Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 849
1.
Cardiovasc Drugs Ther ; 36(1): 15-29, 2022 02.
Article En | MEDLINE | ID: mdl-33064235

PURPOSE: Cardiac dysfunction can occur as a sequela of a state of prolonged pressure overload and postischemic injury. Flavonoids such as quercetin may be protective against cardiovascular disease. This study aimed to systematically assess the effects of quercetin on cardiac function in pressure overload and postischemia-reperfusion injury in rodents. METHODS: A systematic search of the literature up to May 2020 was conducted in PubMed, Ovid Medline, EBSCOhost, Scopus, and the Cochrane Library to identify relevant published studies on quercetin and cardiac function using standardized criteria. Meta-analyses were performed on animal studies of pressure overload and ischemia-reperfusion (I/R) injury. RESULTS: The effects of quercetin on cardiac function in both models were qualitatively reported in 14 studies. The effects of quercetin in four pressure-overload model studies involving 73 rodents and eight I/R-injury model studies involving 120 rodents were quantitatively assessed by meta-analysis. Quercetin improved the overall cardiac function in both pressure overload (n = 4 studies, n = 73 rodents; SMD = - 1.50; 95% CI: - 2.66 to - 0.33; P < 0.05; I2 = 74.05%) and I/R injury (n = 8 studies, n = 120 rodents; SMD = - 1.81; 95% CI: - 3.05 to - 0.56; P < 0.01; I2 = 84.93%) models. The improvement was associated with amelioration in cardiac structure in the pressure-overload model and both systolic and diastolic functioning in the I/R-injury model. CONCLUSION: The present meta-analysis suggested that quercetin has beneficial effects for improving cardiac left ventricular dysfunction in both pressure-overload and I/R-injury models.


Myocardial Reperfusion Injury/drug therapy , Quercetin/pharmacology , Ventricular Dysfunction, Left/drug therapy , Animals , Cardiotonic Agents/pharmacology , Disease Models, Animal , Mice , Myocardial Reperfusion Injury/physiopathology , Rats , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure/drug effects
2.
J Cardiovasc Pharmacol ; 79(1): e41-e49, 2022 01 01.
Article En | MEDLINE | ID: mdl-34654786

ABSTRACT: Treatment with trastuzumab, an antihuman epidermal growth factor receptor type 2 humanized monoclonal antibody, has been associated with heart failure in certain patients with cancer; however, the mechanism underlying trastuzumab-induced cardiac dysfunction remains unclear. This study was conducted to clarify the cardiac effects of trastuzumab in cynomolgus monkeys, which are commonly used as cross-reactive species in preclinical safety evaluation. Monkeys were treated with trastuzumab weekly for 1 month (5 doses in total). At first and fifth doses for pressure-volume loop analysis, trastuzumab at 20 mg·kg-1·10 min-1, equivalent to the human therapeutic dose, was administered intravenously to isoflurane-anesthetized animals, followed by 60 mg·kg-1·10 min-1 at a 30-minute interval. The other doses were fixed at 80 mg·kg-1·10 min-1 under unanesthetized conditions. After the first dose, reduced heart rate, decreases in maximal rate of fall of left ventricular pressure, and prolonged time constant for isovolumic relaxation, which are predictors of drug-induced changes in lusitropy, were observed at 20 and 60 mg·kg-1. The changes after the fifth dose were comparable with those after the first dose, indicating trastuzumab did not show exacerbation of cardiac function during the 1-month trial. No significant changes in slope of preload recruitable stroke work, which is a load-independent inotropic parameter, were observed at either dose. In conclusion, trastuzumab-induced little inotropic effect but induced negative chronotropic or lusitropic effects in monkeys, which might be associated with impaired left ventricular diastolic function.


Antineoplastic Agents, Immunological/toxicity , Heart Rate/drug effects , Trastuzumab/toxicity , Ventricular Dysfunction, Left/chemically induced , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects , Animals , Antineoplastic Agents, Immunological/administration & dosage , Cardiotoxicity , Dose-Response Relationship, Drug , Drug Administration Schedule , Macaca fascicularis , Male , Risk Assessment , Risk Factors , Time Factors , Trastuzumab/administration & dosage , Ventricular Dysfunction, Left/physiopathology
3.
Pak J Pharm Sci ; 34(5(Special)): 2059-2064, 2021 Sep.
Article En | MEDLINE | ID: mdl-34862874

To investigate the impacts of Yangxin decoction on the expressions of matrix metalloproteinase 9 (MMP-9), calcineurin (CaN), T cell activated nuclear factor 3 (NFAT3) and zinc finger transcription factor 4 (GATA4) in myocardial tissue of rats with chronic heart failure (CHF). 50 healthy SD rats were randomly divided into the normal control group (n = 10) and the operation group (n = 40). After successful modeling, the rats were randomly divided into 4 groups. And they were treated with Yangxin decoctions of low concentration (1.5 g/kg), medium concentration (2.5 g/kg), high concentration (3.5 g/kg) and distilled water (for 4 weeks). The LVSP, SAP, DAP and LVEDP in Yangxin decoction treatment groups were significantly superior to the model group. The LVEF, LVIDd and LVIDs in Yangxin decoction treatment groups were significantly superior to the model group. The activity of CaN in each group treated with Yangxin decoction was significantly lower than that in the model group. The expression levels of MMP-9, NFAT3, GATA4 protein in each group treated with Yangxin decoction were significantly lower than that in the model group.. Yangxin decoction can significantly improve the cardiac function, reduce CaN activity, decrease the expression levels of MMP-9, NFAT3 and GATA4, inhibit CaN/NFAT3 signaling pathway, increase myocardial remodeling and protect myocardial tissue in rats with CHF.


Calcineurin/metabolism , Drugs, Chinese Herbal/pharmacology , GATA4 Transcription Factor/metabolism , Heart Failure/drug therapy , Matrix Metalloproteinase 9/metabolism , Myocardium/enzymology , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Ventricular Function, Left/drug effects , Animals , Chronic Disease , Disease Models, Animal , Female , Heart Failure/enzymology , Heart Failure/physiopathology , Male , Rats, Sprague-Dawley , Recovery of Function , Stroke Volume/drug effects , Ventricular Pressure/drug effects , Ventricular Remodeling/drug effects
4.
J Cardiovasc Pharmacol ; 78(1): e65-e76, 2021 07 01.
Article En | MEDLINE | ID: mdl-33929390

ABSTRACT: There is increasing evidence that angiotensin (1-7) [Ang (1-7)] is an endogenous biologically active component of the renin-angiotensin system. However, the role of the Ang (1-7)-MasR axis in postresuscitation myocardial dysfunction (PRMD) and its associated mechanism are still unclear. In this study, we investigated the effect of the Ang (1-7)-MasR axis on myocardial injury after cardiac arrest-cardiopulmonary resuscitation-restoration of spontaneous circulation. We established a model of oxygen/glucose deprivation-reperfusion in myocardial cells in vitro and a rat model of cardiac arrest-cardiopulmonary resuscitation-restoration of spontaneous circulation in vivo. The cell apoptosis rate and the expression of the superoxide anion 3-nitrotyrosine were decreased in the Ang (1-7) group in vitro and in vivo. The mean arterial pressure was decreased, whereas +LVdp/dtmax and -LVdp/dtmax were increased in rats in the Ang (1-7) group. The mRNA and protein levels of Ang II type 1 receptor, MasR, phosphoinositide 3-kinase, protein kinase B, and endothelial nitric oxide synthase were increased in the Ang (1-7) group in vivo. These results indicate that the Ang (1-7)-MasR axis can alleviate PRMD by reducing myocardial tissue damage and oxidative stress through activation of the phosphoinositide 3-kinase-protein kinase B-endothelial nitric oxide synthase signaling pathway and provide a new direction for the clinical treatment of PRMD.


Angiotensin I/pharmacology , Cardiopulmonary Resuscitation/adverse effects , Heart Arrest/therapy , Heart Diseases/prevention & control , Myocytes, Cardiac/drug effects , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Heart Arrest/physiopathology , Heart Diseases/enzymology , Heart Diseases/etiology , Heart Diseases/physiopathology , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Proto-Oncogene Mas/agonists , Proto-Oncogene Mas/genetics , Proto-Oncogene Mas/metabolism , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Return of Spontaneous Circulation , Signal Transduction , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
5.
Eur J Pharmacol ; 899: 173978, 2021 May 15.
Article En | MEDLINE | ID: mdl-33691164

Metabolic syndrome is linked to an increased risk of cardiovascular complications by a mechanism involving mainly decreased nitric oxide (NO) bioavailability and impaired NO-soluble guanylate cyclase (sGC)- cyclic guanosine monophosphate (cGMP) signalling (NO-sGC-cGMP). To further develop this scientific point, this study aimed to investigate the effects of long-term treatment with BAY 41-2272 (a sGC stimulator) on cardiovascular reactivity of spontaneously hypertensive rats (SHR) as a model of metabolic syndrome. SHR were randomly divided into 3 groups: control group, cafeteria diet (CD)-fed group and CD-fed group treated daily with BAY 41-2272 (5 mg/kg) by gastric gavage for 12 weeks. In vivo measurements of body weight, abdominal circumference, blood pressure and glucose tolerance test were performed. At the end of the feeding period, ex vivo cumulative concentration-response curves were performed on isolated perfused heart (isoproterenol (0.1 nM - 1 µM)) and thoracic aorta (phenylephrine (1 nM-10 µM), acetylcholine (1 nM-10 µM), and sodium nitroprusside (SNP) (0.1 nM-0.1 µM)). We showed that chronic CD feeding induced abdominal obesity, hypertriglyceridemia, glucose intolerance and exacerbated arterial hypertension in SHR. Compared to control group, CD-fed group showed a decrease in ß-adrenoceptor-induced cardiac inotropy, in coronary perfusion pressure and in aortic contraction to phenylephrine. While relaxing effects of acetylcholine and SNP were unchanged. BAY 41-2272 long-term treatment markedly prevented arterial hypertension development and glucose intolerance, enhanced the α1-adrenoceptor-induced vasoconstriction, and restored cardiac inotropy and coronary vasodilation. These findings suggest that BAY 41-2272 may be a potential novel drug for preventing metabolic and cardiovascular complications of metabolic syndrome.


Cardiovascular Diseases/prevention & control , Enzyme Activators/pharmacology , Metabolic Syndrome/prevention & control , Pyrazoles/pharmacology , Pyridines/pharmacology , Soluble Guanylyl Cyclase/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Cardiovascular Diseases/enzymology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Coronary Circulation/drug effects , Cyclic GMP/metabolism , Disease Models, Animal , Enzyme Activation , Glucose Intolerance/enzymology , Glucose Intolerance/etiology , Glucose Intolerance/physiopathology , Glucose Intolerance/prevention & control , Hypertension/enzymology , Hypertension/etiology , Hypertension/physiopathology , Hypertension/prevention & control , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/etiology , Hypertriglyceridemia/physiopathology , Hypertriglyceridemia/prevention & control , Isolated Heart Preparation , Male , Metabolic Syndrome/enzymology , Metabolic Syndrome/etiology , Metabolic Syndrome/physiopathology , Nitric Oxide Synthase Type II/metabolism , Obesity, Abdominal/enzymology , Obesity, Abdominal/etiology , Obesity, Abdominal/physiopathology , Obesity, Abdominal/prevention & control , Rats, Inbred SHR , Vasoconstriction/drug effects , Vasodilation/drug effects , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
6.
Cardiovasc Toxicol ; 21(7): 517-532, 2021 07.
Article En | MEDLINE | ID: mdl-33723718

Calcium dysregulation and mitochondrial dysfunction are key elements in the development of sepsis-induced cardiac dysfunction. Evidences have suggested that inhibition of Wnt/ß-Catenin signalling prevents cardiac dysfunction and remodelling in surgical, hypertension and pressure overload models. The present study investigated the effects of Wnt/ß-Catenin inhibitor on calcium overload and mitochondrial dysfunction in rat sepsis model of cardiomyopathy. Induction of sepsis by cecal ligation puncture (CLP) resulted in the up-regulation of cardiac ß-catenin transcriptional levels and cardiac dysfunction depicted by increased serum lactate dehydrogenase, CK-MB levels reduced maximum (dp/dt max.) and minimum developed pressure (dp/dt min.), increased LVEsDP and relaxation constant tau values. Moreover, oxidative and inflammatory stress, immune cell infiltration, increased myeloperoxidase activity, enhanced caspase-3 activity and fibronectin protein levels were observed in septic rat's heart. Also, septic rat's heart displayed mitochondrial dysfunction due to mPTP opening, increased calcium up-regulation in left ventricular apex tissues and whole heart, increased collagen staining, necrosis and structural damage. Pre-treatment with Wnt/ß-Catenin antagonist attenuated sepsis-induced serum and tissue biochemical changes, cardiac dysfunction and structural alterations by inhibiting mitochondrial mPTP opening and restricting calcium overloading in cardiac tissue.


Calcium/metabolism , Cardiomyopathies/prevention & control , Coinfection/drug therapy , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Pyrvinium Compounds/pharmacology , Sepsis/drug therapy , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Coinfection/metabolism , Coinfection/microbiology , Cytokines/metabolism , Disease Models, Animal , Fibrosis , Inflammation Mediators/metabolism , Male , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Permeability Transition Pore/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Oxidative Stress/drug effects , Rats, Wistar , Sepsis/metabolism , Sepsis/microbiology , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects , beta Catenin/genetics
7.
Circ Heart Fail ; 14(3): e007351, 2021 03.
Article En | MEDLINE | ID: mdl-33663236

BACKGROUND: New heart failure therapies that safely augment cardiac contractility and output are needed. Previous apelin peptide studies have highlighted the potential for APJ (apelin receptor) agonism to enhance cardiac function in heart failure. However, apelin's short half-life limits its therapeutic utility. Here, we describe the preclinical characterization of a novel, orally bioavailable APJ agonist, BMS-986224. METHODS: BMS-986224 pharmacology was compared with (Pyr1) apelin-13 using radio ligand binding and signaling pathway assays downstream of APJ (cAMP, phosphorylated ERK [extracellular signal-regulated kinase], bioluminescence resonance energy transfer-based G-protein assays, ß-arrestin recruitment, and receptor internalization). Acute effects on cardiac function were studied in anesthetized instrumented rats. Chronic effects of BMS-986224 were assessed echocardiographically in the RHR (renal hypertensive rat) model of cardiac hypertrophy and decreased cardiac output. RESULTS: BMS-986224 was a potent (Kd=0.3 nmol/L) and selective APJ agonist, exhibiting similar receptor binding and signaling profile to (Pyr1) apelin-13. G-protein signaling assays in human embryonic kidney 293 cells and human cardiomyocytes confirmed this and demonstrated a lack of signaling bias relative to (Pyr1) apelin-13. In anesthetized instrumented rats, short-term BMS-986224 infusion increased cardiac output (10%-15%) without affecting heart rate, which was similar to (Pyr1) apelin-13 but differentiated from dobutamine. Subcutaneous and oral BMS-986224 administration in the RHR model increased stroke volume and cardiac output to levels seen in healthy animals but without preventing cardiac hypertrophy and fibrosis, effects differentiated from enalapril. CONCLUSIONS: We identify a novel, potent, and orally bioavailable nonpeptidic APJ agonist that closely recapitulates the signaling properties of (Pyr1) apelin-13. We show that oral APJ agonist administration induces a sustained increase in cardiac output in the cardiac disease setting and exhibits a differentiated profile from the renin-angiotensin system inhibitor enalapril, supporting further clinical evaluation of BMS-986224 in heart failure.


Apelin Receptors/agonists , Cardiac Output/drug effects , Heart Failure/physiopathology , Intercellular Signaling Peptides and Proteins/pharmacology , Stroke Volume/drug effects , Animals , Bioluminescence Resonance Energy Transfer Techniques , CHO Cells , Cricetulus , Dogs , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Haplorhini , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Phosphorylation , Radioligand Assay , Rats , Tritium , Ventricular Pressure/drug effects , beta-Arrestins/drug effects , beta-Arrestins/metabolism
8.
Pharmacol Res Perspect ; 9(1): e00703, 2021 02.
Article En | MEDLINE | ID: mdl-33421306

Pulmonary arterial hypertension (PAH) is a progressive cardiovascular disease with high mortality. However, there were no efficient medical drugs for PAH to enormously improve the survival and quality of life measures. The present study aimed to explore the protective effect of baicalin against experimental PAH in vivo and vitro. All the experimental rats received intraperitoneal injection of monocrotaline (MCT) to induce PAH model. Baicalin was given by intragastric administration from 2 days after MCT injection. Forty animals were randomly divided into four groups: Control, MCT, saline-, and baicalin-treated groups (n = 10 in each). Post-operation, hemodynamic data, and index of right ventricular hypertrophy (RVHI) were recorded to evaluate the inhibition of baicalin on MCT-induced PAH. Furthermore, pulmonary artery smooth muscle cells (PASMCs) model induced by tumor necrosis factor-α (TNF-α) was used to observe the inhibition of vascular cells proliferation in vitro. The results demonstrated that baicalin significantly attenuated MCT-induced right ventricular systolic pressure (RVSP), the index of right ventricular hypertrophy, and vessel wall thickness; inhibit inflammatory and cell proliferation induced by MCT or TNF-α, respectively. In addition, we found that baicalin might protect against experimental PAH via regulating the TNF-α/BMPR2 signaling pathway.


Antihypertensive Agents/therapeutic use , Bone Morphogenetic Protein Receptors, Type II/metabolism , Flavonoids/therapeutic use , Pulmonary Arterial Hypertension/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Vascular Remodeling/drug effects , Actins/metabolism , Animals , Antihypertensive Agents/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Flavonoids/pharmacology , Interleukin-1beta/genetics , Interleukin-6/genetics , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/physiopathology , Rats, Wistar , Signal Transduction/drug effects , Smad Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects
9.
Cardiovasc Diabetol ; 20(1): 6, 2021 01 07.
Article En | MEDLINE | ID: mdl-33413355

BACKGROUND: In the EMPA-REG OUTCOME trial (Empagliflozin Cardiovascular Outcome Event Trial) treatment with the sodium-glucose cotransporter-2 (SGLT2) inhibitor empagliflozin significantly reduced heart failure hospitalization (HHF) in patients with type 2 diabetes mellitus (T2D) and established cardiovascular disease. The early separation of the HHF event curves within the first 3 months of the trial suggest that immediate hemodynamic effects may play a role. However, hitherto no data exist on early effects of SGLT2 inhibitors on hemodynamic parameters and cardiac function. Thus, this study examined early and delayed effects of empagliflozin treatment on hemodynamic parameters including systemic vascular resistance index, cardiac index, and stroke volume index, as well as echocardiographic measures of cardiac function. METHODS: In this placebo-controlled, randomized, double blind, exploratory study patients with T2D were randomized to empagliflozin 10 mg or placebo for a period of 3 months. Hemodynamic and echocardiographic parameters were assessed after 1 day, 3 days and 3 months of treatment. RESULTS: Baseline characteristics were not different in the empagliflozin (n = 22) and placebo (n = 20) group. Empagliflozin led to a significant increase in urinary glucose excretion (baseline: 7.3 ± 22.7 g/24 h; day 1: 48.4 ± 34.7 g/24 h; p < 0.001) as well as urinary volume (1740 ± 601 mL/24 h to 2112 ± 837 mL/24 h; p = 0.011) already after one day compared to placebo. Treatment with empagliflozin had no effect on the primary endpoint of systemic vascular resistance index, nor on cardiac index, stroke volume index or pulse rate at any time point. In addition, echocardiography showed no difference in left ventricular systolic function as assessed by left ventricular ejections fraction and strain analysis. However, empagliflozin significantly improved left ventricular filling pressure as assessed by a reduction of early mitral inflow velocity relative to early diastolic left ventricular relaxation (E/e') which became significant at day 1 of treatment (baseline: 9.2 ± 2.6; day 1: 8.5 ± 2.2; p = 0.005) and remained apparent throughout the study. This was primarily attributable to reduced early mitral inflow velocity E (baseline: 0.8 ± 0.2 m/s; day 1: 0.73 ± 0.2 m/sec; p = 0.003). CONCLUSIONS: Empagliflozin treatment of patients with T2D has no significant effect on hemodynamic parameters after 1 or 3 days, nor after 3 months, but leads to rapid and sustained significant improvement of diastolic function. Trial registration EudraCT Number: 2016-000172-19; date of registration: 2017-02-20 (clinicaltrialregister.eu).


Benzhydryl Compounds/therapeutic use , Cardiovascular Diseases/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects , Aged , Benzhydryl Compounds/adverse effects , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/physiopathology , Diabetes Mellitus, Type 2/diagnosis , Double-Blind Method , Female , Germany , Glucosides/adverse effects , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Male , Middle Aged , Patient Admission , Pilot Projects , Prospective Studies , Recovery of Function , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Stroke Volume/drug effects , Time Factors , Treatment Outcome , Vascular Resistance/drug effects
10.
Cardiovasc Res ; 117(14): 2794-2806, 2021 12 17.
Article En | MEDLINE | ID: mdl-33135077

AIMS: Emipagliflozin (EMPA) is a potent inhibitor of the renal sodium-glucose co-transporter 2 (SGLT2) and an effective treatment for type-2 diabetes. In patients with diabetes and heart failure, EMPA has cardioprotective effects independent of improved glycaemic control, despite SGLT2 not being expressed in the heart. A number of non-canonical mechanisms have been proposed to explain these cardiac effects, most notably an inhibitory action on cardiac Na+/H+ exchanger 1 (NHE1), causing a reduction in intracellular [Na+] ([Na+]i). However, at resting intracellular pH (pHi), NHE1 activity is very low and its pharmacological inhibition is not expected to meaningfully alter steady-state [Na+]i. We re-evaluate this putative EMPA target by measuring cardiac NHE1 activity. METHODS AND RESULTS: The effect of EMPA on NHE1 activity was tested in isolated rat ventricular cardiomyocytes from measurements of pHi recovery following an ammonium pre-pulse manoeuvre, using cSNARF1 fluorescence imaging. Whereas 10 µM cariporide produced near-complete inhibition, there was no evidence for NHE1 inhibition with EMPA treatment (1, 3, 10, or 30 µM). Intracellular acidification by acetate-superfusion evoked NHE1 activity and raised [Na+]i, reported by sodium binding benzofuran isophthalate (SBFI) fluorescence, but EMPA did not ablate this rise. EMPA (10 µM) also had no significant effect on the rate of cytoplasmic [Na+]i rise upon superfusion of Na+-depleted cells with Na+-containing buffers. In Langendorff-perfused mouse, rat and guinea pig hearts, EMPA did not affect [Na+]i at baseline nor pHi recovery following acute acidosis, as measured by 23Na triple quantum filtered NMR and 31P NMR, respectively. CONCLUSIONS: Our findings indicate that cardiac NHE1 activity is not inhibited by EMPA (or other SGLT2i's) and EMPA has no effect on [Na+]i over a wide range of concentrations, including the therapeutic dose. Thus, the beneficial effects of SGLT2i's in failing hearts should not be interpreted in terms of actions on myocardial NHE1 or intracellular [Na+].


Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Myocytes, Cardiac/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Sodium/metabolism , Animals , Guinea Pigs , HCT116 Cells , HEK293 Cells , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Isolated Heart Preparation , Male , Membrane Potentials , Mice , Myocytes, Cardiac/metabolism , Rats, Wistar , Sodium-Hydrogen Exchanger 1/metabolism , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
11.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 373-381, 2021 02.
Article En | MEDLINE | ID: mdl-33029649

Protamine causes cardiac depression, which may be mediated by tumor necrosis factor alpha (TNF-α). Ulinastatin, a human urinary protease inhibitor, inhibits TNF-α. Here, we aimed to investigate whether ulinastatin prevented protamine-induced myocardial depression by inhibiting TNF-α. Rat hearts were perfused using a Langendorff system, and three protocols were followed. Protocol 1: The hearts were divided into saline, ulinastatin-low, and ulinastatin-high groups. Protamine was administered to each group, and myocardial contractility was the primary outcome. Protocol 2: The hearts were allotted to saline or ulinastatin group. Protamine was administered to each group. TNF-α expression in the coronary effluent and myocardial tissue was measured. Protocol 3: The hearts were allotted to saline and ulinastatin groups. Recombinant rat-TNF-α was administered to each group. Protamine alone reduced the maximum left ventricular pressure derivative (LV dP/dt max) by 45 ± 4%. In contrast, the reduction in LV dP/dt max was 4 ± 3% in the ulinastatin-high group. Compared with that in the saline group, the increase in TNF-α in the coronary effluent was attenuated in the ulinastatin group. Recombinant TNF-α alone reduced LV dP/dt max (- 21 ± 14%). In contrast, when TNF-α was added in the presence of ulinastatin, the decrease in LV dP/dt max was prevented significantly (- 3 ± 8%). We showed, for the first time, that ulinastatin protected against protamine-induced myocardial damage, both by inhibiting TNF-α synthesis and by directly preventing the cardiodepressant action of TNF-α.


Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Glycoproteins/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Cardiotonic Agents/pharmacology , Cardiotoxicity/metabolism , Cardiotoxicity/physiopathology , Glycoproteins/pharmacology , Heart Rate/drug effects , Male , Protamines , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
12.
Am J Respir Cell Mol Biol ; 63(6): 843-855, 2020 12.
Article En | MEDLINE | ID: mdl-32915674

The potential benefit of heart rate reduction (HRR), independent of ß-blockade, on right ventricular (RV) function in pulmonary hypertension (PH) remains undecided. We studied HRR effects on RV fibrosis and function in PH and RV pressure-loading models. Adult rats were randomized to 1) sham controls, 2) monocrotaline (MCT)-induced PH, 3) SU5416 + hypoxia (SUHX)-induced PH, or 4) pulmonary artery banding (PAB). Ivabradine (IVA) (10 mg/kg/d) was administered from 2 weeks after PH induction or PAB. Exercise tolerance, echocardiography, and pressure-volume hemodynamics were obtained at a terminal experiment 3 weeks later. RV myocardial samples were analyzed for putative mechanisms of HRR effects through fibrosis, profibrotic molecular signaling, and Ca++ handling. The effects of IVA versus carvedilol on human induced pluripotent stem cell-derived cardiomyocytes beat rate and relaxation properties were evaluated in vitro. Despite unabated severely elevated RV systolic pressures, IVA improved RV systolic and diastolic function, profibrotic signaling, and RV fibrosis in PH/PAB rats. RV systolic-elastance (control, 121 ± 116; MCT, 49 ± 36 vs. MCT+IVA, 120 ± 54; PAB, 70 ± 20 vs. PAB+IVA, 168 ± 76; SUHX, 86 ± 56 vs. SUHX +IVA, 218 ± 111; all P < 0.05), the time constant of RV relaxation, echo indices of RV function, and fibrosis (fibrosis: control, 4.6 ± 1%; MCT, 13.4 ± 6.5 vs. MCT+IVA, 6.7 ± 2.6%; PAB, 11.4 ± 4.5 vs. PAB+IVA, 6.4 ± 5.1%; SUHX, 10 ± 4.6 vs. SUHX+IVA, 3.9 ± 2.2%; all P < 0.001) were improved by IVA versus controls. IVA had a dose-response effect on induced pluripotent stem cell-derived cardiomyocytes beat rate by delaying Ca++ loss from the cytoplasm. In experimental PH or RV pressure loading, HRR improves RV fibrosis, function, and exercise endurance independent of ß-blockade. The balance between adverse tachycardia and bradycardia requires further study, but judicious HRR may provide a promising strategy to improve RV function in clinical PH.


Heart Rate/drug effects , Hypertension, Pulmonary/chemically induced , Ivabradine/pharmacology , Ventricular Function, Right/drug effects , Animals , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics , Humans , Hypertension, Pulmonary/pathology , Induced Pluripotent Stem Cells/drug effects , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Ventricular Pressure/drug effects
13.
Can J Physiol Pharmacol ; 98(10): 700-707, 2020 Oct.
Article En | MEDLINE | ID: mdl-32516552

Doxorubicin (DOX) is a highly efficient chemotherapeutic drug limited by its cardiotoxicity. Galectin-3 (Gal-3) overexpression is associated with several cardiovascular diseases. In this study, the in vivo models of DOX-treated rats and the in vitro model of DOX-treated H9C2 cells were used. DOX induced cardiac injury and dysfunction accompanied with the upregulation of Gal-3 at the end of the experiment, while inhibition of Gal-3 with modified citrus pectin (MCP) exhibited a dramatic improvement in cardiac function of the DOX-treated rats, as manifested by increased left ventricular systolic pressure and ±dp/dtmax and decreased left ventricular end-diastolic pressure. The plasma levels of myocardial injury markers such as lactate dehydrogenase, creatine kinase, creatine kinase-MB, and cardiac troponin I were decreased after MCP treatment. In parallel, MCP attenuated myocardial tissue markers of oxidative stress such as hydrogen peroxide and malondialdehyde restored the activities of superoxide dismutase, catalase, and glutathione peroxidase and upregulated antioxidant peroxiredoxin-4 (Prx-4). To further verify the role of Prx-4, it was downregulated by siRNA-mediated knockdown in H9C2 cells. MCP could not reverse DOX-induced oxidative stress in Prx-4-knock-down cells. In conclusion, Gal-3 mediated DOX-induced cardiotoxicity and Gal-3 inhibition attenuated DOX-induced cardiac dysfunction by upregulating the expression of Prx-4 to reduce myocardial oxidative stress.


Cardiovascular Agents/pharmacology , Galectin 3/antagonists & inhibitors , Heart Diseases/prevention & control , Myocytes, Cardiac/drug effects , Pectins/pharmacology , Peroxiredoxins/metabolism , Animals , Cardiotoxicity , Cell Line , Disease Models, Animal , Doxorubicin , Galectin 3/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/physiopathology , Male , Myocytes, Cardiac/enzymology , Oxidative Stress/drug effects , Peroxiredoxins/genetics , Rats, Sprague-Dawley , Up-Regulation , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
14.
Cardiovasc Drugs Ther ; 34(4): 487-501, 2020 08.
Article En | MEDLINE | ID: mdl-32377826

BACKGROUND: Previous studies have demonstrated that a high-carbohydrate intake could induce metabolic syndrome (MetS) in male rats with marked cardiac functional abnormalities. In addition, studies mentioned some benefits of insulin application on these complications, but there are considerable disagreements among their findings. Therefore, we aimed to extend our knowledge on the in-vitro influence of insulin on left ventricular dysfunction and also in the isolated cardiomyocytes from MetS rats. RESULTS: At the organ function level, an acute insulin application (100-nM) provided an important beneficial effect on the left ventricular developed pressure in MetS rats. Furthermore, to treat the freshly isolated cardiomyocytes from MetS rats with insulin provided marked recoveries in elevated resting intracellular Ca2+-level, as well as significant prevention of prolonged action potential through an augmentation in depressed K+-channel currents. Insulin also normalized the cellular levels of increased ROS and phosphorylation of PKCα, together with normalizations of apoptotic markers in MetS cardiomyocytes through the insulin-mediated regulation of phospho-Akt. Since not only elevated PKCα-activity but also reductions in phospho-Akt are key modulators of titin-based cardiomyocyte stiffening in hyperglycemia, insulin treatment of the cardiomyocytes prevented the activation of titin via the above pathways. Furthermore, CK2α-activation and NOS-phosphorylation could be prevented with insulin treatment. Mechanistically, we found that impaired insulin signaling and elevated PKCα and CK2α activities, as well as depressed Akt phosphorylation, are key modulators of titin-based cardiomyocyte stiffening in MetS rats. CONCLUSION: We propose that restoring normal kinase activities and also increases in phospho-Akt by insulin can contribute marked recoveries in MetS heart function, indicating a promising approach to modulate titin-associated factors in heart dysfunction associated with type-2 diabetes mellitus. Graphical Abstract.


Casein Kinase II/metabolism , Connectin/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance , Insulin/pharmacology , Metabolic Syndrome/drug therapy , Myocytes, Cardiac/drug effects , Ventricular Dysfunction, Left/drug therapy , Ventricular Function, Left/drug effects , Action Potentials/drug effects , Animals , Calcium Signaling/drug effects , Disease Models, Animal , Isolated Heart Preparation , Male , Metabolic Syndrome/enzymology , Metabolic Syndrome/physiopathology , Myocytes, Cardiac/enzymology , Oxidative Stress/drug effects , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure/drug effects
15.
Int J Legal Med ; 134(5): 1741-1752, 2020 Sep.
Article En | MEDLINE | ID: mdl-32377925

Cocaine-related deaths occur regularly in forensic routine work. In cases in which the detected concentration of cocaine is rather low and other causes of death apart from intoxication can be ruled out, the question arises if adulterants of cocaine might have played a crucial role. In the present study, cardiac effects of cocaine, of the adulterant levamisole and of mixtures of both were evaluated using the isolated perfused Langendorff heart. While exposed to the substances, functional parameters heart rate, left ventricular pressure and coronary flow were documented. Relevant alterations of these parameters were found for cocaine as well as for levamisole. Exposing the hearts to a mixture of both resulted in a combination of these effects; the emergence of new alterations or an obvious aggravation were not detected. Nevertheless, the results imply that the consumption of cocaine adulterated with levamisole bares an increased risk for cardiac complications, especially in the presence of preexisting cardiac pathologies.


Cocaine/pharmacology , Drug Contamination , Isolated Heart Preparation , Levamisole/pharmacology , Animals , Coronary Circulation/drug effects , Heart Rate/drug effects , Male , Models, Animal , Rats , Rats, Wistar , Ventricular Pressure/drug effects
16.
Am J Hypertens ; 33(8): 775-783, 2020 08 04.
Article En | MEDLINE | ID: mdl-32301965

BACKGROUND: Aberrant activation of epidermal growth factor receptor (EGFR) signaling pathway is associated with the pathogenesis of pulmonary hypertension (PH). However, the effect of icotinib, a first generation of EGFR tyrosine kinase inhibitor (EGFR-TKI), on PH remains to be elucidated. METHODS: PH rat model was established by a single intraperitoneal injection of monocrotaline (MCT, 60 mg/kg). Icotinib (15, 30, and 60 mg/kg/day) was administered by oral gavage from the day of MCT injection. After 4 weeks, hemodynamic parameters and histological changes of the pulmonary arterial vessels were assessed, and the phenotypic switching of pulmonary arterial smooth muscle cells (PASMCs) was determined in vivo. Moreover, the effects of icotinib (10 µM) on epidermal growth factor (EGF, 50 ng/ml)-stimulated proliferation, migration, and phenotypic switching of human PASMCs were explored in vitro. RESULTS: Icotinib significantly reduced the right ventricular systolic pressure and right ventricle hypertrophy index in rats with MCT-induced PH. Moreover, icotinib improved MCT-induced pulmonary vascular remodeling. The expression of contractile marker (smooth muscle 22 alpha (SM22α)) and synthetic markers (osteopontin (OPN) and vimentin) in pulmonary artery was restored by icotinib treatment. In vitro, icotinib suppressed EGF-induced PASMCs proliferation and migration. Meanwhile, icotinib inhibited EGF-induced downregulation of α-smooth muscle actin and SM22α and upregulation of OPN and Collagen I in PASMCs, suggesting that icotinib could inhibit EGF-induced phenotypic switching of PASMCs. Mechanistically, these effects of icotinib were associated with the inhibition of EGFR-Akt/ERK signaling pathway. CONCLUSIONS: Icotinib can attenuate MCT-induced pulmonary vascular remodeling and improve PH. This effect of icotinib might be attributed to preventing PASMC dysfunction by inhibiting EGFR-Akt/ERK signaling pathway.


Crown Ethers/pharmacology , ErbB Receptors/antagonists & inhibitors , Hypertension, Pulmonary/physiopathology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/drug effects , Quinazolines/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Epidermal Growth Factor/pharmacology , Hypertension, Pulmonary/chemically induced , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Microfilament Proteins/drug effects , Microfilament Proteins/metabolism , Monocrotaline/toxicity , Muscle Proteins/drug effects , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/physiopathology , Osteopontin/drug effects , Osteopontin/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/physiopathology , Rats , Signal Transduction , Vascular Remodeling/drug effects , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects , Vimentin/drug effects , Vimentin/metabolism
17.
J Cardiovasc Pharmacol ; 75(5): 460-474, 2020 05.
Article En | MEDLINE | ID: mdl-32195757

Estrogenic deficiency is considered a risk of coronary disease in women. The phytoestrogen genistein could be a safe preventive strategy. The first aim of this work was to validate a model of cardiac stunning in which natural estrogenic deficiency rats, ie, adult young male (YM) and aged female (AgF), are compared with young female rats (YF). The second aim was to study whether the in vivo administration of genistein prevents the stunning in estrogenic deficiency rats. The third aim was to evaluate whether in our estrogenic deficiency model exists a synergy between genistein and estradiol. The fourth aim was to characterize the underlying mechanisms of genistein. Stunning was induced by ischemia/reperfusion (I/R) in isolated hearts inside a calorimeter. The left ventricular pressure (P) and total heat rate (Ht) were simultaneously measured, while diastolic contracture and muscle economy (P/Ht) were calculated. During R, P/Ht and P recovered less in AgF and YM than in YF rat hearts. Genistein through i.p. (GST-ip) improved P and P/Ht in AgF and YM, but not in YF. In YM, the cardioprotections of GST-ip and estradiol were synergistic. After ischemia, GST-ip increased SR Ca leak causing diastolic contracture. The GST-ip cardioprotection neither was affected by blockade of PI3K-Akt, NO synthases, or phosphatases, but it was sensitive to blockade of protein-kinase C and mKATP channels. Results suggest that (1) estrogenic deficiency worsens cardiac stunning, (2) GST-ip was more cardioprotective in estrogenic deficiency and synergistic with estradiol, and (3) cardioprotection of GST-ip depends on the protein-kinase C and mKATP channel pathway activation.


Energy Metabolism/drug effects , Genistein/pharmacology , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Myocardial Stunning/prevention & control , Myocytes, Cardiac/drug effects , Phytoestrogens/pharmacology , Potassium Channels/metabolism , Age Factors , Animals , Calcium Signaling , Disease Models, Animal , Estradiol/pharmacology , Female , Isolated Heart Preparation , Male , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Stunning/enzymology , Myocardial Stunning/pathology , Myocardial Stunning/physiopathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Protein Kinase C/metabolism , Rats, Sprague-Dawley , Sex Factors , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
18.
J Cardiovasc Pharmacol ; 75(6): 596-602, 2020 06.
Article En | MEDLINE | ID: mdl-32168153

Chrysin (CH) is the main ingredient of many medicinal plants. Our previous study showed that CH could suppress hypoxia-induced pulmonary arterial smooth muscle cells proliferation and alleviate chronic hypoxia-induced pulmonary hypertension by targeting store-operated Ca entry (SOCE)-[Ca]i pathway. In this study, we investigated the effect of CH on monocrotaline-induced pulmonary hypertension (MCTPH) and the mechanism behind it. Results show that, in MCTPH model rats, (1) CH significantly reduced the enhancement of right ventricular pressure, right ventricular hypertrophy, and pulmonary vascular remodeling; (2) CH markedly suppressed the promotion of SOCE and [Ca]i in pulmonary arterial smooth muscle cells; and (3) CH obviously inhibited the MCT-upregulated proliferating cell nuclear antigen, TRPC1, TRPC4, and TRPC6 expression in distal pulmonary arteries. These results demonstrate that CH likely alleviates MCTPH by targeting TRPC1,4,6-SOCE-[Ca]i pathway.


Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Calcium Signaling/drug effects , Flavonoids/pharmacology , Hypertension, Pulmonary/drug therapy , Monocrotaline , Muscle, Smooth, Vascular/drug effects , Vascular Remodeling/drug effects , Animals , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/metabolism , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects , Ventricular Remodeling/drug effects
19.
Braz J Med Biol Res ; 53(3): e8761, 2020.
Article En | MEDLINE | ID: mdl-32159612

Nitric oxide (NO) inhibition by high-dose NG-nitro-L-arginine methyl ester (L-NAME) is associated with several detrimental effects on the cardiovascular system. However, low-dose L-NAME increases NO synthesis, which in turn induces physiological cardiovascular benefits, probably by activating a protective negative feedback mechanism. Aerobic exercise, likewise, improves several cardiovascular functions in healthy hearts, but its effects are not known when chronically associated with low-dose L-NAME. Thus, we tested whether the association between low-dose L-NAME administration and chronic aerobic exercise promotes beneficial effects to the cardiovascular system, evaluating the cardiac remodeling process. Male Wistar rats were randomly assigned to control (C), L-NAME (L), chronic aerobic exercise (Ex), and chronic aerobic exercise associated to L-NAME (ExL). Aerobic training was performed with progressive intensity for 12 weeks; L-NAME (1.5 mg·kg-1·day-1) was administered by orogastric gavage. Low-dose L-NAME alone did not change systolic blood pressure (SBP), but ExL significantly increased SBP at week 8 with normalization after 12 weeks. Furthermore, ExL promoted the elevation of left ventricle (LV) end-diastolic pressure without the presence of cardiac hypertrophy and fibrosis. Time to 50% shortening and relaxation were reduced in ExL, suggesting a cardiomyocyte contractile improvement. In addition, the time to 50% Ca2+ peak was increased without alterations in Ca2+ amplitude and time to 50% Ca2+ decay. In conclusion, the association of chronic aerobic exercise and low-dose L-NAME prevented cardiac pathological remodeling and induced cardiomyocyte contractile function improvement; however, it did not alter myocyte affinity and sensitivity to intracellular Ca2+ handling.


Calcium/analysis , Enzyme Inhibitors/pharmacology , Myocardial Contraction/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/drug effects , Physical Conditioning, Animal/physiology , Adiposity , Animals , Body Weight/physiology , Enzyme Inhibitors/administration & dosage , Hemodynamics , Male , Models, Animal , Motor Activity/physiology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , NG-Nitroarginine Methyl Ester/administration & dosage , Nitric Oxide Synthase/metabolism , Rats, Wistar , Ventricular Pressure/drug effects
20.
Cardiovasc Diabetol ; 19(1): 13, 2020 02 08.
Article En | MEDLINE | ID: mdl-32035482

BACKGROUND AND AIMS: Sodium-glucose linked cotransporter-2 (SGLT2) inhibitors reduce the likelihood of hospitalization for heart failure and cardiovascular death in both diabetic and non-diabetic individuals with reduced ejection fraction heart failure. Because SGLT2 inhibitors lead to volume contraction with reductions in both preload and afterload, these load-dependent factors are thought to be major contributors to the cardioprotective effects of the drug class. Beyond these effects, we hypothesized that SGLT2 inhibitors may also improve intrinsic cardiac function, independent of loading conditions. METHODS: Pressure-volume (P-V) relationship analysis was used to elucidate changes in intrinsic cardiac function, independent of alterations in loading conditions in animals with experimental myocardial infarction, a well-established model of HFrEF. Ten-week old, non-diabetic Fischer F344 rats underwent ligation of the left anterior descending (LAD) coronary artery to induce myocardial infarction (MI) of the left ventricle (LV). Following confirmation of infarct size with echocardiography 1-week post MI, animals were randomized to receive vehicle, or the SGLT2 inhibitor, empagliflozin. Cardiac function was assessed by conductance catheterization just prior to termination 6 weeks later. RESULTS: The circumferential extent of MI in animals that were subsequently randomized to vehicle or empagliflozin groups was similar. Empagliflozin did not affect fractional shortening (FS) as assessed by echocardiography. In contrast, load-insensitive measures of cardiac function were substantially improved with empagliflozin. Load-independent measures of cardiac contractility, preload recruitable stroke work (PRSW) and end-systolic pressure volume relationship (ESPVR) were higher in rats that had received empagliflozin. Consistent with enhanced cardiac performance in the heart failure setting, systolic blood pressure (SBP) was higher in rats that had received empagliflozin despite its diuretic effects. A trend to improved diastolic function, as evidenced by reduction in left ventricular end-diastolic pressure (LVEDP) was also seen with empagliflozin. MI animals treated with vehicle demonstrated myocyte hypertrophy, interstitial fibrosis and evidence for changes in key calcium handling proteins (all p < 0.05) that were not affected by empagliflozin therapy. CONCLUSION: Empagliflozin therapy improves cardiac function independent of loading conditions. These findings suggest that its salutary effects are, at least in part, due to actions beyond a direct effect of reduced preload and afterload.


Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Heart Failure/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects , Animals , Calcium Signaling/drug effects , Disease Models, Animal , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Rats, Inbred F344
...