Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Contemporary Reviews in Cardiovascular Medicine

Epidemiology and Pathophysiology of Mitral Valve Prolapse


New Insights Into Disease Progression, Genetics, and Molecular Basis
Francesca N. Delling, MD; Ramachandran S. Vasan, MD

M itral valve (MV) prolapse (MVP) is a common disorder,


afflicting 2% to 3% of the general population.1,2 It is
characterized by typical fibromyxomatous changes in the mitral
the numerous reported correlates, only the association with
leaner body mass has been reproducibly associated with MVP
in the literature.1 Abnormal autonomic function has been
leaflet tissue with superior displacement of 1 or both leaflets reported as the mechanism explaining symptoms in patients
into the left atrium.3,4 With a prevalence of 2% to 3%, MVP is with MVP,18 but given its absence in asymptomatic MVP
expected to affect ≈7.8 million individuals in the United States patients, it remains unclear whether MVP is directly related to
and >176 million people worldwide. MVP can be associated autonomic dysfunction or any reported association is purely
with significant mitral regurgitation (MR), bacterial endocardi- incidental.19 Hypomagnesemia and dysregulation of the renin-
tis, congestive heart failure, and even sudden death.5–7 angiotensin-aldosterone system have also been demonstrated
MVP is a clinical entity that is not fully understood, despite in MVP syndrome, albeit in small patient samples.20,21
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

being known for more than a century. A “midsystolic click” The classic auscultatory finding in MVP is a dynamic,
was first described in 1887 by Cuffer and Barbillon.8 In 1963, midsystolic to late systolic click frequently associated with a
Barlow and Pocock9 demonstrated the presence of MR by high-pitched, late systolic murmur. A careful physical exami-
angiography in patients with the “click-murmur” syndrome. nation is highly sensitive for making a diagnosis of MVP, but
Criley et al10 subsequently coined the term MVP. its specificity is limited (with echocardiography used as the
MVP may be familial or sporadic. Despite being the most gold standard).22 Redundant leaflets or chordae may produce
common cause of isolated MR requiring surgical repair,11 an audible click without echocardiographic evidence of leaf-
little is known about the genetic mechanisms underlying the let prolapse, giving false-positive physical findings. Finally,
pathogenesis and progression of MVP. Studies on the heri- echocardiographic prolapse may exist without significant aus-
table features of MVP have been limited by the analysis of cultatory findings.22 Patients with physical examination find-
relatively small pedigrees and by self-referral and selection ings that suggest MVP should undergo confirmatory testing
biases, including a preponderance of data from hospital-based with 2D echocardiography.
cohorts.12,13 Nonetheless, the majority of data favor an auto- In the early days of 2D echocardiography, the diagnosis of
somal-dominant pattern of inheritance in a large proportion MVP occurred with a prevalence ranging from 5% to 15% and
of individuals with MVP.12,13 Despite the variability in clini- in as many as 35% of those undergoing imaging.23–25 In part,
cal features, familial MVP might be considered a prevalent this overdiagnosis was the result of the erroneous assump-
mendelian cardiac abnormality in humans. Although filamin- tion that the MV was planar; thus, any sonographic view that
A has been identified as causing an X-linked form of MVP,14 showed excursion of the leaflets superior to the mitral annu-
the causative genes for the more common form of autosomal- lus was deemed pathological. Pivotal echocardiographic work
dominant MVP have yet to be defined. In this review, we sum- in the late 1980s redefined normal mitral anatomy.26 Using
marize our current knowledge of the diagnosis, epidemiology, 3-dimensional echocardiographic imaging, Levine and col-
prognosis, pathophysiology, and genetics of MVP, with a leagues26 established that the mitral annulus was in fact saddle
focus on potential future research directions. shaped. Therefore, in the anterior-posterior axis, the mitral
annulus is concaved upward, whereas medially to laterally,
Diagnosis of MVP the annulus is concaved downward. This mitral geometry cre-
Physical examination and 2-dimensional (2D) echocardiog- ates the possibility that, in a sonographic 4-chamber view,
raphy are the diagnostic gold standards for MVP.15,16 Various the leaflets can appear to “break” the annular plane (creating
symptoms (including atypical chest pain, exertional dyspnea, the appearance of prolapse) when in reality they are normal.
palpitations, syncope, and anxiety) and clinical findings (low Echocardiographic MVP has since been defined as single-
blood pressure, leaner build, and electrocardiographic repo- leaflet or bileaflet prolapse of at least 2 mm beyond the long-
larization abnormalities) have been associated with MVP, and axis annular plane, with or without mitral leaflet thickening
their constellation has been called the MVP syndrome.1,17 Of (Figure 1A). Prolapse with thickening of the leaflets >5 mm is

From the Framingham Heart Study, Framingham, MA (F.N.D., R.S.V.); Department of Medicine (Cardiovascular Division), Beth Israel Deaconess
Medical Center, Harvard Medical School, Boston, MA (F.N.D.); and Cardiology Section, and Preventive Medicine Section, Boston University School of
Medicine, Boston, MA (R.S.V.).
Correspondence to Francesca Nesta Delling, MD, Beth Israel Deaconess Medical Center, Cardiovascular Division, 330 Brookline Ave, E/SH-458,
Boston, MA 02215. E-mail fdelling@bidmc.harvard.edu
(Circulation. 2014;129:2158-2170.)
© 2014 American Heart Association, Inc.
Circulation is available at http://circ.ahajournals.org DOI: 10.1161/CIRCULATIONAHA.113.006702

2158
Delling and Vasan   Epidemiology and Pathophysiology of MVP   2159

Cardiac magnetic resonance (CMR) represents a novel,


albeit still not widely used, noninvasive imaging method that
identifies MVP with a sensitivity and specificity of 100%
with 2D TTE used as the gold standard (Figure 2A).32 In
addition, CMR can quantify MR using phase-contrast veloc-
ity mapping.33 Because CMR can reliably provide quantita-
tive determination of ventricular volumes and function, it is
becoming an important clinical tool for following up patients
with MVP-related moderate to severe MR and for surgical
decision making.34 Finally, CMR provides novel insight into
the biology of the MV and its linked myocardium through
improved spatial resolution provided by 3-dimensional
acquisition of images with delayed gadolinium enhance-
ment.32 Such enhancement occurs when the kinetics of gado-
linium excretion is different in 2 adjacent compartments so
that over time 1 compartment enhances more than the other.
This has been a powerful tool for delineating infarcted and
Figure 1. Prolapse of the intermediate posterior mitral valve scal- scarred myocardium, which excrete gadolinium more slowly
lop (P2) shown in a long-axis view of (A) a 2-dimensional (2D)
than viable tissue. The presence of gadolinium enhancement
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

transthoracic echocardiogram, (B) a 2D transesophageal echo-


cardiogram (TEE) with (C) associated severe, eccentric, anteriorly has been shown in both the MV and in papillary muscle
directed mitral regurgitation, and (D) a 3-dimensional TEE surgi- tips in patients with MVP but not in normal control sub-
cal view. AO indicates aorta; LA, left atrium; LV, left ventricle; and jects (Figure 2B).32 It has been speculated that the papil-
RV, right ventricle.
lary muscle is altered in MVP by repetitive traction exerted
by the prolapsing leaflets,35 which has been shown experi-
called classic prolapse, whereas prolapse with lesser degrees mentally to lower the threshold for arrhythmias.36 Although
of leaflet thickening is regarded as nonclassic prolapse.26 more frequent complex arrhythmia on 24-hour ambulatory
Transthoracic echocardiography (TTE) may not adequately Holter monitor has been demonstrated in MVP patients with
visualize the entire MV anatomy. Anatomically, the posterior scarring of the papillary muscles,32 its clinical significance
and anterior leaflets of the MV each may be divided into 3 sec- remains to be established.
tions. Carpentier’s widely recognized nomenclature describes
3 posterior leaflet scallops, the lateral (P1), middle (P2), and Clinical Classification and Prevalence of MVP
medial (P3), and 3 anterior segments, the lateral (A1), middle MVP can be distinguished into primary or nonsyndromic
(A2), and medial (A3; Figure 1D).27,28 Most cases of prolapse MVP and secondary or syndromic MVP. In the latter case,
involve the posterior middle scallop, which is easily identi- MVP occurs in the presence of connective tissue disorders
fied on long-axis TTE images (Figure 1A). However, the such as Marfan syndrome (MFS), Loeys-Dietz syndrome,
posterior lateral scallop (P1) is not clearly seen on long-axis Ehlers-Danlos syndrome, osteogenesis imperfecta, pseudo-
images but is best visualized in the apical 4-chamber view. xanthoma elasticum, and the recently reported aneurysms-
As noted above, superior leaflet displacement in a 4-chamber osteoarthritis syndrome.37–42 MVP has also been observed
view should not be regarded as diagnostic of prolapse. Thus, in hypertrophic cardiomyopathy (HCM) and may con-
TTE can confirm the diagnosis of MVP but may not be able tribute to the pathophysiology of obstruction typical of
to exclude prolapse of all scallops. Although the Carpentier this myopathy.43
nomenclature is based on leaflet indentation, in the Duran
classification, scallops are grouped on the basis of chordal
attachments.29 Specifically, the anterior leaflet is divided into
2 segments (A1 and A2) and the posterior into 4 segments (P1,
PM1, P2, and PM2). Segments A1, P1, and PM1 attach to the
anterolateral papillary muscle, and segments A2, P2, and PM2
attach to the posteromedial papillary muscle. The modified
Carpentier classification30 is a combination of the Carpentier
and Duran nomenclatures. Although the Duran and modified
Carpentier classifications are anatomically more precise than
the classic Carpentier scheme, they are less widely used.
By taking into account several planes of imaging, 2D Figure 2. Cardiac magnetic resonance steady state free pro-
cessing long-axis view of bileaflet mitral valve prolapse (A).
transesophageal echocardiography (TEE) is more effective Short-axis view with 3-dimensional late-gadolinium enhance-
in identifying prolapsing MV segments (Figure 1B).28 Three- ment (3D-LGE) showing fibrosis of the papillary muscle tips.
dimensional TEE has the additional advantage of simulating Adapted from Han et al32 with permission from the publisher.
the surgeon’s view of the MV, with the aortic valve at the 11 Copyright © 2008 Elsevier B.V. Authorization for this adaptation
has been obtained both from the owner of the copyright in the
o’clock position (Figure 1D), and has become an essential tool original work and from the owner of copyright in the translation
in the intraoperative setting.31 or adaptation.
2160  Circulation  May 27, 2014

Nonsyndromic MVP elasticum,38,52 although the true prevalence of the disease is


Based on revised echocardiographic diagnostic criteria, the
26 unclear because standard diagnostic criteria were not used in
prevalence of MVP and its clinical associations were examined the initial imaging studies performed on these patients.
in the community-based Framingham Heart Study (FHS).1
The sample analyzed consisted of 3491 participants in whom Hypertrophic Cardiomyopathy
routine 2D echocardiograms were available and adequate for The largest study assessing the prevalence of MVP in HCM
the evaluation of the MV. Forty-seven individuals (1.3%) had observed it in 3% (of 528 people with HCM), which might
classic and 37 (1.1%) had nonclassic MVP, yielding an esti- suggest that HCM and MVP are 2 distinct conditions that may
mated overall prevalence of 2.4%. The prevalence of MVP was coexist in some cases.53 However, the prevalence of other MV
distributed fairly evenly among individuals in each decade of abnormalities (leaflet elongation and increased thickness) is
age from 30 to 80 years of age. With respect to sex, MVP was much higher in HCM, estimated at 66% in 1 study.54 This sug-
equally distributed between men and women. These findings gests that MV abnormalities are intrinsic to HCM,54 either as
differed from older studies based on M-mode diagnostic cri- a primary trait or as a secondary adaptive response to shear
teria or observations of pedigrees12,13 that reported that MVP stress in a turbulent outflow tract or paracrine effects arising in
preferentially afflicted women and older individuals. Although the adjacent hypertrophic ventricle (see below).55
the genetic predisposition to develop MVP may be present at
birth, MVP is not found in newborns,44 and its prevalence is Prognosis of MVP
low among children (0.3%)45 and young adults (0.6%).46 These
findings suggest that MVP is a progressive disease affecting A Controversial Past
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

predominantly middle-aged individuals.1 Participants with The prognosis of MVP has varied in the published literature.
MVP in FHS were leaner compared with those without MVP.1 In the community-based FHS sample, MVP was described as
An important limitation of the FHS sample is that it is pre- a benign entity with a low occurrence of adverse sequelae.1
dominantly white. A similar prevalence of MVP was described Specifically, none of the individuals with MVP had a his-
in a population-based sample of American Indians (the Strong tory of heart failure, 1 patient (1.2%) had atrial fibrillation,
Heart Study),2 and in a different sample of Canadians of South 1 patient (1.2%) had cerebrovascular disease, and 3 patients
Asian, European, and Chinese descent (the SHARE study).47 (3.6%) had syncope. The prevalence of these outcomes in the
Although these recent studies were based on revised echocar- subjects without prolapse was 0.7%, 1.7%, 1.5%, and 3.0%,
diographic criteria, the prevalence of MVP in blacks was based respectively. The frequencies of chest pain, dyspnea, and elec-
on older M-mode criteria and nonstandard 2D echocardio- trocardiographic abnormalities were similar among individu-
graphic views.48 A systematic review of the published literature als with and without MVP. Individuals with MVP had a greater
did not reveal prior studies that have evaluated the prevalence degree of MR than those without prolapse, but typically the
of MVP in Hispanic samples. valvular regurgitation was classified as trace or mild.1 In prior
Tricuspid valve prolapse has been observed in up to 40% studies,5,6 MVP was portrayed as a disease with frequent and
to 50% of patients with primary or nonsyndromic MVP,49 but serious complications, including stroke, atrial fibrillation,
isolated tricuspid prolapse has rarely been reported. heart failure, and MR requiring surgery. These discrepancies
may be due to selection biases inherent in evaluating symp-
Syndromic MVP: MFS and Other Connective tomatic patients at referral tertiary care centers compared with
Tissue Disorders observations made on healthier asymptomatic volunteers.1,5,6
The prevalence of at least mild MV pathology in MFS has been Changes in diagnostic criteria for MVP over time may have
estimated to be ≈75%, whereas the prevalence of more severe further exacerbated these differences in the prevalence of
myxomatous MV thickening with prolapse is closer to 25% in MVP.26 Subsequently, a community-based study from the
these individuals.50 The prevalence of MVP in patients with Mayo Clinic conducted in a primary care setting has under-
Ehlers-Danlos syndrome using standard echocardiographic scored the clinical heterogeneity of MVP, including a widely
criteria appears to be much lower (6%).37 The prevalence varying prognostic spectrum.56 Based on primary (depressed
of MV disease also appears to be lower in patients with the left ventricular ejection fraction, moderate/severe MR) and
Loeys-Dietz syndrome (relative to MFS).51 One group reported secondary (age >50 years, mild MR, left atrial enlargement,
a direct comparison of MVP prevalence in 71 individuals with atrial fibrillation, and flail leaflet) risk factors, different groups
transforming growth factor-β (TGF-β) receptor 2 (TGFBR2) of MVP with varying prognoses were identified with regard
mutations (characteristic of Loeys-Dietz syndrome) with that to cardiovascular morbidity and mortality.56 Overall, young
in 243 people with fibrillin-1 (FBN1) mutations (typical of (<50 years of age), medically treated patients presenting with
MFS) and in 50 unaffected family members.51 The investiga- normal left ventricular function and no symptoms have excel-
tors observed a substantially higher prevalence of both MVP lent survival, even with severe MR.56,57 The benefit of early
and MR in the cohort with FBN1 mutations than in the group surgery (ie, valve repair in asymptomatic patients) versus a
with TGFBR2 mutations (45% and 56% versus 21% and 35%, watchful wait was suggested in observational studies58 but
respectively). Among affected individuals with the aneurysms- remains controversial.57
osteoarthritis syndrome, MV abnormalities were common and
ranged from mild to severe; 10 of 22 (45%) had MVP and 6 Impact of MR
of 22 (27%) had MR.42 The presence of MVP has also been The common denominator of the studies evaluating prog-
described in osteogenesis imperfecta and pseudoxanthoma nosis of MVP is the role of MR at the time of diagnosis in
Delling and Vasan   Epidemiology and Pathophysiology of MVP   2161

determining the risk for adverse events (such as congestive men) and the development of complications in MVP. However,
heart failure, atrial fibrillation, ischemic neurological event, women represent a large proportion of patients with moderate
and endocarditis) and the need for surgery on follow-up.1,5–7,59,60 or severe MR. In these severe forms, the assessment of left ven-
The Mayo Clinic series highlighted that over a follow-up tricular enlargement in women is challenged by the differences
period of 1.5 years, MR volume increased >8 mL in 51% of 74 in weight and height between the 2 sexes and the frequent use
individuals with MVP. In this clinical series, the progression of an absolute rather than a body size–adjusted left ventricular
of the valvular lesion (particularly a new flail leaflet) and an size measurement. Consequently, for the same degree of MR,
increase in the mitral annular diameter were the 2 independent women undergo mitral surgery less frequently and later than
predictors of an increase in the regurgitant volume over time.60 men. As a consequence, women exhibit excess long-term mor-
Although mitral leaflet thickness >5 mm on M-mode echocar- tality but equivalent survival after valve surgery compared with
diography has been associated with increased risk for sudden men.63 Thus, there are important sex-related differences in the
death, endocarditis, and MR in patients with classic prolapse morphology, presentation, and prognosis of MVP.
in some series,5,6 a more recent larger series using 2D echo-
cardiography reported that mitral leaflet thickness was not an Pathology and Pathophysiology
independent predictor of mortality and valvular morbidity.7
In this community-based study of 833 individuals diagnosed Myxomatous Valve Degeneration
with asymptomatic MVP and followed up longitudinally in MVP is characterized by progressive increases in the area and
Olmsted County, cardiac mortality was best predicted by the length of the MV tissue and typically progresses with a natu-
presence of MR and left ventricular dysfunction at the time of ral history spanning decades, causing leaflets to thicken ana-
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

diagnosis. Risk factors for cardiac morbidity (defined as the tomically and prolapse superiorly into the left atrium beyond
occurrence of heart failure, thromboembolic events, endocar- the mitral annulus in systole, leading to MR (Figure 1C).
ditis, atrial fibrillation, and need for cardiac surgery) included Histologically, the mitral leaflets in MVP are characterized by
age ≥50 years, left atrial enlargement, MR, the presence of a myxomatous degeneration. A detailed explanation of myxo-
flail leaflet, and prevalent atrial fibrillation at the time of the matous changes requires an understanding of the histology
baseline echocardiogram.7 and the development of the normal MV.

Impact of a Flail Leaflet Normal MV Histology and Alterations in MVP


The presence of a flail MV leaflet has been associated with The extracellular matrix (ECM) constitutes the fibroskeleton
a widely varying prognosis.61 Survival in medically treated of a normal MV. Normal valve tissue is divided into 3 layers:
asymptomatic patients with MVP presenting with a flail leaflet the atrialis on the atrial side; the spongiosa, which is the mid-
and normal left ventricular function is excellent.61 Thus, such dle layer; and the fibrosa on the ventricular side (Figure 3).64
patients are at relatively low risk of cardiovascular morbidity. The atrialis, a dense sheet of elastic fibers, provides elasticity
The indications for valve surgery in this group include the devel- to the valve leaflet. The spongiosa is rich in glycosaminogly-
opment of atrial fibrillation (4%/y) and heart failure (5.7%/y). cans and proteoglycans within a fine, interweaving, spongy
Older age, the presence of symptoms, and a left ventricular elastin network. It functions to resist compression between the
ejection fraction <60% at the time of initial diagnosis increase outer layers, gives flexibility to the valve leaflet, and dampens
the risk of developing heart failure and atrial fibrillation and the vibrations resulting from valve closure. The fibrosa, which
are markers of the need for valve surgery and mortality.61,62 As is the thickest part of the leaflet, is made up primarily of orga-
for chronic severe MR in general, management decisions for nized collagen fibers that give the valve its tensile strength.64
patients with flail leaflet are based largely on the presence or
absence of clinical symptoms, the functional state of the left
ventricle, and the feasibility of successful MV repair.16

Sex-Related Differences in Outcomes


As noted, the prevalence of MVP was similar in the 2 sexes in
the FHS, a referral-free, community-based sample.1 Conversely,
in the Olmsted County population, characterized by a mixed
spectrum of community-dwelling and referred patients, with
the use of similar echocardiographic criteria, women were
diagnosed with MVP more often than men and at a younger
age.56 However, complications (such as the development of a
flail leaflet) have been reported more frequently in men.62 The
Mayo Clinic group also underscored the anatomic and func-
Figure 3. Schematic of normal mitral valve histology. ECM indi-
tional differences between the 2 sexes in the context of MVP. cates extracellular matrix; GAG, glycosaminoglycans; VEC, val-
Women present with more anterior and bileaflet prolapse, more vular endothelial cells; and VIC, valvular interstitial cells. Adapted
thickened leaflets, fewer flail leaflets, and less MR compared from Bischoff and Aikawa66 with permission from the publisher.
with men.63 These milder clinical features in women have led Copyright © 2011 Springer. Authorization for this adaptation
has been obtained both from the owner of the copyright in the
to the speculation that an interplay may exist between load- original work and from the owner of copyright in the translation or
ing conditions on the valve (such as higher blood pressure in adaptation.
2162  Circulation  May 27, 2014

Valvular leaflets are populated on the surface by endothelial dilatation; and thin, slightly elongated chordae. Patients fre-
cells (VECs) and by interstitial cells (VICs) within the valve quently present at an older age with chordal rupture and flail
(Figure 3). VICs, which originate from endothelial progenitor leaflet after a shorter (if any) clinical history. Although most of
cells, are considered the principal initiators of collagen synthe- the mitral leaflet is thin, localized myxomatous degeneration
sis and degradation in the valve leaflets.65 Quiescent VICs are and thickening occur within the flail scallop, mainly of the
noncontractile, α-smooth muscle actin–negative, fibroblast- posterior leaflet.70 Although Barlow’s disease and fibroelastic
like cells that can synthesize and degrade matrix enzymes.65 deficiency are treated with very different surgical approaches,
Enzymes secreted by the VICs include matrix metalloprotein- it is unclear whether they represent 2 histopathological fea-
ases (MMPs) such as collagenase (MMP-1) and gelatinases tures of the same syndrome or 2 genetically distinct entities.
(MMP2 and MMP9), as well as tissue inhibitors of MMPs.65
Quiescent VICs maintain a tight balance between degradation Molecular Biology of Valve Changes in MVP
and synthesis of the matrix proteins, thus allowing normal Some clues to the various signaling pathways involved in
valve leaflet strength and function. abnormal valve biology in MVP can be gleaned from our
Myxomatous degeneration is characterized by the expansion understanding of normal heart valve development. Early sep-
of the middle spongiosa layer of the valve (resulting from an tation of the cardiac tube into distinct chambers is achieved
accumulation of proteoglycans), structural alterations of colla- through regional swellings of the ECM, known as cardiac
gen in all components of the leaflet, and structurally abnormal cushions, which form the primordial valves.66 Reciprocal sig-
chordae.3,4 Dysregulation of ECM components plays a key role naling between the endocardial and myocardial cell layers in
in mediating these changes. In MVP, the VICs acquire proper- the cardiac cushion (mediated in part by members of the TGF-
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

ties of activated myofibroblasts characterized by the expression β family) induces a transformation of the endothelial cells
of vimentin and α-smooth muscle actin but not SM1 or SM2 (VECs) into interstitial or mesenchymal cells (VICs).71 This
(markers of differentiated smooth muscle cells).3,66 Activated transformation is also known as endothelial to mesenchymal
myofibroblasts are responsible for increased concentrations of transition.66 Sox9 is a transcription factor activated when the
various proteolytic enzymes, including MMPs, which degrade endothelial cells undergo mesenchymal transformation, and
collagen and elastin at a rate exceeding the rate of production Sox9-deficient mesenchymal cells fail to express ErbB3, an
seen in quiescent VICs.67 In addition, cells staining for the enzyme required for the proliferation of cardiac cushion cells.72
pan-hematopoietic marker CD45+ can also be found in myxo- The mesenchymal cells then migrate into the cardiac cushions
matous valve tissue66 and may represent fibrocytes capable of and differentiate into the fibrous tissue of the valves.72
differentiating into myofibroblasts that can both secrete matrix Several genes have been shown to play pivotal roles in the
and degrade collagen and elastin. formation of the heart valves: calcineurin, with the signaling
The chordae tendineae in myxomatous valves do not appear and downstream activation of a family of transcription factors
to have increased cellularity, although they contain increased called nuclear factor of activated T cells (the absence of acti-
amounts of glycosaminoglycans.68 In a study by Grande-Allen vation of the nuclear factor of activated T cells leads to fatal
et al,68 myxomatous chordae contained significantly more defects in cardiac valve formation)73; Wnt/β-catenin signal-
chondroitin/dermatan 6-sulfate and slightly more hyaluronan ing, which determines the fate of the endocardial cells during
than control chordae. In contrast to leaflets, which contained valve development;74 fibroblast growth factor-4 (FGF-4); the
predominantly hyaluronan, the predominant glycosamino- homeobox gene Sox4; and the downstream modulator of TGF-
glycan class in chordae was chondroitin/dermatan sulfate. β superfamily signaling SMAD6,67,75 the disruption of which
Myxomatous chordae contained more water and less col- leads to abnormally thickened, gelatinous valves. Defects in ≥1
lagen than control chordae. These findings may account for of these genes and their signaling cascades may also conceiv-
the reduced tensile strength at which the valve leaflets, and ably lead to myxomatous change and mechanically weakened
especially the chordae, fail in MVP.69 Chordal rupture is a valves in adult life.66 Similar to syndromic MVP (see below),
frequent pathological finding in MVP and may be secondary TGF-β upregulation appears to have a pivotal role in various
to mechanical weakening of the chordae, combined with the biological pathways in the pathogenesis of primary or nonsyn-
abnormal hemodynamic stresses arising from the redundancy dromic MVP. Specifically, TGF-β is known to activate VICs
of the valve leaflets.68,69 toward a pathological synthetic phenotype, as shown both in
Finally, degenerative processes have 2 main histological animal models76 and in human in vitro studies. Geirsson et al77
phenotypes recognized in the surgical literature70: diffuse demonstrated TGF-β signaling dysregulation in clinical speci-
myxomatous degeneration (or Barlow’s disease) and fibroelas- mens of sporadic MVP cases undergoing MV repair. TGF-
tic deficiency. Barlow’s disease is characterized by thickened β–induced ECM production in cultured valvular interstitial
and diffusely redundant myxomatous leaflet tissue with dis- cells was dependent on SMAD2/3 and p38 signaling and was
rupted collagen and elastic layers, leading to prolapse of most inhibited by angiotensin II receptor blockers. In another study
of the mitral leaflet segments, severe mitral annular enlarge- of human MVP surgical specimens by Hulin et al,78 upregula-
ment, and elongated (rarely ruptured) chordae.70 Patients usu- tion of TGF-β2 was secondary to the reduced expression of
ally present young or are middle-aged at the time of surgery metallothioneins, genes involved in the response to oxidative
after a long history of murmur or MR. Fibroelastic deficiency stress. In turn, TGF-β2 upregulation led to downregulation of
is characterized by decreased connective tissue deficient in genes of the ADAMTS family (responsible for degradation
collagen, elastin, and proteoglycans; thin, smooth, translu- of proteoglycans), ultimately causing excessive ECM remod-
cent leaflets without excess tissue and only moderate annulus eling. Finally, upregulation of bone morphogenetic protein
Delling and Vasan   Epidemiology and Pathophysiology of MVP   2163

has also been shown to mediate the activation of VICs from excess TGF-β activity has implications for the identification
healthy quiescent cells to a pathological synthetic phenotype of novel genes that may be implicated in the pathogenesis of
in microarray data of clinical MVP specimens.79 MV disease. Furthermore, these findings raise the possibil-
Endothelial to mesenchymal transition can be induced ity that myxomatous MV disease could potentially be treated
in vitro and is increased in vivo in response to mechanical with therapies that reduce TGF-β overexpression. Although
stretch, suggesting that endothelial to mesenchymal transition treatment with TGF-β–neutralizing antibodies successfully
not only occurs in normal valve development but also plays a improves many phenotypic manifestations of MFS in murine
role in adaptation to pathophysiologic conditions.66 The abil- models, this therapy cannot be readily translated to the treat-
ity of valves to remodel and “reset the clock” in response to a ment of the human form of the disease in the absence of US
reduction in mechanical stretch can be derived from the clini- Food and Drug Administration–approved “humanized” anti-
cal context: MV repair is typically very durable, suggesting bodies that block TGF-β. Because of the extensive interac-
that an annuloplasty ring, by reducing the mechanical load on tions between angiotensin II and TGF-β signaling pathways,
the valves and chordae, improves long-term valve function. mice with a FBN1 mutation were treated with losartan, a selec-
The mesenchymal differentiation potential of the VECs can tive angiotensin II type 1 receptor antagonist, in an attempt
be directed toward their transformation into osteogenic and to reduce TGF-β signaling.86 Remarkably, the mutant mice
chondrogenic phenotypes, reflecting an ability of these cells treated with losartan had dramatic improvement in their rate
to generate VICs that reside in specific regions of the valve.66 of aortic root growth compared with mutant mice treated with
The multilineage differentiation potential of the VECs, com- β-adrenergic receptor blockers in doses with similar hemody-
bined with a robust capacity for self-renewal, strongly sug- namic effects.86 In the recently published Cozaar in Marfan
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

gests that at least a subset of the VECs are likely progenitor Patients Reduces Aortic Enlargement (COMPARE) trial,87
cells.66 Such progenitor cells may be essential for the health losartan reduced aortic root dilatation rate compared with pla-
and longevity of the valve and may become activated during cebo in adult humans with MFS. Another trial comparing the
the disease process. Whether these cells can be harnessed or effect of losartan and atenolol on aortic dilatation and MVP in
manipulated to prevent or limit valve disease is an exciting children and young adults with MFS is ongoing.88,89
direction for future translational research. Among other connective tissue disorders commonly asso-
Altered ECM turnover is also crucial in the pathogenesis ciated with MV disease, the Loeys-Dietz syndrome typically
of ruptured chordae tendineae in MVP. Although the heart is involves marked arterial tortuosity and distinct craniofacial
a vascular-rich organ, most of the cardiac valve complex is abnormalities such as hypertelorism and cleft palate. It is
avascular (similar to cartilage and tendons).80 Work by Kimura caused by heterozygous mutations in either the TGFBR1 gene
et al81 showed a differential local expression of tenomoduline or the TGFBR2 gene that encode subunits of the TGF-β recep-
(a recently isolated antiangiogenic factor) measured in the tor.39 Immunostaining of diseased tissues from affected indi-
chordae tendinae. Specifically, tenomoduline is locally absent viduals with the syndrome shows evidence of increased TGF-β
in the ruptured zones of the chordae, favoring abnormal vessel activity such as increased nuclear accumulation of phosphory-
formation also in combination with enhanced expression of lated SMAD2 and increased connective tissue growth factor,
vascular endothelial growth factor-A. Moreover, in contrast which is induced by TGF-β.39 In the aneurysms-osteoarthritis
to what was observed in normal or nonruptured areas, higher syndrome, characterized by aortic aneurysms, arterial tortu-
numbers of inflammatory cells positive for CD11b, CD14, and osity, craniofacial features (similar to the Loeys-Dietz), and
vimentin and with an augmented expression of MMP-2 and early-onset osteoarthritis,42 inactivating heterozygous muta-
-13 were detected in association with the downregulation of tions in MADH3 encoding SMAD3 (positive regulator of
tenomoduline.81 TGF-β signaling) have been identified, once again pointing
to the link between TGF-β overexpression and myxomatous
Molecular Biology of Syndromic MVP MV disease.
Syndromic MVP associated with connective tissue disorders
has been shown to manifest myxomatous changes similar Nonmyxomatous Valve Elongation: HCM and
to primary or nonsyndromic MVP.82 MFS is associated with MV Disease
mutations in the FBN1 gene (chromosome 15q15-q21).83 It Elongation and pathological thickening of the MV are com-
can also be caused by inactivating mutations of the TGFBR2 monly seen in HCM, a genetic disorder typically caused by
gene, located on chromosome 3p24.2-p25.84 A recent study mutations in sarcomere genes and characterized by unex-
addressed the importance of TGF-β dysregulation in the con- plained thickening of the LV walls and by LV outflow tract
nective tissue for the development of MVP in MFS. Ng et obstruction.55 For years, it has been recognized that the sim-
al85 tested the hypothesis that the FBN1–TGF-β pathway is ple impingement of the MV on the interventricular septum
implicated in the pathogenesis of MVP in a murine model of (resulting from high systolic velocities in the vicinity of the
MFS. Increased TGF-β signaling was observed in the prolaps- leaflets generated by the predominant upper septal hypertro-
ing valves of FBN1-deficient mice. Treatment with a TGF-β– phy) contributes to systolic anterior motion of the MV or SAM
neutralizing antibody successfully normalized both the length (Venturi effect).55 Whereas the Venturi effect likely contributes
and the thickness of the MV leaflets in these mutant mice, to the propagation and worsening of SAM, it is not adequate
further supporting the hypothesis that the valve abnormalities to initiate SAM, indicating that complementary mechanisms
in this mouse model were caused by increased TGF-β lev- centered on structural MV abnormalities likely contribute
els.85 The responsiveness of the MV length and thickness to to SAM.55 Experimental in vitro studies and computational
2164  Circulation  May 27, 2014

models have demonstrated that isolated anterior and internal yield of genetic studies.26 On the basis of this newer MVP
displacement of the papillary muscles, combined with leaflet phenotype, 3 loci for autosomal-dominant, nonsyndromic
elongation (especially of the posterior leaflet), is able to rec- MVP have been identified on chromosomes 16, 11, and
reate SAM.55 The degree of SAM is related to leaflet length, 13.95–97 Although filamin-A has been identified as causing an
even in the absence of septal hypertrophy. Typically, basal and X-linked form of MVP,14 the genes for the more common form
midanterior MV leaflet elongation causes SAM with prolapse, of autosomal-dominant MVP have yet to be defined (Table 1).
whereas distal anterior leaflet elongation creates SAM with In 1999, the first genetic locus for MVP was mapped to
a mobile flap. Leaflet elongation without papillary muscle chromosome 16p11.2-p12.1 (MMVP1) in a family with the
displacement creates prolapse.55 Several mechanisms may trait segregating in an autosomal-dominant fashion.95 Genetic
contribute to MV disease in HCM, including the primary sar- linkage studies yielded maximum multipoint LOD scores of
comeric gene mutation, the response to shear stress in a tur- 5.4 and 5.6. This was confirmed by haplotype analysis dem-
bulent outflow tract, and concomitant but unrelated familial onstrating that a chromosomal region of ≈5 cM containing the
MV disease.55 Paracrine factors such as periostin have also locus (a genetic distance equivalent to 5 million DNA base
been implicated in the pathogenesis of MV disease in HCM.90 pairs) was present in all affected individuals.95 In 2003, Freed
Periostin is a TGF-β1–inducible secreted protein origi- et al96 identified a second locus for MVP (MMVP2) at chromo-
nally identified in mouse osteoblasts.91 In the heart, perios- some 11p15.4. The risk haplotype comprised a 4.3-cM region
tin is expressed physiologically in embryonic cardiac valves, on this chromosome. In 2005, a new locus for autosomal-
whereas it is re-expressed abundantly in the adult left ventricle dominant MVP was mapped to chromosome 13q31.3-q32
after pressure overload or myocardial infarction.91 Markedly with a multipoint LOD score of 3.17 (MMVP3).97 Haplotype
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

elevated levels of periostin are indeed secreted by nonmyocyte analysis showed that a portion of the chromosome contain-
elements in the left ventricular walls and expressed in HCM ing the locus was present in all affected members of the fam-
mice.90 Periostin promotes VIC proliferation, differentiation, ily. The discovery of MMVP3 not only confirmed the genetic
and matrix production, therefore potentially driving leaflet heterogeneity of MVP but also provided important clinical
elongation in HCM.55,90 lessons. Specifically, phenotyping of the chromosome 13 ped-
igree revealed a spectrum of expression that included valve
Genetics of Nonsyndromic MVP morphologies previously considered to be normal variants
A familial basis for MVP has long been recognized, with an but now for the first time were recognized as having the same
autosomal-dominant mode of inheritance, a variable pen- genetic substrate in the familial context.97 Prodromal mor-
etrance influenced by age and sex, and a marked heterogene- phologies shared 2 salient features with fully diagnostic MVP:
ity of clinical presentation even among the affected members an anteriorly displaced coaptation point and posterior leaflet
within a family.12,13,92 asymmetry (Figure 4). The term prodromal used in the initial
Because MVP is found in many but certainly not all patients literature may not be ideal outside the familial context because
with MFS, it was suggested that primary MVP may be due its prognostic significance is unclear at this time. Therefore,
to a mutation of FBN1. However, studies have failed to link these morphologies are better defined as abnormal anterior
nonsyndromic familial MVP with variants in fibrillar or other coaptation (AAC) to underline their similarity with fully diag-
collagen genes.93,94 Negative genetic linkage results may have nostic MVP. Individuals with minimal systolic displacement
been related to a lack of systematic examination of the entire shared the posterior leaflet asymmetry with individuals with
human genome and to phenotypic ambiguity. More recently, full-blown MVP, but their coaptation point was posterior (as
our understanding of the 3-dimensional shape of the MV has in normal individuals). Individuals with AAC and minimal
improved the specificity of MVP diagnosis and in turn the systolic displacement shared either the complete or a major

Table 1.  Summary of Linkage Studies of Nonsyndromic MVP


MMVP1 MMVP2 MMVP3 XMVD
Probands, n 17 1 1 N/A
Pedigrees, n 4 1 1 1
Total subjects in pedigrees with 79 28 47 92
echocardiography+DNA, n
Affected individuals, n 25 12 9 21
Ethnicity Ashkenazi Jewish (pedigree 1), Western European Western European French origin
western France (pedigrees 2,4), descent descent
eastern France (pedigree 3)
LOD score >5 >3 >3 >6
Chromosome 16 11 13 X
Gene map locus 16p12.1-p11.2 11p15.4 13.q31.3-q32.1 Xq28
MMVP indicates myxomatous mitral valve prolapse; MVP, mitral valve prolapse; N/A, non applicable; and XMVD, X-linked
myxomatous valvular dystrophy. Adapted from Grau et al.8 Copyright © 2007 John Wiley & Sons, Inc. Authorization for this
adaptation has been obtained both from the owner of the copyright in the original work and from the owner of copyright in the
translation or adaptation.
Delling and Vasan   Epidemiology and Pathophysiology of MVP   2165
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

Figure 4. Two-dimensional parasternal long-axis images of posteriorly coapting leaflets (anterior leaflet [AL]; posterior leaflet [PL]) in a
normal individual (A) vs increased coaptation height and an elongated posterior leaflet in an individual with abnormal anterior coapta-
tion (AAC) features (B) and in a patient with bileaflet mitral valve prolapse (MVP) into the left atrium (LA; C). Schematics (D) showing the
projections of anterior (A) and posterior (P) leaflets onto the mitral annular diameter (D). C indicates the coaptation height relative to the
annulus and is calculated as P/D or C/LVID, where LVID is left ventricular internal diameter. AO indicates aorta; LV, left ventricle; and RV,
right ventricle.

portion of the at-risk haplotype with fully diagnostic MVP. myxomatous valvular dystrophy gene, with a highly informa-
This same AAC morphology was also observed in the fam- tive LOD score of 6.57 (Table 1). Coupling genealogical sur-
ily linked to chromosome 11.97 Quantitatively, the height of veys of the larger family with linkage analyses, Kyndt et al14
coaptation relative to the annulus or left ventricular diameter refined the previously mapped locus on chromosome Xq28
(Figure 4) correlated well with the ratio of posterior to ante- to a 2.5-Mb region. Screening of candidate genes revealed a
rior leaflet length (r=0.83–0.85) in the members of the family P637Q missense mutation in the filamin-A gene in the affected
with a genetic linkage signal on chromosome 11. Thus, the members of the larger family.14 Mutational analyses of the
spectrum of valvular abnormalities (AAC or minimal systolic filamin-A gene in the other families identified 3 additional fila-
displacement) may represent, in the familial context, early dis- min-A gene mutations: 2 more missense mutations (G288R,
ease expression in gene carriers, a stage of progression, or the V711D) and a 1944–base pair in-frame deletion.14 Both male
result of disease-modifying factors. and female carriers were affected, but the affected female car-
Recognizing early forms of MVP may be important because riers had a less severe phenotype.
the condition often manifests clinically in the fifth or sixth Filamins are large cytoplasmic proteins that play an
decade of life as a severe cardiac event.5,6 It is conceivable that important role in cross-linking cortical actin filaments into a
earlier targeted intervention to reduce hemodynamic stresses dynamic 3-dimensional structure and thereby transmit extra-
on the MV leaflets in genetically susceptible individuals (as cellular signals through their interactions with the integrin
shown in a murine model of MFS with aortic dilatation and in receptors.99 These proteins not only serve a structural role in
surgical specimens of nonsyndromic MVP)77,85,86 may poten- cytoskeletal organization but also appear to serve as hubs or
tially prevent the progression of MVP and severe MR requir- docking platforms for second messengers important in signal
ing surgery, although this premise remains to be tested. transduction. The filamin group of proteins contains 3 mem-
A rare form of myxoid heart disease, X-linked myxomatous bers: A, B, and C. Filamin-A and filamin-B are reported as
valvular dystrophy, was first described >3 decades ago.14,98 It ubiquitously expressed in tissues, whereas filamin-C expres-
is characterized by multivalvular myxomatous degeneration, sion is restricted to the cardiac and the skeletal muscle.99
although the histopathological features of the MV do not dif- The filamins are present as homodimeric or heterodimeric
fer significantly from the severe form of autosomal-dominant Y-shaped proteins with each chain consisting of an actin-
MVP. In 1 large family, X-linked myxomatous valvular dys- binding region at the amino terminus. The core of the pro-
trophy cosegregated with hemophilia A.98 This relatively large tein consists of 24 highly homologous immunoglobulin-like
familial study that included >92 individuals over 4 genera- repeats followed by a carboxyl integrin binding domain.99
tions, with a full penetrance for men (men were either clearly Gene knockout studies have indicated the importance of these
affected or not), was the first investigation that mapped this proteins in diverse developmental processes, and filamin-A
rare clinical dystrophy to a sex chromosome, Xq28. Thus, the appears to be the major family member responsible for car-
genetic linkage analysis, facilitated by the X-linked mode of diac and vascular development.100 Hemizygous mice for the
inheritance and by the linkage with a mild form of hemophilia filamin-A–null allele show embryonic lethality and a wide
A in this pedigree, permitted rapid mapping of this X-linked range of cardiovascular malformations, including incomplete
2166  Circulation  May 27, 2014

septation of the outflow tract that leads to a common arterial Table 2.  Summary of Future Research Directions
trunk and double outlet, abnormally thickened and malformed
MVP pathophysiology and genetics
outflow tract valves, atrial and ventricular septal defects, type
 Identify the genetic variants responsible for nonsyndromic autosomal-
B interruption of the aortic arch, abnormal endothelial organi-
dominant MVP
zation in blood vessels, abnormal vascular permeability, and  Develop functional studies with animal models to corroborate the clinical
thickening of the MV.100 Compensation of the other filamin relevance of identified genetic variants
genes does not seem to occur, nor have cardiovascular defects  Develop new nonsurgical therapies based on a better understanding of
been described in mouse mutants for either the filamin-B or genetic determinants of MVP and related biochemical pathways
the filamin-C gene.101,102 Filamin-A appears as a functional MVP epidemiology
hub in many signaling pathways that may contribute to the  Better characterize the natural history of nondiagnostic MVP morphologies
development of valvular disease. Filamin-A coordinates local-  Identify genetic and environmental risk factors responsible for accelerated
ization and activation of TGF-β receptor–activated SMADs, MVP progression
particularly SMAD2, to act as a positive regulator of TGF-  Examine whether specific risk factors act differently at different stages of
MVP progression
β signaling.103 One potential mechanism underlying cardiac
 Evaluate the duration of the different stages of MVP progression
valvular dystrophy could involve increased TGF-β signaling  Assess whether response to future nonsurgical therapies (losartan?) varies
secondary to perturbed filamin-A–SMAD interactions with on the basis of different MVP stages (nondiagnostic MVP morphologies vs
consequent dysregulation of endothelial-to mesenchymal full-blown MVP)
transition.104 Filamin-A mutations may provide a link between MVP indicates mitral valve prolapse.
MFS and nonsyndromic MVP, conditions that are character-
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

ized by increased TGF-β signaling. Specifically, myxomatous


MVs found in fibrillin-1–deficient mice (which model MFS) valve development.100 Further opportunities for understand-
display excessive TGF-β activation and upregulated expres- ing human MV disease may derive from a study of the King
sion of filamin-A.85,105 Charles Spaniel, a species in which MVP naturally occurs
with a frequency of 1% to 5%, similar to that in humans.106 In
addition, the zebrafish has recently been identified as an ideal
Remaining Questions model for genetic knockdown experiments and understanding
MVP Pathophysiology and Genetics of valve development.107 Although zebrafish display differ-
In this review, we present a unifying theory for the pathogen- ences in atrioventricular canal and valve morphology com-
esis of MVP based on our knowledge of the biology of valves pared with other model organisms (such as mice and chicken),
and the dynamic interplay of differentiating cells and growth they share the same molecular and cellular mechanisms for
factors. However, a full understanding of the molecular basis valve development with humans.107 Thus, several signaling
of MVP requires 2 additional steps (Table 2): identification of pathways implicated in MVP also contribute to atrioven-
the genetic variants responsible for nonsyndromic autosomal- tricular canal formation in zebrafish (eg, the TGF-β/SMAD,
dominant MVP, including the role of both susceptibility and Notch1b, and vascular endothelial growth factor/calcineurin/
modifier genes, and functional studies with animal models to nuclear factor of activated T cells signaling pathways).107
corroborate the clinical relevance of identified mutations. Identification of the mutations responsible for autosomal-
MVP appears to be the result of multiple genetic pathways, dominant MVP and their corroboration through functional
as illustrated by the identification of several genes in syndromic studies will provide a potential screening tool to help clini-
MVP83,84 and 3 loci for nonsyndromic MVP.95–97 The identifica- cians identify asymptomatic patients who may progress to
tion of filamin-A mutations in an X-linked form of valvular significant MR, perhaps among the individuals with diagnos-
dystrophy14 highlights the importance of the cytoskeleton not tic MVP without MR or among AAC individuals who do not
only in providing structural integrity but also in critical cellular meet standard diagnostic criteria but share features of exces-
signaling pathways, specifically the TGF-β pathway. Advances sive leaflet motion with the fully affected family members
in DNA sequencing technologies may lead to the identifica- (Figure 5).97 Advances in the understanding of MVP patho-
tion of the MMVP1, MMVP2, and MMVP3 genes in the near physiology outlined in this review may lead to new medical
future. Large-scale collections of MVP patients and genome- therapies aimed at preventing the progression of disease by
wide association studies will allow identification of additional targeting different cells and signaling pathways (Figure 5):
MVP genes and will finally elucidate the pathways leading to from excessive TGF-β signaling (similar to what has been
the occurrence of MVP. Identification of the genes involved shown in the MFS and in cultured human MV cells of non-
in the development of MVP is important because the disease syndromic MVP, in which angiotensin I receptor blockade
typically manifests later in life, and earlier intervention in sus- leads to regulation of TGF-β and limitation of clinical disease
ceptible individuals may potentially prevent progression to a progression) to manipulation of progenitor cells (among the
clinically severe stage, a premise that remains to be tested. In endothelial cells responsible for both valvular embryogen-
vitro studies of surgical specimens have shown for the first time esis and response to hemodynamic stress in adult life) to the
that the myxomatous changes characteristic of MVP are phar- development of tissue-engineered heart valves. Such medical
macologically preventable,77 which offers great hope for the interventions may hold the potential for limiting disease pro-
development of therapies based on future genetic discoveries. gression at an early stage in nondiagnostic MVP phenotypes
Mouse models have proved to be essential for demon- or avoiding surgical treatment and clinical complications in
strating the importance of filamin-A mutations in cardiac fully diagnostic MVP individuals (Figure 5).
Delling and Vasan   Epidemiology and Pathophysiology of MVP   2167

Figure 5. Proposed temporal spectrum of mitral


valve (MV) prolapse (MVP) progression with poten-
tial interventions/nonsurgical therapies depending
on progression stage (bottom). A and P indicate
projections of anterior and posterior leaflets onto
the mitral annulus; C; mitral leaflet coaptation
height; MR, mitral regurgitation; MSD, minimal sys-
tolic displacement; and TGF-β, transforming growth
factor-β.
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

Progression of MVP Conclusions


Although the natural history of MVP has been studied since MVP is a common clinical phenotype and remains the most
the 1980s, these investigations have focused on individuals common valvular pathology requiring surgery. Multiple loci
with fully diagnostic MVP and its clinical outcomes, includ- for autosomal-dominant nonsyndromic MVP and a gene
ing cardiac death, heart failure, endocarditis, or severe MR responsible for a rare X-linked form of MVP have been dis-
requiring surgery.5,6 The natural history of early echocar- covered. Studies in a mouse MFS model and in clinical speci-
diographic stages of MVP leading to diagnostic MV leaflet mens of excised myxomatous MVs have underlined the role
displacement and to subsequent clinical outcomes has yet of excessive TGF-β signaling in the development of degen-
to be described in both tertiary care and community-based erative MV disease and the potential of angiotensin I recep-
studies (Table 2). In the familial context, previously non- tor blockade in limiting MVP progression. These discoveries
diagnostic morphologies that share features of excessive overall have exponentially accelerated our understanding of
leaflet motion with fully diagnostic MVP have been shown MVP. However, many questions remain unanswered in rela-
to represent mild or early stages of phenotypic expression tion to both MVP pathophysiology and epidemiology, and
in gene carriers.97 The existence of early MVP morpholo- future studies are needed to address these important issues.
gies in the general population would raise the possibility of
echocardiographic screening and provide an opportunity for Sources of Funding
potential intervention at an early phase of disease. As sug- This work was supported by the Founders Affiliate American Heart
gested in Figure 5, the progression of MVP may occur in Association Clinical Research Program, by the National Institute
stages over a lifetime, beginning with a genetic substrate, of Health K23HL116652 (Dr Delling) and by the National Heart,
leading to mild, nondiagnostic valve morphologies, develop- Lung, and Blood Institute’s Framingham Heart Study (contract
N01-HC-25195) R01HL080124 and RO1HL0107385 (Dr Vasan).
ing into full expression of the MVP phenotype, and culmi-
nating in severe MR requiring valve surgery. The duration of
the individual stages of the disease can range from months to Disclosures
None.
years, the shorter progression duration being a consequence
of the development of a flail mitral leaflet. Various risk fac-
tors, including genetic modifiers, environmental factors References
(smoking, diet, body mass index, hypertension), and non- 1. Freed LA, Levy D, Levine RA, Larson MG, Evans JC, Fuller DL, Lehman
B, Benjamin EJ. Prevalence and clinical outcome of mitral-valve prolapse.
modifiable characteristics such as race or sex, may influence N Engl J Med. 1999;341:1–7.
the progression from one stage of MVP to the other and the 2. Devereux RB, Jones EC, Roman MJ, Howard BV, Fabsitz RR, Liu JE,
duration of the different stages. Palmieri V, Welty TK, Lee ET. Prevalence and correlates of mitral valve
prolapse in a population-based sample of American Indians: the Strong
Several questions about the epidemiology of MVP remain Heart Study. Am J Med. 2001;111:679–685.
unanswered (Table 2). Specifically, it is unknown whether 3. Rabkin E, Aikawa M, Stone JR, Fukumoto Y, Libby P, Schoen FJ.
early, nondiagnostic MVP morphologies progress within or Activated interstitial myofibroblasts express catabolic enzymes and
outside the familial context; whether some risk factors more mediate matrix remodeling in myxomatous heart valves. Circulation.
2001;104:2525–2532.
than others contribute to progression and at which stages; and 4. Tamura K, Fukuda Y, Ishizaki M, Masuda Y, Yamanaka N, Ferrans VJ.
whether the response to future nonsurgical therapies varies as Abnormalities in elastic fibers and other connective-tissue components of
MVP progresses from early disease to significant MR. Further floppy mitral valve. Am Heart J. 1995;129:1149–1158.
5. Devereux RB, Kramer-Fox R, Shear MK, Kligfield P, Pini R, Savage
longitudinal studies with a focus on specific clinical, demo-
DD. Diagnosis and classification of severity of mitral valve prolapse:
graphic, and ethnic subgroups are needed to answer these methodologic, biologic, and prognostic considerations. Am Heart J.
important questions. 1987;113:1265–1280.
2168  Circulation  May 27, 2014

6. Zuppiroli A, Rinaldi M, Kramer-Fox R, Favilli S, Roman MJ, valve prolapse in the general population, 1: epidemiologic features: the
Devereux RB. Natural history of mitral valve prolapse. Am J Cardiol. Framingham Study. Am Heart J. 1983;106:571–576.
1995;75:1028–1032. 24. Savage DD, Devereux RB, Garrison RJ, Castelli WP, Anderson SJ, Levy
7. Avierinos JF, Detaint D, Messika-Zeitoun D, Mohty D, Enriquez-Sarano D, Thomas HE, Kannel WB, Feinleib M. Mitral valve prolapse in the gen-
M. Risk, determinants, and outcome implications of progression of mitral eral population, 2: clinical features: the Framingham Study. Am Heart J.
regurgitation after diagnosis of mitral valve prolapse in a single commu- 1983;106:577–581.
nity. Am J Cardiol. 2008;101:662–667. 25. Warth DC, King ME, Cohen JM, Tesoriero VL, Marcus E, Weyman AE.
8. Grau JB, Pirelli L, Yu PJ, Galloway AC, Ostrer H. The genetics of mitral Prevalence of mitral valve prolapse in normal children. J Am Coll Cardiol.
valve prolapse. Clin Genet. 2007;72:288–295. 1985;5:1173–1177.
9. Barlow JB, Pocock WA. The significance of late systolic murmurs and 26. Levine RA, Stathogiannis E, Newell JB, Harrigan P, Weyman AE.

mid-late systolic clicks. Md State Med J. 1963;12:76–77. Reconsideration of echocardiographic standards for mitral valve prolapse:
10. Criley JM, Lewis KB, Humphries JO, Ross RS. Prolapse of the mitral lack of association between leaflet displacement isolated to the apical four
valve: clinical and cine-angiocardiographic findings. Br Heart J. chamber view and independent echocardiographic evidence of abnormal-
1966;28:488–496. ity. J Am Coll Cardiol. 1988;11:1010–1019.
11. Waller BF, Morrow AG, Maron BJ, Del Negro AA, Kent KM, McGrath FJ, 27. Carpentier A. Cardiac valve surgery: the “French correction”. J Thorac
Wallace RB, McIntosh CL, Roberts WC. Etiology of clinically isolated, Cardiovasc Surg. 1983;86:323–337.
severe, chronic, pure mitral regurgitation: analysis of 97 patients over 30 28. Foster GP, Isselbacher EM, Rose GA, Torchiana DF, Akins CW, Picard
years of age having mitral valve replacement. Am Heart J. 1982;104(pt MH. Accurate localization of mitral regurgitant defects using multiplane
1):276–288. transesophageal echocardiography. Ann Thorac Surg. 1998;65:1025–1031.
12. Devereux RB, Brown WT, Kramer-Fox R, Sachs I. Inheritance of mitral 29. Kumar N, Kumar M, Duran CM. A revised terminology for recording sur-
valve prolapse: effect of age and sex on gene expression. Ann Intern Med. gical findings of the mitral valve. J Heart Valve Dis. 1995;4:70–75.
1982;97:826–832. 30. Shah PM. Current concepts in mitral valve prolapse–diagnosis and man-
13. Strahan NV, Murphy EA, Fortuin NJ, Come PC, Humphries JO.
agement. J Cardiol. 2010;56:125–133.
Inheritance of the mitral valve prolapse syndrome: discussion of a three- 31. Biaggi P, Jedrzkiewicz S, Gruner C, Meineri M, Karski J, Vegas A, Tanner
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

dimensional penetrance model. Am J Med. 1983;74:967–972. FC, Rakowski H, Ivanov J, David TE, Woo A. Quantification of mitral
14. Kyndt F, Gueffet JP, Probst V, Jaafar P, Legendre A, Le Bouffant F, Toquet valve anatomy by three-dimensional transesophageal echocardiography
C, Roy E, McGregor L, Lynch SA, Newbury-Ecob R, Tran V, Young I, in mitral valve prolapse predicts surgical anatomy and the complexity of
Trochu JN, Le Marec H, Schott JJ. Mutations in the gene encoding fila- mitral valve repair. J Am Soc Echocardiogr. 2012;25:758–765.
min A as a cause for familial cardiac valvular dystrophy. Circulation. 32. Han Y, Peters DC, Salton CJ, Bzymek D, Nezafat R, Goddu B, Kissinger
2007;115:40–49. KV, Zimetbaum PJ, Manning WJ, Yeon SB. Cardiovascular magnetic
15. Bonow RO, Carabello BA, Chatterjee K, de Leon AC Jr, Faxon DP, Freed resonance characterization of mitral valve prolapse. JACC Cardiovasc
MD, Gaasch WH, Lytle BW, Nishimura RA, O’Gara PT, O’Rourke Imaging. 2008;1:294–303.
RA, Otto CM, Shah PM, Shanewise JS. 2008 Focused update incor- 33. Gelfand EV, Hughes S, Hauser TH, Yeon SB, Goepfert L, Kissinger
porated into the ACC/AHA 2006 guidelines for the management of KV, Rofsky NM, Manning WJ. Severity of mitral and aortic regurgita-
patients with valvular heart disease: a report of the American College tion as assessed by cardiovascular magnetic resonance: optimizing cor-
of Cardiology/American Heart Association Task Force on Practice relation with Doppler echocardiography. J Cardiovasc Magn Reson.
Guidelines (Writing Committee to Revise the 1998 Guidelines for the 2006;8:503–507.
Management of Patients With Valvular Heart Disease): endorsed by the 34. Delling FN, Kang LL, Yeon SB, Kissinger KV, Goddu B, Manning WJ,
Society of Cardiovascular Anesthesiologists, Society for Cardiovascular Han Y. CMR predictors of mitral regurgitation in mitral valve prolapse.
Angiography and Interventions, and Society of Thoracic Surgeons. JACC Cardiovasc Imaging. 2010;3:1037–1045.
Circulation. 2008;118:e523–e661. 35. Sanfilippo AJ, Harrigan P, Popovic AD, Weyman AE, Levine RA. Papillary
16. Bonow RO, Carabello BA, Kanu C, de Leon AC Jr, Faxon DP, Freed muscle traction in mitral valve prolapse: quantitation by two-dimensional
MD, Gaasch WH, Lytle BW, Nishimura RA, O’Gara PT, O’Rourke RA, echocardiography. J Am Coll Cardiol. 1992;19:564–571.
Otto CM, Shah PM, Shanewise JS, Smith SC Jr, Jacobs AK, Adams 36. Gornick CC, Tobler HG, Pritzker MC, Tuna IC, Almquist A, Benditt DG.
CD, Anderson JL, Antman EM, Fuster V, Halperin JL, Hiratzka LF, Electrophysiologic effects of papillary muscle traction in the intact heart.
Hunt SA, Nishimura R, Page RL, Riegel B. ACC/AHA 2006 guidelines Circulation. 1986;73:1013–1021.
for the management of patients with valvular heart disease: a report 37. Dolan AL, Mishra MB, Chambers JB, Grahame R. Clinical and echo-
of the American College of Cardiology/American Heart Association cardiographic survey of the Ehlers-Danlos syndrome. Br J Rheumatol.
Task Force on Practice Guidelines (Writing Committee to Revise the 1997;36:459–462.
1998 Guidelines for the Management of Patients With Valvular Heart 38. Hortop J, Tsipouras P, Hanley JA, Maron BJ, Shapiro JR. Cardiovascular
Disease): developed in collaboration with the Society of Cardiovascular involvement in osteogenesis imperfecta. Circulation. 1986;73:54–61.
Anesthesiologists: endorsed by the Society for Cardiovascular 39. Loeys BL, Schwarze U, Holm T, Callewaert BL, Thomas GH, Pannu
Angiography and Interventions and the Society of Thoracic Surgeons. H, De Backer JF, Oswald GL, Symoens S, Manouvrier S, Roberts AE,
Circulation. 2006;114:e84–e231. Faravelli F, Greco MA, Pyeritz RE, Milewicz DM, Coucke PJ, Cameron
17. Devereux RB, Kramer-Fox R, Brown WT, Shear MK, Hartman N,
DE, Braverman AC, Byers PH, De Paepe AM, Dietz HC. Aneurysm syn-
Kligfield P, Lutas EM, Spitzer MC, Litwin SD. Relation between clini- dromes caused by mutations in the TGF-beta receptor. N Engl J Med.
cal features of the mitral prolapse syndrome and echocardiographically 2006;355:788–798.
documented mitral valve prolapse. J Am Coll Cardiol. 1986;8:763–772. 40. Roman MJ, Devereux RB, Kramer-Fox R, Spitzer MC. Comparison of
18. Boudoulas H. Mitral valve prolapse: etiology, clinical presentation and cardiovascular and skeletal features of primary mitral valve prolapse and
neuroendocrine function. J Heart Valve Dis. 1992;1:175–188. Marfan syndrome. Am J Cardiol. 1989;63:317–321.
19. Chesler E, King RA, Edwards JE. The myxomatous mitral valve and sud- 41. Rubegni P, Mondillo S, De Aloe G, Agricola E, Bardelli AM, Fimiani M.
den death. Circulation. 1983;67:632–639. Mitral valve prolapse in healthy relatives of patients with familial pseudo-
20. Lichodziejewska B, Kłoś J, Rezler J, Grudzka K, Dłuzniewska M, Budaj xanthoma elasticum. Am J Cardiol. 2000;85:1268–1271.
A, Ceremuzyński L. Clinical symptoms of mitral valve prolapse are 42. van de Laar IM, Oldenburg RA, Pals G, Roos-Hesselink JW, de Graaf
related to hypomagnesemia and attenuated by magnesium supplementa- BM, Verhagen JM, Hoedemaekers YM, Willemsen R, Severijnen LA,
tion. Am J Cardiol. 1997;79:768–772. Venselaar H, Vriend G, Pattynama PM, Collee M, Majoor-Krakauer D,
21. Zdrojewski TR, Wyrzykowski B, Krupa-Wojciechowska B. Renin-
Poldermans D, Frohn-Mulder IM, Micha D, Timmermans J, Hilhorst-
aldosterone regulation during upright posture in young men with mitral Hofstee Y, Bierma-Zeinstra SM, Willems PJ, Kros JM, Oei EH, Oostra
valve prolapse syndrome. J Heart Valve Dis. 1995;4:236–241. BA, Wessels MW, Bertoli-Avella AM. Mutations in SMAD3 cause a syn-
22. Weis AJ, Salcedo EE, Stewart WJ, Lever HM, Klein AL, Thomas JD. dromic form of aortic aneurysms and dissections with early-onset osteoar-
Anatomic explanation of mobile systolic clicks: implications for the clini- thritis. Nat Genet.2011;43:121–126.
cal and echocardiographic diagnosis of mitral valve prolapse. Am Heart J. 43. Maron BJ, Epstein SE. Hypertrophic cardiomyopathy: recent observations
1995;129:314–320. regarding the specificity of three hallmarks of the disease: asymmetric
23. Savage DD, Garrison RJ, Devereux RB, Castelli WP, Anderson SJ,
septal hypertrophy, septal disorganization and systolic anterior motion of
Levy D, McNamara PM, Stokes J 3rd, Kannel WB, Feinleib M. Mitral the anterior mitral leaflet. Am J Cardiol. 1980;45:141–154.
Delling and Vasan   Epidemiology and Pathophysiology of MVP   2169

44. Nascimento R, Freitas A, Teixeira F, Pereira D, Cardoso A, Dinis M, 66. Bischoff J, Aikawa E. Progenitor cells confer plasticity to cardiac valve
Mendonça I. Is mitral valve prolapse a congenital or acquired disease? Am endothelium. J Cardiovasc Transl Res. 2011;4:710–719.
J Cardiol. 1997;79:226–227. 67. Salhiyyah K, Yacoub MH, Chester AH. Cellular mechanisms in mitral
45. Hickey AJ, Wilcken DE. Age and the clinical profile of idiopathic mitral valve disease. J Cardiovasc Transl Res. 2011;4:702–709.
valve prolapse. Br Heart J. 1986;55:582–586. 68. Grande-Allen KJ, Griffin BP, Calabro A, Ratliff NB, Cosgrove DM 3rd,
46. Flack JM, Kvasnicka JH, Gardin JM, Gidding SS, Manolio TA, Jacobs Vesely I. Myxomatous mitral valve chordae, II: selective elevation of gly-
DR Jr. Anthropometric and physiologic correlates of mitral valve prolapse cosaminoglycan content. J Heart Valve Dis. 2001;10:325–332.
in a biethnic cohort of young adults: the CARDIA study. Am Heart J. 69. Barber JE, Ratliff NB, Cosgrove DM 3rd, Griffin BP, Vesely I.

1999;138(pt 1):486–492. Myxomatous mitral valve chordae, I: mechanical properties. J Heart Valve
47. Theal M, Sleik K, Anand S, Yi Q, Yusuf S, Lonn E. Prevalence of mitral Dis. 2001;10:320–324.
valve prolapse in ethnic groups. Can J Cardiol. 2004;20:511–515. 70. Fornes P, Heudes D, Fuzellier JF, Tixier D, Bruneval P, Carpentier A.
48. Zua MS, Dziegielewski SF. Epidemiology of symptomatic mitral valve Correlation between clinical and histologic patterns of degenerative mitral
prolapse in black patients. J Natl Med Assoc. 1995;87:273–275. valve insufficiency: a histomorphometric study of 130 excised segments.
49. Ogawa S, Hayashi J, Sasaki H, Tani M, Akaishi M, Mitamura H, Sano Cardiovasc Pathol. 1999;8:81–92.
M, Hoshino T, Handa S, Nakamura Y. Evaluation of combined valvular 71. Brown CB, Boyer AS, Runyan RB, Barnett JV. Requirement of type
prolapse syndrome by two-dimensional echocardiography. Circulation. III TGF-beta receptor for endocardial cell transformation in the heart.
1982;65:174–180. Science. 1999;283:2080–2082.
50. Taub CC, Stoler JM, Perez-Sanz T, Chu J, Isselbacher EM, Picard MH, 72. Akiyama H, Chaboissier MC, Behringer RR, Rowitch DH, Schedl A,
Weyman AE. Mitral valve prolapse in Marfan syndrome: an old topic Epstein JA, de Crombrugghe B. Essential role of Sox9 in the pathway that
revisited. Echocardiography. 2009;26:357–364. controls formation of cardiac valves and septa. Proc Natl Acad Sci U S A.
51. Attias D, Stheneur C, Roy C, Collod-Béroud G, Detaint D, Faivre L, 2004;101:6502–6507.
Delrue MA, Cohen L, Francannet C, Béroud C, Claustres M, Iserin F, 73. de la Pompa JL, Timmerman LA, Takimoto H, Yoshida H, Elia AJ, Samper
Khau Van Kien P, Lacombe D, Le Merrer M, Lyonnet S, Odent S, Plauchu E, Potter J, Wakeham A, Marengere L, Langille BL, Crabtree GR, Mak
H, Rio M, Rossi A, Sidi D, Steg PG, Ravaud P, Boileau C, Jondeau G. TW. Role of the NF-ATc transcription factor in morphogenesis of cardiac
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

Comparison of clinical presentations and outcomes between patients with valves and septum. Nature. 1998;392:182–186.
TGFBR2 and FBN1 mutations in Marfan syndrome and related disorders. 74. Hurlstone AF, Haramis AP, Wienholds E, Begthel H, Korving J, Van Eeden
Circulation. 2009;120:2541–2549. F, Cuppen E, Zivkovic D, Plasterk RH, Clevers H. The Wnt/beta-catenin
52. Lebwohl MG, Distefano D, Prioleau PG, Uram M, Yannuzzi LA,
pathway regulates cardiac valve formation. Nature. 2003;425:633–637.
Fleischmajer R. Pseudoxanthoma elasticum and mitral-valve prolapse. N 75. Schilham MW, Oosterwegel MA, Moerer P, Ya J, de Boer PA, van de
Engl J Med. 1982;307:228–231. Wetering M, Verbeek S, Lamers WH, Kruisbeek AM, Cumano A, Clevers
53. Petrone RK, Klues HG, Panza JA, Peterson EE, Maron BJ. Coexistence of H. Defects in cardiac outflow tract formation and pro-B-lymphocyte
mitral valve prolapse in a consecutive group of 528 patients with hypertro- expansion in mice lacking Sox-4. Nature. 1996;380:711–714.
phic cardiomyopathy assessed with echocardiography. J Am Coll Cardiol. 76. Disatian S, Ehrhart EJ 3rd, Zimmerman S, Orton EC. Interstitial cells from
1992;20:55–61. dogs with naturally occurring myxomatous mitral valve disease undergo
54. Klues HG, Maron BJ, Dollar AL, Roberts WC. Diversity of structural phenotype transformation. J Heart Valve Dis. 2008;17:402–411.
mitral valve alterations in hypertrophic cardiomyopathy. Circulation. 77. Geirsson A, Singh M, Ali R, Abbas H, Li W, Sanchez JA, Hashim S,
1992;85:1651–1660. Tellides G. Modulation of transforming growth factor-beta signaling and
55. Hagège AA, Bruneval P, Levine RA, Desnos M, Neamatalla H, Judge DP. extracellular matrix production in myxomatous mitral valves by angioten-
The mitral valve in hypertrophic cardiomyopathy: old versus new con- sin II receptor blockers. Circulation.2012;126:S189–S197.
cepts. J Cardiovasc Transl Res. 2011;4:757–766. 78. Hulin A, Deroanne CF, Lambert CA, Dumont B, Castronovo V, Defraigne
56. Avierinos JF, Gersh BJ, Melton LJ 3rd, Bailey KR, Shub C, Nishimura JO, Nusgens BV, Radermecker MA, Colige AC. Metallothionein-
RA, Tajik AJ, Enriquez-Sarano M. Natural history of asymptomatic mitral dependent up-regulation of TGF-β2 participates in the remodelling of the
valve prolapse in the community. Circulation. 2002;106:1355–1361. myxomatous mitral valve. Cardiovasc Res. 2012;93:480–489.
57. Rosenhek R, Rader F, Klaar U, Gabriel H, Krejc M, Kalbeck D,
79. Sainger R, Grau JB, Branchetti E, Poggio P, Seefried WF, Field BC,
Schemper M, Maurer G, Baumgartner H. Outcome of watchful Acker MA, Gorman RC, Gorman JH 3rd, Hargrove CW 3rd, Bavaria JE,
waiting in asymptomatic severe mitral regurgitation. Circulation. Ferrari G. Human myxomatous mitral valve prolapse: role of bone mor-
2006;113:2238–2244. phogenetic protein 4 in valvular interstitial cell activation. J Cell Physiol.
58. Ling LH, Enriquez-Sarano M, Seward JB, Orszulak TA, Schaff HV,
2012;227:2595–2604.
Bailey KR, Tajik AJ, Frye RL. Early surgery in patients with mitral 80. Millington-Sanders C, Meir A, Lawrence L, Stolinski C. Structure of chor-
regurgitation due to flail leaflets: a long-term outcome study. Circulation. dae tendineae in the left ventricle of the human heart. J Anat. 1998;192(pt
1997;96:1819–1825. 4):573–581.
59. Enriquez-Sarano M, Avierinos JF, Messika-Zeitoun D, Detaint D, Capps 81. Kimura N, Shukunami C, Hakuno D, Yoshioka M, Miura S, Docheva
M, Nkomo V, Scott C, Schaff HV, Tajik AJ. Quantitative determinants D, Kimura T, Okada Y, Matsumura G, Shin’oka T, Yozu R, Kobayashi J,
of the outcome of asymptomatic mitral regurgitation. N Engl J Med. Ishibashi-Ueda H, Hiraki Y, Fukuda K. Local tenomodulin absence, angio-
2005;352:875–883. genesis, and matrix metalloproteinase activation are associated with the
60. Enriquez-Sarano M, Basmadjian AJ, Rossi A, Bailey KR, Seward JB, rupture of the chordae tendineae cordis. Circulation. 2008;118:1737–1747.
Tajik AJ. Progression of mitral regurgitation: a prospective Doppler echo- 82. Bowers D. Pathogenesis of primary abnormalities of the mitral valve in
cardiographic study. J Am Coll Cardiol. 1999;34:1137–1144. Marfan’s syndrome. Br Heart J. 1969;31:679–683.
61. Grigioni F, Tribouilloy C, Avierinos JF, Barbieri A, Ferlito M, Trojette F, 83. Dietz HC, Cutting GR, Pyeritz RE, Maslen CL, Sakai LY, Corson GM,
Tafanelli L, Branzi A, Szymanski C, Habib G, Modena MG, Enriquez- Puffenberger EG, Hamosh A, Nanthakumar EJ, Curristin SM, Stetten G,
Sarano M; MIDA Investigators. Outcomes in mitral regurgitation due to Meyers DA, Francomano CA. Marfan syndrome caused by a recurrent de
flail leaflets: a multicenter European study. JACC Cardiovasc Imaging. novo missense mutation in the fibrillin gene. Nature. 1991;352:337–339.
2008;1:133–141. 84. Mizuguchi T, Collod-Beroud G, Akiyama T, Abifadel M, Harada N,

62. Ling LH, Enriquez-Sarano M, Seward JB, Tajik AJ, Schaff HV, Bailey Morisaki T, Allard D, Varret M, Claustres M, Morisaki H, Ihara M,
KR, Frye RL. Clinical outcome of mitral regurgitation due to flail leaflet. Kinoshita A, Yoshiura K, Junien C, Kajii T, Jondeau G, Ohta T, Kishino
N Engl J Med. 1996;335:1417–1423. T, Furukawa Y, Nakamura Y, Niikawa N, Boileau C, Matsumoto N.
63. Avierinos JF, Inamo J, Grigioni F, Gersh B, Shub C, Enriquez-Sarano M. Heterozygous TGFBR2 mutations in Marfan syndrome. Nat Genet.
Sex differences in morphology and outcomes of mitral valve prolapse. 2004;36:855–860.
Ann Intern Med. 2008;149:787–795. 85. Ng CM, Cheng A, Myers LA, Martinez-Murillo F, Jie C, Bedja D,

64. Schoen FJ. Evolving concepts of cardiac valve dynamics: the continuum Gabrielson KL, Hausladen JM, Mecham RP, Judge DP, Dietz HC. TGF-
of development, functional structure, pathobiology, and tissue engineer- beta-dependent pathogenesis of mitral valve prolapse in a mouse model of
ing. Circulation. 2008;118:1864–1880. Marfan syndrome. J Clin Invest. 2004;114:1586–1592.
65. Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial 86. Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL, Cooper TK, Myers
cell phenotypes in regulating heart valve pathobiology. Am J Pathol. L, Klein EC, Liu G, Calvi C, Podowski M, Neptune ER, Halushka MK,
2007;171:1407–1418. Bedja D, Gabrielson K, Rifkin DB, Carta L, Ramirez F, Huso DL, Dietz
2170  Circulation  May 27, 2014

HC. Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse 96. Freed LA, Acierno JS Jr, Dai D, Leyne M, Marshall JE, Nesta F,
model of Marfan syndrome. Science. 2006;312:117–121. Levine RA, Slaugenhaupt SA. A locus for autosomal dominant
87. Groenink M, den Hartog AW, Franken R, Radonic T, de Waard V,
mitral valve prolapse on chromosome 11p15.4. Am J Hum Genet.
Timmermans J, Scholte AJ, van den Berg MP, Spijkerboer AM, 2003;72:1551–1559.
Marquering HA, Zwinderman AH, Mulder BJ. Losartan reduces aortic 97. Nesta F, Leyne M, Yosefy C, Simpson C, Dai D, Marshall JE, Hung J,
dilatation rate in adults with Marfan syndrome: a randomized controlled Slaugenhaupt SA, Levine RA. New locus for autosomal dominant mitral
trial. Eur Heart J. 2013;34:3491–3500. valve prolapse on chromosome 13: clinical insights from genetic studies.
88. Lacro RV, Dietz HC, Wruck LM, Bradley TJ, Colan SD, Devereux
Circulation. 2005;112:2022–2030.
RB, Klein GL, Li JS, Minich LL, Paridon SM, Pearson GD, Printz BF, 98. Kyndt F, Schott JJ, Trochu JN, Baranger F, Herbert O, Scott V, Fressinaud
Pyeritz RE, Radojewski E, Roman MJ, Saul JP, Stylianou MP, Mahony L; E, David A, Moisan JP, Bouhour JB, Le Marec H, Bénichou B. Mapping
Pediatric Heart Network Investigators. Rationale and design of a random- of X-linked myxomatous valvular dystrophy to chromosome Xq28. Am
ized clinical trial of beta-blocker therapy (atenolol) versus angiotensin II J Hum Genet. 1998;62:627–632.
receptor blocker therapy (losartan) in individuals with Marfan syndrome. 99. Nakamura F, Stossel TP, Hartwig JH. The filamins: organizers of cell
Am Heart J. 2007;154:624–631. structure and function. Cell Adh Migr. 2011;5:160–169.
89. Lacro RV, Guey LT, Dietz HC, Pearson GD, Yetman AT, Gelb BD, Loeys 100. Norris RA, Moreno-Rodriguez R, Wessels A, Merot J, Bruneval P, Chester
BL, Benson DW, Bradley TJ, De Backer J, Forbus GA, Klein GL, Lai AH, Yacoub MH, Hagège A, Slaugenhaupt SA, Aikawa E, Schott JJ,
WW, Levine JC, Lewin MB, Markham LW, Paridon SM, Pierpont ME, Lardeux A, Harris BS, Williams LK, Richards A, Levine RA, Markwald
Radojewski E, Selamet Tierney ES, Sharkey AM, Wechsler SB, Mahony L; RR. Expression of the familial cardiac valvular dystrophy gene, filamin-
Pediatric Heart Network Investigators. Characteristics of children and young A, during heart morphogenesis. Dev Dyn. 2010;239:2118–2127.
adults with Marfan syndrome and aortic root dilation in a randomized trial 101. Dalkilic I, Schienda J, Thompson TG, Kunkel LM. Loss of FilaminC
comparing atenolol and losartan therapy. Am Heart J. 2013;165:828–835.e3. (FLNc) results in severe defects in myogenesis and myotube structure.
90. Rajan S, Williams SS, Jagatheesan G, Ahmed RP, Fuller-Bicer G,
Mol Cell Biol. 2006;26:6522–6534.
Schwartz A, Aronow BJ, Wieczorek DF. Microarray analysis of gene 102. Zhou X, Tian F, Sandzén J, Cao R, Flaberg E, Szekely L, Cao Y, Ohlsson
expression during early stages of mild and severe cardiac hypertrophy. C, Bergo MO, Borén J, Akyürek LM. Filamin B deficiency in mice
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

Physiol Genomics. 2006;27:309–317. results in skeletal malformations and impaired microvascular develop-
91. Norris RA, Borg TK, Butcher JT, Baudino TA, Banerjee I, Markwald RR. ment. Proc Natl Acad Sci U S A. 2007;104:3919–3924.
Neonatal and adult cardiovascular pathophysiological remodeling and 103. Sasaki A, Masuda Y, Ohta Y, Ikeda K, Watanabe K. Filamin associates
repair: developmental role of periostin. Ann N Y Acad Sci. 2008;1123:30–40. with Smads and regulates transforming growth factor-beta signaling. J
92. Zuppiroli A, Roman MJ, O’Grady M, Devereux RB. A family study of Biol Chem. 2001;276:17871–17877.
anterior mitral leaflet thickness and mitral valve prolapse. Am J Cardiol. 104. Cushing MC, Liao JT, Anseth KS. Activation of valvular interstitial cells
1998;82:823–826, A10. is mediated by transforming growth factor-beta1 interactions with matrix
93. Henney AM, Tsipouras P, Schwartz RC, Child AH, Devereux RB, Leech molecules. Matrix Biol. 2005;24:428–437.
GJ. Genetic evidence that mutations in the COL1A1, COL1A2, COL3A1, 105. Charitakis K, Basson CT. Degenerating heart valves: fill them up with
or COL5A2 collagen genes are not responsible for mitral valve prolapse. filamin? Circulation. 2007;115:2–4.
Br Heart J. 1989;61:292–299. 106. Oyama MA, Chittur SV. Genomic expression patterns of mitral valve
94. Wordsworth P, Ogilvie D, Akhras F, Jackson G, Sykes B. Genetic segrega- tissues from dogs with degenerative mitral valve disease. Am J Vet Res.
tion analysis of familial mitral valve prolapse shows no linkage to fibrillar 2006;67:1307–1318.
collagen genes. Br Heart J. 1989;61:300–306. 107. Peal DS, Lynch SN, Milan DJ. Patterning and development of the atrio-
95. Disse S, Abergel E, Berrebi A, Houot AM, Le Heuzey JY, Diebold B, ventricular canal in zebrafish. J Cardiovasc Transl Res. 2011;4:720–726.
Guize L, Carpentier A, Corvol P, Jeunemaitre X. Mapping of a first locus
for autosomal dominant myxomatous mitral-valve prolapse to chromo- Key Words: mitral valve ◼ mitral valve insufficiency ◼ mitral valve
some 16p11.2-p12.1. Am J Hum Genet. 1999;65:1242–1251. prolapse
Epidemiology and Pathophysiology of Mitral Valve Prolapse: New Insights Into Disease
Progression, Genetics, and Molecular Basis
Francesca N. Delling and Ramachandran S. Vasan

Circulation. 2014;129:2158-2170
doi: 10.1161/CIRCULATIONAHA.113.006702
Downloaded from http://circ.ahajournals.org/ by guest on April 27, 2018

Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2014 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/129/21/2158

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located,
click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation is online at:


http://circ.ahajournals.org//subscriptions/

You might also like