Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2015

Open Access 01.12.2015 | Original investigation

The soluble guanylate cyclase activator cinaciguat prevents cardiac dysfunction in a rat model of type-1 diabetes mellitus

verfasst von: Csaba Mátyás, Balázs Tamás Németh, Attila Oláh, László Hidi, Ede Birtalan, Dalma Kellermayer, Mihály Ruppert, Sevil Korkmaz-Icöz, Gábor Kökény, Eszter Mária Horváth, Gábor Szabó, Béla Merkely, Tamás Radovits

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2015

Abstract

Background

Diabetes mellitus (DM) leads to the development of diabetic cardiomyopathy, which is associated with altered nitric oxide (NO)—soluble guanylate cyclase (sGC)—cyclic guanosine monophosphate (cGMP) signalling. Cardioprotective effects of elevated intracellular cGMP-levels have been described in different heart diseases. In the current study we aimed at investigating the effects of pharmacological activation of sGC in diabetic cardiomyopathy.

Methods

Type-1 DM was induced in rats by streptozotocin. Animals were treated either with the sGC activator cinaciguat (10 mg/kg/day) or with placebo orally for 8 weeks. Left ventricular (LV) pressure–volume (P–V) analysis was used to assess cardiac performance. Additionally, gene expression (qRT-PCR) and protein expression analysis (western blot) were performed. Cardiac structure, markers of fibrotic remodelling and DNA damage were examined by histology, immunohistochemistry and TUNEL assay, respectively.

Results

DM was associated with deteriorated cGMP signalling in the myocardium (elevated phosphodiesterase-5 expression, lower cGMP-level and impaired PKG activity). Cardiomyocyte hypertrophy, fibrotic remodelling and DNA fragmentation were present in DM that was associated with impaired LV contractility (preload recruitable stroke work (PRSW): 49.5 ± 3.3 vs. 83.0 ± 5.5 mmHg, P < 0.05) and diastolic function (time constant of LV pressure decay (Tau): 17.3 ± 0.8 vs. 10.3 ± 0.3 ms, P < 0.05). Cinaciguat treatment effectively prevented DM related molecular, histological alterations and significantly improved systolic (PRSW: 66.8 ± 3.6 mmHg) and diastolic (Tau: 14.9 ± 0.6 ms) function.

Conclusions

Cinaciguat prevented structural, molecular alterations and improved cardiac performance of the diabetic heart. Pharmacological activation of sGC might represent a new therapy approach for diabetic cardiomyopathy.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12933-015-0309-x) contains supplementary material, which is available to authorized users.
Béla Merkely and Tamás Radovits contributed equally to this study
Abkürzungen
α-MHC
myosin heavy chain alfa
ANF
atrial natriuretic factor
BAX
Bcl2-associated X protein
Bcl-2
B-cell CLL/lymphoma 2
β-MHC
myosin heavy chain beta
BW
body weight
Col1
Collagen 1a1
Col3
Collagen 3a1
CO
cardiac output
dP/dtmax
maximal slope of systolic pressure increment
dP/dtmin
maximal slope of diastolic pressure decrement
EDPVR
end-diastolic pressure–volume relationship
Ees
slope of the left ventricular end-systolic pressure–volume relationship
EF
ejection fraction
EIA
enzyme immunoassay
eNOS
endothelial nitric oxide synthase
ESPVR
end systolic pressure–volume relationship
GADPH
glyceraldehyde-3-phosphate dehydrogenase
H&E
hematoxylin and eosin
HR
heart rate
HSP70a1
heat shock 70 kD protein 1A
HW
heart weight
LV
left ventricle
LVEDP
left ventricular end-diastolic pressure
LVSP
left ventricular systolic pressure
MAP
mean arterial pressure
MMP-2
matrix metallopeptidase 2
MMP-9
matrix metallopeptidase 9
MT
Masson’s trichrome
NO
nitric oxide
NT
nitrotyrosine
PDE-5
phosphodiesterase 5
PKG
protein kinase G
PRSW
preload recruitable stroke work
P–V
pressure–volume
ROS
reactive oxygen species
RNS
reactive nitrogen species
sGC
soluble guanylate cyclase
SOD-2
superoxide-dismutase 2
STZ
streptozotocin
SW
stroke work
TIMP-1
tissue inhibitor of matrix metallopeptidase 1
TIMP-2
tissue inhibitor of matrix metallopeptidase 2
VASP
vasodilator-stimulated phosphoprotein

Background

Diabetes mellitus (DM) is associated with cardiovascular complications, such as myocardial infarction, chronic heart failure or vascular diseases. It acts as an independent risk factor for coronary atherosclerosis and ischemic heart disease, however, the altered metabolic state—due to elevated glucose levels—has a direct impact on cardiac structure and function independently of coronary artery disease. Thus DM leads to the development of a special disease entity, termed diabetic cardiomyopathy [1]. Although, both systolic and diastolic dysfunction [25] as well as several key morphological and cellular/subcellular features (including myocardial fibrotic remodelling, cardiomyocyte hypertrophy, nitro-oxidative stress, inflammation and apoptosis) have been described in diabetic cardiomyopathy, the exact mechanisms in the pathophysiology are still unknown [1, 69].
In the cardiovascular system, under physiological conditions, nitric oxide (NO) is produced by the endothelial nitric oxide synthase (eNOS) in endothelial cells and diffuses into target cells, such as vascular smooth muscle cells or cardiomyocytes. It activates its intracellular receptor, the soluble guanylate cyclase enzyme (sGC) which results in the rapid formation of the second messenger cyclic guanosine monophosphate (cGMP). cGMP activates the cGMP-dependent protein kinase (PKG), which mediates most of its physiological effects, such as vasodilatation or inhibition of platelet aggregation [10]. The NO–sGC–cGMP–PKG signalling pathway has been described to get disturbed in DM through several mechanisms, such as increased formation of reactive nitrogen (RNS) and oxygen species (ROS) (nitro-oxidative stress), eNOS uncoupling and decreased NO bioavailability, increased expression of the cGMP-degrading enzyme phosphodiesterase (PDE)-5 [1]. Our research group and others demonstrated protective effects of enhanced cGMP-signalling via pharmacological inhibition of PDE-5 in several cardiovascular diseases [1115] and in DM in particular [3, 16].
Nitro-oxidative stress directly deteriorates the structure of sGC resulting in a heme-deficient, NO-insensitive and inactive form of the enzyme [17]. Therefore, drugs that are able to activate the NO-insensitive form of sGC thus reactivating it might have cardioprotective effects in various pathological conditions through the enhancement of the impaired cGMP-signalling. The sGC activator cinaciguat (BAY 58-2667) has been reported to bind to the heme pocket of NO-insensitive sGC [17] thus increasing its cGMP-producing activity and augmenting cGMP-PKG signalling. Recent studies described its beneficial effects in experimental myocardial infarction [18], myocardial ischemia/reperfusion injury [19, 20], endothelial dysfunction induced by nitro-oxidative stress [21], vascular neointima formation [22] or in prevention of cardiomyocyte hypertrophy [23].
The safety and tolerability of cinaciguat have been assessed by phase-I human clinical trials [24]. Although cinaciguat has demonstrated efficacy in a proof-of-concept study in patients with acute decompensated heart failure [25], the phase-IIb clinical studies for the same indication has been terminated prematurely due to the high number of adverse events (mainly a significant drop of blood pressure) during acute intravenous application [26, 27]. The investigators concluded that rather a chronic, long term oral administration of the compound could improve endothelial and preserve myocardial function in heart failure patients [28].
Based upon the concept of cardioprotection by enhanced cGMP-signalling via chronic pharmacological activation of the sGC enzyme, we investigated in the present study whether long-term administration of the sGC-activator cinaciguat can preserve left ventricular (LV) systolic and diastolic function and prevent DM-associated myocardial alterations in the rat model of streptozotocin (STZ)-induced experimental type-1 DM.

Methods

Animals

The investigation conformed to the EU Directive 2010/63/EU guidelines and the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication No. 85–23, revised 1996) and was reviewed and approved by the appropriate institutional and national ethical committees (permission number: 22.1/1162/3/2010). All animals received humane care.
8-week-old male Sprague–Dawley rats (n = 46; body weight (BW): 225–250 g) (Charles River, Sulzfeld, Germany) were housed individually in a room (constant temperature, 12/12 h light/dark cycles) and received standard laboratory rat diet and water ad libitum.

Induction of DM

Type-1 DM was induced with a single i.p. injection of STZ (60 mg/kg) in our rats as described previously [3]. STZ was freshly dissolved in citrate buffer (0.1 M). Control animals received only the buffer. 72 h after the injection a drop of blood was collected from the tail vein and blood glucose concentration was determined by using a digital blood glucose meter (Accu-Chek® Sensor, Roche, Mannheim, Germany). Animals with a random blood glucose level >15 mmol/l were considered to be diabetic and were included into the study.

Experimental groups, chronic treatment protocol

After confirmation of DM, rats were randomised into four groups: vehicle-treated control (Co; n = 12), cinaciguat-treated control (CoCin; n = 12), vehicle-treated diabetic (DiabCo; n = 12) and cinaciguat-treated diabetic (DiabCin; n = 10) groups. Animals were treated for 8 weeks with 0.5 % methylcellulose (Co, DiabCo) vehicle or with the sGC activator cinaciguat in suspension p.o. (CoCin, DiabCin; 10 mg/kg/day), starting immeadiately after DM confirmation. Water bottles were filled every morning with the same amount of fresh tap water and daily water intake was measured. Animal cages were handled with care and were not moved after water bottle replacement to prevent spilling of water from the bottles. Body weight of the animals were recorded every 2 days and the dose of cinaciguat was adjusted accordingly.

Hemodynamic measurements

24–28 h after the last administration of cinaciguat/vehicle, LV pressure–volume (P–V) analyses were carried out using a pressure-conductance microcatheter system (MPVS-Ultra, Millar Instruments, Houston, TX, USA) to evaluate LV performance. Animals were anesthetised with a mixture of xylazine (3 mg/kg) and ketamin (100 mg/kg) i.p. [3, 29]. Mean arterial blood pressure (MAP), heart rate (HR), maximal LV systolic pressure (LVSP), LV end-diastolic pressure (LVEDP), maximal slope of systolic pressure increment (dP/dtmax) and diastolic pressure decrement (dP/dtmin), time constant of LV pressure decay (Tau), ejection fraction (EF), stroke work (SW) and cardiac output (CO) were calculated. The slope (Ees) of the LV end-systolic P–V relationships (ESPVR; according to the parabolic curvilinear model) and preload recruitable stroke work (PRSW) were calculated as load-independent indexes of LV contractility while the slope of the LV end-diastolic P–V relationship (EDPVR) was calculated as reliable index of LV diastolic stiffness. For detailed hemodynamic description see Additional file 1.

Biochemical measurements

A drop of blood was collected from the tail vein and blood glucose concentration was determined using a digital blood glucose meter (Accu-Chek® Sensor, Roche). At the end of the hemodynamic measurements blood was collected from the inferior caval vein, plasma samples were prepared and stored at −80 °C.
Plasma cGMP levels were determined by enzyme immunoassay (EIA) using a commercial kit (Amersham cGMP EIA Biotrak System, GE Healthcare, Buckinghamshire, UK).

Myocardial mRNA analysis

We performed quantitative real-time polymerase chain reaction (qRT-PCR) experiments as described previously [30]. Frozen LV samples were homogenised, total RNA was isolated. Reverse transcription was performed and cDNA samples were amplified on the StepOnePlus™ Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) using TaqMan® Universal PCR MasterMix and TaqMan® Gene Expression Assays (Applied Biosystems) for the following targets: atrial natriuretic factor (ANF), α myosin heavy chain (MHC) and β-MHC (ratio of β/α-MHC expression was assessed as pathological cardiomyocyte hypertrophy marker), antiapoptotic mediator B cell CLL/lymphoma-2 (Bcl-2), proaptotic mediator Bcl2-associated X protein (BAX), eNOS, mediators of cardiac remodelling such as collagen 1a1 (Col1), 3a1 (Col3) and fibronectin, matrix metallopeptidase (MMP)-2 and MMP-9 and their endogenous inhibitors tissue inhibitor of MMP (TIMP)-1 and TIMP-2, members of different antioxidant systems like catalase, thioredoxin-1, gluthatione-reductase, superoxide dismutase (SOD)-2 and heat shock 70 kD protein 1A (HSP70a1). Gene expression data were normalised to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The mRNA expression levels were calculated using the CT comparative method (2−ΔCT) and adjusted to a positive calibrator. For detailed description see Additional file 1.

Immunoblot analysis

LV tissue samples were homogenised, protein concentration was determined and equal amounts of protein were separated via gel-electrophoresis. Proteins were transferred to nitrocellulose membranes. After blocking, membranes were incubated with primary antibodies against various targets: eNOS (1:1000, SC-654, SantaCruz Biotechnology, Santa Cruz, CA, USA), sGC β1 (1:1000, NB100-91798, Novus Biologicals, Cambridge, UK), PDE-5 (1:2000, ALX-210-099, Enzo Life Sciences, Farmingdale, NY, USA), PKG (1:2000, ADI-KAP-PK005-F, Enzo Life Sciences), vasodilator-stimulated phosphoprotein (VASP) and phospho-VASP (1:1000, 3112 and 1:2000, 3114, Cell Signaling, Danvers, MA, USA) as marker of PKG activity, profibrotic mediator transforming growth factor (TGF)-β1 (1:250, SC-146, SantaCruz Biotechnology), MMP-2 (1:5000, NB200-193, Novus Biologicals) and MMP-9 (1:1000, SC-6840, SantaCruz Biotechnology). After washing, membranes were incubated in horseradish peroxidase-conjugated secondary antibody. Immunoblots were developed by enhanced chemiluminescence detection. Protein band densities were quantified using GeneTools software (Syngene, Frederick, MD, USA). After adjusting protein band densities to GAPDH (1:10000, MAB374, Millipore, Billerica, MA, USA), band density values were normalised to the average value of the Co group for statistical analysis. Detailed description of immunoblot analysis is available as Additional file 1.

Histology, immunohistochemistry

LV sections were stained with hematoxylin-eosin (H&E) and Masson’s trichrome (MT) to examine histopathological characteristics and interstitial myocardial fibrotic remodelling, respectively. To evaluate cardiomyocyte hypertrophy, transverse transnuclear widths (cardiomyocyte diameter) were measured of 100 longitudinally oriented, mono-nucleated cardiomyocytes on H&E stained LV sections cut on the same plane. Immunohistochemistry for the fibrosis marker fibronectin (Sigma-Aldrich, Budapest, Hungary), for the profibrotic mediator TGF-β1 (Santa Cruz Biotechnology), for the sGC derived second messenger cGMP (AbD Serotec, Düsseldorf, Germany) and for the nitro-oxidative stress marker nitrotyrosine (NT) (Millipore) were performed and analysed. Histological and immunohistochemical analyses were performed by two blinded observers. For detailed description see Additional file 1.

Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay

TUNEL assay was performed to detect DNA strand breaks using a commercial kit (DeadEnd™ Colorimetric TUNEL System, Promega, Mannheim, Germany). TUNEL positive cell nuclei were counted in 20 randomly selected fields of each section by two blinded observers. Data were normalised to the mean value of the Co group for statistical analysis.

Drugs

The sGC activator cinaciguat (BAY 58-2667; 4-({(4-carboxybutyl)[2-(2-{[4-(2-phenylethyl)benzyl]oxy}phenyl)ethyl]amino}methyl)benzoic acid) was provided by Bayer HealthCare (Wuppertal, Germany) for oral application. Cinaciguat was suspended in 0.5 % methylcellulose solution. STZ was purchased from Sigma-Aldrich (Taufkirchen, Germany).

Statistics

Data are presented as means and standard errors of the mean (SEM). After testing normal distribution of the data, two-factorial analysis of variance (ANOVA) (with diabetes and drug treatment as factors) was carried out. A P value of <0.05 was used as a criterion of significance. Where F value of diabetes × treatment interaction reached the level of significance Tukey HSD post hoc testing was performed to evaluate differences between the groups. Data that did not show normal distribution were transformed logarithmically before performing two-factorial ANOVA.

Results

Body weight, heart weight (HW) and glucose levels

All animals survived the study period and reached the end-point of the investigation. HW and BW significantly decreased in both diabetic groups while HW to BW ratio increased in the diabetic groups (Table 1). When compared with controls, DM led to significantly increased blood glucose levels and daily water intake. Cinaciguat treatment in diabetic rats did not influence blood glucose levels, but led to attenuated water intake (Table 1). Time-course of body weight changes is available as Additional file 2: Figure S1.
Table 1
Basic characteristics of study groups
Characteristic
Co
CoCin
DiabCo
DiabCin
Pdiabetes
Ptreatment
Pinteraction
Blood glucose (mmol/l)
5.8 ± 0.1
6.2 ± 0.1
30.8 ± 0.5*
29.3 ± 1.4*
<0.001
0.460
0.209
Water intake (ml/gBW/day)
0.078 ± 0.002
0.090 ± 0.002
0.787 ± 0.006*
0.597 ± 0.013*
<0.001
<0.001
<0.001
Heart weight (g)
1.20 ± 0.06
1.24 ± 0.07
0.91 ± 0.04*
0.84 ± 0.03*
<0.001
0.805
0.278
Body weight (g)
480.7 ± 17.6
477.8 ± 20.9
293.5 ± 11.1*
247.6 ± 14.8*
<0.001
0.150
0.202
Heart weight/body weight
0.249 ± 0.008
0.259 ± 0.008
0.311 ± 0.009*
0.348 ± 0.015*
<0.001
0.026
0.196
The values of blood glucose, daily water intake, heart weight, body weight (BW) and heart weight to body weight ratio are shown of the study groups—vehicle-treated controls (Co), cinaciguat-treated controls (CoCin), vehicle-treated diabetic (DiabCo) and cinaciguat-treated diabetic (DiabCin) groups. Values are mean ± SEM of 10–12 experiments per group
* P < 0.05 vs. Co
P < 0.05 vs. DiabCo (Tukey HSD test)

Effects of cinaciguat on plasma and myocardial cGMP levels in DM

Cinaciguat treatment had no effect on plasma cGMP levels in control animals (Fig. 1a), however it resulted in a pronounced increase of plasma cGMP in DM (Fig. 1a). According to cGMP immunohistochemistry, the cGMP content of LV myocardium was significantly lower in DM than in controls (Fig. 1b, c). However, chronic treatment with cinaciguat restored cGMP to the control level (Fig. 1b, c).

Effects of diabetes mellitus and cinaciguat treatment on myocardial nitro-oxidative stress

DM was associated with increased NT immunoreactivity in LV myocardium referring to pronounced nitro-oxidative stress which was significantly alleviated by cinaciguat treatment (Fig. 2a, b). Increased expression of HSP70a1, catalase, gluthatione-reductase and thioredoxin-1 was found in DM (Fig. 2c). HSP70a1 and gluthatione-reductase mRNA expression in the DiabCin group remained on the level of healthy controls (Fig. 2c). SOD-2 expression did not show any difference among the groups (Fig. 2c).

Cinaciguat protects against DM related alteration of the NO-sGC-cGMP-PKG signalling

Protein expression of eNOS and sGC β1 did not differ between healthy and diabetic rats (Fig. 3a) while eNOS gene expression was significantly lower in both diabetic groups (Fig. 3b). We detected elevated PDE-5 and PKG protein expression in the DiabCo group (Fig. 3a) whereas the p-VASP/VASP ratio (marker of PKG activity) was significantly reduced, showing severe deterioration of PKG signalling in DM (Fig. 3a). Application of cinaciguat in diabetic animals significantly reduced the expression of PDE-5, markedly increased PKG activity (as indicated by elevated p-VASP/VASP ratio) (Fig. 3a), while the expression of PKG did not differ between the two diabetic groups (Fig. 3a).

Cinaciguat treatment protects against DM related fibrotic remodelling of the myocardium

DM was associated with dysregulation of the MMP system indicated by markedly increased MMP-9/TIMP-1 and reduced MMP-2/TIMP-2 gene expression ratios (Fig. 4a). These alterations were attenuated in the DiabCin group (Fig. 4a). Although fibronectin expression remained unchanged, Col1 and Col3 expression levels were significantly lower in both diabetic groups (Fig. 4a). The profibrotic TGF-β1 showed increased expression in the diabetic animals, which was significantly ameliorated by cinaciguat (Fig. 4c). Expression of MMP-9 showed a twofold increase in DM, while MMP-2 remained unchanged (Fig. 4c). Cinaciguat did not significantly alter these parameters (Fig. 4c). We found severe interstitial fibrosis of diabetic myocardium indicated by increased MT staining (Fig. 4c). Additionally, elevated immunoreactivity was observed against the profibrotic mediator TGF-β1 and fibrosis marker fibronectin in the diabetic heart (Fig. 4c). Application of cinaciguat reduced MT staining intensity of diabetic myocardium (Fig. 4c) while TGF-β1 immunoreactivity strongly tended to decrease (P = 0.051) (Fig. 4c).
Myocardial mRNA expression data from LV myocardium of diabetic rats showed a marked increase of ANF and the β-MHC/α-MHC ratio (Fig. 5a). Treatment with cinaciguat caused a significant decrease of ANF expression in DM (Fig. 5a) while β-MHC/α-MHC ratio showed slight decrease (Fig. 5a). When compared with controls, increased cardiomyocyte width was observed in the DiabCo group indicative of cardiomyocyte hypertrophy. This increase was completely prevented by cinaciguat (Fig. 5b, c). The mRNA expression levels of proapoptotic BAX and antiapoptotic Bcl-2 did not differ among the study groups resulting in unchanged BAX/Bcl-2 ratio (Fig. 5a). DM was associated with increased TUNEL positivity in LV myocardium referring to pronounced DNA fragmentation (Fig. 5b, c). TUNEL positivity was effectively reduced in the DiabCin group (Fig. 5b, c).

In vivo cardiac function is improved by cinaciguat in DM

In comparison with non-diabetic controls DiabCo group showed remarkably reduced MAP, LVSP, EF, SW, dP/dtmax and impaired dP/dtmin values, while LVEDP and Tau increased, indicating LV systolic and diastolic dysfunction (Table 2). HR significantly decreased in the diabetic groups while CO was not significantly different among the study groups (Table 2). MAP, LVSP, SW, dP/dtmax and dP/dtmin remained unchanged in the DiabCin group, however drug treatment markedly improved LVEDP and Tau in DM (Table 2). As a result of cinaciguat treatment EF tended towards improvement (P = 0.054) in DM (Table 2).
Table 2
Basic hemodynamic data of the study groups
Variable
Co
CoCin
DiabCo
DiabCin
Pdiabetes
Ptreatment
Pinteraction
HR (beats/min)
231 ± 11
246 ± 12
208 ± 8*
204 ± 10*
0.003
0.608
0.351
MAP (mmHg)
80.0 ± 2.0
81.0 ± 3.5
63.7 ± 2.5*
64.4 ± 3.1*
<0.001
0.845
0.969
LVSP (mmHg)
99.5 ± 2.6
103.5 ± 2.1
85.5 ± 1.3*
82.3 ± 2.3*
<0.001
0.858
0.102
SW (mmHg µl)
14,561 ± 1060
13,293 ± 948
9789 ± 592*
12,032 ± 1067
0.003
0.611
0.072
CO (µl/min)
42,347 ± 2472
41,306 ± 2804
33,360 ± 2162
38,605 ± 4288
0.057
0.484
0.297
EF (%)
70.42 ± 2.50
68.17 ± 2.70
58.09 ± 2.54*
68.02 ± 2.56
0.021
0.146
0.024
dP/dtmax (mmHg/s)
6539 ± 240
6804 ± 188
4933 ± 207*
4785 ± 230*
<0.001
0.791
0.350
dP/dtmin (mmHg/s)
−6135 ± 362
−6570 ± 446
−3883 ± 133*
−3723 ± 248*
<0.001
0.679
0.374
LVEDP (mmHg)
7.0 ± 0.6
7.2 ± 0.4
9.7 ± 0.7*
6.8 ± 0.3†
0.026
0.082
0.034
Tau (Weiss; ms)
10.3 ± 0.3
10.1 ± 0.3
17.3 ± 0.8*
14.9 ± 0.6*†
<0.001
0.016
0.054
Heart rate (HR), mean arterial pressure (MAP), maximal left ventricular (LV) systolic pressure (LVSP), stroke work (SW), cardiac output (CO), ejection fraction (EF), maximal slope of systolic pressure increment (dP/dtmax) and diastolic pressure decrement (dP/dtmin), LV end-diastolic pressure (LVEDP) and time constant of LV pressure decay (Tau) are shown. Groups: vehicle-treated controls (Co), cinaciguat-treated controls (CoCin), vehicle-treated diabetic (DiabCo) and cinaciguat-treated diabetic (DiabCin) animals. Values are mean ± SEM of 9–11 experiments per group
* P < 0.05 vs. Co
P < 0.05 vs. DiabCo (Tukey HSD test)
The values of load-independent, P–V-loop derived contractility indexes (Ees, PRSW) were significantly reduced in diabetic animals indicating severe contractile dysfunction (Fig. 6a, b) Treatment with cinaciguat led to a significant increase in PRSW (Fig. 6b) while Ees showed a strong tendency towards improvement in DM (P = 0.092) (Fig. 6b).
In comparison with Co group the slope of EDPVR tended to increase (P = 0.063) in diabetic animals, which was significantly reduced by cinaciguat (Fig. 6b). Cinaciguat had no hemodynamic effects in non-diabetic rats.

Discussion

In the current study we investigated the effects of the sGC activator cinaciguat on DM related cardiac alterations in an experimental type-1 diabetic animal model. We found that chronic application of cinaciguat did not affect blood glucose levels, however it effectively raised cGMP levels and restored cGMP-PKG signalling in the myocardium. These molecular changes were associated with attenuated cardiomyocyte hypertrophy, less severe fibrotic remodelling and markedly improved systolic and diastolic cardiac performance.
Rat model of STZ induced type-1 DM is widely used in research studies aiming at the characterisation of diabetic cardiomyopathy and its possible new therapeutic approaches. STZ is taken up via glucose transporter 2 to the pancreatic beta cells which ensures its selective toxicity for beta cells and the ability to induce insulin-dependent DM. In the present study increased blood glucose and daily water intake levels indicated obvious evidence of pronounced DM in STZ-treated rats which was concordant with earlier data [31].
The most important cause in DM that presumably leads to severe complications, including diabetic cardiomyopathy, is hyperglycemia. The imbalance in glucose homeostasis triggers numerous mechanisms and signalling pathways which result in cardiovascular dysfunction. Despite the fact, that many studies investigated its pathological background so far, the exact pathomechanisms of diabetic cardiomyopathy are still poorly understood. It is known that hyperglycemia increases production of ROS and RNS and subsequent upregulation of endogenous antioxidant systems and overexpression of heat shock proteins [32] through several mechanisms [1]. As a marker of excessive nitro-oxidative stress [33] a significantly higher NT positivity was shown in the diabetic myocardium which might have contributed to the observed induction of endogenous antioxidant systems (see thiorexodin-1, catalase or gluthatione-reductase gene expression levels) and to a remarkable heat shock response (increased HSP70a1 level). Pronounced nitro-oxidative stress can lead to the oxidation of the sGC enzyme which causes the loss of its heme prosthetic group and results in a NO-insensitive (inactive) form of sGC. Additionally, an imbalance of the different types of phosphodiesterases was presented in the failing heart which might have contributed to the rapid degradation of intracellular cGMP [34]. Decreased cGMP levels and subsequent impairment of NO-cGMP-PKG signalling is a well-known feature occuring in the diabetic myocardium. This observation was confirmed by our results showing reduced myocardial cGMP content in diabetic rats while sGC β1 and eNOS protein expression remained unchanged. Moreover, NO-cGMP-PKG signalling showed a severe deterioration in the diabetic group indicated by a reduced gene expression level of eNOS, increased PDE-5 protein levels (in accordance with previous data [35]) and significantly reduced phosphorylation of the PKG target VASP (lower p-VASP/VASP ratio, an index of PKG activity [23]). However, elevated PKG protein expression was observed which might reflect an ineffective compensatory mechanism in the LV. The presumable loss of the widely described cGMP related cytoprotection was associated with a four times higher extent of DNA damage in DM compared with controls (indicated by the TUNEL reaction). The observed discrepancy between impairment of myocardial cGMP-signalling and unchanged plasma cGMP levels in DiabCo vs. Co animals might be the consequence of diabetes-associated increase in ANF levels and the subsequent activation of natriuretic peptide receptors and particulate guanylate cyclase (pGC) in organs other than the heart, as plasma cGMP is seen as an overspill of intracellular cGMP from various tissues [36].
The sGC activator cinaciguat has been described to exert disease specific effects since it preferentially activates the oxidised, inactive form of the enzyme (that is present mainly in diseased conditions) thus restores its cGMP producing ability [17] in various pathological conditions associated with excessive nitro-oxidative stress [19, 21]. With the application of cinaciguat we achieved massive cytoprotective effects in DM, that was confirmed by lower NT staining (indicative of attenuated nitro-oxidative stress), normalised expression of endogenous antioxidant and heat shock protein systems as well as lower extent of DNA breaks (less severe TUNEL positivity). The mechanism by which cinaciguat decreased nitro-oxidative stress remains unclear and needs further investigations. A possible explanation might be the cGMP-induced down-regulation of NADPH-oxidases, as proposed by a recent experimental work [37]. By reducing oxidative damage to the myocardium with cinaciguat, pathologically high levels of PDE-5 enzyme reversed to the level of control animals, which might have contributed to the accumulation of cGMP in the diabetic heart and plasma, too. The augmented cGMP production consequently reinforced the impaired PKG activity (proven by markedly increased myocardial p-VASP/VASP ratio) that further contributed to the cardioprotective effects of cinaciguat in DM.
Pathological remodelling is a well-known phenomenon in the diabetic myocardium, however there are several remaining questions regarding the underlying mechanisms to be clarified. A number of key pathophysiological features such as cardiac hypertrophy, fibrotic remodelling, myocardial tissue injury with DNA fragmentation are lying in the background of diabetic cardiomyopathy [1]. In our diabetic rat model pathological cardiomyocyte hypertrophy was developed demonstrated by increased cardiomyocyte diameter, increased ANF gene expression values and a shift in the β/α-MHC expression ratio. Chronic pharmacological activation of sGC showed antihypertrophic effects not only on the histological level (decreased cardiomyocyte diameter) but also in the molecular environment (significantly reduced ANF gene expression, strong tendency towards reversed shift in β/α-MHC ratio). Our results are concordant with data from a recent publication [23] demonstrating antihypertrophic and antifibrotic effects of cinaciguat in cultured cells. Fibrotic remodelling of the diabetic myocardium is strongly associated with dysregulation of MMPs [1], increased fibroblast proliferation [38], decreased Col1 and Col3 mRNA expression [39] and TGF-β1 signalling [40]. In accordance with previous literature data we found an upregulation of the profibrotic mediator TGF-β1, downregulation of Col1 and Col3 mRNA levels, intense MT and fibronectin staining (as fibrosis markers) and increased DNA fragmentation in diabetic cardiomyopathy. Data from our experiments revealed that treatment with the sGC activator cinaciguat effectively prevented pathological changes in the myocardium by improving MMP dysregulation (reverting altered MMP-9/TIMP-1 and MMP-2/TIMP-2 ratios), by decreasing myocardial level of TGF-β1 and preventing fibrotic changes, as well as repressing myocardial TUNEL positivity. Interestingly, we did not find any differences in BAX/Bcl-2 ratio among the study groups which suggest that classical apoptotic pathways are not involved in the observed beneficial effects. A possible explanation might be a cross-talk between sGC-cGMP axis and TGF-β-dependent extracellular signal-regulated kinase signalling (as reported by Beyer et al. [41]), however this hypothesis requires further investigations.
Several clinical and experimental studies have focused on the investigation of cardiac function in DM. Both systolic and diastolic dysfunctions have been described in type-1 diabetes in the literature using different non-invasive and invasive methods [2, 31]. We found similar alterations in our current study regarding DM related cardiac dysfunction. We observed a significant reduction of MAP (probably as a consequence of diabetic polyuria [42]) in DM compared with controls that was not affected by chronic cinaciguat treatment. Whether cinaciguat administration acutely affects MAP in diabetic rats, needs further non-invasive investigations. DiabCo group showed significantly deteriorated systolic function (as reflected by decreased LVSP, SW, EF and dP/dtmax). EF and dP/dtmax are widely used to assess contractile function, however they are known to be affected by pre- and afterload [29]. Therefore, in the present study we determined P–V-loop derived, pre- and afterload independent contractility parameters (Ees, PRSW) that are more informative on intrinsic myocardial contractile function [29]. The specific indexes of LV contractility showed severe systolic dysfunction in the DiabCo group. As a result of sGC activation a remarkable improvement of systolic function has been achieved in diabetic animals: a significant increase of PRSW clearly reflects improved LV contractility, whereas EF and SW were restored to the levels of healthy controls. When investigating diastolic function we found significantly deteriorated LV active relaxation (as shown by impaired dP/dtmin and prolonged Tau) and markedly elevated LV diastolic stiffness (indicated by higher LVEDP and strong tendency towards increased EDPVR) in DM compared with controls. Cinaciguat effectively improved not only active relaxation (by remarkably shortening Tau) but also diastolic stiffness (by reverting LVEDP and EDPVR values).
Study limitations: Results in the current study are limited to young, male, type-1 diabetic rats. Rats did not receive any glucose-lowering medication. Acute effects of pharmacological activation of sGC on hemodynamics were not investigated. A possible explanation of cinaciguat’s beneficial effects in DM could be the improvement of cardiac microvascular perfusion, however that needs further investigations.

Conclusions

To our knowledge, this is the first study reporting the effects of pharmacological sGC activation on diabetic cardiomyopathy and DM related myocardial dysfunction. We confirmed that the sGC activator cinaciguat improves pathological features of diabetic cardiomyopathy without affecting blood glucose levels. Molecular mechanisms underlying the advantagous effects of sGC activation include the upregulation of NO-sGC-cGMP-PKG axis which might crosstalk with antioxidant, antihypertrophic and antifibrotic cascades hence preventing cardiac complications of diabetic metabolism and improving DM related cardiac dysfunction. Based on the data presented here pharmacological sGC activation might be a potential therapeutic approach to improve cardiovascular dysfunction in DM.

Authors’ contributions

CM, SK, GK, GS, BM and TR were involved in study design and conception. CM, BTN, AO, LH, EB, DK, MR, EMH  and TR were involved in data acquisition. CM, BTN, AO, GK and TR conducted data analysis. CM and TR wrote the first draft of the manuscript. SK, GK, GS, BM and TR reviewed the manuscript for important intellectual content. CM, BM and TR are responsible for the integrity of the work as a whole. All authors read and approved the final manuscript.

Acknowledgements

Technical assistance of Henriett Biró, Gábor Fritz, Gábor Alt, Tímea Fischinger, Krisztina Fazekas and Viktória Gregor is gratefully acknowledged. The authors gratefully acknowledge for Johannes-Peter Stasch from Bayer HealthCare (Wuppertal, Germany) for providing cinaciguat. This work was supported by the Hungarian Scientific Research Fund (OTKA PD100245 to T.R.), by the János Bolyai Research Scholarship of the Hungarian Academy of Sciences (to T.R.) and by the European Social Fund in the framework of TÁMOP 4.2.4. A/1-11-1-2012-0001’National Excellence Program’ (fellowship to B.T.N.).

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Huynh K, Bernardo BC, McMullen JR, Ritchie RH. Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther. 2014;142:375–415.CrossRefPubMed Huynh K, Bernardo BC, McMullen JR, Ritchie RH. Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther. 2014;142:375–415.CrossRefPubMed
2.
Zurück zum Zitat Radovits T, Korkmaz S, Loganathan S, et al. Comparative investigation of the left ventricular pressure-volume relationship in rat models of type 1 and type 2 diabetes mellitus. Am J Physiol Heart Circ Physiol. 2009;297:H125–33.CrossRefPubMed Radovits T, Korkmaz S, Loganathan S, et al. Comparative investigation of the left ventricular pressure-volume relationship in rat models of type 1 and type 2 diabetes mellitus. Am J Physiol Heart Circ Physiol. 2009;297:H125–33.CrossRefPubMed
3.
Zurück zum Zitat Radovits T, Bomicke T, Kokeny G, et al. The phosphodiesterase-5 inhibitor vardenafil improves cardiovascular dysfunction in experimental diabetes mellitus. Br J Pharmacol. 2009;156:909–19.PubMedCentralCrossRefPubMed Radovits T, Bomicke T, Kokeny G, et al. The phosphodiesterase-5 inhibitor vardenafil improves cardiovascular dysfunction in experimental diabetes mellitus. Br J Pharmacol. 2009;156:909–19.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Poirier P, Bogaty P, Garneau C, Marois L, Dumesnil JG. Diastolic dysfunction in normotensive men with well-controlled type 2 diabetes: importance of maneuvers in echocardiographic screening for preclinical diabetic cardiomyopathy. Diabetes Care. 2001;24:5–10.CrossRefPubMed Poirier P, Bogaty P, Garneau C, Marois L, Dumesnil JG. Diastolic dysfunction in normotensive men with well-controlled type 2 diabetes: importance of maneuvers in echocardiographic screening for preclinical diabetic cardiomyopathy. Diabetes Care. 2001;24:5–10.CrossRefPubMed
5.
Zurück zum Zitat Joffe II, Travers KE, Perreault-Micale CL, et al. Abnormal cardiac function in the streptozotocin-induced non-insulin-dependent diabetic rat: noninvasive assessment with doppler echocardiography and contribution of the nitric oxide pathway. J Am Coll Cardiol. 1999;34:2111–9.CrossRefPubMed Joffe II, Travers KE, Perreault-Micale CL, et al. Abnormal cardiac function in the streptozotocin-induced non-insulin-dependent diabetic rat: noninvasive assessment with doppler echocardiography and contribution of the nitric oxide pathway. J Am Coll Cardiol. 1999;34:2111–9.CrossRefPubMed
7.
Zurück zum Zitat Mourmoura E, Vial G, Laillet B, et al. Preserved endothelium-dependent dilatation of the coronary microvasculature at the early phase of diabetes mellitus despite the increased oxidative stress and depressed cardiac mechanical function ex vivo. Cardiovasc Diabetol. 2013;12:49.PubMedCentralCrossRefPubMed Mourmoura E, Vial G, Laillet B, et al. Preserved endothelium-dependent dilatation of the coronary microvasculature at the early phase of diabetes mellitus despite the increased oxidative stress and depressed cardiac mechanical function ex vivo. Cardiovasc Diabetol. 2013;12:49.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Pearson JT, Jenkins MJ, Edgley AJ, et al. Acute Rho-kinase inhibition improves coronary dysfunction in vivo, in the early diabetic microcirculation. Cardiovasc Diabetol. 2013;12:111.PubMedCentralCrossRefPubMed Pearson JT, Jenkins MJ, Edgley AJ, et al. Acute Rho-kinase inhibition improves coronary dysfunction in vivo, in the early diabetic microcirculation. Cardiovasc Diabetol. 2013;12:111.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Cai L. Suppression of nitrative damage by metallothionein in diabetic heart contributes to the prevention of cardiomyopathy. Free Radic Biol Med. 2006;41:851–61.CrossRefPubMed Cai L. Suppression of nitrative damage by metallothionein in diabetic heart contributes to the prevention of cardiomyopathy. Free Radic Biol Med. 2006;41:851–61.CrossRefPubMed
10.
Zurück zum Zitat Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991;43:109–42.PubMed Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991;43:109–42.PubMed
11.
Zurück zum Zitat Radovits T, Arif R, Bomicke T, et al. Vascular dysfunction induced by hypochlorite is improved by the selective phosphodiesterase-5-inhibitor vardenafil. Eur J Pharmacol. 2013;710:110–9.CrossRefPubMed Radovits T, Arif R, Bomicke T, et al. Vascular dysfunction induced by hypochlorite is improved by the selective phosphodiesterase-5-inhibitor vardenafil. Eur J Pharmacol. 2013;710:110–9.CrossRefPubMed
12.
Zurück zum Zitat Szabo G, Radovits T, Veres G, et al. Vardenafil protects against myocardial and endothelial injuries after cardiopulmonary bypass. Eur J Cardiothorac Surg. 2009;36:657–64.CrossRefPubMed Szabo G, Radovits T, Veres G, et al. Vardenafil protects against myocardial and endothelial injuries after cardiopulmonary bypass. Eur J Cardiothorac Surg. 2009;36:657–64.CrossRefPubMed
13.
Zurück zum Zitat Hirschberg K, Radovits T, Loganathan S, et al. Selective phosphodiesterase-5 inhibition reduces neointimal hyperplasia in rat carotid arteries after surgical endarterectomy. J Thorac Cardiovasc Surg. 2009;137:1508–14.CrossRefPubMed Hirschberg K, Radovits T, Loganathan S, et al. Selective phosphodiesterase-5 inhibition reduces neointimal hyperplasia in rat carotid arteries after surgical endarterectomy. J Thorac Cardiovasc Surg. 2009;137:1508–14.CrossRefPubMed
14.
Zurück zum Zitat Loganathan S, Radovits T, Hirschberg K, et al. Effects of selective phosphodiesterase-5-inhibition on myocardial contractility and reperfusion injury after heart transplantation. Transplantation. 2008;86:1414–8.CrossRefPubMed Loganathan S, Radovits T, Hirschberg K, et al. Effects of selective phosphodiesterase-5-inhibition on myocardial contractility and reperfusion injury after heart transplantation. Transplantation. 2008;86:1414–8.CrossRefPubMed
15.
Zurück zum Zitat Kukreja RC, Salloum FN, Das A. Cyclic guanosine monophosphate signaling and phosphodiesterase-5 inhibitors in cardioprotection. J Am Coll Cardiol. 2012;59:1921–7.PubMedCentralCrossRefPubMed Kukreja RC, Salloum FN, Das A. Cyclic guanosine monophosphate signaling and phosphodiesterase-5 inhibitors in cardioprotection. J Am Coll Cardiol. 2012;59:1921–7.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Schafer A, Fraccarollo D, Pfortsch S, et al. Improvement of vascular function by acute and chronic treatment with the PDE-5 inhibitor sildenafil in experimental diabetes mellitus. Br J Pharmacol. 2008;153:886–93.PubMedCentralCrossRefPubMed Schafer A, Fraccarollo D, Pfortsch S, et al. Improvement of vascular function by acute and chronic treatment with the PDE-5 inhibitor sildenafil in experimental diabetes mellitus. Br J Pharmacol. 2008;153:886–93.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Stasch JP, Schmidt PM, Nedvetsky PI, et al. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels. J Clin Invest. 2006;116:2552–61.PubMedCentralCrossRefPubMed Stasch JP, Schmidt PM, Nedvetsky PI, et al. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels. J Clin Invest. 2006;116:2552–61.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Korkmaz S, Radovits T, Barnucz E, et al. Pharmacological activation of soluble guanylate cyclase protects the heart against ischemic injury. Circulation. 2009;120:677–86.CrossRefPubMed Korkmaz S, Radovits T, Barnucz E, et al. Pharmacological activation of soluble guanylate cyclase protects the heart against ischemic injury. Circulation. 2009;120:677–86.CrossRefPubMed
19.
Zurück zum Zitat Radovits T, Korkmaz S, Miesel-Groschel C, et al. Pre-conditioning with the soluble guanylate cyclase activator Cinaciguat reduces ischaemia-reperfusion injury after cardiopulmonary bypass. Eur J Cardiothorac Surg. 2011;39:248–55.CrossRefPubMed Radovits T, Korkmaz S, Miesel-Groschel C, et al. Pre-conditioning with the soluble guanylate cyclase activator Cinaciguat reduces ischaemia-reperfusion injury after cardiopulmonary bypass. Eur J Cardiothorac Surg. 2011;39:248–55.CrossRefPubMed
20.
Zurück zum Zitat Salloum FN, Das A, Samidurai A, et al. Cinaciguat, a novel activator of soluble guanylate cyclase, protects against ischemia/reperfusion injury: role of hydrogen sulfide. Am J Physiol Heart Circ Physiol. 2012;302:H1347–54.PubMedCentralCrossRefPubMed Salloum FN, Das A, Samidurai A, et al. Cinaciguat, a novel activator of soluble guanylate cyclase, protects against ischemia/reperfusion injury: role of hydrogen sulfide. Am J Physiol Heart Circ Physiol. 2012;302:H1347–54.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Korkmaz S, Loganathan S, Mikles B, et al. Nitric oxide- and heme-independent activation of soluble guanylate cyclase attenuates peroxynitrite-induced endothelial dysfunction in rat aorta. J Cardiovasc Pharmacol Ther. 2013;18:70–7.CrossRefPubMed Korkmaz S, Loganathan S, Mikles B, et al. Nitric oxide- and heme-independent activation of soluble guanylate cyclase attenuates peroxynitrite-induced endothelial dysfunction in rat aorta. J Cardiovasc Pharmacol Ther. 2013;18:70–7.CrossRefPubMed
22.
Zurück zum Zitat Hirschberg K, Tarcea V, Pali S, et al. Cinaciguat prevents neointima formation after arterial injury by decreasing vascular smooth muscle cell migration and proliferation. Int J Cardiol. 2013;167:470–7.CrossRefPubMed Hirschberg K, Tarcea V, Pali S, et al. Cinaciguat prevents neointima formation after arterial injury by decreasing vascular smooth muscle cell migration and proliferation. Int J Cardiol. 2013;167:470–7.CrossRefPubMed
23.
Zurück zum Zitat Irvine JC, Ganthavee V, Love JE, et al. The soluble guanylyl cyclase activator bay 58-2667 selectively limits cardiomyocyte hypertrophy. PLoS One. 2012;7:e44481.PubMedCentralCrossRefPubMed Irvine JC, Ganthavee V, Love JE, et al. The soluble guanylyl cyclase activator bay 58-2667 selectively limits cardiomyocyte hypertrophy. PLoS One. 2012;7:e44481.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Frey R, Muck W, Unger S, Artmeier-Brandt U, Weimann G, Wensing G. Pharmacokinetics, pharmacodynamics, tolerability, and safety of the soluble guanylate cyclase activator cinaciguat (BAY 58-2667) in healthy male volunteers. J Clin Pharmacol. 2008;48:1400–10.CrossRefPubMed Frey R, Muck W, Unger S, Artmeier-Brandt U, Weimann G, Wensing G. Pharmacokinetics, pharmacodynamics, tolerability, and safety of the soluble guanylate cyclase activator cinaciguat (BAY 58-2667) in healthy male volunteers. J Clin Pharmacol. 2008;48:1400–10.CrossRefPubMed
25.
Zurück zum Zitat Lapp H, Mitrovic V, Franz N, et al. Cinaciguat (BAY 58-2667) improves cardiopulmonary hemodynamics in patients with acute decompensated heart failure. Circulation. 2009;119:2781–8.CrossRefPubMed Lapp H, Mitrovic V, Franz N, et al. Cinaciguat (BAY 58-2667) improves cardiopulmonary hemodynamics in patients with acute decompensated heart failure. Circulation. 2009;119:2781–8.CrossRefPubMed
26.
Zurück zum Zitat Erdmann E, Semigran MJ, Nieminen MS, et al. Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but also causes hypotension in acute decompensated heart failure. Eur Heart J. 2013;34:57–67.CrossRefPubMed Erdmann E, Semigran MJ, Nieminen MS, et al. Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but also causes hypotension in acute decompensated heart failure. Eur Heart J. 2013;34:57–67.CrossRefPubMed
27.
28.
Zurück zum Zitat Gheorghiade M, Greene SJ, Filippatos G, et al. Cinaciguat, a soluble guanylate cyclase activator: results from the randomized, controlled, phase IIb COMPOSE programme in acute heart failure syndromes. Eur J Heart Fail. 2012;14:1056–66.CrossRefPubMed Gheorghiade M, Greene SJ, Filippatos G, et al. Cinaciguat, a soluble guanylate cyclase activator: results from the randomized, controlled, phase IIb COMPOSE programme in acute heart failure syndromes. Eur J Heart Fail. 2012;14:1056–66.CrossRefPubMed
29.
Zurück zum Zitat Pacher P, Nagayama T, Mukhopadhyay P, Batkai S, Kass DA. Measurement of cardiac function using pressure-volume conductance catheter technique in mice and rats. Nat Protoc. 2008;3:1422–34.PubMedCentralCrossRefPubMed Pacher P, Nagayama T, Mukhopadhyay P, Batkai S, Kass DA. Measurement of cardiac function using pressure-volume conductance catheter technique in mice and rats. Nat Protoc. 2008;3:1422–34.PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Radovits T, Olah A, Lux A, et al. Rat model of exercise-induced cardiac hypertrophy: hemodynamic characterization using left ventricular pressure-volume analysis. Am J Physiol Heart Circ Physiol. 2013;305:H124–34.CrossRefPubMed Radovits T, Olah A, Lux A, et al. Rat model of exercise-induced cardiac hypertrophy: hemodynamic characterization using left ventricular pressure-volume analysis. Am J Physiol Heart Circ Physiol. 2013;305:H124–34.CrossRefPubMed
31.
Zurück zum Zitat Litwin SE, Raya TE, Anderson PG, Daugherty S, Goldman S. Abnormal cardiac function in the streptozotocin-diabetic rat. Changes in active and passive properties of the left ventricle. J Clin Invest. 1990;86:481–8.PubMedCentralCrossRefPubMed Litwin SE, Raya TE, Anderson PG, Daugherty S, Goldman S. Abnormal cardiac function in the streptozotocin-diabetic rat. Changes in active and passive properties of the left ventricle. J Clin Invest. 1990;86:481–8.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Snoeckx LH, Cornelussen RN, Van Nieuwenhoven FA, Reneman RS, Van Der Vusse GJ. Heat shock proteins and cardiovascular pathophysiology. Physiol Rev. 2001;81:1461–97.PubMed Snoeckx LH, Cornelussen RN, Van Nieuwenhoven FA, Reneman RS, Van Der Vusse GJ. Heat shock proteins and cardiovascular pathophysiology. Physiol Rev. 2001;81:1461–97.PubMed
34.
Zurück zum Zitat Lukowski R, Krieg T, Rybalkin SD, Beavo J, Hofmann F. Turning on cGMP-dependent pathways to treat cardiac dysfunctions: boom, bust, and beyond. Trends Pharmacol Sci. 2014;35:404–13.CrossRefPubMed Lukowski R, Krieg T, Rybalkin SD, Beavo J, Hofmann F. Turning on cGMP-dependent pathways to treat cardiac dysfunctions: boom, bust, and beyond. Trends Pharmacol Sci. 2014;35:404–13.CrossRefPubMed
35.
Zurück zum Zitat Fang L, Radovits T, Szabo G, Mozes MM, Rosivall L, Kokeny G. Selective phosphodiesterase-5 (PDE-5) inhibitor vardenafil ameliorates renal damage in type 1 diabetic rats by restoring cyclic 3′,5′ guanosine monophosphate (cGMP) level in podocytes. Nephrol Dial Transplant. 2013;28:1751–61.CrossRefPubMed Fang L, Radovits T, Szabo G, Mozes MM, Rosivall L, Kokeny G. Selective phosphodiesterase-5 (PDE-5) inhibitor vardenafil ameliorates renal damage in type 1 diabetic rats by restoring cyclic 3′,5′ guanosine monophosphate (cGMP) level in podocytes. Nephrol Dial Transplant. 2013;28:1751–61.CrossRefPubMed
36.
Zurück zum Zitat Hamet P, Pang SC, Tremblay J. Atrial natriuretic factor-induced egression of cyclic guanosine 3′:5′-monophosphate in cultured vascular smooth muscle and endothelial cells. J Biol Chem. 1989;264:12364–9.PubMed Hamet P, Pang SC, Tremblay J. Atrial natriuretic factor-induced egression of cyclic guanosine 3′:5′-monophosphate in cultured vascular smooth muscle and endothelial cells. J Biol Chem. 1989;264:12364–9.PubMed
37.
Zurück zum Zitat Koka S, Das A, Salloum FN, Kukreja RC. Phosphodiesterase-5 inhibitor tadalafil attenuates oxidative stress and protects against myocardial ischemia/reperfusion injury in type 2 diabetic mice. Free Radic Biol Med. 2013;60:80–8.CrossRefPubMed Koka S, Das A, Salloum FN, Kukreja RC. Phosphodiesterase-5 inhibitor tadalafil attenuates oxidative stress and protects against myocardial ischemia/reperfusion injury in type 2 diabetic mice. Free Radic Biol Med. 2013;60:80–8.CrossRefPubMed
39.
Zurück zum Zitat Van Linthout S, Seeland U, Riad A, et al. Reduced MMP-2 activity contributes to cardiac fibrosis in experimental diabetic cardiomyopathy. Basic Res Cardiol. 2008;103:319–27.CrossRefPubMed Van Linthout S, Seeland U, Riad A, et al. Reduced MMP-2 activity contributes to cardiac fibrosis in experimental diabetic cardiomyopathy. Basic Res Cardiol. 2008;103:319–27.CrossRefPubMed
40.
Zurück zum Zitat Cai L, Li W, Wang G, Guo L, Jiang Y, Kang YJ. Hyperglycemia-induced apoptosis in mouse myocardium: mitochondrial cytochrome C-mediated caspase-3 activation pathway. Diabetes. 2002;51:1938–48.CrossRefPubMed Cai L, Li W, Wang G, Guo L, Jiang Y, Kang YJ. Hyperglycemia-induced apoptosis in mouse myocardium: mitochondrial cytochrome C-mediated caspase-3 activation pathway. Diabetes. 2002;51:1938–48.CrossRefPubMed
41.
Zurück zum Zitat Beyer C, Zenzmaier C, Palumbo-Zerr K, et al. Stimulation of the soluble guanylate cyclase (sGC) inhibits fibrosis by blocking non-canonical TGFbeta signalling. Ann Rheum Dis. 2014. doi: 10.1136/annrheumdis-2013-204508 (Published ahead of print 23 February 2014). Beyer C, Zenzmaier C, Palumbo-Zerr K, et al. Stimulation of the soluble guanylate cyclase (sGC) inhibits fibrosis by blocking non-canonical TGFbeta signalling. Ann Rheum Dis. 2014. doi: 10.​1136/​annrheumdis-2013-204508 (Published ahead of print 23 February 2014).
42.
Zurück zum Zitat Wang S, Mitu GM, Hirschberg R. Osmotic polyuria: an overlooked mechanism in diabetic nephropathy. Nephrol Dial Transplant. 2008;23:2167–72.CrossRefPubMed Wang S, Mitu GM, Hirschberg R. Osmotic polyuria: an overlooked mechanism in diabetic nephropathy. Nephrol Dial Transplant. 2008;23:2167–72.CrossRefPubMed
Metadaten
Titel
The soluble guanylate cyclase activator cinaciguat prevents cardiac dysfunction in a rat model of type-1 diabetes mellitus
verfasst von
Csaba Mátyás
Balázs Tamás Németh
Attila Oláh
László Hidi
Ede Birtalan
Dalma Kellermayer
Mihály Ruppert
Sevil Korkmaz-Icöz
Gábor Kökény
Eszter Mária Horváth
Gábor Szabó
Béla Merkely
Tamás Radovits
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2015
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-015-0309-x

Weitere Artikel der Ausgabe 1/2015

Cardiovascular Diabetology 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.