TGFbRIIb Mutations Trigger Aortic Aneurysm Pathogenesis by

Transcription

TGFbRIIb Mutations Trigger Aortic Aneurysm Pathogenesis by
DOI: 10.1161/CIRCGENETICS.112.964064
TGFbRIIb Mutations Trigger Aortic Aneurysm Pathogenesis by Altering
TGFb2 Signal Transduction
Running title: Bee et al.; TGFbRIIb mutations in aortic aneurysms
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Deve
vere
ve
reux
re
ux,, MD
ux
MD;;
Katharine J. Bee, PhD; David C. Wilkes, PhD; Richard B. De
Devereux,
Craig T. Basson, MD, PhD; Cathy J. Hatcher, PhD
C
enter for M
olec
eccularr C
ardiiollogy,, G
reen
nbeerg
gD
ivission off C
arrdiolog
ogyy,
og
Center
Molecular
Cardiology,
Greenberg
Division
Cardiology,
W il
We
illl Cornell
Corn
Co
rnel
rn
elll Medical
el
Medi
Me
d ca
di
call College,
Coll
Co
lleg
eg
ge,
e New
e York,
ew
Yor
ork,
k, NY
NY
Weill
Corresponding authors:
Cathy J. Hatcher, PhD
Craig T. Basson, MD, PhD
Greenberg Division of Cardiology
Novartis Institutes for BioMedical Research
Weill Cornell Medical College
220 Massachusetts Avenue
525 E. 68th Street
Cambridge, MA 02139
New York, NY 10065
Tel: 212-746-2201
Tel: 617-871-7652
Fax: 212-746-6669
Fax: 617-871-5203
E-mail: [email protected]
E-mail: [email protected]
Journal Subject Code: [109] Clinical genetics; [89] Genetics of cardiovascular disease; [147]
Growth factors/cytokines
1
DOI: 10.1161/CIRCGENETICS.112.964064
Abstract:
Background - Thoracic aortic aneurysm (TAA) is a common progressive disorder involving
gradual dilation of the ascending and/or descending thoracic aorta that eventually leads to
dissection or rupture. Nonsydromic TAA can occur as a genetically triggered, familial disorder
that is usually transmitted in a monogenic autosomal dominant fashion and is known as familial
TAA (FTAA). Genetic analyses of families affected with TAA have identified several
chromosomal loci and further mapping of FTAA genes has highlighted disease-causing
mutations in at least four genes: myosin heavy chain 11 (MYH11), a-smooth muscle actin
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
(ACTA2), transforming growth factor beta receptors I and II (TGFERI and TGFERII).
)
Methods and Results - We evaluated 100 probands to determine the mu
muta
mutation
tati
ta
tion
ti
on frequency
freq
fr
eque
eq
uenncy in
ue
n
MYH11, ACTA2, TGFbRI and TGFbRII in an unbiased population of indivi
individuals
idu
dual
alss wi
al
with
th ggenetically
e e
en
mediated TAA.
AA.
AA
A. In
n tthis
his study,
hi
stud
udy, 9% of patients had a mu
ud
m
mutation
tation in one
onne off the
the genes analyzed. 33% of
patients had
dm
mutations
utations inn ACTA2,
ACTA
AC
TA22, 3% iin
TA
n MYH11,
MYH
MY
H11, 1%
% in 7*)ȕ5II
7*)ȕ
7*
)ȕ5
)ȕ
5II
II and
ndd nno
o mu
m
mutations
taati
tion
onss were
on
were fo
found
o
in 7*)ȕ5, Additionally,
Add
dditional
ally,, w
al
wee id
identified
denttif
i ie
ied mu
mut
mutations
tat on
tati
onss inn a 755 ba
base
ase ppair
a r alt
ai
alternatively
ternat
attiv
i elyy spliced
spplicedd TGFbRII
TG
exon, exon 1a WKDWSURGXFHVWKH7*)ȕ5,,E
WKDWSURGXFH
FHVV WKKH
FH
H7*
7*
*)ȕ
ȕ5,
5 ,E isoform
issoffor
isof
orm
m and
an
nd accounted
a co
ac
coun
unnte
tedd fo
forr 2% ooff patients with
O r in vvitro
Ou
itroo analyses
it
a al
an
a ys
y es ind
diccat
atee that
t at tthe
th
hee 7*
*)ȕȕ5,,EEac
acti
t va
vati
t ngg mut
utat
atio
onss aalter
ltter
e rec
ecep
ep
p
mutations. Our
indicate
7*)ȕ5,,Eactivating
mutations
receptor
pon
po
on TGFb2
TGFb
TG
Fb22 si
Fb
sign
signaling.
gnal
alin
al
ingg.
in
function upon
Conclusions - We propose that TGFbRIIb expression is a regulatory mechanism for TGFb2
signal transduction. Dysregulation of the TGFb2 signaling pathway, as a consequence of
TGFbRIIb mutations, results in aortic aneurysm pathogenesis.
Key words: aneurysm; aorta; cardiovascular diseases; genetics; TGF-beta pathway aneurysm
2
DOI: 10.1161/CIRCGENETICS.112.964064
Introduction
Thoracic aortic aneurysm (TAA) is a common progressive disorder involving gradual dilation of
the ascending and/or descending thoracic aorta that eventually leads to dissection or rupture.
TAAs are often clinically silent and unsuspected until dissection or rupture occurs. The result is
significant morbidity and mortality despite advances in surgical and percutaneous treatments for
aortic disease. Although TAA is often a feature of Mendelian complex connective tissue
disorders such as Marfan syndrome (MFS), Ehlers-Danlos syndrome type IV (EDS), or LoeysDownloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Dietz syndrome (LDS), most TAAs occur as isolated nonsyndromic disorders.
Nonsydromic
isorders. Nonsydrom
omi
om
TAA can also be a familial disorder that is usually transmitted in a monogenic
autosomal
onogeniic aaut
tosomal
al
dominant fashion.
approximately
TAA.
a hi
ashi
hion
on. These
on
T es
Th
e e genetically
ge
triggered TAAs
As account
account for appro
roximately 20% of TA
ro
A 1-6
Genetic analyses
alysses of familial
al
famillial TAA
TAA
AA ((FTAA)
FTAA
A ) havee iidentified
denntifieed sseveral
e eral
ev
al chr
chromosomal
romos
osomaal lo
os
loci.
ocii. F
Further
urthh
ur
FTA
AA ggenes
enees has
has hi
high
ig ligh
ghte
gh
tedd di
te
ise
seas
asee-caaus
as
usin
i g muta
in
m
mu
uta
tation
onns in
i at
at least
leas
astt four
as
four ggenes:
enes
en
es::
mapping off FTAA
highlighted
disease-causing
mutations
myosin heavy
a y chain 11 ((MYH11),
avy
MYH1
MY
H111)
1 , a-smooth
h muscl
muscle
le actin
actiin ((ACTA2),
AC
CTA
TA2)
2), tran
transforming
sforming
f
g growth
growth factor
faa
-10
10
beta receptors I and
d II ((TGF
TGFERII and
d TGFERII
RII).
I) 77-10
Recentt studies
R
t di hhave also
l iid
identified
d tif
tifi
ifii d mutations
t
in two novel genes, MYLK and SMAD3, that are linked to syndromic aortic aneurysms and
dissections.11, 12 Also, disease genes remain to be determined at additional loci such as AAT1
(also known as FAA1) on chromosome 11q2313 and AAT2 (also known as TAAD1) on
chromosome 5q13.14
Because of the identification of TGFERI and TGFERII mutations in aortic aneurysm
syndromes such as LDS, considerable attention has been devoted to the role that TGFE may play
in FTAA pathogenesis. The TGFE receptor superfamily is comprised of cytokines that control
numerous diverse cellular processes including cell proliferation, differentiation, angiogenesis and
modification of the extracellular matrix (ECM).13-16 Canonical TGFȕ signaling is initiated when
3
DOI: 10.1161/CIRCGENETICS.112.964064
a TGFE ligand binds to TGFERII, resulting in the recruitment of TGFERI. Upon ligand binding,
TGFERII activates TGFERI via trans-phosphorylation of its kinase domain and propagates
downstream signaling actions. Receptor-regulated (R-) Smads are substrates of the TGFERI
kinase and cytoplasmic phosphorylation of R-Smads allows for translocation of the Smad
complexes to the nucleus in order to regulate transcription of target genes.17
Previous studies identified mutations in TGFȕRI, TGFȕRII, ACTA2, and MYH11 in
individuals with familial TAA. In most cases, genetic screenings for mutations in these genes
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
ith
ther
th
er with
wit
ithh an eextensive
x en
xt
ensi
sivv
si
have focused primarily on patients referred to genetic subspecialists either
ective ti
iss
ssue
u disorder
ue
dis
i or
orde
d r and,
family history or with obvious features of a complex Mendelian connective
tissue
therefore, these
hesse ppatients
hes
atien
nts hhave
ave an increased likelihoodd of harboring
ng a m
mutation.
utation. However, su
such
u
reppresent a sm
re
mall su
ubseet of thos
se wit
th ge
eneetica
caall
call
llyy me
ediated TAA
AA. T
AA
he vvast
ast m
ajj
individuals represent
small
subset
those
with
genetically
mediated
TAA.
The
majority
p
with lim
mit
i ed or unknown
un
history and
and
n are without
with
thou
th
o t evidence of a
of patients present
limited
familyy history
ynd
ynd
ndro
romi
ro
micc di
mi
diso
sord
so
rder
rd
er. Th
er
Thes
esee pa
es
pati
tien
ti
ents
en
ts rrepresent
epre
ep
rese
re
sent
se
nt ddiagnostic
iagn
ia
gnos
gn
osti
os
ticc di
ti
dile
lemm
le
mmas
mm
as ffor
or ppracticing
ract
ra
ctic
ct
icin
ic
ingg
in
complex syndromic
disorder.
These
patients
dilemmas
physicians. This study addresses the potential impact of genetic testing for these four TAA
genes on clinical management of TAA patients. We determined the frequency of mutations in
these four TAA genes in an unbiased population that is more representative of the population of
individuals with genetically mediated TAA seen in cardiovascular clinical practice.
Methods
Patient cohort collection
The cohort of patients enrolled in this study consisted of 100 consecutive adult probands from a
clinical population with non-syndromic, potentially genetically triggered aortic aneurysms.
FTAA patients were collected from those presenting to cardiologists and cardiothoracic surgeons
at Weill Cornell Medical Center. Written informed consent was obtained from all subjects
4
DOI: 10.1161/CIRCGENETICS.112.964064
according to a protocol approved by the institutional review board of Weill Cornell Medical
College. To enroll, subjects needed to have been diagnosed with thoracic aortic dilation,
aneurysm, or dissection and meet at least one of these criteria:
1.
Age at diagnosis of aortic disease less than 50 years
2.
Positive family history of aortic aneurysm or dissection in at least one 1st or 2nd
degree relative
3.
Features of a connective tissue disorder, such as arachnodactyly, pectus
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
carinatum, or pectus excavatum.
These inclusion criteria were established to represent patients that might
reasonably
clinically
ght reasonab
bly
ly bbee cl
cli
in
in
suspected too have
genetically
excluded
hav
avee a ge
av
gen
neti
ticcally mediated disorder. Pa
ti
Patients were excl
lud
u ed if they met clinical
clinii
diagnostic criteria
MFS,
rare
syndromes
well
criteeria for MF
cr
FS, LDS,
LDS, or
o EDS
D since
DS
sin
nce eetiologies
tiolo
logies
es for
for these
theese ra
aree sy
yndrom
o ess are
om
are w
ell
known and do not
ott generally
generrallly ppresent
rese
re
s nt ddiagnostic
iagnosti
ia
ticc di
ti
ddilemmas
lemmass ttoo physicians.
lemm
phhyssici
iccians
ians.
DNA Isolation
and Mutation
Analysis
t
tion
Mutatiion A
naly
lysis
ly
Blood or saliva
patients.
ali
al
li a samples
le were
ere obtained
btaii d ffrom
r
patients
atiient Genomic
Ge mii DNA
DNA was
as isolated
isolat
l ed
d from
f
lymphoblasts separated from whole blood (QIAamp DNA Blood kit, Qiagen) and saliva
(Oragene-DNA kit, DNA Genotek) per manufacturer’s instructions. Exons of ACTA2, MYH11,
TGFȕRI and TGFȕRII were PCR amplified with gene-specific primers from genomic DNA
isolated from each patient. Primer sequences are available upon request. Additional mutational
analyses of TGFȕRII focused on an alternatively spliced exon, exon 1a that substitutes a 26
amino acid peptide for Val51 in the receptor’s extracellular domain. This resultant TGFȕRII is
often referred to as TGFȕRIIb, and the specific properties and function of TGFȕRIIb are not well
documented.18
PCR products were purified by vacuum filtration using a MultiScreen-PCR filter plate
5
DOI: 10.1161/CIRCGENETICS.112.964064
(Millipore) per manufacturer’s instructions. Purified PCR products were bidirectionally
sequenced and analyzed on an automated sequencer (ABI 3130XL) with BigDye Terminator
v3.1 (Applied Biosystems). Exons with sequence variants were analyzed in family members
when available. In addition, a minimum of 200 control chromosomes from a population of
‘normal’ samples (mixed ethnicity unaffected individuals without known aortic disease) were
also analyzed either by restriction fragment length polymorphism (RFLP) analysis or denaturing
high-performance liquid chromatography (dHPLC) on a WAVE Nucleic Acid Fragment
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Analysis System (Transgenomic). Additional sets of ethnically matched
used
ed controls were us
e as
noted in the results section below.
Sequence
were
mutations
u nce vvariants
uen
aria
ar
iaantss w
ere considered mutation
onns if they (1) caused
ed
d a non-synonomous
amino acid change
could
potentially
protein
(2)
were
absent
population
chaange or cou
uld
d pot
tenti
tiially alter
alter pro
oteein structure,
sttruct
ctur
ure,
ur
e (2
e,
2) wer
re ab
bsent ffrom
room a po
opull
of at least 200
ethnically
matched
control
chromosomes,
co-segregated
with
disease
2 et
ethn
hnic
hn
i ally
ic
ym
atch
at
tch
hed
d con
ontr
on
trol chr
tr
h om
hr
omosom
omees, and
and (3)
(33 co
co-se
o segr
gregat
gr
ated
at
ed
d wit
ithh di
it
dise
seas
a e in
the family if
samples
also
available
analysis.
addition,
i family
y member samp
ples were als
l o avai
ilabl
ble for an
alys
ly is
i . IIn
n ad
ddition,, we examined
exam
m
the National
Biotechnology
Information
all Center
C te ffor
o Bi
Biot
Biotechnolog
hnoll
Inffo atiio (NCBI)
(NCB
(N
CBI)
I) single
ingll nucleotide
n cl
cleotide
l tid
ide polymorphism
pol
oll morphism
phi
his
(SNP) database of 4356 chromosomes for ACTA2, MYH11, TGFȕRI and TGFȕRII
polymorphisms in genome build 37.3 released in October 2011.
RNA Analyses
Total RNA was isolated from either lymphocytes or homogenized human aortic tissue using
TRIzol reagent (Invitrogen) per manufacturer’s instructions. RNA was subjected to reverse
transcriptase PCR (RT-PCR; One-Step RT-PCR Kit, Qiagen) to preferentially amplify either the
TGFȕRII or TGFȕRIIb isoform with exon-specific primers surrounding, or internal to, the
alternatively spliced exon. RT-PCR reactions were performed under the following conditions:
50oC for 30 mins (cDNA synthesis), 95oC for 15 mins (polymerase heat activation) followed by
6
DOI: 10.1161/CIRCGENETICS.112.964064
94oC for 45 secs, 52oC for 80 secs and 72oC for 60 secs for 35 cycles.
Plasmid Constructs
Full-length cDNAs of both TGFȕRII and TGFȕRIIb were reverse transcribed as described above
from RNA extracted from patient fibroblasts using primers immediately flanking the coding
region of TGFȕRII. RT-PCR products were cloned into the pCR2.1-TOPO vector using the
TOPO TA cloning kit (Invitrogen). Plasmid DNA was isolated (QIAprep Miniprep kit, Qiagen).
The entire coding region of TGFȕRII and TGFERIIb in each construct was sequenced biDownloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
directionally in each cDNA construct to confirm the correct full-length
h sequence for bot
both.
th. The
(Quik
ikCh
Changee kit,
kit
itt,
H56N TGFȕRIIb mutation was generated by site-directed mutagenesiss (QuikChange
Stratagene) per
peer manufacturer’s
m nu
ma
n fa
facttur
urer’s instructions. The D40N
D40
4 N TGFȕRIIb
TGFȕ
FȕRIIbb mu
mutation
uta
tation was generated by
Overlap Extension
x ension PCR.1199 Initial
xte
In
nitiial PCR
P R reactions
PC
reacctiionns were
re per
performed
rfo
form
r ed
rm
d und
under
derr the
he follo
following
l wi
lo
winng con
conditions:
nd
m
minute,
t , 550
0oC for
for 2 mi
minutes,
inute
tes, and 772
2oC for 2 minutes
minute
i tes for 25 cycles.
cyc
y les. P
PCR
CR
R products
prodductt were
95oC for 1 minute
subjected to
o gel
gel eelectrophoresis
lect
le
ctro
ct
roph
ro
phor
ores
or
esis
es
is aand
nd ggel
el purified
pur
urif
ifie
iedd (Q
(QIA
(QIAquick
IAqu
quic
qu
ickk Gel
Gel Extraction
Extr
Ex
trac
tr
acti
ac
tion
on Kit,
Kit
it,, Qiagen)
Qiag
Qi
agen
ag
en)) pe
en
per
manufacturer’s
’ iinstructions.
i
Subsequent
S b
PCR reactions
i
were performed
f
d using
i purified
ifi d DNA
DN
from the initial PCR product using the same cycle conditions.
Wildtype TGFȕRII as well as wildtype and mutant isoforms of TGFȕRIIb were
subcloned into XhoI and BamHI sites of the pcDNA 3.1(-) expression vector 3’ to a cassette
encoding a Kozak sequence in order to generate TGFȕRII- or TGFȕRIIb-cDNA3.1. PCR
amplification with XhoI-Kozak-TGFEII-F and BamHI-TGFERII-R primers facilitated cloning
into pcDNA3.1 (Invitrogen). The entire coding region of each TGFȕRII and TGFȕRIIb construct
was bi-directionally sequenced to confirm the correct full-length sequence.
Cell Culture, Transfection and TGFE
E stimulation
A skin biopsy containing primary dermal fibroblasts from an individual harboring the H56N
7
DOI: 10.1161/CIRCGENETICS.112.964064
TGFȕRII mutation was cultured in Dulbecco’s Modification of Eagle’s Medium (DMEM,
Mediatech) supplemented with 10% fetal bovine serum (FBS) and 0.1 mg/mL primocin. Cells
were maintained and studied at low passage (passages 2-5). Normal human dermal fibroblasts
(NHDF) and L6 rat myoblast cells were obtained from ATCC and grown in DMEM with 10%
FBS.
Low passage primary dermal fibroblasts (passages 2-5) were serum starved for 24 hours
and then stimulated with 5 ng/mL recombinant human (rh) TGFȕ1 or rhTGFȕ2 (R&D Systems)
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
in the presence of 10% serum. Following stimulation, cells were washed
PBS
ed twice in cold P
B
BS
containing 1 mM Na 3 VO 4 , and lysed at 0, 0.5, 1, 2, 4, and 24 hours post-stimulation
ost-sttim
i ullattion in R
RIPA
IP
M TrisHCl
Tris
Tr
isHC
is
HC
HCl
C pH7.6,
pH
H7.6, 150 mM NaCl, 1% IIGEPAL,
G PAL, 0.5% sodium
GE
soodi
d um deoxycholate, 0.1%
0
buffer (50 mM
SDS) supplemented
lemented
lem
em
mented with protease
protteaase in
iinhibitors
hibi
bittorrs (1X
bi
1X
X Protease
Prooteasse In
Inh
Inhibitor
hibiito
or C
Cocktail
ock
ckta
tail Tablets,
ta
Tab
ablets
ab
tss, Roche;
R
Roche
oche 1%
otein Ph
Phos
hos
o phat
attas
asee In
IInhibitor
hi or C
hi
hibito
Cocktail,
ock
ktail
tail
ta
il, Mi
Millipore)
illi
llipor
ll
ipore)
e)) ffor
or sub
subsequent
ubse
ub
sequen
sequ
ent Western
We n blot
We
blott aana
bl
n
na
Ser/Thr Protein
Phosphatase
analysis
as described
d below. Cell sti
stimulation
imulati
l ion was pe
pperformed
rfformedd iin
n tripl
triplicate.
p ic
i ate.
3x10
05 L6 cells
ell
lls were
ere plat
plated
l ed
d one da
dday bbefore
eff
transfection
tr sff tii and
ndd transfected
t
fected
d with
ith
ithh 500
500 ng of
either wildtype (wt) or mutant TGFȕRII-cDNA3.1 constructs (wt-TGFȕRII-, wt-TGFȕRIIb-,
H56N-TGFȕRIIb- or D40N-TGFȕRIIb-cDNA3.1) in low serum media using LipofectAMINE
(Invitrogen). Cells either remained unstimulated or were stimulated with 50 pM TGFȕ1 or 50
pM TGFȕ2 for 0, 0.5, 1 or 2 hours and lysed as described above in RIPA buffer supplemented
with protease inhibitors for Western blot analysis. All cell transfections and stimulations were
performed in triplicate.
SDS-PAGE and Western Blotting
Lysate protein concentrations were determined (Coomassie Plus Bradford Assay kit, Pierce). 20
mg of cell lysate were electrophoresed on 10% polyacrylamide gels (Pierce) and transferred to
8
DOI: 10.1161/CIRCGENETICS.112.964064
PVDF membrane (GE Healthcare). Protein expression was determined by Western blotting with
primary antibodies to anti-phosphorylated Smad2 (pSMAD2, Cell Signaling) or anti-b-actin.
(Sigma). Bound antibodies were detected by incubation with goat anti-rabbit secondary antibody
(Cell Signaling) followed by chemiluminescence (ECL Plus, GE Healthcare). Densitometry was
performed on a BioRad Gel Doc MultiAnalyst system.
Statistical Analysis
All values are expressed as mean + SEM or % and 95% confidence intervals for categorical
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
variables. Statistical analyses were performed using ANOVA and Student’s
udent’s t-test. P<0.05
P<0.
0.05
0.
0 was
considered significant. A Sharp-Wilk normality test of our Western blot
lot data
datta reve
revealed
aled
l tthat
hat it
ha
followed normal
o ma
orma
mall di
dist
distributions
stri
st
ribu
ri
b tiions with a P>0.01 and ha
bu
had
ad eequal
qual variance. Bi
B
Binomial
nomial power
calculationss indicate
inndicate that oour
ur ppower
ower
e to de
er
ddetect
teectt mut
mutations
utatio
ons with
witth a prevalence
prrevalen
en
ncee aass lo
low
ow ass 1%
% iiss 555% in
cont
n rol ch
nt
hro
romo
moso
s mees aand
nd 95
995%
% in
in 5588
888 con
onntrol
ol chr
hrom
hr
omosoomes
omos
es. A
es
lso
so, bi
bino
nom
m
a sample off 2000 co
control
chromosomes
control
chromosomes.
Also,
binomial
power calculations
u
ulations
indicate th
that
hat our ppower
ower to ddetect
etect NC
NCB
NCBI
BI SN
S
SNPs
P withh a prevalence
Ps
prevalence of 5/4356
5/4355
chromosomes
mes is
is 21%
21% iin 200
200 chromosomes
ch
h
and
d 49
49%
% iin
n 588
588 chromosomes,
chromosomes
hr
whereas
hhereas our
o r ddet
detection
et
power is 37% in 200 chromosomes and 74% in 588 chromosomes with a SNP having a
prevalence of 10/4356 chromosomes.
Results
Patient Cohort
The cohort consisted of predominantly male patients (77 men and 23 women) (Table 1). This
distribution is consistent with previous studies in which men predominate in a series of clinically
apparent genetically meditated TAAs.1 The majority of patients were Caucasian (80/100) and
the remainder were of Hispanic, African-American, Native American or Asian descent. Patients
9
DOI: 10.1161/CIRCGENETICS.112.964064
ranged in age from 21-93 years old with an average age of 53. The average weight of patients
was 85.1 kg and average height was 177.3 cm with an average body surface area of 2m2. Of the
100 patients, 64 were diagnosed at less than 50 years of age. Sixty-seven had a family history of
TAA or thoracic aortic dissection. Only 14 exhibited connective tissue abnormalities, including
joint hypermobility, pectus excavatum and pectus carinatum, but none met or nearly met
diagnostic criteria for Marfan or other syndromes.20 At the time of enrollment, 42 patients had
previously undergone aortic surgery. Aortic dissections had been reported in 27 of the patients.
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Aneurysm Gene Mutational Analyses
All patients in this cohort underwent sequencing-based mutational analyses
ACTA2,
alyses off tthe
h AC
he
ACTA
TA22,
TA
MYH11, 7*)ȕ5,and
*)ȕ
*)ȕ
)ȕ5,
5,an
5,
andd 7*)ȕ5,,
an
7*)ȕ
7*
) 5,, genes.
Sequencing
ACTA2
missense
mutations,
T108M,
R118Q,
uen
ncing of thee AC
A
CTA
TA
A2 ggene
ene re
rrevealed
eveealed
d tthree
hreee mis
isse
is
seens
n e mu
m
tattioonss, T10
08M
8M, R
118
18Q
Q and
G270E (Fig.
g 1)
g.
1).
). Tw
Two in
intronic
ntr
t on
onic
i ppolymorphisms
ic
o ym
ol
ymor
orphisms
or
ms w
were
eree se
see
seen,
en,
n hhowever,
ow
wever
err, no eexonic
xoni
nicc poly
ni
polymorphisms
ymo
morp
rphi
his
hi
were detected.
ted. The R118Q
8Q
Q mutatio
mutation
i n in
i ffamily
amil
ilyy JNW
il
JNW ha
hhass been ppreviously
reviiouslly repo
reported
p rted in two oth
other
TAA families.
iies 21 T1
T108M
T108
08M
M andd G270E
G270
G2
70E
E were
ere not
ot ffo
found
ndd in
i 200
200 Caucasian/Northern
C
Caa casian/Northern
ian/N
/Northhe E
European
ropeann
control chromosomes. All three affected probands had TAAs, two of which led to acute
dissections, and all required surgery. Both the R118Q (Family JNW) and G270E (Family ANS)
mutations co-segregated with disease in families. Family members of SY92, who carried the
T108M mutation, were unavailable for genetic analysis. Patient SY92 also had an atrial septal
defect (ASD) and patent ductus arteriosus (PDA). None of the ACTA2 mutations were found in
the NCBI SNP database.
Three mutations and 13 polymorphisms were identified in the MYH11 gene (Fig. 2),
including two missense mutations (R1590Q and E1899D), and one splice site alteration: a seven
nucleotide substitution located five base pairs 3’ to exon 27. None of the variants were detected
10
DOI: 10.1161/CIRCGENETICS.112.964064
in 200 Caucasian/Northern European control chromosomes. The mutations co-segregated with
aortic disease in families JNE and ANHH. Family members of patient ANO II-2 (carrying a
seven nucleotide substitution located five base pairs 3’ to exon 27) were unavailable for genetic
analysis. Individual ANHH II-2 exhibited TAAs and also a bicuspid aortic valve, however,
ANO II-2 had a tricuspid aortic valve. Individual JNE II-1 exhibited TAA resulting in two
dissections. None of these mutations were identified in the NCBI SNP database except for
E1899D MYH11 that was found in 10/4356 chromosomes corresponding to a frequency of
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
0.23%. The significance of this polymorphism is unknown given that the NCBI database
se
includes patients with cardiovascular disease whose aortic aneurysm status
tatus iiss unknown.
unkn
k own. In our
study, we screened
chromosomes
who weree uunaffected
individuals
c en
creen
ened
ed
d 20
2000 ch
hromosomes from controll patients
pa
naffected individua
a
without known
own aortic disease.
ow
diseease..
Members
were
screened
mutations
TGFȕR
type
m s of tthe
mbers
he ccohort
ohor
oh
hortt we
w
re aalso
lso
ls
s scre
reeened
re
ened
e ffor
or m
utat
ut
atio
at
io
ons iin
n both
both
h TG
GFȕR ty
ype I aand
ndd type II
genes, TGFȕRI
FȕȕRII and TGFȕRII.
F
TGFȕ
FȕRI
RIII. In
In TGFȕRI,
T FȕRI
TG
RI, three
th
hree exonic
exoniic ppolymorphisms
olym
y orph
phhis
i ms and
andd no mutations were
w
found. Two
polymorphisms
mutation
TGFȕRII
o eexonic
onic
nii pol
l morphisms
hi
andd one m
tation
tatii were
ere detected
detected
d iinn TG
TGFȕ
TGF
FȕRI
RIII (F
(Fig
(Fi
ig 3).
33)) The
Thh
T
TGFȕRII missense mutation (A414T) in patient KNA II-2 is located in the protein’s kinase
domain, co-segregates with disease in the family, and was absent in 206 ethnically matched
(Ashkenazi Jewish) control chromosomes. All four family members carrying the mutation were
diagnosed with aortic aneurysm. Three of the four also have pectus excavatum, one of whom
was also diagnosed with an ostium secundum ASD. There was no evidence of other LDS
features such as vascular tortuosity or bifid uvula in probands or family members. This 7*)ȕ5,,
mutation was not found in the NCBI SNP database.
TGFȕRII has an additional alternatively spliced isoform containing a 75 base pair exon
(exon 1a) in its extracellular domain that produces TGFȕRIIb. TGFȕRII exon 1a was also
11
DOI: 10.1161/CIRCGENETICS.112.964064
sequenced in all 100 individuals. No polymorphisms were found in exon 1a, but two missense
mutations were identified. The D40N mutation (family KNK) was not detected in 224
Caucasian/Northern European control chromosomes. The H56N mutation (family ANV) cosegregated with disease in the family, and was absent from 588 control chromosomes including
384 ethnically matched (Ashkenazi Jewish) chromosomes. KNK II-1 had a TAA and also a
bicuspid aortic valve. ANV II-1 had an aortic aneurysm and pectus carinatum. H56N was not
found in the NCBI SNP database, but D40N 7*)ȕ5,,E, identified in patient KNKII-1 for which
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
we were unable to determine cosegregation with disease, was found in
chromosomes
n 5/4356 chromoso
ome
m
corresponding to a frequency of 0.11%. The significance of this polymorphism
unknown
morph
phhi m is
phis
i unkno
now
no
wn
given that the
h NC
he
NCBI
BI database
BI
dataaba
da
base includes patients withh cardiovascular
cardiovascular disease
dis
issea
e se whose aortic
aneurysm status
patients
tatus
ta
atu
us is unknown.
unknoown. Inn our
ouur study,
stud
udy, we
ud
we sc
sscreened
creeneed 2224
24 cchromosomes
hrom
omosom
mes from
m co
control
ontrol ppat
at
who were unaff
individuals
uunaffected
ffec
ff
eccted
teed indi
divi
vidu
idu
d al
als
ls without
wiith
thou
out known
ou
know
ownn aortic
ow
ao
orti
rttic
ic disease.
dis
isea
ease
ease
se.
In total,
o , 9 of 100 probands
otal,
proba
b nds
d were ffound
oundd to hhave
ave a mutation in
i one off the
th
he four ggenes
enes
analyzed. No
genotype-phenotype
were
observed
N genot
ot pe phenot
he t pe correlations
latii
ere obser
bs ed
d amongstt the
th
h probands’
obb ds’’ available
a ail
i
family members, and mutations within the same gene did not necessarily correspond to any
specific phenotypic variation. Families ANV, KNK, and KNA each have TGFȕRII mutations
yet exhibit considerable interfamilial variation. Four of the seven individuals with a TGFȕRII
mutation (including those with a mutation in the alternatively spliced exon) exhibited noncardiovascular connective tissue abnormality. Two were diagnosed with a congenital heart
defect.
Although MYH11 mutations have been previously associated with PDA9, none of the individuals
with MYH11 mutations detected in the current study were known to have PDA. Livedo
reticularis or iris flocculi were not observed in individuals in this study with ACTA2 mutations.
12
DOI: 10.1161/CIRCGENETICS.112.964064
TGFȕRII Expression and Activity
Previous studies22 established that TGFER kinase domain mutations inactivate the receptor
although downstream TGFE signaling is paradoxically increased. Similarly, we identified the
A414T TGFERII mutation in a patient and examined its kinase activity through in vitro
expression of A414T-TGFERII in a luciferase vector. Mutant TGFERII was inactive (data not
shown). In this study, we sought to understand how the novel TGFERIIb mutations we observed
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
outside of the TGFERII kinase domain might alter TGFE signaling. Tissue expression patterns
of TGFȕRII alternative splicing have not been established. Using RT-PCR,
-PC
PCR,
PC
R, we
we de
dete
determined
term
te
rmin
rm
ined
in
ed that
aort
rtic
rt
ic w
alll an
al
aand
d
both spliced isoforms, TGFȕRII and TGFȕRIIb, are expressed in the humann ao
aortic
wall
lymphocytes
e (Fig.
es
(F 4) aand
n aalso
nd
lso iinn cultured
c ltur
cu
ured dermal fibroblasts
fibrooblastts and
d ao
aaortic
rtic smoot
smooth
othh muscle
ot
musccle cellss (not
shown).
d
ffec
ff
eccts ooff mu
uta
tant
ntt TGFȕRIIb
TGF
GFȕ
ȕRI
RIIb
I oonn TGFȕ
Ib
TG
GFȕ si
ign
gnal
alin
al
in
ng, w
To determine
the eeffects
mutant
signaling,
wee compared rel
relative
GFȕ1 and TG
TGF
Fȕ2 si
sign
gnal
gn
alin
al
ing (indicated
ing
(ind
(i
(ind
ndic
dic
icat
atted
ated
ed bby
y pSMA
pS
SMAD2
MA
AD2
D2 llevels)
e el
ev
els)
s) iin
s)
n norm
no
orm
rmal
al ddermal
ermal
levels of TGFȕ1
TGFȕ2
signaling
pSMAD2
normal
fibroblasts with dermal fibroblasts isolated from individual ANV I-1 who is heterozygous for the
H56N TGFȕRIIb mutation (Fig 5). Although the specific contributions of canonical TGFE
signaling via Smads vs. noncanonical TGFE signaling via MEK/ERK pathways to the
pathogenesis of specific aneurysm and aneurysm related phenotypes remain under active
investigation,23-25 pSMAD2 provides a valuable biomarker of TGFE activity.26 Upon TGFȕ1
stimulation of normal dermal fibroblasts, we observed an increase in pSMAD2 levels peaking at
0.5 hours post-stimulation before declining by 4 hours. By contrast, TGFȕ1 stimulation of ANV
I-1 dermal fibroblasts resulted in delayed SMAD2 phosphorylation that peaked at 2 hours. Upon
stimulation with TGFȕ2, normal dermal fibroblasts exhibited similar kinetics to TGFȕ1
stimulation; pSMAD2 levels peaked at 0.5 hours although high levels of pSMAD2 persisted
13
DOI: 10.1161/CIRCGENETICS.112.964064
even 4 hours post–stimulation. TGFȕ2 stimulation of ANV I-1 dermal fibroblasts exhibited
distinct kinetics of SMAD2 phosphorylation. Although pSMAD2 levels peaked at 0.5 hours,
these levels rapidly declined by 1 hour and were markedly reduced at all time points compared to
normal dermal fibroblasts.
Since dermal fibroblasts from individual ANV I-1 express both wt- and mutant
TGFȕRIIb isoforms, one cannot distinguish the activities of each isoform in these cells.
Therefore, we utilized L6 rat myoblast cells lacking both endogenous TGFȕRIIb and TGFȕRIII
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
to compare 7*)ȕVLJQDOLQJDFWLYLWLHVin wt- and mutant TGFȕRIIb. L6
6 myoblasts were
transfected with constructs encoding wt-TGFȕRII, wt-TGFȕRIIb, H56N-TGFȕRIIb,
6N-TG
TGF
TG
FȕRI
RIIb
Ib,, or D40NIb
D40
TGFȕRIIb and
stimulated
preliminary
examination
a d tthen
heen stim
st
ti ul
ulated with 50 pM TGFȕ1 or
or TGFȕ2. Our pr
prel
e iminary examinatio
o of
pSMAD2 levels
post-stimulation
evels measured during
ev
durrinng the
the
h first
firrst 2 hhours
ouurs pos
ost-st
stim
imul
im
ulatio
ul
ion showed
sho
howeed that
ho
att 7*)ȕ5,,
7 )ȕ5,
7*
5,,
activity peaked
hours
a d att 0.
aked
00.5
5 ho
our
urss LQ7*)ȕ-stimulated
L 7*)
LQ
* ȕ
ȕ-stimul
ulat
ul
ated
at
ted
e L6
L6 cells,
cellls, whereas
ce
wherea
wh
h ea
eass it
i peaked
peake
kedd aatt 1 hhour
ke
ourr iin
ou
n
7*)ȕ-stimulated
cells. Therefore,
m
mulated
Thereffore,, in subsequent
subbsequ
q ent studies,
studdie
i s,, we assessed
assessed
d peak
p ak
pe
k 7*)ȕreceptor
7*)ȕ
ȕrecept
po
activity in genetically
geneticall
tii ll engineered
gii
ed
d L6 cells
ell
lls following
ffollo
oll
ll iing
n stimulation
stim
sti
im llation
atiio with
iith
th
h either
ithhe TGFȕ1
TGF
TG
Fȕ1 for
fo 00.5
5 hours
(Fig. 6) or TGFȕ2 for 1 hour (Fig. 7). L6 cells transfected with either wt-TGFȕRII or wtTGFȕRIIb exhibited comparable responsiveness to TGFȕ1 (Fig 6A, n=3). Similarly,
introduction of neither H56N-TGFȕRIIb nor D40N-TGFȕRIIb significantly modified TGFȕ1
responsiveness (Fig 6B, n=3). However, when cells were stimulated with TGFȕ2 (Fig. 7),
pSmad2 levels were reduced by 74% in cells transfected with wt-TGFȕRIIb compared to those
transfected with wt-TGFȕRII (Fig 7A; n=3, p=0.02). Introduction of either H56N-TGFȕRIIb or
D40N-TGFȕRIIb, then, ablated this reduction in receptor activity, and both resulted in a nearly
three-fold increase in pSmad2 levels (Fig 7B; n=3, p=0.009 and p=0.02, respectively).
14
DOI: 10.1161/CIRCGENETICS.112.964064
Discussion
FTAA is a clinically heterogeneous disorder exhibiting variation in both age of onset and degree
of aortic dilatation prior to dissection. FTAA can be part of a complex syndrome, such as LDS,
or an isolated finding. The four genes analyzed in this study (ACTA2, 0<+7*)ȕ5,, and
7*)ȕ5,,) were initially identified as associated with syndromic FTAA, and the cause of FTAA
in many families remains unknown. The utility of mutational analyses in clinical strategies for
isolated FTAA diagnostic workup is unclear.
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
The principal goal of our study was to address the potential value
genetic
lue of clinical gene
neettiic
testing of ACTA2, MYH11, TGFȕRI, and TGFȕRII in nonsyndromic FTAA
TAA
A in
in order
ordder ttoo improve
impr
patient caree an
and
diagnosis.
FTAA
nd di
diag
agno
ag
nosiis. Although these four FT
no
TAA causative ge
ggenes
ness are known to be
ne
prevalent in
molecular
n cohorts
coohorts ascertained
ascerrtaained
ed
d ffor
orr m
ollecuulaar ggenetic
en
netic
ic studies,
stu
udi
diees, their
th
heir contribution
co
ontriibutionn to disease
diseeasse in a
population relevant
clinical
studied.
study,
relev
van
nt to cli
lini
nica
icall practice
practiicee hhas
pr
as nnot
ott ppreviously
revi
viou
iousl
slly be
been
en stu
tudi
tu
died
di
ed
d. In th
this
is stu
udy,
dy wee
determined the frequency
mutations
in these
cohort
frequ
q ency
y off muta
tions
i
thhese four
four TA
TAA
A ge
ggenes
nes in
i a coho
h rt routinely
y seen iin
cardiology clinical
Individuals
known
MFS,
LDS,
linii l practice.
practice
tii
IIndi
ndi
di id als
ls ddiagnosed
ia
ed
d with
ith
ithh kno
kn n MFS
MF
S LDS
LDS or EDS
EDS were
ere
excluded. In this study, 9% of patients had a mutation in one of the genes analyzed. 3% of
patients had mutations in ACTA2, 3% in MYH11, and 3% in TGFȕRII. No mutations were found
in TGFȕRI, consistent with the reported rarity of TGFȕRI mutations outside of LDS.27-29
Previous studies reported higher rates of mutation (14% in ACTA2 and 5-10% in
TGFȕRII) than observed here upon screening the same genes.9, 10, 30 Our study differs from those
studies whose cohorts may have been ascertained through family-based programs and/or medical
genetic clinics to which patients are largely referred if they are believed to have signs or
symptoms of known disorders, such as LDS, MFS, and EDS. Patients in those studies are more
likely to harbor a mutation in one of these genes, and our cohort may be more representative of
15
DOI: 10.1161/CIRCGENETICS.112.964064
the patient population routinely presenting to cardiovascular clinical practices.
Our study provides an estimate of the potential value of genetic testing for mutations in
known aortic aneurysm disease genes as part of the diagnostic workup of these patients who are
often seen by the general cardiologist or cardiothoracic surgeon. The 95% confidence interval
for the point estimate of 9% in our population is consistent with finding a potentially causative
mutation in 5-16% of such patients in cardiovascular clinical practices. Genetic testing can be a
valuable adjunct for diagnostic management of aortic aneurysm since this disorder often goes
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
undiagnosed until a dissection or rupture occurs. Individuals identifiedd by genetic testingg as
a at
risk for aortic aneurysm development can undergo interval imaging earlier
monitor
arlier tto
o moni
ito
t r th
thee
progression
dissection.
n off aaortic
orti
or
tiic di
ddilation
laati
tion
o and to facilitate intervention
interve
vent
ve
n ion prior to rupture
ruppture
tu and/or dissection
tu
This study provides
foundation
future
that
prro
rovi
v des a foun
unndattioon ffor
o fut
or
turee sstudies
tuddiees th
hat will
wil
illl likely
lik
like
kely
ly provide
proovide
dee insight
iins
nsig
ghhtt into
into
nt how
h w
ho
enhanced diagnos
algorithms
improve
ddiagnostic
ossti
ticc algo
gori
rith
ithms
thms iincorporating
ncor
orpo
or
porating
po
ng routine
rou
o tiine TAA
ou
TAA
A ggenetic
enetticc ttesting
en
esti
es
tinng ccan
ti
a imp
an
mpro
rove
ve ppatient
at
at
outcomes and survival. Rega
FTAA
Regardless,
g rddless,, our stud
study
dy critically
crit
i icallly
ly hhighlights
ig
ghl
h ig
i hts the
th
he need
d ffor
or further FTA
gene identification,
triggered
aneurysm
study
ffication
icatiio since
in mostt genetically
geneticall
tii ll tri
ig
d aortic
rtiic ane
r sm patients
tii ts iin our
o r st
t d hhad no
evidence of mutation in any of the genes analyzed. With clinical deployment of exonic and
genome wide sequencing that do not rely on family-based analyses, cohorts such as the one
followed here will provide a rich source for such gene identification.
Although no genotype-phenotype correlation was found in this study, the statistical
power to detect correlations may have been inadequate due to the small number of individuals
with a mutation. Nonetheless, the study already highlights certain clinical diagnostic hazards.
For instance, PDA has been strongly associated with MYH11 mutations. In fact, though, we
observed PDA in the setting of ACTA2 mutations as well. Thus, the presence of PDA should not
provoke presumption of MYH11 mutations.
16
DOI: 10.1161/CIRCGENETICS.112.964064
TGFȕ signaling has become an emerging target for novel therapies for aortic aneurysms.
Previous studies have established that dysregulated TGFE signaling contributes to aortic
aneurysms.22, 31 However, a paradox in the mode of pathogenesis obfuscates a clear functional
role for TGFE in aneurysm development.16 Identification of TGFȕR mutations in LDS and
characterization of novel 7*)ȕ5,,b mutations in an alternatively spliced gene segment in our
study highlight the contribution of enhanced TGFE signaling to FTAA. Although previously
identified TGFȕR mutations modified the receptors’ kinase domain, this study identifies novel
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
mutations in an alternatively spliced segment of TGFȕRII that is not involved
nvvol
olve
vedd in kinase
ve
kin
inas
a e ac
as
acti
activity.
ti
Functional analyses of several kinase domain mutations have revealed
d conse
consequent
equ
quen
e t lo
en
loss
loss-ofss-oofss
function that
a ne
at
nev
nevertheless
verthe
heelesss ddisplayed
isplayed a paradoxical enh
enhancement
nhancement ooff TG
TGFȕ
GFȕ signaling in patient
patie
e By
e.
By contrast,
contrastt, mu
utaationns in the aalternatively
lterrnative
vely sspliced
plic
pl
i ed se
ic
egm
mennt off TG
GFȕRI
RIIb de
ddescribed
escc
aortic tissue.
mutations
segment
TGFȕRIIb
here are unique
i e since th
ique
they
y aaugment
uggment
nt rec
rreceptor
ecep
ptor activity
activi
activity,
ty,, and th
ty
these
hes
e e findings
findings prompted
prompted
ro ted us to evaluate
eva
m ca
mica
mi
call si
sign
gnif
gn
ific
if
ican
ic
ance
an
ce of
of th
thes
esee mu
es
muta
tati
ta
tion
ti
onss.
on
the biochemical
significance
these
mutations.
Prior mutational analyses of TGFERII have rarely included the alternatively spliced
segment.9, 27 Little is known about the function of this alternative receptor isoform.32, 33 A
previous study asserted that TGFERII requires an accessory receptor, TGFERIII, for efficient
binding of TGFE2 and subsequent signaling.34 Rotzer et al.33 proposed that TGFERIIb alone is
capable of binding TGFE1, TGFE2 and TGFE3 whereas del Re et al.32 suggested that TGFERIIb
alone is capable of binding only TGFE1 and TGFE3. In contrast to the binding data, del Re et al.
further proposed that TGFERIIb mediates in vitro TGFE2 signaling in a dose-dependent
manner.32 We demonstrate that the segment encoded by exon 1a does not alter receptor function
upon stimulation with TGFE1, but does alter TGFE2 signaling. TGFERIIb has a lower TGFE2–
stimulated activity than TGFERII, and mutations in this alternatively spliced segment reverse
17
DOI: 10.1161/CIRCGENETICS.112.964064
this effect, increasing receptor activity levels similar to that of prototypical TGFERII. We then
propose that TGFERIIb expression is a regulatory mechanism for TGFE2 signal transduction,
and dysregulation of the TGFE2 signaling pathway resulting from TGFERIIb mutations can
contribute to aneurysm pathogenesis.
Regulation of the TGFE signaling pathway is important in determining cellular outcome
and the underlying mechanisms are complex. This pathway depends upon several factors
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
LQFOXGLQJWKHVWRLFKLRPHWULFEDODQFHRI7*)ȕOLJDQGVDQGUHFHSWRUVH[SUHVVHGZLWKLQWKHFHOO
n TG
TGF
TGFb1
Fb1 si
sign
signaling
gnal
gn
a in
al
ingg in
Although TGFERIIb binds TGFE1,32, 33 we did not observe a change in
response to mutant TGFERIIb expression. Upon TGFE2 stimulation of cells
ls eexpressing
xpre
xp
ress
re
ssin
ss
ingg either
in
et
ei
wt-TGFERII
II orr wt-TG
II
wt-TGFERIIb,
GFE
ERI
R Ib,, wee obs
observed
bsser
bser
erveed sign
significantly
g if
gn
i ican
ntlyy le
less
s TGF
ss
TGFERIIb
G ERIIIb
GF
I activity
act
ctiv
ct
ivit
iv
i y re
relative
ela
l ti
tive
v too
TGFERII activity.
cti
cti
tivi
vity
vi
t . However,
How
owev
ow
ev
ver, mutant
mutaant
n TGFERIIb
TG
GFERIIb
b iisoforms
sofo
form
ms ab
ablated
blaatedd thiss rreduction
ed
duction
ucc n in
in receptor
reecept
ptorr
activity by increasing TGFE2-stimulated
TGF
FE22-st
stim
st
im
muullatted TGFERIIb
TGF
GFE
ER
RIIIb activity
act
ctiv
ivityy too levels
iv
lev
e el
elss equivalent
equi
eq
uiva
ui
vale
va
l nt to that of wtw
TGFERII. T
The
he incre
increase
easse in
nT
TGFE2
G E2 ssi
GF
signaling
ign
nal
a in
ng that
th
hat
a we
we ob
observe
bseerv
rvee ma
may be rrelated
elat
el
ated
at
ed to
to complex
comp
co
m lex
stoichiometric interactions at the cell surface between TGFE ligands and various TGFERII
isoforms as suggested by del Re et al.32. The precise mechanism whereby TGFE ligand binding
may induce receptor activation is conflicting. Some models propose that TGFE ligands bind
TGFERII dimers that recruit TGFERI dimers to form a heterotetrameric signaling complex.35
Other models, which propose the existence of inactive preformed complexes of TGFERI and
TGFERII dimers36, 37, are supported by potential cooperative TGFb2 ligand binding to a
TGFERI-TGFERIIb complex in which the receptors make physical contact32. Krishnaveni et al.
suggest that TGFERIIb favors heterodimerization with TGFERII because this interaction is more
robust.38 Overall, these data suggest a complex TGFE signaling process further depending upon
18
DOI: 10.1161/CIRCGENETICS.112.964064
the stoichiometric interactions between TGFE ligands and various receptor isoforms. Further
investigation in vivo of these interactions will add to our understanding of aortic aneurysm
pathogenesis.
Aberrant TGFȕsignaling that result from type I and II receptor mutations has been
implicated in the pathogenesis of cardiovascular disorders involving TAAs. We showed that
TGFȕ2 signaling is decreased in cells expressing TGFȕRIIb and mutations in this receptor result
in increased TGFȕ2 signaling. Identification of TGFȕRIIb activating mutations in two TAA
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
patients supports the hypothesis that an increase in TGFȕ signaling contributes
ntributes to aortic
pathogenesis. Furthermore, this evidence highlights the scientific and clinic
clinical
i all iimport
mport off
expanding diag
alternative
ddiagnostic
ag
gno
nost
stic
st
ic strategies
strat
ateegies to include the altern
at
nat
ative segmentt of TGFȕRIIb
TGFȕ
TG
FȕRIIb in genetic
screening of
individuals
with
Taken
findings
suggest
that
TGFȕRIIb
of in
ndividuals w
ith TAA.
TAA
A. Take
k n together,
to
ogeeth
her,, tthese
hesee fi
findin
ngs sug
ugggeestt tha
at TG
TGF
FȕRI
RIIb
RI
expression is like
likely
important
regulatory
k ly an impo
p rtant regu
g latory mechanism
mechhani
nism off TGFȕ2
TGF
TG
Fȕ2 signaling
signal
i al
aling in
in the aorta, and
ann that
there may be
contributions
TGFE1
TGFE2
signaling
be ddifferential
iffe
if
fere
rent
re
ntia
nt
iall co
cont
ntri
nt
ribu
buti
tion
onss of T
on
GFE
GF
E1 and
and TG
TGF
FE2 si
sign
gnal
gn
alin
ingg to aneurysm
ane
neur
urys
ur
ysm
ys
m
pathogenesis.
Acknowledgments: We are grateful to the family members, cardiologists and cardiothoracic
surgeons at Weill Cornell Medical Center for their participation in this study. We thank Dr.
Harry Ostrer for contributing the Ashkenazi Jewish control samples.
Funding Sources: This work was supported by grants from NIH [R01 HL61785 (C.T.B.,
C.J.H.)], the Snart Cardiovascular Fund [C.J.H.] and Raymond and Beverly Sackler [C.J.H.].
Conflict of Interest Disclosures: None.
References:
1. Coady MA, Davies RR, Roberts M, Goldstein LJ, Rogalski MJ, Rizzo JA, et al. Familial
patterns of thoracic aortic aneurysms. Arch Surg. 1999;134:361-367.
2. Palmieri V, Bella JN, Arnett DK, Roman MJ, Oberman A, Kitzman DW, et al. Aortic root
19
DOI: 10.1161/CIRCGENETICS.112.964064
dilatation at sinuses of valsalva and aortic regurgitation in hypertensive and normotensive
subjects: The Hypertension Genetic Epidemiology Network Study. Hypertension. 2001;37:12291235.
3. Bella JN, MacCluer JW, Roman MJ, Almasy L, North KE, Welty TK, et al. Genetic
influences on aortic root size in American Indians: the Strong Heart Study. Arterioscler Thromb
Vasc Biol. 2002;22:1008-1011.
4. Albornoz G, Coady MA, Roberts M, Davies RR, Tranquilli M, Rizzo JA, et al. Familial
thoracic aortic aneurysms and dissections--incidence, modes of inheritance, and phenotypic
patterns. Ann Thorac Surg. 2006;82:1400-1405.
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
5. Devereux RB, de Simone G, Arnett DK, Best LG, Boerwinkle E, Howard BH, et al. TwoDimensional Echocardiographic Aortic Root Dimensions in Adolescents and Adults: Normal
Limits in Relation to Age, Body Size and Gender. J Am Coll Cardiol. 2010;55:A87.E824.
2010;55:A87.E824
24..
24
6. Devereux RB, Cooper RN, Weder A, Seto TB, Hanis C, Mosley TH,
and
H, et al.
all. Prevalence
Prevallence
cee an
Correlates of Aortic Root Dilatation in Normotensive and Hypertensive
Adults:
The
Family
ve Ad
dul
ults
ts:: T
ts
he F
amil
am
Blood Pressure
Program.
s re P
sure
rogr
ro
gram
am. Circulation.
am
C rculation. 2009;120:S540.
Ci
2009;120:S54
540.
54
0
7. Zhu L, Vranckx
Van
Kien
P,, La
Lalande
Boisset
N,, Ma
Mathieu
Mutations
Vrran
nckx R, Khau
Khhau V
an Ki
ien P
alaandde A, B
oiiss
sset
et N
M
thiieu
ie F, et al
aal.. Mu
Mutati
ions in
myosin heavy
thoracic
avyy chain
cha
h in 111 cause
caause a syndrome
synd
ndro
nd
r me associating
asssocciaatingg tho
hooraciic aortic
ao
ortic aaneurysm/aortic
neuuryysm//ao
a rtticc dissection
di
dissec
issec
and patent ductu
arteriosus.
dductus
us ar
art
te osus
terios
us. Nat
N t Genet.
Na
Gene
neet. 2006;38:343-349.
2006
0 ;3
06
38:
8 3443-34
3 9.
8. Hasham SN,, Willingg MC,
MC
C, Guo
Guo DC,
D , Muilenburg
DC
M il
Mu
ilenbu
b rgg A,
A, He
H R,, Tran
Tran VT,
VT, et al.
all. Mapping
Mapp
ppingg a locus
pp
locu for
familial thoracic
aortic
Circulation.
oracic aort
or
rtic
ic aaneurysms
neeurys
neur
urys
ysms
mss aand
nd ddissections
isse
is
sect
se
ctio
ct
ions
io
ns (TAAD2)
(TA
TAAD
AD2)
AD
2) tto
o 3p24-25.
3p24
3p
24-2
24
-225.
5. Ci
C
irc
rcul
rc
ulat
ul
atio
at
i n.
2003;107:3184-3190.
1844 31
18
3190
90
9. Pannu H, Fadulu VT, Chang J, Lafont A, Hasham SN, Sparks E, et al. Mutations in
transforming growth factor-beta receptor type II cause familial thoracic aortic aneurysms and
dissections. Circulation. 2005;112:513-520.
10. Guo DC, Pannu H, Tran-Fadulu V, Papke CL, Yu RK, Avidan N, et al. Mutations in smooth
muscle alpha-actin (ACTA2) lead to thoracic aortic aneurysms and dissections. Nat Genet.
2007;39:1488-1493.
11. Wang L, Guo DC, Cao J, Gong L, Kamm KE, Regalado E, et al. Mutations in myosin light
chain kinase cause familial aortic dissections. Am J Hum Genet. 2010;87:701-707.
12. van de Laar IM, Oldenburg RA, Pals G, Roos-Hesselink JW, de Graaf BM, Verhagen JM, et
al. Mutations in SMAD3 cause a syndromic form of aortic aneurysms and dissections with earlyonset osteoarthritis. Nat Genet. 2011;43:121-126.
13. Vaughan CJ, Casey M, He J, Veugelers M, Henderson K, Guo D, et al. Identification of a
chromosome 11q23.2-q24 locus for familial aortic aneurysm disease, a genetically
heterogeneous disorder. Circulation. 2001;103:2469-2475.
20
DOI: 10.1161/CIRCGENETICS.112.964064
14. Guo D, Hasham S, Kuang SQ, Vaughan CJ, Boerwinkle E, Chen H, et al. Familial thoracic
aortic aneurysms and dissections: genetic heterogeneity with a major locus mapping to 5q13-14.
Circulation. 2001;103:2461-2468.
15. Massague J. TGF-beta signal transduction. Annu Rev Biochem. 1998;67:753-791.
16. Jones JA, Spinale FG, Ikonomidis JS. Transforming growth factor-beta signaling in thoracic
aortic aneurysm development: a paradox in pathogenesis. J Vasc Res. 2009;46:119-137.
17. Lonn P, Moren A, Raja E, Dahl M, Moustakas A. Regulating the stability of TGFbeta
receptors and Smads. Cell Res. 2009;19:21-35.
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
18. Hirai R, Fijita T. A human transforming growth factor-beta type II receptor that contains an
insertion in the extracellular domain. Exp Cell Res. 1996;223:135-141.
19. Pogulis RJ, Vallejo AN, Pease LR. Recombination and Mutagenesis
Extension
siis by Overlap
Ove
verl
rlap
rl
ap E
xttenss
PCR. In: Rapley R, ed. The Nucleic Acids Protocols Handbook. Totowa,
Press;
wa, NJ
NJ: Humana
Humana P
res
re
2000:857-864.
20. Loeys BL,
Callewaert
Devereux
al. The
BL
L, Dietz
Dietz HC,
HC
H
C, Braverman
B aver
Br
av
ver
erma
m n AC
AC, Ca
C
lllew
e aerrt BL,
L,, De
De Backer
Baack
Back
cker
er JJ,, Deve
vere
ve
reeux
u R
RB,
B, eett al
B,
revised Ghent
Med
ent nosology for
en
forr the
thhe Marfan
Mar
a fan syn
ar
ssyndrome.
yndrrome
me. J M
edd Ge
Genet. 22010;47:476-485.
0100;4
47::476-48
4 5.
48
21. Guo DC,
V,, Reg
Regalado
Mutations
in
C, Papke
Papk
pke
pk
ke CL,
CL, Tran-Fadulu
T an-F
Tr
Fad
a ullu V
gal
alad
lad
ado ES,
ES Av
ES
Avidan
Avid
idan
id
an N
N,, Jo
John
Johnson
h so
hn
son RJ,
RJ et aal.
RJ
l. M
Mutati
uttat
ati
t
smooth muscle
disease,
scle alpha-actinn (ACTA2)
(A
ACT
C A2
A ) cause
caus
ca
usse coronary
co
oro
rona
nary
na
ry aartery
rteery di
rt
dise
seas
se
a e, sstroke,
as
trrok
oke,
e aand
e,
n Moyamoyaa
nd
disease, along
Am
Hum
Genet.
2009;84:617-627.
o g with thoracic
ong
i aortic di
disease. A
mJH
um G
enet. 20
009
09;8
;8
84:61
6 77 62
6 7.
22. Loeys BL,
ER, Judge
DP, Podowski
BL Chen
BL
Ch J,
J Neptune
Nept
Nept ne ER
J ddge
g DP
P
Podo
oddo ski
ki M,
M Holm
Hollm T,
T et al.
all A syndrome
s ndrome
dr
off
altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by
mutations in TGFBR1 or TGFBR2. Nat Genet. 2005;37:275-281.
23. Habashi JP, Doyle JJ, Holm TM, Aziz H, Schoenhoff F, Bedja D, et al. Angiotensin II type 2
receptor signaling attenuates aortic aneurysm in mice through ERK antagonism. Science.
2011;332:361-365.
24. Holm TM, Habashi JP, Doyle JJ, Bedja D, Chen Y, van Erp C, et al. Noncanonical TGFbeta
signaling contributes to aortic aneurysm progression in Marfan syndrome mice. Science.
2011;332:358-361.
25. Gomez D, Al Haj Zen A, Borges LF, Philippe M, Gutierrez PS, Jondeau G, et al. Syndromic
and non-syndromic aneurysms of the human ascending aorta share activation of the Smad2
pathway. J Pathol. 2009;218:131-142.
26. Lin F, Yang X. TGF-beta signaling in aortic aneurysm: another round of controversy. J
Genet Genomics. 2010;37:583-591.
27. Tran-Fadulu V, Pannu H, Kim DH, Vick GW, 3rd, Lonsford CM, Lafont AL, et al. Analysis
of multigenerational families with thoracic aortic aneurysms and dissections due to TGFBR1 or
21
DOI: 10.1161/CIRCGENETICS.112.964064
TGFBR2 mutations. J Med Genet. 2009;46:607-613.
28. Singh KK, Rommel K, Mishra A, Karck M, Haverich A, Schmidtke J, et al. TGFBR1 and
TGFBR2 mutations in patients with features of Marfan syndrome and Loeys-Dietz syndrome.
Hum Mutat. 2006;27:770-777.
29. Stheneur C, Collod-Beroud G, Faivre L, Gouya L, Sultan G, Le Parc JM, et al. Identification
of 23 TGFBR2 and 6 TGFBR1 gene mutations and genotype-phenotype investigations in 457
patients with Marfan syndrome type I and II, Loeys-Dietz syndrome and related disorders. Hum
Mutat. 2008;29:E284-295.
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
30. Akutsu K, Morisaki H, Okajima T, Yoshimuta T, Tsutsumi Y, Takeshita S, et al. Genetic
analysis of young adult patients with aortic disease not fulfilling the diagnostic criteria for
Marfan syndrome. Circ J. 2010;74:990-997.
31. Mizuguchi T, Collod-Beroud G, Akiyama T, Abifadel M, Harada N,
Morisaki
N M
oris
or
isak
is
akii T,
ak
T eett al
al.
Heterozygous TGFBR2 mutations in Marfan syndrome. Nat Genet. 2004;36:855-860.
004;3
36:
6 85
8555 86
8600.
32. del Re E, Ba
receptor, the
Babi
Babitt
biitt JL,
JL,
L Pirani
Pir
irani A, Schneyer AL, Linn HY.
H . In the absence
HY
absen
nce of type III receptor
transforming
II-B
receptor
requires
receptor
bind
ng growth
ng
growth factor
facttor ((TGF)-beta
fa
TGF)
TG
F))-be
beeta ttype
yp
pe II
I-B
B rec
ceptorr re
requ
q ir
ires
es tthe
he ttype
ypee I re
ece
c ptor
pttor
or tto
o bin
n
TGF-beta2.. J B
Biol
iol Chem. 22004;279:22765-22772.
004;;2799:2
227765--222772
72.
33. Rotzer D,
M,, Lu
Lutz
M,, Li
Lindemann
D,, S
Sebald
D Roth
Roth M
Ro
L
tz M
tz
L
nddem
emann
m
D
ebal
eb
bald
ld W,
W Knaus
Kna
nauus P.
P. Type
T pe III
Ty
II TGF-beta
TGF-be
b ta
be
ta receptorrec
ece
independent
alternatively
n signalling off TGF-beta2
nt
T FTG
F-be
beta
be
t 2 via
ta
via TbetaRII-B,
Tbet
Tb
etaR
et
aRII
aR
III-B,, an
an alte
tern
te
rnnat
ativ
iv
vel
elyy sp
sspliced
lice
li
cedd TGF-beta type
ce
typp II
receptor. Embo
2001;20:480-490.
m J. 2001;2
mbo
; 0:48
4800-49
48
4990.
34. Lopez-Casillas
JL, Massag
Massague
Betaglycan
Casillas
C
Ca
sill
ill F,
F Wrana
Wr
JL
M
a
e JJ. Betagl
B
etagll can presents
nt ligand
li ndd to
t the
th
h TGF
TGF beta
beta
signaling receptor. Cell. 1993;73:1435-1444.
35. ten Dijke P, Yamashita H, Ichijo H, Franzen P, Laiho M, Miyazono K, et al. Characterization
of type I receptors for transforming growth factor-beta and activin. Science. 1994;264:101-104.
36. Chen RH, Moses HL, Maruoka EM, Derynck R, Kawabata M. Phosphorylation-dependent
interaction of the cytoplasmic domains of the type I and type II transforming growth factor-beta
receptors. J Biol Chem. 1995;270:12235-12241.
37. Zhu HJ, Sizeland AM. Extracellular domain of the transforming growth factor-beta receptor
negatively regulates ligand-independent receptor activation. J Biol Chem. 1999;274:2922029227.
38. Krishnaveni MS, Hansen JL, Seeger W, Morty RE, Sheikh SP, Eickelberg O. Constitutive
homo- and hetero-oligomerization of TbetaRII-B, an alternatively spliced variant of the mouse
TGF-beta type II receptor. Biochem Biophys Res Commun. 2006;351:651-657.
22
DOI: 10.1161/CIRCGENETICS.112.964064
Table 1. Description of Cohort
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Gender (n)
Male
Female
77
23
Ethnicity (n)
Caucasian
Caucasian/Hispanic
Caucasian/Native American
African American
African American/Hispanic
Asian
Other
80
6
2
2
2
2
6
Age at Enrollment (years)
Mean
Median
Range
53
52
21-93
Body Surface
ace
ac
ce Area (BSA
(BSA,
A, m2)
Mean (n=65)
n=6
n=6
65)
Median
1.9
1.99
99
22.00
2.
00
Aortic Surgery
gery (n)
4
42
Dissection (n)
(n)
27
Inclusion Criteria (n)
Diagnoses <50 years old
Connective Tissue Abnormality
Family History
64
14
67
Figure Legends:
Figure 1. Mutational analysis of ACTA2 in families ANS, JNW and SY92. A. Automated
sequence analyses of ACTA2 from affected individuals in families ANS, JNW and SY92 are
shown. A 1-bp deletion in families ANS, JNW and SY92 (arrow) generates a missense mutation
indicated by the amino acid code. B. Pedigrees of families ANS, JNW and SY92 with an ACTA2
23
DOI: 10.1161/CIRCGENETICS.112.964064
mutation indicate that mutations co-segregate with disease. Mutations are present only in family
members affected with TAA. Arrow indicates family proband.
Figure 2. Mutational analysis of MYH11 in families JNE, ANHH and ANO. A. Automated
sequence analyses of MYH11 from affected individuals in families JNE, ANHH and ANO are
shown. A 1-bp deletion in families JNE and ANHH (arrow) generates a missense mutation
indicated by the amino acid code. A 7-bp intronic substitution in family ANO (arrow) produces
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
a splice site alteration located at the nucleotide position relative to the preceding exon. B.
Pedigrees of families JNE, ANHH and ANO are shown and indicate that
MYH11
hat M
YH11
YH
11 mutations
muttati
tioons coti
segregate with
w h ddisease.
issea
ease
se.. Mu
se
Mutations are present only iin
n family memberss aaffected
ffected with TAA.
Arrow indicates
proband.
catees family pr
ca
roban
nd.
Figure 3. TGFbRII
and TG
TGFbIIb
mutations
T
GFbIIIb mutati
ions in
in families
famil
i ie
il
i s KNA,
KNA,
A, ANV
ANV
V and
d KNK.
KNK
N . A. Automated
Autom
m
sequence analyses
KNA,
ANV
nall ses off T
TGFbRII
GFbR
GF
bRII
II and
ndd TGFbRIIb
TGF
GFbbRI
RIIb
Ib from
f
aff
affected
ffected
d indi
iindividuals
ndi
di id
d als
ls iin families
famili
ili KNA
KNA A
and KNK are shown. A 1-bp deletion in families KNA, ANV and KNK (arrow) generates a
missense mutation indicated by the amino acid code. B. Pedigree of family KNA with a
TGFERII mutation and families ANV and KNK with TGFbRIIb mutations indicate that
mutations co-segregate with disease. Mutations are present only in family members affected with
TAA. Arrow indicates family proband.
Figure 4. mRNA expression of TGFERII isoforms in aortic tissue and lymphocytes. RT-PCR
performed using various pairs of TGFbRII and TGFbRIIb isoform specific primers to amplify
24
DOI: 10.1161/CIRCGENETICS.112.964064
exons 1, 1a, 2 and 3 (a), exons 1, 2 and 3 (b) and exons 1a, 2 and 3 (c) from isolated human
aortic tissue (lanes 1-2) and lymphocyte (lanes 3-4) RNA.
Figure 5. TGFE1 and TGFE2 signaling in dermal fibroblasts. A-B. Cultured fibroblasts were
isolated from normal patient with wt-TGFbRII and ANV I-1 patient with H56N-TGFbRIIb
mutation. Representative Western blot analyses shown for pSMAD2 protein expression in wtTGFbRII or H56N-TGFbRIIb fibroblasts stimulated with either TGFb1 (A) or TGFb2 (B) for
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
indicated time points. Corresponding b-actin blots shown. (A) H56N-TGFbRIIb
-TGFbRIIb dermal
dermaal
fibroblasts exhibit delayed TGFE1 signaling compared to normal dermal
fibroblasts.
H56Nmal fibr
b oblasts.. (B)
(B) H
TGFbRIIb der
ddermal
e ma
er
m l fi
ffibroblasts
brroblaasts exhibit decreased TG
TGFE2
GFE2 signaling
g comp
compared
mp
pared to normal dermal
derm
m
fibroblasts.
Figure 6. ,QYLWUR7*)ȕ-VWLPXODWLRQGRHVQRWDOWHU7*)ȕ5,,DQG7*)ȕ5,,EDFWLYLW\.
Q YLW
LWUR
UR 7*)
*)ȕ
ȕ-VW
ȕ
VWLP
VW
LPXO
LP
XODW
XO
DWLR
DW
LRQQGR
LR
GRHV
GR
HV QRW DOW
OWHU
HU 7*)
*)ȕ5
ȕ ,, DQG 7*)
ȕ5
*)ȕ5
ȕ5,,
ȕ5
,,EEDF
,,
DFWL
DF
WLYL
WL
YLW\
YL
W\. A.
W\
A.
Relative quantification of pSmad2 expression after in vitro 7*)ȕVWLPXOation. Bar graph
displays densitometric quantification of pSmad2 protein expression relative to b-actin in
untransfected control, wt-7*)ȕ5,,DQGZW-7*)ȕ5,,E/FHOOVQ 1RVLJQLILFDQWFKDQJHLQ
7*)ȕ5,,RU7*)ȕ5,,E activity compared to control cells. B. Relative quantification of pSmad2
expression after in vitro 7*)ȕVWLPXODWLRQ%DUJUDSKGLVSOD\VGHQVLWRPHWULFTXDQWLILFDWLRQRI
pSmad2 relative to b-actin in control wt-7*)ȕ5,,E+1-7*)ȕ5,,EDQG'1-7*)ȕ5,,E/
cells (n=3). No significant change in aFWLYLW\LQPXWDQW7*)ȕ5,,EFHOOVFRPSDUHGWRZW7*)ȕ5,,EFHOOVLQresponse to 0.5 hour S07*)ȕVWLPXODWLRQ. Values normalized to control
L6 cells. All data presented as mean+SEM. P values shown. NS= not significant.
25
DOI: 10.1161/CIRCGENETICS.112.964064
Figure 7. In vitro 7*)ȕ-VWLPXODWLRQDOWHUV7*)ȕ5,,DQG7*)ȕ5,,EDFWLYLW\ A. Relative
quantification of pSmad2 expression after in vitro 7*)ȕVWLPXODWLRQ%DUJUDSKGLVSOD\V
densitometric quantification of pSmad2 protein expression relative to b-actin in untransfected
control, wt-7*)ȕ5,,DQGZW-7*)ȕ5,,E/FHOOVQ wt-7*)ȕ5,,EFHOOVH[KLELWGHFUHDVHG
7*)ȕVLJQDOLQJFRPSDUHGWRZW-7*)ȕ5,,FHOOV B. Relative quantification of pSmad2
expression after in vitro 7*)ȕVWLPXODWLRQ%DUJUDSKGLVSOD\VGHQVLWRPHWULFTXDQWLILFDWLRQRI
pSmad2 relative to b-actin in control wt-7*)ȕ5,,E+1-7*)ȕ5,,EDQG'1-7*)ȕ5,,E/
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
cells (n=3). Mutant 7*)ȕ5,,E cells exhibit increased response after 1 hour S07*)ȕ
S07*)ȕ
)ȕ
)ȕ
stimulation compared to wt-7*)ȕ5,,EFHOOV Values normalized to control
L6
cells.
ntroll L
6 cell
lls. All
ll
Alll data
dat
presented aass m
mean+SEM.
eann+SEM
ea
SE
EM. P values shown.
26
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
TGFβRIIb Mutations Trigger Aortic Aneurysm Pathogenesis by Altering TGFβ2 Signal
Transduction
Katharine J. Bee, David C. Wilkes, Richard B. Devereux, Craig T. Basson and Cathy J. Hatcher
Downloaded from http://circgenetics.ahajournals.org/ by guest on November 17, 2016
Circ Cardiovasc Genet. published online October 24, 2012;
Circulation: Cardiovascular Genetics is published by the American Heart Association, 7272 Greenville Avenue, Dallas,
TX 75231
Copyright © 2012 American Heart Association, Inc. All rights reserved.
Print ISSN: 1942-325X. Online ISSN: 1942-3268
The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circgenetics.ahajournals.org/content/early/2012/10/24/CIRCGENETICS.112.964064
Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in
Circulation: Cardiovascular Genetics can be obtained via RightsLink, a service of the Copyright Clearance Center,
not the Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information about
this process is available in the Permissions and Rights Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Circulation: Cardiovascular Genetics is online at:
http://circgenetics.ahajournals.org//subscriptions/