You are on page 1of 10

Hemodynamic Modulation of Endocardial Thromboresistance

Navin K. Kapur, MD; Clayton B. Deming, MS; Sunil Kapur, BS; Ce Bian, MD;
Hunter C. Champion, MD, PhD; J. Kevin Donahue, MD; David A. Kass, MD; Jeffrey J. Rade, MD

Background—Patients with heart failure are at increased risk for thromboembolic events, including stroke. Historically
attributed to blood stasis, little is known about the adverse effects of elevated chamber filling pressure on endocardial
function, which could predispose to intracardiac thrombus formation.
Methods and Results—We investigated changes in the expression of thrombomodulin, a key component of the
anticoagulant protein C pathway, in rats subjected to acute atrial pressure overload caused by aortic banding. Acute
elevation of left atrial filling pressure, without an associated decline in ventricular systolic function, caused a 70%
inhibition of atrial endocardial thrombomodulin expression and resulted in increased local thrombin generation.
Targeted restoration of atrial thrombomodulin expression with adenovirus-mediated gene transfer successfully reduced
thrombin generation to baseline levels. In vitro co-culture studies revealed that thrombomodulin downregulation is
caused by the paracrine release of transforming growth factor-␤ from cardiac connective tissue in response to
mechanical stretch. This was confirmed in vivo by administration of a neutralizing transforming growth factor-␤
antibody, which effectively prevented thrombomodulin downregulation during acute pressure overload.
Conclusions—These findings suggest that increased hemodynamic load adversely affects endocardial function and is a
potentially important contributor to thromboembolus formation in heart failure. (Circulation. 2007;115:67-75.)
Key Words: endocardium 䡲 heart failure 䡲 thrombosis 䡲 thrombomodulin

C ongestive heart failure is a growing public health


problem that affects more than 5 million people in the
United States. Thromboembolic events, including stroke,
Clinical Perspective p 75
binds thrombin and renders it incapable of enzymatically
are a major cause of morbidity and mortality in patients cleaving fibrinogen or activating cellular thrombin receptors,
with heart failure.1 The annual risk of a thromboembolic but enables the activation of circulating protein C. Activated
event with mild to moderate heart failure and sinus rhythm protein C (APC) degrades factors Va and XIIIa of the
ranges from 1.5% to 2.5% and exceeds 5% in severe coagulation cascade, potently inhibiting further thrombin
generation. Deletion of the thrombomodulin gene causes
disease.2,3 Thromboembolus formation in heart failure has
lethal thrombosis in mice, and the acquired loss of thrombo-
historically been ascribed to blood stasis within the dilated
modulin in humans is thought to contribute to the thrombotic
cardiac chambers.4 Stasis alone, however, cannot fully
manifestations of bacterial sepsis, radiation enteropathy, and
explain intracardiac thrombus formation, as even patients
coronary atherosclerosis.9 –11
with heart failure and preserved systolic function remain at
We recently identified pressure-induced vascular stretch as
increased risk for stroke compared with those without a novel and potent inhibitory stimulus for endothelial throm-
heart failure.5 Although recent evidence does suggest that bomodulin expression.12 This was first observed in rabbit
heart failure may be associated with a mild degree of vein segments implanted into the arterial circulation; throm-
hypercoagulation,6 what has received relatively little at- bomodulin protein expression decreased by 95% and resulted
tention and yet may be a significant contributor to intra- in increased local thrombin generation and microthrombus
cardiac thrombus formation is the third arm of Virchow’s formation.13 On the basis of these studies, we hypothesize
triad— dysfunction of the endocardial endothelium. that pressure-induced chamber stretch might also negatively
Thrombomodulin is a 100-kDa membrane glycoprotein regulate endocardial thrombomodulin expression and contrib-
that is a major regulator of vascular thromboresistance.7 It is ute to intracardiac thrombus formation during heart failure.
expressed in abundance by vascular endothelial cells, includ- To explore this, we characterized endocardial thrombomodu-
ing those that comprise the endocardium.8 Thrombomodulin lin expression and function in a rat model of acute pressure

Received April 3, 2006; accepted October 13, 2006.


From the Department of Medicine, Division of Cardiology of the Johns Hopkins School of Medicine, Baltimore, Md.
The online-only Data Supplement, consisting of supplemental Methods, Figure I, and Figure II, is available with this article at
http://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.106.640698/DC1.
Correspondence to Jeffrey J. Rade, MD, Division of Cardiology, Johns Hopkins School of Medicine, Carnegie 568, 600 N. Wolfe St, Baltimore, MD
21287. E-mail jjrade@jhmi.edu
© 2007 American Heart Association, Inc.
Circulation is available at http://www.circulationaha.org DOI: 10.1161/CIRCULATIONAHA.106.640698

67
Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015
68 Circulation January 2/9, 2007

overload. We then used adenovirus-mediated gene transfer to expression in cultured cells and TGF-␤ gene expression in both
determine the relationship between thrombomodulin function tissue samples and cultured cells were normalized to 18S ribosomal
and endocardial thromboresistance and identified an impor- RNA (TaqMan Ribosomal RNA Reagents with VIC-labeled probe;
Applied Biosystems).
tant role of paracrine effects of transforming growth factor-␤
(TGF-␤) in regulation of endocardial thrombomodulin Western Blot and Immunohistochemical Analyses
expression. Western blot analysis was performed with anti-thrombomodulin
antibodies (#3381 and #2380; American Diagnostica) and an anti-
Methods CD31 antibody (M0823; Dako, Carpinteria, Calif) as described.13
Bands were detected by autoradiography and quantified densito-
Rat Model of Pressure Overload metrically with UN-SCAN-IT software (Silk Scientific, Orem,
Animal protocols were approved by the Johns Hopkins Animal Care Utah). Immunohistochemical analysis was performed on formalin-
and Use Committee. A well-characterized rat model was used with fixed and paraffin-embedded sections of rat left atria with antibodies
modification.14 Six-week-old male Wistar rats that weighed ⬇100g against rat thrombomodulin (#3381; American Diagnostica), human
(Charles River Laboratories, Wilmington, Mass) were anesthetized TGF-␤ latent associated peptide (EF01; R&D), and rat prolyl
with 1% to 2% isoflurane and mechanically ventilated. Through a 4-hydoxylase (6-9H6; Acris Antibodies, Hiddenhausen, Germany) as
right thoracotomy, a 23-gauge needle was placed adjacent to the described.8
ascending aorta and a 4-0 silk suture ligature was applied around
both. The needle was then removed to create an immediate ⬎70% In Situ Protein C and Thrombin Activity Assays
luminal stenosis. Sham control animals underwent thoracotomy
Capacity to generate APC was measured on freshly-harvested whole
without suture placement. To investigate the effects of TGF-␤, rats
left atria with a previously described protocol with modifiction.13
were administered 1 mg/kg of either a neutralizing anti–TGF-␤
antibody (MAB240; R&D Systems, Minneapolis, Minn) or an IgG1 The atria were washed in Hank’s buffered salt solution and then
isotype control antibody (MAB005; R&D Systems) via peritoneal incubated in 250 ␮L Hank’s buffered salt solution that contained 40
injection 24 hours before surgery, with a second dose of 0.5 mg/kg nmol/L human ␣-thrombin (Sigma-Aldrich, St Louis, Mo) and 1
administered 48 hours after surgery. The antibody dose was based on ␮mol/L human protein C (American Diagnostica) at 37°C. After 60
previous studies in the rat that demonstrated effective neutralization minutes, thrombin was neutralized by excess lepirudin (Berlex
of TGF-␤.15 Laboratories, Montville, NJ). Aliquots measuring 100 ␮L were
At euthanization, echocardiographic measurements were obtained incubated at room temperature with a 3-mmol/L solution of the
with a Sonos 550 Ultrasound System (Hewlett-Packard, Andover, chromogenic substrate S-2366 (DiaPharma, West Chester, Ohio).
Mass) equipped with a 15-MHz probe (Acuson, Siemens, Malvern, The rate of substrate conversion was determined spectrophotometri-
Pa) with validated techniques in rodents.16 Ventricular pressures cally with a Vmax Kinetic Microplate Reader (Molecular Devices,
were measured directly via cardiac puncture with a 21-gauge needle Sunnyvale, Calif) and the amount of protein C activation calculated
attached to a pressure monitor (SpaceLabs, Redmond, Wash). by comparison with a human APC (American Diagnostica) standard
curve. Bound thrombin activity on the atrial endocardial surface was
Atrial Gene Transfer measured as previously described with some modification.13 Freshly
excised whole atria were washed in Hank’s buffered salt solution,
The construction of adenovirus vectors that express human throm-
then incubated in 250 ␮L of substrate buffer (50 mmol/L Tris-HCl,
bomodulin (AdTMh5) and no transgene (AdNull) have been previ-
175 nmol/L NaCl, and 2 mmol/L CaCl2 [pH 7.8] that contained 333
ously described.13 The left atrium was exposed via a left thoracot-
omy and 400 ␮L of a solution of 20% Pluronic F-127 (Molecular ␮mol/L of the chromogenic substrate, S-2238 (DiaPharma) at 37°C
Probes, Eugene, Ore), 0.5% trypsin, and 1010 plaque-forming for 30 minutes. The change in absorbance at 405 nm before and after
units/mL of the indicated adenovirus vector were directly applied to lepirudin treatment represented thrombin-specific substrate conver-
the epicardial surface of the left atrium with a camel-hair paintbrush. sion. Bound thrombin activity was then calculated by comparison
The applied solution was allowed to gel for 10 minutes before with a human ␣-thrombin standard curve.
closure of the thoracotomy. Five days after atrial gene transfer, the
ascending aorta was banded as described. The expression of recom- In Vitro Stretch Experiments
binant human thrombomodulin protein in transduced atria was Human atrial endocardial cells isolated in our laboratory (see Data
assessed in tissue lysates by ELISA as described.13 Supplement) and human cardiac fibroblasts purchased from Cell
Applications (San Diego, Calif) of passage 2 to 3 were plated onto
Assessment of Neurohormonal Activation type I collagen-coated 6-well Bioflex plates (Flexcell International,
At the time of euthanization, levels of angiotensin II, atrial natriuretic Hillsborough, NC) and grown in either EGM-2 (endocardial cells) or
peptide, and brain natriuretic peptide were determined in extracted FGM-2 (fibroblasts) culture media (BioWhittaker). When nearly
serum with EIA kits that recognize rat peptides (Phoenix Pharma- confluent, cells were refreshed in basal media and subjected to 0% to
ceuticals, Belmont, Calif) in accordance with the manufacturer’s 10% cyclic strain delivered at 1 Hz for 24 hours at 37°C and 5% CO2
instructions. Serum tumor necrosis factor-␣ concentrations were with a FX-4000T Tension Plus System (Flexcell International). For
measured by rat tumor necrosis factor-␣ ELISA kit (BioSource, co-culture experiments, atrial endocardial cells were grown on
Camarillo, Calif) in accordance with the manufacturer’s instructions. 24-mm diameter Transwell inserts with a 0.4-␮m pore size (Corning,
Corning, NY) in EGM-2 medium. When confluent, the inserts were
Real-Time Quantitative Polymerase Chain Reaction placed into the individual wells of the Bioflex plates with cardiac
RNA from freshly harvested tissue or cultured cells was extracted fibroblasts in basal medium (BioWhittaker). The cardiac fibroblasts
using TRIZOL Reagent (Invitrogen, Carlsbad, Calif). Real-time were then subjected to 0% to 10% cyclic strain at 1 Hz for 24 hours.
quantitative polymerase chain reaction (PCR) was performed with a To investigate the effects of TGF-␤, 0.5 ␮g/mL of either a neutral-
7900HT Sequence Detection System (Applied Biosystems, Foster izing anti–TGF-␤ (MAB240; R&D) or IgG1 isotype control anti-
City, Calif). Triplicate samples were subjected to reverse transcrip- body (MAB005; R&D) were added to the medium before stretching
tion and real-time PCR with TaqMan One-Step RT-PCR Master Mix the fibroblasts at 10% cyclic strain delivered at 1 Hz for 24 hours.
Reagents with gene-specific primers and probes designed by Primer
Express software (Applied Biosystems) based on published nucleo- Statistical Analysis
tide sequences (see Data Supplement). Thrombomodulin gene ex- All data are presented as mean⫾SEM. Comparison between 2
pression was normalized to endothelial-specific RNA content by groups is by 2-tailed t tests and between multiple groups is by 1-way
quantification of rat CD31 gene expression. Thrombomodulin gene ANOVA with a Bonferroni correction for intergroup comparisons.

Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015


Kapur et al Endocardial Thromboresistance 69

Figure 1. Effects of suture banding of the ascending aorta 96 hours after surgery. A, Left ventricular systolic and diastolic pressures
measured by cardiac puncture (n⫽8 per group). B, Left atrial (LA) diameter, left ventricular diastolic posterior wall thickness (LVPWd),
and left ventricular internal diastolic diameter (LVIDd) as measured by echocardiography (n⫽8 per group). C, Serum levels of atrial na-
triuretic peptide (ANP; n⫽6 to 8 per group), brain natriuretic peptide (BNP; n⫽7 to 8 per group), angiotensin II (AT II; n⫽7 to 8 per
group), and tumor necrosis factor-␣ (TNF-␣; n⫽3 to 4 per group).

Statistical relationships not otherwise indicated are to be assumed confirmed a 70% decline in thrombomodulin expression
nonsignificant (P⬎0.05). (P⬍0.002; Figure 2B). Left atrial tissue stained with an
The authors had full access to and take responsibility for the
integrity of the data. All authors have read and agree to the
anti-mouse thrombomodulin antibody verified decreased
manuscript as written. thrombomodulin expression by the atrial endocardial en-
dothelium (Figure 2C). These data suggest that endocardial
Results thrombomodulin expression is directly modulated by
Effects of Acute Pressure Overload on changes in chamber loading rather than by systemic factors
Endocardial Thrombomodulin Expression induced by acute heart failure.
The ascending aortae of 6-week-old rats were suture- We also determined whether acute pressure overload
banded to induce a ⬎70% luminal constriction. Because altered the atrial expression of other molecules known to
this degree of acute increase in afterload results in pulmo- modulate endocardial thromboresistance (Figure 3). Aortic
nary edema and death within 7 days of surgery, all banding did not alter the expression of endothelial nitric
hemodynamic and echocardiographic measurements were oxide synthase but was associated with increased tissue
obtained 96 hours after banding. Compared with sham- factor pathway inhibitor and decreased tissue factor ex-
operated controls, left ventricular systolic pressure in- pression. The net effect of these changes would be ex-
creased 40% in banded animals, whereas left ventricular pected to reduce local thrombin generation at the endocar-
diastolic pressure, equivalent to mean left atrial pressure, dial surface. Interestingly, banding also increased the
increased by nearly 400% (P⬍0.0001 for both; Figure 1A). expressions of both tissue plasminogen activator and
Ventricular wall thickness and diastolic diameter were plasminogen activator inhibitor-1.
unchanged, whereas left atrial diameter increased slightly
(Figure 1B). Systolic function was not depressed in this Consequences of Thrombomodulin
acute period, with slight increases in ejection fractions Downregulation on Endocardial
(77⫾9% banded versus 65⫾5% sham controls, P⬍0.01), Thromboresistance
which likely reflect acute compensation to high after- Thrombomodulin exerts its anticoagulant effect via acti-
load.17 Serum atrial natriuretic peptide and brain natriuret-
vation of circulating protein C. Endocardial capacity to
ic peptide levels were ⬎3-fold higher in banded rats,
generate APC was measured in resected whole left atria 96
consistent with the induction of heart failure, whereas
hours after surgery. Aortic banding reduced the capacity to
angiotensin II and tumor necrosis factor-␣ levels did not
generate APC in the left atria of banded rats by ⬎35%
differ from controls (Figure 1C).
compared with sham controls (P⫽0.01; Figure 4A). To
To determine the impact of acute hemodynamic changes
on endocardial thrombomodulin expression, tissue from determine the effect on in situ thrombin generation, the
the left atrium and ventricle of banded rats was subjected activity of thrombin bound to the atrial endocardial surface
to quantitative PCR analysis and compared with sham was then quantified. Thrombin generated at sites of vas-
controls (Figure 2A). Aortic tissue, distal to the suture cular injury binds to fibrin strands within a developing clot
ligature, was analyzed as an additional control. Data were and is protected from inactivation by circulating inhibitors.
normalized to CD31, an endothelial-specific adhesion Bound thrombin activity is therefore proportional to the
molecule whose expression does not change with pressure- degree of thrombus that is present and is capable of
induced cell stretch.18 Thrombomodulin expression in the detecting the presence of microscopic amounts of fibrin
left atrium of banded rats declined by 60% compared with clot.12 Bound thrombin activity in the atria of banded rats
sham controls (P⫽0.01), whereas thrombomodulin expres- was significantly higher than in atria from sham-operated
sion in the left ventricle or in the distal aorta was controls and approached that observed in atria subjected to
unchanged. Western blot analysis of left atrial tissue direct mechanical forcep injury (Figure 4B).
Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015
70 Circulation January 2/9, 2007

Figure 2. Effect of acute pressure overload on endocardial thrombomodulin (TM) expression. A, Thrombomodulin gene expression, nor-
malized to CD31, was determined by quantitative PCR of tissue extracts obtained from the left atrium (n⫽10 per group), left ventricle
(n⫽10 per group), and the distal aorta (n⫽8 per group) of banded and sham rats 96 hours after surgery. B, Thrombomodulin protein
expression, normalized to CD31, was determined by densitometric analysis of Western blots of left atrial tissue extracts obtained from
banded and sham rats (n⫽4 to 5 per group) 96 hours after surgery. The right panel depicts a representative Western blot. C, Histologi-
cal sections of left atrial tissue obtained from sham and banded rats 96 hours after surgery and stained with an anti-mouse thrombo-
modulin antibody that shows decreased endocardial thrombomodulin protein expression.

We then determined the effects of thrombomodulin average of 76.5⫾24.9 ng of human thrombomodulin per
expression restoration on capacity to generate APC and milligram of atrial tissue in AdTMh5-transduced atria
bound thrombin activity. This was accomplished with a (n⫽4), whereas no human thrombomodulin was detected
“gene painting” technique, in which solutions that contain in AdNull-transduced atria (n⫽3; P⬍0.004). These results
adenovirus vectors that express either human thrombo- were confirmed by Western blot analysis (Figure 4C). To
modulin (AdTM5) or no transgene (AdNull) were applied verify that acute pressure overload does not attenuate
to the epicardial surfaces of the rat left atria. Previous transgene expression, transduced atria from suture-banded
studies in the pig revealed that this technique results in rats were assayed for the expression of both human and
efficient transmural transduction of atrial cells, including native rat thrombomodulin by quantitative PCR 96 hours
the endocardial endothelium.19 Figure I illustrates the after surgery. Figure 4D demonstrates that human throm-
bomodulin gene expression remains robust throughout the
degree of gene transfer that can be attained with this
experimental period in AdTMh5-transduced atria, whereas
method in the rat. Three days after transduction, protein
no human thrombomodulin gene expression could be
lysates of left atrial tissue subjected to ELISA that detects
detected in AdNull-transduced atria. Restoration of throm-
only the human form of thrombomodulin13 revealed an
bomodulin expression with adenovirus-mediated gene
transfer effectively prevented the loss of capacity to
generate APC induced by aortic banding (Figure 4A) and
reduced the levels of bound thrombin activity to baseline
values (Figure 4B). Taken together, these data support the
concept that downregulation of endocardial thrombomodu-
lin contributes to increased local thrombin generation.

Effects of Stretch on Thrombomodulin Expression


in Isolated Endocardial Endothelial Cells
We have previously shown that thrombomodulin down-
regulation in vein segments exposed to arterial pressure
Figure 3. Effect of acute pressure overload on the expression of
molecules that modulate endocardial thromboresistance. Gene results from pressure-induced vascular stretch and not
expression, normalized to CD31, was determined by quantitative from the direct effects of pressure per se.12 To determine
PCR of tissue extracts obtained from the left atrium of banded whether stretch can directly inhibit thrombomodulin gene
and sham rats 96 hours after surgery (n⫽10 per group). eNOS expression, endocardial endothelial cells isolated from
indicates endothelial nitric oxide synthase; TF, tissue factor;
TFPI, tissue factor pathway inhibitor; tPA, tissue plasminogen human right atrial appendages were grown on collagen-
activator; and PAI-1, plasminogen activator inhibitor-1. coated silastic membranes and subjected to increasing
Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015
Kapur et al Endocardial Thromboresistance 71

Figure 4. Effects of thrombomodulin expres-


sion on endocardial thromboresistance. A,
Capacity to generate APC was assessed 96
hours after surgery in the left atrium of
sham-operated and banded rats (n⫽10 per
group) as well as in banded rats whose left
atria were transduced with either AdNull or
AdTMh5 5 days before surgical banding
(n⫽5 per group). B, Bound thrombin activity,
proportional to microthrombus burden, was
measured 96 hours after surgery in the left
atria of sham-operated and banded rats
(n⫽10 per group) as well as in banded rats
whose left atria were transduced with either
AdNull or AdTMh5 5 days before surgical
banding (n⫽5 per group). C, Rat LA were
transduced with either AdNull (n⫽5) or
AdTMh5 (n⫽8) 5 days before surgical band-
ing; 96 hours after banding, thrombomodulin
gene expression was determined in atrial
tissue by quantitative PCR with species-
specific probes and primers. D, Rat left atria
were transduced with either AdNull or
AdTMh5, an adenovirus vector expressing
human thrombomodulin. Three days after
transduction, atrial tissue was subjected to
Western blot analysis with antibodies that
recognize the human isoform of thrombo-
modulin and rat CD31. The depicted blot is
representative of 3 experiments.

amounts of cyclic stretch for 24 hours. Surprisingly, cyclic Modulation of Thrombomodulin Expression by
stretch resulted in a trend toward increased thrombomodu- Paracrine Release of TGF-␤
lin gene expression (Figure 5A). Similar results were
TGF-␤ is a multifunctional stretch-induced growth factor
observed in stretched endothelial cells isolated from both
rabbit jugular veins and human umbilical veins (data not known to be intimately involved in cardiac remodeling
shown). Because endocardial endothelial cells in situ lie induced by heart failure20 and has been reported to inhibit
adjacent to cardiac connective tissue, we next tested thrombomodulin protein expression in human umbilical
whether this interaction might influence thrombomodulin vein endothelial cells.21 TGF-␤ gene expression in cardiac
expression by stretch. Human atrial endocardial cells were fibroblasts exhibited a dose-dependent increase in re-
cultured on stationary filters suspended in the media of sponse to cyclic stretch (Figure 6A). In addition, throm-
wells that contain human cardiac fibroblast subjected to bomodulin gene expression in human atrial endocardial
increasing amounts of cyclic stretch. Stretching of cardiac cells was inhibited in a dose-dependent manner after
fibroblasts resulted in a dose-dependent decrease in throm- 24-hour exposure to recombinant TGF-␤1 (Figure 6B). To
bomodulin expression in the stationary endocardial cells determine whether TGF-␤ is the paracrine factor respon-
(Figure 5B). Experiments that substituted human aortic sible for thrombomodulin downregulation, human endo-
smooth cells for cardiac fibroblasts yielded identical re- cardial cells were plated on stationary filters submerged in
sults (data not shown). These results indicate that endo- the media of cardiac fibroblasts subjected to 10% cyclic
cardial thrombomodulin expression is modulated in para- stretch for 24 hours in the presence of a neutralizing
crine fashion by a soluble factor released by cardiac anti–TGF-␤ antibody or an isotype control antibody.
connective tissue in response to stretch. Figure 6C shows that neutralization of TGF-␤ effectively

Figure 5. Effect of cyclic stretch on endocar-


dial thrombomodulin expression. A, Human
atrial endothelial cells were plated on
collagen-coated Bioflex plates and subjected
to increasing amounts of cyclic stretch at 1
Hz for 24 hours. Thrombomodulin gene
expression, normalized to rRNA, was deter-
mined by quantitative PCR (n⫽5 per group).
B, Human atrial endocardial cells were
plated on stationary filters, submerged in the
media of human cardiac fibroblasts, and
subjected to increasing amounts of cyclic
stretch at 1 Hz for 24 hours. Thrombomodu-
lin gene expression, normalized to rRNA,
was determined by quantitative PCR (n⫽4
per group).

Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015


72 Circulation January 2/9, 2007

Figure 6. Effect of TGF-␤ on endocardial thrombomodulin expression. A, Human cardiac fibroblasts were subjected to increasing
amounts of cyclic stretch for 24 hours. TGF-␤ gene expression, normalized to rRNA, was determined by quantitative PCR (n⫽6 per
group). B, Human atrial endocardial cells were incubated with increasing concentrations of recombinant human TGF-␤1 for 24 hours.
Thrombomodulin gene expression, normalized to rRNA, was determined by quantitative PCR (n⫽4 per group). C, Human atrial endo-
cardial cells were plated on stationary filters, submerged in the media of human cardiac fibroblasts, and subjected to 10% cyclic
stretch for 24 hours in the presence of 0.5 ␮g/mL of neutralizing TGF-␤ or isotype control antibodies. Thrombomodulin gene expres-
sion, normalized to rRNA, was determined by quantitative PCR (n⫽4 per group).

prevented the downregulation of endocardial thrombo- downregulation, rats were administered either a neutraliz-
modulin gene expression. ing anti–TGF-␤ antibody or an isotype control antibody in
To determine whether paracrine release of TGF-␤ is the perioperative period. Neutralization of TGF-␤ effec-
responsible for in vivo modulation of thrombomodulin, we tively prevented thrombomodulin downregulation in the
first assessed TGF-␤ expression in the cardiovascular left atrium 96 hours after aortic banding (Figure 7C).
tissue of rats subjected to aortic banding. Compared with Importantly, there was no meaningful difference in the
sham-operated controls, TGF-␤ gene expression in the left hemodynamic response to banding between rats adminis-
atrium of banded rats increased by ⬎200% (P⬍0.001), tered the anti–TGF-␤ antibody versus the isotype control
whereas there was no significant change in TGF-␤ expres- antibody (left ventricular end-diastolic pressure 17.7⫾0.6
sion in the left ventricle or in the distal aorta (Figure 7A). versus 15.8⫾0.2 mm Hg, respectively; P⫽0.02). These
Banding was also associated with increased TGF-␤ acti- data confirm that the effects of pressure overload on
vation, as evidenced by positive immunostaining for endocardial thrombomodulin expression and thromboresis-
latency-associated peptide, the cleaved propeptide of the tance are mediated via paracrine release of TGF-␤.
TGF-␤ precursor molecule that remains noncovalently
attached to active TGF-␤. Latency-associated peptide Discussion
staining localized predominantly to fibroblast-appearing The major findings of this study are (1) acute elevations in
cells in the subendocardial space that also expressed an filling pressure adversely affect atrial endocardial throm-
abundance of the prolyl 4-hydroxylase, an enzyme in- bomodulin expression; (2) downregulation of endocardial
volved in collagen synthesis (Figure 7B). To determine thrombomodulin expression impairs local protein C acti-
whether TGF-␤ inhibition could prevent thrombomodulin vation and contributes to local thrombin generation; and

Figure 7. Effect of TGF-␤ on endocardial


thrombomodulin expression in heart failure.
A, TGF-␤ gene expression, normalized to
rRNA, was determined by quantitative PCR
of tissue extracts obtained from the left
atrium (n⫽10 per group), left ventricle (n⫽8
per group), and the distal aorta (n⫽8 per
group) of banded and sham-operated rats
96 hours after surgery. B, Sham and banded
atria were immunostained for TGF-␤ activa-
tion with an antibody that recognized the
cleaved latency associated peptide (LAP).
TGF-␤ activation localized to fibroblast-
appearing cells in the subendocardial space
that abundantly express rat prolyl
4-hydroxylase (rPH). C, Rats were adminis-
tered 1 mg/kg of either a neutralizing anti–
TGF-␤ or isotype control antibody 24 hours
before aortic banding and a second dose of
0.5 mg/kg 48 hours postoperatively. Throm-
bomodulin gene expression, normalized to
CD31, was determined 96 hours after sur-
gery by quantitative PCR of left atrial tissue
extracts and compared with sham-operated
controls (n⫽6 per group).

Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015


Kapur et al Endocardial Thromboresistance 73

(3) endocardial thrombomodulin expression is negatively observed in the left atrium but not in the left ventricle and
regulated during acute pressure overload by the paracrine appeared to originate predominantly from cardiac fibro-
effects of TGF-␤ secreted by cardiac connective tissue in blasts. This was likely the result of the relatively greater
response to pressure-induced chamber stretch. hemodynamic load imposed on the atrium and explains
It has long been recognized that patients with heart why endocardial thrombomodulin expression in the ven-
failure develop intracardiac thrombi that cause cerebral, tricle did not change. A recognized limitation of this model
pulmonary, and peripheral arterial thromboembolization. is the absence of ventricular remodeling and systolic
Thrombus formation has traditionally been viewed as a dysfunction common to most chronic heart failure models.
consequence of blood stasis that results from impaired Although the model mostly replicates features of diastolic
ventricular function. If the presence of atrial fibrillation is heart failure, it was precisely the absence of systolic
excluded, however, the degree of left ventricular dysfunc- dysfunction that obviated potential confounding effects of
tion only weakly correlates with thromboembolic risk.3 blood stasis on intracardiac thrombin generation. In mod-
This suggests that factors other than stasis contribute to els of chronic systolic dysfunction, TGF-␤ expression is
intracardiac thrombus formation in patients with heart increased in ventricular tissue,26 and it is reasonable to
failure, of which the overwhelming majority remain in predict that thrombomodulin expression in the overlying
sinus rhythm. We believe that ours is the first study to endocardium would also be reduced. Adaptive responses,
convincingly demonstrate that elevated cardiac filling however, may complicate this issue, as there is evidence
pressures can adversely affect endocardial function and that myocardial TGF-␤ expression may vary according to
predispose to intracardiac thrombus formation independent the cause of heart failure and may change over time as
of blood stasis. heart failure progresses.27,28 This raises the possibility that
Thrombomodulin is but 1 of several anticoagulant mol- the risk and chamber origin of thrombus formation may
ecules expressed by endothelial cells that protect against change over time, and the risk may be greater for different
pathological thrombosis. The level of thrombomodulin types of heart failure. With the observed association
expression varies among endothelial cell types and there- between TGF-␤ expression and endocardial dysfunction as
fore its relative contribution to vascular thromboresistance a conceptual framework, future studies that use chronic
may differ between vascular beds.22,23 Several elements of models of systolic heart failure can be used to investigate
the present study suggest that thrombomodulin is a critical these possibilities.
contributor to endocardial thromboresistance. The first is Extremely little is known about the molecular mecha-
that thrombomodulin is expressed in abundance by the nism by which TGF-␤ regulates thrombomodulin expres-
endocardial endothelium. In addition to the rat, we have sion in endothelial cells. TGF-␤ signal transduction is
also found that in situ endocardial thrombomodulin expres- initiated via binding to specific serine/threonine type I and
sion is also robust in the rabbit8 and in humans (Figure II). type II receptor complexes located on the endothelial cell
Second, increased local thrombin generation in our model surface.29 Receptor engagement causes the phosphoryla-
was proportional to the downregulation of thrombomodu- tion of several intracellular effector molecules known as
lin and occurred despite changes in the expression of other Smads. Whereas most of the Smad proteins positively
anticoagulant molecules that would be expected to inhibit regulate TGF-␤ signal transduction, Smad6 and Smad7 are
thrombus formation. Third, and most important, targeted known to exert inhibitory effects. In the only published
restoration of thrombomodulin expression with study to date that investigates the mechanism of thrombo-
adenovirus-mediated gene transfer effectively reduced lo- modulin modulation by TGF-␤, antisense inhibition of
cal thrombin generation. Smad7 potentiated the effects of TGF-␤, whereas antisense
In prior studies with rabbit vein grafts, we identified inhibition of a splice variant of Smad6, known as Smad6s,
pressure-induced vascular stretch as a novel and potent unexpectedly blunted the downregulation of thrombo-
negative regulator of in vivo thrombomodulin expression. modulin in human umbilical endothelial cells by TGF-␤.30
The present study extends these findings by providing As Smad6 and Smad7 are differentially expressed in the
evidence of a second, and uniquely important, vascular bed endothelium of normal and atherosclerotic arteries,30 they
in which stretch modulates thrombomodulin expression may be able to differentially modulate the paracrine effects
and by identifying paracrine release of TGF-␤ as the of TGF-␤ on thrombomodulin expression in various vas-
critical molecular mediator. TGF-␤ is a multifunctional cular beds and disease states. What remains completely
dimeric polypeptide growth factor involved in a diverse unknown at the present time is the nature of the signaling
array of biological processes that include embryogenesis, pathway distal to Smad activation that may regulate
tumor growth, wound healing, and tissue remodeling.24 thrombomodulin gene transcription.
TGF-␤ is intimately involved in the adaptive response of In addition to heart failure, our findings may have
cardiac and vascular tissue to pressure overload. In systolic implications for understanding the thrombogenic potential
heart failure, for example, TGF-␤ expression is known to of other forms of cardiovascular disease, such as atrial
be markedly increased in cardiac myocytes and fibroblasts, fibrillation. Thrombus formation in atrial fibrillation is
where it is a recognized autocrine and paracrine mediator also primarily ascribed to blood stasis in a left atrial
of hypertrophy and fibrosis.25 appendage that is not contracting. However, it is well
In the model of acute pressure overload used for this known that atrial fibrillation in patients with structurally
study, a marked increase in local TGF-␤ expression was normal hearts, ie, “lone atrial fibrillation,” is associated
Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015
74 Circulation January 2/9, 2007

with a relatively small risk (⬍0.5% per year) of thrombo- 12. Sperry JL, Deming CB, Bian C, Walinsky PL, Kass DA, Kolodgie FD,
embolic stroke compared with atrial fibrillation that occurs Virmani R, Kim AY, Rade JJ. Wall tension is a potent negative
regulator of in vivo thrombomodulin expression. Circ Res. 2003;92:
in the setting of hypertension, heart failure, or valvular 41– 47.
heart disease (5% to 12% per year).31 As these latter 13. Kim AY, Walinsky PL, Kolodgie FD, Bian C, Sperry JL, Deming CB,
conditions are all characterized by pressure-induced atrial Peck JG, Ang GB, Sohn RH, Esmon CT, Virmani R, Stuart RS, Rade
distension, it is possible that endocardial dysfunction and JJ. Early loss of thrombomodulin expression impairs vein graft throm-
boresistance: implications for vein graft failure. Circ Res. 2002;90:
altered thrombomodulin expression may contribute sub- 205–212.
stantially to the thromboembolic risk in these patients. 14. Schunkert H, Dzau VJ, Tang SS, Hirsch AT, Apstein CS, Lorell BH.
This concept is supported by data from a rat model of atrial Increased rat cardiac angiotensin converting enzyme activity and
mRNA expression in pressure overload left ventricular hypertrophy.
fibrillation in which atrial thrombomodulin expression
Effects on coronary resistance, contractility, and relaxation. J Clin
decreased by 35% after 8 hours of rapid atrial pacing.32 Invest. 1990;86:1913–1920.
Interestingly, the left atrial pressure in this model triples 15. Koyanagi M, Egashira K, Kubo-Inoue M, Usui M, Kitamoto S, Tomita
during pacing,33 which raises the possibility that the effects H, Shimokawa H, Takeshita A. Role of transforming growth
factor-beta1 in cardiovascular inflammatory changes induced by
on thrombomodulin expression were primarily the result of
chronic inhibition of nitric oxide synthesis. Hypertension. 2000;35:
hemodynamic rather than electrical influences. 86 –90.
16. Litwin SE, Katz SE, Weinberg EO, Lorell BH, Aurigemma GP,
Sources of Funding Douglas PS. Serial echocardiographic-Doppler assessment of left ven-
This work was supported in part by an American Heart Association tricular geometry and function in rats with pressure-overload hyper-
Grant-in-Aid (0555515U) to Dr Rade and by National Heart, Lung trophy: chronic angiotensin-converting enzyme inhibition attenuates
the transition to heart failure. Circulation. 1995;91:2642–2654.
and Blood Institute Grant HL-080142 to Dr Rade. The Flow
17. Sarnoff SJ, Mitchell JH, Gilmore JP, Remensnyder JP. Homeometric
Cytometry Cell Sorting Core Laboratory of the Bloomberg School of
autoregulation in the heart. Circ Res. 1960;8:1077–1091.
Public Health is supported by National Institute of Allergy and
18. Lauth M, Berger MM, Cattaruzza M, Hecker M. Pressure-induced
Infectious Disease Grant 5P30AI042855.
upregulation of preproendothelin-1 and endothelin B receptor
expression in rabbit jugular vein in situ: implications for vein graft
Disclosures failure? Arterioscler Thromb Vasc Biol. 2000;20:96 –103.
None. 19. Kikuchi K, McDonald AD, Sasano T, Donahue JK. Targeted modifi-
cation of atrial electrophysiology by homogeneous transmural atrial
References gene transfer. Circulation. 2005;111:264 –270.
20. Rosenkranz S. TGF-beta1 and angiotensin networking in cardiac
1. Fuster V, Gersh BJ, Giuliani ER, Tajik AJ, Brandenburg RO, Frye RL.
remodeling. Cardiovasc Res. 2004;63:423– 432.
The natural history of idiopathic dilated cardiomyopathy. Am J Cardiol.
21. Ohji T, Urano H, Shirahata A, Yamagishi M, Higashi K, Gotoh S,
1981;47:525–531.
Karasaki Y. Transforming growth factor beta 1 and beta 2 induce
2. Dunkman WB, Johnson GR, Carson PE, Bhat G, Farrell L, Cohn JN.
Incidence of thromboembolic events in congestive heart failure. The down-modulation of thrombomodulin in human umbilical vein endo-
V-HeFT VA Cooperative Studies Group. Circulation. 1993;87: thelial cells. Thromb Haemost. 1995;73:812– 818.
VI94 –VI101. 22. Maruyama I, Bell CE, Majerus PW. Thrombomodulin is found on
3. Dries DL, Rosenberg YD, Waclawiw MA, Domanski MJ. Ejection endothelium of arteries, veins, capillaries, and lymphatics, and on
fraction and risk of thromboembolic events in patients with systolic synciotrophoblast of human placenta. J Cell Biol. 1985;101:363–371.
dysfunction and sinus rhythm: evidence for gender differences in the 23. DeBault LE, Esmon NL, Olson JR, Esmon CT. Distribution of the
studies of left ventricular dysfunction trials. J Am Coll Cardiol. thrombomodulin antigen in the rabbit vasculature. Lab Invest. 1986;
1997;29:1074 –1080. 54:172–178.
4. Meltzer RS, Visser CA, Fuster V. Intracardiac thrombi and systemic 24. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth
embolization. Ann Intern Med. 1986;104:689 – 698. factor beta in human disease. N Engl J Med. 2000;342:1350 –1358.
5. Gottdiener JS, McClelland RL, Marshall R, Shemanski L, Furberg 25. Azhar M, Schultz JJ, Grupp I, Dorn GW, Meneton P, Molin DG,
CD, Kitzman DW, Cushman M, Polak J, Gardin JM, Gersh BJ, Gittenberger-de Groot AC, Doetschman T. Transforming growth
Aurigemma GP, Manolio TA. Outcome of congestive heart failure in factor beta in cardiovascular development and function. Cytokine
elderly persons: influence of left ventricular systolic function. The Growth Factor Rev. 2003;14:391– 407.
Cardiovascular Health Study. Ann Intern Med. 2002;137:631– 639. 26. Hao J, Ju H, Zhao S, Junaid A, Scammell-La Fleur T, Dixon IM.
6. Gibbs CR, Blann AD, Watson RD, Lip GY. Abnormalities of hemo- Elevation of expression of Smads 2, 3, and 4, decorin and TGF-beta
rheological, endothelial, and platelet function in patients with chronic in the chronic phase of myocardial infarct scar healing. J Mol Cell
heart failure in sinus rhythm: effects of angiotensin-converting Cardiol. 1999;31:667– 678.
enzyme inhibitor and beta-blocker therapy. Circulation. 2001;103: 27. Aharinejad S, Krenn K, Paulus P, Schafer R, Zuckermann A, Grimm
1746 –1751. M, Abraham D. Differential role of TGF-beta1/bFGF and ET-1 in
7. Esmon CT. The protein C anticoagulant pathway. Arterioscler graft fibrosis in heart failure patients. Am J Transplant. 2005;5:
Thromb Vasc Biol. 1992;12:135–145. 2185–2192.
8. Deming CB, Kim AY, Bian C, Regard JB, Rade JJ. cDNA cloning of 28. Hein S, Arnon E, Kostin S, Schonburg M, Elsasser A, Polyakova V,
rabbit thrombomodulin and characterization of gene expression in Bauer EP, Klovekorn WP, Schaper J. Progression from compensated
cardiovascular tissue. DNA Seq. 2003;14:399 – 405. hypertrophy to failure in the pressure-overloaded human heart:
9. Faust SN, Levin M, Harrison OB, Goldin RD, Lockhart MS, structural deterioration and compensatory mechanisms. Circulation.
Kondaveeti S, Laszik Z, Esmon CT, Heyderman RS. Dysfunction of 2003;107:984 –991.
endothelial protein C activation in severe meningococcal sepsis. 29. Bertolino P, Deckers M, Lebrin F, ten Dijke P. Transforming growth
N Engl J Med. 2001;345:408 – 416. factor-beta signal transduction in angiogenesis and vascular disorders.
10. Richter KK, Fink LM, Hughes BM, Sung C-C, Hauer-Jensen M. Is the Chest. 2005;128:585S-590S.
loss of thrombomodulin involved in the mechanism of chronicity in 30. Sandusky G, Berg DT, Richardson MA, Myers L, Grinnell BW.
late radiation enteropathy? Radiother Oncol. 1997;44:65–71. Modulation of thrombomodulin-dependent activation of human
11. Laszik ZG, Zhou XJ, Ferrell GL, Silva FG, Esmon CT. Down- protein C through differential expression of endothelial Smads. J Biol
regulation of endothelial expression of endothelial cell protein C Chem. 2002;277:49815– 49819.
receptor and thrombomodulin in coronary atherosclerosis. 31. Hart RG, Halperin JL. Atrial fibrillation and stroke: concepts and
Am J Pathol. 2001;159:797– 802. controversies. Stroke. 2001;32:803– 808.

Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015


Kapur et al Endocardial Thromboresistance 75

32. Yamashita T, Sekiguchi A, Iwasaki YK, Sagara K, Hatano S, Iinuma 33. Yamashita T, Murakawa Y, Hayami N, Fukui E, Kasaoka Y, Inoue M,
H, Aizawa T, Fu LT. Thrombomodulin and tissue factor pathway Omata M. Short-term effects of rapid pacing on mRNA level of
inhibitor in endocardium of rapidly paced rat atria. Circulation. 2003; voltage-dependent K(⫹) channels in rat atrium: electrical remodeling
108:2450 –2452. in paroxysmal atrial tachycardia. Circulation. 2000;101:2007–2014.

CLINICAL PERSPECTIVE
Patients with congestive heart failure, atrial fibrillation, and valvular heart disease are at risk for thromboembolic events,
including stroke. Thromboembolus formation in these patients has historically been attributable to blood stasis within the
cardiac chambers. The present study reveals that elevated cardiac chamber filling pressure, which is common to each of
these conditions, can adversely affect endocardial function and contribute to intracardiac thrombus formation. This may
help explain why patients with heart failure and preserved ventricular function remain at risk for thromboembolic stroke,
whereas patients with atrial fibrillation and structurally normal hearts are at relatively low risk. The present study also
identifies transforming growth factor-␤, released in paracrine fashion by pressure-induced chamber stretch, as a potent
negative regulator of endocardial thromboresistance. Targeted inhibition of the transforming growth factor-␤ signaling
pathway could represent a novel therapeutic alternative for prevention of thromboembolus formation in selected cardiac
patients currently being treated with systemic anticoagulation.

Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015


Hemodynamic Modulation of Endocardial Thromboresistance
Navin K. Kapur, Clayton B. Deming, Sunil Kapur, Ce Bian, Hunter C. Champion, J. Kevin
Donahue, David A. Kass and Jeffrey J. Rade

Circulation. 2007;115:67-75; originally published online December 26, 2006;


doi: 10.1161/CIRCULATIONAHA.106.640698
Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2006 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7322. Online ISSN: 1524-4539

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circ.ahajournals.org/content/115/1/67

Data Supplement (unedited) at:


http://circ.ahajournals.org/content/suppl/2006/12/15/CIRCULATIONAHA.106.640698.DC1.html

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

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation is online at:


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

Downloaded from http://circ.ahajournals.org/ by guest on March 28, 2015

You might also like