Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38672089

ABSTRACT

There is ample evidence on the benefit of angiotensin receptor-neprilysin inhibitors (ARNIs) in heart failure, yet data regarding the potential protective action of ARNIs in hypertensive heart disease are sparse. The aim of this study was to show whether an ARNI exerts a protective effect in a model of Nω-nitro-L-arginine methyl ester (L-NAME)-induced hypertension with a hypertensive heart and to compare this potential benefit with an angiotensin-converting enzyme inhibitor, captopril. Five groups of adult male Wistar rats were studied (14 per group) for four weeks: untreated controls; ARNI (68 mg/kg/day); L-NAME (40 mg/kg/day); L-NAME treated with ARNI; and L-NAME treated with captopril (100 mg/kg/day). L-NAME administration induced hypertension, accompanied by increased left ventricular (LV) weight and fibrotic rebuilding of the LV in terms of increased concentration and content of hydroxyproline in insoluble collagen and in total collagen and with a histological finding of fibrosis. These alterations were associated with a compromised systolic and diastolic LV function. Treatment with either an ARNI or captopril reduced systolic blood pressure (SBP), alleviated LV hypertrophy and fibrosis, and prevented the development of both systolic and diastolic LV dysfunction. Moreover, the serum levels of prolactin and prolactin receptor were reduced significantly by ARNI and slightly by captopril. In conclusion, in L-NAME-induced hypertension, the dual inhibition of neprilysin and AT1 receptors by ARNI reduced SBP and prevented the development of LV hypertrophy, fibrosis, and systolic and diastolic dysfunction. These data suggest that ARNI could provide protection against LV structural remodeling and functional disorders in hypertensive heart disease.

2.
Int J Mol Sci ; 25(2)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38255922

ABSTRACT

We aimed to determine effects of aliskiren, a direct renin inhibitor, loaded onto polymeric nanoparticles on the (pro)renin receptor (Atp6ap2), angiotensin II type 1 receptor (Agtr1), and angiotensin-converting enzyme (ACE) gene expression in the heart of spontaneously hypertensive rats (SHR). Twelve-week-old male SHRs were divided into an untreated group and groups treated with powdered aliskiren or aliskiren-loaded nanoparticles (25 mg/kg/day). After three weeks, the accumulation of aliskiren, distribution of polymeric nanoparticles, gene expression of Atp6ap2 and Agtr1 receptors and ACE, and protein expression of NADPH oxidase along with the conjugated diene (CD) concentration were analyzed. The accumulation of aliskiren in the heart was higher in the aliskiren-loaded nanoparticle group than in the powdered group. The fluorescent signals of nanoparticles were visible in cardiomyocytes, vessel walls, and erythrocytes. Aliskiren-loaded nanoparticles decreased the gene expression of Atp6ap2 and ACE, while not affecting Agtr1. Both forms of aliskiren decreased the protein expression of NADPH oxidase, with a more pronounced effect observed in the aliskiren-loaded nanoparticle group. CD concentration was decreased only in the aliskiren-loaded nanoparticle group. We hypothesize that aliskiren-loaded nanoparticle-mediated downregulation of Atp6ap2 and ACE may contribute to a decrease in ROS generation with beneficial effects in the heart. Moreover, polymeric nanoparticles may represent a promising tool for targeted delivery of aliskiren.


Subject(s)
Amides , Fumarates , Nanoparticles , Prorenin Receptor , Male , Animals , Rats , Rats, Inbred SHR , NADPH Oxidases/genetics , Myocytes, Cardiac , Polyenes , Gene Expression
3.
Biol Res ; 56(1): 55, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37875978

ABSTRACT

BACKGROUND: Angiotensin converting enzyme 2 (ACE2) plays a crucial role in the infection cycle of SARS-CoV-2 responsible for formation of COVID-19 pandemic. In the cardiovascular system, the virus enters the cells by binding to the transmembrane form of ACE2 causing detrimental effects especially in individuals with developed hypertension or heart disease. Zofenopril, a H2S-releasing angiotensin-converting enzyme inhibitor (ACEI), has been shown to be effective in the treatment of patients with essential hypertension; however, in conditions of ACE2 inhibition its potential beneficial effect has not been investigated yet. Therefore, the aim of the study was to determine the effect of zofenopril on the cardiovascular system of spontaneously hypertensive rats, an animal model of human essential hypertension and heart failure, under conditions of ACE2 inhibition induced by the administration of the specific inhibitor MLN-4760 (MLN). RESULTS: Zofenopril reduced MLN-increased visceral fat to body weight ratio although no changes in systolic blood pressure were recorded. Zofenopril administration resulted in a favorable increase in left ventricle ejection fraction and improvement of diastolic function regardless of ACE2 inhibition, which was associated with increased H2S levels in plasma and heart tissue. Similarly, the acute hypotensive responses induced by acetylcholine, L-NAME (NOsynthase inhibitor) and captopril (ACEI) were comparable after zofenopril administration independently from ACE2 inhibition. Although simultaneous treatment with zofenopril and MLN led to increased thoracic aorta vasorelaxation, zofenopril increased the NO component equally regardless of MLN treatment, which was associated with increased NO-synthase activity in aorta and left ventricle. Moreover, unlike in control rats, the endogenous H2S participated in maintaining of aortic endothelial function in MLN-treated rats and the treatment with zofenopril had no impact on this effect. CONCLUSIONS: Zofenopril treatment reduced MLN-induced adiposity and improved cardiac function regardless of ACE2 inhibition. Although the concomitant MLN and zofenopril treatment increased thoracic aorta vasorelaxation capacity, zofenopril increased the participation of H2S and NO in the maintenance of endothelial function independently from ACE2 inhibition. Our results confirmed that the beneficial effects of zofenopril were not affected by ACE2 inhibition, moreover, we assume that ACE2 inhibition itself can lead to the activation of cardiovascular compensatory mechanisms associated with Mas receptor, nitrous and sulfide signaling.


Subject(s)
Captopril , Cardiovascular System , Humans , Rats , Animals , Captopril/pharmacology , Rats, Inbred SHR , Angiotensin-Converting Enzyme 2/pharmacology , Pandemics , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Blood Pressure , Essential Hypertension
4.
Gen Physiol Biophys ; 42(2): 209-215, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36896950

ABSTRACT

This study investigated whether chronic isoproterenol administration could induce kidney alterations and whether ivabradine, a heart rate (HR)-reducing substance exerting cardiovascular protection, is able to attenuate potential kidney damage. Twenty-eight Wistar rats were divided into non-diseased controls, rats treated with ivabradine, rats treated with isoproterenol, and rats treated with isoproterenol plus ivabradine. Six weeks of isoproterenol administration was associated with decreased systolic blood pressure (SBP) (by 25%) and glomerular, tubulointerstitial and vascular/perivascular fibrosis due to enhanced type I collagen volume (7-, 8-, and 4-fold, respectively). Ivabradine reduced HR (by 15%), partly prevented SBP decline (by 10%) and site-specifically mitigated kidney fibrosis by decreasing type I collagen volume in all three sites investigated (by 69, 58, and 67%, respectively) and the ratio of type I collagen-to-type III collagen in glomerular and vascular/perivascular sites (by 79 and 73%, respectively). We conclude that ivabradine exerts protection against kidney remodelling in isoproterenol-induced kidney damage.


Subject(s)
Collagen Type I , Kidney Diseases , Rats , Animals , Ivabradine/pharmacology , Isoproterenol/toxicity , Rats, Wistar , Kidney , Fibrosis , Heart Rate
5.
Front Pharmacol ; 13: 978337, 2022.
Article in English | MEDLINE | ID: mdl-36176443

ABSTRACT

Lactacystin is a specific proteasome inhibitor that blocks the hydrolysis of intracellular proteins by ubiquitin/proteasome system inhibition. The administration of lactacystin to rats induced hypertension and remodeling of the left ventricle and aorta. This study tested whether lactacystin induces structural and fibrotic rebuilding of the kidneys and whether melatonin and captopril can prevent these potential changes. Six weeks of lactacystin administration to rats increased their average systolic blood pressure (SBP). In the kidneys, lactacystin reduced glomerular density, increased the glomerular tuft area, and enhanced hydroxyproline concentrations. It also elevated the intraglomerular proportion including the amounts of collagen (Col) I and Col III. Lactacystin also raised the tubulointerstitial amounts of Col I and the sum of Col I and Col III with no effect on vascular/perivascular collagen. Six weeks of captopril treatment reduced SBP, while melatonin had no effect. Both melatonin and captopril increased glomerular density, reduced the glomerular tuft area, and lowered the hydroxyproline concentration in the kidneys. Both drugs reduced the proportion and total amounts of intraglomerular and tubulointerstitial Col I and Col III. We conclude that chronic lactacystin treatment stimulated structural and fibrotic remodeling of the kidneys, and melatonin and captopril partly prevented these alterations. Considering the effect of lactacystin on both the heart and kidneys, chronic treatment with this drug may be a prospective model of cardiorenal damage suitable for testing pharmacological drugs as protective agents.

6.
Int J Mol Sci ; 24(1)2022 Dec 23.
Article in English | MEDLINE | ID: mdl-36613727

ABSTRACT

In addition to their LDL-cholesterol-lowering effect, statins have pleiotropic beneficial effects on the cardiovascular system. However, long-term treatment with statins may be associated with serious side effects. With the aim to make statin therapy more effective, we studied the effects of simvastatin- and coenzyme-Q10-loaded polymeric nanoparticles on the lipid profile and nitric oxide (NO)/reactive oxygen species (ROS) balance in the heart and aorta of adult male obese Zucker rats. The rats were divided into an untreated group, a group treated with empty nanoparticles, and groups treated with simvastatin-, coenzyme Q10 (CoQ10)-, or a combination of simvastatin- and CoQ10-loaded nanoparticles (SIMV+CoQ10). After 6 weeks, the lipid profile in the plasma and the concentration of conjugated dienes in the liver were determined. Nitric oxide synthase (NOS) activity, Akt, endothelial NOS (eNOS), phosphorylated eNOS (p-eNOS), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and nuclear factor kappaB (NF-kappaB) protein expressions were measured in the heart and aorta. All simvastatin, CoQ10, and SIMV+CoQ10 treatments decreased plasma LDL levels, but only the combined SIMV+CoQ10 treatment increased NOS activity and the expression of Akt, eNOS, and p-eNOS in both the heart and the aorta. Interestingly, NADPH oxidase in the heart and NF-kappaB protein expression in the aorta were decreased by all treatments, including nanoparticles alone. In conclusion, only combined therapy with SIMV- and CoQ10-loaded nanoparticles increased NOS activity and upregulated the Akt-eNOS pathway in obese Zucker rats, which may represent a promising tool for the treatment of cardiometabolic diseases.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Metabolic Syndrome , Male , Rats , Animals , Simvastatin/pharmacology , Simvastatin/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt , Metabolic Syndrome/drug therapy , NF-kappa B , Rats, Zucker , Nitric Oxide Synthase Type III/metabolism , Obesity/drug therapy , Lipids
7.
Nanomaterials (Basel) ; 10(10)2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33050346

ABSTRACT

This study aimed to develop the method for determination of the ultra-small superparamagnetic iron oxide nanoparticle (USPION)-originated iron (UOI) in the tissues of rats on the basis of the magnetic characteristics (MC) in the liver, left heart ventricle (LHV), kidneys, aorta and blood of Wistar-Kyoto (WKY). Rats were treated intravenously by USPIONs dispersed in saline (transmission electron microscope (TEM) mean size ~30 nm, hydrodynamic size ~51 nm, nominal iron content 1 mg Fe/mL) at the low iron dose of 1 mg/kg. MC in the form of the mass magnetisation (M) versus the magnetic field (H) curves and temperature dependences of M (determined using the SQUID magnetometer), histochemical determination of iron (by Perl's method) and USPION-induced superoxide production (by lucigenin-enhanced chemiluminescence) were investigated 100 min post-infusion. USPIONs significantly elevated superoxide production in the liver, LHV, kidney and aorta vs. the control group. Histochemical staining confirmed the presence of iron in all solid biological samples, however, this method was not suitable to unequivocally confirm the presence of UOI. We improved the SQUID magnetometric method and sample preparation to allow the determination of UOI by measurements of the MC of the tissues at 300 K in solid and liquid samples. The presence of the UOI was confirmed in all the tissues investigated in USPIONs-treated rats. The greatest levels were found in blood and lower amounts in the aorta, liver, LHV and kidneys. In conclusion, we have improved SQUID-magnetometric method to make it suitable for detection of low amounts of UOI in blood and tissues of rats.

8.
Front Med (Lausanne) ; 7: 325, 2020.
Article in English | MEDLINE | ID: mdl-32754607

ABSTRACT

Hypertension-induced renal injury is characterized by structural kidney alterations and function deterioration. Therapeutics for kidney protection are limited, thus novel renoprotectives in hypertension are being continuously sought out. Ivabradine, an inhibitor of the If current in the sinoatrial node reducing heart rate (HR), was shown to be of benefit in various cardiovascular pathologies. Yet, data regarding potential renoprotection by ivabradine in hypertension are sparse. Thirty-six adult male Wistar rats were divided into non-diseased controls and rats with NG-nitro-L-arginine methyl ester (L-NAME)-induced hypertension to assess ivabradine's site-specific effect on kidney fibrosis. After 4 weeks of treatment, L-NAME increased the average systolic blood pressure (SBP) (by 27%), decreased glomerular density (by 28%) and increased glomerular tuft area (by 44%). Moreover, L-NAME induced glomerular, tubulointerstitial, and vascular/perivascular fibrosis by enhancing type I collagen volume (16-, 19- and 25-fold, respectively). L-NAME also increased the glomerular type IV collagen volume and the tubular injury score (3- and 8-fold, respectively). Ivabradine decreased average SBP and HR (by 8 and 12%, respectively), increased glomerular density (by 57%) and reduced glomerular tuft area (by 30%). Importantly, ivabradine decreased type I collagen volume at all three of the investigated sites (by 33, 38, and 72%, respectively) and enhanced vascular/perivascular type III collagen volume (by 67%). Furthermore, ivabradine decreased the glomerular type IV collagen volume and the tubular injury score (by 63 and 34%, respectively). We conclude that ivabradine attenuated the alterations of glomerular density and tuft area and modified renal fibrosis in a site-specific manner in L-NAME-hypertension. It is suggested that ivabradine may be renoprotective in hypertensive kidney disease.

9.
Molecules ; 24(15)2019 Jul 25.
Article in English | MEDLINE | ID: mdl-31349653

ABSTRACT

Aliskiren, a renin inhibitor, has been shown to have cardioprotective and blood pressure (BP) lowering effects. We aimed to determine the effects of nanoparticle-loaded aliskiren on BP, nitric oxide synthase activity (NOS) and structural alterations of the heart and aorta developed due to spontaneous hypertension in rats. Twelve week-old male spontaneously hypertensive rats (SHR) were divided into the untreated group, group treated with powdered or nanoparticle-loaded aliskiren (25 mg/kg/day) and group treated with nanoparticles only for 3 weeks by gavage. BP was measured by tail-cuff plethysmography. NOS activity, eNOS and nNOS protein expressions, and collagen content were determined in both the heart and aorta. Vasoactivity of the mesenteric artery and wall thickness, inner diameter, and cross-sectional area (CSA) of the aorta were analyzed. After 3 weeks, BP was lower in both powdered and nanoparticle-loaded aliskiren groups with a more pronounced effect in the latter case. Only nanoparticle-loaded aliskiren increased the expression of nNOS along with increased NOS activity in the heart (by 30%). Moreover, nanoparticle-loaded aliskiren decreased vasoconstriction of the mesenteric artery and collagen content (by 11%), and CSA (by 25%) in the aorta compared to the powdered aliskiren group. In conclusion, nanoparticle-loaded aliskiren represents a promising drug with antihypertensive and cardioprotective effects.


Subject(s)
Amides/administration & dosage , Antihypertensive Agents/administration & dosage , Cardiovascular System/drug effects , Drug Carriers , Fumarates/administration & dosage , Nanoparticles , Animals , Biomarkers , Blood Pressure/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Heart/drug effects , Hypertension/diagnosis , Hypertension/drug therapy , Hypertension/etiology , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Nitric Oxide/metabolism , Polyesters/chemistry , Rats , Rats, Inbred SHR
10.
Gen Physiol Biophys ; 38(3): 265-270, 2019 May.
Article in English | MEDLINE | ID: mdl-31184313

ABSTRACT

This study investigated the effect of lisinopril (angiotensin-converting enzyme inhibitor) on potential behavioural alterations in spontaneously hypertensive rats (SHR). Three groups of 15-17-week-old rats were investigated for 2 weeks: Wistar control group, SHR group and SHR+lisinopril group. Systolic blood pressure (SBP) was normal in Wistar rats, SHR expressed hypertension and lisinopril normalized the SBP. We observed increased time spent in and increased frequency of entries to the central area of the open field in SHR, while lisinopril induced a trend to reduce the time spent in the central area of the open field and reduced the frequency of entries there. There was a positive correlation between SBP and reduced anxiety-like behaviour in normotensive rats; no correlations in the SHR or SHR+lisinopril groups were observed. We conclude that lisinopril normalized the increase in SBP and partly reversed the alterations of anxiety-like behaviour in SHR.


Subject(s)
Antihypertensive Agents/pharmacology , Anxiety/drug therapy , Behavior, Animal/drug effects , Hypertension/drug therapy , Hypertension/psychology , Lisinopril/pharmacology , Animals , Anxiety/prevention & control , Blood Pressure , Hypertension/physiopathology , Rats , Rats, Inbred SHR , Rats, Wistar
11.
Molecules ; 24(9)2019 Apr 30.
Article in English | MEDLINE | ID: mdl-31052164

ABSTRACT

Myocardial infarction (MI) remains the leading cause of death worldwide. We aimed to investigate the effect of NO deficiency on selective biochemical parameters within discreet myocardial zones after experimentally induced MI. To induce MI, the left descending coronary artery was ligated in two groups of 16-week-old WKY rats. In one group, NO production was inhibited by L-NAME (20 mg/kg/day) administration four weeks prior to ligation. Sham operations were performed on both groups as a control. Seven days after MI, we evaluated levels of nitric oxide synthase (NOS) activity, eNOS, iNOS, NFÒ¡B/p65 and Nrf2 in ischemic, injured and non-ischemic zones of the heart. Levels of circulating TNF-α and IL-6 were evaluated in the plasma. MI led to increased NOS activity in all investigated zones of myocardium as well as circulating levels of TNF-α and IL-6. L-NAME treatment decreased NOS activity in the heart of sham operated animals. eNOS expression was increased in the injured zone and this could be a compensatory mechanism that improves the perfusion of the myocardium and cardiac dysfunction. Conversely, iNOS expression increased in the infarcted zone and may contribute to the inflammatory process and irreversible necrotic changes.


Subject(s)
Adaptation, Physiological , Blood Pressure , Hypertension/metabolism , Hypertension/physiopathology , Nitric Oxide/metabolism , Animals , Biomarkers , Blood Pressure/drug effects , Body Weight , Cytokines/metabolism , Enzyme Activation , Gene Expression , Hypertension/etiology , NG-Nitroarginine Methyl Ester/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Organ Size , Rats
12.
Oxid Med Cell Longev ; 2016: 9814038, 2016.
Article in English | MEDLINE | ID: mdl-27148433

ABSTRACT

Activation of nuclear factor-κB (NF-κB) by increased production of reactive oxygen species (ROS) might induce transcription and expression of different antioxidant enzymes and also of nitric oxide synthase (NOS) isoforms. Thus, we aimed at studying the effect of NF-κB inhibition, caused by JSH-23 (4-methyl-N (1)-(3-phenyl-propyl)-benzene-1,2-diamine) injection, on ROS and NO generation in hereditary hypertriglyceridemic (HTG) rats. 12-week-old, male Wistar and HTG rats were treated with JSH-23 (bolus, 10 µmol, i.v.). After one week, blood pressure (BP), superoxide dismutase (SOD) activity, SOD1, endothelial NOS (eNOS), and NF-κB (p65) protein expressions were higher in the heart of HTG rats compared to control rats. On the other hand, NOS activity was decreased. In HTG rats, JSH-23 treatment increased BP and heart conjugated dienes (CD) concentration (measured as the marker of tissue oxidative damage). Concomitantly, SOD activity together with SOD1 expression was decreased, while NOS activity and eNOS protein expression were increased significantly. In conclusion, NF-κB inhibition in HTG rats led to decreased ROS degradation by SOD followed by increased oxidative damage in the heart and BP elevation. In these conditions, increased NO generation may represent rather a counterregulatory mechanism activated by ROS. Nevertheless, this mechanism was not sufficient enough to compensate BP increase in HTG rats.


Subject(s)
Myocardium/metabolism , Transcription Factor RelA/metabolism , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Gene Expression/drug effects , Glutathione/analysis , Heart Ventricles/metabolism , Hyperlipoproteinemia Type IV/pathology , Hyperlipoproteinemia Type IV/veterinary , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Phenylenediamines/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factor RelA/genetics
13.
Food Funct ; 6(7): 2187-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26039974

ABSTRACT

We aimed to determine the effects of curcumin on liver fibrosis and to clarify the role of nuclear factor-κB (NF-κB) and inducible nitric oxide synthase (iNOS) in a model of microsurgical cholestasis in the early stage of extrahepatic biliary atresia. Twelve-week-old Wistar rats were divided into four groups (n = 8): sham-operated rats (received olive oil after laparotomy); a curcumin group (received curcumin, 200 mg kg(-1) per day, after laparotomy); a biliary duct ligated group (BDL, received olive oil after operation); and a biliary duct ligated/curcumin group (BDL curc, received curcumin, 200 mg kg(-1) per day, after operation). After 3 weeks of treatment, curcumin did not modify blood plasma markers as well as the expressions of iNOS and NF-κB (p65) in the livers of the sham group. Interestingly, there was a significant increase in the extent of both liver and kidney fibrosis. On the other hand, despite a decrease in the expression of iNOS and NF-κB (p65), treatment with curcumin did not affect fibrosis enlargement due to bile duct ligation in the liver. In the BDL group, treatment with curcumin decreased the level of blood plasma markers investigated. In conclusion, treatment with curcumin was able to improve the functional properties of hepatocytes and to inhibit the upregulations of both NF-κB and iNOS in the BDL group; however, no beneficial effect was observed on the liver fibrosis developed in this model of cholestasis. Thus, in the studied model of microsurgical cholestasis, other factors different from NF-κB and iNOS are responsible for fibrotic processes in the liver.


Subject(s)
Cholestasis/surgery , Curcumin/administration & dosage , Liver Cirrhosis/drug therapy , Animals , Bile Ducts/surgery , Cholestasis/drug therapy , Cholestasis/genetics , Cholestasis/metabolism , Disease Models, Animal , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/surgery , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar
14.
Food Funct ; 5(9): 2202-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25051230

ABSTRACT

We aimed to analyse the effects of alcohol-free Alibernet red wine extract (AWE) on nitric oxide synthase (NOS) activity and pro-inflammatory markers such as nuclear factor-κB (NFκB) and inducible NOS (iNOS) protein expression in experimental metabolic syndrome. Young 6 week-old male Wistar Kyoto (WKY) and obese, spontaneously hypertensive rats (SHR/N-cp) were divided into control groups and groups treated with AWE (24.2 mg per kg per day) for 3 weeks (n = 6 in each group). Total NOS activity and endothelial NOS (eNOS), iNOS and NFκB (p65) protein expressions were determined in the heart left ventricle and aorta by Western blot and immunohistochemical analysis. All parameters investigated significantly increased in the aorta of SHR/N-cp rats. Pro-inflammatory markers such as NFκB and iNOS were increased in the left ventricle as well. AWE treatment did not affect total NOS activity and eNOS expression in the aorta; however, it was able to decrease NFκB and iNOS protein expression in both the left ventricle and aorta. In conclusion, in the cardiovascular system, Alibernet red wine extract decreased NFκB and iNOS protein expressions elevated as a consequence of developed metabolic syndrome. This effect may represent one of the protective, anti-inflammatory properties of Alibernet red wine polyphenols on cardiovascular risk factors related to metabolic syndrome.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Metabolic Syndrome/drug therapy , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Wine/analysis , Animals , Anti-Inflammatory Agents/analysis , Down-Regulation , Humans , Male , NF-kappa B/immunology , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/immunology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar
15.
Oxid Med Cell Longev ; 2012: 806285, 2012.
Article in English | MEDLINE | ID: mdl-22720118

ABSTRACT

We aimed to perform a chemical analysis of both Alibernet red wine and an alcohol-free Alibernet red wine extract (AWE) and to investigate the effects of AWE on nitric oxide and reactive oxygen species production as well as blood pressure development in normotensive Wistar Kyoto (WKY) and spontaneously hypertensive rats (SHRs). Total antioxidant capacity together with total phenolic and selected mineral content was measured in wine and AWE. Young 6-week-old male WKY and SHR were treated with AWE (24,2 mg/kg/day) for 3 weeks. Total NOS and SOD activities, eNOS and SOD1 protein expressions, and superoxide production were determined in the tissues. Both antioxidant capacity and phenolic content were significantly higher in AWE compared to wine. The AWE increased NOS activity in the left ventricle, aorta, and kidney of SHR, while it did not change NOS activity in WKY rats. Similarly, increased SOD activity in the plasma and left ventricle was observed in SHR only. There were no changes in eNOS and SOD1 expressions. In conclusion, phenolics and minerals included in AWE may contribute directly to increased NOS and SOD activities of SHR. Nevertheless, 3 weeks of AWE treatment failed to affect blood pressure of SHR.


Subject(s)
Hypertension/metabolism , Nitric Oxide/metabolism , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Wine/analysis , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Aorta/drug effects , Aorta/enzymology , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Hypertension/pathology , Kidney/drug effects , Kidney/enzymology , Male , Minerals/analysis , Minerals/pharmacology , Nitric Oxide Synthase Type III/metabolism , Plant Extracts/chemistry , Polyphenols/analysis , Polyphenols/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Vitis/chemistry
16.
J Hypertens ; 28 Suppl 1: S45-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20823716

ABSTRACT

OBJECTIVE: We aimed to analyze effects of nuclear factor-kappa B (NF-kappaB) inhibition on blood pressure (BP) regulation and cardiovascular remodelling. DESIGN: Adult 12-week-old male Wistar Kyoto rats (WKY) were treated with the nitric oxide synthase (NOS) inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME, 40 mg/kg/day) for seven weeks. From the fourth week of L-NAME treatment, the NF-kappaB inhibitor lactacystin (1 microg/kg) was applied once a week. Furthermore, age-matched WKY received L-NAME or lactacystin alone for 7 or 3 weeks, respectively. METHODS: Total NOS activity was determined in the left ventricle (LV) and aorta. The concentration of conjugated dienes, fibrosis, and collagen I and III levels were determined in the LV. The cross-sectional area (CSA) and wall thickness to internal diameter ratio (WT/ID) were measured in the aorta. RESULTS: L-NAME treatment increased BP significantly (145 +/- 2 mmHg vs. 110 +/- 1 mmHg in controls). The addition of lactacystin resulted in further significant increase in BP (161 +/- 3 mmHg). Similarly, lactacystin potentiated the increased conjugated dienes concentration induced by L-NAME. Whereas L-NAME alone did not affect NOS activity, the addition of lactacystin decreased it in both tissues investigated. The addition of lactacystin did not affect LV hypertrophy, fibrosis, and collagen I and III, already increased by L-NAME; however, it further amplified CSA in the aorta increased by L-NAME alone. WT/ID increased significantly only after the addition of lactacystin. CONCLUSION: Decreased NOS activity along with increased oxidative load may be responsible for decreased NO bio-availability and further BP increase after NF-kappaB inhibition in L-NAME-induced hypertension. Increased CSA and WT/ID could contribute to this hypertensive process.


Subject(s)
Hypertension/chemically induced , NF-kappa B/antagonists & inhibitors , NG-Nitroarginine Methyl Ester/adverse effects , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Blood Pressure , Male , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Inbred WKY , Ventricular Remodeling/drug effects
17.
J Pineal Res ; 48(2): 102-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20041987

ABSTRACT

The mechanisms responsible for the antihypertensive effect of melatonin are not completely understood. To elucidate the possible role of the nitric oxide (NO) pathway in the hemodynamic actions of melatonin, the effects of this indolamine on vascular function during hypertension induced by the NO-synthase (NOS) inhibitor, N(omega)-nitro-L-arginine-methyl ester (L-NAME) were investigated. Four groups of male adult Wistar rats were employed: control, L-NAME (40 mg/kg), melatonin (10 mg/kg) and L-NAME + melatonin for 5 wks. Systolic and diastolic blood pressure were measured invasively in the carotid artery. Conjugated dienes concentration (an oxidative load marker), NOS RNA expression and its activity and RNA expression of cyclooxygenase-(COX)-1 and COX-2 were determined in the aorta. Acetylcholine-induced responses and their NO-mediated component were evaluated in femoral and mesenteric artery. Moreover, endothelium-derived constricting factor (EDCF)-dependent vasoconstriction and inner diameter were determined in the femoral artery. Chronic L-NAME treatment induced hypertension, elevated the oxidative load and inhibited NOS activity. Moreover, impaired NO-dependent relaxation, augmented EDCF-constriction, increased COX-2 expression and reduced arterial inner diameter were observed. Melatonin added to L-NAME treatment completely prevented elevation of the oxidative load in the aorta. However, melatonin was not able to prevent NOS activity decline, elevation of COX-2 expression or the impairment of vascular responses (except moderate improvement in relaxation of small mesenteric arteries) and it exerted only slight antihypertensive effect. In conclusion, in addition to the reduction of the oxidative load, the restoration of the NO pathway seems to play an important role in the antihypertensive effect of melatonin.


Subject(s)
Hypertension/physiopathology , Melatonin/pharmacology , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cyclooxygenase 2/biosynthesis , Endothelins/pharmacology , Hypertension/chemically induced , Male , NG-Nitroarginine Methyl Ester , Nitric Oxide Synthase/metabolism , Rats , Rats, Wistar
18.
J Hypertens Suppl ; 27(6): S11-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19633445

ABSTRACT

OBJECTIVE: Melatonin was shown to reduce blood pressure, enhance nitric oxide availability and scavenge free radicals. There is, however, a shortage of data with respect to the effect of melatonin on pathological left ventricular remodelling associated with haemodynamic overload. DESIGN: We investigated whether melatonin was able to prevent left ventricular hypertrophy (LVH) and fibrosis associated with N(G)-nitro-L-arginine-methyl ester (L-NAME)-induced hypertension. METHODS: Four groups of male Wistar rats were investigated: control, L-NAME (50 mg/kg per day), melatonin (10 mg/kg per day) and L-NAME plus melatonin. Blood pressure was measured non-invasively each week. After 5 weeks of treatment the animals were killed and nitric oxide synthase (NOS) activity, endothelial and inducible NOS expression, the level of collagenous proteins, hydroxyproline and conjugated dienes in the left ventricle were determined. RESULTS: The administration of L-NAME inhibited NOS activity, increased conjugated dienes concentration, elevated blood pressure and induced LVH and fibrosis (indicated by increased collagenous proteins and hydroxyproline levels). The addition of melatonin to L-NAME treatment failed to prevent the attenuation of NOS activity and the development of LVH and prevented hypertension only partly. The administration of melatonin, however, completely prevented the increase in conjugated dienes concentration and the development of left ventricular fibrosis. NOS expression was not different among experimental groups. CONCLUSION: Melatonin prevented the development of left ventricular fibrosis and the increase in oxidative load in rats with L-NAME-induced hypertension. The antifibrotic effect of melatonin seems to be independent of its effects on NOS activity and might be linked to its antioxidant properties.


Subject(s)
Fibrosis/prevention & control , Heart Ventricles/pathology , Hypertrophy/prevention & control , Melatonin/pharmacology , NG-Nitroarginine Methyl Ester/metabolism , Animals , Blood Pressure , Free Radical Scavengers/pharmacology , Heart Ventricles/drug effects , Hemodynamics , Hydroxyproline/metabolism , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...