25.07.2013 Views

Syndromic and non-syndromic aneurysms of the ... - INSERM U698

Syndromic and non-syndromic aneurysms of the ... - INSERM U698

Syndromic and non-syndromic aneurysms of the ... - INSERM U698

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Journal <strong>of</strong> Pathology<br />

J Pathol 2009; 218: 131–142<br />

Published online 6 January 2009 in Wiley InterScience<br />

(www.interscience.wiley.com) DOI: 10.1002/path.2516<br />

Original Paper<br />

<strong>Syndromic</strong> <strong>and</strong> <strong>non</strong>-<strong>syndromic</strong> <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> human<br />

ascending aorta share activation <strong>of</strong> <strong>the</strong> Smad2 pathway<br />

Delphine Gomez, 1† Ayman Al Haj Zen, 1† Luciano F Borges, 2 Monique Philippe, 1 Paulo Sampaio Gutierrez, 2<br />

Guillaume Jondeau, 1,3 Jean-Baptiste Michel1 <strong>and</strong> Roger Vranckx1 *<br />

1<strong>INSERM</strong>, <strong>U698</strong>, Paris, F-75018 France; Univ Paris 7-Denis Diderot, Paris, F-75018, France<br />

2Laboratory <strong>of</strong> Pathology, Instituto do Coraçào, University <strong>of</strong> Sao Paulo. School <strong>of</strong> Medicine, Brazil<br />

3AP-HP, Hôpital Xavier Bichat, Department <strong>of</strong> Cardiovascular Medicine <strong>and</strong> Surgery <strong>and</strong> <strong>the</strong> Reference Center for Marfan <strong>and</strong> Related Syndromes,<br />

Paris, France<br />

*Correspondence to:<br />

Roger Vranckx, <strong>INSERM</strong>, <strong>U698</strong>,<br />

Hôpital Xavier Bichat (Secteur<br />

Claude Bernard), 46 Rue Henri<br />

Huchard, FR-75877 Paris Cedex<br />

18, France.<br />

E-mail: roger.vranckx@inserm.fr<br />

† These authors contributed<br />

equally to this work.<br />

No conflicts <strong>of</strong> interest were<br />

declared.<br />

Received: 20 May 2008<br />

Revised: 16 December 2008<br />

Accepted: 20 December 2008<br />

Introduction<br />

Abstract<br />

Common features such as elastic fibre destruction, mucoid accumulation, <strong>and</strong> smooth muscle<br />

cell apoptosis are co-localized in <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> ascending aorta <strong>of</strong> various aetiologies.<br />

Recent experimental studies reported an activation <strong>of</strong> TGF-β in <strong>aneurysms</strong> related to<br />

Marfan (<strong>and</strong> Loeys-Dietz) syndrome. Here we investigate TGF-β signalling in normal <strong>and</strong><br />

pathological human ascending aortic wall in <strong>syndromic</strong> <strong>and</strong> <strong>non</strong>-<strong>syndromic</strong> aneurysmal<br />

disease. Aneurysmal ascending aortic specimens, classified according to aetiology: <strong>syndromic</strong><br />

MFS (n = 15, including two mutations in TGFBR2 ), associated with BAV (n = 15) or<br />

degenerative forms (n = 19), were examined. We show that <strong>the</strong> amounts <strong>of</strong> TGF-β1 protein<br />

retained within <strong>and</strong> released by aneurysmal tissue were greater than for control aortic<br />

tissue, whatever <strong>the</strong> aetiology, contrasting with an unchanged TGF-β1 mRNA level. The<br />

increase in stored TGF-β1 was associated with enhanced LTBP-1 protein <strong>and</strong> mRNA levels.<br />

These dysregulations <strong>of</strong> <strong>the</strong> extracellular lig<strong>and</strong> are associated with higher phosphorylated<br />

Smad2 <strong>and</strong> Smad2 mRNA levels in <strong>the</strong> ascending aortic wall from all types <strong>of</strong> aneurysm.<br />

This activation correlated with <strong>the</strong> degree <strong>of</strong> elastic fibre fragmentation. Surprisingly, <strong>the</strong>re<br />

was no consistent association between <strong>the</strong> nuclear location <strong>of</strong> pSmad2 <strong>and</strong> extracellular<br />

TGF-β1 <strong>and</strong> LTBP-1 staining <strong>and</strong> between <strong>the</strong>ir respective mRNA expressions. In parallel,<br />

decorin was focally increased in aneurysmal media, whereas biglycan was globally decreased<br />

in aneurysmal aortas. In conclusion, this study highlights independent dysregulations <strong>of</strong><br />

TGF-β retention <strong>and</strong> Smad2 signalling in <strong>syndromic</strong> <strong>and</strong> <strong>non</strong>-<strong>syndromic</strong> <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong><br />

ascending aorta.<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John<br />

Wiley & Sons, Ltd.<br />

Keywords: Marfan syndrome; bicuspid aortic valves; TGF-β; LTBP-1; proteoglycans;<br />

PAI-1; decorin<br />

In contrast with abdominal aortic aneurysm, mainly<br />

due to a<strong>the</strong>rosclerosis, aetiologies <strong>of</strong> aneurysm <strong>of</strong><br />

<strong>the</strong> ascending aorta are less well characterized <strong>and</strong><br />

appear to be multiple [1]. However, whatever <strong>the</strong><br />

aetiology [2] — genetic, <strong>syndromic</strong> forms associated<br />

with mutations in FBN1, TGFBR1 or TGFBR2 [3–5]<br />

(mainly Marfan syndrome [6]), MYH11 [7], ACTA2<br />

[8], GLUT10 [9]; <strong>aneurysms</strong> associated with <strong>the</strong> bicuspid<br />

aortic valve (BAV) [10]; or degenerative forms<br />

[11] — <strong>the</strong>y all present common histopathological<br />

features such as elastolysis <strong>and</strong> collagenolysis, accumulation<br />

<strong>of</strong> mucoid material [12,13], <strong>and</strong> smooth<br />

muscle cell (SMC) apoptosis [14–16]. In this context,<br />

it has been recently proposed that transforming<br />

growth factor-beta (TGF-β) plays a predominant<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

www.pathsoc.org.uk<br />

pathophysiological role in Marfan mutation-induced<br />

<strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> ascending aorta [6].<br />

(TGF-β, a multifunctional cytokine, plays a key<br />

role in embryo morphogenesis, differentiation, apoptosis,<br />

<strong>and</strong> extracellular matrix integrity [17–19].<br />

These effects result from <strong>the</strong> binding <strong>of</strong> TGF-β1<br />

to transmembrane receptors with a cytoplasmic serine/threonine<br />

kinase domain. TGF-β1 <strong>and</strong> TGF-β<br />

receptor types I <strong>and</strong> II activate several pathways,<br />

including Smad-dependent <strong>and</strong> Smad-independent<br />

pathways [20]. Smads are a family <strong>of</strong> transcriptional<br />

factors critical for transmitting immediate signals from<br />

TGF-β receptors to <strong>the</strong> nucleus, where <strong>the</strong>y regulate<br />

<strong>the</strong> expression <strong>of</strong> multiple targets [21]: matrix proteins<br />

(collagen, fibronectin [22]), matrix metalloproteinases<br />

(MMP-2 [23]), <strong>and</strong> members <strong>of</strong> <strong>the</strong> fibrinolytic system<br />

[plasminogen activator inhibitor-1 (PAI-1) [24]].


132 DGomezet al<br />

In addition to regulating extracellular matrix component<br />

syn<strong>the</strong>sis, TGF-β1 interplays with various extracellular<br />

matrix molecules (LTBP-1, decorin) that regulate<br />

its activity. TGF-β1 is stored in its latent form<br />

<strong>and</strong> is associated with LTBP-1 (latent TGF-β1 binding<br />

protein 1) [25]. LTBP-1 also possesses domains<br />

that interact with matrix macromolecules (fibrillin-1,<br />

fibronectin) [26]. Thus, <strong>the</strong> associations between TGFβ1<br />

<strong>and</strong> LTBP-1 <strong>and</strong> between LTBP-1 <strong>and</strong> matrix proteins<br />

determine <strong>the</strong> sequestration/release <strong>of</strong> TGF-β1<br />

within <strong>the</strong> extracellular matrix, which is an important<br />

mechanism that controls TGF-β signalling. A previous<br />

study has suggested that <strong>the</strong> genetic deficiency<br />

<strong>of</strong> fibrillin-1 in Marfan syndrome (MFS), <strong>and</strong> <strong>the</strong><br />

resulting extracellular matrix <strong>and</strong> micr<strong>of</strong>ibril degradation<br />

observed in <strong>the</strong> ascending aortic aneurysm, could<br />

induce anomalous latent TGF-β1 sequestration <strong>and</strong><br />

cause excessive activation <strong>of</strong> TGF-β1 in <strong>the</strong> vascular<br />

wall [27].<br />

A recent report in a mouse model <strong>of</strong> MFS related<br />

to an FBN1 mutation has suggested that TGF-β signalling<br />

is a molecular target in aortic dilatation [28,29].<br />

An enhancement <strong>of</strong> <strong>the</strong> TGF-β signalling pathway<br />

was detected in <strong>the</strong> aortic wall <strong>of</strong> patients presenting<br />

mutations in TGFBR1 <strong>and</strong> TGFBR2 [4,5,13].<br />

However, <strong>the</strong>se mutations appear to alter <strong>the</strong> transmission<br />

<strong>of</strong> <strong>the</strong> subcellular TGF-β signal [3,5]. There<br />

have been no reports on human aortic wall derived<br />

from <strong>aneurysms</strong> <strong>of</strong> o<strong>the</strong>r aetiologies, presumably not<br />

directly related to TGF-β signalling (eg BAV <strong>and</strong><br />

degenerative aneurysm).<br />

Therefore, <strong>the</strong> purpose <strong>of</strong> <strong>the</strong> present study was to<br />

clarify fur<strong>the</strong>r <strong>the</strong> relationship between extracellular<br />

TGF-β <strong>and</strong> <strong>the</strong> Smad2 signalling pathway in situ in<br />

<strong>the</strong> wall <strong>of</strong> ascending aortic <strong>aneurysms</strong> <strong>of</strong> various<br />

aetiologies.<br />

Materials <strong>and</strong> methods<br />

Patients <strong>and</strong> aortic specimens<br />

The clinical research protocol was approved by <strong>the</strong><br />

local ethical committee (CCP 05.04.32, 10.23.2007,<br />

Ambroise Paré, Boulogne, France) <strong>and</strong> all patients<br />

signed informed consent. Aneurysmal ascending aortic<br />

specimens were collected during aortic surgery<br />

(Hôpital Bichat). Forty-six specimens were divided<br />

into three groups according to <strong>the</strong>ir clinical features<br />

<strong>and</strong> genetic background: MFS (n = 15, mean age<br />

39 ± 15 years, including two TGFBR2 mutations);<br />

BAV (n = 15, mean age 59 ± 13 years); <strong>and</strong> degenerative<br />

(n = 19, mean age 68 ± 20 years). Clinical<br />

<strong>and</strong> biological data associated with this series have<br />

been recently reported [2]. All specimens were from<br />

<strong>aneurysms</strong> <strong>of</strong> more than 5 cm in diameter. Normal<br />

thoracic aortas were obtained from normal organ transplant<br />

donors (n = 10), with <strong>the</strong> authorization <strong>of</strong> <strong>the</strong><br />

French Biomedicine Agency. Aneurysmal tissues were<br />

sampled in <strong>the</strong> outer curvature, <strong>the</strong> most dilated part<br />

<strong>of</strong> <strong>the</strong> ascending aorta.<br />

Histopathological evaluation<br />

The aortic specimens were fixed in 4% (v/v) buffered<br />

paraformaldehyde for 48 h at room temperature,<br />

embedded in paraffin, <strong>and</strong> serial sections (5 µm thickness)<br />

were obtained from each specimen. OCTembedded<br />

frozen specimens were also conserved at<br />

−20 ◦ C. The sections were stained with Movat’s pentachrome<br />

for general evaluation <strong>of</strong> tissue morphology,<br />

orcein for visualization <strong>of</strong> elastic fibres, or Alcian blue<br />

for proteoglycans. The aortic elastic fibre network was<br />

assessed by two blinded observers, at three different<br />

sites <strong>of</strong> <strong>the</strong> medial layer <strong>of</strong> <strong>the</strong> ascending aortic wall<br />

at magnification ×200, using a scale ranging from 0<br />

(indicating no fragmentation in <strong>the</strong> elastic fibres) to 5<br />

(indicating total disappearance <strong>of</strong> elastic fibres or wide<br />

diffuse disruption) for each site [30] <strong>and</strong> a mean score<br />

was calculated for each patient.<br />

Immunohistochemistry<br />

TGF-β <strong>and</strong> pSmad2<br />

The sections were treated for antigen retrieval by<br />

heating with citrate buffer, pH 6. Endogenous peroxidase<br />

activity was quenched with 3% (v/v) hydrogen<br />

peroxide. Non-specific binding was blocked by<br />

incubation with normal horse serum. Slides were<br />

incubated overnight at 4 ◦ C with anti-TGF-β polyclonal<br />

(5 µg/ml; Abcam), anti-pSmad2 polyclonal<br />

(0.5 µg/ml; Chemicon), anti-TGF-β2 <strong>and</strong> anti-TGF-β3<br />

(5 <strong>and</strong> 2 µg/ml, respectively; Abcam) or anti-activin-A<br />

(Serotec). Since LTBP-1 was not detectable on fixed<br />

sections, LTBP-1 immunohistochemistry (5 µg/ml;<br />

R&D Systems) was performed on frozen sections.<br />

Decorin <strong>and</strong> biglycan<br />

For anti-decorin <strong>and</strong> anti-biglycan antibodies (both<br />

from R&D Systems), a pretreatment step with chondroitinase<br />

ABC was performed <strong>and</strong> <strong>the</strong>n immunochemistry<br />

was done as described above. The dilution<br />

<strong>of</strong> <strong>the</strong> antibody was 5 µg/ml.<br />

Smooth muscle cell phenotype<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

On serial sections, we performed immunostaining <strong>of</strong><br />

three differentiation markers <strong>of</strong> SMCs, which are<br />

widely used to determine <strong>the</strong>ir phenotype. Antibodies<br />

against human SM α-actin (0.1 µg/ml; Dako),<br />

human SMC myosin heavy chains SM2 (1 µg/ml;<br />

Abcam), <strong>and</strong> <strong>non</strong>-muscle embryonic myosin (MHC<br />

IIB) (2 µg/ml; Abcam) were used for this purpose.<br />

The labelling <strong>of</strong> primary antibodies was achieved<br />

by using a biotinylated link antibody (Vectastain<br />

ABC complex) <strong>and</strong> staining was visualized using <strong>the</strong><br />

DAB substrate chromogen system (DakoCytomation).<br />

Sections were counterstained with Mayer’s haematoxylin<br />

(Sigma). The specificity <strong>of</strong> immunolabelling<br />

was tested by omitting <strong>the</strong> primary antibody <strong>and</strong> by<br />

using unspecific IgG.


Smad2 activation in ascending aortic aneurysm 133<br />

Morphometry<br />

TGF-β1<br />

The per cent area positive for TGF-β1 immunostaining<br />

in <strong>the</strong> medial layer was quantified using <strong>the</strong> colour<br />

detecting mode <strong>of</strong> an image analysis system (Quantimet<br />

500, Leica), taking all aneurysm cases, irrespective<br />

<strong>of</strong> aetiology, as one group <strong>and</strong> controls as <strong>the</strong><br />

o<strong>the</strong>r. Most fields were composed exclusively <strong>of</strong> <strong>the</strong><br />

media <strong>and</strong> only <strong>the</strong>se were considered for <strong>the</strong> measurements.<br />

Thus, <strong>the</strong> area <strong>of</strong> TGF-β1 staining directly<br />

reflects its proportion relative to <strong>the</strong> total medial area<br />

studied <strong>and</strong> <strong>the</strong> results are expressed as such.<br />

pSmad2<br />

The number <strong>of</strong> cells with nuclear staining for pSmad2<br />

within medial aortic tissue sections was obtained by<br />

counting three fields per section. Data are expressed<br />

as <strong>the</strong> percentage <strong>of</strong> total nuclei which were p-Smad2positive.<br />

Real-time quantitative PCR<br />

The medial layers <strong>of</strong> aneurysmal <strong>and</strong> <strong>non</strong>-aneurysmal<br />

ascending aortic specimens were isolated by adventicectomy.<br />

Pieces <strong>of</strong> media were incubated in 0.1%<br />

collagenase <strong>and</strong> elastase solution. Cells were collected<br />

after filtration <strong>and</strong> total RNA was extracted from control<br />

<strong>and</strong> aneurysmal samples by using <strong>the</strong> RNeasy kit<br />

(QIAGEN), according to <strong>the</strong> manufacturer’s instructions.<br />

Real-time PCR was performed in <strong>the</strong> Light-<br />

Cycler system with SYBR Green detection (Roche<br />

Applied Science). mRNA levels were normalized to<br />

GAPDH mRNA.<br />

Immunoblotting<br />

Proteins were extracted from medial tissues by homogenization<br />

in a hypotonic lysis buffer [50 mM Tris<br />

(pH 8), 150 mM NaCl, 1% Triton X-100, 1% sodium<br />

deoxycholate, 5 mM EDTA] <strong>and</strong> containing protease<br />

inhibitors (Sigma). Extracts were separated<br />

in <strong>non</strong>-reducing conditions by 10% sodium dodecyl<br />

sulphate-polyacrylamide gel electrophoresis (SDS-<br />

PAGE). The membrane was blocked with 5% milk<br />

powder in TBS-T [Tris-buffered saline (pH 7.4)–0.1%<br />

Tween 20] <strong>and</strong> was <strong>the</strong>n incubated overnight (4 ◦ C)<br />

with primary antibodies: TGF-β1 (10 ng/ml; Santa<br />

Cruz), LTBP-1 (1 µg/ml; Santa Cruz) or GAPDH<br />

(1.6 µg/ml; Biovalley). The membrane was <strong>the</strong>n incubated<br />

with peroxidase-conjugated anti-mouse or antigoat<br />

IgG (Jackson Laboratories) for 1 h. The signal<br />

was detected by using a chemiluminescence kit<br />

(ECL+ kit; Amersham).<br />

ELISA<br />

Small pieces <strong>of</strong> aortic media were incubated for 24 h<br />

in RPMI culture medium containing 1% L-glutamine<br />

<strong>and</strong> 1% penicillin, streptomycin, <strong>and</strong> amphotericin<br />

at 37 ◦ C (5% CO2), to study extracellular mediators/markers<br />

released by arterial tissue as previously<br />

described in an experimental model [31] <strong>and</strong><br />

for human aneurysmal tissue <strong>of</strong> <strong>the</strong> ascending aorta<br />

[2,12]. The volume <strong>of</strong> culture medium was adjusted<br />

to <strong>the</strong> sample wet weight (6 ml per gram). Tissueculture<br />

media were collected <strong>and</strong> centrifuged (3000 g,<br />

15 min, 20 ◦ C). Concentrations <strong>of</strong> TGF-β1 <strong>and</strong> PAI-<br />

1 were determined in <strong>the</strong> tissue-culture media <strong>and</strong><br />

aortic medial extracts using ELISA kits (R&D systems,<br />

HYPHEN BioMed, respectively) following <strong>the</strong><br />

manufacturers’ instructions. TGF-β1 essays were performed,<br />

in parallel, on acidified (HCl, 1.5 N) <strong>and</strong> <strong>non</strong>acidified<br />

samples.<br />

Statistical analysis<br />

Since <strong>the</strong> number <strong>of</strong> samples in each group was<br />

limited, <strong>non</strong>-parametric statistical tests <strong>and</strong> expression<br />

were chosen. The significance <strong>of</strong> differences between<br />

groups was tested using <strong>the</strong> Mann–Whitney test <strong>and</strong><br />

results are expressed as box plots, in which <strong>the</strong> median<br />

is shown. Upper <strong>and</strong> lower limits <strong>of</strong> boxes represent<br />

inter-quartiles (25th <strong>and</strong> 75th), whereas upper <strong>and</strong><br />

lower bars show percentiles (tenth <strong>and</strong> 90th). The<br />

results <strong>of</strong> mRNA quantifications are expressed as<br />

means ± SD. A value <strong>of</strong> p ≤ 0.05 was considered<br />

significant<br />

Results<br />

TGF-β1 storage in ascending aortic aneurysm<br />

TGF-β1 immunostaining was mainly localized in <strong>the</strong><br />

adventitia, where its distribution was similar in control<br />

<strong>and</strong> aneurysmal aortas (Figure 1A). In all groups <strong>of</strong><br />

<strong>aneurysms</strong>, increased TGF-β1 staining was observed<br />

in <strong>the</strong> media. This staining showed a gradient across<br />

<strong>the</strong> media, being more intense in <strong>the</strong> outer part.<br />

This increase in staining in aneurysmal aortas was<br />

confirmed by morphometric analysis showing higher<br />

TGF-β1 expression in <strong>the</strong> aneurysmal medial layer<br />

than in controls (8.7% ± 0.02 versus 1.8% ± 0.02,<br />

respectively; p < 0.0001). There was no evidence<br />

<strong>of</strong> differences between aetiologies (Figure 1B, a–f).<br />

The increase in staining in <strong>the</strong> aneurysmal aorta was<br />

markedly decreased by incubation <strong>of</strong> <strong>the</strong> anti-TGF-β<br />

antibody with human recombinant TGF-β1, proving<br />

<strong>the</strong> specificity <strong>of</strong> TGF-β1 staining (Figure 1B, g, h).<br />

Immunostaining for <strong>the</strong> is<strong>of</strong>orms <strong>of</strong> TGF-β (TGF-β2<br />

<strong>and</strong> -β3) <strong>and</strong> for activin-A (which can lead to a similar<br />

activation <strong>of</strong> <strong>the</strong> Smad signalling pathway [32])<br />

was performed. TGF-β2, TGF-β3, <strong>and</strong> activin-A were<br />

weakly expressed in <strong>the</strong> media <strong>of</strong> both aneurysmal <strong>and</strong><br />

control aortas. However, this background expression<br />

was homogeneous throughout <strong>the</strong> media <strong>and</strong> no gradient<br />

was observed. No difference was found between<br />

aneurysmal aortic walls <strong>and</strong> controls for <strong>the</strong>se factors<br />

(Figure 1C).<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.


134 DGomezet al<br />

Figure 1. Expression <strong>of</strong> TGF-β1 in ascending aortic aneurysm. (A) Representative photomicrographs <strong>of</strong> TGF-β1 immunostaining<br />

in sections <strong>of</strong> control <strong>and</strong> aneurysmal ascending aorta. TGF-β1 staining is observed in <strong>the</strong> outer media <strong>and</strong> adventitia on <strong>the</strong><br />

right (original magnification × 40). The TGF-β1 expression, which is greater in aneurysmal aorta, is extracellular <strong>and</strong> is associated<br />

with matrix fibres. (Inset: original magnification × 1000.) (B) Representative photomicrographs <strong>of</strong> TGF-β1 immunostaining in <strong>the</strong><br />

media <strong>of</strong> ascending aortic <strong>aneurysms</strong> <strong>of</strong> different aetiologies (haematoxylin counterstaining): (a) <strong>non</strong>-aneurysmal aorta; (b) negative<br />

control; (c) MFS related to FBN1 mutations; (d) MFS related to TGFBR2 mutations; (e) BAV; (f) degenerative aneurysm. TGF-β1<br />

staining is more intense in aneurysmal media (original magnification × 200). The increase in TGF-β staining in degenerative<br />

aneurysmal media obtained with an anti-TGF-β antibody (5 µg/ml) (g) was reversed when <strong>the</strong> antibody was incubated with<br />

recombinant TGF-β1 (150 ng/ml) (h). (C) Immunostaining for TGF-β2, TGF-β3, <strong>and</strong> activin-A is uniformly weak <strong>and</strong> similar in<br />

aneurysmal ascending aorta <strong>and</strong> control (original magnification × 40)<br />

Quantification <strong>of</strong> TGF-β1 mRNA levels was performed<br />

in extracts <strong>of</strong> aortic medial tissue. No significant<br />

difference was found in <strong>the</strong> TGF-β1 mRNA<br />

level in aneurysmal media compared with control<br />

(p < 0.01) (Figure 2A). Paradoxically, immunoblotting<br />

<strong>of</strong> TGF-β1 indicated an increase in <strong>the</strong> TGF-β1<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

protein level in aneurysmal extracts (Figure 2B). A 25<br />

kD b<strong>and</strong> corresponding to <strong>the</strong> active form <strong>of</strong> TGF-β1<br />

was detected but its level was similar in aneurysmal<br />

<strong>and</strong> control extracts. In contrast, a 250 kD b<strong>and</strong>, corresponding<br />

to latent TGF-β1, probably stored within <strong>the</strong><br />

large latency complex (LTBP-1/latent TGF-β1) was


Smad2 activation in ascending aortic aneurysm 135<br />

Figure 2. Expression, storage, <strong>and</strong> activation <strong>of</strong> TGF-β1 in <strong>the</strong> media <strong>of</strong> ascending aortic aneurysm. (A) Quantification by real-time<br />

PCR <strong>of</strong> TGF-β1 mRNA levels in medial extracts from aneurysmal <strong>and</strong> <strong>non</strong>-aneurysmal aortas. The TGF-β1 mRNA level was not<br />

different between aneurysmal <strong>and</strong> control samples. TGF-β1 mRNA levels were normalized to GAPDH mRNA. Results are expressed<br />

as relative TGF-β1 mRNA levels. (B) Immunoblotting was performed in <strong>non</strong>-reducing conditions with a monoclonal anti-TGF-β1<br />

antibody (Santa Cruz). Results were normalized to GAPDH levels. (C) Quantification <strong>of</strong> <strong>the</strong> LTBP-1 level in medial extracts from<br />

aneurysmal <strong>and</strong> control aortas. LTBP-1 mRNA expression was increased in aneurysmal samples compared with control. LTBP-1<br />

mRNA levels were normalized to GAPDH mRNA. Results are expressed in relative LTBP-1 mRNA levels. LTBP-1 immunoblotting<br />

was performed in <strong>non</strong>-reducing conditions. The LTBP-1 protein level was increased in aneurysmal samples compared with controls.<br />

(D) Photomicrographs <strong>of</strong> LTBP-1 immunostaining on frozen sections <strong>of</strong> control <strong>and</strong> severely dilated aneurysmal ascending aorta<br />

(haematoxylin counterstaining). LTBP-1 staining was more intense in <strong>the</strong> media <strong>of</strong> aneurysmal sections, compared with controls,<br />

for all groups <strong>of</strong> <strong>aneurysms</strong>. LTBP-1 <strong>and</strong> TGF-β1 staining exhibited a similar distribution within <strong>the</strong> media <strong>of</strong> aneurysmal aortas.<br />

(E) Quantification by ELISA <strong>of</strong> TGF-β1 released into culture medium by aneurysmal <strong>and</strong> <strong>non</strong>-aneurysmal aortas. TGF-β1 release<br />

was measured in acidified <strong>and</strong> <strong>non</strong>-acidified tissue-culture medium. TGF-β1 was increased in acidified tissue-culture medium <strong>of</strong><br />

aneurysmal samples compared with controls. Results are expressed in ng/g <strong>of</strong> tissue wet weight (left). There was no significant<br />

difference in active TGF-β1 level (<strong>non</strong>-acidified samples) between aneurysmal <strong>and</strong> control samples (right)<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.


136 DGomezet al<br />

predominant in aneurysmal media. Next, <strong>the</strong> expression<br />

<strong>and</strong> location within <strong>the</strong> aortic wall <strong>of</strong> LTBP-1<br />

were investigated. An increase in LTBP-1 mRNA <strong>and</strong><br />

protein levels was observed in aneurysmal media compared<br />

with controls (p < 0.01) (Figure 2C). In <strong>the</strong><br />

same way, LTBP-1 immuno-staining was increased<br />

in aneurysmal media (Figure 2D), in all groups <strong>of</strong><br />

ascending aortic <strong>aneurysms</strong>. LTBP-1 staining was<br />

mainly localized within <strong>the</strong> aneurysmal media <strong>and</strong><br />

was observed in areas presenting an increase in TGFβ1<br />

immuno-staining. TGF-β1 release into conditioned<br />

media by aneurysmal aortic wall was measured by<br />

ELISA in both acidified (quantification <strong>of</strong> total TGFβ1)<br />

<strong>and</strong> <strong>non</strong>-acidified (quantification <strong>of</strong> active TGFβ1)<br />

samples (Figure 2E). TGF-β1 was increased in<br />

acidified aneurysmal samples compared with controls<br />

(p < 0.001), whatever <strong>the</strong> aetiology. In contrast, no<br />

significant differences were found in active TGF-β1<br />

levels between aneurysmal <strong>and</strong> control samples. The<br />

same results were observed in tissue extracts from <strong>the</strong><br />

aortic medial layer (data not shown).<br />

Smad2-pathway signalling activation in aneurysmal<br />

aortas<br />

In order to determine whe<strong>the</strong>r <strong>the</strong> increase in TGFβ1<br />

is associated with an up-regulation <strong>of</strong> <strong>the</strong> TGFβ1<br />

signalling pathway, we investigated <strong>the</strong> expression<br />

<strong>of</strong> pSmad2 <strong>and</strong> PAI-1 (pathway effector <strong>and</strong><br />

target, respectively). pSmad2 immuno-staining was<br />

much greater in <strong>the</strong> media <strong>of</strong> <strong>the</strong> ascending aortic<br />

<strong>aneurysms</strong> <strong>and</strong> was specifically localized in <strong>the</strong><br />

nucleus <strong>of</strong> <strong>the</strong> vascular smooth muscle cells. Contrasting<br />

with specific TGF-β1 localization, which was<br />

more intense in <strong>the</strong> outer media, pSmad2-positive<br />

nuclei were present uniformly throughout <strong>the</strong> aneurysmal<br />

media (Figure 3A). The proportion <strong>of</strong> cell nuclei<br />

positive for pSmad2 was higher in all types <strong>of</strong> ascending<br />

aortic aneurysm compared with <strong>non</strong>-aneurysmal<br />

aorta: MFS: 38 ± 16%; BAV: 46 ± 21%; <strong>and</strong> degenerative:<br />

55 ± 21%; compared with control: 11 ± 18%<br />

(Figure 3B), p < 0.01. There was no statistically significant<br />

difference between aneurysmal aetiologies.<br />

Semi-quantitative scoring <strong>of</strong> elastic fibre degradation<br />

revealed a variable relationship between this parameter<br />

<strong>and</strong> <strong>the</strong> proportion <strong>of</strong> pSmad2-positive nuclei<br />

in <strong>the</strong> different aneurysmal groups. However, <strong>the</strong>re<br />

was an overall positive correlation between <strong>the</strong> elastic<br />

lamellar fragmentation score <strong>and</strong> <strong>the</strong> proportion<br />

<strong>of</strong> pSmad2-positive nuclei in <strong>the</strong> aneurysmal aortic<br />

wall, when all aetiologies were considered (r = 0.45,<br />

p < 0.001) (Figure 3B). Smad2 phosphorylation <strong>and</strong><br />

nuclear translocation were associated with an overexpression<br />

<strong>of</strong> Smad2 in aneurysmal aortas (Figure 3C).<br />

Indeed, a significant increase in <strong>the</strong> Smad2 mRNA<br />

level was observed, whatever <strong>the</strong> aetiology (p < 0.01).<br />

To confirm Smad2 pathway activation, we measured<br />

one <strong>of</strong> its targets, PAI-1, known to be up-regulated by<br />

TGF-β [24]. The PAI-1 mRNA level was increased in<br />

aneurysmal aortas compared with controls (p < 0.01)<br />

(Figure 4A). In <strong>the</strong> same way, <strong>the</strong> PAI-1 concentration<br />

was increased in conditioned media <strong>of</strong> aneurysmal<br />

aortas (p < 0.001) (Figure 4B).<br />

Smooth muscle cell differentiation markers<br />

The state <strong>of</strong> SMC differentiation is an index <strong>of</strong> <strong>the</strong><br />

balance between syn<strong>the</strong>sis <strong>and</strong> proteolysis <strong>of</strong> extracellular<br />

matrix proteins [33], <strong>and</strong> TGF-β is one <strong>of</strong> <strong>the</strong><br />

cytokines regulating <strong>the</strong> differentiation <strong>of</strong> SMCs [34].<br />

We observed that in <strong>the</strong> areas with a high proportion<br />

<strong>of</strong> pSmad2-positive nuclei, embryonic myosin (MHC<br />

IIB) was markedly increased (Figure 5).<br />

However, <strong>the</strong> mature form <strong>of</strong> MHC, SM2-MHC,<br />

was also expressed, toge<strong>the</strong>r with MHC IIB (Figure 5,<br />

right panel). α-SMC actin was expressed in all <strong>the</strong><br />

SMCs <strong>of</strong> <strong>the</strong> media <strong>and</strong> <strong>the</strong>refore did not appear to be<br />

influenced by Smad2 activation. Again, <strong>the</strong>se results<br />

were similar in all groups <strong>of</strong> <strong>aneurysms</strong>.<br />

The expression <strong>of</strong> small leucine-rich<br />

proteoglycans: tissue modulators <strong>of</strong> TGF-β activity<br />

Staining for decorin was observed in <strong>the</strong> adventitia<br />

<strong>and</strong> <strong>the</strong> intima <strong>of</strong> aneurysmal aortas <strong>and</strong> controls,<br />

<strong>and</strong> was similar in <strong>the</strong> control <strong>and</strong> aneurysmal wall.<br />

In contrast, staining for decorin was absent from<br />

<strong>the</strong> normal media, whereas it was focally positive<br />

in media from <strong>the</strong> aneurysmal aorta (Figure 6A).<br />

It co-localized with fibrillar collagen (Figure 6B).<br />

Biglycan staining was associated with elastic lamellae<br />

in controls <strong>and</strong> was correspondingly decreased in<br />

areas with important elastic lamellar fragmentation<br />

<strong>and</strong> disorganization <strong>of</strong> arterial structure in <strong>aneurysms</strong><br />

(Figure 6C). These observations were made in all<br />

ascending aortic <strong>aneurysms</strong>. Therefore, activation <strong>of</strong><br />

<strong>the</strong> TGF-β pathway in <strong>the</strong> media <strong>of</strong> aneurysmal aortas<br />

did not appear to be associated with any consistent<br />

increase in its natural tissue regulators, decorin <strong>and</strong><br />

biglycan.<br />

Discussion<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

Our data indicate that Smad2 activation <strong>and</strong> increase<br />

in stored TGF-β1 are concomitantly observed in <strong>the</strong><br />

media <strong>of</strong> human ascending aortic aneurysm. Smad2<br />

expression <strong>and</strong> its signalling pathway were activated<br />

in both <strong>syndromic</strong>, as previously described [4,13,28],<br />

<strong>and</strong> <strong>non</strong>-<strong>syndromic</strong> forms <strong>of</strong> aneurysmal disease. In<br />

contrast, <strong>the</strong>re was no consistent association between<br />

<strong>the</strong> nuclear location <strong>of</strong> pSmad2 <strong>and</strong> extracellular<br />

TGF-β1 staining <strong>and</strong> between <strong>the</strong>ir respective mRNA<br />

expression.<br />

Our data indicate that <strong>the</strong> observed increase in<br />

TGF-β1, in pathological sections, corresponds to an<br />

enhancement <strong>of</strong> <strong>the</strong> storage <strong>and</strong> sequestration <strong>of</strong> TGFβ1<br />

in <strong>the</strong> extracellular matrix. In contrast, <strong>the</strong> expression<br />

<strong>and</strong> activation <strong>of</strong> TGF-β1 were unchanged in<br />

aneurysmal media compared with control aorta. In


Smad2 activation in ascending aortic aneurysm 137<br />

Figure 3. Activation <strong>of</strong> <strong>the</strong> TGF-β-signalling Smad2 pathway in <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> ascending aorta. (A) Representative<br />

photomicrographs <strong>of</strong> phosphorylated Smad2 immunostaining in sections <strong>of</strong> aneurysmal <strong>and</strong> control ascending aorta. The<br />

staining is localized within vascular SMC nuclei. pSmad2-positive nuclei are far more numerous in aneurysmal than in control aorta.<br />

The lumen is placed on <strong>the</strong> left (original magnification ×40). (B) Serial sections <strong>of</strong> ascending aorta stained with orcein <strong>and</strong> Alcian<br />

blue. Breakdown <strong>of</strong> elastic fibre architecture can be seen in <strong>the</strong> aneurysmal aortic wall, as well as areas <strong>of</strong> mucoid degeneration.<br />

Adjacent serial sections showing nuclear immunostaining for phosphorylated Smad2 in SMCs in <strong>the</strong> vicinity <strong>of</strong> zones <strong>of</strong> mucoid<br />

degeneration. The proportion <strong>of</strong> pSmad2-positive nuclei is increased in <strong>the</strong> media <strong>of</strong> aneurysmal aorta (degenerative aneurysm)<br />

compared with control aorta. This pSmad2 increase is observed in all types <strong>of</strong> aortic aneurysm (original magnification × 200).<br />

Top right: bar graph displaying <strong>the</strong> percentage <strong>of</strong> pSmad2-positive nuclei in patient groups: <strong>non</strong>-aneurysmal control aorta<br />

(n = 10); Marfan syndrome [n = 15, including TGFBR2 mutations (n = 2)]; bicuspid aortic valve ‘BAV’ (n = 15); <strong>and</strong> degenerative<br />

aneurysm (n = 19) ( ∗∗∗ p < 0.001). Bottom right: scatter plot with <strong>the</strong> predicted regression line showing <strong>the</strong> positive correlation<br />

between pSmad2-positive nuclei <strong>and</strong> elastic lamellar breakdown in all groups <strong>of</strong> ascending aortic <strong>aneurysms</strong> taken toge<strong>the</strong>r<br />

(p < 0.001). Elastic network disintegration was evaluated by semi-quantitative scoring (see <strong>the</strong> Materials <strong>and</strong> methods section).<br />

(C) Quantification <strong>of</strong> Smad2 mRNA levels in medial extracts from aneurysmal <strong>and</strong> control aortas. The Smad2 mRNA level was<br />

increased in aneurysmal aorta compared with control, in all types <strong>of</strong> aneurysm. Smad2 mRNA levels were normalized to GAPDH<br />

mRNA. Results are expressed as relative Smad2 mRNA levels<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.


138 DGomezet al<br />

Figure 4. Quantification <strong>of</strong> PAI-1 (plasminogen activator inhibitor 1), a target <strong>of</strong> <strong>the</strong> Smad2 signalling pathway. (A) Quantification<br />

<strong>of</strong> <strong>the</strong> PAI-1 mRNA level in medial extracts. PAI-1 mRNA levels were increased in aneurysmal aortas compared with control,<br />

whatever <strong>the</strong> aetiology. PAI-1 mRNA levels were normalized to GAPDH mRNA. (B) Quantification <strong>of</strong> PAI-1 using ELISA in tissue<br />

culture media <strong>of</strong> aneurysmal <strong>and</strong> control aortas<br />

physiological conditions, TGF-β1 storage, release, <strong>and</strong><br />

activation are tightly regulated [35]. A latent form<br />

<strong>of</strong> TGF-β1 is syn<strong>the</strong>sized <strong>and</strong> secreted by SMCs<br />

<strong>and</strong> is associated with LTPB-1 within <strong>the</strong> extracellular<br />

matrix. In <strong>the</strong> present study, overexpression <strong>of</strong><br />

LTBP-1 (mRNA <strong>and</strong> protein) was observed. Despite<br />

<strong>the</strong> impossibility <strong>of</strong> performing analysis on serial sections<br />

(fixed versus frozen sections), <strong>the</strong> localization<br />

<strong>of</strong> LTBP-1 staining appears to be similar to that <strong>of</strong><br />

TGF-β1 staining. This suggests that <strong>the</strong> overexpression<br />

<strong>of</strong> LTBP-1 could be a major component responsible<br />

for <strong>the</strong> differential storage <strong>of</strong> TGF-β1 within<br />

<strong>the</strong> aneurysmal media. Moreover, LTBP-1 interacts<br />

with fibronectin in fibrillar structures [36]. Recent<br />

findings showed that fibronectin is overexpressed in<br />

<strong>the</strong> outer curvature <strong>of</strong> dilated ascending aortas from<br />

Marfan <strong>and</strong> BAV patients [37]. This overexpression<br />

<strong>of</strong> fibronectin could also participate in <strong>the</strong> increase<br />

in TGF-β1 storage/sequestration. On <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>,<br />

<strong>the</strong> production <strong>of</strong> small leucine-rich proteoglycans (eg<br />

decorin <strong>and</strong> biglycan) can retain <strong>the</strong> active form <strong>of</strong><br />

TGF-β1 [38,39]. In <strong>the</strong> present study, we observed a<br />

decrease in biglycan staining associated with important<br />

elastic lamellar fragmentation <strong>and</strong> heterogeneous<br />

increase in decorin staining. Thus, <strong>the</strong>se global modifications<br />

<strong>of</strong> <strong>the</strong> integrity <strong>of</strong> <strong>the</strong> extracellular matrix,<br />

<strong>the</strong> localized process <strong>of</strong> matrix repair <strong>and</strong> <strong>the</strong> overexpression<br />

<strong>of</strong> LTBP-1, could induce a differential heterogeneous<br />

storage <strong>of</strong> TGF-β1 in <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong><br />

ascending aorta. These processes appear not to affect<br />

TGF-β1 activation, since no difference in active TGFβ1<br />

levels was observed, in any aetiological group <strong>of</strong><br />

<strong>aneurysms</strong>. These findings contrast with <strong>the</strong> hypo<strong>the</strong>sis<br />

suggesting that matrix degradation <strong>and</strong> micr<strong>of</strong>ibril<br />

proteolysis are involved in <strong>the</strong> activation <strong>of</strong> TGF-β1<br />

in aneurysmal disease [27].<br />

TGF-β signalling was shown to be increased in <strong>the</strong><br />

ascending aorta <strong>of</strong> Marfan patients with mutations in<br />

FBN1 [29], as well as in transgenic experimental models<br />

[28]. Fibrillin-1 is <strong>the</strong> most abundant micr<strong>of</strong>ibril<br />

present in elastic fibres <strong>and</strong> plays an essential role in<br />

<strong>the</strong> regulated deposition <strong>of</strong> tropoelastin molecules during<br />

development [40], but it can also participate in <strong>the</strong><br />

regulation <strong>of</strong> TGF-β bioavailability [41]. Habashi et al<br />

have shown that treatment <strong>of</strong> <strong>the</strong> transgenic model<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

Figure 5. Differentiation state <strong>of</strong> medial smooth muscle cells<br />

in <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> ascending aorta. Serial histological sections<br />

show representative ascending aortic walls <strong>of</strong> controls (left<br />

panel) <strong>and</strong> <strong>aneurysms</strong> with severe architectural disruption (right<br />

panel). Expression <strong>of</strong> MHC IIB (<strong>the</strong> <strong>non</strong>-muscle embryonic<br />

myosin) is strongly induced in SMCs close to areas <strong>of</strong> severe<br />

disruption <strong>of</strong> <strong>the</strong> aortic architecture (E, F) <strong>and</strong> co-localizes<br />

with increased pSMAD2 staining (C, D). Nei<strong>the</strong>r SM2-MHC<br />

(G, H) nor α-SM-actin (I, J) expression is modified with<br />

increased pSmad2 staining. (A, B) Alcian blue stain (original<br />

magnification × 200). These results are from a degenerative<br />

aneurysm. The same observations were made, whatever <strong>the</strong><br />

aetiology


Smad2 activation in ascending aortic aneurysm 139<br />

Figure 6. Decorin <strong>and</strong> biglycan expression patterns in <strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> ascending aorta. (A) Decorin staining is localized in <strong>the</strong><br />

intima (left) <strong>and</strong> adventitia (right) <strong>of</strong> both control <strong>and</strong> aneurysmal aorta. In aneurysmal aorta, decorin is expressed focally in <strong>the</strong><br />

media (original magnification × 40). (B) Serial sections <strong>of</strong> a representative aneurysm characterized by diffuse destruction <strong>of</strong> elastic<br />

lamellae <strong>and</strong> extensive fibrosis, revealing <strong>the</strong> co-localization <strong>of</strong> collagen accumulation (yellow/yellowish green) <strong>and</strong> decorin/TGF-β1<br />

staining (original magnification × 40). (C) Elastin fibre breakdown is associated with a corresponding loss <strong>of</strong> biglycan staining<br />

(original magnification × 200)<br />

with TGF-β antibody can prevent dilatation <strong>of</strong> <strong>the</strong><br />

aortic <strong>aneurysms</strong> [29], possibly by preventing SMC<br />

apoptosis. In contrast, in <strong>the</strong>ir experimental study, Dai<br />

et al demonstrated that overexpression <strong>of</strong> TGF-β in a<br />

rat model <strong>of</strong> aneurysm prevented fur<strong>the</strong>r progression<br />

<strong>of</strong> dilatation [42]. Therefore, <strong>the</strong> exact role <strong>of</strong> TGF-β<br />

in aneurysmal disease remains to be fur<strong>the</strong>r clarified.<br />

Increased Smad2 signalling was also present in <strong>the</strong><br />

aortic wall from patients with TGFBR2 mutations,<br />

as described by Loeys et al [4,13]. Our findings are<br />

in agreement with <strong>the</strong>se observations <strong>and</strong> extend <strong>the</strong><br />

occurrence <strong>of</strong> TGF-β accumulation <strong>and</strong> Smad2 activation<br />

to all aetiologies <strong>of</strong> aortic <strong>aneurysms</strong>, including<br />

<strong>non</strong>-<strong>syndromic</strong> forms.<br />

We showed an increase in <strong>the</strong> nuclear location<br />

<strong>of</strong> phosphorylated Smad2. Surprisingly, pSmad2 was<br />

present in nuclei uniformly distributed throughout <strong>the</strong><br />

media in aneurysmal aorta as previously described<br />

[13], whereas <strong>the</strong> localization <strong>of</strong> increased TGF-β1<br />

protein was restricted to <strong>the</strong> adventitia <strong>and</strong> outer<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.


140 DGomezet al<br />

media in <strong>the</strong> aneurysmal aortas. Smad2 activation was<br />

associated with an increase in Smad2 mRNA expression.<br />

These findings, in addition to active TGF-β1<br />

quantification, indicate that <strong>the</strong> increase in stored TGFβ1<br />

<strong>and</strong> <strong>the</strong> activation <strong>of</strong> Smad2 are not tightly linked.<br />

Moreover, <strong>the</strong> unchanged TGF-β1 mRNA expression<br />

excludes any activation <strong>of</strong> Smad2 induced by autocrine<br />

production <strong>of</strong> TGF-β1 by vSMCs. Therefore, <strong>the</strong> activation<br />

<strong>of</strong> Smad2 may be not only <strong>the</strong> consequence <strong>of</strong><br />

increased release <strong>and</strong> activation <strong>of</strong> extracellular TGFβ1,<br />

but may also involve a parallel autonomous intracellular<br />

hypersignal. Smad overexpression <strong>and</strong> activation<br />

could also be induced by o<strong>the</strong>r extracellular<br />

lig<strong>and</strong>s such as angiotensin II [43,44]. The enhancement<br />

<strong>of</strong> stored TGF-β1 is probably concomitant to<br />

aneurysm development but not <strong>the</strong> main cause <strong>of</strong> <strong>the</strong><br />

aneurysmal disease. Smad2 activation appears to be a<br />

common starting point for all <strong>the</strong>se different forms <strong>of</strong><br />

aneurysm, including those <strong>of</strong> patients bearing mutations<br />

in TGFBR2 [3]. This opposition between Smad2<br />

activation <strong>and</strong> <strong>the</strong> loss <strong>of</strong> function <strong>of</strong> TGFβRII [3]<br />

also suggests that Smad2 activation is not only dependent<br />

on TGF-β1 release from <strong>the</strong> extracellular matrix,<br />

but that <strong>the</strong>re is also a dissociation between TGF-β1<br />

release, TGFβRII functionality, <strong>and</strong> Smad intracellular<br />

signalling.<br />

Different aetiology-associated mechanisms may<br />

induce TGF-β/Smad activation (eg degenerative processes,<br />

mutations in various genes, a defect in SMC<br />

anchorage [45]), all leading to <strong>the</strong> same events, eg<br />

SMC apoptosis, mucoid degeneration, <strong>and</strong> matrix<br />

degradation. Smad2 activation was globally correlated<br />

with elastic fibre destruction in our study. Matrix metalloproteinase<br />

(MMP) activation is implicated in collagen<br />

<strong>and</strong> elastic fibre degradation in <strong>aneurysms</strong> <strong>of</strong><br />

<strong>the</strong> ascending aorta [12,46,47]. Moreover, <strong>the</strong> TGF-β1<br />

pathway is known to regulate several MMPs (MMP-<br />

2, MMP-9 [48]), <strong>the</strong>refore indirectly participating in<br />

matrix degradation.<br />

We have shown here that Smad2 activation colocalized<br />

with <strong>the</strong> embryonic form <strong>of</strong> myosin expression<br />

in <strong>the</strong> media <strong>of</strong> ascending aortic <strong>aneurysms</strong>. MHC<br />

IIB is a <strong>non</strong>-muscle type MHC abundantly expressed<br />

in immature SMCs during fetal life <strong>and</strong> its expression<br />

is decreased in well-differentiated cells [49]. It<br />

can re-appear in response to wall injury [50,51]. In<br />

contrast with our results, TGF-β has been reported<br />

elsewhere to induce SMC differentiation in experimental<br />

studies [34]. This discrepancy may be explained by<br />

<strong>the</strong> fact that different responses to TGF-β have been<br />

reported in SMCs originating from <strong>the</strong> cardiac neural<br />

crest [52] compared with SMCs <strong>of</strong> mesodermal origin<br />

[53]. Alternatively, <strong>the</strong> alterations <strong>of</strong> <strong>the</strong> extracellular<br />

matrix network may induce SMC dedifferentiation<br />

[54].<br />

One <strong>of</strong> <strong>the</strong> limitations <strong>of</strong> our study is <strong>the</strong> difficulty in<br />

obtaining aortic samples from patients with TGFBR2<br />

mutations [3]. It is noteworthy that <strong>aneurysms</strong> not<br />

related to FBN1 or TGFBR2 mutations, but associated<br />

with BAV or degenerative diseases, ie <strong>non</strong>-<strong>syndromic</strong><br />

forms, also showed up-regulation <strong>of</strong> Smad2 signalling.<br />

Our study was performed on <strong>the</strong> dilated part <strong>of</strong> <strong>the</strong><br />

aneurysmal aortas. However, investigation <strong>of</strong> Smad2<br />

signalling <strong>and</strong> TGF-β1 storage may be performed in<br />

<strong>the</strong> <strong>non</strong>-dilated part <strong>of</strong> future aneurysmal samples <strong>and</strong><br />

compared with <strong>the</strong> present results. Taken toge<strong>the</strong>r,<br />

<strong>the</strong>se data indicate that in addition to <strong>the</strong> common<br />

histopathological features <strong>of</strong> all types <strong>of</strong> human<br />

<strong>aneurysms</strong> <strong>of</strong> <strong>the</strong> ascending aorta involving extracellular<br />

matrix breakdown associated with mucoid degeneration<br />

[12,14], <strong>the</strong>re is probably also a common activation<br />

<strong>of</strong> <strong>the</strong> Smad2 pathway within smooth muscle<br />

cells, dependent or not on TGF-β1 activation. This<br />

study thus extends <strong>and</strong> generalizes to all <strong>aneurysms</strong> <strong>of</strong><br />

<strong>the</strong> ascending aorta, including <strong>non</strong>-<strong>syndromic</strong> forms,<br />

<strong>the</strong> previous results describing <strong>the</strong> activation <strong>of</strong> <strong>the</strong><br />

Smad2 pathway in <strong>syndromic</strong> aortic <strong>aneurysms</strong> [4,28].<br />

In view <strong>of</strong> this observation, it could be <strong>of</strong> interest to<br />

extend <strong>the</strong> preventive treatment used in Marfan syndrome<br />

to o<strong>the</strong>r forms <strong>of</strong> aneurysmal dilatation <strong>of</strong> <strong>the</strong><br />

ascending aorta, as recently proposed [55].<br />

Acknowledgements<br />

This study was supported by grants from <strong>the</strong> French Society<br />

<strong>of</strong> Cardiology, <strong>the</strong> French Federation <strong>of</strong> Cardiology, <strong>and</strong> <strong>the</strong><br />

National Research Agency (ANR). This study was part <strong>of</strong><br />

<strong>the</strong> FAD (Fighting Aneurysmal Disease) project supported by<br />

<strong>the</strong> European Community (FP-7 200647, http://www.fightinganeurysm.org).<br />

We would like to thank Mary Osborne-Pellegrin<br />

for editing <strong>the</strong> manuscript <strong>and</strong> surgeons <strong>of</strong> <strong>the</strong> cardiac <strong>and</strong><br />

vascular surgery departments for providing us with normal<br />

<strong>and</strong> pathological aortic tissues. Paulo Sampaio Gutierrez was<br />

sponsored by CNPq, Brazil. Luciano de Figueiredo Borges<br />

was sponsored by FAPESP, Brazil <strong>and</strong> Paris 7-Denis Diderot<br />

University, France.<br />

References<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

1. Isselbacher EM. Thoracic <strong>and</strong> abdominal aortic <strong>aneurysms</strong>.<br />

Circulation 2005;111:816–828.<br />

2. Touat Z, Lepage L, Ollivier V, Nataf P, Hvass U, Labreuche J,<br />

et al. Dilation-dependent activation <strong>of</strong> platelets <strong>and</strong> prothrombin<br />

in human thoracic ascending aortic aneurysm. Arterioscler Thromb<br />

Vasc Biol 2008;28:940–946.<br />

3. Mizuguchi T, Collod-Beroud G, Akiyama T, Abifadel M, Harada<br />

N, Morisaki T, et al. Heterozygous TGFBR2 mutations in Marfan<br />

syndrome. Nature Genet 2004;36:855–860.<br />

4. Loeys BL, Chen J, Neptune ER, Judge DP, Podowski M, Holm T,<br />

et al. A syndrome <strong>of</strong> altered cardiovascular, crani<strong>of</strong>acial,<br />

neurocognitive <strong>and</strong> skeletal development caused by mutations in<br />

TGFBR1 or TGFBR2. Nature Genet 2005;37:275–281.<br />

5. Loeys BL, Schwarze U, Holm T, Callewaert BL, Thomas GH,<br />

Pannu H, et al. Aneurysm syndromes caused by mutations in <strong>the</strong><br />

TGF-beta receptor. N Engl J Med 2006;355:788–798.<br />

6. Robinson PN, Arteaga-Solis E, Baldock C, Collod-Beroud G,<br />

Booms P, De Paepe A, et al. The molecular genetics <strong>of</strong> Marfan<br />

syndrome <strong>and</strong> related disorders. JMedGenet2006;43:769–787.<br />

7. Zhu L, Vranckx R, Khau Van Kien P, Lal<strong>and</strong>e A, Boisset N,<br />

Mathieu F, et al. Mutations in myosin heavy chain 11 cause a<br />

syndrome associating thoracic aortic aneurysm/aortic dissection<br />

<strong>and</strong> patent ductus arteriosus. Nature Genet 2006;38:343–349.<br />

8. Guo DC, Pannu H, Tran-Fadulu V, Papke CL, Yu RK, Avidan N,<br />

et al. Mutations in smooth muscle alpha-actin (ACTA2) lead


Smad2 activation in ascending aortic aneurysm 141<br />

to thoracic aortic <strong>aneurysms</strong> <strong>and</strong> dissections. Nature Genet<br />

2007;39:1488–1493.<br />

9. Coucke PJ, Willaert A, Wessels MW, Callewaert B, Zoppi N, De<br />

Backer J, et al. Mutations in <strong>the</strong> facilitative glucose transporter<br />

GLUT10 alter angiogenesis <strong>and</strong> cause arterial tortuosity syndrome.<br />

Nature Genet 2006;38:452–457.<br />

10. Roos-Hesselink JW, Scholzel BE, Heijdra RJ, Spitaels SE, Meijboom<br />

FJ, Boersma E, et al. Aortic valve <strong>and</strong> aortic arch pathology<br />

after coarctation repair. Heart 2003;89:1074–1077.<br />

11. Klein DG. Thoracic aortic <strong>aneurysms</strong>. J Cardiovasc Nurs<br />

2005;20:245–250.<br />

12. Borges LF, Touat Z, Leclercq A, Al Haj Zen A, Jondeau G,<br />

Franc B, et al. Tissue diffusion <strong>and</strong> retention <strong>of</strong> metalloproteinases<br />

in ascending aortic <strong>aneurysms</strong> <strong>and</strong> dissections. Hum Pathol 2008;<br />

Oct 28 [Epub ahead <strong>of</strong> print].<br />

13. Maleszewski JJ, Miller DV, Lu J, Dietz HC, Halushka MK.<br />

Histopathologic findings in ascending aortas from individuals<br />

with Loeys–Dietz syndrome (LDS). Am J Surg Pathol<br />

2009;33:194–201.<br />

14. de Figueiredo Borges L, Jaldin RG, Dias RR, Stolf NA, Michel<br />

JB, Gutierrez PS. Collagen is reduced <strong>and</strong> disrupted in human<br />

<strong>aneurysms</strong> <strong>and</strong> dissections <strong>of</strong> ascending aorta. Hum Pathol<br />

2008;39:437–443.<br />

15. Klima T, Spjut HJ, Coelho A, Gray AG, Wukasch DC, Reul GJ<br />

Jr, et al. The morphology <strong>of</strong> ascending aortic <strong>aneurysms</strong>. Hum<br />

Pathol 1983;14:810–817.<br />

16. Bonderman D, Gharehbaghi-Schnell E, Wollenek G, Maurer G,<br />

Baumgartner H, Lang IM. Mechanisms underlying aortic dilatation<br />

in congenital aortic valve malformation. Circulation<br />

1999;99:2138–2143.<br />

17. Bonyadi M, Rusholme SA, Cousins FM, Su HC, Biron CA,<br />

Farrall M, et al. Mapping <strong>of</strong> a major genetic modifier <strong>of</strong><br />

embryonic lethality in TGF beta 1 knockout mice. Nature Genet<br />

1997;15:207–211.<br />

18. Owens GK, Wise G. Regulation <strong>of</strong> differentiation/maturation in<br />

vascular smooth muscle cells by hormones <strong>and</strong> growth factors.<br />

Agents Actions Suppl 1997;48:3–24.<br />

19. Corde<strong>non</strong>si M, Dupont S, Maretto S, Insinga A, Imbriano C,<br />

Piccolo S. Links between tumor suppressors: p53 is required<br />

for TGF-beta gene responses by cooperating with Smads. Cell<br />

2003;113:301–314.<br />

20. Derynck R, Zhang YE. Smad-dependent <strong>and</strong> Smad-independent<br />

pathways in TGF-beta family signalling. Nature<br />

2003;425:577–584.<br />

21. Shi Y, Massague J. Mechanisms <strong>of</strong> TGF-beta signaling from cell<br />

membrane to <strong>the</strong> nucleus. Cell 2003;113:685–700.<br />

22. Ignotz RA, Massague J. Transforming growth factor-beta stimulates<br />

<strong>the</strong> expression <strong>of</strong> fibronectin <strong>and</strong> collagen <strong>and</strong> <strong>the</strong>ir<br />

incorporation into <strong>the</strong> extracellular matrix. J Biol Chem<br />

1986;261:4337–4345.<br />

23. Ma C, Tarnuzzer RW, Chegini N. Expression <strong>of</strong> matrix metalloproteinases<br />

<strong>and</strong> tissue inhibitor <strong>of</strong> matrix metalloproteinases in<br />

meso<strong>the</strong>lial cells <strong>and</strong> <strong>the</strong>ir regulation by transforming growth<br />

factor-beta1. Wound Repair Regen 1999;7:477–485.<br />

24. Dong C, Zhu S, Wang T, Yoon W, Goldschmidt-Clermont PJ.<br />

Upregulation <strong>of</strong> PAI-1 is mediated through TGF-beta/Smad<br />

pathway in transplant arteriopathy. J Heart Lung Transplant<br />

2002;21:999–1008.<br />

25. Oklu R, Hesketh R. The latent transforming growth factor beta<br />

binding protein (LTBP) family. Biochem J 2000;352:601–610.<br />

26. Isogai Z, Ono RN, Ushiro S, Keene DR, Chen Y, Mazzieri R,<br />

et al. Latent transforming growth factor beta-binding protein 1<br />

interacts with fibrillin <strong>and</strong> is a micr<strong>of</strong>ibril-associated protein. J<br />

Biol Chem 2003;278:2750–2757.<br />

27. ten Dijke P, Arthur HM. Extracellular control <strong>of</strong> TGFbeta<br />

signalling in vascular development <strong>and</strong> disease. Nature Rev Mol<br />

Cell Biol 2007;8:857–869.<br />

28. Neptune ER, Frischmeyer PA, Arking DE, Myers L, Bunton TE,<br />

Gayraud B, et al. Dysregulation <strong>of</strong> TGF-beta activation contributes<br />

to pathogenesis in Marfan syndrome. Nature Genet<br />

2003;33:407–411.<br />

29. Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL,<br />

Cooper TK, et al. Losartan, an AT1 antagonist, prevents aortic<br />

aneurysm in a mouse model <strong>of</strong> Marfan syndrome. Science<br />

2006;312:117–121.<br />

30. Schlatmann TJ, Becker AE. Pathogenesis <strong>of</strong> dissecting aneurysm<br />

<strong>of</strong> aorta. Comparative histopathologic study <strong>of</strong> significance <strong>of</strong><br />

medial changes. Am J Cardiol 1977;39:21–26.<br />

31. Delbosc S, Haloui M, Louedec L, Dupuis M, Cubizolles M,<br />

Podust VN, et al. Proteomic analysis permits <strong>the</strong> identification <strong>of</strong><br />

new biomarkers <strong>of</strong> arterial wall remodeling in hypertension. Mol<br />

Med 2008;14:383–394.<br />

32. Moustakas A, Souchelnytskyi S, Heldin CH. Smad regulation in<br />

TGF-beta signal transduction. J Cell Sci 2001;114:4359–4369.<br />

33. Owens GK. Regulation <strong>of</strong> differentiation <strong>of</strong> vascular smooth<br />

muscle cells. Physiol Rev 1995;75:487–517.<br />

34. Sinha S, Ho<strong>of</strong>nagle MH, Kingston PA, McCanna ME, Owens GK.<br />

Transforming growth factor-beta1 signaling contributes to development<br />

<strong>of</strong> smooth muscle cells from embryonic stem cells. Am J<br />

Physiol Cell Physiol 2004;287:C1560–C1568.<br />

35. O’Rourke MF, Nichols WW. Aortic diameter, aortic stiffness,<br />

<strong>and</strong> wave reflection increase with age <strong>and</strong> isolated systolic<br />

hypertension. Hypertension 2005;45:652–658.<br />

36. Taipale J, Saharinen J, Hedman K, Keski-Oja J. Latent transforming<br />

growth factor-beta 1 <strong>and</strong> its binding protein are components<br />

<strong>of</strong> extracellular matrix micr<strong>of</strong>ibrils. J Histochem Cytochem<br />

1996;44:875–889.<br />

37. Della Corte A, De Santo LS, Montagnani S, Quarto C, Romano G,<br />

Amarelli C, et al. Spatial patterns <strong>of</strong> matrix protein expression<br />

in dilated ascending aorta with aortic regurgitation: congenital<br />

bicuspid valve versus Marfan’s syndrome. J Heart Valve Dis<br />

2006;15:20–7; discussion 27.<br />

38. Kinsella MG, Bressler SL, Wight TN. The regulated syn<strong>the</strong>sis <strong>of</strong><br />

versican, decorin, <strong>and</strong> biglycan: extracellular matrix proteoglycans<br />

that influence cellular phenotype. Crit Rev Eukaryot Gene Expr<br />

2004;14:203–234.<br />

39. Yamaguchi Y, Mann DM, Ruoslahti E. Negative regulation <strong>of</strong><br />

transforming growth factor-beta by <strong>the</strong> proteoglycan decorin.<br />

Nature 1990;346:281–284.<br />

40. Trask TM, Trask BC, Ritty TM, Abrams WR, Rosenbloom J,<br />

Mecham RP. Interaction <strong>of</strong> tropoelastin with <strong>the</strong> amino-terminal<br />

domains <strong>of</strong> fibrillin-1 <strong>and</strong> fibrillin-2 suggests a role for <strong>the</strong> fibrillins<br />

in elastic fiber assembly. JBiolChem2000;275:24400–24406.<br />

41. Kaartinen V, Warburton D. Fibrillin controls TGF-beta activation.<br />

Nature Genet 2003;33:331–332.<br />

42. Dai J, Losy F, Guinault AM, Pages C, Anegon I, Desgranges P,<br />

et al. Overexpression <strong>of</strong> transforming growth factor-beta1<br />

stabilizes already-formed aortic <strong>aneurysms</strong>: a first approach to<br />

induction <strong>of</strong> functional healing by endovascular gene <strong>the</strong>rapy.<br />

Circulation 2005;112:1008–1015.<br />

43. Kamada Y, Tamura S, Kiso S, Fukui K, Doi Y, Ito N, et al.<br />

Angiotensin II stimulates <strong>the</strong> nuclear translocation <strong>of</strong> Smad2 <strong>and</strong><br />

induces PAI-1 mRNA in rat hepatic stellate cells. Hepatol Res<br />

2003;25:296–305.<br />

44. Rodriguez-Vita J, Sanchez-Lopez E, Esteban V, Ruperez M,<br />

Egido J, Ruiz-Ortega M. Angiotensin II activates <strong>the</strong> Smad<br />

pathway in vascular smooth muscle cells by a transforming<br />

growth factor-beta-independent mechanism. Circulation<br />

2005;111:2509–2517.<br />

45. Milewicz DM, Guo DC, Tran-Fadulu V, Lafont AL, Papke CL,<br />

Inamoto S, et al. Genetic basis <strong>of</strong> thoracic aortic <strong>aneurysms</strong> <strong>and</strong><br />

dissections: focus on smooth muscle cell contractile dysfunction.<br />

Annu Rev Genomics Hum Genet 2008;9:283–302.<br />

46. Boyum J, Fellinger EK, Schmoker JD, Trombley L, McPartl<strong>and</strong><br />

K, Ittleman FP, et al. Matrix metalloproteinase activity in<br />

thoracic aortic <strong>aneurysms</strong> associated with bicuspid <strong>and</strong> tricuspid<br />

aortic valves. J Thorac Cardiovasc Surg 2004;127:686–691.<br />

47. Koullias GJ, Ravich<strong>and</strong>ran P, Korkolis DP, Rimm DL, Elefteriades<br />

JA. Increased tissue microarray matrix metalloproteinase<br />

expression favors proteolysis in thoracic aortic <strong>aneurysms</strong> <strong>and</strong><br />

dissections. Ann Thorac Surg 2004;78:2106–2110; discussion<br />

2110–2111.<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.


142 DGomezet al<br />

48. Kim ES, Kim MS, Moon A. TGF-beta-induced upregulation <strong>of</strong><br />

MMP-2 <strong>and</strong> MMP-9 depends on p38 MAPK, but not ERK<br />

signaling in MCF10A human breast epi<strong>the</strong>lial cells. Int J Oncol<br />

2004;25:1375–1382.<br />

49. Owens GK, Kumar MS, Wamh<strong>of</strong>f BR. Molecular regulation <strong>of</strong><br />

vascular smooth muscle cell differentiation in development <strong>and</strong><br />

disease. Physiol Rev 2004;84:767–801.<br />

50. Aikawa M, Sivam PN, Kuro-o M, Kimura K, Nakahara K,<br />

Takewaki S, et al. Human smooth muscle myosin heavy chain<br />

is<strong>of</strong>orms as molecular markers for vascular development <strong>and</strong><br />

a<strong>the</strong>rosclerosis. Circ Res 1993;73:1000–1012.<br />

51. Ueda M, Becker AE, Naruko T, Kojima A. Smooth muscle cell<br />

de-differentiation is a fundamental change preceding wound<br />

healing after percutaneous transluminal coronary angioplasty in<br />

humans. Coron Artery Dis 1995;6:71–81.<br />

J Pathol 2009; 218: 131–142 DOI: 10.1002/path<br />

Copyright © 2009 Pathological Society <strong>of</strong> Great Britain <strong>and</strong> Irel<strong>and</strong>. Published by John Wiley & Sons, Ltd.<br />

52. Bergwerff M, Verberne ME, DeRuiter MC, Poelmann RE,<br />

Gittenberger-de Groot AC. Neural crest cell contribution to <strong>the</strong><br />

developing circulatory system: implications for vascular morphology?<br />

Circ Res 1998;82:221–231.<br />

53. Topouzis S, Majesky MW. Smooth muscle lineage diversity<br />

in <strong>the</strong> chick embryo. Two types <strong>of</strong> aortic smooth muscle<br />

cell differ in growth <strong>and</strong> receptor-mediated transcriptional<br />

responses to transforming growth factor-beta. Dev Biol<br />

1996;178:430–445.<br />

54. Karnik SK, Brooke BS, Bayes-Genis A, Sorensen L, Wy<strong>the</strong> JD,<br />

Schwartz RS, et al. A critical role for elastin signaling in vascular<br />

morphogenesis <strong>and</strong> disease. Development 2003;130:411–423.<br />

55. Brooke BS, Habashi JP, Judge DP, Patel N, Loeys B, Dietz HC<br />

3rd. Angiotensin II blockade <strong>and</strong> aortic-root dilation in Marfan’s<br />

syndrome. N Engl J Med 2008;358:2787–2795.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!