ArticlePDF Available

Factor Xa Inhibition with Apixaban Does Not Influence Cardiac Remodelling in Rats with Heart Failure After Myocardial Infarction

Authors:

Abstract and Figures

Background Heart failure (HF) is considered to be a prothrombotic condition and it has been suggested that coagulation factors contribute to maladaptive cardiac remodelling via activation of the protease-activated receptor 1 (PAR1). We tested the hypothesis that anticoagulation with the factor Xa (FXa) inhibitor apixaban would ameliorate cardiac remodelling in rats with HF after myocardial infarction (MI).Methods and ResultsMale Sprague-Dawley rats were either subjected to permanent ligation of the left ascending coronary artery (MI) or sham surgery. The MI and sham animals were randomly allocated to treatment with placebo or apixaban in the chow (150 mg/kg/day), starting 2 weeks after surgery. Cardiac function was assessed using echocardiography and histological and molecular markers of cardiac hypertrophy were assessed in the left ventricle (LV). Apixaban resulted in a fivefold increase in anti-FXa activity compared with vehicle, but no overt bleeding was observed and haematocrit levels remained similar in apixaban- and vehicle-treated groups. After 10 weeks of treatment, LV ejection fraction was 42 ± 3% in the MI group treated with apixaban and 37 ± 2 in the vehicle-treated MI group (p > 0.05). Both vehicle- and apixaban-treated MI groups also displayed similar degrees of LV dilatation, LV hypertrophy and interstitial fibrosis. Histological and molecular markers for pathological remodelling were also comparable between groups, as was the activity of signalling pathways downstream of the PAR1 receptor.ConclusionFXa inhibition with apixaban does not influence pathological cardiac remodelling after MI. These data do not support the use of FXa inhibitor in HF patients with the aim to amend the severity of HF. Graphical Abstract
Content may be subject to copyright.
ORIGINAL ARTICLE
Factor Xa Inhibition with Apixaban Does Not Influence Cardiac
Remodelling in Rats with Heart Failure After Myocardial Infarction
Salva R. Yurista
1
&Herman H. W. Silljé
1
&Kirsten T. Nijholt
1
&Martin M. Dokter
1
&Dirk J. van Veldhuisen
1
&
Rudolf A. de Boer
1
&B. Daan Westenbrink
1
#The Author(s) 2020
Abstract
Background Heart failure (HF) is considered to be a prothrombotic condition and it has been suggested that coagulation factors
contribute to maladaptive cardiac remodelling via activation of the protease-activated receptor 1 (PAR1). We tested the hypoth-
esis that anticoagulation with the factor Xa (FXa) inhibitor apixaban would ameliorate cardiac remodelling in rats with HF after
myocardial infarction (MI).
Methods and Results Male Sprague-Dawley rats were either subjected to permanent ligation of the left ascending coronary artery
(MI) or sham surgery. The MI and sham animals were randomly allocated to treatment with placebo or apixaban in the chow
(150 mg/kg/day), starting 2 weeks after surgery. Cardiac function was assessed using echocardiography and histological and
molecular markers of cardiac hypertrophy were assessed in the left ventricle (LV). Apixaban resulted in a fivefold increase in
anti-FXa activity compared with vehicle, but no overt bleeding was observed and haematocrit levels remained similar in
apixaban- and vehicle-treated groups. After 10 weeks of treatment, LV ejection fraction was 42 ± 3% in the MI group treated
with apixaban and 37 ± 2 in the vehicle-treated MI group (p> 0.05). Both vehicle- and apixaban-treated MI groups also displayed
similar degrees of LV dilatation, LV hypertrophy and interstitial fibrosis. Histological and molecular markers for pathological
remodelling were also comparable between groups, as was the activity of signalling pathways downstream of the PAR1 receptor.
Conclusion FXa inhibition with apixaban does notinfluence pathological cardiac remodelling after MI. These datado not support
the use of FXa inhibitor in HF patients with the aim to amend the severity of HF.
Keywords Anticoagulant .Heart failure .Cardiac function .Cardiac remodelling
Introduction
Heart failure is a major global health problem that is reaching
epidemic proportions in the near future [1,2]. Despite the large
range of pharmacological and device-based therapies available,
mortality and morbidity remain high [3]. Patients with heart fail-
ure (HF) are at increased risk of stroke and other thromboembolic
events and are also more likely to succumb from these events
[49]. The higher incidence of thromboembolic events is ob-
served both in patients with sinus rhythm with atrial fibrillation
[10,11], suggesting that HF should be considered as a
prothrombotic or hypercoagulable state. Interestingly, coagula-
tion factors such as thrombin and FXa can exert direct effects on
the heart, which are thought to promote inflammation, endothe-
lial dysfunction and maladaptive cardiac remodelling [12].
Anticoagulants could therefore offer therapeutic benefits in HF
patients beyond the prevention of thromboembolic events [10].
The effects of thrombin and FXa on myocardial tissues are
thoughttobegovernedbyprotease-activatedreceptors
(PARs), which coordinate a myriad of cellular responses in
multiple cell types. PAR1 and PAR2 are expressed in cardiac
tissue and it has been proposed that these receptors contribute
to the progression of HF [1315]. Indeed, cardiomyocyte-
specific overexpression of PAR1 induces cardiac hypertrophy
Electronic supplementary material The online version of this article
(https://doi.org/10.1007/s10557-020-06999-7) contains supplementary
material, which is available to authorized users.
*B. Daan Westenbrink
b.d.westenbrink@umcg.nl
1
Department of Cardiology, University Medical Center Groningen,
University of Groningen, PO Box 30.001, Groningen 9700 RB,
The Netherlands
https://doi.org/10.1007/s10557-020-06999-7
Published online: 26 May 2020
Cardiovascular Drugs and Therapy (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
that rapidly progressed into dilated cardiomyopathy [16].
Conversely, deletion of the PAR1 receptor attenuates cardiac
remodelling after a myocardial infarction (MI) in mice [16].
Nevertheless, direct evidence for enhanced activity of PARs
in HF is sparse and the exact role of PAR signalling in cardiac
remodelling remains poorly defined. Furthermore, it is un-
known whether PAR receptor activation in HF is amendable
by anticoagulant therapy. In this study, we tested the hypoth-
esis that inhibition of PAR signalling by the direct FXa inhib-
itor apixaban could attenuate cardiac remodelling in rats with
established LV dysfunction after MI.
Methods
Experimental Protocol
Male Sprague-Dawley rats (Envigo, The Netherlands) were
randomized to treatment with chow containing apixaban or
control chow, starting 2 weeks after MI surgery. We chose
this protocol to represent a population of stable chronic HF
after a large myocardial infarction, which still represents the
majority of patients with chronic HF. Here, we examined the
effects of clinically relevant doses of apixaban on cardiac re-
modelling in rats with HF after MI [17]. Treatment allocation
was stratified according to left ventricular ejection fraction
(LVEF) to ensure that the baseline cardiac function is similar
in the apixaban and the vehicle groups. After 10 weeks of
treatment, rats were anaesthetized, blood was drawn and the
hearts were rapidly excised for further analysis. Rats with an
infarct size of less than 15% were excluded from analysis as
these small infarcts are haemodynamically fully compensated
[18]. The experimental protocol is illustrated in Scheme 1.
Ethical Statement
The experimental protocol was approved by the Animal Ethical
Committee of the University of Groningen (IvD number: 16487-
02-001). The investigation conforms to the Guide for the Care
and Use of Laboratory Animals published by the US National
Institutes of Health (NIH publication no. 8523, revised 1996).
We followed ARRIVE guidelines when reporting this study.
MI Surgery
Rats were randomized to HF or sham surgery under isoflurane
(2.5%) inhalation anaesthesia. After left-sided thoracotomy,
HF was induced by permanent ligating of the proximal portion
of the left coronary artery as previously described [19]. Sham-
operated rats underwent the same procedure but without cor-
onary ligation.
Investigational Drug
Apixaban was kindly supplied by Bristol-Myers Squibb
(BMS), USA. Apixaban was mixed with standard rat chow
(R/M-H V1534-70, Ssniff, Germany) in a final concentration
of 1.95 g/kg intended to reach an average dose of 150 mg/kg.
Standard rat chow (R/M-H V1534-70, Ssniff, Germany) was
used as the control (vehicle).
Echocardiography
Two weeks after surgery and 1 week before termination, the
M-mode and 2D echocardiography were performed using a
Vivid 7 echo machine (GE Healthcare) equipped with a 10-
MHz phase array linear transducer for serial assessment of
cardiac structure and function as previously described [19].
Invasive Haemodyamic Measurements
Prior to sacrifice, invasive haemodynamics were analysed by
aortic and LV catheterization as previously described [19]. The
right carotid artery was isolated and punctured and a 1.9-F rat
pressure-volume catheter (Scisense, London, Ontario, Canada)
was inserted into the right carotid artery. The tip of the catheter
was advanced through the aorta into the LV cavity. Heart rate
(HR), left ventricular end-systolic (LVESP) and end-diastolic
(LVEDP) pressures, and maximal rates of increase and decrease
in developed LV pressure (dP/dtmax and dP/dtmin) were deter-
mined. The data were acquired using a PowerLab data acquisi-
tion system (ADInstruments, Colorado Springs, CO) and
analysed with a LabChart 8 software.
2 weeks
MI / sham
Surgery Echo Echo
11 weeks 12 weeks0
Male Sprague-
Dawley rats
Sacrifice
Scheme 1 Schematic representation of the experimental protocol
954 Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Infarct Size, Cardiomyocyte Size and Interstitial
Fibrosis Measurement
Rats were euthanized under isoflurane anaesthesia. The heart
was rapidly excised and weighed. The mid-papillary slice of
the LV was fixed in 4% formaldehyde and paraffin-embed-
ded. The infarct size was calculated using midline length
methods as the percentage of the scar length to the total LV
circumference on Massonstrichromestained section, as de-
scribed previously [19,20]. Furthermore, Massonstrichrome
staining was also used to evaluate the extent of interstitial
fibrosis. A Hamamatsu microscope was used to capture the
whole tissue section and an Aperio ImageScope software was
used to quantify fibrosis in the non-infarcted LV [19]. Finally,
sections were stained with FITC-labelled wheat germ agglu-
tinin (WGA) to determine cardiomyocyte size as previously
described [19]. Cell size from transversally cut
cardiomyocytes in the non-infarcted LV was measured using
an image analyser (Zeiss KS400, Germany) and a quantified
using ImageJ software (National Institutes of Health,
Bethesda, MD, USA). The investigators analysing the data
were blinded to the treatment allocation.
Blood and Urine Measurements
Blood samples were obtained via a tail vein under isoflurane
anaesthesia to determine anti-factor Xa activity (AXA), pro-
thrombin time (PT) and activated partial thromboplastin time
(APTT) levels. At sacrifice, 8 ml of blood was drawn from the
abdominal aorta (either anti-coagulated with sodium citrate or
EDTA), and urine was collected directly from the bladder.
Complete blood count was determined on the day of sacrifice
using a Sysmex Hematology Analyzer (Symex XN-10,
Sysmex Corporation, Japan).
Prothrombin Time, Activated Partial Thromboplastin
Time and Anti-Factor Xa Activity Assay
To determine the effect of apixaban on PT and APTT levels,
each sample was tested using Innovin (PT) and Actin FS
(APTT) reagents (Siemens Healthcare Diagnostics). Plasma
apixaban concentration was assessed using a chromogenic
anti-factor Xa activity (AXA) assay, the Berichrom Heparin
Assay (Dade Behring, Marburg, Germany) as this is the most
reliable method to measure the pharmacodynamics of
apixaban [21].
Urine Occult Blood Test
Haemoglobin/red blood cells were determined by a semiquan-
titative method using Combur10 Test Sticks (Roche
Diagnostics GmbH, Mannheim, Germany).
Quantitative Real-time PCR
RNA was extracted from the non-infarcted LV using TRIzol
reagent (Invitrogen Corp., Carlsbad, CA, USA), as previously
described [19,22] and the NanoDrop device was used to mea-
sure RNA concentration. Random primer mix was used to
prepare first-stranded DNA and thereafter used as a template
for quantitative real-time reverse-transcriptase PCR (qRT-
PCR) (25 ng/reaction). mRNA levels obtained by a qRT-
PCR using a C1000 Thermal Cycler CFX384 Real-Time
PCR Detection System (Bio-Rad Laboratories, Veenendaal,
The Netherlands). 36B4 reference gene was used to correct all
measured mRNA expression. Primer sequences can be found
in Supplementary table S1.
Western Blot
Frozen non-infarcted LV tissue was homogenized in ice-cold
lysis buffer containing phosphatase inhibitor cocktail 1
(Sigma) and protease inhibitor (ROCHE) as described previ-
ously [19]. Bio-Rad DC Protein Assay (Bio-Rad Laboratories,
Veenendaal, The Netherlands) was used for protein quantifi-
cations with bovine albumin as a standard, as described before
[19]. Immunoblotting was performed using primary antibod-
ies from commercial suppliers (Supplementary table S2).
Immunoblots were incubated with appropriate secondary an-
tibodies for 1 h at room temperature. Signals were detected by
ECL (ParkinElmer, Waltham, MA, USA). Blots were quanti-
fied using ImageJ software (National Institutes of Health,
Bethesda, MD, USA). The density of each band was normal-
ized to GADPH acting as a loading control and presented as
fold change over Sham-veh group.
RhoA Activity Assay
RhoA activity was measured according to the manufacturers
protocol (BK124; Cytoskeleton Inc.), as previously described
[23].
Statistical Analysis
Data are presented as means ± standard errors of the mean
(SEM). To compare normally distributed parameters, one-
way analysis of variance (ANOVA) followed by Tukeyspost
hoc test was used. When data were not normally distributed, a
non-parametric Kruskal-Wallis test followed by a Mann-
Whitney Utest with correction for multiple comparisons
was used. Wilcoxon signed-rank test was used to evaluate
LVEF post-MI vs before termination. Differences were con-
sidered significant at p< 0.05. IBM SPSS Statistics for
Windows, Version 23.0 (IBM Corp, USA), was used to per-
form all statistical analysis.
955Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Results
We performed MI or sham surgery on a total of 78 male
Sprague-Dawley rats; 21 rats (27%) died during the surgical
procedure; all remaining rats survived the rest of the study. A
total of 4 rats with an infarct < 15% of the LV were excluded
from further analysis, leaving a total of 53 rats for the analysis.
The final group sizes were 8 for the sham-apixaban group, 13
for the sham-vehicle group, 17 for the MI-apixaban group and
15 for the MI-vehicle group.
Efficacy and Safety of Apixaban in Rats with HF After
MI
Efficacy of Apixaban
Daily food intake and water intake were comparable among the
groups (Table 1). As expected, plasma PT and APTT were not
affected by apixaban treatment (Table 1)[24]. In rats treated with
apixaban, the plasma AXA activity was increased by fivefold as
compared with that in vehicle-treated rats (Table 1).
Safety of Apixaban
We did not observe any bleeding events during the study, nor
did we observe reductions in haemoglobin levels (Table 1)or
detect blood in the urine (Supplementary fig. S1).
Furthermore, apixaban did not affect other haematological
parameters such as white blood cells (WBC), red blood cells
(RBC), haematocrit (HCT) and platelet (PLT) counts
(Table 1). Body weight was similar in apixaban- and
vehicle-treated groups (Table 1).
Effect of Apixaban on Cardiac Function
The average MI size was 38 ± 2% and was comparable be-
tween MI-vehicle and MI-apixaban (Fig. 1a, b). MI surgery
resulted in cardiac dilatation and a marked reduction in LVEF
(Fig. 1b, c). At the initiation of therapy, 2 weeks after MI, all
indices of left ventricular function were comparable between
the MI-Apixaban and the MI-vehicle groups. As expected, a
progressive deterioration in LVEF was observed in the MI-
vehicle group over the 10-week treatment period (Fig. 1e).
Treatment with apixaban did not alter cardiac function and
all echocardiographic parameters remained comparable be-
tween the MI-vehicle- and the MI-apixaban-treated animals
(Fig. 1, Table 2). All other relevant echocardiographic param-
eters at week 11 are depicted in Table 2. The infarcted rats
were haemodynamically compromised, as reflected by a de-
crease in contractility and relaxation (dP/dt max-min), and an
increase in LVEDP. Treatment with apixaban did not alter
these parameters (Table 3). Moreover, no significant differ-
ence was found in systolic blood pressure or diastolic blood
pressure among the groups (Table 3).
Effect of Apixaban on Cardiac Histology and
Molecular Markers for Remodelling and Fibrosis
The biventricular weight/tibia lengths ratio was calculated as a
marker of hypertrophy and was found to be significantly
Table 1 General characteristics
and haematological parameters Parameters Sham-vehicle Sham-
apixaban
MI-vehicle MI-apixaban
Food intake (g/day) 31.9 ± 0.5 29.9 ± 0.7 30.3 ± 0.5 29.6 ± 0.3
Water intake (ml/day) 32.0 ± 0.9 29.9 ± 0.6 30.9 ± 1.0 28.5 ± 0.4
Body weight change (g) 91.15 ± 3.2 93.25 ± 4.8 94.73 ± 4.8 93.24 ± 4.8
AXA (ng/ml) 21 ± 2 109 ± 17
19 ± 1 103 ± 11
#
PT (s) 11.12 ± 0.12 11.00 ± 0.10 11.31 ± 0.21 11.17 ± 0.17
APTT (s) 15.48 ± 1.13 16.74 ± 0.62 17.24 ± 1.20 16.91 ± 0.94
WBC (10^9/l) 9.34 ± 2.18 11.34 ± 0.68 11.23 ± 1.24 10.66 ± 0.80
RBC (10^12/l) 9.01 ± 0.17 8.94 ± 0.10 8.99 ± 0.10 9.32 ± 0.23
HCT (mmol/l) 10.02 ± 0.25 9.84 ± 0.12 9.79 ± 0.13 10.00 ± 0.12
HGB (l/l) 0.503 ± 0.02 0.489 ± 0.01 0.493 ± 0.01 0.505 ± 0.01
PLT (10^9/l) 1025.60 ± 53.50 911.00 ± 47.85 826.56 ± 57.10 946.50 ± 57.56
Data are presented as means ± SEM
p< 0.05 vs Sham-veh
#
p<0.05vsMI-veh
AXA, anti-factor Xa activity; PT, prothrombin time; APTT, activated partial thromboplastin time; WBC, white
blood cell count; RBC, red blood cell count; HCT, haematocrit; HGB: haemoglobin; PLT, absolute automated
platelet count
956 Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Post-MI Before termination
0
30
40
50
60
70
Ejection fraction (%)
MI-Vehicle
MI-Apixaban
VehApixVehApix
0
25
30
35
40
Biventricular weight / TL
(mg/mm)
Sham MI
*
*
Ejection fraction (%)
Veh Apix Veh Apix
0
20
40
60
80
Sham MI
*
*
Veh Apix Veh Apix
0
6
8
10
LVIDd (mm)
Sham MI
*
*
Infarct size (%)
Veh Apix Veh Apix
0
10
20
30
40
50
Sham
MI
Sham-Vehicle Sham-Apixaban MI-Vehicle MI-Apixaban
Masson trichrome
a
cdb
ef
Fig. 1 Effect of apixaban on cardiac function. aRepresentative LV
sections stained with Massons trichrome. bQuantification of infarct
size from Massons trichromestained section; n=817/group. cLeft
ventricular internal dimensions in diastole (LVIDd) at week 11; n=8
17/group.dEjection fraction of the LV at week 11. eLongitudinal change
of LV ejection fraction post-MI and before termination; n=817/group. f
Ratio of biventricular weight to tibia length; n=817/group. Data are
presented as means ± SEM. *p< 0.05 vs sham with the same treatment;
p<0.05vsMIpost-MI
Table 2 Echocardiography
parameters in sham-operated and
post-myocardial infarction rats at
week 11
Sham-veh Sham-
apixaban
MI-vehicle MI-apixaban
IVSd (mm) 2.09 ± 0.15 2.46 ± 0.13 1.89 ± 0.10* 1.81 ± 0.13*
LVIDd (mm) 7.67 ± 0.22 7.34 ± 0.18 9.29 ± 0.31* 8.99 ± 0.33*
LVPWd (mm) 2.18± 0.13 2.23 ± 0.27 1.88 ± 0.12 1.85 ± 0.12
IVSs (mm) 3.45 ± 0.18 3.64 ± 0.27 2.39 ± 0.15* 2.31 ± 0.21*
LVIDs (mm) 4.52 ± 0.25 4.38 ± 0.22 7.52 ± 0.37* 7.04 ± 0.35*
LVPWs (mm) 3.14 ± 0.13 2.86 ± 0.34 2.37 ± 0.10* 2.42 ± 0.14
FS (%) 41.41 ± 2.37 37.40 ± 1.68 19.02 ± 1.22* 22.17 ± 1.76*
EF (%) 70 ± 2 66 ± 2 37 ± 2* 42 ± 3*
Data are presented as means ± SEM
*p< 0.05 vs sham with the same treatment
IVS, interventricular in diastole (d) and systole (s), respectively; LVID, left ventricular internal dimensions in both
diastole (d) and systole (s); LVPW, the thickness of left ventricle posterior wall in diastole (d) and systole (s); FS,
fractional shortening; EF, left ventricular ejection fraction
957Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
increased in MI-vehicle rats compared with that in sham-
operated rats (Fig. 1e).TheMIratsdemonstratedincreased
cardiomyocyte cross-sectional area and fibrosis compared
with the control rats (Fig. 2ac). However, apixaban therapy
did not affect the extent of cardiac hypertrophy nor did it
influence the degree of LV fibrosis (Fig. 2ac).
MI surgery increased the myocardial expression of atrial
natriuretic peptide (ANP), and brain natriuretic peptide (BNP)
and increased the relative expression of foetal (β-MHC) com-
pared to with that of adult (α-MHC)myosinheavychainiso-
form (i.e. β-MHC/α-MHC ratio), as markers for foetal gene
reprogramming in heart failure (Fig. 2d). The mRNA levels of
cardiac fibrosis markers collagen, type I, alpha 1 (COL1A1)
and tissue inhibitor of metalloproteinases 1 (TIMP1) were also
significantly increased in the hearts of rats following MI, com-
pared to with that in the hearts of sham rats (Fig. 2e). Apixaban
treatment had no effect on ANP, BNP, β-MHC/α-MHC ratio,
COL1A1 or TIMP1 mRNA levels (Fig. 2d, e).
Effect of Apixaban on PAR1 Signalling Pathways
Next, we aimed to determine whether apixaban influenced the
activity of thrombin-related pathways downstream of the
PAR1 receptor. Binding of thrombin to the PAR1 receptor
results in the activation of RhoA, which in turn phosphory-
lates Rho-associated coiled-coil kinase (ROCK) [25]. The
RhoA/ROCK pathway has been shown to contribute to cardi-
ac remodelling in HF [26,27] It has also been reported that
PAR1 activates AKT and ERK, two well-established regula-
tors of cardiac remodelling, independent from RhoA [16,28].
Next, we determined the effects of Apixaban on myocardi-
al RhoA activity. As expected, RhoA activity was increased
after MI, but the RhoA activity did not differ between the MI-
vehicle and the MI-apixaban group (Fig. 3a). Furthermore,
apixaban did not influence the phosphorylation levels of
AKT (Fig. 3b, d), nor did it affect the activation of ERK1/2
(data not shown). Finally, the LV protein expression levels of
the PAR1 receptor were comparable between all groups
(Fig. 3b, c), indicating that the results above could not be
explained by aberrant expression of the PAR1 receptor.
Discussion
We tested the hypothesis that treatment with FXa inhibitors
apixaban would improve cardiac function and ameliorate car-
diac remodelling in rats with HF after a large transmural an-
terior MI. For this purpose, we used a well-established model
of chronic post-MI HF and ensured that the degree of LV
dysfunction was similar in the apixaban and vehicle-treated
groups at the initiation of therapy. Furthermore, we used a
clinically relevant dose of apixaban that also appeared to be
safe and effective, as evidenced by a consistent 5-fold increase
in AXA and by the absence of (occult) bleeding. We demon-
strate that treatment with apixaban did not influence LV func-
tion, nor did it influence LV dilatation of LV hypertrophy.
Moreover, histological and molecular markers for pathologi-
cal LV remodelling were also not influenced by apixaban and
the activity of signal transduction pathways downstream of
the PAR1 was unaltered. These findings suggest that the in-
hibition of FXa with a safe, effective and clinically relevant
dose of apixaban does not influence cardiac remodelling in the
chronic phase after MI. In addition, our findings suggest that
the role of thrombin-mediated PAR1 receptor activation to the
pathophysiology of HF is limited. Our findings are in line with
the results from the COMMANDER HF trial and do not sup-
port the use of FXa inhibitor in HF patients with the aim to
amend the severity of HF.
HF reflects a pro-coagulant state, because all prerequisites
for thrombosis as described in Virchows law are met:
Table 3 Haemodynamic
parameters in sham-operated and
post-myocardial infarction rats
Sham-veh Sham-
apixaban
MI-vehicle MI-apixaban
HR (bpm) 287 ± 15 291 ± 13 299 ± 11 291 ± 8
SBP (mmHg) 119.24 ± 4.37 115.87 ± 6.01 115.77 ± 3.25 110.42 ± 1.93
DBP (mmHg) 84.69 ± 3.45 73.63 ± 2.36 82.66 ± 2.19 75.31 ± 2.46
LVESP (mmHg) 104.49 ± 8.30 110.77 ± 5.78 110.35 ± 3.17 105.94 ± 3.45
LVEDP (mmHg) 12.10 ± 2.24 11.40 ± 1.02 16.30 ± 0.80* 15.85 ± 0.76*
dP/dt max (mmHg/s) 6848 ± 239 6432 ± 504 5427 ± 173* 5021 ± 297*
dP/dt min (mmHg/s) 7683 ± 302 6766 ± 456 5285 ± 224* 5304 ± 250*
Data are presented as means ± SEM
*p< 0.05 vs sham with the same treatment
HR, heart rate; bpm, beat per minute; SBP, systolic blood pressure; DBP, diastolic blood pressure; LVESP, left
ventricular end-systolic pressure; LVEDP, left ventricular end-diastolic pressure; dP/dtmax and dP/dtmin,the
maximal rate of increase and decrease of left ventricular pressure, respectively
958 Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
0
1
2
3
4
5
mRNA level (fold change)
Vehicle
Apixaban
Sham Sham ShamMI MI MI
*
*
*
***
0.0
0.5
1.0
1.5
2.0
2.5
mRNA level (fold change)
Vehicle
Apixaban
Sham ShamMI MI
*
**
*
% Fibrosis
Veh Apix Veh Apix
0
5
10
15
20
25
Sham MI
*
*
Cardiomyocyte CSA (µm2)
Veh Apix Veh Apix
0
200
400
600
800
1000
Sham MI
*
*
ab
cd e
Fig. 2 Effect of apixaban on cardiac histology and molecular markers for
remodelling and fibrosis. aQuantification of cardiomyocyte cross-
sectional area from WGA-stained section; n=817/group. b
Representative LV sections stained with WGA and Massonstrichrome
to assess cardiomyocyte hypertrophy and fibrosis. cQuantification of
fibrosis in non-infarcted LV from Massonstrichromestained section;
n=817/group. d,eMeasurement of mRNA levels to assess molecular
markers for remodelling and fibrosis in non-infarcted LV, respectively,
normalized to 36b4; n=817/group. Data are presented as means ± SEM.
*p< 0.05 vs sham with the same treatment
RhoA activity (fold change)
Veh Apix Veh Apix
0.0
0.5
1.0
1.5
2.0
2.5
Sham MI
*
*
PAR1 relative expression
(fold change)
Veh Apix Veh Apix
0.0
0.5
1.0
1.5
Sham MI
pAKT/AKT relative
expression (fold change)
Veh Apix Veh Apix
0.0
0.5
1.0
1.5
Sham MI
PAR 1
pAKT
AKT
GAPDH
Sham-Veh Sham-Apix MI-Veh MI-Apix
ab
cd
Fig. 3 Effect of apixaban on
PAR1 signalling pathways. a
RhoA activity; n=6/group. b
Western blot analysis of PAR1,
total and phosphorylated Akt; n=
6/group. cQuantification of
PAR1 protein levels; n=6/group.
dQuantification of Akt
phosphorylation protein levels;
n= 6/group. Apix, apixaban. The
density of each band was
normalized to GADPH acting as a
loading control and presented as
fold change over Sham-veh
group. Data are presented as
means ± SEM. *p< 0.05 vs sham
with the same treatment
959Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
abnormalities in blood flow, in the vessel wall and in blood
constituents [29,30]. Aberrant platelet activation and in-
creased levels of pro-coagulant factors reflect abnormalities
in blood constituents. Impaired contractility and dilated cham-
bers perturb myocardial blood flow and endothelial damage
and reduced endothelium-dependent vasodilation reflect the
changes in the vessel wall [31]. It has been previously recog-
nized that patients with HF have an increased risk of throm-
boembolic events, both systemic and venous. The
prothrombotic molecules, such as fibrinogen and von
Willebrand factors, have been found to be elevated in subjects
with HF. Moreover, previous studies showed that the throm-
botic risk associated with HF appears to increase with the
severity of the disease [32,33]. A large Danish prospective
cohort study demonstrated that the risk of stroke and throm-
boembolic event was increased in HF patients with HF inde-
pendent of the CHA2DS2-VASc score. Furthermore, in pa-
tients with HF and a CHA2DS2-VASc score 4, the absolute
risk of thromboembolic events was even higher in patients
without than with concomitant AF [34]. In addition, the prog-
nosis of HF markedly deteriorates after a thromboembolic
event occurs, suggesting that interventions to reduce throm-
boembolic events may improve prognosis in HF patients [35].
Another mechanism by which the hypercoagulable state could
contribute to the progression of HF is coronary microvascular
embolization leading to MI [36,37].
Anticoagulants are often prescribed in HF patients without
AF that are considered to be at high risk for stroke, such as
those with an LV thrombus. There is, however, very little
evidence to support the lenient prescription of anticoagulants
in patients with HF and sinus rhythm [38]. In the Warfarin
versus Aspirin in Reduced Cardiac Ejection Fraction
(WARCEF) trial, warfarin did not result in a meaningful re-
duction in the rates of ischaemic stroke, intracerebral haemor-
rhage or death from any cause [39]. Furthermore, the effect of
FXa inhibitor rivaroxaban on clinical outcomes was recently
tested in the COMMANDER HF (A Study to Assess the
Effectiveness and Safety of Rivaroxaban in Reducing the
Risk of Death, Myocardial Infarction, or Stroke in
Participants with Heart Failure and Coronary Artery Disease
Following an Episode of Decompensated Heart Failure)trial
[40]. The authors randomized > 5000 patients with chronic
ischaemic HF with reduced ejection fraction but without atrial
fibrillation to a low dose of rivaroxaban or placebo.
Rivaroxaban was safe and well tolerated, but did not affect
the incidence of the combined endpoint of all-cause mortality,
stroke or myocardial infarction. In both the WARCEF and the
COMMANDER HF study, anticoagulation did reduce the in-
cidence of ischaemic stroke, suggesting that the lack of effect
was not dose related. Our results confirm and extend upon the
results of the COMMANDER HF trial and provide mechanis-
tic underpinnings that explain the neutral outcomes.
Furthermore, our findings suggest that clinically relevant
doses of FXa inhibitors do not influence PAR mediated sig-
nalling and cardiac remodelling.
To the best of our knowledge, our study is the first to study
the effect of Xa inhibition on cardiac remodelling in rats with
established LV dysfunction. Several studies have, however,
evaluated the early effect of FXa inhibition on myocardial
ischaemia/reperfusion injury and post-infarct remodelling.
The indirect FXa inhibitor fondaparinux has been shown to
reduceinfarctsizefollowing2hofreperfusioninaratmodel
of myocardial ischaemia-reperfusion [41]. The effects of FXa
inhibitors are more variable as Flierl et al. did not observe
infarct size reduction when rivaroxaban was given in rats with
permanent coronary ligation [42]. However, a similar study by
Bode et al. indicated that the administration of rivaroxaban
immediately after surgery resulted ina reduction in infarct size
and improvements in cardiac function. Conversely, when
rivaroxaban was initiated 3 days after MI surgery, no effect
on infarct size or cardiac function was observed [43]. Taken
together, the available evidence on the effect of FXa inhibitors
on cardiac function is in line with our observations and dis-
putes their utility in chronic HF setting.
There are four PAR isoforms, but PAR1 and PAR2 are the
predominant isoforms in the heart [13,16]. PAR1 is activated
by thrombin and FXa, but PAR2 is only activated by FXa.
Both receptors are expressed in cardiomyocytes and cardiac
fibroblast. Cleavage of PARs results in activation of several G
proteincoupled receptors and their downstream signalling
pathways, including RhoA/ROCK, the MAPK pathways,
ERK 1/2 and ERK5 [16]. Several lines of evidence indicate
that PARs are activated in HF and contribute to disease pro-
gression. First, PAR1 expression and the activity of its down-
stream signal transduction pathways are increased in murine
models of chronic HF and in the ventricles of HF patients with
ischaemic or idiopathic dilated cardiomyopathy [14,15].
Second, the activation of PARs in cultured cardiomyocytes
induces pathological hypertrophy, reflected by increases in
cell size and sarcomeric organization, the activation of the
foetal gene program and perturbations in cardiac calcium han-
dling [28]. Third, PAR1 activation in cardiac fibroblasts in-
duces a pro-fibrotic state reflected by enhanced proliferation
and increased expression of transforming growth factor Beta
(TGF-ß) [13,28,44]. Fourth, PAR1 activation is strongly pro-
inflammatory as it induces the expression of interleukin (IL)-
6, IL-8 and monocyte chemoattractant protein (MCP-1) [12,
13]. Yet, the most robust evidence comes from studies in
PAR1-knockout (KO) mice and mice with overexpressing
the PAR1. In a PAR1-KO mouse model, reduced cardiac re-
modelling was observed after I/R injury; however, infarct size
was not affected. Accordingly, mice with cardiomyocyte-
specific overexpression of PAR1 exhibited eccentric hypertro-
phy and dilated cardiomyopathy [16]. Additionally, PAR sig-
nalling activates RhoA/ROCK pathway [25,45] which has
been shown to mediate fibrosis in the heart [26]. Protein levels
960 Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
of ROCKs as well as RhoA activity were significantly in-
creased in CHF patients [27]. Furthermore, the deletion of
ROCK attenuates HF progression and improves the cardiac
performance in mice [46,47]. These findings provided a clear
rationale for our studies.
However, in contrast to our expectations, we did not ob-
serve any changes in protein levels for PAR1 protein, nor did
we detect differences in the activation of the AKT and ERK 1/
2 signal transduction pathways downstream of PAR1 [48].
These findings are in consistence with a previous study [49]
in which PAR1 protein expression did not differ at 12 weeks
after MI induction or apixaban treatment. Moreover, it has
been previously published that upregulated AKT exerts
cardioprotective effects in models of preconditioning resulting
in limiting infarct size [50,51]. However, AKT is not regulat-
ed in a chronic ischaemia setting [52], and this finding was
also seen in our study. Our observation that ERK1/2 protein
levels were also not affected in post-MI LV dysfunction is in
accordance with similar studies in post-MI LV dysfunction
[53]. Consistent with other reports, we were able to detect a
clear increase in RhoA activity after MI, but this was not
affected by treatment with apixaban.
Other studies have demonstrated evidence that FXa inhib-
itors can influence PAR receptor signalling. For instance,
Bukowska et al. showed that FXa inhibitor rivaroxaban re-
duced MAP kinase activity and diminished the upregulation
of PARs, ICAM-1, LOX-1 and IL-8 and in human atrial tissue
cultures in media containing activated FXa [54]. Interestingly,
FXa has also been shown to activate ERK1/2 and induce pro-
inflammatory cytokines in alveolar epithelial cells, which was
suppressed by FXa inhibitor edoxaban [55]. In addition, the
FXa inhibitor rivaroxaban did inhibit cardiac FXa activity and
reduced cardiac fibrosis in a model of transverse aortic con-
striction [56]. Our results may therefore have been different in
other disease models or if we would have induced a murine
model prone to develop cardiac thrombi [57].
Study Limitations
Despite strengths related to the direct measures of cardiac struc-
ture, function and other haematological parameters, our study
does have limitations. First, in accordance with the previous
study [49], we did not observe changes in PAR1 protein levels
in post-MI HF model. Furthermore, the activity of downstream
signalling pathways of the PAR1 was not increased after MI and
apixaban did not alter this. It is possible that the activation of
PAR1 and other thrombin-related pathways is more pronounced
in other models or settings. Second, we started the treatment
2 weeks after MI when infarct healing had completed; we cannot
exclude that FXa inhibition could be beneficial during earlier
stages of post-infarct remodelling. Third, we employed apixaban
at a dose that was within the safety range (FDA application no.
202155Orig1s000). Based on the AXA levels, this dosage is
comparable with a 5-mg dose in humans [17]. The outcome of
our study may have been different when a higher dose had been
used. The clinical relevance of a study with high-dose apixaban
is, however, limited as anticoagulants have a narrow therapeutic
window and the benefits are often offset by the associated in-
creased risk of bleeding events [39]. Our study does not exclude
the possibility that thrombin-related pathways and PARs contrib-
ute to cardiac remodelling in HF. Our findings do, however,
question the utility of FXa inhibitors as a pharmacological ther-
apy to attenuate cardiac remodelling. Importantly, our study is in
line with the neutral effects of the COMMANDER HF trial, and
the molecular insights do not hint towards a direct of effect of
apixaban on cardiac muscle structure and function.
Conclusions
FXa inhibition with apixaban does not influence pathological
cardiac remodelling after a MI. These data do not support the
use of FXa inhibitor in HF patients with the aim to amend the
severity of HF.
Clinical Perspectives
It has been suggested that coagulation factors such as factor
Xa (FXa) and thrombin promote maladaptive cardiac remod-
elling and could promote heart failure (HF) development via
activation of the protease-activated receptors (PARs) in myo-
cardial tissue. If this hypothesis is true, cardiac remodelling
would be amendable by treatment with anticoagulants. To test
this hypothesis, rats with HF after myocardial infarction (MI)
were treated with the FXa inhibitor apixaban or a matching
vehicle. While apixaban was effective in inhibiting FXa activ-
ity, it did not affect the activity of PAR1 signalling pathways,
nor did it affect cardiac function and cardiac remodelling after
MI. These findings are in line with the results of the recent
COMMANDER HF trial and do not support the use of FXa
inhibitors in HF patients with the aim to improve heart func-
tion or to modulate the clinical course of HF.
Acknowledgements We acknowledge Bristol-Myers Squibb for supply-
ing apixaban and thank Dr. William Achanzar for the support and expert
advice on determining the correct dosage for this study. We thank
Marloes Schouten, Janny Takens and Silke Oberdorf for the expert tech-
nical assistance and advice.
AuthorsContributions SRY, HHWS, RAdB and BDW designed the
research study. SRY and MMD performed the research. SRY and
BDW analysed the data. SRY and BDW wrote the paper. HHWS,
KTN, DJvV and RAdB revised the paper for important intellectual con-
tent. All authors read and approved the final manuscript.
961Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Funding Information The study was funded by a competitive grant from
Bristol-Myers Squibb/Pfizer Alliance, within the European Thrombosis
Investigator-Initiated Research Program (ERISTA) (grant BMS ISR
#CV185-638).
Dr. Yurista is supported by a grant from the Indonesia Endowment
Fund for Education (LPDP no. 20150722083422). Dr. de Boer is sup-
ported by the Netherlands Heart Foundation (CVON DOSIS, grant 2014-
40; CVON SHE-PREDICTS-HF, grant 2017-21; and CVON RED-
CVD, grant 2017-11) and the Innovational Research Incentives Scheme
programme of the Netherlands Organization for Scientific Research
(NWO VIDI, grant 917.13.350). Dr. Westenbrink is supported by
The Netherlands Organisation for Scientific Research (NWO VENI,
grant 016.176.147).
Compliance and Ethical Standard
Ethics Approval and Consent to Participate The study was approved by
the University of Groningen Ethical Committee on Animal
Experimentation (IvD number: 16487-02-001). The investigation con-
forms to the Guide for the Care and Use of Laboratory Animals published
by the US National Institutes of Health and in accordance with national
regulations.
Competing Interests SRY, HHWS, KTN, MMD, DJvV and BDW do
not report conflicts of interest relative to this report. The UMCG, which
employs Dr. De Boer, has received research grants and/or fees from
AstraZeneca, Abbott, Bristol-Myers Squibb, Novartis, Roche, Trevena
and ThermoFisher GmbH. Dr. de Boer received personal fees from
MandalMed Inc., Novartis and Servier.
Disclaimer The funder had no role in the design and conduct of the
study; collection, management, analysis and interpretation of the data;
preparation, review or approval of the manuscript; and decision to submit
the manuscript for publication.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as
long as you give appropriate credit to the original author(s) and the
source, provide a link to the Creative Commons licence, and indicate if
changes were made. The images or other third party material in this article
are included in the article's Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the
article's Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
References
1. Zannad F. Risingincidence ofheart failure demands action. Lancet.
2018;391:5189.
2. Savarese G, Lund LH. Global Public Health Burden of Heart
Failure. Card Fail Rev. 2017;3:711.
3. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats
AJS, et al. 2016 ESC Guidelines for the diagnosis and treatment of
acute and chronic heart failure. Eur Heart J. 2016;37:2129200.
4. Lip GY, Gibbs CR. Does heart failure confer a hypercoagulable
state? Virchows triad revisited. J Am Coll Cardiol. 1999;33:
14246.
5. Loh E, Sutton MSJ, Wun C-CC, Rouleau JL, Flaker GC, Gottlieb
SS, et al. Ventricular dysfunction and the risk of stroke after myo-
cardial infarction. N Engl J Med. 1997;336:2517.
6. Abdul-Rahim AH, Perez A-C, Fulton RL, Jhund PS, Latini R,
Tognoni G, et al. Risk of stroke in chronic heart failure patients
without atrial fibrillation. Circulation. 2015;131:148694.
7. Kang S-H, Kim J, Park JJ, Oh I-Y, Yoon C-H, Kim H-J, et al. Risk
of stroke in congestive heart failure with and without atrial fibrilla-
tion. Int J Cardiol. 2017;248:1827.
8. Adelborg K, Szépligeti S, Sundbøll J, Horváth-Puhó E, Henderson
VW, Ording A, et al. Risk of stroke in patients with heart failure.
Stroke. 2017;48:11618.
9. Greenberg B, Neaton JD, Anker SD, Byra WM, Cleland JGF, Deng
H, et al. Association of rivaroxaban with thromboembolic events in
patients with heart failure, coronary disease, and sinus rhythm: a
post hoc analysis of the COMMANDER HF trial. JAMA Cardiol.
2019;4:51523.
10. Zannad F, Greenberg B, Cleland JGF, Gheorghiade M, van
Veldhuisen DJ, Mehra MR, et al. Rationale and design of a ran-
domized, double-blind, event-driven, multicentre study comparing
the efficacy and safety of oral rivaroxaban with placebo for reduc-
ing the risk of death, myocardial infarction or stroke in subjects
with heart failure and signi. Eur J Heart Fail. 2015;17:73542.
11. Westenbrink BD, Alings M, Connolly SJ, Eikelboom J, Ezekowitz
MD, Oldgren J, et al. Anemia predicts thromboembolic events,
bleeding complications and mortality in patients with atrial fibrilla-
tion: insights from the RE-LY trial. J Thromb Haemost. 2015;13:
699707.
12. Spronk HMH, de Jong AM, Crijns HJ, Schotten U, Van Gelder IC,
ten Cate H. Pleiotropic effects of factor Xa and thrombin: what to
expect from novel anticoagulants. Cardiovasc Res. 2014;101:34451.
13. Spronk HMH, De Jong AM, Verheule S, De Boer HC, Maass AH,
Lau DH, et al. Hypercoagulability causes atrial fibrosis and pro-
motes atrial fibrillation. Eur Heart J. 2017;38:3850.
14. Moshal KS, Tyagi N, Henderson B, Ovechkin AV, Tyagi SC.
Protease-activated receptor and endothelial-myocyte uncoupling
in chronic heart failure. Am J Physiol Heart Circ Physiol.
2005;288:H27707.
15. Moshal KS, Tyagi N, Moss V, Henderson B, Steed M, Ovechkin A,
et al. Early induction of matrix metalloproteinase-9 transduces sig-
naling in human heart end stage failure. J Cell Mol Med. 2005;9:
70413.
16. Pawlinski R, Tencati M, Hampton CR, Shishido T, Bullard TA,
Casey LM, et al. Protease-activated receptor-1 contributes to cardi-
ac remodeling and hypertrophy. Circulation. 2007;116:2298306.
17. Shin H, Cho M-C, Kim RB, Kim C-H, Choi N-C, Kim S-K, et al.
Laboratory measurement of apixaban using anti-factor Xa assays in
acute ischemic stroke patients with non-valvular atrial fibrillation. J
Thromb Thrombolysis. 2018;45:2506.
18. van der Meer P, Lipsic E, HenningRH, Boddeus K,van der Velden
J, Voors AA, et al. Erythropoietin induces neovascularization and
improves cardiac function in rats with heart failure after myocardial
infarction. J Am Coll Cardiol. 2005;46:12533.
19. Yurista SR, Silljé HHW, Oberdorf-Maass SU, Schouten E, Pavez
Giani MG, Hillebrands J, et al. Sodium-glucose co-transporter 2
inhibition with empagliflozin improves cardiac function in non-
diabetic rats with left ventricular dysfunction after myocardial in-
farction. Eur J Heart Fail. 2019;21:86273.
20. Takagawa J, Zhang Y, Wong ML, Sievers RE, Kapasi NK, Wang
Y, et al. Myocardial infarct size measurement in the mouse chronic
infarction model: comparison of area- and length-based approaches.
J Appl Physiol. 2007;102:210411.
21. Osanai H, Ajioka M, Masutomi T, Kuwayama T, Ishihama S,
Sakamato Y, et al. Measurement of anti-factor Xa activity in pa-
tients on apixaban for non-valvular atrial fibrillation. Circ J.
2015;79:258490.
22. Yurista SR, Silljé HHW, van Goor H, Hillebrands J-L, Heerspink
HJL, de Menezes Montenegro L, et al. Effects of SodiumGlucose
Co-transporter 2 Inhibition with Empaglifozin on Renal Structure
962 Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
and Function in Non-diabetic Rats with Left Ventricular
Dysfunction After Myocardial Infarction. Cardiovasc Drugs Ther.
2020;34:31121.
23. Jin W, Goldfine AB, Boes T, Henry RR, Ciaraldi TP, Kim E-Y,
et al. Increased SRF transcriptional activity in human and mouse
skeletal muscle is a signature of insulin resistance. J Clin Invest.
2011;121:91829.
24. Van Blerk M, Bailleul E, Chatelain B, Demulder A, Devreese K,
Douxfils J, et al. Influence of apixaban on commonly used coagu-
lation assays: results from the Belgian national External Quality
Assessment Scheme. Int J Lab Hematol. 2017;39:4028.
25. Greenberg DL, Mize GJ, Takayama TK. Protease-Activated
Receptor Mediated RhoA Signaling and Cytoskeletal
Reorganization in LNCaP Cells . Biochemistry. 2003;42:7029.
26. Lauriol J, Keith K, Jaffre F, Couvillon A, Saci A, Goonasekera SA,
et al. RhoA signaling in cardiomyocytes protects against stress-
induced heart failure but facilitates cardiac fibrosis. Sci Signal.
2014;7:ra100ra100
27. Dong M, Liao JK, Fang F, Lee AP-W, Yan BP-Y, Liu M, et al.
Increased Rho kinase activity in congestive heart failure. Eur J
Heart Fail. 2012;14:96573.
28. Sabri A, Muske G, Zhang H, Pak E, Darrow A, Andrade-Gordon P,
et al. Signaling properties and functions of two distinct cardiomyo-
cyte protease-activated receptors. Circ Res. 2000;86:105461.
29. Gurbel PA, Tantry US. Antiplatelet and anticoagulant agents in
heart failure. JACC Heart Fail. 2014;2:114.
30. Zeitler EP, Eapen ZJ. Anticoagulation in heart failure: a review. J
Atr Fibrillation. 2015;8:1250.
31. Gheorghiade M, Vaduganathan M, Fonarow GC, Greene SJ,
Greenberg BH, Liu PP, et al. Anticoagulation in heart failure: cur-
rent status and future direction. Heart Fail Rev. 2013;18:797813.
32. Howell M. Congestive heart failure and outpatient risk of venous
thromboembolism: a retrospective, case-control study. J Clin
Epidemiol. 2001;54:8106.
33. Weill-Engerer S, Meaume S, Lahlou A, Piette F, Saint-Jean O,
Sachet A, et al. Risk factors for deep vein thrombosis in inpatients
aged 65 and older: a case-control multicenter study. J Am Geriatr
Soc. 2004;52:1299304.
34. Melgaard L, Gorst-Rasmussen A, Lane DA, Rasmussen LH,
Larsen TB, Lip GYH. Assessment of the CHA 2 DS 2 -VASc score
in predicting ischemic stroke, thromboembolism, and death in pa-
tients with heart failure with and without atrial fibrillation. JAMA.
2015;314:10308.
35. Mebazaa A, Spiro TE, Büller HR, Haskell L, Hu D, Hull R, et al.
Predicting the risk of venous thromboembolism in patients hospi-
talized with heart failure. Circulation. 2014;130:4108.
36. Jiao Z-Y, Zhang D-P, Xia K, Wang L-F, Yang X-C. Clinical anal-
ysis of acute myocardial infarction caused by coronary embolism. J
Thorac Dis. 2017;9:2898903.
37. Raphael CE, Heit JA, Reeder GS, Bois MC, Maleszewski JJ,
Tilbury RT, et al. Coronary embolus. JACC Cardiovasc Interv.
2018;11:17280.
38. Gianstefani S, Douiri A, Delithanasis I, Rogers T, Sen A, Kalra S,
et al. Incidence and predictors of early left ventricular thrombus
after ST-elevation myocardial infarction in the contemporary era
of primary percutaneous coronary intervention. Am J Cardiol.
2014;113:11116.
39. Homma S, Thompson JLP, Pullicino PM, Levin B, Freudenberger
RS, Teerlink JR, et al. Warfarin and aspirin in patients with heart
failure and sinus rhythm. N Engl J Med. 2012;366:185969.
40. Zannad F, Anker SD, Byra WM, Cleland JGF, Fu M, Gheorghiade
M, et al. Rivaroxaban in patients with heart failure, sinus rhythm,
and coronary disease. N Engl J Med. 2018;379:133242.
41. Macchi L, Ben MW, Guillou S, Tamareille S, Lamon D, Prunier D,
et al. The synthetic pentasaccharide fondaparinux attenuates
myocardial ischemia-reperfusion injury in rats via STAT-3.
Shock. 2014;41:16671.
42. Flierl U, Fraccarollo D, Micka J, Bauersachs J, Schäfer A. The
direct factor Xa inhibitor Rivaroxaban reduces platelet activation
in congestive heart failure. Pharmacol Res. 2013;74:4955.
43. Bode MF, Auriemma AC, Grover SP, Hisada Y, Rennie A, Bode
WD, et al. The factor Xa inhibitor rivaroxaban reduces cardiac
dysfunction in a mouse model of myocardial infarction. Thromb
Res. 2018;167:12834.
44. Sabri A, Short J, Guo J, Steinberg SF. Protease-activated receptor-
1-mediated DNA synthesis in cardiac fibroblast is via epidermal
growth factor receptor transactivation: distinct PAR-1 signaling
pathways in cardiac fibroblasts and cardiomyocytes. Circ Res.
2002;91:5329.
45. Klarenbach SW, Chipiuk A, Nelson RC, Hollenberg MD, Murray
AG. Differential actions of PAR2 and PAR1, in stimulating human
endothelial cell exocytosis and permeability: the role of Rho-
GTPases. Circ Res. 2003;92:2728.
46. Okamoto R, Li Y, Noma K, Hiroi Y, Liu PY, Taniguchi M, et al.
FHL2 prevents cardiac hypertrophy in mice with cardiac-specific
deletion of ROCK2. FASEB J. 2013;27:143949.
47. Zhang YM, Bo J, Taffet GE, Chang J, Shi J, Reddy AK, et al.
Targeted deletion of ROCK1 protects the heart against pressure
overload by inhibiting reactive fibrosis. FASEB J. 2006;20:91625.
48. Reséndiz JC, Kroll MH, Lassila R. Protease-activated receptor-in-
duced Akt activationregulation and possible function. J Thromb
Haemost. 2007;5:248493.
49. Shi G, Yang X, Pan M, Sun J, Ke H, Zhang C, et al. Apixaban
attenuates ischemia-induced myocardial fibrosis by inhibition of
Gq/PKC signaling. Biochem Biophys Res Commun. 2018;500:
5506.
50. HAUSENLOY D, YELLON D. Survival kinases in ischemic pre-
conditioning and postconditioning. Cardiovasc Res. 2006;70:240
53.
51. Cantley LC. The phosphoinositide 3-kinase pathway. Science.
2002;296:16557.
52. Ravingerová T, Matejíková J, Neckár J, Andelová E, Kolár F.
Differential role of PI3K/Akt pathway in the infarct size limitation
and antiarrhythmic protection in the rat heart. Mol Cell Biochem.
2007;297:11120.
53. Tenhunen O, Soini Y, Ilves M, Rysä J, Tuukkanen J, Serpi R, et al.
p38 kinase rescues failing myocardium after myocardial infarction:
evidence for angiogenic and anti-apoptotic mechanisms. FASEB J.
2006;20:19079.
54. Bukowska A, Zacharias I, Weinert S, Skopp K, Hartmann C, Huth
C, et al. Coagulation factor Xa induces an inflammatory signalling
by activation of protease-activated receptors in human atrial tissue.
Eur J Pharmacol. 2013;718:11423.
55. Bukowska A, Schild L, Bornfleth P, Peter D, Wiese-Rischke C,
Gardemann A, et al. Activated clotting factor X mediates mitochon-
drial alterations and inflammatory responses via protease-activated
receptor signaling in alveolar epithelial cells. Eur J Pharmacol.
2020;869:172875.
56. Guo X, Kolpakov MA, Hooshdaran B, Schappell W, Wang T,
Eguchi S, et al. Cardiac expression of factor X mediates cardiac
hypertrophy and fibrosis in pressure overload. JACC Basic Transl
Sci. 2020;5:6983.
57. Bukowska A, Felgendreher M, Scholz B, Wolke C, Schulte JS,
Fehrmann E, et al. CREM-transgene mice: an animal model of
atrial fibrillation and thrombogenesis. Thromb Res. 2018;163:
1729.
PublishersNote Springer Nature remains neutral with regard to juris-
dictional claims in published maps and institutional affiliations.
963Cardiovasc Drugs Ther (2021) 35:953–963
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... HF or sham surgery was performed under isoflurane (2.5%) inhalation anesthesia as described previously. 28,29 After left-sided thoracotomy, HF was induced by permanent ligating of the proximal portion of the left coronary. Sham-operated rats underwent the same procedure but without the actual ligation. ...
... Echocardiography, invasive hemodynamics, and cardiac gravimetric and histological measurements were performed as described elsewhere. 15,[27][28][29][30] Statistical Analysis ...
Article
Full-text available
Background Accumulating evidence suggests that the failing heart reprograms fuel metabolism toward increased utilization of ketone bodies and that increasing cardiac ketone delivery ameliorates cardiac dysfunction. As an initial step toward development of ketone therapies, we investigated the effect of chronic oral ketone ester (KE) supplementation as a prevention or treatment strategy in rodent heart failure models. Methods Two independent rodent heart failure models were used for the studies: transverse aortic constriction/myocardial infarction (MI) in mice and post-MI remodeling in rats. Seventy-five mice underwent a prevention treatment strategy with a KE comprised of hexanoyl-hexyl-3-hydroxybutyrate KE (KE-1) diet, and 77 rats were treated in either a prevention or treatment regimen using a commercially available β-hydroxybutyrate-(R)-1,3-butanediol monoester (DeltaG; KE-2) diet. Results The KE-1 diet in mice elevated β-hydroxybutyrate levels during nocturnal feeding, whereas the KE-2 diet in rats induced ketonemia throughout a 24-hour period. The KE-1 diet preventive strategy attenuated development of left ventricular dysfunction and remodeling post-transverse aortic constriction/MI (left ventricular ejection fraction±SD, 36±8 in vehicle versus 45±11 in KE-1; P =0.016). The KE-2 diet therapeutic approach also attenuated left ventricular dysfunction and remodeling post-MI (left ventricular ejection fraction, 41±11 in MI-vehicle versus 61±7 in MI-KE-2; P <0.001). In addition, ventricular weight, cardiomyocyte cross-sectional area, and the expression of ANP (atrial natriuretic peptide) were significantly attenuated in the KE-2–treated MI group. However, treatment with KE-2 did not influence cardiac fibrosis post-MI. The myocardial expression of the ketone transporter and 2 ketolytic enzymes was significantly increased in rats fed KE-2 diet along with normalization of myocardial ATP levels to sham values. Conclusions Chronic oral supplementation with KE was effective in both prevention and treatment of heart failure in 2 preclinical animal models. In addition, our results indicate that treatment with KE reprogrammed the expression of genes involved in ketone body utilization and normalized myocardial ATP production following MI, consistent with provision of an auxiliary fuel. These findings provide rationale for the assessment of KEs as a treatment for patients with heart failure.
... Following fixation and embedding processes, sections were cut using a microtome and were thereafter processed onto slides. To determine (cardio) myocyte cross sectional area, deparaffinised 3-μm thick sections were stained with 4′,6-diamidino-2-phenylindole (DAPI) (Vector Laboratories, USA) and wheat germ agglutinin (WGA) (Sigma-Aldrich, USA), as described before [60]. Stained transverse sections were visualised with fluorescence microscopy to generate images for analysis (Zeiss KS400, Germany). ...
Article
Full-text available
Erythropoietin (EPO) is a haematopoietic hormone that regulates erythropoiesis, but the EPO-receptor (EpoR) is also expressed in non-haematopoietic tissues. Stimulation of the EpoR in cardiac and skeletal muscle provides protection from various forms of pathological stress, but its relevance for normal muscle physiology remains unclear. We aimed to determine the contribution of the tissue-specific EpoR to exercise-induced remodelling of cardiac and skeletal muscle. Baseline phenotyping was performed on left ventricle and m. gastrocnemius of mice that only express the EpoR in haematopoietic tissues (EpoR-tKO). Subsequently, mice were caged in the presence or absence of a running wheel for 4 weeks and exercise performance, cardiac function and histological and molecular markers for physiological adaptation were assessed. While gross morphology of both muscles was normal in EpoR-tKO mice, mitochondrial content in skeletal muscle was decreased by 50%, associated with similar reductions in mitochondrial biogenesis, while mitophagy was unaltered. When subjected to exercise, EpoR-tKO mice ran slower and covered less distance than wild-type (WT) mice (5.5 ± 0.6 vs. 8.0 ± 0.4 km/day, p < 0.01). The impaired exercise performance was paralleled by reductions in myocyte growth and angiogenesis in both muscle types. Our findings indicate that the endogenous EPO-EpoR system controls mitochondrial biogenesis in skeletal muscle. The reductions in mitochondrial content were associated with reduced exercise capacity in response to voluntary exercise, supporting a critical role for the extra-haematopoietic EpoR in exercise performance.
Article
Background: PAR1 (protease-activated receptor-1) contributes to acute thrombosis, but it is not clear whether the receptor is involved in deleterious inflammatory and profibrotic processes in heart failure. Here, we employ the pepducin technology to determine the effects of targeting PAR1 in a mouse heart failure with reduced ejection fraction model. Methods: After undergoing transverse aortic constriction pressure overload or sham surgery, C57BL/6J mice were randomized to daily sc PZ-128 pepducin or vehicle, and cardiac function, inflammation, fibrosis, and molecular analyses conducted at 7 weeks RESULTS: After 7 weeks of transverse aortic constriction, vehicle mice had marked increases in macrophage/monocyte infiltration and fibrosis of the left ventricle as compared with Sham mice. PZ-128 treatment significantly suppressed the inflammatory cell infiltration and cardiac fibrosis. Despite no effect on myocyte cell hypertrophy, PZ-128 afforded a significant reduction in overall left ventricle weight and completely protected against the transverse aortic constriction-induced impairments in left ventricle ejection fraction. PZ-128 significantly suppressed transverse aortic constriction-induced increases in an array of genes involved in myocardial stress, fibrosis, and inflammation. Conclusions: The PZ-128 pepducin is highly effective in protecting against cardiac inflammation, fibrosis, and loss of left ventricle function in a mouse model.
Article
Full-text available
A Kinase Interacting Protein 1 (AKIP1) is a signalling adaptor that promotes physiological hypertrophy in vitro. The purpose of this study is to determine if AKIP1 promotes physiological cardiomyocyte hypertrophy in vivo. Therefore, adult male mice with cardiomyocyte-specific overexpression of AKIP1 (AKIP1-TG) and wild type (WT) littermates were caged individually for four weeks in the presence or absence of a running wheel. Exercise performance, heart weight to tibia length (HW/TL), MRI, histology, and left ventricular (LV) molecular markers were evaluated. While exercise parameters were comparable between genotypes, exercise-induced cardiac hypertrophy was augmented in AKIP1-TG vs. WT mice as evidenced by an increase in HW/TL by weighing scale and in LV mass on MRI. AKIP1-induced hypertrophy was predominantly determined by an increase in cardiomyocyte length, which was associated with reductions in p90 ribosomal S6 kinase 3 (RSK3), increments of phosphatase 2A catalytic subunit (PP2Ac) and dephosphorylation of serum response factor (SRF). With electron microscopy, we detected clusters of AKIP1 protein in the cardiomyocyte nucleus, which can potentially influence signalosome formation and predispose a switch in transcription upon exercise. Mechanistically, AKIP1 promoted exercise-induced activation of protein kinase B (Akt), downregulation of CCAAT Enhancer Binding Protein Beta (C/EBPβ) and de-repression of Cbp/p300 interacting transactivator with Glu/Asp rich carboxy-terminal domain 4 (CITED4). Concludingly, we identified AKIP1 as a novel regulator of cardiomyocyte elongation and physiological cardiac remodelling with activation of the RSK3-PP2Ac-SRF and Akt-C/EBPβ-CITED4 pathway. These findings suggest that AKIP1 may serve as a nodal point for physiological reprogramming of cardiac remodelling.
Article
Full-text available
Background The use of sodium–glucose co-transporter 2 inhibitors (SGLT2i) is currently expanding to cardiovascular risk reduction in non-diabetic subjects, but renal (side-)effects are less well studied in this setting.Methods Male non-diabetic Sprague Dawley rats underwent permanent coronary artery ligation to induce MI, or sham surgery. Rats received chow containing empagliflozin (EMPA) (30 mg/kg/day) or control chow. Renal function and electrolyte balance were measured in metabolic cages. Histological and molecular markers of kidney injury, parameters of phosphate homeostasis and bone resorption were also assessed.ResultsEMPA resulted in a twofold increase in diuresis, without evidence for plasma volume contraction or impediments in renal function in both sham and MI animals. EMPA increased plasma magnesium levels, while the levels of glucose and other major electrolytes were comparable among the groups. Urinary protein excretion was similar in all treatment groups and no histomorphological alterations were identified in the kidney. Accordingly, molecular markers for cellular injury, fibrosis, inflammation and oxidative stress in renal tissue were comparable between groups. EMPA resulted in a slight increase in circulating phosphate and PTH levels without activating FGF23–Klotho axis in the kidney and bone mineral resorption, measured with CTX-1, was not increased.ConclusionsEMPA exerts profound diuretic effects without compromising renal structure and function or causing significant electrolyte imbalance in a non-diabetic setting. The slight increase in circulating phosphate and PTH after EMPA treatment was not associated with evidence for increased bone mineral resorption suggesting that EMPA does not affect bone health.
Article
Full-text available
Activated factor X is a key component of the coagulation cascade, but whether it directly regulates pathological cardiac remodeling is unclear. In mice subjected to pressure overload stress, cardiac factor X mRNA expression and activity increased concurrently with cardiac hypertrophy, fibrosis, inflammation and diastolic dysfunction, and responses blocked with a low coagulation-independent dose of rivaroxaban. In vitro, neurohormone stressors increased activated factor X expression in both cardiac myocytes and fibroblasts, resulting in activated factor X-mediated activation of protease-activated receptors and pro-hypertrophic and -fibrotic responses, respectively. Thus, inhibition of cardiac-expressed activated factor X could provide an effective therapy for the prevention of adverse cardiac remodeling in hypertensive patients.
Article
Full-text available
There is growing evidence for the contribution of the activated coagulation factor X (FXa) in the development of chronic inflammatory lung diseases. Therefore, we aimed to investigate effects of exogenous FXa on mitochondrial and metabolic function as well as the induction of inflammatory molecules in type II alveolar epithelial cells. Effects of FXa on epithelial cells were investigated in A549 cell line. Activation of extracellular signal-regulated kinase (ERK) and induction of inflammatory molecules were examined by immunoblot and gene expression analysis. Mitochondrial function was assessed by the measurement of oxygen consumption during maximal oxidative phosphorylation and quantitative determination of cardiolipin oxidation. Apoptosis was tested using a caspase 3 antibody. Metabolic activity and lactate dehydrogenase assay were applied for the detection of cellular viability. FXa activated ERK1/2 and induced an increase in the expression of pro-inflammatory cytokines, which was prevented by an inhibitor of FXa, edoxaban, or an inhibitor of protease-activated receptor 1, vorapaxar. Exposure to FXa caused mitochondrial alteration with restricted capacity for ATP generation, which was effectively prevented by edoxaban, vorapaxar and GB83 (inhibitor of protease-activated receptor 2). Of note, exposure to FXa did not initiate apoptosis in epithelial cells. FXa-dependent pro-inflammatory state and impairment of mitochondria did not reach the level of significance in lung epithelial cells. However, these effects might limit regenerative potency of lung epithelial cells, particular under clinical circumstances where lung injury causes exposure to clotting factors.
Article
Full-text available
Aims Sodium–glucose co‐transporter 2 (SGLT2) inhibition reduces heart failure hospitalizations in patients with diabetes, irrespective of glycaemic control. We examined the effect of SGLT2 inhibition with empagliflozin (EMPA) on cardiac function in non‐diabetic rats with left ventricular (LV) dysfunction after myocardial infarction (MI). Methods and results Non‐diabetic male Sprague–Dawley rats underwent permanent coronary artery ligation to induce MI, or sham surgery. Rats received chow containing EMPA that resulted in an average daily intake of 30 mg/kg/day or control chow, starting before surgery (EMPA‐early) or 2 weeks after surgery (EMPA‐late). Cardiac function was assessed using echocardiography and histological and molecular markers of cardiac remodelling and metabolism were assessed in the left ventricle. Renal function was assessed in metabolic cages. EMPA increased urine production by two‐fold without affecting creatinine clearance and serum electrolytes. EMPA did not influence MI size, but LV ejection fraction (LVEF) was significantly higher in the EMPA‐early and EMPA‐late treated MI groups compared to the MI group treated with vehicle (LVEF 54%, 52% and 43%, respectively, all P < 0.05). EMPA also attenuated cardiomyocyte hypertrophy, diminished interstitial fibrosis and reduced myocardial oxidative stress. EMPA treatment reduced mitochondrial DNA damage and stimulated mitochondrial biogenesis, which was associated with the normalization of myocardial uptake and oxidation of glucose and fatty acids. EMPA increased circulating ketone levels as well as myocardial expression of the ketone body transporter and two critical ketogenic enzymes, indicating that myocardial utilization of ketone bodies was increased. Together these metabolic changes were associated with an increase in cardiac ATP production. Conclusion Empagliflozin favourably affects cardiac function and remodelling in non‐diabetic rats with LV dysfunction after MI, associated with substantial improvements in cardiac metabolism and cardiac ATP production. Importantly, it did so without renal adverse effects. Our data suggest that EMPA might be of benefit in heart failure patients without diabetes.
Article
Full-text available
Background Heart failure is associated with activation of thrombin-related pathways, which predicts a poor prognosis. We hypothesized that treatment with rivaroxaban, a factor Xa inhibitor, could reduce thrombin generation and improve outcomes for patients with worsening chronic heart failure and underlying coronary artery disease. Methods In this double-blind, randomized trial, 5022 patients who had chronic heart failure, a left ventricular ejection fraction of 40% or less, coronary artery disease, and elevated plasma concentrations of natriuretic peptides and who did not have atrial fibrillation were randomly assigned to receive rivaroxaban at a dose of 2.5 mg twice daily or placebo in addition to standard care after treatment for an episode of worsening heart failure. The primary efficacy outcome was the composite of death from any cause, myocardial infarction, or stroke. The principal safety outcome was fatal bleeding or bleeding into a critical space with a potential for causing permanent disability. Results Over a median follow-up period of 21.1 months, the primary end point occurred in 626 (25.0%) of 2507 patients assigned to rivaroxaban and in 658 (26.2%) of 2515 patients assigned to placebo (hazard ratio, 0.94; 95% confidence interval [CI], 0.84 to 1.05; P=0.27). No significant difference in all-cause mortality was noted between the rivaroxaban group and the placebo group (21.8% and 22.1%, respectively; hazard ratio, 0.98; 95% CI, 0.87 to 1.10). The principal safety outcome occurred in 18 patients who took rivaroxaban and in 23 who took placebo (hazard ratio, 0.80; 95% CI, 0.43 to 1.49; P=0.48). Conclusions Rivaroxaban at a dose of 2.5 mg twice daily was not associated with a significantly lower rate of death, myocardial infarction, or stroke than placebo among patients with worsening chronic heart failure, reduced left ventricular ejection fraction, coronary artery disease, and no atrial fibrillation. (Funded by Janssen Research and Development; COMMANDER HF ClinicalTrials.gov number, NCT01877915.)
Article
Full-text available
Apixaban is effective and safe for preventing stroke, and its usage has increased exponentially in recent years. However, data concerning the therapeutic range of apixaban is limited. This study determined the trough and peak levels of apixaban-specific anti-factor Xa activity (AFXaA) in acute ischemic stroke patients with non-valvular atrial fibrillation (NVAF) in Korea. The study included 85 patients who received apixaban. Blood samples were taken to measure the trough and peak levels of AFXaA using a chromogenic anti-factor assay, as well as prothrombin time (PT) and activated partial thromboplastin time (aPTT). We also reviewed complications such as major bleeding of patients treated with apixaban. In patients given a 5.0-mg apixaban dose, the median trough and peak levels of AFXaA were 104.5 and 202.0 ng/mL. In patients given a 2.5-mg apixaban dose, the median trough and peak AFXaA levels were 76.0 and 151.0 ng/mL. The PT showed a positive correlation with increased AFXaA activity at both levels (Trough R = 0.486, Peak R = 0.592), but the aPTT had no relationship with AFXaA activity at both levels (Trough R = 0.181, Peak R = 0.129). Two cases with intracranial bleeding belonged to the highest AFXaA quartile (Trough, p = 0.176; Peak, p = 0.053). In conclusion, we determined the trough and peak levels of AFXaA in patients with NVAF while being treated with the apixaban in Korea. Our results could be used as a starting point when setting the reference ranges for laboratories using anti-Xa assay. Large-scale studies are needed to establish the reference range for AFXaA in patients with NVAF.
Article
Importance Whether anticoagulation benefits patients with heart failure (HF) in sinus rhythm is uncertain. The COMMANDER HF randomized clinical trial evaluated the effects of adding low-dose rivaroxaban to antiplatelet therapy in patients with recent worsening of chronic HF with reduced ejection fraction, coronary artery disease (CAD), and sinus rhythm. Although the primary end point of all-cause mortality, myocardial infarction, or stroke did not differ between rivaroxaban and placebo, there were numerical advantages favoring rivaroxaban for myocardial infarction and stroke. Objective To examine whether low-dose rivaroxaban was associated with reduced thromboembolic events in patients enrolled in the COMMANDER HF trial. Design, Setting, and Participants Post hoc analysis of the COMMANDER HF multicenter, randomized, double-blind, placebo-controlled trial in patients with CAD and worsening HF. The trial randomized 5022 patients postdischarge from a hospital or outpatient clinic after treatment for worsening HF between September 2013 and October 2017. Patients were required to be receiving standard care for HF and CAD and were excluded for a medical condition requiring anticoagulation or a bleeding history. Patients were randomized in a 1:1 ratio. Analysis was conducted from June 2018 and January 2019. Intervention Patients were randomly assigned to receive 2.5 mg of rivaroxaban given orally twice daily or placebo in addition to their standard therapy. Main Outcomes and Measures For this post hoc analysis, a thromboembolic composite was defined as either (1) myocardial infarction, ischemic stroke, sudden/unwitnessed death, symptomatic pulmonary embolism, or symptomatic deep venous thrombosis or (2) all of the previous components except sudden/unwitnessed deaths because not all of these are caused by thromboembolic events. Results Of 5022 patients, 3872 (77.1%) were men, and the overall mean (SD) age was 66.4 (10.2) years. Over a median (interquartile range) follow-up of 19.6 (11.7-30.8) months, fewer patients assigned to rivaroxaban compared with placebo had a thromboembolic event including sudden/unwitnessed deaths: 328 (13.1%) vs 390 (15.5%) (hazard ratio, 0.83; 95% CI, 0.72-0.96; P = .01). When sudden/unwitnessed deaths were excluded, the results analyzing thromboembolic events were similar: 153 (6.1%) vs 190 patients (7.6%) with an event (hazard ratio, 0.80; 95% CI, 0.64-0.98; P = .04). Conclusions and Relevance In this study, thromboembolic events occurred frequently in patients with HF, CAD, and sinus rhythm. Rivaroxaban may reduce the risk of thromboembolic events in this population, but these events are not the major cause of morbidity and mortality in patients with recent worsening of HF for which rivaroxaban had no effect. While consistent with other studies, these results require confirmation in prospective randomized clinical trials. Trial Registration ClinicalTrials.gov identifier: NCT01877915
Article
Introduction: Rivaroxaban selectively inhibits factor Xa (FXa), which plays a central role in blood coagulation. In addition, FXa activates protease-activated receptor-2 (PAR-2). We have shown that PAR-2-/- mice exhibit less cardiac dysfunction after cardiac injury. Material and methods: Wild-type (WT) and PAR-2-/- mice were subjected to left anterior descending artery (LAD) ligation to induce cardiac injury and heart failure. Mice received either placebo or rivaroxaban chow either starting at the time of surgery or 3 days after surgery and continued up to 28 days. Cardiac function was measured by echocardiography pre-surgery and 3, 7 and 28 days after LAD ligation. We also measured anticoagulation, intravascular thrombi, infarct size, cardiac hypertrophy and inflammation at various times. Results: Rivaroxaban increased the prothrombin time and inhibited the formation of intravascular thrombi in mice subjected to LAD ligation. WT mice receiving rivaroxaban immediately after surgery had similar infarct sizes at day 1 as controls but exhibited significantly less impairment of cardiac function at day 3 and beyond compared to the placebo group. Rivaroxaban also inhibited the expansion of the infarct at day 28. Rivaroxaban did not significantly affect the expression of inflammatory mediators or a neutrophil marker at day 2 after LAD ligation. Delaying the start of rivaroxaban administration until 3 days after surgery failed to preserve cardiac function. In addition, rivaroxaban did not reduce cardiac dysfunction in PAR-2-/- mice. Conclusions: Early administration of rivaroxaban preserves cardiac function in mice after LAD ligation.
Article
It was previously found that patients with symptom of myocardial dysfunction had increased levels of thrombin. Apixaban is one of the novel oral anticoagulant drugs widely used in clinic. As the inhibitor of FXa (prothrombin), it inhibits prothrombin conversion into thrombin leading to thrombin deficiency in vivo. However, the effects of apixaban on myocardial fibrosis were still unclear, and the concomitant molecular mechanisms remain to be investigated. Here, we showed that myocardial fibrosis-bearing mice induced by continuous myocardial ischemia (MI) had higher levels of thrombin. Orally administration of apixaban significantly abrogated fibrosis condition and thrombin levels. In vitro, thrombin induced collagen deposition in primary cardiac fibroblasts in a dose-dependent manner. Mechanistic experiments showed that thrombin induced collagen deposition by activation of the Par-1-coupled Gq/PKC signaling. Genetic ablation of Gq or pharmacological inhibition of PKC effectively blunted thrombin-induced collagen deposition in cardiac fibroblasts. Moreover, administration of PKC inhibitor or Gq antagonist obviously blocked MI-induced myocardial fibrosis in mice. To conclude, apixaban attenuates MI-induced myocardial fibrosis by inhibition of thrombin-dependent Par-1/Gq/PKC signaling axis.
Article
Coronary embolism is the underlying cause of 3% of acute coronary syndromes but is often not considered in the differential of acute coronary syndromes. It should be suspected in the case of high thrombus burden despite a relatively normal underlying vessel or recurrent coronary thrombus. Coronary embolism may be direct (from the aortic valve or left atrial appendage), paroxysmal (from the venous circulation through a patent foramen ovale), or iatrogenic (following cardiac intervention). Investigations include transesophageal echocardiography to assess the left atrial appendage and atrial septum and continuous electrocardiographic monitoring to assess for paroxysmal atrial fibrillation. The authors review the historic and contemporary published data about this important cause of acute coronary syndromes. The authors propose an investigation and management strategy for work-up and anticoagulation strategy for patients with suspected coronary embolism.