ArticlePDF AvailableLiterature Review

SMAD6-deficiency in human genetic disorders

Authors:

Abstract and Figures

SMAD6 encodes an intracellular inhibitor of the bone morphogenetic protein (BMP) signalling pathway. Until now, SMAD6-deficiency has been associated with three distinctive human congenital conditions, i.e., congenital heart diseases, including left ventricular obstruction and conotruncal defects, craniosynostosis and radioulnar synostosis. Intriguingly, a similar spectrum of heterozygous loss-of-function variants has been reported to cause these clinically distinct disorders without a genotype–phenotype correlation. Even identical nucleotide changes have been described in patients with either a cardiovascular phenotype, craniosynostosis or radioulnar synostosis. These findings suggest that the primary pathogenic variant alone cannot explain the resultant patient phenotype. In this review, we summarise clinical and (patho)genetic (dis)similarities between these three SMAD6-related conditions, compare published Madh6 mouse models, in which the importance and impact of the genetic background with respect to the observed phenotype is highlighted, and elaborate on the cellular key mechanisms orchestrated by SMAD6 in the development of these three discrete inherited disorders. In addition, we discuss future research needed to elucidate the pathogenetic mechanisms underlying these diseases in order to improve their molecular diagnosis, advance therapeutic strategies and facilitate counselling of patients and their families.
Content may be subject to copyright.
REVIEW ARTICLE OPEN
SMAD6-deciency in human genetic disorders
Ilse Luyckx
1,2
, Aline Verstraeten
1
, Marie-José Goumans
3
and Bart Loeys
1,2
SMAD6 encodes an intracellular inhibitor of the bone morphogenetic protein (BMP) signalling pathway. Until now, SMAD6-
deciency has been associated with three distinctive human congenital conditions, i.e., congenital heart diseases, including left
ventricular obstruction and conotruncal defects, craniosynostosis and radioulnar synostosis. Intriguingly, a similar spectrum of
heterozygous loss-of-function variants has been reported to cause these clinically distinct disorders without a genotypephenotype
correlation. Even identical nucleotide changes have been described in patients with either a cardiovascular phenotype,
craniosynostosis or radioulnar synostosis. These ndings suggest that the primary pathogenic variant alone cannot explain the
resultant patient phenotype. In this review, we summarise clinical and (patho)genetic (dis)similarities between these three SMAD6-
related conditions, compare published Madh6 mouse models, in which the importance and impact of the genetic background with
respect to the observed phenotype is highlighted, and elaborate on the cellular key mechanisms orchestrated by SMAD6 in the
development of these three discrete inherited disorders. In addition, we discuss future research needed to elucidate the
pathogenetic mechanisms underlying these diseases in order to improve their molecular diagnosis, advance therapeutic strategies
and facilitate counselling of patients and their families.
npj Genomic Medicine (2022) 7:68 ; https://doi.org/10.1038/s41525-022-00338-5
The protein SMAD6, encoded by SMAD6 (OMIM: 602931), belongs
to the SMAD family of proteins involved in the bone morphoge-
netic proteins (BMP) signalling cascade. Even though these
molecules were initially discovered for their ability to induce
bone formation, it is now clear that BMPs are important in the
embryogenesis and development of many organ systems, as well
as in maintenance of adult tissue homoeostasis. SMAD6 is an
intracellular inhibitor of, predominantly, the BMP signalling
pathway, yet it cross-talks with the closely related transforming
growth factor-β(TGF-β) signalling pathway
1,2
.
Over the past 10 years, genetic variants in SMAD6 were
demonstrated to impinge on the risk of human genetic
disorders
313
such as cardiovascular diseases, including congenital
heart defects (CHD), craniosynostosis (CRS) and radioulnar
synostosis (RUS). CHD is among the most common birth defects,
affecting 613:1000 live-born infants
1416
. In association with
SMAD6-deciency, it encompasses a range of cardiac and outow
tract abnormalities. Complex lesions consisting of multiple defects
are often severe, even critical, for which treatment with advanced
surgery for denitive correction of malformations or (palliative)
medication is imperative
17
. In addition, adult patients with a sole
congenital aortic valve defect associate with more late-onset
vascular complications like a pathological widening of the thoracic
aorta (~thoracic aortic aneurysm (TAA))
18
. TAAs are also life-
threatening as they are (1) clinically silent
19
, (2) entail a high risk
for acute dissection and/or rupture (mortality rates 70%)
20
, and
(3) no therapy currently exists that can stop TAA development or
progression
21
. CRS, which is a skull defect aficting 1:21002500
live births
22,23
, is a second SMAD6-related disease. Surgical
correction is frequently necessary to prevent complications
24
such as developmental delay, facial abnormality, sensory, respira-
tory and neurological dysfunction, anomalies affecting the eye,
and psychological disturbances
25
. Finally, congenital RUS, also
referred to as fused forearm bones, is a rare condition with ~500
cases reported in literature
9,13,26
. This malformation, usually
diagnosed before the age of 5 years, is not life-threatening, but
corrective surgery and/or medication to control pain might, in
some cases, improve the quality of life
13
.
The therapeutic strategies for SMAD6 mutation-positive patients
mainly focus on disease monitoring in order to dene the
appropriate time for intervention, medication to control pain, and
surgical repair
19,24,2730
. Even though surgery is effective, it is
associated with risks, requires early detection of at-risk patients,
only provides relief late in the disease course, and does not target
the underlying driver(s) of the disease. Hence, there is a need for a
better (molecular) understanding for early diagnosis, and to
empower new therapies to prevent disease progression. With this
review, we provide a comprehensive overview on SMAD6-
deciency in human genetic disorders by summarising the clinical,
(patho)genetic and cellular (dis)similarities observed in human
and mouse models. We conclude with future directions of
research needed to improve patient management based on the
underlying SMAD6-related molecular disease signature.
CLINICAL PHENOTYPE OF PATIENTS WITH SMAD6-DEFICIENCY
The clinical presentation of heterozygous SMAD6 variant-positive
patients is extremely heterogeneous as illustrated by the different
affected organ systems, the varying degree of severity, and the
range of associated complications. Table 1summarises the clinical
phenotype of probands with disease-causative SMAD6 variants. All
disease-related clinical denitions are summarised in Table 2.
Cardiovascular diseases
The cardiovascular phenotype (cases, N=31) (probands, N=28,
Table 1) include left ventricular outow tract defects (N=21/28,
75%)
4,5,7,8,10
, conotruncal defects (N=4/28, 14%)
5
, and defects
dened as others(N=3/28, 11%)
5
as they cannot be categorised
1
Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
2
Department of Human
Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
3
Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands.
email: bart.loeys@uantwerpen.be
www.nature.com/npjgenmed
Published in partnership with CEGMR, King Abdulaziz University
1234567890():,;
Table 1. Clinical phenotype of probands with disease-causative SMAD6 variants.
Study Patient cohort Major defect (sub-category) Number of
probands
NS/S Age Additional features within the same organ system
Tan et al.
10
Cardiovascular
malformations
Bicuspid aortic valve (LVO) 1 NS 1.5 y
a
Aortic stenosis
Cardiovascular
malformations
Bicuspid aortic valve with
coarctation of the aorta (LVO)
1NS30y
a
Aortic stenosis
Timberlake et al.
11
;
Timberlake et al.
12
Craniosynostosis Craniosynostosis 17 NS Paediatric
b
Metopic synostosis (N=12), sagittal synostosis (N=3),
metopic +sagittal synostosis (N=2)
Jin et al.
5
Congenital heart defect Tetralogy of Fallot (CTD) 3 Unknown
c
Paediatric
b
Pulmonary stenosis (subvalvular (N=2), valvar (N=1)), ventricular
septal defect (malalignment) (N=1), coronary artery anomaly (right)
(N=1), left aortic arch with normal branching pattern (N=1), patent
foramen ovale (N=1)
Congenital heart defect Transposition of the Great
Arteries (CTD)
1 Unknown
c
Paediatric
b
Atypical coronary arteries in D-loop Transposition of the Great
Arteries, transposition D-loop of the Great Arteries with intact
ventricular septum, left aortic arch with a normal branching pattern
Congenital heart defect Hypoplastic left heart
syndrome (LVO)
1 Unknown
c
Paediatric
b
Aortic arch hypoplasia, aortic atresia, hypoplasia ascending aorta,
hypoplastic left ventricle (subnormal cavity volume), mitral atresia,
restrictive patent foramen ovale
Congenital heart defect Coarctation of the aorta (LVO) 1 Unknown
c
Paediatric
b
Atrial septal defect (secundum), left-sided patent ductus arteriosus,
tubular hypoplasia of aorta, ventricular septal defect (malalignment,
muscular outlet)
Congenital heart defect Bicuspid aortic valve with
coarctation of the aorta (LVO)
1 Unknown
c
Paediatric
b
Aortic arch hypoplasia, hypoplastic left ventricle (subnormal cavity
volume), patent foramen ovale
Congenital heart defect Other 1 Unknown
c
Paediatric
b
Left aortic arch with normal branching pattern, partially anomalous
pulmonary veins, sinus venosus septal defect (superior type)
Congenital heart defect Other 1 Unknown
c
Paediatric
b
Vascular ring, aberrant left subclavian artery, abnormal branching
right aortic arch, right aortic arch left ligament
Gillis et al.
4
; Luyckx
et al.
7
; Park et al.
8
Cardiovascular
malformations
Bicuspid aortic valve-related
thoracic aortic aneurysm
15 NS Average 64.1 y Aortic valve calcication (N=1), coarctation of the aorta (N=2),
aortic regurgitation (N=1), aortic stenosis (N=1)
Kloth et al.
6d
Cardiovascular
malformations
Coarctation of the aorta (LVO) 1 S
e
6 y Suspected tricuspid aor tic valve
Other 1 NS 10 y Dysplastic and stenotic pulmonary valve, dilated cardiomyopathy,
stenotic left main coronary artery
Yang et al.
13
; Shen et al.
9
Radioulnar synostosis Radioulnar synostosis 77 NS Average 5 y Lateral left radioulnar synostosis (N=17), lateral right radioulnar
synostosis (N=6), bilateral radioulnar synostosis (N=54)
Calpena et al.
3
Craniosynostosis Craniosynostosis 26 S
f
/NS
g
Not reported Metopic synostosis (N=15), sagittal synostosis (N=6), right coronal
synostosis (N=2), sagittal +left coronal synostosis (N=1),
sagittal +bicoronal synostosis (N=2)
NS non-syndromic, Ssyndromic, LVO left ventricular obstruction, CTD conotruncal defect, yyears, NA not applicable.
This table excludes patients with known disease-related genetic hits at other loci in addition to SMAD6.
a
Unclear if the aorta has been evaluated.
b
Age is not specied.
c
Two patients had extra-cardiac abnormalities (i.e., syndromic cases).
d
Only paper describing bi-allelic variants.
e
Consanguineous family with facial dysmorphism, unilateral hypoplasia, bilateral radioulnar synostosis, bilateral toe 2/3 syndactyly, very dry and scaly skin, dysrhythmic electro-encephalogram without seizure
activity and mild intellectual disability.
f
Seven syndromic probands.
g
Nineteen non-syndromic probands.
I. Luyckx et al.
2
npj Genomic Medicine (2022) 68 Published in partnership with CEGMR, King Abdulaziz University
1234567890():,;
into the traditional subgroups that arise from disruption of shared
embryonic processes. Left ventricular outow tract defect refers to
hypoplastic left heart syndrome (HLHS) (N=1/21, 5%), coarctation
of the aorta (CoA) (N=2/21, 10%), and bicuspid aortic valve (BAV),
which is associated with congenital CoA and/or late-onset TAA
(N =17/21, 81%) in all patients, except for one toddler with
isolated BAV (N=1/21, 5%; 1.5 years old). Conotruncal defects
include Tetralogy of Fallot (N=3/4, 75%) and D-loop transposition
of the Great Arteries (N=1/4, 25%). The remaining three patients
presented with either a vascular ring, partially anomalous
pulmonary veins combined with sinus venosus septal defect, or
stenotic pulmonary valve and stenotic left main coronary artery
accompanied with dilated cardiomyopathy.
Craniosynostosis
The clinical outcome of CRS (cases, N=49) (probands, N=43,
Table 1)
3,11,12
involves syndromic (N=7/43, 16%) and non-
Table 2. Clinical description of the disease-related anomalies.
Anomalies Clinical description
Absent corpus callosum A congenital brain defect with partial or complete absence of the region that connects the two
cerebral hemispheres.
Atrial septal defect A congenital heart defect resulting from incomplete atrial septation.
Atrioventricular septal defect A congenital heart defect resulting from incomplete septation of the atrioventricular canal.
Bicuspid aortic valve A congenital heart defect in which the aortic valve has only two leaets instead of the normal three.
Caudal vertebrae dysplasia A congenital defect of a total or partial failure of the development of the caudal vertebrae.
Coarctation of the aorta A congenital heart defect in which blood ow is blocked by aortic narrowing usually at the region of
the ductus arteriosus.
Coronal synostosis A congenital skull defect in which the coronal suture close prematurely leading to attening of the
head (unicoronal), or a short head with wide appearance (bicoronal).
Dilated cardiomyopathy A condition in which the heart becomes enlarged and cannot pump blood effectively.
D-loop Refers to the normal rightward (dextro =D) loop or bend of the embryonic heart tube and indicates
that the inow portion of the right ventricle is to the right of the morphological left ventricle.
Frontal bossing A condition indicating a protuberance of the frontal bones of the forehead.
Hypoplastic left heart syndrome A congenital heart defect in which the hearts left side (including the aorta, aortic valve, left ventricle
and mitral valve) is underdeveloped.
Macrocephaly A condition in which circumference of the head is more than two standard deviations above the
mean value for a given age and gender.
Metopic synostosis A congenital skull defect in which the metopic suture close prematurely leading to a forehead with
triangular appearance (trigonocephaly).
Microcephaly A condition in which circumference of the head is more than two standard deviations below the
mean value for a given age and gender.
Mitral/pulmonary/tricuspid/aortic valve
regurgitation
A condition in which the valve does not close properly, allowing blood to ow backwards.
Regurgitation is also called insufciency or incompetence.
Patent ductus arteriosus A congenital heart defect in which the ductus arteriosus fails to close after birth.
Patent foramen ovale A congenital heart defect in which the foramen ovale did not close properly at birth, with the result of
an existing hole between the left and right atria of the heart.
Plagiocephaly A condition in which the skull attens on one side.
Polydactyly A congenital skeletal condition in which an individual has more than 5 ngers per hand or 5 toes
per foot.
Premature fusion of the anterior fontanel A congenital skull defect in which the anterior fontanel close prematurely.
Radioulnar synostosis A congenital defect in which the radius and ulna of the forearm is abnormally connected (synostosis).
Sagittal synostosis A congenital skull defect in which the sagittal suture close prematurely leading to a long and narrow
head (scaphocephaly).
Sinus venosus septal defect A congenital heart defect in which a deciency of the common wall between the superior vena cava
and the right upper pulmonary vein is present thereby allowing shunting of blood from the systemic
to the pulmonary circulation.
Stenotic left main coronary artery A condition in which the left main coronary artery is narrowed.
Stenotic pulmonary valve A condition in which the pulmonary valve is narrowed.
Tetralogy of Fallot A congenital heart defect characterised by right ventricular outow tract obstruction, right ventricular
hypertrophy, ventricular septal defect and overriding aorta.
Thoracic aortic aneurysm A condition in which the aortic diameter is more than two standard deviations above the mean value
for a given age and gender.
Transposition of the Great Arteries A congenital heart defect referring to ventriculoarterial discordance, i.e., aorta arises from a
morphological right ventricle, and the pulmonary artery arises from a morphological left ventricle.
Vascular ring A congenital heart defect in which the aorta or its branches forms a ring around the trachea and the
oesophagus.
Ventricular septal defect A congenital heart defect resulting from incomplete ventricular septation.
Ventriculomegaly A condition in which the brain ventricles are abnormally enlarged.
I. Luyckx et al.
3
Published in partnership with CEGMR, King Abdulaziz University npj Genomic Medicine (2022) 68
syndromic presentations (N=36/43, 84%) in which single and
multiple fusion events of almost all sutures have been identied.
Most common presentation was metopic synostosis (N=27/43,
63%), followed by sagittal synostosis (N=9/43, 21%), right
unicoronal synostosis (N=2/43, 5%), combined metopic and
sagittal synostosis (N=2/43, 5%), combined sagittal and bicoronal
synostosis (N =2/46, 4%), and combined sagittal and left
unicoronal synostosis (N=1/43, 2%). Remarkably, raised intracra-
nial pressure following cranial reconstruction, which is usually a
rather infrequent complication in simple synostosis of midline
sutures, should be specically monitored in SMAD6 variant-
positive patients
3
. Other recurrent brain or skull anomalies in
(non-)syndromic subjects comprise ventriculomegaly and absent
corpus callosum, macrocephaly, and mild microcephaly, and mild-
to-moderate neurodevelopmental delay (consisting of speech,
educational and global delay)
3,11,12
. More subtle learning difcul-
ties were observed in 36% of the non-syndromic patients
(N=14)
11
, while gross and ne motor delays were only observed
occasionally
3,11
. In syndromic cases
3
, cardiac defects were
common (N=5/7, 71%), but seem to have another pattern which
is further discussed in the following section on the clinical overlap.
Congenital radioulnar synostosis
Patients with RUS
9,13
(cases, N=93) (probands, N=77, Table 1)
are most frequently characterised by bilateral RUS (69%; isolated
(N=42/61), familial (N=22/32)), followed by unilateral left-sided
RUS in sporadic patients (N=15/42, 36%) versus right-sided RUS
(N=4/42, 10%), while no susceptibility for left- or right-sided RUS
was observed in families (N=5/32, 16%). Affected individuals
within a single pedigree can show both bilateral and unilateral
RUS. No syndromic cases have been reported thus far, yet some
subordinate clinical ndings have been described in 14
families
9,13
: three families presented with axial skeletal deformities
(including cervical fusion, rib malformation, caudal vertebrae
dysplasia and vertebral malformations), two families had poly-
dactyly (pre- and pro-axial type), ve families exhibited CHD
(encompassing patent ductus arteriosus, mitral/tricuspid/aortic
regurgitation, atrial septal defect, BAV, left ventricular hypertrophy
and mitral/pulmonary valve insufciency), and four families
showed skull-related defects (containing frontal bossing, plagio-
cephaly and premature closure of anterior fontanel, but no
unequivocal description of CRS). Remarkably, in six out of the 14
families a variant-positive SMAD6 carrier without RUS but with
skeleton-, skull-, or CHD-related features
9,13
was reported: two
affected individuals from two families displayed cervical fusion or
caudal vertebrae dysplasia, two patients from two families
exhibited with premature closure of anterior fontanel, frontal
bossing and plagiocephaly or solely plagiocephaly, one subject
presented with polydactyly and, nally, one affected had patent
ductus arteriosus together with mitral regurgitation.
Clinical overlap?
Studies have, in addition to the clinical indication for study
enrolment, to some extent also assessed the presence of other
SMAD6-related clinical associations. Patients with cardiovascular
disease did not exhibit CRS and/or RUS. A child, from a
consanguineous family harbouring a pathogenic homozygous
SMAD6 variant, was reported to present with CoA, suspected
tricuspid aortic valve, bilateral RUS, renal anomalies, facial
dysmorphism and global development delay
6
. In view of
consanguinity, homozygosity at other loci as an explanation for
these multisystemic features cannot be excluded. CRS cases did
not exhibit RUS, but ve syndromic CRS cases presented with a
CHD
3
of which none seem to mimic the more severe conotruncal
and outow tract defects seen in the cardiovascular disease
cohorts. For example, defects in three patients resolved sponta-
neously. One patient with atrioventricular septal defect required
surgery at the age of three years, yet his variant-positive mother
Fig. 1 Graphical representation of identied heterozygous SMAD6 variants in probands with bicuspid aortic valve-related aortopathy,
congenital heart disease, (non-)syndromic craniosynostosis and non-syndromic radioulnar synostosis. SMAD6 protein has several
domains: MH1-domain (grey; inhibitory effect on signalling), PY motif (orange), PLDLDS motif (yellow), MH2-domain (grey; inhibitory effect on
signalling) and L3-loop (blue; determines specicity for interaction with type I receptors)
88,89
. Of note: (1) all variants were called pathogenic
in the original publications but there is different criteria for calling pathogenicity(e.g., by applying different allele frequency thresholds, and/
or using specic functional tests); (2) identical protein changes (underlined) are described in patients with distinctive phenotypes.
I. Luyckx et al.
4
npj Genomic Medicine (2022) 68 Published in partnership with CEGMR, King Abdulaziz University
had a normal echocardiogram. The fth patient had BAV (N=1/
46, 2%) with right bundle branch block. This observation exactly
matches the epidemiological number of 2% for BAV in the general
population though
31
. None of the seven extra screened asympto-
matic parents of SMAD6 variant-positive children with CRS showed
any evidence for BAV or TAA
3
. Finally, no BAV or TAA has been
identied so far in the non-syndromic CRS cohort (personal
communication with A. Wilkie, Oxford). Hence, no clinical overlap
of a variant-positive SMAD6 carrier with cardiovascular disease or
CRS with any abnormality affecting the other organ systems has
been observed to date.
Finally, the phenotypic picture in 14 families with RUS is more
complicated as both CHD as well as skull and skeletal
abnormalities have been observed occasionally (N=12/93,
13%)
9,13
. Although based on their nature and incidence, we
cannot rule out an alternative cause for some abnormalities (e.g.,
valve insufciency, left ventricular hypertrophy and rib/vertebral
malformation), the occurrence of skeletal- (N=4/93, 4%), skull-
(N=9/93, 10%), or CHD-related (N=3/93, 3%) abnormalities in
families with RUS does hint to some clinical overlap. For example,
two variant-positive SMAD6 carriers from two families without RUS
presented with axial skeletal deformities, either cervical fusion or
caudal vertebrae dysplasia. Extra skull features were observed in
another ve families, including frontal bossing (N=4/93, 4%),
plagiocephaly (N=3/93, 3%), and premature fusion of the anterior
fontanel (N=2/93, 2%). Plagiocephaly and premature fusion of
the anterior fontanel was reported in a variant-positive family
member without RUS. And nally, three families had CHD too,
namely patent ductus arteriosus (N=1/93, 1%), atrial septal defect
(N=1/93, 1%), and BAV (N=1/93, 1%).
GENETIC (DIS)SIMILARITIES BETWEEN SMAD6-RELATED
DISORDERS
Intriguingly, similar, or even identical, heterozygous loss-of-
function variants in SMAD6 cause these three distinct disorders
(Fig. 1and Supplementary Table 1)
313
. The variant spectrum
includes rare truncating and missense variants locating in the
functional MH1- and MH2-domain of the protein with no
phenotypic correlation with respect to variant type nor location.
Identical nucleotide changes (N=6) have been described in
patients with cardiovascular disease (N=3), CRS (N=5) or RUS
(N=10). Moreover, the phenotype within these families are,
predominantly, restricted to one affected organ system. For
example, the p.(Gly156Valfs*23) variant causes BAV-related aorto-
pathy (N=1), sagittal synostosis (N=1), and non-syndromic RUS
(N=4), for which no clinical overlap has been documented except
for frontal bossing in one family with left-sided RUS. Hence, the
molecular nding cannot predict the clinical presentation of a
patient, and, as such, it is likely that (a) factor(s) inherited together
with the primary SMAD6 mutation drives the resultant patient
phenotype. The latter seems likely as within one family
concordance of the phenotype is frequently observed.
Cardiovascular disease
The aetiology of CHD is multifactorial, involving genetic and
environmental factors such as smoking, alcohol abuse and
infection transmitted by the mother during pregnancy
32
. Familial
studies have demonstrated that the CHD recurrence risk in family
members of affected individuals depends on the type of lesion
33
.
Pathogenic variants cause autosomal dominant, autosomal
recessive, or X-linked traits with variable penetrance and clinical
expressivity. About 132 denitive and strong candidate genes for
CHD in numerous functional classes like chromatin modication,
transcription factors and signal transduction, amongst others,
have been reported. The predominant disease-causative effect is
through loss-of-function
34
. To date, 50% of the patients remain
molecularly undiagnosed though, and the yield is even lower in
non-syndromic cases
34
. Interestingly, pathogenic SMAD6 variants
have been shown to be enriched in isolated paediatric and adult
CHD patients, in which most patients exhibited left ventricular
outow tract defects (Table 3). So far, patients with recessive
variants do not seem to present with a more severe cardiovascular
phenotype as compared to subjects harbouring heterozygous
variants. However, this observation is based on only two cases,
and no functional analyses have been performed
6
.ASMAD6
genetic uptake of 4.6% was reached in more severely affected
BAV-related aortopathy patients, i.e., BAV patients who underwent
surgical repair for aneurysmal disease before the age of 50, and
with a positive family history for cardiovascular disease. The
estimated penetrance for the disease was 82.4%. SMAD6 is the
most important BAV/TAA gene identied thus far, as none of the
approximately 30 denitive and candidate genes for BAV and/or
TAA explain more than 1% of these patients
7
. The emerging BAV/
TAA disease-related pathways include impaired cardiac transcrip-
tion factor activity (e.g., GATA5)
35,36
, perturbed extracellular matrix
homoeostasis (e.g., LOX)
37
, aberrant TGF-β(e.g., TGFBR1)
19
and
NOTCH (e.g., NOTCH1)
38
signalling, deciency of the vascular
smooth muscle cell contractile apparatus (e.g., ACTA2)
39
, and
altered endothelial cell function (e.g., ROBO4)
40
. Taken together,
carrying a pathogenic SMAD6 might be insufcient to denitively
cause cardiovascular disease in all cases, and, as such, more
research is required to identify the missing information, and to
understand how it contributes to disease.
A clinical and genetic association between BAV, HLHS, and CoA
have already been thoroughly discussed in familial studies
41,42
,
and some examples of monozygotic twins with discordant
phenotypes, i.e., one has BAV while the other present with HLHS,
have been described
43,44
. As SMAD6-deciency results in a
spectrum of, mainly, left ventricular outow tract defects, one
could hypothesise the existence of additional genetic hits in
families. Particular emphasis might be given to ascertain essential
cardiac transcription complexes, and to investigate the
Table 3. SMAD6 variant-positive patients with congenital heart disease.
Tan et al.
10
Jin et al.
5
Gillis et al.
4
Luyckx et al.
7
Total
Left ventricular outow tract 2/83 (2.4%) 3/797 (0.4%) 11/441 (2.5%) 3/65 (4.6%) 19/1386 (1.4%)
D-loop transposition of the Great Arteries 0/65 (0%) 1/251 (0.4%) –– 1/316 (0.3%)
Conotruncal defects 0/78 (0%) 3/872 (0.3%) –– 3/950 (0.3%)
Heterotaxy 0/10 (0%) 0/272 (0%) –– 0/282 (0%)
Others 0/200 (0%) 2/679 (0.3%) –– 2/879 (0.2%)
Total 2/436 (0.5%) 9/2871 (0.3%) 11/441 (2.5%) 3/65 (4.6%)
This table excludes (1) patients with known disease-related genetic hits at other loci, in addition to SMAD6, and (2) case reports lacking information on the total
number of screened patients.
I. Luyckx et al.
5
Published in partnership with CEGMR, King Abdulaziz University npj Genomic Medicine (2022) 68
accessibility of these factors onto DNA in patient-derived material
in order to reveal novel crucial clues on the pathogenesis of CHD
disease.
Craniosynostosis
CRS is a heterogeneous disease inuenced by mechanical and
extrinsic forces as well as genetic components affecting the
intrinsic properties of the suture
45
. In families, an autosomal
dominant mode of transmission is mostly observed, but in about
half of the cases a de novo variant is found. The genetic uptake is
highest in syndromic cases, while isolated cases (i.e., 75% of all
patients) largely remain molecularly undiagnosed
45,46
. Approxi-
mately one quarter of CRS cases harbour a disease-causative
variant in one of the known genes, mostly in FGFR2,FGFR3,TCF12,
ERF, EFNB1,orTWIST1, causing either a loss- or gain-of-function.
These gene products are involved in signal transduction pathways
like FGF signalling (FGR2, FGFR3), Eph/ephrin signalling (EFNB1)
and ERK-MAPK activity (ERF) or they bind DNA to regulate gene
expression (TWIST1, TCF12). As SMAD6 variants account for 5.8% of
all (non-)syndromic patients with metopic synostosis, it became,
by far, the largest monogenic contributor to metopic synostosis
yet identied. Furthermore, SMAD6 variants seem less commonly
associated with other types of suture fusion
3
, making it in
particular relevant to screen patients with metopic synostosis for
SMAD6 deciency.
SMAD6-related CRS has been associated with reduced pene-
trance (overall penetrance, 1624%)
3,11,12
. As such, a two-locus
inheritance model for CRS (i.e., metopic, sagittal and combined
metopic and sagittal) was proposed by Timberlake et al., in which
near complete-penetrance was reached upon co-occurrence with
a common BMP2 SNP risk allele (C) (rs1884302)
11
. Upon merger of
datasets, this association still holds true, yet the initial signal has
weakened due to non-replication in an independent cohort. One
explanation might be the underrepresentation of the risk allele
(frequency ~0.33, gnomAD: European non-Finnish) in non-
penetrant SMAD6 variant harbouring individuals in the discovery
studies, which was not observed in a third study
3,11,12
. Addition-
ally, rs1884302 was found to strongly associate with sagittal
synostosis
47
, and more recent data for metopic synostosis reveal
no equivalent association for this SNP
48
. Extra work is necessary to
explore on such relationship between SMAD6 variant-positive
patients with sagittal synostosis, and a larger sample size is
needed to dissect whether this interaction is truly digenic
inheritance or is merely an additive effect of the GWAS signal,
modifying the penetrance of SMAD6 pathogenic variants. Addi-
tional light was shed onto this digenic inheritance model by
revealing the presence of this common SNP in SMAD6 mutation-
positive patients with either BAV-related aortopathy (N=4)
7
or
radioulnar synostosis (N=7)
13
but in the absence of any sign of
CRS. Altogether, current data suggest that the pathogenic SMAD6
variant alone might be insufcient to denitively cause CRS in all
cases, and it still remains to be further investigated what the extra
hits, and what the underlying mechanisms are.
Table 4. Overview of the published Madh6 knock-out mouse models with the phenotypic characterisation of Madh6
/
animals.
Galvin et al.
54
Estrada et al.
55
Wylie et al.
56
Generation model Embryonic stem cells with transgene
interrupting SMAD6 function (i.e., insertion of
LacZ/neomycin resistance cassette into the
terminus of the exon encoding the MH2-
domain)
Stem cells from Galvin et al. Stem cells from Galvin et al.
Biological consequence Madh6-LacZ fused transcript See Galvin et al.
54
See Galvin et al.
54
Parents of breeding Heterozygous Heterozygous Heterozygous
Lethality Partial lethality of madh6
/
mice (P21; 313%
~genetic background
a
)
Lethality of madh6
/
mice
(P0; 5%, but all died <24 h)
Lethality of madh6
/
mice
(P0; 8%, but all died within
26 days of birth)
Cardiac phenotype Hyperplasia cardiac valves, enlarged mitral
valve, enlarged pulmonary valve, abnormal
truncus arteriosus septation
b
, aortic
ossication
c
, hypertension
Not investigated No hyperplastic valves, or
other major defects explaining
cause of death
Vascular phenotype Decreased vasodilation
c
, abnormal
thrombosis
d
Not investigated Blood vessel haemorrhages in
skin and brown fat pads
Craniofacial phenotype Not observed Domed skull, shortened snout Nothing obvious that could
explain the cause of death
Axial skeletal phenotype Not observed Posterior transformation of cervical
vertebrae, bilateral ossication centres in
lumbar vertebrae, bid sternebrae
Nothing obvious that could
explain the cause of death
Appendicular skeletal
phenotype
Not observed Smaller size, abnormal growth plate
development
e
Nothing obvious that could
explain the cause of death
Genetic background 129/SvEv × BALB/cBy, 129/SvEv × C57Bl/6,
inbred 129/SvEv
C57BL/6J x BALB/c CD1
Madh6 is the murine orthologue of human SMAD6.
a
Background sensitivity: inbred 129S/SvEv (3% versus expected 25%), mixed 129S6/SvEvTac × BALB/cByJ (9% versus expected 25%), mixed 129S6/
SvEvTac × C57BL6/J (13% versus expected 25%).
b
A subset of homozygotes exhibit abnormal septation of the outow tract leading to a severely narrowed ascending aorta, and an enlarged pulmonary trunk
or the reverse.
c
Only observed in the surviving animals starting at 6 weeks of age.
d
Surviving homozygotes display occasional thrombotic lesions as well as focal ischaemia in the lung, liver and kidney.
e
Abnormal growth plate development: delayed onset of hypertrophic differentiation and mineralisation at midgestation, but expanded hypertrophic zone at
late gestation.
I. Luyckx et al.
6
npj Genomic Medicine (2022) 68 Published in partnership with CEGMR, King Abdulaziz University
Radioulnar synostosis
Since the 70s, congenital RUS is recognised as an inheritable
disease segregating in an autosomal dominant manner
49,50
.In
total, 10% of the RUS patients were identied with a monogenetic
cause (e.g., NOG) or with aneuploidy syndromes, in which the
syndromic subjects presented with additional abnormalities in the
skeleton, heart, urinary tract, blood and males had extra X and Y
chromosomes
51
. At present, SMAD6 deciency is, by far, the most
important known disease gene for non-syndromic RUS, as it
explains 42% of familial cases and 16% of sporadic patients
9,13
.
The penetrance of disease is incomplete, and has been reported
around 2025%
9,13
. Other genetic causes include two pathogenic
variants in NOG, explaining less than 1% of the patients
13
.NOG
encodes noggin, which is a major BMP antagonist. Dysregulation
of BMP signalling due to NOG deciency in mice showed
interference with hedgehog signalling for BMP-induced inter-
digital cell death
52
, and for axial skeleton development
53
. The
contribution of genetic variability in SMAD6 and NOG to syndromic
RUS is yet unexplored. In sum, literature indicates that radioulnar
synostosis is not exclusively caused by one pathogenic SMAD6
variant in all cases. Again, more investigation is needed to ll our
gap in knowledge about the extra hits and underlying
mechanisms.
Current challenges in SMAD6-related diagnosis and
counselling
Patient management for SMAD6-related disorders is challenging
as rare pathogenic loss-of-function variants associate with (1)
reduced penetrance, (2) extreme variability in phenotypical
expression, and (3) distinctive clinical entities without
genotypephenotype correlation, as outlined above. Hence, every
single case should be discussed thoroughly in a multidisciplinary
team based on phenotype, family history, inheritance pattern, and
pathogenicity of the variant. Given the possibility of a devastating
cardiovascular outcome, echocardiographic evaluation is currently
indicated in a SMAD6 variant-positive proband, irrespective of the
clinical indication for referral. A genetic test is best offered to
family members of SMAD6 variant-positive patients with cardio-
vascular disease or RUS as some clinical overlap with the
cardiovascular disease might exist. In contrast, variant-positive
SMAD6 carriers in CRS cohorts are frequently unaffected making a
genetic test uninformative. There is currently some preliminary
evidence that phenotypes are quite consistent in a single family.
Nevertheless, more insight is needed before we can abandon
echocardiographic evaluation in relatives of SMAD6 variant-
positive probands with CRS. Another counselling challenge is
caused by the observation that the general population well-
tolerates loss-of-function SMAD6 variants (pLI =0, gnomAD
v2.1.1), despite the overwhelming overrepresentation of such
variants in disease cohorts as compared to this control popula-
tion
35,1113
. This is in particular challenging for CRS given the low
penetrance of CRS in individuals heterozygous for pathogenic
SMAD6 variants
3,11
.
Lastly, diagnostic and research laboratories also encounter
difculties for variant interpretation, in particular for missense
variants. In this regard, Calpena et al.
3
have provided a ltering
strategy able to discriminate high-penetrant rare pathogenic
missense variants, as proven in functional tests assessing protein
stability and/or impaired BMP signalling activity. Even though very
useful, this approach will not classify all type of variants (e.g., 5
untranslated region), and current bio-informatic tools are not
sufcient sensitive to assess variants with moderate effects, which
are likely to explain, to some extent, the variability in expressivity
and unpredictable penetrance. Nonetheless, implementation of
exible, preferably high-throughput, functional assays for variant
interpretation, combined with further renement of bio-informatic
tools, is necessary to address this challenge.
LESSONS FROM MOUSE MODELS
Genetically modied mouse models have, with success, been used
to interrogate the pathomechanisms underlying rare human
disorders. At present, three mouse models lacking the murine
orthologue of SMAD6, i.e., Madh6, have been studied (Table 4).
The Madh6-mutant mice were produced by a LacZ/neomycin
resistance cassette inserted into the terminus of the exon
encoding the MH2-domain of Smad6
54
. Each model is unique by
its respective genetic background as all models were generated
using embryonic stem cells created by Galvin et al.
In the model on a mixed 129/SvEv × BALB/cBy background
54
,
homozygous animals exhibited hyperplasia of the cardiac valves,
with the mitral and pulmonary valve being more extremely
affected, septation defects, and lethality. The latter was observed
due to an underrepresentation of homozygotes at the time of
weaning. Surviving animals developed aortic ossication with
notable cartilaginous metaplasia and trabeculation of the aortic
media (from 6 weeks of age), decreased vasodilation and
hypertension. Subsequent in-depth characterisation revealed an
excess of mesenchymal cells in the cardiac valves in all
homozygotes, while the following was only observed in a subset
of the animals: (1) abnormal septation of the outow tract, i.e., a
severely narrowed ascending aorta and an enlarged pulmonary
trunk or the reverse, (2) thrombotic lesions and ischaemia in lung,
liver, and kidney, (3) subepicardial vascular malformations in the
ventricular wall with loss of multiple smooth muscle cell layers in
large vessels, and (4) thickening of the endocardium. Interestingly,
a background sensitivity for the survival of homozygotes up to
weaning was observed by comparing mouse models on different
genetic backgrounds (i.e., 129/SvEv × BALB/cBy, 129/SvEv × C57Bl/
6, inbred 129/SvEv), which corresponded to the severity of cardiac
defects. Heterozygotes were not further studied, and no gender-
specic analyses were performed. Even though similar anomalies
were described in humans, it is still unanswered whether these
mice also present BAV, aortic valve calcication, hypoplastic left
heart and what the relative position of the aorta and pulmonary
artery is. No gross non-cardiovascular anomalies were described,
yet this has not been investigated into detail.
The next-studied knock-out mouse model
55
, on a C57BL/
6J × BALB/c background, was generated to investigate the
consequences of Smad6 loss during cartilage development.
Homozygotes displayed craniofacial anomalies like a domed skull
and shortened snout, but no defects in cranial sutures were found.
Abnormalities in the skeleton were observed too, such as posterior
transformation of cervical vertebrae (C7), atter thoracic vertebral
bodies, presence of bilateral ossication centres in lumbar
vertebrae, and bid sternebrae due to incomplete sternal band
fusion. In addition, homozygotes were smaller in size, as
conrmed by shorter appendicular bones, and stage-specic
defects in endochondral bone formation were found like the
delayed onset of hypertrophy at midgestation and expanded
hypertophic zone at late gestation. Furthermore, signicant
embryonic and neonatal lethality was observed, as merely 5% of
the progeny were homozygous and all live-born pups died within
24 h after birth due to an unspecied cause. Heterozygotes were
not examined in this model, alike with other organ systems,
especially no data on the cardiovascular system in the homo-
zygotes were reported.
The last published model
56
was generated on a CD1 back-
ground to elucidate the effects of Smad6 loss on blood vessel
development. Wylie et al. reported on embryonic and postnatal
lethality of homozygotes (all died by P26), in addition to regions
of haemorrhages in skin and brown fat pads without any sign of
hyperplastic valve thickening in these animals. The observed
vessel phenotype was a consequence of disrupted endothelial cell
junctions, thereby compromising vessel wall integrity. No in-depth
I. Luyckx et al.
7
Published in partnership with CEGMR, King Abdulaziz University npj Genomic Medicine (2022) 68
experiments were performed on heterozygous animals, nor other
organ systems were examined.
Altogether, this mouse knock-out data support a role for unique
genetic background-related clinical presentations. Additional gene
expression or pathway analyses in the different Madh6-decient
mouse models might provide essential insights into the pathogen-
esis of these phenotypes. With respect to the observed cardiovas-
cular phenotype in 129/SvEv × BALB/cBy, 129/SvEv × C57Bl/6 and
inbred 129/SvEv Madh6
/
mice, a major codominant modier
gene for lethality might be present. Alike to Tgfβ1
/
mice created
on different genetic backgrounds to study angiogenesis
5759
,
independent but epistatically interacting genetic loci might be
found that determine the incidence of lethality depending on the
model. Interesting modifying genes have already been described
to alter the response of lack to TGF-β1 in mice, suggesting that
proper TGF-βsignalling is key for embryonic survival. Whether by
analogy, improper BMP or TGF-βsignalling explains the incidence
of lethality in Madh6
/
mice with a cardiovascular phenotype
remains to be determined.
CELLULAR MECHANISMS ORCHESTRATED BY SMAD6
Epithelial-to-mesenchymal transition (EMT) is a reversible funda-
mental biological process for (1) the formation of the body plan,
(2) the differentiation of multiple tissues and organs, and (3) to
repair tissues. EMT is an extremely coordinated multifaceted
process, in which cells disrupt their intercellular adhesion
complexes and lose their apicobasal polarity in order to
migrate
60,61
. Two highly conserved and critical regulators of EMT
are the TGF-βand BMP signalling pathway, which either
stimulates or tempers this process, respectively
62
. Hence, SMAD6
modulates EMT by interfering with, predominantly, BMP
signalling
54,63
.
The mechanosensitive BMP signalling pathway (Fig. 2) regulates
cellular lineage commitment, morphogenesis, differentiation,
proliferation and apoptosis
64,65
. BMPs activate numerous path-
ways, of which the SMAD signalling pathway has best been
studied
66
. BMP signalling interferes with its own signalling as
SMAD6, a direct BMP target, selectively recruits SMURF1 to BMP
type 1 receptors
67
or competes with receptor-regulated Smads for
binding to SMAD4
68
, thereby establishing a negative feedback
Fig. 2 Schematic overview of SMAD-(in)dependent bone morphogenetic protein (BMP) signalling pathway (oversimplication). Upon
BMP ligand binding, specic type I and type II receptors form a heterotetrameric complex. The type II receptor phosphorylates the type I
receptor, which, in turn, phosphorylates Smad1, Smad5, and Smad8 (canonical BMP signalling). Phosphorylated Smads propagate the signal
via complex formation with Smad4 and translocates into the nucleus, where it regulates the expression of BMP-responsive target genes. In
addition to Smad activation, activated BMP receptor complexes initiates several intracellular pathways to modulate BMP-dependent cellular
responses like PI3-kinase, ERK, RhoA, and MAPK/JNK. Canonical BMP signalling is intracellularly inhibited by inhibitory Smads (Smad6, Smad7)
and E3 ubiquitin ligases like Smurf1 and Smurf2. Created with BioRender.com. The gure was exported under a paid subscription.
I. Luyckx et al.
8
npj Genomic Medicine (2022) 68 Published in partnership with CEGMR, King Abdulaziz University
loop. A further level of control is achieved by cross-talk with TGF-β,
FGF, MAPK, Hedgehog, PI3K/Akt, Wnt/beta-catenin, retinoic acid
and Notch signalling pathways in order to regulate cellular BMP-
related processes in a very tight spatial and temporal
manner
64,65,69
.
SMAD6 signalling in cardiovascular development
Dysregulation of BMP signalling has extensively been investigated
in numerous cardiovascular diseases
1,70
. Interestingly, SMAD6-
decient patients mainly exhibit defects related to two discrete
cell lineages, namely second heart eld and neural crest cells.
Second heart eld cells are multipotent progenitors originating
from cardiac progenitor cells and contribute to distinct regions of
the myocardium, cardiac endothelial cells and smooth muscle
cells
71
, while neural crest cells are derived from the dorsal aorta
and migrate as multipotent cells into the developing outow tract
to coordinate outow tract septation
72
. During cardiac cushion
development, SMAD6 is specically expressed in endothelial cells
where it functions in (1) maintaining endothelial to mesenchymal
transition (EndMT)
54,63
, (2) stimulating cardiac cushions to grow
73
and (3) interacting with cardiac neural crest cells
74
, cells required
for aorticopulmonary septum formation. As such, this might
explain the marked clinical variability of SMAD6-decient patient
with BAV-related aortopathy as predominant phenotype, and,
emphasises the complexity of CHD, in which gene dosage, timing,
haemodynamic ow, and its interplay with other signalling
pathways like Notch and TGF-βare important too. For example,
endothelial cells can undergo EndMT to become either
myobroblast-like
75
or chrondrocyte- and osteoblast-like cells
76
,
depending on their cellular context.
SMAD6 signalling in cranial suture development
Gene discovery studies, and their subsequent characterisation in
mice, have determined highly conserved molecular pathways and
specic biological processes at different stages in cranial suture
development
45
. Initially, the strongest implication of BMP signal-
ling involvement was shown by BMP type 1 receptor (BMPR1A)
77
,
and by its convergence at key transcriptional factors downstream
of BMP, i.e., Msx2
78
and Twist1
79,80
, to regulate cell proliferation,
mesenchyme condensation, osteoblast differentiation, and osteo-
genesis. Subsequent work further supported a role of SMAD-
dependent signalling by the identication of causal mutations in
SKI
81
and SMAD3
82
in ShprintzenGoldberg and LoeysDietz
patients, both conditions associated with CRS. Additional evidence
has emerged as SMAD6-deciency increases the risk for CRS, and
in particular for metopic synostosis. In literature, metopic
synostosis has already been hypothesised to be the consequence
of abnormal maturation of neural crest-derived mesenchymal
stem cells via disturbed dynamics of cell identity or migration as a
common predisposing factor, and this can now be further
investigated
83,84
. Alternatively, processes not involved in cranial
suture development but affecting osteogenesis such as osteoblast
and osteoclast activity could be impaired too, and lead to CRS.
SMAD6 signalling in radioulnar development
Studies on BMP signalling in radioulnar development are very
scarce. So far, published data on RUS is limited to genetic
studies
9,13
and clinical descriptive reports lacking in-depth
functional analyses. Our current knowledge is inferred from
studies in axial skeletal development, with molecular pathways
like Wnt, Hedgehog, Notch, and FGF signalling pathways, to be
highly involved
85
. As RUS is believed to be the result of anomalous
differentiation and/or segmentation of the adjacent radius and
ulna, it could be true that BMPs lead to impaired mesenchymal
stem cell differentiation via Runx2 to promote osteoblast
differentiation from mesenchymal precursor cells
86,87
.
SUMMARY AND FUTURE OUTLOOK
In summary, three distinctive human genetic disorders are
caused by SMAD6 deciency without domain-specicor
mutation-type genotypephenotype correlation making proper
patient management difcult. Patients with cardiovascular
disease or craniosynostosis do not show any manifestations in
the other organ system within relatives of a single family,
suggesting that, (an)other factor(s) co-segregating with the
primary SMAD6 variant might explain the resultant phenotype.
To further explore this hypothesis, in-depth investigation into
the identication of the responsible cell type(s) and their
identity, as well as dening the predominant affected signalling
cascade(s) driving these disorders, will be fundamental for our
knowledge. Cell lineage tracing and spatial gene expression
analyses in Madh6-decient mouse models might unravel
important clues to discriminate the aficted processes leading
to cardiovascular disease, craniosynostosis and radioulnar
synostosis. Furthermore, a detailed clinical and genetic assess-
ment of additional SMAD6 variant-positive patients will be
needed, and, in particular, ascertain the complete phenotypic
picture of families with RUS, in which some clinical overlap with
CHD-, skull-, and skeletal-related anomalies might exist.
Other (additional) genetic factor(s) might explain incomplete
penetrance and extreme variability in phenotypical expressivity in
a patient with SMAD6 deciency. For example, rare (or common)
variants located in a regulatory element of the trans-wild-type
SMAD6 allele, or variants in genes (e.g., SMAD7)aficting
expression and/or activity of the BMP and/or the closely related
TGF-βsignalling activity are interesting avenues for further
exploration. It is worthwhile to consider genome-wide association
approaches that look into rare second-hitvariants with large
effect size in SMAD6-decient patients in order to add novel
information to the puzzle. Although this would aid to understand
the molecular basis of disease, the current available number of
SMAD6 mutant patients might not be sufcient to detect (a)
signal(s) even when only extreme phenotypes would be selected.
Nonetheless, in the upcoming years we will condently identify
the SMAD6-related molecular patterns associated with these three
distinctive genetic disorders. This will allow us to detect early at-
risk individuals and empower new therapies.
Reporting summary
Further information on research design is available in the Nature
Research Reporting Summary linked to this article.
DATA AVAILABILITY
All data generated or analysed during this study are included in this publish ed article
(and its supplementary information le).
Received: 29 June 2022; Accepted: 8 November 2022;
REFERENCES
1. Goumans, M. J., Zwijsen, A., Ten Dijke, P. & Bailly, S. Bone morphogenetic proteins
in vascular homeostasis and disease. Cold Spring Harb. Perspect. Biol.10, a031989
(2018).
2. Wang, R. N. et al. Bone morphogenetic protein (BMP) signaling in development
and human diseases. Genes Dis. 1,87105 (2014).
3. Calpena, E. et al. SMAD6 variants in craniosynostosis: genotype and phenotype
evaluation. Genet Med. 22, 14981506 (2020).
4. Gillis, E. et al. Candidate gene resequencing in a large bicuspid aortic valve-
associated thoracic aortic aneurysm cohort: SMAD6 as an important contributor.
Front. Physiol. 8, 400 (2017).
5. Jin, S. C. et al. Contribution of rare inherited and de novo variants in 2,871
congenital heart disease probands. Nat. Genet. 49, 15931601 (2017).
I. Luyckx et al.
9
Published in partnership with CEGMR, King Abdulaziz University npj Genomic Medicine (2022) 68
6. Kloth, K. et al. Biallelic variants in SMAD6 are associated with a complex cardi-
ovascular phenotype. Hum. Genet. 138, 625634 (2019).
7. Luyckx, I. et al. Conrmation of the role of pathogenic SMAD6 variants in bicuspid
aortic valve-related aortopathy. Eur. J. Hum. Genet. 27, 10441053 (2019).
8. Park, J. E. et al. A novel SMAD6 variant in a patient with severely calcied bicuspid
aortic valve and thoracic aortic aneurysm. Mol. Genet. Genom. Med. 7, e620 (2019).
9. Shen, F. et al. A genotype and phenotype analysis of SMAD6 mutant patients
with radioulnar synostosis. Mol. Genet. Genom. Med. 10, e1850 (2022).
10. Tan, H. L. et al. Nonsynonymous variants in the SMAD6 gene predispose to
congenital cardiovascular malformation. Hum. Mutat. 33, 720727 (2012).
11. Timberlake, A. T. et al. Two locus inheritance of non-syndromic midline cra-
niosynostosis via rare SMAD6 and common BMP2 alleles. eLife 5, e20125
(2016).
12. Timberlake, A. T. et al. De novo mutations in inhibitors of Wnt, BMP, and Ras/ERK
signaling pathways in non-syndromic midline craniosynostosis. Proc. Natl Acad.
Sci. USA 114, E7341E7347 (2017).
13. Yang, Y. et al. SMAD6 is frequently mutated in nonsyndromic radioulnar synos-
tosis. Genet Med. 21, 25772585 (2019).
14. Bakker, M. K. et al. Prenatal diagnosis and prevalence of critical congenital heart
defects: an international retrospective cohort study. BMJ Open 9, e028139 (2019).
15. Leirgul, E. et al. Birth prevalence of congenital heart defects in Norway
19942009-a nationwide study. Am. Heart J. 168, 956964 (2014).
16. Liu, Y. et al. Global birth prevalence of congenital heart defects 19702017:
updated systematic review and meta-analysis of 260 studies. Int. J. Epidemiol. 48,
455463 (2019).
17. McCracken, C. et al. Mortality following pediatric congenital heart surgery: an
analysis of the causes of death derived from the national death index. J. Am.
Heart Assoc. 7, e010624 (2018).
18. Verma, S. & Siu, S. C. Aortic dilatation in patients with bicuspid aortic valve. N.
Engl. J. Med. 370, 19201929 (2014).
19. Verstraeten, A., Luyckx, I. & Loeys, B. Aetiology and management of hereditary
aortopathy. Nat. Rev. 14, 197208 (2017).
20. Criado, F. J. Aortic dissection: a 250-year perspective. Tex. Heart Inst. J. 38,
694700 (2011).
21. Senser, E. M., Misra, S. & Henkin, S. Thoracic aortic aneurysm: a clinical review.
Cardiol. Clin. 39, 505515 (2021).
22. Boulet, S. L., Rasmussen, S. A. & Honein, M. A. A population-based study of
craniosynostosis in metropolitan Atlanta, 1989-2003. Am. J. Med. Genet. A 146A,
984991 (2008).
23. Lajeunie, E., Le Merrer, M., Bonaiti-Pellie, C., Marchac, D. & Renier, D. Genetic study
of nonsyndromic coronal craniosynostosis. Am. J. Med. Genet. 55, 500504 (1995).
24. Utria, A. F. et al. The importance of timing in optimizing cranial vault remodeling
in syndromic craniosynostosis. Plast. Reconstr. Surg. 135, 10771084 (2015).
25. Timberlake, A. T. & Persing, J. A. Genetics of nonsyndromic craniosynost osis. Plast.
Reconstr. Surg. 141, 15081516 (2018).
26. Tsai, J. Congenital radioulnar synostosis. Radio. Case Rep. 12, 552554 (2017).
27. Mathijssen, I. M. J. Introduction to updated guideline on treatment and man-
agement of craniosynostosis. J. Craniofac Surg. 32, 370 (2021).
28. Pei, X. & Han, J. Efcacy and feasibility of proximal radioulnar derotational
osteotomy and internal xation for the treatment of congenital radioulnar
synostosis. J. Orthop. Surg. Res. 14, 81 (2019).
29. Rao, P. S. Management of congenital heart disease: state of the art-part II-cya-
notic heart defects. Children 6, 54 (2019).
30. Rao, P. S. Management of congenital heart disease: state of the art; part
I-ACYANOTIC heart defects. Children 6, 54 (2019).
31. Braverman, A. C. et al. The bicuspid aortic valve. Curr. Probl. Cardiol. 30, 470522
(2005).
32. Blue, G. M., Kirk, E. P., Sholler, G. F., Harvey, R. P. & Winlaw, D. S. Congenital heart
disease: current knowledge about causes and inheritance. Med. J. Aust. 197,
155159 (2012).
33. McBride, K. L. et al. Inheritance analysis of congenital left ventricular outow tract
obstruction malformations: Segregation, multiplex relative risk, and heritability.
Am. J. Med Genet. A 134A, 180186 (2005).
34. Morton, S. U., Quiat, D., Seidman, J. G. & Seidman, C. E. Genomic frontiers in
congenital heart disease. Nat. Rev. 19,26
42 (2022).
35. Bonachea, E. M. et al. Rare GATA5 sequence variants identied in individuals with
bicuspid aortic valve. Pediatr. Res. 76, 211216 (2014).
36. Shi, L. M. et al. GATA5 loss-of-function mutations associated with congenital
bicuspid aortic valve. Int. J. Mol. Med. 33, 12191226 (2014).
37. Guo, D. C. et al. LOX mutations predispose to thoracic aortic aneurysms and
dissections. Circ. Res. 118, 928934 (2016).
38. Garg, V. et al. Mutations in NOTCH1 cause aortic valve disease . Nature 437,
270274 (2005).
39. Guo, D. C. et al. Mutations in smooth muscle alpha-actin (ACTA2) cause coronary
artery disease, stroke, and Moyamoya disease, along with thoracic aortic disease.
Am. J. Hum. Genet. 84, 617627 (2009).
40. Gould, R. A. et al. ROBO4 variants predispose individuals to bicuspid aortic valve
and thoracic aortic aneurysm. Nat. Genet. 51,4250 (2019).
41. Parker, L. E. & Landstrom, A. P. Genetic etiology of left-sided obstructive heart
lesions: a story in development. J. Am. Heart Assoc. 10, e019006 (2021).
42. Silberbach, M. et al. Cardiovascular health in turner syndrome: a scientic
statement from the American Heart Association. Circ. Genom. Precis. Med. 11,
e000048 (2018).
43. Hinton, R. B. et al. Hypoplastic left heart syndrome links to chromosomes 10q and
6q and is genetically related to bicuspid aortic valve. J. Am. Coll. Cardiol. 53,
10651071 (2009).
44. Mu, T. S., McAdams, R. M. & Bush, D. M. A case of hypoplastic left heart syndrome
and bicuspid aortic valve in monochorionic twins. Pediatr. Cardiol. 26, 884885
(2005).
45. Twigg, S. R. & Wilkie, A. O. A genetic-pathophysiological framework for cranio-
synostosis. Am. J. Hum. Genet. 97, 359377 (2015).
46. Goos, J. A. C. & Mathijssen, I. M. J. Genetic causes of craniosynostosis: an update.
Mol. Syndromol. 10,623 (2019).
47. Justice, C. M. et al. A genome-wide association study identies susceptibility loci
for nonsyndromic sagittal craniosynostosis near BMP2 and within BBS9. Nat.
Genet. 44, 13601364 (2012).
48. Justice, C. M. et al. A genome-wide association study implicates the BMP7 locus
as a risk factor for nonsyndromic metopic craniosynostosis. Hum. Genet. 139,
10771090 (2020).
49. Rizzo, R. et al. Autosomal dominant and sporadic radio-ulnar synostosis. Am. J.
Med. Genet. 68, 127134 (1997).
50. Spritz, R. A. Familial radioulnar synostosis. J. Med. Genet. 15, 160162 (1978).
51. Mazauric-Stuker, M., Kordt, G. & Brodersen, D. Y aneuploidy: a further case of a
male patient with a 48,XYYY karyotype and literature review. Annales de. genet-
ique 35, 237240 (1992).
52. Murgai, A., Altmeyer, S., Wiegand, S., Tylzanowski, P. & Stricker, S. Cooperation of
BMP and IHH signaling in interdigital cell fate determination. PLoS ONE 13,
e0197535 (2018).
53. Stafford, D. A., Brunet, L. J., Khokha, M. K., Economides, A. N. & Harland, R. M.
Cooperative activity of noggin and gremlin 1 in axial skeleton development.
Development 138, 10051014 (2011).
54. Galvin, K. M. et al. A role for smad6 in development and homeostasis of the
cardiovascular system. Nat. Genet. 24, 171174 (2000).
55. Estrada, K. D., Retting, K. N., Chin, A. M. & Lyons, K. M. Smad6 is essential to limit
BMP signaling during cartilage development. J. Bone Miner. Res. 26, 24982510
(2011).
56. Wylie, L. A., Mouillesseaux, K. P., Chong, D. C. & Bautch, V. L. Developmental
SMAD6 loss leads to blood vessel hemorrhage and disrupted endothelial cell
junctions. Dev. Biol. 442, 199209 (2018).
57. Bonyadi, M. et al. Mapping of a major genetic modier of embryonic lethality in
TGF beta 1 knockout mice. Nat. Genet. 15, 207211 (1997).
58. Tang, Y. et al. Epistatic interactions between modier genes confer strain-specic
redundancy for Tgfb1 in developmental angiogenesis. Genomics 85,6070
(2005).
59. Tang, Y. et al. Genetic modiers interact with maternal determinants in vascular
development of Tgfb1(-/-) mice. Hum. Mol. Genet. 12, 15791589 (2003).
60. Kalluri, R. & Weinberg, R. A. The basics of epithelial-mesenchymal transition. J.
Clin. Investig. 119, 14201428 (2009).
61. Thiery, J. P., Acloque, H., Huang, R. Y. & Nieto, M. A. Epithelial-mesenchymal
transitions in development and disease. Cell 139, 871890 (2009).
62. Kahata, K., Dadras, M. S. & Moustakas, A. TGF-beta family signaling in epithelial
differentiation and epithelial-mesenchymal transition. Cold Spring Harb. Perspect.
Biol.10, a022194 (2018).
63. Desgrosellier, J. S., Mundell, N. A., McDonnell, M. A., Moses, H. L. & Barnett, J. V.
Activin receptor-like kinase 2 and Smad6 regulate epithelial-mesenchymal
transformation during cardiac valve formation. Dev. Biol. 280, 201210 (2005).
64. Garside, V. C., Chang, A. C., Karsan, A. & Hoodless, P. A. Co-ordinating Notch, BMP,
and TGF-beta signaling during heart valve development. Cell. Mol. life Sci.: CMLS
70, 28992917 (2013).
65. Gonzalez, D. M. & Medici, D. Signaling mechanisms of the epithelial-
mesenchymal transition. Sci. Signal 7, re8 (2014).
66. Nishimura, R. et al. The role of Smads in BMP signaling. Front. Biosci.: a J. virtual
Libr. 8, s275s284 (2003).
67. Goto, K., Kamiya, Y., Imamura, T., Miyazono, K. & Miyazawa, K. Selective inhibitory
effects of Smad6 on bone morphogenetic protein type I receptors. J. Biol. Chem.
282, 2060320611 (2007).
I. Luyckx et al.
10
npj Genomic Medicine (2022) 68 Published in partnership with CEGMR, King Abdulaziz University
68. Hata, A., Lagna, G., Massague, J. & Hemmati-Brivanlou, A. Smad6 inhibits BMP/
Smad1 signaling by specically competing with the Smad4 tumor suppressor.
Genes Dev. 12, 186197 (1998).
69. Derynck, R. & Zhang, Y. E. Smad-dependent and Smad-independent pathways in
TGF-beta family signalling. Nature 425, 577584 (2003).
70. Wang, J., Greene, S. B. & Martin, J. F. BMP signaling in congenital heart disease:
new developments and future directions. Birth Defects Res. A Clin. Mol. Teratol. 91,
441448 (2011).
71. Kelly, R. G. The second heart eld. Curr. Top. Dev. Biol. 100,3365 (2012).
72. Plein, A., Fantin, A. & Ruhrberg, C. Neural crest cells in cardiovascular develop-
ment. Curr. Top. Dev. Biol. 111, 183200 (2015).
73. Yamada, M., Szendro, P. I., Prokscha, A., Schwartz, R. J. & Eichele, G. Evidence for a
role of Smad6 in chick cardiac development. Dev. Biol. 215,4861 (1999).
74. Delot, E. C. Control of endocardial cushion and cardiac valve maturation by BMP
signaling pathways. Mol. Genet. Metab. 80,2735 (2003).
75. Kovacic, J. C. et al. Endothelial to mesenchymal transition in cardiovascular dis-
ease: JACC state-of-the-art review. J. Am. Coll. Cardiol. 73, 190209 (2019).
76. Gomez-Stallons, M. V., Wirrig-Schwendeman, E. E., Hassel, K. R., Conway, S. J. &
Yutzey, K. E. Bone morphogenetic protein signaling is required for aortic valve
calcication. Arteriosclerosis Thrombosis Vasc. Biol. 36, 13981405 (2016).
77. Komatsu, Y. et al. Augmentation of Smad-dependent BMP signaling in neural crest
cells causes craniosynostosis in mice. J. Bone Miner. Res. 28, 14221433 (2013).
78. Jabs, E. W. et al. A mutation in the homeodomain of the human MSX2 gene in a
family affected with autosomal dominant craniosynostosis. Cell 75, 443450
(1993).
79. el Ghouzzi, V. et al. Mutations of the TWIST gene in the Saethre-Chotzen syn-
drome. Nat. Genet. 15,4246 (1997).
80. Howard, T. D. et al. Mutations in TWIST, a basic helix-loop-helix transcription
factor, in Saethre-Chotzen syndrome. Nat. Genet. 15,3641 (1997).
81. Doyle, A. J. et al. Mutations in the TGF-beta repressor SKI cause Shprintzen-
Goldberg syndrome with aortic aneurysm. Nat. Genet. 44, 12491254 (2012).
82. Velchev, J. D., Van Laer, L., Luyckx, I., Dietz, H. & Loeys, B. Loeys-Dietz syndrome.
Adv. Exp. Med Biol. 1348, 251264 (2021).
83. Piacentino, M. L., Hutchins, E. J. & Bronner, M. E. Essential function and targets of BMP
signaling during midbrain neural crest delamination. Dev. Biol. 477,251261 (2021).
84. Siismets, E. M. & Hatch, N. E. Cranial neural crest cells and their role in the
pathogenesis of craniofacial anomalies and coronal craniosynostosis. J. Dev. Biol.
8, 18 (2020).
85. Williams, S., Alkhatib, B. & Serra, R. Development of the axial skeleton and
intervertebral disc. Curr. Top. Dev. Biol. 133,4990 (2019).
86. Liu, Q. et al. Recent advances of osterix transcription factor in osteoblast differ-
entiation and bone formation. Front Cell Dev. Biol. 8, 601224 (2020).
87. Phimphilai, M., Zhao, Z., Boules, H., Roca, H. & Franceschi, R. T. BMP signaling is
required for RUNX2-dependent induction of the osteoblast phenotype. J. Bone
Miner. Res. 21, 637646 (2006).
88. Miyazawa, K. & Miyazono, K. Regulation of TGF-beta family signalling by inhibi-
tory smads. Cold Spring Harb Perspect Biol.9, a022095 (2017).
89. Lo, R. S. et al. The L3 loop: a structural motif determining specic interactions
between SMAD proteins and TGF-βreceptors. EMBO J.17, 9961005 (1998).
ACKNOWLEDGEMENTS
This research was supported by funding from the University of Antwerp (Methusalem-
OEC grant GenomedFFB190208). B.L. holds a consolidator grant from the European
Research Council (Genomia ERC-C OG-2017 -77194 5). B.L. a nd A.V. are m embers of the
European Reference Network on rare multisystemic vascular disorders (VASCERN -
project ID: 769036 partly co-funded by the European Union Third Health Programme). I.L.
is supported by the Outreach project (Dutch Heart Foundation).
AUTHOR CONTRIBUTIONS
I.L. and B.L. conceived the idea. I.L. drafted the initial manuscript and revised the
manuscript. A.V., M.J.G. and B.L. contributed to the critical review and editing of the
manuscript. All authors contributed to the review and the nal approval of the
completed version.
COMPETING INTERESTS
The authors declare no competing interests.
ADDITIONAL INFORMATION
Supplementary information The online version contains supplementary material
available at https://doi.org/10.1038/s41525-022-00338-5.
Correspondence and requests for materials should be addressed to Bart Loeys.
Reprints and permission information is available at http://www.nature.com/
reprints
Publishers note Springer Nature remains neutral with regard to jurisdictional claims
in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this license, visit http://
creativecommons.org/licenses/by/4.0/.
© The Author(s) 2022
I. Luyckx et al.
11
Published in partnership with CEGMR, King Abdulaziz University npj Genomic Medicine (2022) 68
... SMAD6 encodes an inhibitor of bone morphogenic protein signaling and is highly expressed in cardiac valves and the aortic root (23,24). SMAD6 mutations have been described in 3-9% of individuals with BAV and TAA, including predicted loss of function mutations that are similar to the frameshifts and indel that we identified in 3 EBAV probands (25,26). The SMAD6 mutant phenotype frequently includes other left ventricular obstructive lesions, such as coarctation and HLHS, as well as conotruncal defects, and these variable phenotypes are recapitulated in mice with knockout of the SMAD6 analog Madh6 (23,(26)(27)(28). ...
... SMAD6 mutations have been described in 3-9% of individuals with BAV and TAA, including predicted loss of function mutations that are similar to the frameshifts and indel that we identified in 3 EBAV probands (25,26). The SMAD6 mutant phenotype frequently includes other left ventricular obstructive lesions, such as coarctation and HLHS, as well as conotruncal defects, and these variable phenotypes are recapitulated in mice with knockout of the SMAD6 analog Madh6 (23,(26)(27)(28). There is some evidence that SMAD6 functions downstream of NOTCH1 signaling to regulate endothelial cell motility and adhesion (28). ...
Preprint
Full-text available
Bicuspid Aortic Valve (BAV) is the most common adult congenital heart lesion with an estimated population prevalence of 1%. We hypothesize that early onset complications of BAV (EBAV) are driven by specific impactful genetic variants. We analyzed whole exome sequences (WES) to identify rare coding variants that contribute to BAV disease in 215 EBAV families. Predicted pathogenic variants of causal genes were present in 111 EBAV families (51% of total), including genes that cause BAV (8%) or heritable thoracic aortic disease (HTAD, 17%) when mutated. After appropriate filtration, we also identified 93 variants in 26 novel genes that are associated with autosomal dominant congenital heart phenotypes, including recurrent deleterious variation of FBN2, MYH6, channelopathy genes, and type 1 and 5 collagen genes. These findings confirm our hypothesis that unique rare genetic variants contribute to early onset complications of BAV disease.
... 6 The established feedback inhibitors of the TGFβ signaling pathway include inhibitory Smad (I-Smad) Smad7, Smad6, E3 ubiquitin protein ligases Smurf1/2, and other RING/U-box type E3 ubiquitin ligases that bind to Smad3 and Smad4 to induce their degradation. [5][6][7][8][9] The role of TGFβ is complicated and context dependent. For example, TGFβ is involved in both proinflammatory and suppressive immune responses, such as its distinct roles in different T cell subsets and differentiation stages, as well as in inflammatory reaction of myeloid cells in different stimulatory conditions. ...
Article
Full-text available
Inflammation plays a crucial role in the development of various disease conditions or is closely associated with them. Inflammatory cytokines like TNF often engage in interactions with other cytokines and growth factors, including TGFβ, to orchestrate inflammatory process. Basal/endogenous TGFβ signaling is a universal presence, yet the precise way TNF communicates with TGFβ signaling to regulate inflammation and influence inflammatory levels in macrophages has remained elusive. To address this question, this study utilized genetic approaches and a combination of molecular and cellular methods, including conditional TGFβ receptor knockout mice, human cells, RNAseq, ATACseq and Cut & Run‐seq. The results reveal that the TGFβ signaling functions as a vital homeostatic pathway, curtailing uncontrolled inflammation in macrophages in response to TNF. Conversely, TNF employs two previously unrecognized mechanisms to suppress the TGFβ signaling. These mechanisms encompass epigenetic inhibition and RBP‐J‐mediated inhibition of the TGFβ signaling pathway by TNF. These mechanisms empower TNF to diminish the antagonistic influence exerted by the TGFβ signaling pathway, ultimately enhancing TNF's capacity to induce heightened levels of inflammation. This reciprocal suppression dynamic between TNF and the TGFβ signaling pathway holds unique physiopathological significance, as it serves as a crucial “braking” mechanism. The balance between TNF levels and the activity of the endogenous TGFβ signaling pathway plays a pivotal role in determining the overall extent of inflammation. The potential for therapeutically augmenting the TGFβ signaling pathway presents an intriguing avenue for countering the impact of TNF and, consequently, developing innovative strategies for inflammation control.
... Importantly, an important inhibitory protein in SAMD proteins, SMAD6, was identified in our study, which is progressively expressed at elevated levels with prolonged pesudotime in the development of TOF. SMAD6 expression levels are tightly correlated with the development of CHD [38]. Ruter et al. showed that SMAD6 is also a necessary element for endothelial cell flow-mediated responses [39]. ...
... Also, given our observation of the impact of SMAD6 loss-of-function on ID1 and SGK1 expression downstream, we analyzed our WES data for either ID1 or SGK1 variants prior to our filtering strategy to find any putative modifier variant; however, we did not find any. SMAD6-deficiency caused by germline loss-of-function mutations has been previously associated with three distinct human congenital conditions [65]. Interestingly, identical nucleotide changes have been described in patients with different pathological phenotypes; indeed, this is the first time that SMAD6 has been related to HHT. ...
Article
Background: Hereditary hemorrhagic telangiectasia (HHT) is a rare vascular disease inherited in an autosomal dominant manner. Disease-causing variants in endoglin (ENG) and activin A receptor type II-like 1 (ACVRL1) genes are detected in around 90% of the patients; also 2% of patients harbor pathogenic variants at SMAD4 and GDF2. Importantly, the genetic cause of 8% of patients with clinical HHT remains unknown. Here, we present new putative genetic drivers of HHT. Methods: To identify new HHT genetic drivers, we performed exome sequencing of 19 HHT patients and relatives with unknown HHT genetic etiology. We applied a multistep filtration strategy to catalog deleterious variants and prioritize gene candidates based on their known relevance in endothelial cell biology. Additionally, we performed in vitro validation of one of the identified variants. Results: We identified variants in the INHA, HIF1A, JAK2, DNM2, POSTN, ANGPTL4, FOXO1 and SMAD6 genes as putative drivers in HHT. We have identified the SMAD6 p.(Glu407Lys) variant in one of the families; this is a loss-of-function variant leading to the activation of the BMP/TGFβ signaling in endothelial cells. Conclusions: Variants in these genes should be considered for genetic testing in patients with HHT phenotype and negative for ACVRL1/ENG mutations.
Article
The BMP signaling pathway plays a crucial role in regulating early embryonic development and tissue homeostasis. SMAD6 encodes a negative regulator of BMP, and rare variants of SMAD6 are recurrently found in individuals with birth defects. However, we observed that a subset of rare pathogenic variants of SMAD6 consistently exhibited positive regulatory effects instead of the initial negative effects on the BMP signaling pathway. We sought to determine whether these SMAD6 variants have common pathogenic mechanisms. Here, we showed that pathogenic SMAD6 variants accompanying this functional reversal exhibit similar increases in deamidation. Mechanistically, increased deamidation of SMAD6 variants promotes the accumulation of the BMP receptor BMPR1A and the formation of new complexes, both of which lead to BMP signaling pathway activation. Specifically, two residues, N262 and N404, in SMAD6 were identified as the crucial sites of deamidation, which was catalyzed primarily by glutamine-fructose-6-phosphate transaminase 2 (GFPT2). Additionally, treatment of cells harboring SMAD6 variants with a deamidase inhibitor restored the inhibitory effect of SMAD6 on the BMP signaling pathway. Conversely, when wild-type SMAD6 was manually simulated to mimic the deamidated state, the reversed function of activating BMP signaling was reproduced. Taken together, these findings show that deamidation of SMAD6 plays a crucial role in the functional reversal of BMP signaling activity, which can be induced by a subset of various SMAD6 variants. Our study reveals a common pathogenic mechanism shared by these variants and provides a potential strategy for preventing birth defects through deamidation regulation, which might prevent the off-target effects of gene editing.
Preprint
Full-text available
Thoracic aortic aneurysm and dissection (TAAD) associates with a high mortality rate. Despite the existence of different mouse models for TAAD, the underlying disease mechanisms remain elusive. Treatment options are limited and mainly consist of surgical repair at critical aortic diameters as current pharmacological interventions are unable to stop disease progression. In humans, loss of function (LOF) of SMAD3 and SMAD6 impairs vascular homeostasis, increasing the risk for TAAD. We developed a zebrafish model for thoracic aortic dissection/rupture by targeting both ohnologs of smad3 and smad6 . At 10 days post fertilization, we found an increased diameter of the ventral aorta in smad3a -/- ; smad3b -/- double knockout zebrafish, while smad6a -/- ; smad6b -/- double knockout zebrafish have a reduced aortic diameter associated with early mortality. We discovered that a smad3a -/- ; smad3b -/- ; smad6a -/- ; smad6b -/- quadruple knockout (qKO) zebrafish model is viable and survives to adulthood, although exposure to stress leads to sudden death. Histological analysis of the adult ventral aorta shows medial elastolysis, aortic dissections and ruptures at sites exposed to high biomechanical stress. RNA-sequencing of 5 days post fertilization qKO zebrafish indicates a profile of reduced negative regulation of proteolysis and upregulation of melanogenesis, a previously unaddressed pathway in this pathology. We confirm that pharmacological modulation of tyrosinase, the enzyme responsible for the production of melanin, influences aortic morphology. Overall, the qKO mutant, thus far the only known zebrafish model of thoracic aortic dissection and rupture, reveals novel SMAD3/6-dependent pathways that impact thoracic aortic homeostasis, in this way opening avenues for the development of novel treatments in TAAD.
Article
Introduction: Increased levels of pro-inflammatory markers have been reported in tissues of individuals with Coronavirus Disease 2019 (COVID-19). We hypothesize that inflamed dental pulp tissues of individuals with previous history of COVID-19 may present a differential inflammatory gene expression profile in comparison to individuals who never had COVID-19. Materials and methods: Dental pulp tissues were collected from 27 individuals referred for endodontic treatment due to symptomatic irreversible pulpitis. Of these, 16 individuals had a history of COVID-19 (6 months to 1 year post infection) and 11 individuals had no previous history of COVID-19 (controls). Total RNA from pulp tissue samples was extracted and subjected to RNA sequencing for comparison of differentially expressed genes (DEGs) among groups. DEGs showing log2(fold-change) > 1 or < -1, and P < .05 were considered significantly dysregulated. Results: RNA sequencing identified 1461 genes as differentially expressed among the groups. Of these, 311 were protein coding genes, 252 (81%) which were upregulated and 59 (19%) which were downregulated in the COVID group compared to controls. The top upregulated genes in COVID group were HSFX1 (4.12 fold-change) and LINGO3 (2.06 fold-change); significantly downregulated genes were LYZ (-1.52 fold-change), CCL15 and IL8 (-1.45 fold-change). Conclusions: Differential gene expression in dental pulp tissues of COVID and non-COVID groups suggests potential contribution of COVID-19 on dysregulating inflammatory gene expression in the inflamed dental pulp.
Article
Full-text available
Background SMAD6 variants have been reported in patients with radioulnar synostosis (RUS). This study aimed to investigate the genotypes and phenotypes for a large cohort of patients with RUS having mutant SMAD6. Methods Genomic DNA samples were isolated from 251 RUS sporadic patients (with their parents) and 27 RUS pedigrees. Sanger sequencing was performed for the SMAD6 coding regions. For positive probands, co-segregation and parental-origin analysis of SMAD6 variants and phenotypic re-evaluation were performed for their family members. Results We identified 50 RUS probands with SMAD6 variants (13 co-segregated with RUS in pedigrees and 37 in RUS-sporadic patients). Based on the new and previous data, we identified SMAD6 mutated in 16/38 RUS pedigrees and 61/393 RUS sporadic patients, respectively. Overall, 93 SMAD6 mutant patients with RUS were identified, among which 29 patients had unilateral RUS, where the left side was more involved than the right side (left:right = 20:9). Female protective effects and non-full penetrance were observed, in which only 6.90% mothers (vs. ~50% fathers) of SMAD6 mutant RUS probands had RUS. Pleiotropy was observed as a re-evaluation of SMAD6 mutant families identified: (a) three families had axial skeletal malformations; (b) two families had polydactyly; and (c) eight families had other known malformations. Conclusion SMAD6 was mutated in 42.11% RUS pedigrees and 15.52% RUS sporadic patients. The RUS patients with SMAD6 variants exhibit both non-full-penetrance, variable expressivity, pleiotropy, female protective effects, and the left side is more susceptible than the right side.
Article
Full-text available
Congenital heart disease is the most common congenital defect observed in newborns. Within the spectrum of congenital heart disease are left‐sided obstructive lesions (LSOLs), which include hypoplastic left heart syndrome, aortic stenosis, bicuspid aortic valve, coarctation of the aorta, and interrupted aortic arch. These defects can arise in isolation or as a component of a defined syndrome; however, nonsyndromic defects are often observed in multiple family members and associated with high sibling recurrence risk. This clear evidence for a heritable basis has driven a lengthy search for disease‐causing variants that has uncovered both rare and common variants in genes that, when perturbed in cardiac development, can result in LSOLs. Despite advancements in genetic sequencing platforms and broadening use of exome sequencing, the currently accepted LSOL‐associated genes explain only 10% to 20% of patients. Further, the combinatorial effects of common and rare variants as a cause of LSOLs are emerging. In this review, we highlight the genes and variants associated with the different LSOLs and discuss the strengths and weaknesses of the present genetic associations. Furthermore, we discuss the research avenues needed to bridge the gaps in our current understanding of the genetic basis of nonsyndromic congenital heart disease.
Article
Full-text available
With increasing life expectations, more and more patients suffer from fractures either induced by intensive sports or other bone-related diseases. The balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption is the basis for maintaining bone health. Osterix (Osx) has long been known to be an essential transcription factor for the osteoblast differentiation and bone mineralization. Emerging evidence suggests that Osx not only plays an important role in intramembranous bone formation, but also affects endochondral ossification by participating in the terminal cartilage differentiation. Given its essentiality in skeletal development and bone formation, Osx has become a new research hotspot in recent years. In this review, we focus on the progress of Osx’s function and its regulation in osteoblast differentiation and bone mass. And the potential role of Osx in developing new therapeutic strategies for osteolytic diseases was discussed.
Article
Full-text available
Craniofacial anomalies are among the most common of birth defects. The pathogenesis of craniofacial anomalies frequently involves defects in the migration, proliferation, and fate of neural crest cells destined for the craniofacial skeleton. Genetic mutations causing deficient cranial neural crest migration and proliferation can result in Treacher Collins syndrome, Pierre Robin sequence, and cleft palate. Defects in post-migratory neural crest cells can result in pre- or post-ossification defects in the developing craniofacial skeleton and craniosynostosis (premature fusion of cranial bones/cranial sutures). The coronal suture is the most frequently fused suture in craniosynostosis syndromes. It exists as a biological boundary between the neural crest-derived frontal bone and paraxial mesoderm-derived parietal bone. The objective of this review is to frame our current understanding of neural crest cells in craniofacial development, craniofacial anomalies, and the pathogenesis of coronal craniosynostosis. We will also discuss novel approaches for advancing our knowledge and developing prevention and/or treatment strategies for craniofacial tissue regeneration and craniosynostosis.
Article
Full-text available
Enrichment of heterozygous missense and truncating SMAD6 variants was previously reported in nonsyndromic sagittal and metopic synostosis, and interaction of SMAD6 variants with a common polymorphism near BMP2 (rs1884302) was proposed to contribute to inconsistent penetrance. We determined the occurrence of SMAD6 variants in all types of craniosynostosis, evaluated the impact of different missense variants on SMAD6 function, and tested independently whether rs1884302 genotype significantly modifies the phenotype. We performed resequencing of SMAD6 in 795 unsolved patients with any type of craniosynostosis and genotyped rs1884302 in SMAD6-positive individuals and relatives. We examined the inhibitory activity and stability of SMAD6 missense variants. We found 18 (2.3%) different rare damaging SMAD6 variants, with the highest prevalence in metopic synostosis (5.8%) and an 18.3-fold enrichment of loss-of-function variants comparedwith gnomAD data (P < 10⁻⁷). Combined with eight additional variants, ≥20/26 were transmitted from an unaffected parent but rs1884302 genotype did not predict phenotype. Pathogenic SMAD6 variants substantially increase the risk of both nonsyndromic and syndromic presentations of craniosynostosis, especially metopic synostosis. Functional analysis is important to evaluate missense variants. Genotyping of rs1884302 is not clinically useful. Mechanisms to explain the remarkable diversity of phenotypes associated with SMAD6 variants remain obscure.
Chapter
Loeys-Dietz syndrome is an autosomal dominant aortic aneurysm syndrome characterized by multisystemic involvement. The most typical clinical triad includes hypertelorism, bifid uvula or cleft palate and aortic aneurysm with tortuosity. Natural history is significant for aortic dissection at smaller aortic diameter and arterial aneurysms throughout the arterial tree. The genetic cause is heterogeneous and includes mutations in genes encoding for components of the transforming growth factor beta (TGFβ) signalling pathway: TGFBR1, TGFBR2, SMAD2, SMAD3, TGFB2 and TGFB3. Despite the loss of function nature of these mutations, the patient-derived aortic tissues show evidence of increased (rather than decreased) TGFβ signalling. These insights offer new options for therapeutic interventions.
Article
Thoracic aortic aneurysms are common. Most thoracic aortic aneurysms are degenerative. However, some are associated with connective tissue disorders, bicuspid aortic valves, or familial/genetic predisposition. Most are asymptomatic, discovered incidentally on imaging. Aortic diameter is the best predictor of the natural history and risk of complications. Treating hypertension and smoking cessation can slow their growth. Surveillance imaging and referral for prophylactic aortic repair based on absolute aneurysm diameter is the primary means to decrease mortality from thoracic aortic aneurysm. We provide a practical evidence-based summary of the pathophysiology, risk factors, associated genetic syndromes, and clinical management of thoracic aortic aneurysms.
Article
The application of next-generation sequencing to study congenital heart disease (CHD) is increasingly providing new insights into the causes and mechanisms of this prevalent birth anomaly. Whole-exome sequencing analysis identifies damaging gene variants altering single or contiguous nucleotides that are assigned pathogenicity based on statistical analyses of families and cohorts with CHD, high expression in the developing heart and depletion of damaging protein-coding variants in the general population. Gene classes fulfilling these criteria are enriched in patients with CHD and extracardiac abnormalities, evidencing shared pathways in organogenesis. Developmental single-cell transcriptomic data demonstrate the expression of CHD-associated genes in particular cell lineages, and emerging insights indicate that genetic variants perturb multicellular interactions that are crucial for cardiogenesis. Whole-genome sequencing analyses extend these observations, identifying non-coding variants that influence the expression of genes associated with CHD and contribute to the estimated ~55% of unexplained cases of CHD. These approaches combined with the assessment of common and mosaic genetic variants have provided a more complete knowledge of the causes and mechanisms of CHD. Such advances provide knowledge to inform the clinical care of patients with CHD or other birth defects and deepen our understanding of the complexity of human development. In this Review, we highlight known and candidate CHD-associated human genes and discuss how the integration of advances in developmental biology research can provide new insights into the genetic contributions to CHD.
Article
BMP signaling plays iterative roles during vertebrate neural crest development from induction through craniofacial morphogenesis. However, far less is known about the role of BMP activity in cranial neural crest epithelial-to-mesenchymal transition and delamination. By measuring canonical BMP signaling activity as a function of time from specification through early migration of avian midbrain neural crest cells, we found elevated BMP signaling during delamination stages. Moreover, inhibition of canonical BMP activity via a dominant negative mutant Type I BMP receptor showed that BMP signaling is required for neural crest migration from the midbrain, independent from an effect on EMT and delamination. Transcriptome profiling on control compared to BMP-inhibited cranial neural crest cells identified novel BMP targets during neural crest delamination and early migration including targets of the Notch pathway that are upregulated following BMP inhibition. These results suggest potential crosstalk between the BMP and Notch pathways in early migrating cranial neural crest and provide novel insight into mechanisms regulated by BMP signaling during early craniofacial development.