Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Pathophysiology of Aortic Stenosis and Mitral

Regurgitation
Gianluca L Perrucci,1,2 Marco Zanobini,1 Paola Gripari,*†1 Paola Songia,1 Bayan Alshaikh,3
Elena Tremoli,1 and Paolo Poggio**1

ABSTRACT
The global impact of the spectrum of valve diseases is a crucial, fast-growing, and underrec-
ognized health problem. The most prevalent valve diseases, requiring surgical intervention, are
represented by calcific and degenerative processes occurring in heart valves, in particular, aortic
and mitral valve. Due to the increasing elderly population, these pathologies will gain weight in
the global health burden. The two most common valve diseases are aortic valve stenosis (AVS) and
mitral valve regurgitation (MR). AVS is the most commonly encountered valve disease nowadays
and affects almost 5% of elderly population. In particular, AVS poses a great challenge due to
the multiple comorbidities and frailty of this patient subset. MR is also a common valve pathology
and has an estimated prevalence of 3% in the general population, affecting more than 176 mil-
lion people worldwide. This review will focus on pathophysiological changes in both these valve
diseases, starting from the description of the anatomical aspects of normal valve, highlighting all
the main cellular and molecular features involved in the pathological progression and cardiac
consequences. This review also evaluates the main approaches in clinical management of these
valve diseases, taking into account of the main published clinical guidelines. © 2017 American
Physiological Society. Compr Physiol 7:799-818, 2017.

Didactic Synopsis 4. To date, the aortic valve stenosis management is repre-


sented by surgical intervention, since no pharmacological
Major teaching points treatments has been proved to be effective.
1. Understanding the anatomical structures of aortic and
mitral valves is necessary to apprehend the pathophysiol- 5. The major pathological stimuli involved in mitral valve
ogy of aortic valve stenosis and mitral valve regurgitation. regurgitation are as follows:
a. Mechanical, such as tension, shear stress, compression,
2. The major pathological processes involved in aortic valve
and flexure
stenosis are as follows:
b. Chemical, such as serotonin and TGF-β signaling
a. Valve interstitial cell phenotypic switching in
osteoblast-like cells
6. The main cardiac pathologies caused by mitral valve regur-
b. Endothelial-to-mesenchymal transition of valve gitation are as follows:
endothelial cells
a. Left atrial pressure overload
c. Inflammatory cell infiltration
b. Pulmonary edema
d. Fibrosis and osteogenesis
c. Pulmonary hypertension
3. The main cardiac implications caused by aortic valve
stenosis are as follows: * Correspondence to paola.gripari@ccfm.it
** Correspondence to paolo.poggio@ccfm.it
a. Left ventricular pressure overload
1 Centro Cardiologico Monzino, IRCCS, Milan, Italy
b. Cardiac hypertrophy 2 Department of Clinical Sciences and Community Health, University

c. Syncope of Milan, Milan, Italy


3 King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia

d. Pulmonary hypertension † Thisauthor has contributed equally to Marco Zanobini.


e. Angina Published online, July 2017 (comprehensivephysiology.com)
DOI: 10.1002/cphy.c160020
f. Heart failure Copyright © American Physiological Society.

Volume 7, July 2017 799


Valve Pathology Consequences Comprehensive Physiology

d. Left ventricle dysfunction Each leaflet takes the name from the correspondent originat-
ing coronary: left, right, and noncoronary. The free edge of the
e. Heart failure
leaflet has a small bulge named the nodule of Arantius, which
meets together with the other nodules when the leaflets are
7. To date, mortality rates are extremely high without sur-
closed, providing the coaptation area that maintain the unidi-
gical intervention. However, patients with acute MR will
rectional blood flow. The rim of each valve leaflet, attached
also benefit from hemodynamically stabilization by after-
to the root, is slightly thicker than its body and it is known
load reduction accomplished either by pharmacological
as the lunula. The lunulae of adjacent cusps overlap during
(e.g., vasodilators) or by mechanical treatments (e.g., intra-
valve closure increasing valve support.
aortic balloon pump).

Introduction Annulus
The global impact of the spectrum of valve diseases is a cru- The aortic annulus cannot be simply defined as the junction
cial, fast-growing, and underrecognized heath problem. The between the AV and the ventricle. It is a more complex struc-
most prevalent valve diseases, requiring surgical intervention, ture due to the semilunar attachment of the leaflets, which
are represented by calcific and degenerative processes occur- extend from the basal attachment in the LV to the distal one
ring in heart valves. Due to the increasing elderly population, in the sinotubular junction, making a crown shape ring rather
these pathologies will gain weight in the global health bur- than a simple circular one.
den. This review will focus on pathophysiological changes in
the two most common valve diseases, namely, aortic valve
stenosis (AVS) and mitral valve regurgitation (MR). Commissures
The junction between each leaflet attachment is called com-
missure. AV contains three commissures at the top of the annu-
Aortic valve lus, which are equally distributed. The commissure between
The aortic valve (AV) is one of the two semilunar valves of the the left and right leaflet is located at the right posterior aspect
heart. Its function is to prevent regurgitation of blood from the of the aortic root, whereas the commissures between the right
aorta to the left ventricle (LV) during diastole. It is composed or left and noncoronary leaflet are located at the right or left
of three leaflets, an annulus, and several commissures and anterior aspect of the aortic root.
resides within the aortic root (Fig. 1).

Leaflets Structure and cell types


The AV is made up of three semilunar leaflets attached right The leaflets are less than 1 mm thick and three different layers
below the sinus of Valsalva, where the coronary arteries arise. compose them. The fibrosa layer is rich in collagen (mostly

Figure 1 Aortic valve anatomy. Structure and nomenclature of aortic root components. The
upper side of the figure represents thoracic aortic traits, with highlighted distinctions between
ascending aorta and aortic arch. The lower part of the figure recapitulates the detailed aortic
root components: the sinotubular junction of aortic root with ascending aorta; the aortic valve
leaflets; the ventriculo-aortic junction of aortic root and left heart ventricle.

800 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

type I and III) and faces the aorta; the spongiosa layer contains Table 1 Echocardiography Criteria for the Identification of Aortic
glycosaminoglycans and resides in the middle of the leaflets Valve Stenosis Stage
functioning as a bearing, whereas the ventricularis layer is
abundant in elastin fibers facing the cardiac ventricle. Aortic stenosis stages
A monolayer of valve endothelial cells (VECs) covers Mild Moderate Severe
the two outer layers, while a mixed population, called valve
interstitial cells (VIC), resides within all three. The majority Aortic jet velocity (m/s) 2.6–2.9 3.0–4.0 >4.0
of VICs are fibroblastic-like, up to 20% are myofibroblastic- Mean gradient (mmHg) <30a 30–50a >50a
like, and less than 5% are smooth muscle cells (37, 154, 169).
<20b 20–40b >40b
Every cell type is committed to several-specific func-
tions. VECs maintain valve homeostasis, while VICs repair AVA (cm2 ) >1.5 1.0–1.5 <1.0
leaflet micro-damages. In particular, VICs secrete new extra- Indexed AVA (cm2 /m2 ) >0.85 0.60–0.85 <0.6
cellular matrix (ECM) proteins (i.e., collagen, elastin, and Velocity ratio >0.50 0.25–0.50 <0.25
glycosaminoglycans) and eliminate damaged ones, at the
a EAE/ASE Guidelines (15).
same time, VECs control VIC activation, shutting them down b AHA/ACC Guidelines (147).
when the repair processes are completed. Together, these two
cell types provide architectural stability and valve elasticity
(37, 169).
The position of the AV, between the two highest-pressure due to the multiple comorbidities and frailty of this patient
chambers of the heart, and its anatomy lead the AV structures subset. In addition, only subjects with a congenital bicuspid
to be exposed to both bloodstream and mechanical forces. AV, 1% to 2% of the population, develop AVS at a younger
Therefore, it is not surprising that together biochemical and age, usually before 60 years of age (191).
mechanical stimuli may alter AV features causing persistent AV degeneration is defined by echocardiography follow-
damages. In fact, hypercholesterolemia, hyperglycemia along ing European Society (15) and American Heart Association
with hypertension could provide hints of stress to the valve guidelines (147) (Table 1). AVS can be viewed as a pro-
(56). All these stimuli may result in a progressive fibrosis gression from aortic valve sclerosis (AVSc), even if only a
process of the leaflets, remarked by early manifestation, such small percentage of people with AVSc will reach severe AVS.
as inflammation, neovascularization, onset of mesenchymal In AVSc, valve thickening and/or calcification do not affect
tissue, and, at final stages, calcification (18, 37, 154). normal leaflet motion (30, 200). However, as the disease pro-
gresses, leaflets become thicker, calcium nodules form, and
new blood vessels appear (33, 200), impairing leaflets motion
Aortic valve stenosis and causing obstruction of the LV outflow tract during systole
(33, 105) (Fig. 2). The classical triad of symptoms is angina,
AVS is the most commonly encountered valve disease nowa- syncope, and heart failure. Usually, patients are asymptomatic
days. It affects almost 3% of the population, its prevalence for a long period, but eventually these symptoms occur lead-
dramatically increases with age, from 0.7% in young and up ing to decompensation, which conveys a poor prognosis with
to 13% in people older than 75 year of age, thus, being con- median mortality of 5, 3, and 2 years, respectively to angina,
sidered an elderly disease (148). AVS poses a great challenge syncope and heart failure (176).

Figure 2 Echocardiography of normal and stenotic aortic valve in systole. 2D-echocardiography of a


healthy (left) and a stenotic (right) aortic valve in short-axis view. Red arrows (left) highlight the normal, thin
leaflets with an optimal orifice area. Red stars (right) highlight thick calcific leaflet edges with a restricted
aortic valve area.

Volume 7, July 2017 801


Valve Pathology Consequences Comprehensive Physiology

Figure 3 Cellular features of stenotic aortic valve. The cartoon depicts the main cellular mechanisms involved in the pathogenesis of aor-
tic valve stenosis. (A) The differentiation of monocytes into osteoclasts. (B) The endothelial damage leading to endothelial-to-mesenchymal
transition (EndMT) into activated myofibroblasts. (C) The differentiation of valve interstitial cells (VIC) into activated myofibroblast and/or
osteoblast-like cells, leading to fibrosis and calcification, respectively. Adapted with permission from (132).

Cellular mechanisms the expression of osteoblast transcription factor Runx2. Once


Runx2 is expressed, cells are committed to an osteoblast
To date, it has been well established that valve stenosis is a
lineage, upregulating osteopontin, bone sialoprotein II, and
complex process due to the sum of cellular and molecular
osteocalcin eventually leading calcification of the AV tissue
events involving valve cells (VICs, VECs, and vascular peri-
(91, 162, 181).
cytes) modification and circulating cells (inflammatory cells
There are evidences indicating that BMP signaling is
and bone-marrow derived cells) action (Fig. 3).
activated in human calcified valves. Moreover, several studies
have also shown higher levels of SMAD 1/5/8 and Runx2 in
human valve leaflet undergoing calcification (6, 134). How-
VIC phenotypic switching in osteoblast-like cells
ever, to date the in vitro studies and the animal model used are
Several studies have described VIC switching in osteoblast- not able to recapitulate all the complex pathway interactions.
like cells, able to form bone tissue (6, 93, 131, 134, 165). The
abnormal activation of quiescent VICs leads to this phe-
Endothelial-to-mesenchymal transition
notype transition. Once activated, VICs become more sen-
sible to osteogenic mediators. Among these pro-ostegenic VICs are not the only cell population able to undergo
molecules, bone morphogenic proteins (BMPs) are the most phenotypic switch, from quiescent to osteoblastic-like cells.
important, since studies on vascular calcification showed their Indeed, VECs have the same capability; however, this
capability to stimulate osteoblasts and trigger bone forma- process needs further steps. Initially, VECs must undergo
tion (18). Experimental models showed transforming growth endothelial-to-mesenchymal transition (EndMT). During
factor-beta (TGF-β) family members, such as BMP-2 and this process, VECs lose their ability to maintain homeostasis
BMP-4, are secreted not only by activated endothelial cells, and most of their endothelial cell properties. In particular,
in response to hemodynamic alterations (35, 193), but also EndMT leads to a decrease or completely lost of endothelial
by aged-rodent VICs (187). BMP protein family exerts their markers expression (i.e., vascular endothelial cadherin,
effect through SMAD and Wnt/β-catenin signaling pathway platelet endothelial cell adhesion molecule (PECAM1), von
activation as well as through Msx2, an osteochondrogenic Willebrand factor) and a simultaneous acquisition of typical
transcription factor (18). All these pathways lead to induce features of mesenchymal cells. Among these properties,

802 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

the VECs, undergoing EndMT, increase the expression of and preventing RAGE activation. In this context, patients with
α-smooth muscle actin (α-SMA), collagen type I, vimentin AVS have low soluble RAGE plasma levels than healthy sub-
as well as migratory properties (161). Several well-known jects (14).
stimuli are involved in the EndMT of VECs, such as In the last years, several studies investigated the role of
TGF-β, transcription factor Msx2, and β-catenin signaling. lymphocyte in calcified leaflets of AVS patients. Intriguingly,
Interestingly, all of them have been linked to the progression it has been shown that isolated T-cells from diseased AVs may
of the AV calcification. However, all the data on these play an active role in AVS pathogenesis rather than mediate a
molecular processes have been obtained from experiments secondary response (208). Moreover, a recent study revealed
on mitral VECs—and not aortic—involving human in vitro the presence of B-cells within AV leaflets of AVS patients
studies and an ovine model (192, 209). The lack of EndMT and showed that these cells were also active (142). All these
studies on AV is mainly because a very small percentage of observations open new questions and further studies, in the
VECs undergo EndMT. Thus, further studies are necessary field of immunology, are needed to clarify a number of still
to better investigate VEC behavior in stenotic human AVs. unraveled key points related to AVS pathogenesis.

Inflammatory cells Fibrosis and osteogenic processes


The unbalanced number of infiltrated macrophages and leuko- As inferred by previously described overview on cell mech-
cytes in healthy versus calcified human AVs supported the anisms, the main processes involved in AVS are fibrosis and
hypothesis of valve calcification and subsequent valve steno- calcification. There are extensive fibrotic zones as well as
sis as an inflammatory disease process (93,142). Inflammatory calcium nodule formation in diseased human (34) and mice
cell infiltration is an early event in valve disease establish- (133) AVs. Due to the overlapping features between fibrosis
ment and it starts at the outer layers of the valve, which are and calcification, Miller and colleagues (132) asserted in a
more vulnerable. Activated VECs, expressing typical mes- comprehensive review that the term “fibrocalcific” could be
enchymal adhesion molecules, facilitate the recruitment and more suitable referring to AVS.
migration of monocytes into the leaflets. An interesting in vivo The early phase of degenerative AVS is characterized by
study, involving the ApoE-deficient mouse model, showed an VIC activation, where VICs acquire a myofibroblastic-like
increased number of macrophages along with the degree of phenotype (133), and overproduction/secretion of ECM com-
valve calcification (143). ponents (217) cause tissue fibrosis. However, specific ECM
In the initial phase of valve degeneration, the presence alterations, in AVS patients, will be further discussed in the
of infiltrated inflammatory cells is often accompanied by following paragraphs.
a higher concentration of proinflammatory cytokines (3, 4) Concerning the calcification, many questions are still
(i.e., tumor necrosis factor-α (TNF-α) interleukin-1β (IL- open and further studies are needed to clarify the mech-
1β) and nuclear factor kappa-B (NF-κB) ligand) and spe- anisms underlying this process. To date, there are several
cific molecules secreted by macrophages and leukocytes (i.e., potential mechanisms involved in calcium nodules deposition
metalloproteinases (MMPs) and endoproteases). In particu- in AVS. It has been suggested that valve calcification may
lar, these latter proteins specifically break down ECM com- progress by processes similar to those occurring in bones
ponents, such as collagen and elastin, well-known processes (138), by accumulation of amorphous calcium deposits [crys-
involved in pathological alteration of valve architecture (93). talline ultrastructure lacking live cells within the calcified
Among proinflammatory cytokines, TNF-α may play a mass (138, 201)], by cellular necrosis, or even by caspase-
pivotal role on downstream mediator of inflammation-induced dependent apoptosis (201). The mechanism most acknowl-
calcification. Several experimental evidences revealed TNF- edged accounted for mineralization of valve tissues resembles
α involvement in stenotic valve pathology. In interleukin-1 in part the skeletal ossification. In particular, only a subset of
receptor antagonist deficient mice (IL-1Ra−/− ) occurs valve cells that express osteogenic markers (4, 133, 134), resem-
thickening, calcification, and sclerosis/stenosis, however, in bling thus osteoblast characteristics, produces the nucleation
the same model also lacking for TNF-α (TNF-α−/− /IL- sites where calcium nodules could grow. However, the signal-
1Ra−/− ) this pathological valve phenotype was abrogated ing cascade responsible for the trigger and the progression of
(85). Moreover, TNF-α has been shown to induce vascular AVS, in vivo, is still unknown.
calcification and MMP activation in diabetic mice (5).
Activation of receptors of advanced glycosylation end
products (RAGE) can increase vascular smooth muscle cell Molecular mediators
calcification rate, both in vitro and in vivo (60,79,197). These Molecular mediators that induce pro-osteogenic differenti-
observations could suggest that RAGE activation may con- ation stimuli are in the spotlight because clinicians and
tribute to the AVS process, since elevated plasma and tissue researchers repute them triggers of AVS. In the next para-
AGE levels, ligands of RAGE, have been found in subjects graphs, we will analyze the main signaling pathways and the
more prone to AVS (62, 96). Additionally, circulating soluble specific molecules that are thought to be involved in AVS
RAGE is considered antiatherogenic being a decoy for AGEs initiation and progression.

Volume 7, July 2017 803


Valve Pathology Consequences Comprehensive Physiology

TGF-𝛽 signaling It is well known that increased levels of BMPs and


phospho-SMAD1-5-8 are found in fibrocalcific AVs (164,
The TGF-β family regulates a broad range of cellular func- 207). Although, the mechanisms contributing in BMP
tions, including growth, development, and tissue homeostasis increase are not completely clear to date. However, recent
(135). A peculiar feature of TGF-β is its localization, since this studies suggest that hemodynamic alterations (i.e. shear
molecule is stored in the ECM as a latent complex. TGF-β acti- stress) may stimulate the BMP2-4 secretion by valve endothe-
vation can be mediated by two different mechanisms, integrin lium on the aortic side (145, 196). In particular, this study
dependent or integrin independent. The former mechanism exploited porcine AV leaflets exposed to altered shear stress
is mediated by mechanical release of TGF-β produced by the conditions, showing a strong increase in both BMP4 and
direct binding of specific integrins with TGF-β latent complex TGF-β expression, which in turn stimulated inflammatory
(9). Otherwise, the integrin-independent mechanisms involve responses, observed as an increase of VEC adhesion molecule
plasmin (119), matrix MMPs (9), and thrombospondin-1 expression. Interestingly, these results were also validated in
(185). These proteins cleave the latent complex and sub- human AVs (7).
sequent conformational rearrangements allow its activation, Regarding the early step of AVS, a hypercholesterolemia
releasing the active form of TGF-β. In the last 20 years, the mouse model showed increased levels of active SMAD1-5-8
integrin-mediated mechanisms of TGF-β activation has been before any calcium nodule formation and valve impairment
deeply characterized and it has been demonstrated that TGF-β (134). In addition, SMAD6-7, molecules acting as inhibitors
acts differently, depending on the ECM compliance (206). of SMAD1-5-8 play a significant role in preventing calcifica-
TGF-β signaling starts with TGF-β dimerization and its tion. Indeed, an important study by Galvin et al. (58) demon-
subsequent binding to TGF-β receptor type I (TGF-βR1) and strated that SMAD6 null mice, starting at 2 weeks after birth,
type II (TGF-βR2). These two form a receptor heterocom- were prone to develop both cardiovascular calcification and
plex, able to recruit and phosphorylate suppressor of mothers aortic ossification.
against decapentaplegic (SMAD) proteins, triggering a wide
range of signal transduction pathways (81). In vitro analy-
sis obtained on VICs showed that TGF-β1 strongly promotes Wnt/𝛽-catenin signaling
cell apoptosis, cellular aggregation, and calcium nodule for- Wnt proteins are a family of secreted glycoproteins used for
mation (42, 89, 203). However, administration of TGF-β1 to short- or long-range signaling. The receptors of Wnt proteins
VICs plated on a less stiff collagen matrix did not induced belong to the Frizzled family and they are able to regulate a
neither osteogenic differentiation nor calcification (214). In variety of cellular processes (e.g., cellular differentiation, tis-
addition, as previously mentioned, it has been also highlighted sue morphogenesis, and tissue homeostasis) (120, 173). The
the ability of TGF-β in EndMT promotion. Paranya et al. Wnt/β-catenin pathway is one of the major axes involved in
(160) demonstrated that ovine aortic VECs treated with TGF- regulation of AV formation and disease (83). β-catenin is a
β exhibited higher levels of typical EndMT markers, increased transcription factors bound to the cytosolic tail of Wnt recep-
α-SMA with subsequent PECAM1 reduction. tor. When Wnt receptor is activated, β-catenin do not get
Experimental evidences have shown that also circulating degraded through ubiquitination but remain stable and subse-
plasma levels of TGF-β were associated with AVS. In par- quently is released to the cytosol. Finally, β-catenin translo-
ticular, a threefold increase, in TGF-β levels, was found in cate to the nucleus where activates the transcription of specific
AVS patients compared to healthy subjects (202). In contrast, genes (120).
at valve level, calcified valves presented similar expression The expression of several modulators of the Wnt signaling
pattern of TGF-β when compared to normal AVs (213). pathway, such as WIF-1, DKK-1, and sFRP-3, has been found
All the collected data on TGF-β signaling, in both ani- upregulated in patients with AVS, suggesting their potential
mal models (133, 134) and AVS patients (89), candidate this role as pathology biomarkers (11). Additionally, increased
molecule as potential early driving mediator of fibrocalcific levels of low-density lipoprotein receptor-related protein 5
processes. (Lrp5) together with increased levels of nuclear β-catenin
were reported in human stenotic valves (28). High levels of
β-catenin were also showed in calcified valves of hypercholes-
Bone morphogenetic protein signaling
terolemic animal models (134,170). All these data support the
Bone morphogenetic proteins (BMP) are growth factors, hypothesis that alteration in Wnt/β-catenin signaling could
belonging to the TGF-β superfamily, not only localized in pave the way to AV degeneration.
bone but deeply involved during AV development and injuries
(36, 193). The BMP pathway starting mechanism is simi-
Reactive oxygen species
lar to the TGF-β one. Two molecule of BMP interact with
BMP receptors (BMPR1 and BMPR2) forming a heteroclom- Reactive oxygen species (ROS) appear to play a pivotal role
plex that phosphorylates SMAD1-5-8 complex and mitogen- in atherosclerosis, stroke, and other cardiovascular diseases
activate protein kinase (MAPK), triggering a myriad of down- (74). Both superoxide (O2 − ) and hydrogen peroxide (H2 O2 )
stream pathways. result to be significantly increased in stenotic AVs, as well as

804 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

the expression of nitric oxide (NO) synthase and the reduction Moreover, it has been described a large presence of inflam-
of antioxidant enzymes (131). matory cell infiltration (i.e., macrophages), in fibrocalcific
Several experimental evidences underline a close relation aortic leaflet, that are able to produce and release large
between ROS activity and the progression of calcific valve amount of AngII (154). In 2004, Helske et al. (75) discovered
impairment. Indeed, it is well known that ROS levels increase that the expression of angiotensin converting enzyme (ACE)
prior to valve dysfunction in mice, suggesting that this event and one of the AngII receptor (angiotensin type I receptors,
does not merely happen because of the calcification-related AT1R) were increased in stenotic valves and colocalized with
modifications (133). Moreover, ROS have been found rele- macrophages and mast cells. Animal models confirmed the
vant in the transduction of pro-osteogenic and pro-fibrotic involvement of RAS in AVS progression using AT1R block-
signaling cascades (41). ers, which leaded to reduce inflammatory cell infiltration and
Despite the clear key-points associating ROS and AV cal- myofibroblast activation, already at the early stages of the
cification initiation and progression, to date, several link are disease (10).
still missing. For example, the precise role of NO synthase
isoforms and NAD(P)H oxidase isoforms, which contribute
RANK/RANKL/OPG axis
to increase ROS generation. In addition, the real contribu-
tions of the different antioxidant mechanisms (e.g., catalase, The interactions between receptor activator of NF-κB
superoxide dismutases, and peroxidases) in their respective (RANK), RANK ligand (RANKL), and osteoprotegerin
subcellular compartments. (OPG) modulates oxidative stress and inflammation with
important consequences on calcification progress in bones,
arteries, and probably causing also impaired aortic leaflet
Nitric oxide bioavailability motion (43). Increased RANK/RANKL activation may influ-
ence calcification on circulating and interstitial cells. Indeed,
The counterpart of ROS is NO that plays a key role in sev-
the production of RANKL by vascular smooth muscle cells,
eral cardiovascular disease. Its bioavailability is frequently
under calcification stimuli, is sufficient to promote both mono-
inversely correlated with ROS levels. The major NO pro-
cyte recruitment (proinflammatory cell) and osteoclast differ-
ducer is represented by the enzyme endothelial nitric oxide
entiation (interstitial cell) in an atherosclerotic mouse model
synthase (eNOS), which has been found upregulated on the
(26). The osteoclast differentiation mediated by RANKL may
aortic side of the impaired valves during the early stages of
be driven by ROS production, even if RANKL induces calci-
valve pathology (65). This side-specific response could be due
fication of vascular smooth muscle cells through the BMP4
to a protective mechanism activated by VECs. The protective
pathway (108,159,183). The last mentioned actor of this axis
role of eNOS, which acts also as inhibitor of VIC proliferation,
is OPG that is an endogenous RANKL decoy receptor able
deeply depends on the expression levels of its cofactors and
to inhibit inflammation and the pro-osteogenic pathway acti-
substrates. However, it has been notice that eNOS overexpres-
vation. Based on animal models and human studies, it has
sion, in a hypercholesterolemic mouse model, could acceler-
been suggested that OPG may act as inhibitor of calcification
ate the atherosclerotic process. This event is strongly related to
in AV leaflets. Indeed, it has been shown that: (i) in OPG−/−
eNOS uncoupling (157), a peculiar phenomenon that leads to
knockout mice aortic calcification was inhibited (152); (ii)
ROS generation and occurs when eNOS is decoupled from its
atherosclerotic lesion progression was quicker in ApoE−/−
cofactors or its substrates are not available (118). Moreover,
OPG−/− double knockout mice then in ApoE−/− model (16);
endogenous inhibitors of eNOS have been shown significantly
(iii) OPG administration prevented calcification of the aorta
increase in patients with stenosis of AV (27). Thus, the modu-
in ldlr−/− knockout mice (140); and (iv) RANKL expression
lation of the molecular system underlying NO bioavailability
was higher in human stenotic valve than in control (92).
may represent a possible therapeutic strategy to decrease AV
calcification progression. Indeed, exogenous NO administra-
tion reduced calcium nodule formation in VICs (99) and the Peroxisome proliferator-activated receptor gamma
administration of statins, in a hypercholesterolemic animal
Peroxisome proliferator-activated receptor gamma (PPARγ)
model, increased eNOS levels with concomitant NO release
is a ligand-dependent transcription factor, member of the
(171).
nuclear hormone receptor superfamily (71). Several findings
depict a strong interplay between PPARγ and AVS (97), (212).
In particular, increased PPARγ expression reduces differen-
Renin—angiotensin system
tiation of progenitor cells into osteoblast-like cell, in vitro
Renin-angiotensin system (RAS) and the principal effector, (97). Accordingly, PPARγ inhibition increases differentia-
angiotensin II (AngII), are well-establish signaling known tion of embryonic stem cells into osteoblasts (212). Despite
to be involved in oxidative stress, inflammation, and several these evidences of PPARγ involvement in AVS, to date no
processes able to trigger the early stages of AVS (125). AngII study have neither deeply investigated nor elucidated this
overexpression is strongly related to hypertension, which is association. However, it has been highlighted that the role
an important risk factor for AVS (148). of PPARγ ligands are important in antioxidant defenses and

Volume 7, July 2017 805


Valve Pathology Consequences Comprehensive Physiology

Figure 4 Aortic valve stenosis pathophysiology. Phenotype of stenotic aortic valves and the involvement of valve stenosis in cardiac
diseases. In the left side are depicted normal and stenotic aortic valves in diastole and systole. In the stenotic picture are present several
leaflet regions of spotty calcification, which stiffen the valve structure and alter the dynamic of valve opening. In the right side of the figure
are mentioned the main cardiac pathologies caused by aortic valve stenosis.

antinflammatory gene expression induction (175). Since Pathophysiology


PPARγ regulates osteoblast modulation and plays an impor-
AVS will lead to narrowing of the aortic orifice and once aortic
tant role as antioxidant and antinflammatory agent, it may be
valve area (AVA) reaches measurements below 2.0 cm2 will
enrolled as therapeutic target for calcific AV disease, but we
cause a progressive increase in pressure gradient (Fig. 4). This
need to be very careful because PPARγ affects a large number
will cause LV pressure overload and subsequent increased
of genes (98).
wall stress. To overcome these stress changes, LV reacts with
compensatory mechanisms. As per Laplace law, wall stress is
proportional to pressure × radius/thickness. In other words,
Notch signaling any increase in pressure will cause an increase in wall stress,
Polymorphisms accounted for Notch1 gene have been so to compensate, the heart has to reduce diameter and/or
reported to be strongly associated with AVS early develop- increase its thickness (hypertrophy), thus normalizing the wall
ment, in particular in bicuspid AVs (59). In Notch1 haploin- stress (30, 63, 174). Usually, it begins with a slow LV remod-
sufficient mice, the calcification of the aortic leaflets occurs eling leading eventually to concentric hypertrophy associated
through the repression of BMP2 expression (117, 146). How- with increased LV mass. Concentric hypertrophy is a mech-
ever, recently has been proposed that decreased levels of anism that increases the number of sarcomeres reducing the
Notch1, in the endothelium, provoke a subsequent decrease intraventricular radius, thus the ventricular volume results
of matrix Gla protein (MGP), which allow increased BMP2/4 reduced.
availability and, at the same time, promotes osteogenic dif- Hypertrophy is considered a double-edged sword, ben-
ferentiation of VICs (8). eficial in some aspects and deleterious in others (30). This
can be simply explained by supply/demand mismatch. Once
hypertrophy begin, the heart requires more oxygen to perform
Matrix metalloproteinases and cathepsins its normal functions, however, the impairment of blood flow,
due to decrease LV compliance consequential to AVS, leads
MMP and cathepsin degrade both matrix and non-matrix pro- to compression of coronary arteries limiting their flow (30).
teins in the extracellular environment. Both of these prote- AVS could also cause syncope, but the exact mechanism
olytic enzyme classes have been found upregulated in AVS tis- is still unclear. However, it could be explained by exercise
sues and therefore corroborating the hypothesis regarding the induced vasodilatation associated with fixed cardiac output
association with fibrocalcific processes occurring at AV lev- state and inability to augment the cardiac output. Other clini-
els. In particular, the expression of MMP-1, MMP-2, MMP-3, cians have postulated that very high intraventricular pressure,
MMP-9, and cathepsins S, K, V, and G has been reported in developed during exercise in people affected by AVS, causes
valve pathology (50,55,76,77,88,93). Although the functional a reflex depressor response causing in turn syncope (i.e.,
role of a single MMP or cathepsin, in valve stenosis, is still vasoplegic syncope) (30). In addition, arrhythmias occurring
unclear, the complex remodeling of ECM could be the main in AVS patient, induced by exercise, lead to ischemia that
process involved in calcified nodule formation and growth, impairs ventricular filling either by reducing filling time or
along with elastin and collagen deposition/brake down (166). loss of atrial kick, causing syncope (30, 95).

806 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

These symptoms can be either due to diastolic or systolic


dysfunctions. The former is due to concentric hypertrophy
and subendocardial fibrosis that will lead to impairment of
LV relaxation. This impairment has deleterious effects on
diastolic filling pressure leading: (i) to reduction of LV com-
pliance; (ii) to increase pressure of the left atrium (LA). Con-
sequently, these modifications eventually lead to pulmonary
congestion and heart failure.
The reduced LV compliance elevates LV diastolic pres-
sure impairing the LV filling, reducing the stroke volume
and subsequently the aortic pressure. These mechanical alter-
ations lead to the activation of neurohormonal pathways to Figure 5 Mitral valve anatomy. Structure and nomenclature of mitral
valve. The mitral valve represents the gate between left atrium and left
compensate the reduction of cardiac output. ventricle. The perfect closure of the valve depends on cardiac papillary
Another potential problem in AVS patients, underesti- muscles, which orchestrate the leaflet traction during systole through
mated in most instances, is represented by pulmonary hyper- chordae tendinae.
tension. The process starts with LV diastolic dysfunction and
the subsequent increase in diastolic filling pressure, which
leads LA and pulmonary venous hypertension. Raise in LA tendineae, and the LV wall along with its papillary muscles
pressure will activate pulmonary vascular protective mech- (PM) compose it (Fig. 5).
anism, by which pulmonary arterioles start to constrict. In
addition, this will cause a further reduction of cardiac output,
due to the secondary obstruction caused by the developed pul- Leaflets
monary hypertension, causing right ventricle (RV) pressure Mitral valve contains two leaflets, one anterior (adjacent to
overload and eventually an overt RV failure (101). the AV) and one posterior (adjacent to the wall). Each leaflet is
The systolic dysfunction exact mechanism is still contro- separated by commissures, anterolateral, and posteromedial,
versial. However, it could be explained by myocyte exhaustion respectively (172). The anterior leaflet is the largest one and
that can no longer compensate for the increase in afterload, attaches only to one third of the annulus, which is in continu-
leading the fall of the ventricular functions (30, 73, 78). ity with the AV. The posterior leaflet is smaller than anterior
one, but it attaches to two third of the annulus, and it contains
multiple indentations (referred as “clefts”), which forms 2 to
Management of AVS 5 scallops (segments). Along the elongated free edge, triscal-
From the prognostic point of view, patients with AVS and loped leaflet is the most common form. These indentations
symptomatic heart failure have the worst outcomes. should not extend all the way through the leaflet to the annu-
In severe symptomatic AVS only mechanical relief will lus; if this is seen, then it is usually associated with patholog-
improve the prognosis. Medical treatment for sever AVS is ical valve regurgitation (128). The posterior lateral segment,
not effective (30, 163). Several studies linked AVS inflamma- referred as P1, is adjacent to the anterolateral commissure, P2
tory processes with atherosclerosis because they share many is central and varies in size, and the medial segment (P3) is
risk factors (94, 144, 158). Indeed, some studies have been adjacent to the posteromedial commissure (32).
trying to prove that statin therapies can retard the progres- The leaflets are divided into several zones: “basal”, where
sion of stenosis. However, all of these studies failed since they are attached to the atrial-ventricular junction, clear, which
randomized controlled trials showed no difference between is the central portion, and “rough”, area where the chordae
statin and placebo group, in term of inducing regression or insert and make up the coaptation area. For tight leaflet coap-
preventing progression of AVS (40). Thus, surgical aortic tation, a redundant leaflet tissue in the rough zone is criti-
valve replacement is the treatment of choice when it comes to cally important, allowing good apposition and preventing any
severe symptomatic AVS or severe AVS with dropped ejec- regurgitant volume.
tion fraction, as it carries low mortality and complication
rates with significant improvement in patient survival. How-
ever, transcutaneous aortic valve implantation is the treatment Annulus
of choice in high-risk surgical patients. The junction between the LA and LV defines the MA and
it gives attachment to the mitral leaflets. It is a dynamic,
anatomically well-defined structure and, in three dimensions,
Mitral valve it has a nonplanar saddle shape (44,103,110,111). The anterior
portion of the MA is continuous with the aortic annulus. The
The mitral valve apparatus is a complex three-dimensional angle between MA-to-aortic annulus changes dynamically
functional unit critical to unidirectional blood flow (44). The over the cardiac cycle, with displacement coupled through
mitral annulus (MA), the mitral valve leaflets, the chordae the fibrous continuum (198). For this reason, the anterior

Volume 7, July 2017 807


Valve Pathology Consequences Comprehensive Physiology

portion is fibrous and less prone to dilation. Beyond this point, While, VICs are fibroblastic-like cells originating by mes-
the remaining two-thirds of the annulus (posterior portion) are enchymal lineage. The principal function of VICs is the con-
mainly muscular, which makes this region more susceptible tinuous synthesis and degradation of the ECM components,
to dilatation, as well as to pathological calcification (128). thus maintaining correct valve ECM homeostasis. Since the
first step in valve degeneration is based on the ECM home-
ostasis alteration, VICs play a pivotal role in the cellular-
Chordae tendinae mediated mitral valve degeneration. Indeed, the transition of
The chordae originate from PMs and attach to the leaflet in VICs from a “quiescent” to an “activated” state has been
a fan-like way. According to the attachment site, they are described as the principal event occurring in human myxo-
divided into three types. Primary chords, involved in mitral matous mitral valve degeneration (167). Activated VICs typ-
valve competence, which are attached to the free edge on the ically display myofibroblast-like features, such as expression
“rough” zone. Secondary chords, involved in left ventricular of cytoskeletal elements (i.e., α-SMA), desmin, markers of
geometry and function, which are bound to the body of the mesenchymal activation (i.e., embryonic nonmuscle myosin,
leaflet on the ventricular side. Tertiary chords, only on pos- SMemb) as well as increased expression of matrix catabolic
terior leaflet, which are attached directly to ventricular wall enzymes including matrix metalloproteinase (MMP)-1, -2, -9,
(149). and -13, and elastases (i.e., cathepsin K). To date, activated
VIC phenotype is widely accepted as pathological trait in
heart valve impairment (114).
Papillary muscles
There are two PMs arising from the LV free wall, one antero-
lateral and one postero-medial. The number of postero-medial Mitral valve regurgitation
PM heads, in the general population, varies from one to three
(179). PMs are vital to mitral valve and LV correct functions Abnormality in any part of mitral valve complex, preventing
(124). normal leaflets cooptation, will result in mitral regurgitation
(MR). MR is now easily grouped and explained by Carpen-
tier’s classification according to leaflet movement: normal
Structure and cell types leaflet movement with annular dilatation or leaflet perfora-
The mitral valve displays a structure consisting in three highly tion (type I); excessive leaflet movement (type II); restric-
conserved stratified layers like the AV. These three layers are tive leaflet movement (type III), diastolic restriction such as
named as atrialis, which constitutes the flow side of the valve, rheumatic disease (IIIa) and systolic restriction as in func-
spongiosa in the middle of valve structure and fibrosa on tional disease (IIIb) (31). Causes of MR are classified into
the opposite surface of flow side and structurally continuous ischemic, due to coronary artery disease and nonischemic.
with the chordae tendineae (112). The atrialis layer presents Most common nonischemic causes of MR are degenerative,
a conspicuous amount of elastin fibers in radially oriented in developed countries, and rheumatic, in developing coun-
architecture, opposing to shear forces occurring on the flow tries (86). Other nonischemic causes are infective endocardi-
side of the leaflet when mitral valve is open. The fibrosa layer tis, cardiomyopathies, inflammatory diseases, drug-induced,
is composed by collagen fibers, resisting to the strong tensile traumatic, and congenital MR (21, 52).
forces acting when the valve is closed. The collagen structures Doppler echocardiography is the main method for assess-
allow the flexure during valve opening/closing cycles. Finally, ment of patients with MR. However, chest X-ray and electro-
the spongiosa layer is rich in proteoglycan and glycosamino- cardiogram might give clues for MR diagnosis. Nonetheless,
glycan. It is able to resist the compressive forces on closed 2D echocardiography is considered the gold standard to con-
valve and provides nutrients to the valve cells by promoting firm and quantify the lesion severity (Table 2), valve and
fluid movement during flexure. Thus, the ECM composition sub-valvular morphology (Fig. 6). 2D echocardiography is
of all the three valve layers is crucial because supports the able to define the possible causes and to assess the function,
leaflet mechanics. In the same manner, also the cellular com- giving the parameters for a good surgical repair.
ponents of mitral valves are widely distributed, according Functional MR induced by annular dilatation, papillary
to their function in the valve structure. Therefore, the sur- muscle displacement, and chordal tethering will cause LV
face of heart valves is covered by VECs, which are CD31 remodeling (1). Degenerative MR is usually related to mitral
positive and perform functions similar to vascular endothe- valve prolapse (MVP) with leaflet thickening and chordae
lial cells (153). Nonetheless, VECs are distinct from vascu- elongation which could be restricted to only one segment,
lar endothelium for several features, such as the perpendic- fibroelastic deficiency, or more diffuse disease affecting multi-
ular alignment in flow environments and the expression of segments, Barlow’s disease (2).
genes associated with chondrogenesis (23, 25). Smooth mus- MVP is a common valve pathology and has an estimated
cle cells are specifically characterized by the expression of prevalence of 2% to 3% in the general population, affect-
smooth muscle myosin heavy chain and arranged as a thin ing more than 176 million people worldwide (45). Despite
layer of cells near the flow side of the valve leaflet (13, 46). the pathology was first described in the late 1800s (45), no

808 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

Table 2 Echocardiography Criteria for the Identification of Mitral Since the mesenchymal cell response to altered mechanical
Valve Regurgitation stimuli lead to VIC activation, therefore causing ECM remod-
eling, VICs are the main cell type involved in the development
Mitral regurgitation and progression of mitral degeneration (167).
The biomechanical stimuli on heart valves are tension,
Qualitative shear stress, compression, and flexure (24, 180). These stim-
Valve morphology Flail leaflet/ruptured papillary uli may be associated with the initiation and extension of the
muscle/large coaptation defect valve regurgitation, however, little is known about VIC
Color flow regurgitant jet Very large central jet or eccentric mechanosensoring and mechanotransduction mechanisms
jet adhering, swirling, and and their associated signaling pathways.
reaching the posterior wall of the
left atrium The considerable role played by high tension in the patho-
logic process of degenerative valve diseases has been recently
CW signal of regurgitant jet Dense/triangular
highlighted. Hypertension or connective tissue disorders, such
Other Large flow convergence zonea as Marfan’s syndrome (19,189,216), are well-known risk fac-
Semi-quantitative tors for human MR. Because the increased tensile forces, in
Vena contracta width (mm) ≥7 (>8 for biplane)b hypertension, and impaired ECM homeostasis, in connective
tissue disorders, lead to VIC activation, causing their phe-
Upstream vein flow Systolic pulmonary vein flow
reversal notypic switch. In turn, this lead to increased glycosamino-
glycan synthesis, altered proteoglycan expression, as well
Inflow E-wave dominant ≥1.5 m/sc
as increased expression of α-SMA, embryonic SMMHC,
Other TVI mitral/TVI aortic >1.4 MMP-1, MMP-13, and cathepsin K (66, 106).
Quantitative Recently, in vivo experiments, on a sheep model, showed
Primary Secondaryd that mitral leaflet affected by turbulent flow easily under-
went myxomatous degeneration (195). Moreover, in vitro
EROA (mm2 ) ≥40 ≥20
cocultures, of VECs and VICs, suggested that nonpatholog-
R Vol (mL/beat) ≥60 ≥30 ical VECs are able to inhibits VICs activation and normal
+ enlargement of cardiac LV, LA “quiescent” VICs are able to limit VEC EndMT (22). How-
chambers/vessels ever, VICs are mostly reactive to tensile forces, due to their
association with the ECM, whereas VECs are responsive to
CW, continuous wave; EROA, effective regurgitant orifice area; LA, left shear stress.
atrium; LV, left ventricle; R Vol, regurgitant volume; TVI, time–velocity
integral.
a At a Nyquist limit of 50 to 60 cm/s.
Chemical stimuli
b For average between apical four- and two-chamber views.
c In the absence of other causes of elevated left atrial pressure and of
Overall, circulating plasmatic compounds and/or released
mitral stenosis. molecules from circulating cells represent the chemical stim-
d Different thresholds are used in secondary MR where an EROA
>20 mm2 and regurgitant volume >30 mL identify a subset of patients uli that are able to trigger any pathological response, when
at increased risk of cardiac events. find themselves in proximity to the valve structures. Among
these compounds, serotonin has been recently investigated as
major risk factor has been identified (190) and the initiating a possible stimulus for in vivo MR (156).
mechanisms driving MVP are not fully understood (192). Typically, circulating serotonin plasma levels are low
because of its rapid removal carried out either by platelet, by
liver or by lung cells. When serotonin synthesis overcomes
Cellular mechanisms these mechanisms of plasmatic clearance, circulating sero-
To date, the literature does not provide specific stimuli trig- tonin levels become sufficiently high to induce valvulopathy
gering mitral valve degeneration. The knowledge available (67). Gustafsson et al. (67) have shown high circulating sero-
provides only possible initiating signaling pathways concern- tonin levels in carcinoid syndrome, which in turn induced
ing mechanical and chemical stimuli, which in turn define the human myxomatous valvulopathy associated with exuber-
activation of specific transcription factors and downstream ant proteoglycans and glycosaminoglycans deposition. More-
gene expression modulation (Fig. 7). over, exogenous serotonin as well as other chemical stimuli
could be delivered to heart valves and provide a detrimental
chemical stimulus. A possible triggering effect, of degenera-
Mechanical stimuli
tive mitral valve pathology, is represented by VEC dysfunc-
As previously mentioned, degenerative mitral valve disease is tion that allows increased adherence of platelets to the surface
accounted for the dysregulation of ECM homeostasis. It is rea- of mitral leaflets (39, 194). VEC denudation resulting from
sonable that the abnormal biomechanical loading represents shear stress has been previously described and is commonly
the main mechanism involved in valve disease development. associated with MR (39, 194).

Volume 7, July 2017 809


Valve Pathology Consequences Comprehensive Physiology

Figure 6 Echocardiography of healthy and prolapsed mitral valve in systole. 2D-echocardiography of a healthy (left) and
regurgitant (central and right) mitral valve in long axis view. The left echocardiography image shows the correct alignment
(coaptation—red arrow) of healthy, thin mitral valve leaflets. The central echocardiography shows impaired closure (red
star). The right color-Doppler image shows the regurgitant volume toward the left atrium.

Molecular mediators distinct proteins, six of them belonging to the superfamily


The serotonin hypothesis of Gq-protein-coupled receptors. The binding of serotonin
with its receptors activates phospholipase C (210), which in
The serotonin formation is provided by two different isoforms turn activates the MAPK extracellular signaling-regulated
of the tryptophan hydroxylase (TPH-1 and -2), which is also kinase (Erk) trigger the proliferative signaling (107). Several
the rate-limiting step in the synthesis of this neurotransmitter studies in different cell types, including VICs, reported that
(205). The serotonin receptor family is constituted by seven serotonin-mediated activation of phospho-Erk (p-Erk) and its

Figure 7 Cellular features of mitral valve regurgitation. The cartoon depicts the main cellular mechanisms involved
in the pathogenesis of mitral valve regurgitation. (A) The endothelial damage leading to EndMT of VECs into myofi-
broblas (MyoFB). (B) The fragmentation of elastin in the atrialis layer. (C) The differentiation of VICs into MyoFB
leading to fibrosis (collagen and glycosaminoglycans deposition depicted in blue in the spongiosa layer). (D) The
collagen fragmentation in the fibrosa layer. Adapted with permission from (109).

810 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

subsequent nuclear translocation lead to Rho kinase-mediated serotonin induces TGF-β1 RNA expression in cultured ovine
mitogenesis (115, 210). As previously mentioned, high cir- aortic VICs by Gq-dependent signaling (90) and by Erk phos-
culating serotonin can induce valve disease, both in humans phorylation in mesangial cells (64).
and in murine models. Indeed, carcinoid syndrome patients, All these results display a strong interplay between sero-
displaying high level of plasmatic serotonin, are more prone tonin signaling, TGF-β pathway, and the expression of effec-
to develop valvulopathy (67). In particular, carcinoid-derived tor genes and proteins involved in myxomatous mitral valve
valvulopathy is characterized by specific pathological degeneration.
feature, such as myofibroblast proliferation, fibrosis, excess
deposition of ECM and myxomatous degeneration (67). Sim-
Development-related gene pathways
ilar valvulopathy occurs in rat model undergoing long-term
injections of serotonin (49, 68). Development-related pathways are highly conserved among
Several drugs upregulating serotonin levels (e.g., anorec- vertebrates, specifically, transcription factor signaling and
tic drugs) or agonist of serotonin receptor (e.g., Parkinson’s their downstream genes. However, valve development reg-
disease treatments) have been shown to be strictly implicated ulating pathways are also involved in other processes, such
in mitral valve disease (184,218). Strong evidence specifically as arteries, cartilage, bone, and tendon formation. Among
links the activation of the serotonin 2B receptor (5HT2BR) these, we will focus our attention especially on BMP2-Sox 9,
in serotoninergic valve disease (54, 80, 177, 178). Constant Wnt-periostin, and Notch 1 (28, 38, 126, 127).
administration of serotonin in a rat model lead to an increased BMPs are members of the TGF-β superfamily and their
expression of 5HT2BR and a parallel decreased level of sero- principal effectors are SMAD proteins, which are activated
tonin transporter (SERT) (51). Indeed, a mouse model lacking by phosphorylation and carry out their functions already
SERT display severe mitral regurgitation, possibly due to the mentioned before (38, 53). Nevertheless, BMPs are consider-
decreased reuptake of serotonin (129). ably involved in development and after-birth chondrogenesis
In 2006, an important study on dogs, firstly reported an through sex determining region Y-box 9 (Sox 9) activation
increased 5HT2BR transcript level in the context of mitral (53, 215). The transcription factor Sox 9 mediates the expres-
valve disease development and progression (155). More- sion of several structural genes including aggrecan, collagen
over, in this animal model, it was also shown a concomi- type II and N-cadherin, all important component of valve
tant decrease of SERT and an overactivation of Erk signaling ECM (215).
(48, 186). Furthermore, Disatian and colleagues reported not BMP2 is thought to play an important role in the devel-
only a several-fold increase in the expression of TPH type 1, opment of the spongiosa layer during heart valve formation
both in human and in a canine mitral valve disease model, but and ECM stratification (38, 112).
also a raise in the expression of this enzyme already in the Experimental evidences showed both in animal models
early-stages of mitral valve disease (47, 48). Furthermore, it and in human the colocalization of Sox 9 with aggrecan,
has also been shown that inhibition of TPH type 1 or 5HT2BR collagen type II and BMP2 in impaired mitral valves (122).
diminishes expression of strain-induced myxomatous proteins In addition, all these molecules are well known to be involved
in mitral valve models, supporting the serotonin implication in in chondrogenesis process and have been found upregulated
the mechanisms involved in mitral valve degeneration (106). in MVP human specimens (182).
Consistently, these important results support the hypoth- Since chondrogenesis process and myxomatous valve dis-
esis of local serotonin as an initiating stimulus in mitral valve play large morphologic similarities, BMP-Sox 9 signaling is a
degeneration. potential candidate pathway to better understand degenerative
mitral valve disease and possibly identify potential therapeu-
tic targets.
TGF-𝛽 signaling
Wnt signaling is highly involved in embryogenesis and is
Among the growth factor superfamily, TGF-β1, -β2, -β3 iso- activated by the specific binding of Wnt with Frizzled (Fz)
forms, BMPs, inhibins and activins constitute the main sub- receptor and stabilized by low-density lipoprotein receptor-
families (84). In particular, the overexpression of TGF-β has related protein (Lrp)-5 or Lrp-6 (116, 123). The activation of
been demonstrated to be strongly involved in several cardiac the Wnt canonical pathway leads to a reduced degradation of
diseases based on fibrosis (100, 102), carcinoid heart disease β-catenin that translocates to the nucleus and activates the T
(204) and valvular pathologies such as AVS as well as MR cell-specific transcription factor/lymphoid enhancer-binding
(61, 70). In particular, several studies have shown high lev- factor 1 (TCF/LEF). Wnt signaling regulates multiple path-
els of TGF-β and overexpression of TGF-βR1 and 2 in MR, ways, in particular, development and postnatal chondrogenic
both in humans (61, 70) and in vivo models (12, 48, 82, 150). and osteogenic processes. Moreover, this axis is also involved
Indeed, these observation have been well characterized and in endochondral bone formation and in maintaining bone den-
TGF-β has been recognized as driving factor of myofibrob- sity (72, 123). Interestingly, it has been recently shown that
lastic activation of VICs under tensile stress (130, 203). Lrp-5 regulates bone formation by the inhibition of TPH type
Many studies have also reported a strong link between 1 and the synthesis of serotonin, which in turn directly inhibits
serotonin and TGF-β, in vitro. It has been shown that osteoclastogenesis (211). Furthermore, a deep implication of

Volume 7, July 2017 811


Valve Pathology Consequences Comprehensive Physiology

Lrp-5-Wnt-β-catenin signaling in human aortic valve degen- AngII is a little peptide resulting from the enzymatic con-
eration has been shown (28, 168). A similar osteogenic phe- version of angiotensin I and performed by angiotensin con-
notypic switch of VICs, in aortic valve disease, through the verting enzyme (ACE). The binding of AngII with the AT1R
expression of bone phenotype markers such as osteopontin leads to the activation of phospholipase C, the generation of
and osteocalcin has also been shown in degenerative mitral the inositol trisphosphate (IP3), a second messengers. Con-
valve disease. In particular, the endochondral bone formation flicting evidence in the direct role for AngII in mitral valve
has been hypothesized to be involved in myxomatous degen- impairment have been collected. AngII was shown to strongly
eration of mitral valves (28). stimulate collagen synthesis in human VICs, in vitro (69). It
Given the fact that osteogenesis and chondrogenesis can- has been reported a weak relation between AngII and ACE
not be active at the same time, it has been speculated that inhibitor, in human mitral valves, contrarily to a strong asso-
Wnt signaling could reinforce a local (mitral valve) serotonin ciation in rat mitral valves (141). Moreover, AngII has been
synthesis, thus enhancing chondrogenesis (153). shown to increase the expression of TGF-β1 in human cardiac
The Notch signaling pathway, commonly shared by a fibroblasts (104) and that the ACE inhibition has been shown
number of multicellular organisms, is based on the specific to reduce TGF-β3 synthesis in VIC (150). Taken these results
binding of several ligands with transmembrane Notch recep- together, a possible role for AngII in MR may be hypothe-
tors (Notch 1-4), which, once activated, induced proteolytic sized, but further investigation should unravel this issue.
cleavage and subsequent release of its intracellular domain
(38). The intracellular domain translocates into the nucleus to
modulate downstream gene expression. Likewise Notch, also Pathophysiology
its ligands are transmembrane proteins, thus this peculiar path- MR can be divided into two different entities, acute and
way require cell-to-cell contact to be properly activated. For chronic, which have different physiological consequences.
this reason, Notch signaling is strongly related to the cellular In addition, factors that affect the degree of MR are the lesion
network formation and also to the epithelial-to-mesenchymal severity (regurgitant orifice area), the driving force (LV sys-
transition that is crucial in the early stage of valve formation tolic pressure) and the left atrial compliance (57). It is worth
(121, 199). In addition, localized activation of Notch signal- mentioning that the principal determinant of MR is the regur-
ing has been shown to play a pivotal role in valve polarity gitant orifice area (Fig. 8).
establishment and in mitral valve atrialis layer development
(38). This signaling pathway has also been demonstrated to
be susceptible to the shear stress (38). As previously men-
Acute MR
tioned, mutations in Notch gene are present in congenital In acute MR, there is no time for compensatory changes,
valve (i.e., bicuspid aortic valve) and in calcific aortic valve due to the regurgitant volume into a small non-compliant
disease, in human (59, 121). Unfortunately, to date, there are LA. The regurgitation will cause a significant increase in LA
no experimental evidence demonstrating the involvement of pressure and the pressure overload will be transmitted to the
Notch signaling in MR, thus further studies are necessary to pulmonary veins and subsequently to the pulmonary artery.
validate this hypothesis. Eventually, patients with MR might develop acute pulmonary
edema when the LA and pulmonary pressure increase to a
point that exceeds oncotic pressure (17, 139). In worst cases,
edema is followed by hemorrhage because the pressure gra-
Other pathways
dient is high enough to permit the extravasation of erythro-
NO production, regulated by the enzyme family of NO syn- cytes. Another pathophysiological change could occur when
thases (NOS), is well known to play a central role in a vari- increase in cardiac output is needed (e.g., during exercise).
ety of biological functions including endothelium-dependent The rapid increase in LA pressure will lead the activation
vasodilation. The three NOS isoforms include eNOS, which of pulmonary vein baroreceptor, which will result in tissue
regulates endothelial NO signaling by calcium/calmodulin, and cerebral acidosis, causing exertional dyspnea. Later on,
neuronal NOS (nNOS) and inducible NOS (iNOS), which is patients might develop dyspnea at mild exertion because of
activated in a variety of pathological processes (151). Release chronic LA pressure overload (17, 139). At the same time,
of NO and its subsequent activation leads to cyclic guanosine MR will lead to volume overload in the LV, increasing its
monophosphate (cGMP) generation, which in turn activates distension and wall stress. Finally, LV failure occurs as a
cGMP-dependent protein kinase G that contribute to smooth result of increase wall stress with no time for compensatory
muscle relaxation, platelet function and cell division (151). hypertrophy resulting in peripheral vascular collapse or acute
The implementation of a porcine model of mildly myxo- pulmonary edema, or both (174).
matous mitral valve degeneration demonstrated the capability
to release NO by mitral VECs (136, 188) after calcium influx
Chronic MR
and eNOS activation (202, 203). In addition, the activity of
ADP, bradykinin, and thrombin-induced NO levels increase In chronic MR, significant regurgitant volume, from LV to LA
in myxomatous mitral valve (137). during systole, increases LA pressure subsequently leading

812 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

Figure 8 Mitral valve regurgitation pathophysiology. Phenotype of regurgitant mitral valve and its involvement in cardiac diseases. In
the left side pictures of normal and regurgitant mitral valve in systole and diastole. In the closed regurgitant mitral valve picture is clear
the improper coaptation of valve leaflets. In the right side of the figure are mentioned the main cardiac pathologies caused by mitral valve
regurgitation.

to LA dilatation to accommodate the excessive volume. The Chronic MR


early cardiac output compensation, in the acute phase, will be
gradually replaced by LV dilatation and eccentric hypertro- Mild and moderate MR will only require a series of clini-
phy to increase LV compliance and volume. This will cause cal examination and echocardiography follow up to evaluate
an increase in the afterload. Such compensatory changes hap- patient status and only if symptoms occur surgical interven-
pen through remodeling of the extramyocardial matrix. In tion will be evaluated. On the other hand, patient with mod-
particular, collagen fiber disruption allows the rearrangement erate to severe MR will require a closer attention, examining
and slippage of myocardial fibers causing chamber dilata- these patients yearly. Unfortunately, like all other valve dis-
tion. Eccentric hypertrophy develops by accumulating new eases, there is no medical therapy that could slow the pro-
sarcomeres in series in the LV, which increases the length of gression of MR. However pharmacological therapy is used
cardiomyocytes without increasing length of sarcomere pre- with functional MR associated with LV dysfunction. In this
serving the effect of preload (57). Myocardial contractility case, angiotensin-converting enzyme inhibitors, β-blockers,
will also be affected by the dilatation, due to the increase in and biventricular pacing have been shown to improve sur-
wall stress, leading to ejection fraction drop (57). Elevation vival rates reducing the degree of MR (20, 29, 113). In any
of LA pressure, through abnormal diastolic pressure gradient, event, when patients develop severe MR and symptoms occur,
will lead to pulmonary venous hypertension and reduction in only surgical intervention will completely restore normal life
cardiac output inducing pulmonary vascular resistance. This expectancy.
will cause a further reduction of cardiac output, due to the
secondary obstruction caused by the developed pulmonary
hypertension, which will lead to RV pressure overload and
eventually an overt RV failure (87, 101). Both diastolic and
Conclusion
systolic dysfunction will cause eventually heart failure, which Calcific and degenerative valve pathologies are an under-
conveys a decrease in survival and require immediate surgical recognized heath problem. The aging population and the lack
intervention. of effective pharmacological therapies will increase drasti-
cally the burden on the global health system over the next
decades. Up to now, only clinical examination and cardiac
Management of MR imaging recognize these pathologies and, most of the time,
these diseases are recognized only at advanced stages due to
Acute MR
symptoms occurrence. Once patients experience any symp-
Mortality rates are extremely high without surgical inter- toms, the only available options are based on surgical interven-
vention. However, patient with acute MR will also benefit tion, from percutaneous to open heart surgery. However, these
from hemodynamically stabilization by afterload reduction procedures do not cure the diseases, but treat only the symp-
accomplished either by pharmacological (e.g., vasodilators) toms, leaving the underling pathological processes untouched.
or by mechanical treatments (e.g., intra-aortic balloon Further studies that will establish innovative, low-cost screen-
pump). ing procedure will allow the identification of affected patients

Volume 7, July 2017 813


Valve Pathology Consequences Comprehensive Physiology

at earlier stages and, in turn, the possibility to find and inter- Task Force on Practice Guidelines (writing committee to revise the
1998 Guidelines for the Management of Patients With Valvular Heart
vene on the triggers of these debilitating pathologies. Disease): Developed in collaboration with the Society of Cardiovas-
cular Anesthesiologists: endorsed by the Society for Cardiovascular
Angiography and Interventions and the Society of Thoracic Surgeons.
Acknowledgements 18.
Circulation 114: e84-231, 2006.
Bostrom KI, Rajamannan NM, Towler DA. The regulation of valvular
and vascular sclerosis by osteogenic morphogens. Circ Res 109: 564-
The authors thank the Fondazione Gigi and Pupa Ferrari 577, 2011.
ONLUS for the financial support. The authors thank Giovanni 19. Boudoulas H. Etiology of valvular heart disease. Expert Rev Cardiovasc
Ther 1: 523-532, 2003.
Fumagalli. 20. Breithardt OA, Sinha AM, Schwammenthal E, Bidaoui N, Markus
KU, Franke A, Stellbrink C. Acute effects of cardiac resynchronization
therapy on functional mitral regurgitation in advanced systolic heart
References 21.
failure. J Am Coll Cardiol 41: 765-770, 2003.
Bursi F, Enriquez-Sarano M, Jacobsen SJ, Roger VL. Mitral regurgi-
tation after myocardial infarction: a review. Am J Med 119: 103-112,
1. Acker MA. Should moderate or greater mitral regurgitation be repaired 2006.
in all patients with LVEF <30%? Mitral valve repair in patients 22. Butcher JT, Nerem RM. Valvular endothelial cells regulate the phe-
with advanced heart failure and severe functional mitral insufficiency notype of interstitial cells in co-culture: Effects of steady shear stress.
reverses left ventricular remodeling and improves symptoms. Circ Tissue Eng 12: 905-915, 2006.
Heart Fail 1: 281-284, 2008. 23. Butcher JT, Penrod AM, Garcia AJ, Nerem RM. Unique morphology
2. Adams DH, Rosenhek R, Falk V. Degenerative mitral valve regurgita- and focal adhesion development of valvular endothelial cells in static
tion: Best practice revolution. Eur Heart J 31: 1958-1966, 2010. and fluid flow environments. Arterioscler Thromb Vasc Biol 24: 1429-
3. Aikawa E, Nahrendorf M, Figueiredo JL, Swirski FK, Shtatland T, 1434, 2004.
Kohler RH, Jaffer FA, Aikawa M, Weissleder R. Osteogenesis asso- 24. Butcher JT, Simmons CA, Warnock JN. Mechanobiology of the aortic
ciates with inflammation in early-stage atherosclerosis evaluated by heart valve. J Heart Valve Dis 17: 62-73, 2008.
molecular imaging in vivo. Circulation 116: 2841-2850, 2007. 25. Butcher JT, Tressel S, Johnson T, Turner D, Sorescu G, Jo H, Nerem
4. Aikawa E, Nahrendorf M, Sosnovik D, Lok VM, Jaffer FA, Aikawa M, RM. Transcriptional profiles of valvular and vascular endothelial cells
Weissleder R. Multimodality molecular imaging identifies proteolytic reveal phenotypic differences: Influence of shear stress. Arterioscler
and osteogenic activities in early aortic valve disease. Circulation 115: Thromb Vasc Biol 26: 69-77, 2006.
377-386, 2007. 26. Byon CH, Sun Y, Chen J, Yuan K, Mao X, Heath JM, Anderson PG,
5. Al-Aly Z, Shao JS, Lai CF, Huang E, Cai J, Behrmann A, Cheng Tintut Y, Demer LL, Wang D, Chen Y. Runx2-upregulated recep-
SL, Towler DA. Aortic Msx2-Wnt calcification cascade is regulated by tor activator of nuclear factor kappaB ligand in calcifying smooth
TNF-alpha-dependent signals in diabetic Ldlr−/− mice. Arterioscler muscle cells promotes migration and osteoclastic differentiation of
Thromb Vasc Biol 27: 2589-2596, 2007. macrophages. Arterioscler Thromb Vasc Biol 31: 1387-1396, 2011.
6. Alexopoulos A, Bravou V, Peroukides S, Kaklamanis L, Varakis J, 27. Cagirci G, Cay S, Canga A, Karakurt O, Yazihan N, Kilic H, Topaloglu
Alexopoulos D, Papadaki H. Bone regulatory factors NFATc1 and S, Aras D, Demir AD, Akdemir R. Association between plasma asym-
Osterix in human calcific aortic valves. Int J Cardiol 139: 142-149, metrical dimethylarginine activity and severity of aortic valve stenosis.
2010. J Cardiovasc Med (Hagerstown) 12: 96-101, 2011.
7. Ankeny RF, Thourani VH, Weiss D, Vega JD, Taylor WR, Nerem RM, 28. Caira FC, Stock SR, Gleason TG, McGee EC, Huang J, Bonow
Jo H. Preferential activation of SMAD1/5/8 on the fibrosa endothelium RO, Spelsberg TC, McCarthy PM, Rahimtoola SH, Rajamannan NM.
in calcified human aortic valves—association with low BMP antago- Human degenerative valve disease is associated with up-regulation of
nists and SMAD6. PLoS One 6: e20969, 2011. low-density lipoprotein receptor-related protein 5 receptor-mediated
8. Ann EJ, Kim HY, Choi YH, Kim MY, Mo JS, Jung J, Yoon JH, Kim bone formation. J Am Coll Cardiol 47: 1707-1712, 2006.
SM, Moon JS, Seo MS, Hong JA, Jang WG, Shore P, Komori T, Koh 29. Capomolla S, Febo O, Gnemmi M, Riccardi G, Opasich C, Caporotondi
JT, Park HS. Inhibition of Notch1 signaling by Runx2 during osteoblast A, Mortara A, Pinna GD, Cobelli F. Beta-blockade therapy in chronic
differentiation. J Bone Miner Res 26: 317-330, 2011. heart failure: Diastolic function and mitral regurgitation improvement
9. Annes JP, Munger JS, Rifkin DB. Making sense of latent TGFbeta by carvedilol. Am Heart J 139: 596-608, 2000.
activation. J Cell Sci 116: 217-224, 2003. 30. Carabello BA, Paulus WJ. Aortic stenosis. Lancet 373: 956-966, 2009.
10. Arishiro K, Hoshiga M, Negoro N, Jin D, Takai S, Miyazaki M, Ishihara 31. Carpentier A. Cardiac valve surgery—the “French correction”. J Thorac
T, Hanafusa T. Angiotensin receptor-1 blocker inhibits atherosclerotic Cardiovasc Surg 86: 323-337, 1983.
changes and endothelial disruption of the aortic valve in hypercholes- 32. Carpentier AF, Lessana A, Relland JY, Belli E, Mihaileanu S, Berrebi
terolemic rabbits. J Am Coll Cardiol 49: 1482-1489, 2007. AJ, Palsky E, Loulmet DF. The “physio-ring”: An advanced concept in
11. Askevold ET, Gullestad L, Aakhus S, Ranheim T, Tonnessen T, Solberg mitral valve annuloplasty. Ann Thorac Surg 60: 1177-1185; discussion
OG, Aukrust P, Ueland T. Secreted Wnt modulators in symptomatic 1185-1176, 1995.
aortic stenosis. J Am Heart Assoc 1: e002261, 2012. 33. Cary T, Pearce J. Aortic stenosis: Pathophysiology, diagnosis, and med-
12. Aupperle H, Marz I, Thielebein J, Schoon HA. Expression of trans- ical management of nonsurgical patients. Crit Care Nurse 33: 58-72,
forming growth factor-beta1, -beta2 and -beta3 in normal and diseased 2013.
canine mitral valves. J Comp Pathol 139: 97-107, 2008. 34. Chakraborty S, Wirrig EE, Hinton RB, Merrill WH, Spicer DB, Yutzey
13. Barber JE, Kasper FK, Ratliff NB, Cosgrove DM, Griffin BP, Vesely I. KE. Twist1 promotes heart valve cell proliferation and extracellular
Mechanical properties of myxomatous mitral valves. J Thorac Cardio- matrix gene expression during development in vivo and is expressed in
vasc Surg 122: 955-962, 2001. human diseased aortic valves. Dev Biol 347: 167-179, 2010.
14. Basta G, Corciu AI, Vianello A, Del Turco S, Foffa I, Navarra T, 35. Chang K, Weiss D, Suo J, Vega JD, Giddens D, Taylor WR, Jo H.
Chiappino D, Berti S, Mazzone A. Circulating soluble receptor for Bone morphogenic protein antagonists are coexpressed with bone mor-
advanced glycation end-product levels are decreased in patients with phogenic protein 4 in endothelial cells exposed to unstable flow in
calcific aortic valve stenosis. Atherosclerosis 210: 614-618, 2010. vitro in mouse aortas and in human coronary arteries: Role of bone
15. Baumgartner H, Hung J, Bermejo J, Chambers JB, Evangelista A, morphogenic protein antagonists in inflammation and atherosclerosis.
Griffin BP, Iung B, Otto CM, Pellikka PA, Quinones M. Echocardio- Circulation 116: 1258-1266, 2007.
graphic assessment of valve stenosis: EAE/ASE recommendations for 36. Chen D, Zhao M, Mundy GR. Bone morphogenetic proteins. Growth
clinical practice. Eur J Echocardiogr 10: 1-25, 2009. Factors 22: 233-241, 2004.
16. Bennett BJ, Scatena M, Kirk EA, Rattazzi M, Varon RM, Averill M, 37. Chen JH, Simmons CA. Cell-matrix interactions in the pathobiology of
Schwartz SM, Giachelli CM, Rosenfeld ME. Osteoprotegerin inacti- calcific aortic valve disease: Critical roles for matricellular, matricrine,
vation accelerates advanced atherosclerotic lesion progression and cal- and matrix mechanics cues. Circ Res 108: 1510-1524, 2011.
cification in older ApoE−/− mice. Arterioscler Thromb Vasc Biol 26: 38. Combs MD, Yutzey KE. Heart valve development: Regulatory networks
2117-2124, 2006. in development and disease. Circ Res 105: 408-421, 2009.
17. Bonow RO, Carabello BA, Kanu C, de Leon AC, Jr., Faxon DP, Freed 39. Corcoran BM, Black A, Anderson H, McEwan JD, French A, Smith P,
MD, Gaasch WH, Lytle BW, Nishimura RA, O’Gara PT, O’Rourke RA, Devine C. Identification of surface morphologic changes in the mitral
Otto CM, Shah PM, Shanewise JS, Smith SC, Jr., Jacobs AK, Adams valve leaflets and chordae tendineae of dogs with myxomatous degen-
CD, Anderson JL, Antman EM, Fuster V, Halperin JL, Hiratzka LF, eration. Am J Vet Res 65: 198-206, 2004.
Hunt SA, Nishimura R, Page RL, Riegel B. ACC/AHA 2006 guidelines 40. Cowell SJ, Newby DE, Prescott RJ, Bloomfield P, Reid J, Northridge
for the management of patients with valvular heart disease: A report DB, Boon NA. A randomized trial of intensive lipid-lowering therapy
of the American College of Cardiology/American Heart Association in calcific aortic stenosis. N Engl J Med 352: 2389-2397, 2005.

814 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

41. Cucoranu I, Clempus R, Dikalova A, Phelan PJ, Ariyan S, Dikalov S, 65. Guerraty MA, Grant GR, Karanian JW, Chiesa OA, Pritchard WF,
Sorescu D. NAD(P)H oxidase 4 mediates transforming growth factor- Davies PF. Hypercholesterolemia induces side-specific phenotypic
beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. changes and peroxisome proliferator-activated receptor-gamma path-
Circ Res 97: 900-907, 2005. way activation in swine aortic valve endothelium. Arterioscler Thromb
42. Cushing MC, Liao JT, Anseth KS. Activation of valvular interstitial Vasc Biol 30: 225-231, 2010.
cells is mediated by transforming growth factor-beta1 interactions with 66. Gupta V, Werdenberg JA, Lawrence BD, Mendez JS, Stephens EH,
matrix molecules. Matrix Biol 24: 428-437, 2005. Grande-Allen KJ. Reversible secretion of glycosaminoglycans and pro-
43. D’Amelio P, Isaia G, Isaia GC. The osteoprotegerin/RANK/RANKL teoglycans by cyclically stretched valvular cells in 3D culture. Ann
system: A bone key to vascular disease. J Endocrinol Invest 32: 6-9, Biomed Eng 36: 1092-1103, 2008.
2009. 67. Gustafsson BI, Hauso O, Drozdov I, Kidd M, Modlin IM. Carcinoid
44. Dal-Bianco JP, Levine RA. Anatomy of the mitral valve apparatus: heart disease. Int J Cardiol 129: 318-324, 2008.
Role of 2D and 3D echocardiography. Cardiol Clin 31: 151-164, 2013. 68. Gustafsson BI, Tommeras K, Nordrum I, Loennechen JP, Brunsvik A,
45. Delling FN, Vasan RS. Epidemiology and pathophysiology of mitral Solligard E, Fossmark R, Bakke I, Syversen U, Waldum H. Long-term
valve prolapse: New insights into disease progression, genetics, and serotonin administration induces heart valve disease in rats. Circulation
molecular basis. Circulation 129: 2158-2170, 2014. 111: 1517-1522, 2005.
46. Disatian S, Ehrhart EJ, III, Zimmerman S, Orton EC. Interstitial cells 69. Hafizi S, Taylor PM, Chester AH, Allen SP, Yacoub MH. Mitogenic
from dogs with naturally occurring myxomatous mitral valve disease and secretory responses of human valve interstitial cells to vasoactive
undergo phenotype transformation. J Heart Valve Dis 17: 402-411; agents. J Heart Valve Dis 9: 454-458, 2000.
discussion 412, 2008. 70. Hagler MA, Hadley TM, Zhang H, Mehra K, Roos CM, Schaff HV,
47. Disatian S, Lacerda C, Orton EC. Tryptophan hydroxylase 1 expres- Suri RM, Miller JD. TGF-beta signalling and reactive oxygen species
sion is increased in phenotype-altered canine and human degenerative drive fibrosis and matrix remodelling in myxomatous mitral valves.
myxomatous mitral valves. J Heart Valve Dis 19: 71-78, 2010. Cardiovasc Res 99: 175-184, 2013.
48. Disatian S, Orton EC. Autocrine serotonin and transforming growth 71. Hamblin M, Chang L, Fan Y, Zhang J, Chen YE. PPARs and the
factor beta 1 signaling mediates spontaneous myxomatous mitral valve cardiovascular system. Antioxid Redox Signal 11: 1415-1452, 2009.
disease. J Heart Valve Dis 18: 44-51, 2009. 72. Hartmann C, Tabin CJ. Dual roles of Wnt signaling during chondroge-
49. Droogmans S, Roosens B, Cosyns B, Degaillier C, Hernot S, Weyt- nesis in the chicken limb. Development 127: 3141-3159, 2000.
jens C, Garbar C, Caveliers V, Pipeleers-Marichal M, Franken PR, 73. Hein S, Arnon E, Kostin S, Schonburg M, Elsasser A, Polyakova V,
Bossuyt A, Schoors D, Lahoutte T, Van Camp G. Dose dependency Bauer EP, Klovekorn WP, Schaper J. Progression from compensated
and reversibility of serotonin-induced valvular heart disease in rats. hypertrophy to failure in the pressure-overloaded human heart: Struc-
Cardiovasc Toxicol 9: 134-141, 2009. tural deterioration and compensatory mechanisms. Circulation 107:
50. Edep ME, Shirani J, Wolf P, Brown DL. Matrix metalloproteinase 984-991, 2003.
expression in nonrheumatic aortic stenosis. Cardiovasc Pathol 9: 281- 74. Heistad DD, Wakisaka Y, Miller J, Chu Y, Pena-Silva R. Novel aspects
286, 2000. of oxidative stress in cardiovascular diseases. Circ J 73: 201-207, 2009.
51. Elangbam CS, Job LE, Zadrozny LM, Barton JC, Yoon LW, Gates LD, 75. Helske S, Lindstedt KA, Laine M, Mayranpaa M, Werkkala K, Lommi
Slocum N. 5-hydroxytryptamine (5HT)-induced valvulopathy: Com- J, Turto H, Kupari M, Kovanen PT. Induction of local angiotensin
positional valvular alterations are associated with 5HT2B receptor and II-producing systems in stenotic aortic valves. J Am Coll Cardiol 44:
5HT transporter transcript changes in Sprague-Dawley rats. Exp Toxicol 1859-1866, 2004.
Pathol 60: 253-262, 2008. 76. Helske S, Syvaranta S, Kupari M, Lappalainen J, Laine M, Lommi J,
52. Enriquez-Sarano M, Akins CW, Vahanian A. Mitral regurgitation. Turto H, Mayranpaa M, Werkkala K, Kovanen PT, Lindstedt KA. Pos-
Lancet 373: 1382-1394, 2009. sible role for mast cell-derived cathepsin G in the adverse remodelling
53. Fischer L, Boland G, Tuan RS. Wnt-3A enhances bone morphogenetic of stenotic aortic valves. Eur Heart J 27: 1495-1504, 2006.
protein-2-mediated chondrogenesis of murine C3H10T1/2 mesenchy- 77. Helske S, Syvaranta S, Lindstedt KA, Lappalainen J, Oorni K, Mayran-
mal cells. J Biol Chem 277: 30870-30878, 2002. paa MI, Lommi J, Turto H, Werkkala K, Kupari M, Kovanen PT.
54. Fitzgerald LW, Burn TC, Brown BS, Patterson JP, Corjay MH, Valen- Increased expression of elastolytic cathepsins S, K, and V and their
tine PA, Sun JH, Link JR, Abbaszade I, Hollis JM, Largent BL, Hartig inhibitor cystatin C in stenotic aortic valves. Arterioscler Thromb Vasc
PR, Hollis GF, Meunier PC, Robichaud AJ, Robertson DW. Possi- Biol 26: 1791-1798, 2006.
ble role of valvular serotonin 5-HT(2B) receptors in the cardiopathy 78. Hess OM, Villari B, Krayenbuehl HP. Diastolic dysfunction in aortic
associated with fenfluramine. Mol Pharmacol 57: 75-81, 2000. stenosis. Circulation 87: IV73-IV76, 1993.
55. Fondard O, Detaint D, Iung B, Choqueux C, Adle-Biassette H, Jarraya 79. Hofmann Bowman MA, Gawdzik J, Bukhari U, Husain AN, Toth PT,
M, Hvass U, Couetil JP, Henin D, Michel JB, Vahanian A, Jacob MP. Kim G, Earley J, McNally EM. S100A12 in vascular smooth mus-
Extracellular matrix remodelling in human aortic valve disease: The cle accelerates vascular calcification in apolipoprotein E-null mice by
role of matrix metalloproteinases and their tissue inhibitors. Eur Heart activating an osteogenic gene regulatory program. Arterioscler Thromb
J 26: 1333-1341, 2005. Vasc Biol 31: 337-344, 2011.
56. Freeman RV, Otto CM. Spectrum of calcific aortic valve disease: Patho- 80. Hofmann C, Penner U, Dorow R, Pertz HH, Jahnichen S, Horowski
genesis, disease progression, and treatment strategies. Circulation 111: R, Latte KP, Palla D, Schurad B. Lisuride, a dopamine receptor ago-
3316-3326, 2005. nist with 5-HT2B receptor antagonist properties: absence of cardiac
57. Gaasch WH, Meyer TE. Left ventricular response to mitral regur- valvulopathy adverse drug reaction reports supports the concept of a
gitation: Implications for management. Circulation 118: 2298-2303, crucial role for 5-HT2B receptor agonism in cardiac valvular fibrosis.
2008. Clin Neuropharmacol 29: 80-86, 2006.
58. Galvin KM, Donovan MJ, Lynch CA, Meyer RI, Paul RJ, Lorenz JN, 81. Huang F, Chen YG. Regulation of TGF-beta receptor activity. Cell
Fairchild-Huntress V, Dixon KL, Dunmore JH, Gimbrone MA, Jr., Falb Biosci 2: 9, 2012.
D, Huszar D. A role for smad6 in development and homeostasis of the 82. Hulin A, Deroanne CF, Lambert CA, Dumont B, Castronovo
cardiovascular system. Nat Genet 24: 171-174, 2000. V, Defraigne JO, Nusgens BV, Radermecker MA, Colige AC.
59. Garg V, Muth AN, Ransom JF, Schluterman MK, Barnes R, King IN, Metallothionein-dependent up-regulation of TGF-beta2 participates in
Grossfeld PD, Srivastava D. Mutations in NOTCH1 cause aortic valve the remodelling of the myxomatous mitral valve. Cardiovasc Res 93:
disease. Nature 437: 270-274, 2005. 480-489, 2012.
60. Gawdzik J, Mathew L, Kim G, Puri TS, Hofmann Bowman MA. Vas- 83. Hurlstone AF, Haramis AP, Wienholds E, Begthel H, Korving J, Van
cular remodeling and arterial calcification are directly mediated by Eeden F, Cuppen E, Zivkovic D, Plasterk RH, Clevers H. The Wnt/beta-
S100A12 (EN-RAGE) in chronic kidney disease. Am J Nephrol 33: catenin pathway regulates cardiac valve formation. Nature 425: 633-
250-259, 2011. 637, 2003.
61. Geirsson A, Singh M, Ali R, Abbas H, Li W, Sanchez JA, Hashim S, 84. Hyytiainen M, Penttinen C, Keski-Oja J. Latent TGF-beta binding pro-
Tellides G. Modulation of transforming growth factor-beta signaling teins: Extracellular matrix association and roles in TGF-beta activation.
and extracellular matrix production in myxomatous mitral valves by Crit Rev Clin Lab Sci 41: 233-264, 2004.
angiotensin II receptor blockers. Circulation 126: S189-197, 2012. 85. Isoda K, Matsuki T, Kondo H, Iwakura Y, Ohsuzu F. Deficiency
62. Geroldi D, Falcone C, Emanuele E. Soluble receptor for advanced of interleukin-1 receptor antagonist induces aortic valve disease in
glycation end products: From disease marker to potential therapeutic BALB/c mice. Arterioscler Thromb Vasc Biol 30: 708-715, 2010.
target. Curr Med Chem 13: 1971-1978, 2006. 86. Iung B, Baron G, Butchart EG, Delahaye F, Gohlke-Barwolf C, Levang
63. Gould KL, Carabello BA. Why angina in aortic stenosis with normal OW, Tornos P, Vanoverschelde JL, Vermeer F, Boersma E, Ravaud P,
coronary arteriograms? Circulation 107: 3121-3123, 2003. Vahanian A. A prospective survey of patients with valvular heart disease
64. Grewal JS, Mukhin YV, Garnovskaya MN, Raymond JR, Greene EL. in Europe: The Euro Heart Survey on Valvular Heart Disease. Eur Heart
Serotonin 5-HT2A receptor induces TGF-beta1 expression in mesan- J 24: 1231-1243, 2003.
gial cells via ERK: Proliferative and fibrotic signals. Am J Physiol 276: 87. Jameson AG. Pathologic physiology in mitral valve disease. Ann N Y
F922-F930, 1999. Acad Sci 118: 486-489, 1965.

Volume 7, July 2017 815


Valve Pathology Consequences Comprehensive Physiology

88. Jian B, Jones PL, Li Q, Mohler ER, III, Schoen FJ, Levy RJ. Matrix 110. Levine RA, Handschumacher MD, Sanfilippo AJ, Hagege AA, Har-
metalloproteinase-2 is associated with tenascin-C in calcific aortic rigan P, Marshall JE, Weyman AE. Three-dimensional echocardio-
stenosis. Am J Pathol 159: 321-327, 2001. graphic reconstruction of the mitral valve, with implications for the
89. Jian B, Narula N, Li QY, Mohler ER, III, Levy RJ. Progression of aortic diagnosis of mitral valve prolapse. Circulation 80: 589-598, 1989.
valve stenosis: TGF-beta1 is present in calcified aortic valve cusps and 111. Levine RA, Triulzi MO, Harrigan P, Weyman AE. The relationship of
promotes aortic valve interstitial cell calcification via apoptosis. Ann mitral annular shape to the diagnosis of mitral valve prolapse. Circula-
Thorac Surg 75: 457-465; discussion 465-456, 2003. tion 75: 756-767, 1987.
90. Jian B, Xu J, Connolly J, Savani RC, Narula N, Liang B, Levy RJ. 112. Lincoln J, Lange AW, Yutzey KE. Hearts and bones: Shared regulatory
Serotonin mechanisms in heart valve disease I: Serotonin-induced up- mechanisms in heart valve, cartilage, tendon, and bone development.
regulation of transforming growth factor-beta1 via G-protein signal Dev Biol 294: 292-302, 2006.
transduction in aortic valve interstitial cells. Am J Pathol 161: 2111- 113. Linde C, Leclercq C, Rex S, Garrigue S, Lavergne T, Cazeau S,
2121, 2002. McKenna W, Fitzgerald M, Deharo JC, Alonso C, Walker S, Braun-
91. Johnson RC, Leopold JA, Loscalzo J. Vascular calcification: Pathobio- schweig F, Bailleul C, Daubert JC. Long-term benefits of biventricular
logical mechanisms and clinical implications. Circ Res 99: 1044-1059, pacing in congestive heart failure: Results from the MUltisite STim-
2006. ulation in cardiomyopathy (MUSTIC) study. J Am Coll Cardiol 40:
92. Kaden JJ, Bickelhaupt S, Grobholz R, Haase KK, Sarikoc A, Kilic 111-118, 2002.
R, Brueckmann M, Lang S, Zahn I, Vahl C, Hagl S, Dempfle CE, 114. Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial
Borggrefe M. Receptor activator of nuclear factor kappaB ligand and cell phenotypes in regulating heart valve pathobiology. Am J Pathol
osteoprotegerin regulate aortic valve calcification. J Mol Cell Cardiol 171: 1407-1418, 2007.
36: 57-66, 2004. 115. Liu Y, Suzuki YJ, Day RM, Fanburg BL. Rho kinase-induced nuclear
93. Kaden JJ, Dempfle CE, Grobholz R, Fischer CS, Vocke DC, Kilic R, translocation of ERK1/ERK2 in smooth muscle cell mitogenesis caused
Sarikoc A, Pinol R, Hagl S, Lang S, Brueckmann M, Borggrefe M. by serotonin. Circ Res 95: 579-586, 2004.
Inflammatory regulation of extracellular matrix remodeling in calcific 116. Logan CY, Nusse R. The Wnt signaling pathway in development and
aortic valve stenosis. Cardiovasc Pathol 14: 80-87, 2005. disease. Annu Rev Cell Dev Biol 20: 781-810, 2004.
94. Kamath AR, Pai RG. Risk factors for progression of calcific aor- 117. Luna-Zurita L, Prados B, Grego-Bessa J, Luxan G, del Monte G, Ben-
tic stenosis and potential therapeutic targets. Int J Angiol 17: 63-70, guria A, Adams RH, Perez-Pomares JM, de la Pompa JL. Integration of
2008. a Notch-dependent mesenchymal gene program and Bmp2-driven cell
95. Kapoor WN. Syncope. In: Walker HK, Hall WD, Hurst JW, editors. invasiveness regulates murine cardiac valve formation. J Clin Invest
Clinical Methods: The History, Physical, and Laboratory Examina- 120: 3493-3507, 2010.
tions. Boston: 1990. 118. Luo S, Lei H, Qin H, Xia Y. Molecular mechanisms of endothelial NO
96. Katz R, Budoff MJ, Takasu J, Shavelle DM, Bertoni A, Blumenthal synthase uncoupling. Curr Pharm Des 20: 3548-3553, 2014.
RS, Ouyang P, Wong ND, O’Brien KD. Relationship of metabolic 119. Lyons RM, Keski-Oja J, Moses HL. Proteolytic activation of latent
syndrome with incident aortic valve calcium and aortic valve calcium transforming growth factor-beta from fibroblast-conditioned medium.
progression: The Multi-Ethnic Study of Atherosclerosis (MESA). Dia- J Cell Biol 106: 1659-1665, 1988.
betes 58: 813-819, 2009. 120. MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: Compo-
97. Kawaguchi H, Akune T, Yamaguchi M, Ohba S, Ogata N, Chung UI, nents, mechanisms, and diseases. Dev Cell 17: 9-26, 2009.
Kubota N, Terauchi Y, Kadowaki T, Nakamura K. Distinct effects 121. MacGrogan D, Luna-Zurita L, de la Pompa JL. Notch signaling in
of PPARgamma insufficiency on bone marrow cells, osteoblasts, and cardiac valve development and disease. Birth Defects Res Part A Clin
osteoclastic cells. J Bone Miner Metab 23: 275-279, 2005. Mol Teratol 91: 449-459, 2011.
98. Keen HL, Ryan MJ, Beyer A, Mathur S, Scheetz TE, Gackle BD, Faraci 122. MacLea HB, Boon JA, Bright JM. Doppler echocardiographic evalua-
FM, Casavant TL, Sigmund CD. Gene expression profiling of potential tion of midventricular obstruction in cats with hypertrophic cardiomy-
PPARgamma target genes in mouse aorta. Physiol Genomics 18: 33-42, opathy. J Vet Intern Med 27: 1416-1420, 2013.
2004. 123. Macsai CE, Foster BK, Xian CJ. Roles of Wnt signalling in bone
99. Kennedy JA, Hua X, Mishra K, Murphy GA, Rosenkranz AC, Horowitz growth, remodelling, skeletal disorders and fracture repair. J Cell
JD. Inhibition of calcifying nodule formation in cultured porcine aor- Physiol 215: 578-587, 2008.
tic valve cells by nitric oxide donors. Eur J Pharmacol 602: 28-35, 124. Madu EC, D’Cruz IA. The vital role of papillary muscles in mitral
2009. and ventricular function: Echocardiographic insights. Clin Cardiol 20:
100. Khan R, Sheppard R. Fibrosis in heart disease: Understanding the role 93-98, 1997.
of transforming growth factor-beta in cardiomyopathy, valvular disease 125. Marchesi C, Paradis P, Schiffrin EL. Role of the renin-angiotensin
and arrhythmia. Immunology 118: 10-24, 2006. system in vascular inflammation. Trends Pharmacol Sci 29: 367-374,
101. Kiefer TL, Bashore TM. Pulmonary hypertension related to left-sided 2008.
cardiac pathology. Pulm Med 2011: 381787, 2011. 126. Markwald RR, Butcher JT. The next frontier in cardiovascular devel-
102. Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac opmental biology—an integrated approach to adult disease? Nat Clin
fibrosis. Cell Mol Life Sci 71: 549-574, 2014. Pract Cardiovasc Med 4: 60-61, 2007.
103. Kopuz C, Erk K, Baris YS, Onderoglu S, Sinav A. Morphometry of the 127. Markwald RR, Norris RA, Moreno-Rodriguez R, Levine RA. Develop-
fibrous ring of the mitral valve. Ann Anat 177: 151-154, 1995. mental basis of adult cardiovascular diseases: Valvular heart diseases.
104. Kupfahl C, Pink D, Friedrich K, Zurbrugg HR, Neuss M, Warnecke C, Ann N Y Acad Sci 1188: 177-183, 2010.
Fielitz J, Graf K, Fleck E, Regitz-Zagrosek V. Angiotensin II directly 128. McCarthy KP, Ring L, Rana BS. Anatomy of the mitral valve:
increases transforming growth factor beta1 and osteopontin and indi- Understanding the mitral valve complex in mitral regurgitation. Eur
rectly affects collagen mRNA expression in the human heart. Cardio- J Echocardiogr 11: i3-i9, 2010.
vasc Res 46: 463-475, 2000. 129. Mekontso-Dessap A, Brouri F, Pascal O, Lechat P, Hanoun N, Lan-
105. Kurtz CE, Otto CM. Aortic stenosis: Clinical aspects of diagnosis and fumey L, Seif I, Benhaiem-Sigaux N, Kirsch M, Hamon M, Adnot S,
management, with 10 illustrative case reports from a 25-year experi- Eddahibi S. Deficiency of the 5-hydroxytryptamine transporter gene
ence. Medicine (Baltimore) 89: 349-379, 2010. leads to cardiac fibrosis and valvulopathy in mice. Circulation 113:
106. Lacerda CM, Maclea HB, Kisiday JD, Orton EC. Static and cyclic 81-89, 2006.
tensile strain induce myxomatous effector proteins and serotonin in 130. Merryman WD, Lukoff HD, Long RA, Engelmayr GC, Jr., Hopkins RA,
canine mitral valves. J Vet Cardiol 14: 223-230, 2012. Sacks MS. Synergistic effects of cyclic tension and transforming growth
107. Launay JM, Birraux G, Bondoux D, Callebert J, Choi DS, Loric S, factor-beta1 on the aortic valve myofibroblast. Cardiovasc Pathol 16:
Maroteaux L. Ras involvement in signal transduction by the serotonin 268-276, 2007.
5-HT2B receptor. J Biol Chem 271: 3141-3147, 1996. 131. Miller JD, Chu Y, Brooks RM, Richenbacher WE, Pena-Silva R,
108. Lee NK, Choi YG, Baik JY, Han SY, Jeong DW, Bae YS, Kim N, Heistad DD. Dysregulation of antioxidant mechanisms contributes to
Lee SY. A crucial role for reactive oxygen species in RANKL-induced increased oxidative stress in calcific aortic valvular stenosis in humans.
osteoclast differentiation. Blood 106: 852-859, 2005. J Am Coll Cardiol 52: 843-850, 2008.
109. Levine RA, Hagege AA, Judge DP, Padala M, Dal-Bianco JP, Aikawa 132. Miller JD, Weiss RM, Heistad DD. Calcific aortic valve stenosis: Meth-
E, Beaudoin J, Bischoff J, Bouatia-Naji N, Bruneval P, Butcher JT, ods, models, and mechanisms. Circ Res 108: 1392-1412, 2011.
Carpentier A, Chaput M, Chester AH, Clusel C, Delling FN, Dietz HC, 133. Miller JD, Weiss RM, Serrano KM, Brooks RM, II, Berry CJ, Zim-
Dina C, Durst R, Fernandez-Friera L, Handschumacher MD, Jensen merman K, Young SG, Heistad DD. Lowering plasma cholesterol lev-
MO, Jeunemaitre XP, Le Marec H, Le Tourneau T, Markwald RR, els halts progression of aortic valve disease in mice. Circulation 119:
Merot J, Messas E, Milan DP, Neri T, Norris RA, Peal D, Perrocheau 2693-2701, 2009.
M, Probst V, Puceat M, Rosenthal N, Solis J, Schott JJ, Schwammen- 134. Miller JD, Weiss RM, Serrano KM, Castaneda LE, Brooks RM,
thal E, Slaugenhaupt SA, Song JK, Yacoub MH. Mitral valve Zimmerman K, Heistad DD. Evidence for active regulation of pro-
disease—morphology and mechanisms. Nat Rev Cardiol 12: 689-710, osteogenic signaling in advanced aortic valve disease. Arterioscler
2015. Thromb Vasc Biol 30: 2482-2486, 2010.

816 Volume 7, July 2017


Comprehensive Physiology Valve Pathology Consequences

135. Miyazono K, Hellman U, Wernstedt C, Heldin CH. Latent high molec- 157. Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue
ular weight complex of transforming growth factor beta 1. Purification N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, Yokoyama
from human platelets and structural characterization. J Biol Chem 263: M. Overexpression of endothelial nitric oxide synthase accelerates
6407-6415, 1988. atherosclerotic lesion formation in apoE-deficient mice. J Clin Invest
136. Moesgaard SG, Olsen LH, Aasted B, Viuff BM, Pedersen LG, Ped- 110: 331-340, 2002.
ersen HD, Harrison AP. Direct measurements of nitric oxide release 158. Palta S, Pai AM, Gill KS, Pai RG. New insights into the progression
in relation to expression of endothelial nitric oxide synthase in iso- of aortic stenosis: Implications for secondary prevention. Circulation
lated porcine mitral valves. J Vet Med A Physiol Pathol Clin Med 54: 101: 2497-2502, 2000.
156-160, 2007. 159. Panizo S, Cardus A, Encinas M, Parisi E, Valcheva P, Lopez-Ongil
137. Moesgaard SG, Olsen LH, Viuff BM, Baandrup U, Pedersen LG, Thom- S, Coll B, Fernandez E, Valdivielso JM. RANKL increases vascular
sen PD, Pedersen HD, Harrison AP. Increased nitric oxide release and smooth muscle cell calcification through a RANK-BMP4-dependent
expression of endothelial and inducible nitric oxide synthases in mildly pathway. Circ Res 104: 1041-1048, 2009.
changed porcine mitral valve leaflets. J Heart Valve Dis 16: 67-75, 160. Paranya G, Vineberg S, Dvorin E, Kaushal S, Roth SJ, Rabkin E, Schoen
2007. FJ, Bischoff J. Aortic valve endothelial cells undergo transforming
138. Mohler ER, III, Gannon F, Reynolds C, Zimmerman R, Keane MG, growth factor-beta-mediated and non-transforming growth factor-beta-
Kaplan FS. Bone formation and inflammation in cardiac valves. Circu- mediated transdifferentiation in vitro. Am J Pathol 159: 1335-1343,
lation 103: 1522-1528, 2001. 2001.
139. Mokadam NA, Stout KK, Verrier ED. Management of acute regurgita- 161. Piera-Velazquez S, Li Z, Jimenez SA. Role of endothelial-mesenchymal
tion in left-sided cardiac valves. Tex Heart Inst J 38: 9-19, 2011. transition (EndoMT) in the pathogenesis of fibrotic disorders. Am J
140. Morony S, Tintut Y, Zhang Z, Cattley RC, Van G, Dwyer D, Stolina Pathol 179: 1074-1080, 2011.
M, Kostenuik PJ, Demer LL. Osteoprotegerin inhibits vascular calcifi- 162. Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH,
cation without affecting atherosclerosis in ldlr(−/−) mice. Circulation III, Sacks MS, Bavaria JE, Ferrari G. Osteopontin-CD44v6 interac-
117: 411-420, 2008. tion mediates calcium deposition via phospho-Akt in valve interstitial
141. Mow T, Poulsen K, Nielsen AH, Baandrup U, Hansen BF, Pedersen cells from patients with noncalcified aortic valve sclerosis. Arterioscler
HD. Species differences in expression of angiotensin II receptors and Thromb Vasc Biol 34: 2086-2094, 2014.
angiotensin-converting enzyme in human, canine and rat mitral valve 163. Poggio P, Folesani G, Raffa GM, Songia P, Valenti V, Myasoedova V,
leaflets. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 124: Parolari A. Antihypertensive treatments in patients affected by aortic
11-17, 1999. valve stenosis. Curr Pharm Des 2016.
142. Natorska J, Marek G, Sadowski J, Undas A. Presence of B cells within 164. Poggio P, Sainger R, Branchetti E, Grau JB, Lai EK, Gorman RC,
aortic valves in patients with aortic stenosis: Relation to severity of the Sacks MS, Parolari A, Bavaria JE, Ferrari G. Noggin attenuates the
disease. J Cardiol 67: 80-85, 2016. osteogenic activation of human valve interstitial cells in aortic valve
143. New SE, Aikawa E. Molecular imaging insights into early inflammatory sclerosis. Cardiovasc Res 98: 402-410, 2013.
stages of arterial and aortic valve calcification. Circ Res 108: 1381- 165. Pohjolainen V, Taskinen P, Soini Y, Rysa J, Ilves M, Juvonen T,
1391, 2011. Ruskoaho H, Leskinen H, Satta J. Noncollagenous bone matrix pro-
144. Ngo MV, Gottdiener JS, Fletcher RD, Fernicola DJ, Gersh BJ. Smoking teins as a part of calcific aortic valve disease regulation. Hum Pathol
and obesity are associated with the progression of aortic stenosis. Am J 39: 1695-1701, 2008.
Geriatr Cardiol 10: 86-90, 2001. 166. Qin X, Corriere MA, Matrisian LM, Guzman RJ. Matrix metallopro-
145. Ni CW, Qiu H, Rezvan A, Kwon K, Nam D, Son DJ, Visvader JE, Jo H. teinase inhibition attenuates aortic calcification. Arterioscler Thromb
Discovery of novel mechanosensitive genes in vivo using mouse carotid Vasc Biol 26: 1510-1516, 2006.
artery endothelium exposed to disturbed flow. Blood 116: e66-e73, 167. Rabkin E, Aikawa M, Stone JR, Fukumoto Y, Libby P, Schoen FJ. Acti-
2010. vated interstitial myofibroblasts express catabolic enzymes and mediate
146. Nigam V, Srivastava D. Notch1 represses osteogenic pathways in aortic matrix remodeling in myxomatous heart valves. Circulation 104: 2525-
valve cells. J Mol Cell Cardiol 47: 828-834, 2009. 2532, 2001.
147. Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP, III, 168. Rajamannan NM. Mechanisms of aortic valve calcification: The LDL-
Guyton RA, O’Gara PT, Ruiz CE, Skubas NJ, Sorajja P, Sundt TM, III, density-radius theory: A translation from cell signaling to physiology.
Thomas JD, Anderson JL, Halperin JL, Albert NM, Bozkurt B, Brindis Am J Physiol Heart Circ Physiol 298: H5-H15, 2010.
RG, Creager MA, Curtis LH, DeMets D, Guyton RA, Hochman JS, 169. Rajamannan NM, Evans FJ, Aikawa E, Grande-Allen KJ, Demer LL,
Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen WK, Stevenson Heistad DD, Simmons CA, Masters KS, Mathieu P, O’Brien KD,
WG, Yancy CW, American College of C, American College of Car- Schoen FJ, Towler DA, Yoganathan AP, Otto CM. Calcific aortic valve
diology/American Heart A, and American Heart A. 2014 AHA/ACC disease: Not simply a degenerative process: A review and agenda for
guideline for the management of patients with valvular heart disease: A research from the National Heart and Lung and Blood Institute Aor-
report of the American College of Cardiology/American Heart Asso- tic Stenosis Working Group. Executive summary: Calcific aortic valve
ciation Task Force on Practice Guidelines. J Thorac Cardiovasc Surg disease-2011 update. Circulation 124: 1783-1791, 2011.
148: e1-e132, 2014. 170. Rajamannan NM, Subramaniam M, Caira F, Stock SR, Spelsberg TC.
148. Nkomo VT, Gardin JM, Skelton TN, Gottdiener JS, Scott CG, Enriquez- Atorvastatin inhibits hypercholesterolemia-induced calcification in the
Sarano M. Burden of valvular heart diseases: A population-based study. aortic valves via the Lrp5 receptor pathway. Circulation 112: I229-234,
Lancet 368: 1005-1011, 2006. 2005.
149. Obadia JF, Casali C, Chassignolle JF, Janier M. Mitral subvalvular 171. Rajamannan NM, Subramaniam M, Stock SR, Stone NJ, Springett
apparatus: Different functions of primary and secondary chordae. Cir- M, Ignatiev KI, McConnell JP, Singh RJ, Bonow RO, Spelsberg TC.
culation 96: 3124-3128, 1997. Atorvastatin inhibits calcification and enhances nitric oxide synthase
150. Obayashi K, Miyagawa-Tomita S, Matsumoto H, Koyama H, Nakan- production in the hypercholesterolaemic aortic valve. Heart 91: 806-
ishi T, Hirose H. Effects of transforming growth factor-beta3 and matrix 810, 2005.
metalloproteinase-3 on the pathogenesis of chronic mitral valvular dis- 172. Ranganathan N, Lam JH, Wigle ED, Silver MD. Morphology of the
ease in dogs. Am J Vet Res 72: 194-202, 2011. human mitral valve. II. The value leaflets. Circulation 41: 459-467,
151. Oliveira-Paula GH, Lacchini R, Tanus-Santos JE. Inducible nitric oxide 1970.
synthase as a possible target in hypertension. Curr Drug Targets 15: 173. Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: A
164-174, 2014. prelude for more. Circ Res 106: 1798-1806, 2010.
152. Orita Y, Yamamoto H, Kohno N, Sugihara M, Honda H, Kawamata 174. Rapaport E. Natural history of aortic and mitral valve disease. Am J
S, Mito S, Soe NN, Yoshizumi M. Role of osteoprotegerin in arterial Cardiol 35: 221-227, 1975.
calcification: Development of new animal model. Arterioscler Thromb 175. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxi-
Vasc Biol 27: 2058-2064, 2007. some proliferator-activated receptor-gamma is a negative regulator of
153. Orton EC, Lacerda CM, MacLea HB. Signaling pathways in mitral macrophage activation. Nature 391: 79-82, 1998.
valve degeneration. J Vet Cardiol 14: 7-17, 2012. 176. Ross J, Jr., Braunwald E. Aortic stenosis. Circulation 38: 61-67, 1968.
154. Otto CM, Kuusisto J, Reichenbach DD, Gown AM, O’Brien KD. Char- 177. Rothman RB, Baumann MH. Serotonergic drugs and valvular heart
acterization of the early lesion of ‘degenerative’ valvular aortic stenosis. disease. Expert Opin. Drug Saf 8: 317-329, 2009.
Histological and immunohistochemical studies. Circulation 90: 844- 178. Rothman RB, Baumann MH, Savage JE, Rauser L, McBride A,
853, 1994. Hufeisen SJ, Roth BL. Evidence for possible involvement of 5-HT(2B)
155. Oyama MA, Chittur SV. Genomic expression patterns of mitral valve receptors in the cardiac valvulopathy associated with fenfluramine
tissues from dogs with degenerative mitral valve disease. Am J Vet Res and other serotonergic medications. Circulation 102: 2836-2841,
67: 1307-1318, 2006. 2000.
156. Oyama MA, Levy RJ. Insights into serotonin signaling mechanisms 179. Rusted IE, Scheifley CH, Edwards JE. Studies of the mitral valve. I.
associated with canine degenerative mitral valve disease. J Vet Intern Anatomic features of the normal mitral valve and associated structures.
Med 24: 27-36, 2010. Circulation 6: 825-831, 1952.

Volume 7, July 2017 817


Valve Pathology Consequences Comprehensive Physiology

180. Sacks MS, David Merryman W, Schmidt DE. On the biomechanics of 198. Timek TA, Green GR, Tibayan FA, Lai DT, Rodriguez F, Liang D,
heart valve function. J Biomech 42: 1804-1824, 2009. Daughters GT, Ingels NB, Jr., Miller DC. Aorto-mitral annular dynam-
181. Sainger R, Grau JB, Branchetti E, Poggio P, Lai E, Koka E, Vernick ics. Ann Thorac Surg 76: 1944-1950, 2003.
WJ, Gorman RC, Bavaria JE, Ferrari G. Comparison of transesophageal 199. Timmerman LA, Grego-Bessa J, Raya A, Bertran E, Perez-Pomares
echocardiographic analysis and circulating biomarker expression pro- JM, Diez J, Aranda S, Palomo S, McCormick F, Izpisua-Belmonte
file in calcific aortic valve disease. J Heart Valve Dis 22: 156-165, JC, de la Pompa JL. Notch promotes epithelial-mesenchymal transition
2013. during cardiac development and oncogenic transformation. Genes Dev
182. Sainger R, Grau JB, Branchetti E, Poggio P, Seefried WF, Field BC, 18: 99-115, 2004.
Acker MA, Gorman RC, Gorman JH, III, Hargrove CW, III, Bavaria 200. Vahanian A, Otto CM. Risk stratification of patients with aortic stenosis.
JE, Ferrari G. Human myxomatous mitral valve prolapse: Role of bone Eur Heart J 31: 416-423, 2010.
morphogenetic protein 4 in valvular interstitial cell activation. J Cell 201. Vattikuti R, Towler DA. Osteogenic regulation of vascular calcification:
Physiol 227: 2595-2604, 2012. An early perspective. Am J Physiol Endocrinol Metab 286: E686-E696,
183. Sasaki H, Yamamoto H, Tominaga K, Masuda K, Kawai T, Teshima- 2004.
Kondo S, Matsuno K, Yabe-Nishimura C, Rokutan K. Receptor acti- 202. Villar AV, Cobo M, Llano M, Montalvo C, Gonzalez-Vilchez F, Martin-
vator of nuclear factor-kappaB ligand-induced mouse osteoclast differ- Duran R, Hurle MA, Nistal JF. Plasma levels of transforming growth
entiation is associated with switching between NADPH oxidase homo- factor-beta1 reflect left ventricular remodeling in aortic stenosis. PloS
logues. Free Radic Biol Med 47: 189-199, 2009. One 4: e8476, 2009.
184. Schade R, Andersohn F, Suissa S, Haverkamp W, Garbe E. Dopamine 203. Walker GA, Masters KS, Shah DN, Anseth KS, Leinwand LA. Valvular
agonists and the risk of cardiac-valve regurgitation. N Engl J Med 356: myofibroblast activation by transforming growth factor-beta: Implica-
29-38, 2007. tions for pathological extracellular matrix remodeling in heart valve
185. Schultz-Cherry S, Murphy-Ullrich JE. Thrombospondin causes activa- disease. Circ Res 95: 253-260, 2004.
tion of latent transforming growth factor-beta secreted by endothelial 204. Waltenberger J, Lundin L, Oberg K, Wilander E, Miyazono K, Heldin
cells by a novel mechanism. J Cell Biol 122: 923-932, 1993. CH, Funa K. Involvement of transforming growth factor-beta in the
186. Scruggs SM, Disatian S, Orton EC. Serotonin transmembrane trans- formation of fibrotic lesions in carcinoid heart disease. Am J Pathol
porter is down-regulated in late-stage canine degenerative mitral valve 142: 71-78, 1993.
disease. J Vet Cardiol 12: 163-169, 2010. 205. Walther DJ, Peter JU, Bashammakh S, Hortnagl H, Voits M, Fink H,
187. Seya K, Yu Z, Kanemaru K, Daitoku K, Akemoto Y, Shibuya H, Fukuda Bader M. Synthesis of serotonin by a second tryptophan hydroxylase
I, Okumura K, Motomura S, Furukawa K. Contribution of bone morpho- isoform. Science 299: 76, 2003.
genetic protein-2 to aortic valve calcification in aged rat. J Pharmacol 206. Wipff PJ, Rifkin DB, Meister JJ, Hinz B. Myofibroblast contraction
Sci 115: 8-14, 2011. activates latent TGF-beta1 from the extracellular matrix. J Cell Biol
188. Siney L, Lewis MJ. Nitric oxide release from porcine mitral valves. 179: 1311-1323, 2007.
Cardiovasc Res 27: 1657-1661, 1993. 207. Wirrig EE, Hinton RB, Yutzey KE. Differential expression of cartilage
189. Singh JP, Evans JC, Levy D, Larson MG, Freed LA, Fuller DL, Lehman and bone-related proteins in pediatric and adult diseased aortic valves.
B, Benjamin EJ. Prevalence and clinical determinants of mitral, tri- J Mol Cell Cardiol 50: 561-569, 2011.
cuspid, and aortic regurgitation (the Framingham Heart Study). Am J 208. Wu HD, Maurer MS, Friedman RA, Marboe CC, Ruiz-Vazquez EM,
Cardiol 83: 897-902, 1999. Ramakrishnan R, Schwartz A, Tilson MD, Stewart AS, Winchester R.
190. Singh RG, Cappucci R, Kramer-Fox R, Roman MJ, Kligfield P, Borer The lymphocytic infiltration in calcific aortic stenosis predominantly
JS, Hochreiter C, Isom OW, Devereux RB. Severe mitral regurgitation consists of clonally expanded T cells. J Immunol 178: 5329-5339, 2007.
due to mitral valve prolapse: Risk factors for development, progression, 209. Wylie-Sears J, Aikawa E, Levine RA, Yang JH, Bischoff J. Mitral valve
and need for mitral valve surgery. Am J Cardiol 85: 193-198, 2000. endothelial cells with osteogenic differentiation potential. Arterioscler
191. Siu SC, Silversides CK. Bicuspid aortic valve disease. J Am Coll Car- Thromb Vasc Biol 31: 598-607, 2011.
diol 55: 2789-2800, 2010. 210. Xu J, Jian B, Chu R, Lu Z, Li Q, Dunlop J, Rosenzweig-Lipson S,
192. Songia P, Branchetti E, Parolari A, Myasoedova V, Ferrari G, Ala- McGonigle P, Levy RJ, Liang B. Serotonin mechanisms in heart valve
manni F, Tremoli E, Poggio P. Mitral valve endothelial cells secrete disease II: The 5-HT2 receptor and its signaling pathway in aortic valve
osteoprotegerin during endothelial mesenchymal transition. J Mol Cell interstitial cells. Am J Pathol 161: 2209-2218, 2002.
Cardiol 98: 48-57, 2016. 211. Yadav VK, Ducy P. Lrp5 and bone formation: A serotonin-dependent
193. Sorescu GP, Song H, Tressel SL, Hwang J, Dikalov S, Smith DA, Boyd pathway. Ann N Y Acad Sci 1192: 103-109, 2010.
NL, Platt MO, Lassegue B, Griendling KK, Jo H. Bone morphogenic 212. Yamashita A, Takada T, Nemoto K, Yamamoto G, Torii R. Transient
protein 4 produced in endothelial cells by oscillatory shear stress suppression of PPARgamma directed ES cells into an osteoblastic lin-
induces monocyte adhesion by stimulating reactive oxygen species eage. FEBS Lett 580: 4121-4125, 2006.
production from a nox1-based NADPH oxidase. Circ Res 95: 773-779, 213. Yetkin E, Tchaikovski V, Erdil N, Alan S, Waltenberger J. Increased
2004. expression of cystatin C and transforming growth factor beta-1 in cal-
194. Stein PD, Wang CH, Riddle JM, Sabbah HN, Magilligan DJ, Jr., cific aortic valves. Int J Cardiol 176: 1252-1254, 2014.
Hawkins ET. Scanning electron microscopy of operatively excised 214. Yip CY, Chen JH, Zhao R, Simmons CA. Calcification by valve inter-
severely regurgitant floppy mitral valves. Am J Cardiol 64: 392-394, stitial cells is regulated by the stiffness of the extracellular matrix.
1989. Arterioscler Thromb Vasc Biol 29: 936-942, 2009.
195. Stephens EH, Nguyen TC, Itoh A, Ingels NB, Jr., Miller DC, Grande- 215. Yoon BS, Lyons KM. Multiple functions of BMPs in chondrogenesis.
Allen KJ. The effects of mitral regurgitation alone are sufficient for J Cell Biochem 93: 93-103, 2004.
leaflet remodeling. Circulation 118: S243-S249, 2008. 216. Yosefy C, Ben Barak A. Floppy mitral valve/mitral valve prolapse and
196. Sucosky P, Balachandran K, Elhammali A, Jo H, Yoganathan AP. genetics. J Heart Valve Dis 16: 590-595, 2007.
Altered shear stress stimulates upregulation of endothelial VCAM-1 217. Yperman J, De Visscher G, Holvoet P, Flameng W. Molecular and
and ICAM-1 in a BMP-4- and TGF-beta1-dependent pathway. Arte- functional characterization of ovine cardiac valve-derived interstitial
rioscler Thromb Vasc Biol 29: 254-260, 2009. cells in primary isolates and cultures. Tissue Eng 10: 1368-1375, 2004.
197. Tanikawa T, Okada Y, Tanikawa R, Tanaka Y. Advanced glycation end 218. Zanettini R, Antonini A, Gatto G, Gentile R, Tesei S, Pezzoli G. Valvular
products induce calcification of vascular smooth muscle cells through heart disease and the use of dopamine agonists for Parkinson’s disease.
RAGE/p38 MAPK. J Vasc Res 46: 572-580, 2009. N Engl J Med 356: 39-46, 2007.

818 Volume 7, July 2017

You might also like