Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
J Appl Physiol (1985) ; 118(9): 1154-60, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25749446

ABSTRACT

We examined the effect of specific and local silencing of sodium/hydrogen exchanger isoform 1 (NHE1) with a small hairpin RNA delivered by lentivirus (L-shNHE1) in the cardiac left ventricle (LV) wall of spontaneously hypertensive rats, to reduce cardiac hypertrophy. Thirty days after the lentivirus was injected, NHE1 protein expression was reduced 53.3 ± 3% in the LV of the L-shNHE1 compared with the control group injected with L-shSCR (NHE1 scrambled sequence), without affecting its expression in other organs, such as liver and lung. Hypertrophic parameters as LV weight-to-body weight and LV weight-to-tibia length ratio were significantly reduced in animals injected with L-shNHE1 (2.32 ± 0.5 and 19.30 ± 0.42 mg/mm, respectively) compared with L-shSCR-injected rats (2.68 ± 0.06 and 21.53 ± 0.64 mg/mm, respectively). Histochemical analysis demonstrated a reduction of cardiomyocytes cross-sectional area in animals treated with L-shNHE1 compared with L-shSCR (309,81 ± 20,86 vs. 424,52 ± 21 µm(2), P < 0.05). Echocardiography at the beginning and at the end of the treatment showed that shNHE1 expression for 30 days induced 9% reduction of LV mass. Also, animals treated with L-shNHE1 exhibited a reduced LV wall thickness without changing LV diastolic dimension and arterial pressure, indicating an increased parietal stress. In addition, midwall shortening was not modified, despite the increased wall tension, suggesting an improvement of cardiac function. Chronic shNHE1 expression in the heart emerges as a possible methodology to reduce pathological cardiac hypertrophy, avoiding potentially undesired effects caused from a body-wide inhibition of NHE1.


Subject(s)
Cardiomegaly/genetics , Cardiomegaly/pathology , Gene Silencing/physiology , Myocardium/metabolism , Sodium-Hydrogen Exchangers/genetics , Animals , Arterial Pressure/genetics , Arterial Pressure/physiology , Cell Line , Diastole/genetics , Diastole/physiology , Echocardiography/methods , HEK293 Cells , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Male , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , Rats , Rats, Inbred SHR/genetics , Rats, Inbred SHR/physiology , Sodium-Hydrogen Exchanger 1
2.
Hypertension ; 63(1): 112-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24126173

ABSTRACT

Myocardial stretch triggers an angiotensin II-dependent autocrine/paracrine loop of intracellular signals, leading to reactive oxygen species-mediated activation of redox-sensitive kinases. Based on pharmacological strategies, we previously proposed that mineralocorticoid receptor (MR) is necessary for this stretch-triggered mechanism. Now, we aimed to test the role of MR after stretch by using a molecular approach to avoid secondary effects of pharmacological MR blockers. Small hairpin interference RNA capable of specifically knocking down the MR was incorporated into a lentiviral vector (l-shMR) and injected into the left ventricular wall of Wistar rats. The same vector but expressing a nonsilencing sequence (scramble) was used as control. Lentivirus propagation through the left ventricle was evidenced by confocal microscopy. Myocardial MR expression, stretch-triggered activation of redox-sensitive kinases (ERK1/2-p90(RSK)), the consequent Na(+)/H(+) exchanger-mediated changes in pHi (HEPES-buffer), and its mechanical counterpart, the slow force response, were evaluated. Furthermore, reactive oxygen species production in response to a low concentration of angiotensin II (1.0 nmol/L) or an equipotent concentration of epidermal growth factor (0.1 µg/mL) was compared in myocardial tissue slices from both groups. Compared with scramble, animals transduced with l-shMR showed (1) reduced cardiac MR expression, (2) cancellation of angiotensin II-induced reactive oxygen species production but preservation of epidermal growth factor-induced reactive oxygen species production, (3) cancellation of stretch-triggered increase in ERK1/2-p90(RSK) phosphorylation, (4) lack of stretch-induced Na(+)/H(+) exchanger activation, and (5) abolishment of the slow force response. Our results provide strong evidence that MR activation occurs after myocardial stretch and is a key factor to promote redox-sensitive kinase activation and their downstream consequences.


Subject(s)
Myocardium/metabolism , Receptors, Mineralocorticoid/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Genetic Vectors , Heart/physiology , Lentivirus , Male , Mitochondria/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sodium-Hydrogen Exchanger 1
3.
Pflugers Arch ; 466(9): 1819-30, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24327206

ABSTRACT

Emerging evidence supports a key role for endothelin-1 (ET-1) and the transactivation of the epidermal growth factor receptor (EGFR) in angiotensin II (Ang II) action. We aim to determine the potential role played by endogenous ET-1, EGFR transactivation and redox-dependent sodium hydrogen exchanger-1 (NHE-1) activation in the hypertrophic response to Ang II of cardiac myocytes. Electrically paced adult cat cardiomyocytes were placed in culture and stimulated with 1 nmol l(-1) Ang II or 5 nmol l(-1) ET-1. Ang II increased ~45 % cell surface area (CSA) and ~37 % [(3)H]-phenylalanine incorporation, effects that were blocked not only by losartan (Los) but also by BQ123 (AT1 and ETA receptor antagonists, respectively). Moreover, Ang II significantly increased ET-1 messenger RNA (mRNA) expression. ET-1 similarly increased myocyte CSA and protein synthesis, actions prevented by the reactive oxygen species scavenger MPG or the NHE-1 inhibitor cariporide (carip). ET-1 increased the phosphorylation of the redox-sensitive ERK1/2-p90(RSK) kinases, main activators of the NHE-1. This effect was prevented by MPG and the antagonist of EGFR, AG1478. Ang II, ET-1 and EGF increased myocardial superoxide production (187 ± 9 %, 149 ± 8 % and 163.7 ± 6 % of control, respectively) and AG1478 inhibited these effects. Interestingly, Los inhibited only Ang II whilst BQ123 cancelled both Ang II and ET-1 actions, supporting the sequential and unidirectional activation of AT1, ETA and EGFR. Based on the present evidence, we propose that endogenous ET-1 mediates the hypertrophic response to Ang II by a mechanism that involves EGFR transactivation and redox-dependent activation of the ERK1/2-p90(RSK) and NHE-1 in adult cardiomyocytes.


Subject(s)
Angiotensin II/metabolism , Cardiomegaly/metabolism , Endothelin-1/metabolism , ErbB Receptors/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cats , Disease Models, Animal , Electric Stimulation , Hypertrophy/metabolism , Myocytes, Cardiac/pathology , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Sodium-Hydrogen Exchangers/metabolism , Transcriptional Activation
4.
Curr Cardiol Rev ; 9(3): 230-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23909633

ABSTRACT

The stretch of cardiac muscle increases developed force in two phases. The first phase, which occurs rapidly, constitutes the well-known Frank-Starling mechanism and it is generally attributed to enhanced myofilament responsiveness to Ca(2+). The second phase or slow force response (SFR) occurs gradually and is due to an increase in the calcium transient amplitude as a result of a stretch-triggered autocrine/paracrine mechanism. We previously showed that Ca(2+) entry through reverse Na(+)/Ca(2+) exchange underlies the SFR, as the final step of an autocrine/paracrine cascade involving release of angiotensin II/endothelin, and a Na(+)/H(+) exchanger (NHE-1) activation-mediated rise in Na+. In the present review we mainly focus on our three latest contributions to the understanding of this signalling pathway triggered by myocardial stretch: 1) The finding that an increased production of reactive oxygen species (ROS) from mitochondrial origin is critical in the activation of the NHE-1 and therefore in the genesis of the SFR; 2) the demonstration of a key role played by the transactivation of the epidermal growth factor receptor; and 3) the involvement of mineralocorticoid receptors (MR) activation in the stretch-triggered cascade leading to the SFR. Among these novel contributions, the critical role played by the MR is perhaps the most important one. This finding may conceivably provide a mechanistic explanation to the recently discovered strikingly beneficial effects of MR antagonism in humans with cardiac hypertrophy and failure.


Subject(s)
Autocrine Communication/physiology , Myocardial Contraction/physiology , Myocardium/metabolism , Paracrine Communication/physiology , Receptors, Mineralocorticoid/metabolism , Angiotensin II/metabolism , Angiotensin II/physiology , Calcium/metabolism , Cardiomegaly/metabolism , Cardiovascular Diseases/physiopathology , Heart Failure/metabolism , Heart Failure/prevention & control , Humans , Reactive Oxygen Species/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchangers/metabolism , Thyroid Hormones/metabolism
5.
Am J Physiol Heart Circ Physiol ; 304(2): H175-82, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23161880

ABSTRACT

Myocardial stretch elicits a rapid increase in developed force, which is mainly caused by an increase in myofilament calcium sensitivity (Frank-Starling mechanism). Over the ensuing 10-15 min, a second gradual increase in force takes place. This slow force response to stretch is known to be the result of an increase in the calcium transient amplitude and constitutes the in vitro equivalent of the Anrep effect described 100 years ago in the intact heart. In the present review, we will update and discuss what is known about the Anrep effect as the mechanical counterpart of autocrine/paracrine mechanisms involved in its genesis. The chain of events triggered by myocardial stretch comprises 1) release of angiotensin II, 2) release of endothelin, 3) activation of the mineralocorticoid receptor, 4) transactivation of the epidermal growth factor receptor, 5) increased formation of mitochondria reactive oxygen species, 6) activation of redox-sensitive kinases upstream myocardial Na(+)/H(+) exchanger (NHE1), 7) NHE1 activation, 8) increase in intracellular Na(+) concentration, and 9) increase in Ca(2+) transient amplitude through the Na(+)/Ca(2+) exchanger. We will present the experimental evidence supporting each of the signaling steps leading to the Anrep effect and its blunting by silencing NHE1 expression with a specific small hairpin interference RNA injected into the ventricular wall.


Subject(s)
Excitation Contraction Coupling , Mechanoreceptors/metabolism , Mechanotransduction, Cellular , Models, Cardiovascular , Muscle Strength , Myocardial Contraction , Myocardium/metabolism , Reflex, Stretch , Animals , Autocrine Communication , Calcium/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Paracrine Communication , RNA Interference , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
6.
J Physiol ; 589(Pt 24): 6051-61, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22174146

ABSTRACT

The increase in myocardial reactive oxygen species after epidermal growth factor receptor transactivation is a crucial step in the autocrine/paracrine angiotensin II/endothelin receptor activation leading to the slow force response to stretch (SFR). Since experimental evidence suggests a link between angiotensin II or its AT1 receptor and the mineralocorticoid receptor (MR), and MR transactivates the epidermal growth factor receptor, we thought to determine whether MR activation participates in the SFR development in rat myocardium. We show here that MR activation is necessary to promote reactive oxygen species formation by a physiological concentration of angiotensin II (1 nmol l(-1)), since an increase in superoxide anion formation of ~50% of basal was suppressed by blocking MR with spironolactone or eplerenone. This effect was also suppressed by blocking AT1, endothelin (type A) or epidermal growth factor receptors, by inhibiting NADPH oxydase or by targeting mitochondria, and was unaffected by glucocorticoid receptor inhibition. All interventions except AT1 receptor blockade blunted the increase in superoxide anion promoted by an equipotent dose of endothelin-1 (1 nmol l(-1)) confirming that endothelin receptors activation is downstream of AT1. Similarly, an increase in superoxide anion promoted by an equipotent dose of aldosterone (10 nmol l(-1)) was blocked by spironolactone or eplerenone, by preventing epidermal growth factor receptor transactivation, but not by inhibiting glucocorticoid receptors or protein synthesis, suggesting non-genomic MR effects. Combination of aldosterone plus endothelin-1 did not increase superoxide anion formation more than each agonist separately. We found that aldosterone increased phosphorylation of the redox-sensitive kinases ERK1/2-p90RSK and the NHE-1, effects that were eliminated by eplerenone or by preventing epidermal growth factor receptor transactivation. Finally, we provide evidence that the SFR is suppressed by MR blockade, by preventing epidermal growth factor receptor transactivation or by scavenging reactive oxygen species, but it is unaffected by glucocorticoid receptor blockade or protein synthesis inhibition. Our results suggest that MR activation is a necessary step in the stretch-triggered reactive oxygen species-mediated activation of redox-sensitive kinases upstream NHE-1.


Subject(s)
Heart/physiology , Muscle, Smooth/physiology , Myocardial Contraction/physiology , Receptors, Mineralocorticoid/physiology , Aldosterone/pharmacology , Angiotensin II/metabolism , Animals , Endothelin-1/pharmacology , ErbB Receptors/metabolism , In Vitro Techniques , Male , Mitochondria, Heart/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Papillary Muscles/physiology , Rats , Rats, Wistar , Receptors, Endothelin/metabolism , Receptors, Mineralocorticoid/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Sodium-Hydrogen Exchangers/metabolism , Stress, Mechanical , Superoxides/metabolism
7.
Hypertension ; 58(5): 912-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22016493

ABSTRACT

The use of antagonists of the mineralocorticoid receptor in the treatment of myocardial hypertrophy and heart failure has gained increasing importance in the last years. The cardiac Na(+)/H(+) exchanger (NHE-1) upregulation induced by aldosterone could account for the genesis of these pathologies. We tested whether aldosterone-induced NHE-1 stimulation involves the transactivation of the epidermal growth factor receptor (EGFR). Rat ventricular myocytes were used to measure intracellular pH with epifluorescence. Aldosterone enhanced the NHE-1 activity. This effect was canceled by spironolactone or eplerenone (mineralocorticoid receptor antagonists), but not by mifepristone (glucocorticoid receptor antagonist) or cycloheximide (protein synthesis inhibitor), indicating that the mechanism is mediated by the mineralocorticoid receptor triggering nongenomic pathways. Aldosterone-induced NHE-1 stimulation was abolished by the EGFR kinase inhibitor AG1478, suggesting that is mediated by transactivation of EGFR. The increase in the phosphorylation level of the kinase p90(RSK) and NHE-1 serine703 induced by aldosterone was also blocked by AG1478. Exogenous epidermal growth factor mimicked the effects of aldosterone on NHE-1 activity. Epidermal growth factor was also able to increase reactive oxygen species production, and the epidermal growth factor-induced activation of the NHE-1 was abrogated by the reactive oxygen species scavenger N-2-mercaptopropionyl glycine, indicating that reactive oxygen species are participating as signaling molecules in this mechanism. Aldosterone enhances the NHE-1 activity via transactivation of the EGFR, formation of reactive oxygen species, and phosphorylation of the exchanger. These results call attention to the consideration of the EGFR as a new potential therapeutic target of the cardiovascular pathologies involving the participation of aldosterone.


Subject(s)
Aldosterone/pharmacology , ErbB Receptors/metabolism , Myocytes, Cardiac/drug effects , Sodium-Hydrogen Exchangers/drug effects , Animals , Cells, Cultured , ErbB Receptors/genetics , Models, Animal , Myocytes, Cardiac/metabolism , Phosphorylation/physiology , Random Allocation , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sensitivity and Specificity , Signal Transduction/drug effects , Sodium-Hydrogen Exchangers/metabolism , Superoxides/metabolism , Transcriptional Activation
8.
Appl Physiol Nutr Metab ; 36(5): 768-72, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21999300

ABSTRACT

A reduction in the risk of coronary heart disease has been associated to moderate red wine consumption. We tested whether a nonalcoholic red wine extract would open mitochondrial K(ATP) channels in guinea pig myocytes. The opening of mitochondrial K(ATP) channels was assessed by endogenous flavoprotein fluorescence. Red wine extract (100 µg·mL(-1)) increased flavoprotein oxidation (10.9% ± 1.2%, n = 20). This effect was prevented by the mitochondrial K(ATP) channel blocker, 5-hydroxydecanoate (500 µmol·L(-1); 0.3% ± 1.1%, n = 13), confirming the hypothesis that red wine extract opens mitochondrial K(ATP) channels.


Subject(s)
Coronary Disease/prevention & control , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels/agonists , Wine/analysis , Animals , Cells, Cultured , Decanoic Acids/pharmacology , Flavoproteins/metabolism , Guinea Pigs , Hydroxy Acids/pharmacology , Kinetics , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Plant Extracts/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Wine/adverse effects
9.
Pflugers Arch ; 462(5): 733-43, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21870055

ABSTRACT

Growing in vitro evidence suggests NHE-1, a known target for reactive oxygen species (ROS), as a key mediator in cardiac hypertrophy (CH). Moreover, NHE-1 inhibition was shown effective in preventing CH and failure; so has been the case for AT1 receptor (AT1R) blockers. Previous experiments indicate that myocardial stretch promotes angiotensin II release and post-translational NHE-1 activation; however, in vivo data supporting this mechanism and its long-term consequences are scanty. In this work, we thought of providing in vivo evidence linking AT1R with ROS and NHE-1 activation in mediating CH. CH was induced in mice by TAC. A group of animals was treated with the AT1R blocker losartan. Cardiac contractility was assessed by echocardiography and pressure-volume loop hemodynamics. After 7 weeks, TAC increased left ventricular (LV) mass by ~45% vs. sham and deteriorated LV systolic function. CH was accompanied by activation of the redox-sensitive kinase p90(RSK) with the consequent increase in NHE-1 phosphorylation. Losartan prevented p90(RSK) and NHE-1 phosphorylation, ameliorated CH and restored cardiac function despite decreased LV wall thickness and similar LV systolic pressures and diastolic dimensions (increased LV wall stress). In conclusion, AT1R blockade prevented excessive oxidative stress, p90(RSK) and NHE-1 phosphorylation, and decreased CH independently of hemodynamic changes. In addition, cardiac performance improved despite a higher work load.


Subject(s)
Cardiomegaly/physiopathology , Reactive Oxygen Species/pharmacology , Sodium-Hydrogen Exchangers/metabolism , Animals , Aorta/surgery , Ligation , Lipid Peroxidation , Losartan/pharmacology , Male , Mice , Mice, Inbred BALB C , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
10.
J Appl Physiol (1985) ; 111(3): 874-80, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21659487

ABSTRACT

Myocardial stretch induces a biphasic force response: a first abrupt increase followed by a slow force response (SFR), believed to be the in vitro manifestation of the Anrep effect. The SFR is due to an increase in Ca²âº transient of unclear mechanism. We proposed that Na⁺/H⁺ exchanger (NHE-1) activation is a key factor in determining the contractile response, but recent reports challenged our findings. We aimed to specifically test the role of the NHE-1 in the SFR. To this purpose small hairpin interference RNA capable of mediating specific NHE-1 knockdown was incorporated into a lentiviral vector (l-shNHE1) and injected into the left ventricular wall of Wistar rats. Injection of a lentiviral vector expressing a nonsilencing sequence (scramble) served as control. Myocardial NHE-1 protein expression and function (the latter evaluated by the recovery of pH(i) after an acidic load and the SFR) were evaluated. Animals transduced with l-shNHE1 showed reduced NHE-1 expression (45 ± 8% of controls; P < 0.05), and the presence of the lentivirus in the left ventricular myocardium, far from the site of injection, was evidenced by confocal microscopy. These findings correlated with depressed basal pH(i) recovery after acidosis [(max)dpH(i)/dt 0.055 ± 0.008 (scramble) vs. 0.009 ± 0.004 (l-shNHE1) pH units/min, P < 0.05], leftward shift of the relationship between J(H⁺) (H⁺ efflux corrected by the intrinsic buffer capacity), and abolishment of SFR (124 ± 2 vs. 101 ± 2% of rapid phase; P < 0.05) despite preserved ERK1/2 phosphorylation [247 ± 12 (stretch) and 263 ± 23 (stretch l-shNHE1) % of control; P < 0.05 vs. nonstretched control], well-known NHE-1 activators. Our results provide strong evidence to propose NHE-1 activation as key factor in determining the SFR to stretch.


Subject(s)
Mechanoreceptors/metabolism , Muscle Strength , Myocardial Contraction , Papillary Muscles/metabolism , RNA Interference , Sodium-Hydrogen Exchangers/metabolism , Acidosis/metabolism , Acidosis/physiopathology , Animals , Down-Regulation , Hydrogen-Ion Concentration , Injections, Intramuscular , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Papillary Muscles/physiopathology , Phosphorylation , RNA, Small Interfering/administration & dosage , Rats , Rats, Wistar , Signal Transduction , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Time Factors
11.
J Appl Physiol (1985) ; 111(2): 566-72, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21596922

ABSTRACT

Cardiac Na(+)/H(+) exchanger (NHE1) hyperactivity is a central factor in cardiac remodeling following hypertension, myocardial infarction, ischemia-reperfusion injury, and heart failure. Treatment of these pathologies by inhibiting NHE1 is challenging because specific drugs that have been beneficial in experimental models were associated with undesired side effects in clinical practice. In the present work, small interference RNA (siRNA) produced in vitro to specifically silence NHE1 (siRNA(NHE1)) was injected once in vivo into the apex of the left ventricular wall of mouse myocardium. After 48 h, left ventricular NHE1 protein expression was reduced in siRNA(NHE1)-injected mice compared with scrambled siRNA by 33.2 ± 3.4% (n = 5; P < 0.05). Similarly, NHE1 mRNA levels were reduced by 20 ± 2.0% (n = 4). At 72 h, siRNA(NHE1) spreading was evident from the decrease in NHE1 expression in three portions of the myocardium (apex, medium, base). NHE1 function was assessed based on maximal velocity of intracellular pH (pH(i)) recovery (dpH(i)/dt) after an ammonium prepulse-induced acidic load. Maximal dpH(i)/dt was reduced to 14% in siRNA(NHE1)-isolated left ventricular papillary muscles compared with scrambled siRNA. In conclusion, only one injection of naked siRNA(NHE1) successfully reduced NHE1 expression and activity in the left ventricle. As has been previously suggested, extensive NHE1 expression reduction may indicate myocardial spread of siRNA molecules from the injection site through gap junctions, providing a valid technique not only for further research into NHE1 function, but also for consideration as a potential therapeutic strategy.


Subject(s)
Cation Transport Proteins/genetics , Gene Silencing/drug effects , Heart/drug effects , Heart/physiology , RNA, Small Interfering/pharmacology , Sodium-Hydrogen Exchangers/genetics , Animals , Buffers , Cation Transport Proteins/drug effects , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Immunochemistry , Injections , Male , Mice , Mice, Inbred BALB C , Myocardium/metabolism , Papillary Muscles/drug effects , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/drug effects , Tissue Culture Techniques , Ventricular Function, Left/genetics , Ventricular Function, Left/physiology
12.
Cell Physiol Biochem ; 27(1): 13-22, 2011.
Article in English | MEDLINE | ID: mdl-21325817

ABSTRACT

BACKGROUND/AIMS: Flow restoration to ischemic myocardium reduces infarct size (IS), but it also promotes reperfusion injury. A burst of reactive oxygen species (ROS) and/or NHE-1 reactivation were proposed to explain this injury. Our study was aimed to shed light on this unresolved issue. METHODS: Regional infarction (40 min-ischemia/2 hs-reperfusion) was induced in isolated and perfused rat hearts. Maximal doses of N-(2-mercaptopropionyl)-glycine (MPG 2mmol/L, ROS scavenger), cariporide (10µmol/L, NHE-1 inhibitor), or sildenafil (1µmol/L, phosphodiesterase5A inhibitor) were applied at reperfusion onset. Their effects on IS, myocardial concentration of thiobarbituric acid reactive substances (TBARS), ERK1/2, p90(RSK), and NHE-1 phosphorylation were analyzed. RESULTS: All treatments decreased IS ∼ 50% vs. control. No further protection was obtained by combining cariporide or MPG with sildenafil. Myocardial TBARS increased after infarction and were decreased by MPG or cariporide, but unaffected by sildenafil. In line with the fact that ROS induce MAPK-mediated NHE-1 activation, myocardial infarction increased ERK1/2, p90(RSK), and NHE-1 phosphorylation. MPG and cariporide cancelled these effects. Sildenafil did not reduce the phosphorylated ERK1/2-p90(RSK) levels but blunted NHE-1 phosphorylation suggesting a direct dephosphorylating action. CONCLUSIONS: 1) Reperfusion injury would result from ROS-triggered MAPK-mediated NHE-1 phosphorylation (and reactivation) during reperfusion; 2) sildenafil protects the myocardium by favouring NHE-1 dephosphorylation and bypassing ROS generation.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Reactive Oxygen Species/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Glycine/analogs & derivatives , Glycine/therapeutic use , Guanidines/therapeutic use , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Reperfusion Injury/drug therapy , Phosphorylation , Piperazines/therapeutic use , Purines/therapeutic use , Rats , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sildenafil Citrate , Sulfhydryl Compounds/therapeutic use , Sulfones/therapeutic use , Thiobarbituric Acid Reactive Substances/analysis , Vasodilator Agents/therapeutic use
13.
Am J Physiol Heart Circ Physiol ; 300(4): H1237-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21297023

ABSTRACT

Inhibition of Na(+)/H(+) exchanger 1 (NHE1) reduces cardiac ischemia-reperfusion (I/R) injury and also cardiac hypertrophy and failure. Although the mechanisms underlying these NHE1-mediated effects suggest delay of mitochondrial permeability transition pore (MPTP) opening, and reduction of mitochondrial-derived superoxide production, the possibility of NHE1 blockade targeting mitochondria has been incompletely explored. A short-hairpin RNA sequence mediating specific knock down of NHE1 expression was incorporated into a lentiviral vector (shRNA-NHE1) and transduced in the rat myocardium. NHE1 expression of mitochondrial lysates revealed that shRNA-NHE1 transductions reduced mitochondrial NHE1 (mNHE1) by ∼60%, supporting the expression of NHE1 in mitochondria membranes. Electron microscopy studies corroborate the presence of NHE1 in heart mitochondria. Immunostaining of rat cardiomyocytes also suggests colocalization of NHE1 with the mitochondrial marker cytochrome c oxidase. To examine the functional role of mNHE1, mitochondrial suspensions were exposed to increasing concentrations of CaCl(2) to induce MPTP opening and consequently mitochondrial swelling. shRNA-NHE1 transduction reduced CaCl(2)-induced mitochondrial swelling by 64 ± 4%. Whereas the NHE1 inhibitor HOE-642 (10 µM) decreased mitochondrial Ca(2+)-induced swelling in rats transduced with nonsilencing RNAi (37 ± 6%), no additional HOE-642 effects were detected in mitochondria from rats transduced with shRNA-NHE1. We have characterized the expression and function of NHE1 in rat heart mitochondria. Because mitochondria from rats injected with shRNA-NHE1 present a high threshold for MPTP formation, the beneficial effects of NHE1 inhibition in I/R resulting from mitochondrial targeting should be considered.


Subject(s)
Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Sodium-Hydrogen Exchangers/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Calcium/pharmacology , Electron Transport Complex IV/ultrastructure , Gene Expression/drug effects , Gene Silencing , Guanidines/pharmacology , Male , Mitochondria, Heart/ultrastructure , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , Sulfones/pharmacology
14.
Pflugers Arch ; 462(1): 29-38, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21301862

ABSTRACT

Myocardial strain triggers an autocrine/paracrine mechanism known to participate in myocardial hypertrophy development. After the onset of stretch, there is a rapid augmentation in developed tension due to an increase in myofilament calcium sensitivity (the Frank Starling mechanism) followed by a gradual increase in tension over the next 10-15 min. This second phase is called the slow force response (SFR) to stretch and is known to be the result of an increase in calcium transient amplitude. In the present review, we will discuss what is known thus far about the SFR, which is the in vitro equivalent of the Anrep effect and the mechanical counterpart of the autocrine/paracrine mechanism elicited by myocardial stretch. The chain of events triggered by myocardial stretch comprises: (1) release of angiotensin II, (2) release/formation of endothelin, (3) NADPH oxidase activation and transactivation of the EGFR, (4) mitochondrial reactive oxygen species production, (5) activation of redox-sensitive kinases, (6) NHE-1 hyperactivity, (7) increase in intracellular Na(+) concentration, and (8) increase in Ca(2+) transient amplitude through the Na(+)/Ca(2+) exchanger. The evidence for each step of the intracellular signaling pathway leading to the development of SFR and their relationship with the mechanisms proposed for cardiac hypertrophy development will be analyzed.


Subject(s)
Autocrine Communication , Cardiomegaly/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , Paracrine Communication , Stress, Mechanical , Angiotensin II/metabolism , Animals , Calcium/metabolism , Endothelins/metabolism , ErbB Receptors/metabolism , Humans , Myocardium/cytology , Myocardium/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchangers/metabolism
15.
Cell Physiol Biochem ; 26(4-5): 531-40, 2010.
Article in English | MEDLINE | ID: mdl-21063091

ABSTRACT

BACKGROUND/AIMS: This study aimed to identify the signaling pathway for the proposed link between phosphodiesterase-5A (PDE5A) inhibition and decreased cardiac Na(+)/H(+) exchanger (NHE-1) activity. METHODS: NHE-1 activity was assessed in rat isolated papillary muscles by the Na(+)-dependent initial pH(i) recovery from a sustained acidosis (ammonium prepulse). ERK1/2, p90RSK and NHE-1 phosphorylation state during acidosis was determined. RESULTS: PDE5A inhibition (1 µmol/L sildenafil, SIL) did not modify basal pH(i) but significantly blunted pH(i) recovery after sustained acidosis. Although preventing ERK1/2- p90RSK signaling pathway (10 µmol/L U0126) mimicked SIL effect, SIL did not blunt the acidosis-mediated increase in kinases activation. SIL+U0126 did not show additive effect on NHE-1 activity. Then, we hypothesized that SIL could be activating phophasatases (PP1 and/or PP2A) to directly dephosphorylate NHE-1 despite preserved ERK1/2-p90RSK activation. Non-specific phosphatases inhibition (1 µmol/L okadaic acid) canceled SIL effect on pH(i) recovery from acidosis. Same result was observed by inhibiting PP2A either with a lower dose of okadaic acid (1 nmol/L) or, more specifically, with 100 µmol/L endothall. Consistently, NHE-1 phosphorylation at Ser703 increased after acidosis, SIL prevented this effect and PP2A inhibition (endothall) reverted SIL effect. CONCLUSION: We suggest that PDE5A inhibitors decrease NHE-1 phosphorylation and activity through a mechanism that involves PP2A activation.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Phosphodiesterase Inhibitors/pharmacology , Protein Phosphatase 1/physiology , Protein Phosphatase 2/physiology , Sodium-Hydrogen Exchangers/metabolism , Acidosis/drug therapy , Animals , Butadienes/pharmacology , Dicarboxylic Acids/pharmacology , Hydrogen-Ion Concentration , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Okadaic Acid/pharmacology , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Phosphorylation , Piperazines/pharmacology , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Purines/pharmacology , Rats , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sildenafil Citrate , Sulfones/pharmacology
16.
Hypertension ; 56(4): 690-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20713918

ABSTRACT

The beneficial effect of phosphodiesterase 5A inhibition in ischemia/reperfusion injury and cardiac hypertrophy is well established. Inhibition of the cardiac Na(+)/H(+) exchanger (NHE-1) exerts beneficial effects on these same conditions, and a possible link between these therapeutic strategies was suggested. Experiments were performed in isolated cat cardiomyocytes to gain insight into the intracellular pathway involved in the reduction of NHE-1 activity by phosphodiesterase 5A inhibition. NHE-1 activity was assessed by the rate of intracellular pH recovery from a sustained acidic load in the absence of bicarbonate. Phosphodiesterase 5A inhibition with sildenafil (1 µmol/L) did not affect basal intracellular pH; yet, it did decrease proton efflux (J(H); in millimoles per liter per minute) after the acidic load (proton efflux: 6.97±0.43 in control versus 3.31±0.58 with sildenafil; P<0.05). The blockade of both protein phosphatase 1 and 2A with 100 nmol/L of okadaic acid reverted the sildenafil effect (proton efflux: 6.77±0.82). In contrast, selective inhibition of protein phosphatase 2A (1 nmol/L of okadaic acid or 100 µmol/L of endothall) did not (3.86±1.0 and 2.61±1.2), suggesting that only protein phosphatase 1 was involved in sildenafil-induced NHE-1 inhibition. Moreover, sildenafil prevented the acidosis-induced increase in NHE-1 phosphorylation without affecting activation of the extracellular signal-regulated kinase 1/2-p90(RSK) pathway. Our results suggest that phosphodiesterase 5A inhibition decreases NHE-1 activity, during intracellular pH recovery after an acidic load, by a protein phosphatase 1-dependent reduction in NHE-1 phosphorylation.


Subject(s)
Phosphodiesterase 5 Inhibitors , Piperazines/pharmacology , Protein Phosphatase 1/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sulfones/pharmacology , Animals , Biological Transport/drug effects , Cats , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Dicarboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Immunoblotting , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Okadaic Acid/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/drug effects , Protein Phosphatase 1/antagonists & inhibitors , Protons , Purines/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sildenafil Citrate
17.
18.
Medicina (B.Aires) ; 70(2): 200-201, Apr. 2010.
Article in Spanish | LILACS | ID: lil-633743
19.
J Physiol ; 588(Pt 9): 1579-90, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20231142

ABSTRACT

Myocardial stretch elicits a biphasic contractile response: the Frank-Starling mechanism followed by the slow force response (SFR) or Anrep effect. In this study we hypothesized that the SFR depends on epidermal growth factor receptor (EGFR) transactivation after the myocardial stretch-induced angiotensin II (Ang II)/endothelin (ET) release. Experiments were performed in isolated cat papillary muscles stretched from 92 to 98% of the length at which maximal twitch force was developed (L(max)). The SFR was 123 +/- 1% of the immediate rapid phase (n = 6, P < 0.05) and was blunted by preventing EGFR transactivation with the Src-kinase inhibitor PP1 (99 +/- 2%, n = 4), matrix metalloproteinase inhibitor MMPI (108 +/- 4%, n = 11), the EGFR blocker AG1478 (98 +/- 2%, n = 6) or the mitochondrial transition pore blocker clyclosporine (99 +/- 3%, n = 6). Stretch increased ERK1/2 phosphorylation by 196 +/- 17% of control (n = 7, P < 0.05), an effect that was prevented by PP1 (124 +/- 22%, n = 7) and AG1478 (131 +/- 17%, n = 4). In myocardial slices, Ang II (which enhances ET mRNA) or endothelin-1 (ET-1)-induced increase in O(2)() production (146 +/- 14%, n = 9, and 191 +/- 17%, n = 13, of control, respectively, P < 0.05) was cancelled by AG1478 (94 +/- 5%, n = 12, and 98 +/- 15%, n = 8, respectively) or PP1 (100 +/- 4%, n = 6, and 99 +/- 8%, n = 3, respectively). EGF increased O(2)() production by 149 +/- 4% of control (n = 9, P < 0.05), an effect cancelled by inhibiting NADPH oxidase with apocynin (110 +/- 6% n = 7), mKATP channels with 5-hydroxydecanoic acid (5-HD; 105 +/- 5%, n = 8), the respiratory chain with rotenone (110 +/- 7%, n = 7) or the mitochondrial permeability transition pore with cyclosporine (111 +/- 10%, n = 6). EGF increased ERK1/2 phosphorylation (136 +/- 8% of control, n = 9, P < 0.05), which was blunted by 5-HD (97 +/- 5%, n = 4), suggesting that ERK1/2 activation is downstream of mitochondrial oxidative stress. Finally, stretch increased Ser703 Na(+)/H(+) exchanger-1 (NHE-1) phosphorylation by 172 +/- 24% of control (n = 4, P < 0.05), an effect that was cancelled by AG1478 (94 +/- 17%, n = 4). In conclusion, our data show for the first time that EGFR transactivation is crucial in the chain of events leading to the Anrep effect.


Subject(s)
ErbB Receptors/physiology , Mechanoreceptors/physiology , Myocardial Contraction/physiology , Transcriptional Activation/physiology , Angiotensin II/biosynthesis , Animals , Cats , Endothelin-1/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Oxidation-Reduction , Papillary Muscles/physiology , Phosphorylation , RNA/biosynthesis , RNA/genetics , Reactive Oxygen Species/metabolism , Receptor Cross-Talk/physiology , Receptors, G-Protein-Coupled/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Sodium-Hydrogen Exchangers/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL