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










Publication year range
1.
J Cardiovasc Pharmacol ; 79(2): 206-216, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35099165

ABSTRACT

ABSTRACT: Accumulating evidence indicates that transient receptor potential (TRP) channels are involved in the pathophysiological process in the heart, and monoterpenes, such as carvacrol, are able to modulate these channels activity. In this article, our purpose was to evaluate the direct cardiac effect of carvacrol on the contractility of cardiomyocytes and isolated right atria from spontaneously hypertensive and Wistar Kyoto rats. In this way, in vitro experiments were used to evaluate the ventricular cardiomyocytes contractility and the Ca2+ transient measuring, in addition to heart rhythm in the right atria. The role of TRPM channels in carvacrol-mediated cardiac activities was also investigated. The results demonstrated that carvacrol induced a significant reduction in ventricular cell contractility, without changes in transient Ca2+. In addition, carvacrol promoted a significant negative chronotropic response in spontaneously hypertensive and Wistar Kyoto rats' atria. Selective blockage of TRPM channels suggests the involvement of TRP melastatin subfamily 2 (TRPM2), TRPM4, and TRPM7 in the carvacrol-mediated cardiac effects. In silico studies were conducted to further investigate the putative role of TRPM4 in carvacrol-mediated cardiac action. FTMap underscores a conserved pocket in both TRPM4 and TRPM7, revealing a potential carvacrol binding site, and morphological similarity analysis demonstrated that carvacrol shares a more than 85% similarity to 9-phenanthrol. Taken together, these results suggest that carvacrol has direct cardiac actions, leading to reduced cellular contractility and inducing a negative chronotropic effect, which may be related to TRPM7 and TRPM4 modulation.


Subject(s)
Hypertension , TRPM Cation Channels , Animals , Calcium/metabolism , Cymenes , Rats , Rats, Inbred SHR , Rats, Inbred WKY , TRPM Cation Channels/metabolism
2.
Oxid Med Cell Longev ; 2020: 4850697, 2020.
Article in English | MEDLINE | ID: mdl-32273944

ABSTRACT

BACKGROUND: Cardiomyopathies remain among the leading causes of death worldwide, despite all efforts and important advances in the development of cardiovascular therapeutics, demonstrating the need for new solutions. Herein, we describe the effects of the redox-active therapeutic Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, AEOL10113, BMX-010 (MnTE-2-PyP5+), on rat heart as an entry to new strategies to circumvent cardiomyopathies. METHODS: Wistar rats weighing 250-300 g were used in both in vitro and in vivo experiments, to analyze intracellular Ca2+ dynamics, L-type Ca2+ currents, Ca2+ spark frequency, intracellular reactive oxygen species (ROS) levels, and cardiomyocyte and cardiac contractility, in control and MnTE-2-PyP5+-treated cells, hearts, or animals. Cells and hearts were treated with 20 µM MnTE-2-PyP5+ and animals with 1 mg/kg, i.p. daily. Additionally, we performed electrocardiographic and echocardiographic analysis. RESULTS: Using isolated rat cardiomyocytes, we observed that MnTE-2-PyP5+ reduced intracellular Ca2+ transient amplitude, without altering cell contractility. Whereas MnTE-2-PyP5+ did not alter basal ROS levels, it was efficient in modulating cardiomyocyte redox state under stress conditions; MnTE-2-PyP5+ reduced Ca2+ spark frequency and increased sarcoplasmic reticulum (SR) Ca2+ load. Accordingly, analysis of isolated perfused rat hearts showed that MnTE-2-PyP5+ preserves cardiac function, increases SR Ca2+ load, and reduces arrhythmia index, indicating an antiarrhythmic effect. In vivo experiments showed that MnTE-2-PyP5+ treatment increased Ca2+ transient, preserved cardiac ejection fraction, and reduced arrhythmia index and duration. MnTE-2-PyP5+ was effective both to prevent and to treat cardiac arrhythmias. CONCLUSION: MnTE-2-PyP5+ prevents and treats cardiac arrhythmias in rats. In contrast to most antiarrhythmic drugs, MnTE-2-PyP5+ preserves cardiac contractile function, arising, thus, as a prospective therapeutic for improvement of cardiac arrhythmia treatment.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/prevention & control , Cardiovascular System/drug effects , Metalloporphyrins/therapeutic use , Oxidation-Reduction/drug effects , Animals , Male , Rats , Rats, Wistar
3.
J Pharm Pharmacol ; 67(12): 1682-95, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26256440

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the cytotoxic effect of new 1,4-naphthoquinone- 1,2,3-triazoles, named C2 to C8 triazole derivatives, towards human cancer cell lines. METHODS: The effect on cell viability was assessed by MTT and propidium iodide assays. The cytotoxic effect of C2 and C3 in K562 and HL-60 cells were analyzed by flow cytometry, DNA fragmentation and reactive oxygen species (ROS) production. Western blot and q-PCR procedures were also performed. KEY FINDINGS: C2 and C3 inhibited both K562 and HL-60 cells growth in a concentration-dependent manner. C2 presented the highest cytotoxic activity with an IC50 of approximately 14 µm and 41 µm for HL-60 and K562 cells, respectively, while being less toxic to normal peripheral blood monocyte cells. Both derivatives induced cellular changes in HL-60 cells, characteristic of apoptosis, such as mitochondrial membrane depolarization, phosphatidylserine externalization, increasing sub-G1 phase, DNA fragmentation, downregulating Bcl-2 protein and upregulating Bax protein. In K562 cells, C2 and C3 induced S-phase arrest of cell cycle, which was associated with upregulation of p21. The effect of these derivatives in HL-60 cells can be related to the ROS intracellular level. CONCLUSION: Taken together our results showed that C2 and C3 triazole derivatives presented the best potential for drug design.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia/drug therapy , Naphthoquinones/pharmacology , Triazoles/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Inhibitory Concentration 50 , K562 Cells , Leukemia/metabolism , Leukemia/pathology , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Naphthoquinones/chemistry , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Structure-Activity Relationship , Triazoles/chemistry
4.
Appl Physiol Nutr Metab ; 40(4): 393-400, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25659569

ABSTRACT

The hypothesis that oral supplementation with virgin coconut oil (Cocos nucifera L.) and exercise training would improve impaired baroreflex sensitivity (BRS) and reduce oxidative stress in spontaneously hypertensive rats (SHR) was tested. Adult male SHR and Wistar Kyoto rats (WKY) were divided into 5 groups: WKY + saline (n = 8); SHR + saline (n = 8); SHR + coconut oil (2 mL·day(-1), n = 8); SHR + trained (n = 8); and SHR + trained + coconut oil (n = 8). Mean arterial pressure (MAP) was recorded and BRS was tested using phenylephrine (8 µg/kg, intravenous) and sodium nitroprusside (25 µg·kg(-1), intravenous). Oxidative stress was measured using dihydroethidium in heart and aorta. SHR + saline, SHR + coconut oil, and SHR + trained group showed higher MAP compared with WKY + saline (175 ± 6, 148 ± 6, 147 ± 7 vs. 113 ± 2 mm Hg; p < 0.05). SHR + coconut oil, SHR + trained group, and SHR + trained + coconut oil groups presented lower MAP compared with SHR + saline group (148 ± 6, 147 ± 7, 134 ± 8 vs. 175 ± 6 mm Hg; p < 0.05). Coconut oil combined with exercise training improved BRS in SHR compared with SHR + saline group (-2.47 ± 0.3 vs. -1.39 ± 0.09 beats·min(-1)·mm Hg(-1); p < 0.05). SHR + saline group showed higher superoxide levels when compared with WKY + saline (774 ± 31 vs. 634 ± 19 arbitrary units (AU), respectively; p < 0.05). SHR + trained + coconut oil group presented reduced oxidative stress compared with SHR + saline in heart (622 ± 16 vs. 774 ± 31 AU, p < 0.05). In aorta, coconut oil reduced oxidative stress in SHR compared with SHR + saline group (454 ± 33 vs. 689 ± 29 AU, p < 0.05). Oral supplementation with coconut oil combined with exercise training improved impaired BRS and reduced oxidative stress in SHR.


Subject(s)
Baroreflex , Hypertension/therapy , Oxidative Stress/drug effects , Physical Conditioning, Animal , Plant Oils/pharmacology , Animals , Blood Pressure/drug effects , Coconut Oil , Heart Rate/drug effects , Lipid Peroxidation/drug effects , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sensitivity and Specificity , Superoxides/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Weight Gain/drug effects
5.
J Renin Angiotensin Aldosterone Syst ; 15(4): 480-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23386282

ABSTRACT

INTRODUCTION: Angiotensin (Ang) A was first identified in human plasma and it differs from Ang II in Ala(1) instead of Asp(1). Here, we hypothesized that the actions of this peptide might explain, at least partially, the limited effects of AT1R antagonists in certain cardiovascular diseases. MATERIALS AND METHODS: The effects of Ang A and Ang II on blood pressure (BP) and heart function were compared. Importantly, participation of AT1R in these effects was evaluated. Furthermore, the effects of these two peptides on ischemia/reperfusion arrhythmias and involvement of calcium in these effects were investigated. RESULTS: Administration of increasing doses of these peptides caused elevations in BP at comparable magnitude. AT1R blockade completely abolished these effects. The actions of these peptides in cardiac function were quite similar although the effects of Ang A were only partially blocked by losartan. Interestingly, Ang II elicited an increase in the duration of ischemia/reperfusion arrhythmias while Ang A had no effect on cardiac rhythm during reperfusion. In accordance, differently to Ang II, Ang A did not induce any significant effect on calcium transient during baseline and ischemic stress conditions. CONCLUSIONS: These data suggest that the existence of alternative peptides of the renin-angiotensin system (RAS) might contribute to the limited effects of angiotensin receptor blockers (ARBs) in certain pathophysiological circumstances.


Subject(s)
Angiotensins/pharmacology , Cardiovascular System/drug effects , Peptides/pharmacology , Renin-Angiotensin System/drug effects , Animals , Blood Pressure/drug effects , Calcium Signaling/drug effects , Heart/drug effects , Heart/physiopathology , Heart Function Tests , Humans , In Vitro Techniques , Male , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats, Wistar
6.
PLoS One ; 8(5): e62452, 2013.
Article in English | MEDLINE | ID: mdl-23658728

ABSTRACT

The use of ß-blockers is mandatory for counteracting heart failure (HF)-induced chronic sympathetic hyperactivity, cardiac dysfunction and remodeling. Importantly, aerobic exercise training, an efficient nonpharmacological therapy to HF, also counteracts sympathetic hyperactivity in HF and improves exercise tolerance and cardiac contractility; the latter associated with changes in cardiac Ca(2+) handling. This study was undertaken to test whether combined ß-blocker and aerobic exercise training would integrate the beneficial effects of isolated therapies on cardiac structure, contractility and cardiomyocyte Ca(2+) handling in a genetic model of sympathetic hyperactivity-induced HF (α2A/α2C- adrenergic receptor knockout mice, KO). We used a cohort of 5-7 mo male wild-type (WT) and congenic mice (KO) with C57Bl6/J genetic background randomly assigned into 5 groups: control (WT), saline-treated KO (KOS), exercise trained KO (KOT), carvedilol-treated KO (KOC) and, combined carvedilol-treated and exercise-trained KO (KOCT). Isolated and combined therapies reduced mortality compared with KOS mice. Both KOT and KOCT groups had increased exercise tolerance, while groups receiving carvedilol had increased left ventricular fractional shortening and reduced cardiac collagen volume fraction compared with KOS group. Cellular data confirmed that cardiomyocytes from KOS mice displayed abnormal Ca(2+) handling. KOT group had increased intracellular peak of Ca(2+) transient and reduced diastolic Ca(2+) decay compared with KOS group, while KOC had increased Ca(2+) decay compared with KOS group. Notably, combined therapies re-established cardiomyocyte Ca(2+) transient paralleled by increased SERCA2 expression and SERCA2:PLN ratio toward WT levels. Aerobic exercise trained increased the phosphorylation of PLN at Ser(16) and Thr(17) residues in both KOT and KOCT groups, but carvedilol treatment reduced lipid peroxidation in KOC and KOCT groups compared with KOS group. The present findings provide evidence that the combination of carvedilol and aerobic exercise training therapies lead to a better integrative outcome than carvedilol or exercise training used in isolation.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carbazoles/pharmacology , Exercise Therapy , Heart Failure/therapy , Myocardial Contraction , Propanolamines/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Animals , Blood Pressure , Calcium Signaling , Carbazoles/therapeutic use , Carvedilol , Cells, Cultured , Combined Modality Therapy , Drug Evaluation, Preclinical , Exercise Tolerance , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Rate , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Oxidative Stress , Physical Conditioning, Animal , Propanolamines/therapeutic use , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Ventricular Remodeling
7.
Seizure ; 22(4): 259-66, 2013 May.
Article in English | MEDLINE | ID: mdl-23340275

ABSTRACT

PURPOSE: Cardiac dysfunction is one of the possible causes of sudden unexpected death in epilepsy (SUDEP). Therefore, the objective of this study was to evaluate cardiac and electrocardiographic parameters in rats with audiogenic epileptic seizures (WAR--Wistar audiogenic rats). METHODS: In vivo arterial pressure, heart rate (HR), autonomic tone and electrocardiography (ECG) were measured in awake animals in order to examine cardiac function and rhythm. Ex vivo, the Langendorff technique was used to analyze the cardiac function and the severity of reperfusion arrhythmias. In vitro, confocal microscopy was used to evaluate calcium transient parameters of isolated ventricular cardiomyocytes. RESULTS: In vivo autonomic tone evaluation revealed enhanced sympathetic activity, changes in cardiac function with increased systolic arterial pressure and higher basal HR in WAR. In addition, ECG analysis demonstrated electrical alterations with prolongation of the QT interval and QRS complex in these animals. Ex vivo, we observed a decrease in systolic tone and HR and an increase in the duration of ischemia/reperfusion arrhythmias in WAR. Moreover, intracellular Ca2+ handling analysis revealed an increase in the peak of calcium and calcium transient decay in audiogenic rats. Treatment with atenolol (ß1-adrenergic antagonist) normalized the systolic tone, reduced cardiac hypertrophy and the associated increase in the susceptibility to reperfusion arrhythmias observed in WAR. CONCLUSION: We present evidence that chronic disturbances in sympathetic tone in WAR cause increases the risk to life-threatening arrhythmias. Our results support a relationship between seizures, cardiac dysfunction and cardiac arrhythmias, which may contribute to the occurrence of SUDEP.


Subject(s)
Acoustic Stimulation/adverse effects , Arrhythmias, Cardiac/physiopathology , Epilepsy, Reflex/physiopathology , Seizures/physiopathology , Animals , Arrhythmias, Cardiac/complications , Blood Pressure/physiology , Electrocardiography/methods , Epilepsy, Reflex/complications , Heart Rate/physiology , Male , Rats , Rats, Wistar , Seizures/complications
8.
Int J Cardiol ; 167(6): 3011-20, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23031286

ABSTRACT

BACKGROUND: Chagas' disease is one of the leading causes of heart failure in Latin American countries. Despite its great social impact, there is no direct evidence in the literature explaining the development of heart failure in Chagas' disease. Therefore, the main objective of the study was to investigate the development of the Chagas' disease towards its chronic phase and correlate with modifications in the cellular electrophysiological characteristics of the infected heart. METHODS AND RESULTS: Using a murine model of Chagas' disease, we confirmed and extended previous findings of altered electrocardiogram and echocardiogram in this cardiomyopathy. The observed changes in the electrocardiogram were correlated with the prolonged action potential and reduced transient outward potassium current density. Reduced heart function was associated with remodeling of intracellular calcium handling, altered extracellular matrix content, and to a set of proteins involved in the control of cellular contractility in ventricular myocytes. Furthermore, disruption of calcium homeostasis was partially due to activation of the PI3Kinase/nitric oxide signaling pathway. Finally, we propose a causal link between the inflammatory mediators and heart remodeling during chagasic cardiomyopathy. CONCLUSION: Altogether our results demonstrate that heart failure in Chagas' disease may occur due to electrical and mechanical remodeling of cardiac myocytes, and suggest that AKT/PI3K/NO axis could be an important pharmacological target to improve the disease outcome.


Subject(s)
Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Nitric Oxide/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Cells, Cultured , Chagas Cardiomyopathy/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/parasitology , Trypanosoma cruzi/physiology
9.
PLoS One ; 7(6): e39997, 2012.
Article in English | MEDLINE | ID: mdl-22768193

ABSTRACT

Autonomic dysfunction is observed in many cardiovascular diseases and contributes to cardiac remodeling and heart disease. We previously reported that a decrease in the expression levels of the vesicular acetylcholine transporter (VAChT) in genetically-modified homozygous mice (VAChT KD(HOM)) leads to decreased cholinergic tone, autonomic imbalance and a phenotype resembling cardiac dysfunction. In order to further understand the molecular changes resulting from chronic long-term decrease in parasympathetic tone, we undertook a transcriptome-based, microarray-driven approach to analyze gene expression changes in ventricular tissue from VAChT KD(HOM) mice. We demonstrate that a decrease in cholinergic tone is associated with alterations in gene expression in mutant hearts, which might contribute to increased ROS levels observed in these cardiomyocytes. In contrast, in another model of cardiac remodeling and autonomic imbalance, induced through chronic isoproterenol treatment to increase sympathetic drive, these genes did not appear to be altered in a pattern similar to that observed in VAChT KD(HOM) hearts. These data suggest the importance of maintaining a fine balance between the two branches of the autonomic nervous system and the significance of absolute levels of cholinergic tone in proper cardiac function.


Subject(s)
Cholinergic Agents/metabolism , Gene Expression Profiling , Heart/physiopathology , Myocardium/metabolism , Myocardium/pathology , Synaptic Transmission , Animals , Disease Models, Animal , Gene Expression Regulation , Gene Knockdown Techniques , Homozygote , Isoproterenol , Lipids/biosynthesis , Mice , Mitochondria/metabolism , Myocardium/enzymology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Purine-Nucleoside Phosphorylase/metabolism , Reproducibility of Results , Superoxides/metabolism , Transcription, Genetic , Up-Regulation/genetics , Vesicular Acetylcholine Transport Proteins/genetics
10.
Int J Hypertens ; 2012: 493129, 2012.
Article in English | MEDLINE | ID: mdl-22518286

ABSTRACT

The Renin-Angiotensin System (RAS) acts at multiple targets and has its synthesis machinery present in different tissues, including the heart. Actually, it is well known that besides Ang II, the RAS has other active peptides. Of particular interest is the heptapeptide Ang-(1-7) that has been shown to exert cardioprotective effects. In this way, great compilations about Ang-(1-7) actions in the heart have been presented in the literature. However, much less information is available concerning the Ang-(1-7) actions directly in cardiomyocytes. In this paper, we show the actual knowledge about Ang-(1-7)-mediated signaling in cardiac cells more specifically we provide a brief overview of ACE2/Ang-(1-7)/Mas axis; and highlight the discoveries made in cardiomyocyte physiology through the use of genetic approaches. Finally, we discuss the protective signaling induced by Ang-(1-7) in cardiomyocytes and point molecular determinants of these effects.

11.
Toxicon ; 57(2): 217-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21115025

ABSTRACT

In this study, we evaluated the effects of PhKv, a 4584 Da peptide isolated from the spider Phoneutria nigriventer venom, in the isolated rat heart and in isolated ventricular myocytes. Ventricular arrhythmias were induced by occlusion of the left anterior descending coronary artery for 15 min followed by 30 min of reperfusion. Administration of native PhKv (240 nM) 1 min before or after reperfusion markedly reduced the duration of arrhythmias. This effect was blocked by atropine, thereby indicating the participation of muscarinic receptors in the antiarrhythmogenic effect of PhKv. Notably, recombinant PhKv (240 nM) was also efficient to attenuate the arrhythmias (3.8 ± 0.9 vs. 8.0 ± 1.2 arbitrary units in control group). Furthermore, PhKv induced a significant reduction in heart rate. This bradycardia was partially blunted by atropine and potentiated by pyridostigmine. To further evaluate the participation of acetylcholine on the PhKv effects, we examined the release of this neurotransmitter from neuromuscular junctions. It was found that Phkv (200 nM) significantly increased the release of acetylcholine in this preparation. Moreover, PhKv (250 nM) did not cause any significant change in action potential or Ca(2+) transient parameters in isolated cardiomyocytes. Altogether, these findings show an important acetylcholine-mediated antiarrhythmogenic effect of the spider PhKv toxin in isolated hearts.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Neurotoxins/pharmacology , Spider Venoms/chemistry , Spiders/chemistry , Acetylcholine/metabolism , Acetylcholine/physiology , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/isolation & purification , Atropine/pharmacology , Calcium Signaling/drug effects , Cloning, Molecular , Electrophysiology , In Vitro Techniques , Male , Myocytes, Cardiac/drug effects , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Neurotoxins/chemistry , Neurotoxins/genetics , Pyridostigmine Bromide/pharmacology , Rats , Rats, Wistar
12.
Toxicon ; 56(8): 1426-35, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20826175

ABSTRACT

Loxosceles spider bites cause many human injuries worldwide. Injections in mice of whole Loxosceles (L.) intermedia venom or a recombinant toxin (rLiD1) produce systemic symptoms similar to those detected in envenomed humans. This animal model was used to characterize the effects of Loxosceles intermedia venom in cardiac tissues. L. intermedia antigens were detected by ELISA in kidney, heart, lung and liver of experimentally envenomed mice. In addition, rLiD1 binding to cardiomyocytes was demonstrated by immunofluorescence and confocal microscopy. Furthermore, isolated perfused heart preparations and ventricular cardiomyocytes from envenomed mice showed heart function impairment, and a significant increase of I(Ca,L) density and intracellular Ca(2+) transients, respectively. Thus, L. intermedia spider venom, as shown through the use of the recombinant toxin rLiD1, causes cardiotoxic effects and a protein from the sphingomyelinase D family plays a key role in heart dysfunction. Thus, L. intermedia spider venom and the Loxtox rLiD1 play a key role in heart dysfunction.


Subject(s)
Cardiotoxins/toxicity , Heart/drug effects , Myocardium/pathology , Phosphoric Diester Hydrolases/toxicity , Spider Venoms/toxicity , Animals , Antigens/analysis , Calcium/metabolism , Cardiotoxins/immunology , Cardiotoxins/isolation & purification , Cells, Cultured , Creatine Kinase/blood , Creatine Kinase, MB Form/blood , Enzyme-Linked Immunosorbent Assay , Female , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Phosphoric Diester Hydrolases/immunology , Phosphoric Diester Hydrolases/isolation & purification , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/pharmacology , Recombinant Fusion Proteins , Spider Venoms/immunology , Spider Venoms/isolation & purification
13.
Toxicon ; 56(7): 1162-71, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20417225

ABSTRACT

In order to better understand the relationship between the primary structure of TsHpt-I - a bradykinin-potentiating peptide (BPP) isolated from the venom of the yellow scorpion Tityus serrulatus, with a non-canonical Lys residue prior to the conservative Pro-Pro doublet - and its cardiovascular effects, a series of ladder peptides were synthesized using the C-terminal portion of TsHpt-I as a template. All synthetic peptides having the Pro-Pro doublet at their C-terminal were able to potentiate the hypotensive effect of bradykinin. Conversely, only those analogues having Lys residue could induce a transient hypotension when intravenously administrated in male rats, indicating that the positive charge located toward the radical of this amino acid residue is crucial for this cardiovascular effect. Differently from all known BPPs, TsHpt-I acts as an agonist of the B(2) receptor and does not inhibit angiotensin-converting enzyme. The capacity of this peptide to activate this subtype of kinin receptor, releasing NO, was also affected by the absence of Lys' side-chain positive charge. Moreover, this study has demonstrated that the minimization of the primary structure of TsHpt-I does not significantly alter the biological effects of this native peptide, which could be of interest for biotechnological purposes.


Subject(s)
Bradykinin/metabolism , Oligopeptides/chemistry , Receptor, Bradykinin B2/agonists , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Hypotension/chemically induced , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Oligopeptides/physiology , Rats , Rats, Wistar , Sequence Analysis, Protein , Structure-Activity Relationship
14.
Mol Cell Biol ; 30(7): 1746-56, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20123977

ABSTRACT

Overwhelming evidence supports the importance of the sympathetic nervous system in heart failure. In contrast, much less is known about the role of failing cholinergic neurotransmission in cardiac disease. By using a unique genetically modified mouse line with reduced expression of the vesicular acetylcholine transporter (VAChT) and consequently decreased release of acetylcholine, we investigated the consequences of altered cholinergic tone for cardiac function. M-mode echocardiography, hemodynamic experiments, analysis of isolated perfused hearts, and measurements of cardiomyocyte contraction indicated that VAChT mutant mice have decreased left ventricle function associated with altered calcium handling. Gene expression was analyzed by quantitative reverse transcriptase PCR and Western blotting, and the results indicated that VAChT mutant mice have profound cardiac remodeling and reactivation of the fetal gene program. This phenotype was attributable to reduced cholinergic tone, since administration of the cholinesterase inhibitor pyridostigmine for 2 weeks reversed the cardiac phenotype in mutant mice. Our findings provide direct evidence that decreased cholinergic neurotransmission and underlying autonomic imbalance cause plastic alterations that contribute to heart dysfunction.


Subject(s)
Cholinergic Agents/metabolism , Heart Failure/metabolism , Primary Dysautonomias/physiopathology , Synaptic Transmission/physiology , Ventricular Remodeling/physiology , Animals , Calcium/metabolism , Echocardiography , Heart Failure/physiopathology , Heart Rate/physiology , Hemodynamics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Phenotype , Receptors, G-Protein-Coupled/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sympathetic Nervous System/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism
15.
Ther Adv Cardiovasc Dis ; 4(2): 83-96, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20051448

ABSTRACT

OBJECTIVE: It has been shown that Ang-(1-7) has cardioprotective actions. To directly investigate the effects of Ang-(1-7) specifically in the heart, we generated and characterized transgenic (TG) rats which express an Ang-(1-7)-producing fusion protein driven by the alpha-MHC promoter. METHODS AND RESULTS: After microinjection of the transgene into fertilized rat zygotes, we obtained four different transgenic lines. Homozygous animals were analyzed with regard to the expression profile of the transgene by ribonuclease protection assay. Transgene expression was detected mainly in the heart with weak or no expression in other organs. Heterozygous TG(hA-1-7)L7301 rats presented a significant increase in cardiac Ang-(1-7) concentration compared with control rats (17.1+/-2.1 versus 3.9+/-1.4 pg/mg protein in SD rats). Radiotelemetry analysis revealed that TG rats presented no significant changes in blood pressure and heart rate compared with normal rats. Overexpression of Ang-(1-7) in the heart produced slight improvement in resting cardiac function (+ dT/dt: 81530+/-1305.0 versus 77470+/-345.5 g/s bpm in SD rats, p < 0.05), which was in keeping with the enhanced [Ca(2+)] handling observed in cardiomyocytes of TG rats. TG(hA-1-7)L7301 rats also showed a greater capacity to withstand stress since TG rats showed a less pronounced deposition of collagen type III and fibronectin induced by isoproterenol treatment in the subendocardial area than in corresponding controls. In addition, hearts from TG rats showed reduced incidence and duration of reperfusion arrhythmias in comparison with SD rats. CONCLUSION: These results indicate that Ang-(1-7) has blood pressure-independent, antifibrotic effects, acting directly in the heart.


Subject(s)
Angiotensin I/metabolism , Gene Expression Regulation , Heart Ventricles/pathology , Peptide Fragments/metabolism , Angiotensin I/genetics , Animals , Arrhythmias, Cardiac/physiopathology , Blood Pressure/physiology , Calcium/metabolism , Disease Models, Animal , Fibrosis , Heart Rate/physiology , Isoproterenol/toxicity , Male , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Peptide Fragments/genetics , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Telemetry/methods
16.
Cell Calcium ; 47(1): 37-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20018372

ABSTRACT

GPR91 is an orphan G-protein-coupled receptor (GPCR) that has been characterized as a receptor for succinate, a citric acid cycle intermediate, in several tissues. In the heart, the role of succinate is unknown. We now report that rat ventricular cardiomyocytes express GPR91. We found that succinate, through GPR91, increases the amplitude and the rate of decline of global Ca(2+) transient, by increasing the phosphorylation levels of ryanodine receptor and phospholamban, two well known Ca(2+) handling proteins. The effects of succinate on Ca(2+) transient were abolished by pre-treatment with adenylyl cyclase and cAMP-dependent protein kinase (PKA) inhibitors. Direct PKA activation by succinate was further confirmed using a FRET-based A-kinase activity reporter. Additionally, succinate decreases cardiomyocyte viability through a caspase-3 activation pathway, effect also prevented by PKA inhibition. Taken together, these observations show that succinate acts as a signaling molecule in cardiomyocytes, modulating global Ca(2+) transient and cell viability through a PKA-dependent pathway.


Subject(s)
Calcium Signaling/drug effects , Cell Survival/drug effects , Myocytes, Cardiac/metabolism , Receptors, G-Protein-Coupled/metabolism , Succinic Acid/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Microscopy, Confocal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Ryanodine Receptor Calcium Release Channel/metabolism
17.
Hypertension ; 55(1): 153-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19996065

ABSTRACT

The renin-angiotensin (Ang) system plays a pivotal role in the pathogenesis of cardiovascular disease, with Ang II being the major effector of this system. Multiple lines of evidence have shown that Ang-(1-7) exerts cardioprotective effects in the heart by counterregulating Ang II actions. The questions that remain are how and where Ang-(1-7) exerts its effects. By using a combination of molecular biology, confocal microscopy, and a transgenic rat model with increased levels of circulating Ang-(1-7) (TGR[A1-7]3292), we evaluated the signaling pathways involved in Ang-(1-7) cardioprotection against Ang II-induced pathological remodeling in ventricular cardiomyocytes. Rats were infused with Ang II for 2 weeks. We found that ventricular myocytes from TGR(A1-7)3292 rats are protected from Ang II pathological remodeling characterized by Ca(2+) signaling dysfunction, hypertrophic fetal gene expression, glycogen synthase kinase 3beta inactivation, and nuclear factor of activated T-cells nuclear accumulation. Moreover, cardiomyocytes from TGR(A1-7)3292 rats infused with Ang II presented increased expression levels of neuronal NO synthase. To provide a signaling pathway involved in the beneficial effects of Ang-(1-7), we treated neonatal cardiomyocytes with Ang-(1-7) and Ang II for 36 hours. Treatment of cardiomyocytes with Ang-(1-7) prevented Ang II-induced hypertrophy by modulating calcineurin/nuclear factor of activated T-cell signaling cascade. Importantly, antihypertrophic effects of Ang-(1-7) on Ang II-treated cardiomyocytes were prevented by N(G)-nitro-l-arginine methyl ester and 1H-1,2,4oxadiazolo4,2-aquinoxalin-1-one, suggesting that these effects are mediated by NO/cGMP. Taken together, these data reveal a key role for NO/cGMP as a mediator of Ang-(1-7) beneficial effects in cardiac cells.


Subject(s)
Angiotensin I/metabolism , Cyclic GMP/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Peptide Fragments/metabolism , Signal Transduction , Angiotensin I/blood , Angiotensin II/administration & dosage , Angiotensin II/toxicity , Animals , Animals, Newborn , Blood Pressure/drug effects , Blood Pressure/genetics , Blood Pressure/physiology , Calcium/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cell Size/drug effects , Cells, Cultured , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Microscopy, Confocal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NFATC Transcription Factors/metabolism , Peptide Fragments/blood , Protein Transport , Rats , Rats, Sprague-Dawley , Rats, Transgenic
18.
Hypertension ; 52(3): 542-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18695148

ABSTRACT

Recently there has been growing evidence suggesting that beneficial effects of angiotensin-(1-7) [Ang-(1-7)] in the heart are mediated by its receptor Mas. However, the signaling pathways involved in these effects in cardiomyocytes are unknown. Here, we investigated the involvement of the Ang-(1-7)/Mas axis in NO generation and Ca(2+) handling in adult ventricular myocytes using a combination of molecular biology, intracellular Ca(2+) imaging, and confocal microscopy. Acute Ang-(1-7) treatment (10 nmol/L) leads to NO production and activates endothelial NO synthase and Akt in cardiomyocytes. Ang-(1-7)-dependent NO raise was abolished by pretreatment with A-779 (1 micromol/L). To confirm that Ang-(1-7) action is mediated by Mas, we used cardiomyocytes isolated from Mas-deficient mice. In Mas-deficient cardiomyocytes, Ang-(1-7) failed to increase NO levels. Moreover, Mas-ablation was accompanied by significant alterations in the proteins involved in the regulation of endothelial NO synthase activity, indicating that endothelial NO synthase and its binding partners are important effectors of the Mas-mediated pathway in cardiomyocytes. We then investigated the role of the Ang-(1-7)/Mas axis on Ca(2+) signaling. Cardiomyocytes treated with 10 nmol/L of Ang-(1-7) did not show changes in Ca(2+)-transient parameters such as peak Ca(2+) transients and kinetics of decay. Nevertheless, cardiomyocytes from Mas-deficient mice presented reduced peak and slower [Ca(2+)](i) transients when compared with wild-type cardiomyocytes. Lower Ca(2+) ATPase of the sarcoplasmic reticulum expression levels accompanied the reduced Ca(2+) transient in Mas-deficient cardiomyocytes. Therefore, chronic Mas-deficiency leads to impaired Ca(2+) handling in cardiomyocytes. Collectively, these observations reveal a key role for the Ang-(1-7)/Mas axis as a modulator of cardiomyocyte function.


Subject(s)
Angiotensin I/metabolism , Calcium Signaling/physiology , Myocytes, Cardiac/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin I/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Calcium Signaling/drug effects , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Peptide Fragments/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...