[110] Innasimuthu AL and Katz WE. Effect of
Bisphosphonates on the Progression of De-
generative Aortic Stenosis. Echocardiography
2011;28:1-7.
[111] Skolnick AH, Osranek M, Formica P and Kron-
zon I. Osteoporosis treatment and progression
of aortic stenosis. Am J Cardiol 2009; 104:
122-124.
[112] Rocha R, Rudolph AE, Frierdich GE, Na-
chowiak DA, Kekec BK, Blomme EA, McMahon
EG and Delyani JA. Aldosterone induces a
vascular inflammatory phenotype in the rat
heart. Am J Physiol Heart Circ Physiol 2002;
283: H1802-1810.
[113] Pitt B, Remme W, Zannad F, Neaton J, Marti-
nez F, Roniker B, Bittman R, Hurley S, Kleiman
J and Gatlin M. Eplerenone, a selective aldos-
terone blocker, in patients with left ventricular
dysfunction after myocardial infarction. N Engl
J Med 2003; 348: 1309-1321.
[114] Stewart RA, Kerr AJ, Cowan BR, Young AA,
Occleshaw C, Richards AM, Edwards C, Whal-
ley GA, Freidlander D, Williams M, Doughty
RN, Zeng I and White HD. A randomized trial of
the aldosterone-receptor antagonist
eplerenone in asymptomatic moderate-severe
aortic stenosis. Am Heart J 2008; 156: 348-
355.
[115] Roberts WC and Ko JM. Frequency by decades
of unicuspid, bicuspid, and tricuspid aortic
valves in adults having isolated aortic valve
replacement for aortic stenosis, with or with-
out associated aortic regurgitation. Circulation
2005; 111: 920-925.
Pathogenesis of aortic stenosis
199 Am J Cardiovasc Dis 2011;1(2):185-199
[116] Iung B, Baron G, Tornos P, Gohlke-Barwolf C,
Butchart EG and Vahanian A. Valvular heart
disease in the community: a European experi-
ence. Curr Probl Cardiol 2007; 32: 609-661.
[117] Novaro GM, Mishra M and Griffin BP. Inci-
dence and echocardiographic features of con-
genital unicuspid aortic valve in an adult popu-
lation. J Heart Valve Dis 2003; 12: 674-678.
[118] Fernandes SM, Sanders SP, Khairy P, Jenkins
KJ, Gauvreau K, Lang P, Simonds H and Colan
SD. Morphology of bicuspid aortic valve in
children and adolescents. J Am Coll Cardiol
2004; 44: 1648-1651.
... Calcific degeneration, by far, is the most common cause of aortic valve stenosis [4] and has an estimated prevalence of 0.4% in the general population and 1.7% in the population >65 years of age, in developed countries [5]. Degenerative calcific AS is an active process characterized by the disruption of the aortic valvular endothelium with endothelial dysfunction, lipid accumulation, and infiltration of lymphocytes and macrophages that release pro-inflammatory molecules; they recruit fibroblasts and activate osteoblasts, leading to valve fibrosis, progressive thickening, that, over time, evolves into severe valve calcification [6]. Therefore, valve calcification leads to restriction of leaflets mobility, valve area reduction, and flow obstruction. ...
... In particular, in a subset of moderate AS patients, there was a mean annualized peak velocity progression of 0.18 m/s/y [95% CI: 0.12-0.23 m/s/y]. Based on the upper 95% CI for progression of 0.23 m/s/y, the authors suggested that an even lower threshold for the definition of rapid progression than the one previously reported, may be used [6]. Table 1. ...
... The continuity equation is based on the assumption that the LVOT is circular, and the parasternal long-axis plane bisects the LVOT. However, the latter often presents a more elliptical shape, representing one of the main limitations of this calculation: in the case of an elliptical shape, the utilization of the antero-posterior diameter, which is generally smaller than the medio-lateral diameter, may result in underestimation of LVOT area and, thus, of stroke volume and effective AVA (6). In this context, the use of three-dimensional (3D) transesophageal echocardiography (TEE) is of fundamental importance and could overcome this limitation: 3D echocardiography indeed enables the measurement of the LVOT medio-lateral diameter and of the LVOT area planimetry. ...
Article
Full-text available
Echocardiography represents the most important diagnostic tool in the evaluation of aortic stenosis. The echocardiographic assessment of its severity should always be performed through a standardized and stepwise approach in order to achieve a comprehensive evaluation. The latest technical innovations in the field of echocardiography have improved diagnostic accuracy, guaranteeing a better and more detailed evaluation of aortic valve anatomy. An early diagnosis is of utmost importance since it shortens treatment delays and improves patient outcomes. Echocardiography plays a key role also in the evaluation of all the structural changes related to aortic stenosis. Detailed evaluation of subtle and subclinical changes in left ventricle function has a prognostic significance: scientific efforts have been addressed to identify the most accurate global longitudinal strain cut-off value able to predict adverse outcomes. Moreover, in recent years the role of artificial intelligence is increasingly emerging as a promising tool able to assist cardiologists in aortic stenosis screening and diagnosis, especially by reducing the rate of aortic stenosis misdiagnosis.
... With respect to heart valves, NO production by VECs and its subsequent action of VICs is an important paracrine regulator of valve cell function [3,10]. More importantly, dysregulation of NO signalling is implicated in the development of heart valve disease [52][53][54][55]. In the initial stages of CAVD, eNOS is upregulated on the aortic-side endothelium [56], where it contributes to pathological neovascularisation of the valve tissue [57]. ...
... /10.5772/intechopen.112649 calcified portion of the valve, the pathology is not localised at the calcification, and altered stiffness characteristics across the whole leaflet ensures that all cells present will have some degree of pathological programming [53]. ...
Chapter
Full-text available
Valvular interstitial cells (VICs) are the primary cellular component of the heart valve. Their function is to maintain the structure of the valve leaflets as they endure some three billion beats in the course of a human lifespan. Valvular pathology is becoming ever more prevalent in our ageing world, and there has never been a greater need for understanding of the pathological processes that underpin these diseases. Despite this, our knowledge of VIC pathology is limited. The scientific enquiry of valve disease necessitates stable populations of VICs in the laboratory. Such populations are commonly isolated from porcine and human tissue. This is achieved by digesting valve tissue from healthy or diseased sources. Understanding of the many VIC phenotypes, and the biochemical cues that govern the transition between phenotypes is essential for experimental integrity. Here we present an overview of VIC physiology, and a tried-and-true method for their isolation and culture. We make mention of several biochemical cues that the researcher may use in their culture media to ensure high quality and stable VIC populations.
... The monocytes differentiate into macrophages that recognize and internalize modified lipoproteins but become foam cells with time. The lipid core is released from foam cells, which can occur via necrosis (in CAD) and/or apoptosis (in both CAD and AVS) [15,16]. The apoptotic process contributes to reduced levels/less efficient efferocytosis, removing apoptotic cells from phagocytic cells, resulting in increased tissue necrosis and exacerbated atherosclerosis [17]. ...
... Signed informed consent was obtained from all patients. This study followed the principles stated in the Declaration of Helsinki, and the ethics committee of the Centro Hospitalar Universitário de São João approved the protocol (reference [7][8][9][10][11][12][13][14][15][16][17], 22 May 2017). A total of 31 patients were selected with a diagnosis of CAD/AVS (12 for the discovery cohort and 19 for testing), and 21 controls (11 for the discovery and 10 for the testing cohorts) were recruited on the occasion of a consultation for assessment of cardiovascular risk factors, where, besides such factors, family history, signs and symptoms were investigated to rule out clinically meaningful CAD/AVS. ...
Article
Full-text available
Coronary artery disease (CAD) and the frequently coexisting aortic valve stenosis (AVS) are heart diseases accounting for most cardiac surgeries. These share many risk factors, such as age, diabetes, hypertension, or obesity, and similar pathogenesis, including endothelial disruption, lipid and immune cell infiltration, inflammation, fibrosis, and calcification. Unsuspected CAD and AVS are sometimes detected opportunistically through echocardiography, coronary angiography, and magnetic resonance. Routine biomarkers for early detection of either of these atherosclerotic-rooted conditions would be important to anticipate the diagnosis. With a noninvasive collection, urine is appealing for biomarker assessment. We conducted a shotgun proteomics exploratory analysis of urine from 12 CAD and/or AVS patients and 11 controls to identify putative candidates to differentiate these diseases from healthy subjects. Among the top 20 most dysregulated proteins, TIMP1, MMP2 and vWF stood out, being at least 2.5× increased in patients with CAD/AVS and holding a central position in a network of protein-protein interactions. Moreover, their assessment in an independent cohort (19 CAD/AVS and 10 controls) evidenced strong correlations between urinary TIMP1 and vWF levels and a common cardiovascular risk factor-HDL (r = 0.59, p < 0.05, and r = 0.64, p < 0.01, respectively).
... Aortic stenosis (AS) is the commonest valvular heart disease in adults, and its prevalence increases exponentially with age [1]. The complex pathophysiology of aortic stenosis is reflected in its impact on the left ventricular (LV) haemodynamics and remodelling [2]. The subsequent LV pressure overload results in increased wall thickness and hypertrophy, which has been proved to be maladaptive and is associated with increased morbidity and mortality [3,4]. ...
Article
Full-text available
Background: Recent data have suggested that global longitudinal strain (GLS) could be useful for risk stratification of patients with severe aortic stenosis (AS). In this study, we aimed to investigate the prognostic role of GLS in patients with AS and also its incremental value in relation to left ventricular ejection fraction (LVEF) and late gadolinium enhancement (LGE). Methods: We analysed all consecutive patients with AS and LGE-CMR in our institution. Survival data were obtained from office of national statistics, a national body where all deaths in England are registered by law. Death certificates were obtained from the general register office. Results: Some 194 consecutive patients with aortic stenosis were investigated with CMR at baseline and followed up for 7.3 ± 4 years. On multivariate Cox regression analysis, only increasing age remained significant for both all-cause and cardiac mortality, while LGE (any pattern) retained significance for all-cause mortality and had a trend to significance for cardiac mortality. Kaplan–Meier survival analysis demonstrated that patients in the best and middle GLS tertiles had significantly better mortality compared to patients in the worst GLS tertiles. Importantly though, sequential Cox proportional-hazard analysis demonstrated that GLS did not have significant incremental prognostic value for all-cause mortality or cardiac mortality in addition to LVEF and LGE. Conclusions: Our study has demonstrated that age and LGE but not GLS are significant poor prognostic indicators in patients with moderate and severe AS.
... 13 NO has direct effect on aortic valve interstitial cell, and it affects restoration of endothelial function and mitigation of AVC. 14 Also, components of the RAAS seem to have regulatory mechanism of valve calcification. 15 Systemic circulatory factors suggested above affect valvular calcification. ...
Article
Background: Racial disparities in transcatheter aortic valve replacement (TAVR) and surgical aortic valve replacement (SAVR) outcomes have been established, but research has predominantly focused on African Americans and Hispanics, leaving a gap in Asian Americans. This study aimed to investigate disparities in aortic valve replacement outcomes among Asian Americans. Methods: Patients who underwent SAVR and TAVR were identified in National Inpatient Sample from the last quarter of 2015-2020. A 1:2 propensity score matching was applied to Asian Americans and Caucasians. In-hospital perioperative outcomes, length of stay, days from admission to operation, and total hospital charge, were compared. Results: In TAVR, 51,394 (84.41 %) were Caucasians and 795 (1.31 %) were Asian Americans. In SAVR, there were 50,080 (78.52 %) Caucasians and 1233 (1.93 %) Asian Americans. No significant difference was found in post-TAVR complications. However, Asian Americans experienced longer waiting time until operation (p = 0.03) and higher costs (p < 0.01) in TAVR. In SAVR, Asian Americans had higher risks of in-hospital mortality (3.91 % vs 2.39 %, p = 0.01), cardiogenic shock (8.71 % vs 6.74 %, p = 0.03), respiratory complications (14.08 % vs 11.2 %, p = 0.01), mechanical ventilation (13.83 % vs 9.09 %, p < 0.01), acute kidney injury (25.47 % vs 20.13 %, p < 0.01), and hemorrhage/hematoma (72.01 % vs 62.95 %, p < 0.01). Additionally, Asian Americans underwent SAVR had longer lengths of stay (p < 0.01) and higher costs (p < 0.01). Conclusions: Asian Americans were underrepresented in aortic valve replacement. Asian Americans, while having similar post-TAVR outcomes to Caucasians, faced greater risks of post-SAVR mortality and surgical complications. These disparities among Asian Americans call for targeted actions to ensure equitable health outcomes.
Article
Calcified aortic valve disease (CAVD) is a common cardiovascular disease in elderly individuals. Although it was previously considered a degenerative disease, it is, in fact, a progressive disease involving multiple mechanisms. Aortic valve endothelial cells, which cover the outermost layer of the aortic valve and are directly exposed to various pathogenic factors, play a significant role in the onset and progression of CAVD. Hemodynamic changes can directly damage the structure and function of valvular endothelial cells (VECs). This leads to inflammatory infiltration and oxidative stress, which promote the progression of CAVD. VECs can regulate the pathological differentiation of valvular interstitial cells (VICs) through NO and thus affect the process of CAVD. Under the influence of pathological factors, VECs can also be transformed into VICs through EndMT, and then the pathological differentiation of VICs eventually leads to the formation of calcification. This review discusses the role of VECs, especially the role of oxidative stress in VECs, in the process of aortic valve calcification.
Article
Full-text available
Aldosterone blockade reduces mortality and morbidity among patients with severe heart failure. We conducted a double-blind, placebo-controlled study evaluating the effect of eplerenone, a selective aldosterone blocker, on morbidity and mortality among patients with acute myocardial infarction complicated by left ventricular dysfunction and heart failure. Patients were randomly assigned to eplerenone (25 mg per day initially, titrated to a maximum of 50 mg per day; 3319 patients) or placebo (3313 patients) [correction] in addition to optimal medical therapy. The study continued until 1012 deaths occurred. The primary end points were death from any cause and death from cardiovascular causes or hospitalization for heart failure, acute myocardial infarction, stroke, or ventricular arrhythmia. During a mean follow-up of 16 months, there were 478 deaths in the eplerenone group and 554 deaths in the placebo group (relative risk, 0.85; 95 percent confidence interval, 0.75 to 0.96; P=0.008). Of these deaths, 407 in the eplerenone group and 483 in the placebo group were attributed to cardiovascular causes (relative risk, 0.83; 95 percent confidence interval, 0.72 to 0.94; P=0.005). The rate of the other primary end point, death from cardiovascular causes or hospitalization for cardiovascular events, was reduced by eplerenone (relative risk, 0.87; 95 percent confidence interval, 0.79 to 0.95; P=0.002), as was the secondary end point of death from any cause or any hospitalization (relative risk, 0.92; 95 percent confidence interval, 0.86 to 0.98; P=0.02). There was also a reduction in the rate of sudden death from cardiac causes (relative risk, 0.79; 95 percent confidence interval, 0.64 to 0.97; P=0.03). The rate of serious hyperkalemia was 5.5 percent in the eplerenone group and 3.9 percent in the placebo group (P=0.002), whereas the rate of hypokalemia was 8.4 percent in the eplerenone group and 13.1 percent in the placebo group (P<0.001). The addition of eplerenone to optimal medical therapy reduces morbidity and mortality among patients with acute myocardial infarction complicated by left ventricular dysfunction and heart failure.
Article
Full-text available
Valve lesions in degenerative calcific aortic stenosis (CAS), a disorder affecting 3% of those older than 75 years, are infiltrated by T lymphocytes. We sought to determine whether the alphabeta TCR repertoire of these valve-infiltrating lymphocytes exhibited features either of a polyclonal nonselective response to inflammation or contained expanded clones suggesting a more specific immune process. TCR beta-chain CDR3-length distribution analysis using PCR primers specific for 23 Vbeta families performed in eight individuals with CAS affecting tri- or bileaflet aortic valves revealed considerable oligoclonal T cell expansion. In five cases, beta-chain nucleotide sequencing in five selected Vbeta families showed that an average of 92% of the valve-infiltrating T cell repertoire consisted of expanded T cell clones, differing markedly in composition from the relatively more polyclonal peripheral CD8 or CD4 T cell subsets found even in this elderly population. Twenty-four of the valve-infiltrating T cell clones also had the same clone identified in blood, some of which were highly expanded. Interestingly, 22 of these 24 shared clones were CD8 in lineage (p = 1.5 x 10(-12)), suggesting a possible relationship to the expanded CD8(+)CD28(-) T cell clones frequently present in the elderly. Additionally, the sequences of several TCR beta-chain CDR3 regions were homologous to TCR beta-chains identified previously in allograft arteriosclerosis. We infer that these findings are inconsistent with a nonselective secondary response of T cells to inflammation and instead suggest that clonally expanded alphabeta T cells are implicated in mediating a component of the valvular injury responsible for CAS.
Article
There is abundant evidence that the endothelium plays a crucial role in the maintenance of vascular tone and structure. One of the major endothelium-derived vasoactive mediators is nitric oxide (NO). Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of NO synthase. ADMA inhibits vascular NO production at concentrations found in pathophysiological conditions (i.e., 3-15 mmol / l); ADMA also causes local vasoconstriction when it is infused intraarterially. The biochemical and physiological pathways related to ADMA are now well understood: dimethylarginines are the result of the degradation of methylated proteins; the methyl group is derived from S-adenosylmethionine. Both ADMA and its regioisomer, SDMA, are eliminated from the body by renal excretion, whereas only ADMA, but not SDMA, is metabolized via hydrolytic degradation to citrulline and dimethylamine by the enzyme dimethylarginine dimethylaminohydrolase (DDAH). DDAH activity and / or expression may therefore contribute to the pathogenesis of endothelial dysfunction in various diseases. ADMA is increased in the plasma of humans with hypercholesterolemia, atherosclerosis, hypertension, chronic renal failure, and chronic heart failure. Increased ADMA levels are associated with reduced NO synthesis as assessed by impaired endothelium-dependent vasodilation. In several prospective and cross-sectional studies, ADMA evolved as a marker of cardiovascular risk. With our increasing knowledge of the role of ADMA in the pathogenesis of cardiovascular disease, ADMA is becoming a goal for pharmacotherapeutic intervention. Among other treatments, the administration of L-arginine has been shown to improve endothelium-
Article
background Calcific aortic stenosis has many characteristics in common with atherosclerosis, in- cluding hypercholesterolemia. We hypothesized that intensive lipid-lowering therapy would halt the progression of calcific aortic stenosis or induce its regression. methods In this double-blind, placebo-controlled trial, patients with calcific aortic stenosis were randomly assigned to receive either 80 mg of atorvastatin daily or a matched placebo. Aortic-valve stenosis and calcification were assessed with the use of Doppler echocardi- ography and helical computed tomography, respectively. The primary end points were change in aortic-jet velocity and aortic-valve calcium score. results Seventy-seven patients were assigned to atorvastatin and 78 to placebo, with a median follow-up of 25 months (range, 7 to 36). Serum low-density lipoprotein cholesterol concentrations remained at 130±30 mg per deciliter in the placebo group and fell to 63±23 mg per deciliter in the atorvastatin group (P
Article
Microthrombi with evidence of organization were observed in 10 of 19 calcified and stenotic aortic valves (53 percent). The organization that results from such thrombi may contribute to the deformity of the valve. Repetitive deposits of microthrombi, followed by organization and calcification, would explain the continuous process of stenosis in previously deformed aortic valves. The formation of such thrombi may be initiated by turbulent flow and other fluid dynamic factors. © 1977 Dun · Donnelley Publishing Corporation 666 Fifth Avenue, New York, New York 10019 All rights reserved.
Article
The aim of the study is to determine whether the selective aldosterone-receptor antagonist eplerenone delays onset of left ventricular (LV) systolic dysfunction or reduces LV hypertrophy in asymptomatic patients with moderate to severe aortic stenosis. Effects of eplerenone on LV diastolic function and progression of valve stenosis were also evaluated. Sixty-five asymptomatic patients with a peak aortic valve velocity >3.0 m/s and normal LV function were randomized double blind to eplerenone, 100 mg daily (n = 33), or placebo (n = 32) for a median of 19 (interquartile range 15 to 25) months. Cardiac magnetic resonance imaging and echocardiography were performed and N-terminal pro-brain natriuretic peptide was measured at baseline and follow-up. Symptomatic deterioration occurred in 13 subjects randomized to eplerenone and 11 to placebo (P = .34). Change in LV mass index (mean change +/- SD -0.3 +/- 14.6 vs +5.1 +/- 15 g/m(2) per year, P = .3), LV ejection fraction (+0.0% +/- 5.7% vs +0.8% +/- 5.7% per year, P = .9), and LV end-systolic volume index (-1.2 +/- 9 vs +0.04 +/- 12 mL/m(2) per year, P = .8) were small and similar for patients randomized to eplerenone and placebo, respectively. Decrease of aortic valve area (-0.11 +/- 0.22 vs -0.18 +/- 0.24 cm(2)/y, P = .2), worsening of LV diastolic dysfunction by echo-Doppler (E/E' +0.49 +/- 0.7 vs +1.32 +/- 2.0/year, P = .4), increase in the plasma level of N-terminal pro-brain natriuretic peptide (+63% vs +12% per year, P = .1), and decline in physical function score (9 +/- 34 vs 12 +/- 37/year, P = .7) were similar for subjects randomized to eplerenone and placebo, respectively. In asymptomatic patients with moderate-severe aortic stenosis, eplerenone did not slow onset of LV systolic or diastolic dysfunction, decrease LV mass, or reduce progression of valve stenosis.
Article
We sought to examine the effect of angiotensin receptor blocker (ARB) on the formation of lesions in the aortic valves of hypercholesterolemic rabbits. Recently, atherosclerosis has been recognized as a mechanism that is responsible for calcific aortic stenosis. The effect of ARBs might help to prevent aortic stenosis because they have multiple antiatherosclerotic effects. Male Japanese white rabbits (n = 36) were separated as follows: control with chow diet (C) and vehicle (V) groups, both of which were fed a 1% cholesterol diet for 8 weeks, and an ARB group (A), which was fed a 1% cholesterol diet for 8 weeks with ARB (olmesartan, 1 mg/kg/day) for the last 4 weeks. This dose of olmesartan did not affect either blood pressure or cholesterol levels. Dietary cholesterol induced fatty deposition with macrophage accumulation and osteopontin coexpression in valve leaflets, whereas ARB decreased macrophage accumulation (% area: V, 9.3 +/- 0.34; A, 1.4 +/- 0.30; p = 0.003) and osteopontin expression (p = 0.017). Angiotensin-converting enzyme was also up-regulated in V and decreased by olmesartan (p = 0.015). Immunohistochemistry with anti-CD31 antibody revealed that dietary cholesterol disrupted and olmesartan preserved endothelial integrity on the lesion-prone aortic side of the valve (% CD31-positive circumference: V, 30 +/- 3.7; A, 62 +/- 4.8; p = 0.003). Numbers of alpha-smooth muscle actin-positive myofibroblasts were increased in V and decreased by olmesartan (p = 0.003). Real-time polymerase chain reaction revealed that increased amounts of messenger ribonucleic acid for osteoblast-specific transcription factor core binding factor alpha-1 in V were diminished by olmesartan. Atherosclerotic changes in the aortic valves of rabbits fed with cholesterol were inhibited by ARB, whereas endo-thelial integrity was preserved and transdifferentiation into myofibroblasts and/or osteoblasts in valve leaflets was inhibited.