Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Hypertension ; 66(2): 389-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26077567

ABSTRACT

Mineralocorticoids have been implicated in the pathogenesis of diastolic heart failure. On the contrary, angiotensin (Ang)-(1-7) has emerged as a potential strategy for treatment of cardiac dysfunction induced by excessive mineralocorticoid receptor activation. A critical question about the cardioprotective effect of Ang-(1-7) in hypertensive models is its dependence on blood pressure (BP) reduction. Here, we addressed this question by investigating the mechanisms involved in Ang-(1-7) cardioprotection against mineralocorticoid receptor activation. Sprague-Dawley (SD) and transgenic (TG) rats that overexpress an Ang-(1-7) producing fusion protein (TG(A1-7)3292) were treated with deoxycorticosterone acetate (DOCA) for 6 weeks. After treatment, SD rats became hypertensive and developed ventricular hypertrophy. These parameters were attenuated in TG-DOCA. SD-DOCA rats developed diastolic dysfunction which was associated at the cellular level with reduced Ca(2+) transient. Oppositely, TG-DOCA myocytes presented enhanced Ca(2+) transient. Moreover, higher extracellular signal-regulated kinase phosphorylation, type 1 phosphatase, and protein kinase Cα levels were found in SD-DOCA cells. In vivo, pressor effects of DOCA can contribute to the diastolic dysfunction, raising the question of whether protection in TG was a consequence of reduced BP. To address this issue, BP in SD-DOCA was kept at TG-DOCA level by giving hydralazine or by reducing the DOCA amount given to rats (Low-DOCA). Under similar BP, diastolic dysfunction and molecular changes were still evident in DOCA-hydralazine and SD-low-DOCA, but not in TG-DOCA. In conclusion, Ang-(1-7) protective signaling against DOCA-induced diastolic dysfunction occurs independently of BP attenuation and is mediated by the activation of pathways involved in Ca(2+) handling, hypertrophy, and survival.


Subject(s)
Angiotensin I/therapeutic use , Blood Pressure/physiology , Desoxycorticosterone Acetate/adverse effects , Heart Failure, Diastolic/chemically induced , Heart Failure, Diastolic/prevention & control , Peptide Fragments/therapeutic use , Angiotensin I/pharmacology , Animals , Blood Pressure/drug effects , Calcium/physiology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Heart Failure, Diastolic/physiopathology , Hydralazine/pharmacology , Hypertension/physiopathology , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Transgenic
2.
J Transl Med ; 13: 76, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25889299

ABSTRACT

BACKGROUND: Activation of protein kinase AKT is required for cardioprotection by ischemic preconditioning, and transgenic overexpression of AKT protects the heart against ischemia. However, it is unknown whether acute pharmacological activation of AKT alone, using a therapeutically relevant strategy, induces cardioprotection. In this study we provide the first evidence to clarify this question. METHODS: We used a recently described specific activator of AKT, the small molecule SC79, to treat rat hearts submitted to ischemia and reperfusion. Initially, isolated rat hearts were perfused with increasing doses of SC79 to verify the magnitude of AKT activation. Low and high doses were determined and used to treat hearts submitted to ischemia (35 minutes) and reperfusion (60 minutes), in a randomized and blinded design. AKT activation was verified by western immunobloting. Metabolic profile was determined by cardiac ATP content and mitochondrial enzyme activity, while cytosolic levels of cytochrome C and caspase-3 activity were used as markers of apoptosis. Ischemic injury was assessed by quantification of infarct size and cardiac release of creatine kinase and lactate dehydrogenase. RESULTS: SC79 activated cardiac AKT within 30 minutes in a dose-dependent fashion. ATP content was largely reduced by ischemia, but was not rescued by SC79. Similarly, mitochondrial enzyme activity was not affected by SC79. SC79 administered before ischemia or at reperfusion did not prevent cytosolic accumulation of cytochrome C and overactivation of caspase-3. Finally, SC79 failed to reduce infarct size or release of cardiac injury biomarkers at reperfusion. CONCLUSION: We conclude that selective AKT activation by the synthetic molecule SC79 does not protect the rat heart against ischemic injury, indicating that acute pharmacological activation of AKT is not sufficient for cardioprotection.


Subject(s)
Enzyme Activators/pharmacology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Proto-Oncogene Proteins c-akt/metabolism , Small Molecule Libraries/pharmacology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Creatine Kinase/metabolism , Enzyme Activation/drug effects , Female , Mitochondria/drug effects , Mitochondria/enzymology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/enzymology , Myocardium/pathology , Rats, Sprague-Dawley
3.
Cells Tissues Organs ; 198(4): 311-7, 2013.
Article in English | MEDLINE | ID: mdl-24247000

ABSTRACT

Protein restriction (PR) is associated with cardiovascular diseases. The purpose of this study was to investigate the effects on single ventricular cardiomyocyte contractile function of a short-term PR after weaning. Male Fischer rats that were 28 days old were randomly divided into a control group (CG, n = 16) and a protein-restricted group (PRG, n = 16). After weaning, CG and PRG animals received isocaloric diets containing 15 and 6% protein, respectively, for 35 days. Biometric parameters were then measured, and the hearts were removed for the analysis of contractile function and calcium transient in isolated cardiomyocytes of the left ventricule (LV), and the quantification of calcium and collagen fibers in LV myocardium. PRG animals had lower body weight (BW) and LV weight (LVW), an increased LVW to BW ratio and a higher proportion of collagen fibers than CG animals. PRG animals exhibited reduced tissue levels of calcium, reduced the length, width and volume of cardiomyocytes and their sarcomere length compared to CG animals. Cardiomyocytes from PRG animals had a lower amplitude of shortening, a slower time to the peak of shortening and a longer time to half-relaxation than those from the CG. Cardiomyocytes from PRG animals also presented a lower peak of calcium transient and a longer calcium transient decay time than CG animals. Taken together, the results indicate that short-term PR after weaning induces a marked structural remodeling of the myocardium parenchyma and stroma that coexists with contractile dysfunctions in single LV cardiomyocytes of rats, which is probably associated with pathological changes of the intracellular calcium kinetics, rather than inadequate available amounts of this mineral in cardiac tissue.


Subject(s)
Calcium/metabolism , Cardiovascular Diseases/etiology , Diet, Protein-Restricted/adverse effects , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Protein-Energy Malnutrition/etiology , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Heart Ventricles/cytology , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein-Energy Malnutrition/metabolism , Protein-Energy Malnutrition/physiopathology , Random Allocation , Rats , Rats, Inbred F344 , Weaning
4.
Hypertension ; 61(2): 425-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23232646

ABSTRACT

High serum levels of aldosterone have been linked to the development of cardiac disease. In contrast, angiotensin (Ang)-(1-7) was extensively shown to possess cardioprotective effects, including the attenuation of cardiac dysfunction induced by excessive mineralocorticoid activation in vivo, suggesting possible interactions between these 2 molecules. Here, we investigated whether there is cross-talk between aldosterone and Ang-(1-7) and its functional consequences for calcium (Ca(2+)) signaling in ventricular myocytes. Short-term effects of aldosterone on Ca(2+) transient were assessed in Fluo-4/AM-loaded myocytes. Confocal images showed that Ang-(1-7) had no effect on Ca(2+) transient parameters, whereas aldosterone increased the magnitude of the Ca(2+) transient. Quite unexpectedly, addition of Ang-(1-7) to aldosterone-treated myocytes further enhanced the amplitude of the Ca(2+) transient suggesting a synergistic effect of these molecules. Aldosterone action on Ca(2+) transient amplitude was mediated by protein kinase A, and was related to an increase in Ca(2+) current (I(Ca)) density. Both changes were not altered by Ang-(1-7). When cardiomyocytes were exposed to aldosterone, increased Ca(2+) spark rate was measured. Ang-(1-7) prevented this change. In addition, a NO synthase inhibitor restored the effect of aldosterone on Ca(2+) spark rate in Ang-(1-7)-treated myocytes and attenuated the synergistic effect of these 2 molecules on Ca(2+) transient. These results indicate that NO plays an important role in this cross-talk. Our results bring new perspectives in the understanding of how 2 prominent molecules with supposedly antagonist cardiac actions cross-talk to synergistically amplify Ca(2+) signals in cardiomyocytes.


Subject(s)
Aldosterone/metabolism , Angiotensin I/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Myocytes, Cardiac/metabolism , Peptide Fragments/metabolism , Aldosterone/pharmacology , Angiotensin I/pharmacology , Animals , Calcium Signaling/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley
5.
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
6.
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
7.
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
8.
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
...