Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Crit Care Med ; 46(8): e805-e810, 2018 08.
Article in English | MEDLINE | ID: mdl-29782355

ABSTRACT

OBJECTIVES: The vascular endothelium is a major target of sepsis-induced events, and endothelial activation accounts for much of the pathology of sepsis. Urinary tract infections and pneumonia caused by Escherichia coli are among of the most common infections causing sepsis in both community and hospital settings. Currently, there are no approved drugs on the market to treat the underlying pathophysiology of sepsis. The aim of this study is to elucidate the molecular mechanism by which E. coli induces endothelial injury as a result of attachment. DESIGN: Laboratory research using a hemodynamic perfusion ex vivo model. SETTING: Research Laboratories of Royal College of Surgeons in Ireland and Beaumont Hospital. PATIENTS: Ex vivo human vascular endothelial cells. INTERVENTIONS: Addition of αVß3 antagonist, cilengitide. MEASUREMENTS AND MAIN RESULTS: Clinical strains of E. coli isolated from patients with sepsis bound to sheared human endothelial cells under static and hemodynamic shear conditions. Binding was dependent on E. coli cell membrane protein outer membrane protein A attaching directly to endothelial cell integrin αVß3. Attachment resulted in disturbances in endothelial barrier integrity, as determined by loss of tight junction protein staining, permeability changes, and ultimately cell death by apoptosis. Using a low concentration of the αVß3 antagonist cilengitide or using a strain deficient in outer membrane protein A resulted in a significant reduction in endothelial dysfunction following infection. CONCLUSIONS: Inhibition of E. coli binding to endothelial cell αVß3 by cilengitide prevents endothelial dysfunction and may, therefore, present as a novel early therapeutic for the treatment of sepsis.


Subject(s)
Endothelial Cells/microbiology , Escherichia coli/growth & development , Integrin alphaVbeta3/antagonists & inhibitors , Sepsis/microbiology , Snake Venoms/pharmacology , Capillary Permeability , Dose-Response Relationship, Drug , Humans
2.
Adv Mater ; 30(15): e1706442, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29504165

ABSTRACT

Electroconductive substrates are emerging as promising functional materials for biomedical applications. Here, the development of biohybrids of collagen and pristine graphene that effectively harness both the biofunctionality of the protein component and the increased stiffness and enhanced electrical conductivity (matching native cardiac tissue) obtainable with pristine graphene is reported. As well as improving substrate physical properties, the addition of pristine graphene also enhances human cardiac fibroblast growth while simultaneously inhibiting bacterial attachment (Staphylococcus aureus). When embryonic-stem-cell-derived cardiomyocytes (ESC-CMs) are cultured on the substrates, biohybrids containing 32 wt% graphene significantly increase metabolic activity and cross-striated sarcomeric structures, indicative of the improved substrate suitability. By then applying electrical stimulation to these conductive biohybrid substrates, an enhancement of the alignment and maturation of the ESC-CMs is achieved. While this in vitro work has clearly shown the potential of these materials to be translated for cardiac applications, it is proposed that these graphene-based biohybrid platforms have potential for a myriad of other applications-particularly in electrically sensitive tissues, such as neural and neural and musculoskeletal tissues.


Subject(s)
Biocompatible Materials/chemistry , Collagen , Electric Conductivity , Graphite , Humans , Myocytes, Cardiac
3.
FASEB J ; 32(4): 1969-1981, 2018 04.
Article in English | MEDLINE | ID: mdl-29183963

ABSTRACT

Most mammalian cells can intercommunicate via connexin-assembled, gap-junctional channels. To regulate signal transmission, connexin (Cx) channel permeability must respond dynamically to physiological and pathophysiological stimuli. One key stimulus is intracellular pH (pHi), which is modulated by a tissue's metabolic and perfusion status. Our understanding of the molecular mechanism of H+ gating of Cx43 channels-the major isoform in the heart and brain-is incomplete. To interrogate the effects of acidic and alkaline pHi on Cx43 channels, we combined voltage-clamp electrophysiology with pHi imaging and photolytic H+ uncaging, performed over a range of pHi values. We demonstrate that Cx43 channels expressed in HeLa or N2a cell pairs are gated biphasically by pHi via a process that consists of activation by H+ ions at alkaline pHi and inhibition at more acidic pHi. For Cx43 channel-mediated solute/ion transmission, the ensemble of these effects produces a pHi optimum, near resting pHi. By using Cx43 mutants, we demonstrate that alkaline gating involves cysteine residues of the C terminus and is independent of motifs previously implicated in acidic gating. Thus, we present a molecular mechanism by which cytoplasmic acid-base chemistry fine tunes intercellular communication and establishes conditions for the optimal transmission of solutes and signals in tissues, such as the heart and brain.-Garciarena, C. D., Malik, A., Swietach, P., Moreno, A. P., Vaughan-Jones, R. D. Distinct moieties underlie biphasic H+ gating of connexin43 channels, producing a pH optimum for intercellular communication.


Subject(s)
Connexin 43/metabolism , Ion Channel Gating , Protons , Animals , Cell Communication , Connexin 43/chemistry , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice
5.
Drug Discov Today ; 21(9): 1437-1445, 2016 09.
Article in English | MEDLINE | ID: mdl-27117348

ABSTRACT

The field of tissue engineering is developing biomimetic biomaterial scaffolds that are showing increasing therapeutic potential for the repair of cardiovascular tissues. However, a major opportunity exists to use them as 3D in vitro models for the study of cardiovascular tissues and disease in addition to drug development and testing. These in vitro models can span the gap between 2D culture and in vivo testing, thus reducing the cost, time, and ethical burden of current approaches. Here, we outline the progress to date and the requirements for the development of ideal in vitro 3D models for blood vessels, heart valves, and myocardial tissue.


Subject(s)
Blood Vessels , Heart Valves , Models, Biological , Myocardium , Animals , Cardiovascular Diseases , Humans , Tissue Engineering
6.
Pathogens ; 4(4): 869-82, 2015 Dec 05.
Article in English | MEDLINE | ID: mdl-26690226

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen often carried asymptomatically on the human body. Upon entry to the otherwise sterile environment of the cardiovascular system, S. aureus can lead to serious complications resulting in organ failure and death. The success of S. aureus as a pathogen in the bloodstream is due to its ability to express a wide array of cell wall proteins on its surface that recognise host receptors, extracellular matrix proteins and plasma proteins. Endothelial cells and platelets are important cells in the cardiovascular system and are a major target of bloodstream infection. Endothelial cells form the inner lining of a blood vessel and provide an antithrombotic barrier between the vessel wall and blood. Platelets on the other hand travel throughout the cardiovascular system and respond by aggregating around the site of injury and initiating clot formation. Activation of either of these cells leads to functional dysregulation in the cardiovascular system. In this review, we will illustrate how S. aureus establish intimate interactions with both endothelial cells and platelets leading to cardiovascular dysregulation.

7.
J Mol Cell Cardiol ; 61: 51-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23602948

ABSTRACT

Acid extrusion on Na(+)-coupled pH-regulatory proteins (pH-transporters), Na(+)/H(+) exchange (NHE1) and Na(+)-HCO3(-) co-transport (NBC), drives Na(+) influx into the ventricular myocyte. This H(+)-activated Na(+)-influx is acutely up-regulated at pHi<7.2, greatly exceeding Na(+)-efflux on the Na(+)/K(+) ATPase. It is spatially heterogeneous, due to the co-localisation of NHE1 protein (the dominant pH-transporter) with gap-junctions at intercalated discs. Overall Na(+)-influx via NBC is considerably lower, but much is co-localised with L-type Ca(2+)-channels in transverse-tubules. Through a functional coupling with Na(+)/Ca(2+) exchange (NCX), H(+)-activated Na(+)-influx increases sarcoplasmic-reticular Ca(2+)-loading and release during intracellular acidosis. This raises Ca(2+)-transient amplitude, rescuing it from direct H(+)-inhibition. Functional coupling is biochemically regulated and linked to membrane receptors, through effects on NHE1 and NBC. It requires adequate cytoplasmic Na(+)-mobility, as NHE1 and NCX are spatially separated (up to 60µm). The relevant functional NCX activity must be close to dyads, as it exerts no effect on bulk diastolic Ca(2+). H(+)-activated Na(+)-influx is up-regulated during ischaemia-reperfusion and some forms of maladaptive hypertrophy and heart failure. It is thus an attractive system for therapeutic manipulation. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".


Subject(s)
Acid-Base Imbalance/metabolism , Cation Transport Proteins/physiology , Excitation Contraction Coupling , Sodium-Hydrogen Exchangers/physiology , Animals , Cardiomegaly/metabolism , Heart Failure/metabolism , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/physiology , Protons , Sodium-Hydrogen Exchanger 1
8.
J Physiol ; 591(9): 2287-306, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23420656

ABSTRACT

Membrane acid extrusion by Na(+)/H(+) exchange (NHE1) and Na(+)-HCO3(-) co-transport (NBC) is essential for maintaining a low cytoplasmic [H(+)] (∼60 nm, equivalent to an intracellular pH (pHi) of 7.2). This protects myocardial function from the high chemical reactivity of H(+) ions, universal end-products of metabolism. We show here that, in rat ventricular myocytes, fluorescent antibodies map the NBC isoforms NBCe1 and NBCn1 to lateral sarcolemma, intercalated discs and transverse tubules (t-tubules), while NHE1 is absent from t-tubules. This unexpected difference matches functional measurements of pHi regulation (using AM-loaded SNARF-1, a pH fluorophore). Thus, myocyte detubulation (by transient exposure to 1.5 m formamide) reduces global acid extrusion on NBC by 40%, without affecting NHE1. Similarly, confocal pHi imaging reveals that NBC stimulation induces spatially uniform pHi recovery from acidosis, whereas NHE1 stimulation induces pHi non-uniformity during recovery (of ∼0.1 units, for 2-3 min), particularly at the ends of the cell where intercalated discs are commonly located, and where NHE1 immunostaining is prominent. Mathematical modelling shows that this induction of local pHi microdomains is favoured by low cytoplasmic H(+) mobility and long H(+) diffusion distances, particularly to surface NHE1 transporters mediating high membrane flux. Our results provide the first evidence for a spatial localisation of [H(+)]i regulation in ventricular myocytes, suggesting that, by guarding pHi, NHE1 preferentially protects gap junctional communication at intercalated discs, while NBC locally protects t-tubular excitation-contraction coupling.


Subject(s)
Myocytes, Cardiac/physiology , Sarcolemma/physiology , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Calcium/physiology , Electric Capacitance , Female , Guinea Pigs , Heart Ventricles , Hydrogen-Ion Concentration , Male , Rats , Rats, Sprague-Dawley
9.
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
10.
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
11.
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
12.
J Mol Cell Cardiol ; 47(5): 716-22, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19646989

ABSTRACT

The sarcolemmal Na+/HCO3- cotransporter (NBC) plays an important role in intracellular pH (pH(i)) regulation in the heart. In the present work we studied, in isolated cat ventricular myocytes, the role of Angiotensin II (Ang II) and reactive oxygen species (ROS) production as potential activators of the NBC. pH(i) was measured in single cells in a medium with HCO3- using the fluorescent pH indicator BCECF. The NH4+ pulse method was used to induce an intracellular acid load and the acid efflux (JH) in the presence of the Na+/H+ exchanger blocker HOE642 (10 microM) was calculated as indicator of NBC activity. The following JH data are presented at pH(i) of 6.8 (* and # indicate p<0.05 after ANOVA vs. control and Ang II, respectively). The basal JH (1.03+/-0.12 mM/min, n=11) was significantly increased in the presence of 100 nM Ang II (1.70+/-0.15 mM/min, n=8*). This effect of Ang II was abolished when we added to the extracellular solution 2 mM MPG (ROS scavenger; 0.80+/-0.08 mM/min, n=11#), 300 microM apocynin (NADPH oxidase blocker; 0.80+/-0.13 mM/min, n=6#), 500 microM 5-hydroxidecanoate (mitochondrial ATP dependent K+ channel, mK(ATP), blocker; 0.97+/-0.21 mM/min, n=9#), or the inhibitor of the MAP kinase ERK pathway U0126 (10 microM; 0.56+/-0.18 mM/min, n=6#). We also determined the phosphorylation of ERK during the first min of acidosis and we detected that Ang II significantly enhanced the ERK phosphorylation levels, an effect that was cancelled by scavenging ROS with MPG. In conclusion, we propose that Ang II enhances the production of ROS through the activation of the NADPH oxidase, which in turn triggers mK(ATP) opening and mitochondrial ROS production ("ROS-induced ROS-release mechanism"). Finally, these mitochondrial ROS stimulate the ERK pathway, leading to the activation of the NBC.


Subject(s)
Angiotensin II/pharmacology , Biological Transport/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Sodium-Bicarbonate Symporters/metabolism , Vasoconstrictor Agents/pharmacology , Animals , Biological Transport/physiology , Cats , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation
13.
Hypertension ; 53(4): 708-14, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19221208

ABSTRACT

The effect of endurance training (swimming 90 min/d for 5 days a week for 60 days) on cardiac hypertrophy was investigated in the spontaneously hypertensive rat (SHR). Sedentary SHRs (SHR-Cs) and normotensive Wistar rats were used as controls. Exercise training enhanced myocardial hypertrophy assessed by left ventricular weight/tibial length (228+/-7 versus 251+/-5 mg/cm in SHR-Cs and exercised SHRs [SHR-Es], respectively). Myocyte cross-sectional area increased approximately 40%, collagen volume fraction decreased approximately 50%, and capillary density increased approximately 45% in SHR-Es compared with SHR-Cs. The mRNA abundance of atrial natriuretic factor and myosin light chain 2 was decreased by the swimming routine (100+/-19% versus 41+/-10% and 100+/-8% versus 61+/-9% for atrial natriuretic factor and myosin light chain 2 in SHR-Cs and SHR-Es, respectively). The expression of sarcoplasmic reticulum Ca(2+) pump was significantly augmented, whereas that of Na(+)/Ca(2+) exchanger was unchanged (93+/-7% versus 167+/-8% and 158+/-13% versus 157+/-7%, sarcoplasmic reticulum Ca(2+) pump and Na(+)/Ca(2+) exchanger in SHR-Cs and SHR-Es, respectively; P<0.05). Endurance training inhibited apoptosis, as reflected by a decrease in caspase 3 activation and poly(ADP-ribose) polymerase-1 cleavage, and normalized calcineurin activity without inducing significant changes in the phosphatidylinositol 3-kinase/Akt pathway. The swimming routine improved midventricular shortening determined by echocardiography (32.4+/-0.9% versus 36.9+/-1.1% in SHR-Cs and SHR-Es, respectively; P<0.05) and decreased the left ventricular free wall thickness/left ventricular cavity radius toward an eccentric model of cardiac hypertrophy (0.59+/-0.02 versus 0.53+/-0.01 in SHR-Cs and SHR-Es, respectively; P<0.05). In conclusion, we present data demonstrating the effectiveness of endurance training to convert pathological into physiological hypertrophy improving cardiac performance. The reduction of myocardial fibrosis and calcineurin activity plus the increase in capillary density represent factors to be considered in determining this beneficial effect.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , Hypertension/pathology , Hypertension/physiopathology , Physical Endurance/physiology , Animals , Apoptosis/physiology , Calcineurin/metabolism , Cardiomegaly/therapy , Down-Regulation/physiology , Gene Expression/physiology , Hypertension/therapy , Male , Rats , Rats, Inbred SHR , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Signal Transduction/physiology , Sodium-Calcium Exchanger/genetics , Swimming/physiology
14.
J Appl Physiol (1985) ; 106(4): 1325-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179646

ABSTRACT

Na(+)/H(+) exchanger (NHE-1) inhibition was demonstrated to induce the regression of cardiac hypertrophy (CH) in several experimental models and to inhibit mitochondrial death pathway in "in-vitro" experiments. Since recent reports show that NHE-1 inhibition delays the transition from CH to failure, and apoptosis plays a key role in this process, we investigated the effect of chronic treatment with the NHE-1 blocker cariporide on CH and apoptosis in the SHR. One month of cariporide treatment (30 mg x kg(-1) x day(-1)) induced the regression of CH (cardiomyocyte cross-sectional area: 468 +/- 20 vs. 285 +/- 9 microm(2) in untreated and cariporide-treated spontaneously hypertensive rats; P < 0.05). Apoptosis was assessed by TUNEL staining, the expression of Bcl-2, Bax, and activation of caspase-3 and PARP-1 by immunoblot. Cariporide treatment decreased the TUNEL-positive cells, the Bax-to-Bcl-2 ratio (3.16 +/- 0.32 vs. 1.70 +/- 0.17, untreated and cariporide-treated, respectively; P < 0.05); caspase-3 and PARP-1 activation (465 +/- 62 vs. 260 +/- 22 and 2,239 +/- 62 vs. 1,683 +/- 85 AU, untreated and cariporide-treated, respectively; P < 0.05). Angiotensin II, a growth factor and apoptotic stimulus, was used to induce O(2)(-) production that activated the ERK1/2-p90(RSK) pathway, increasing NHE-1 phosphorylation. These effects were prevented by losartan, N-(2-mercaptopropionyl)-glycine, and cariporide. In conclusion, we present data demonstrating that chronic NHE-1 inhibition with cariporide decreases both hypertrophy and apoptosis susceptibility in the spontaneously hypertensive rat heart. The antiapoptotic effect would be the consequence of two different actions of cariporide: the prevention of cytosolic Na(+) and Ca(2+) overload due to the inhibition of the sarcolemmal NHE-1 and a direct mitochondrial effect preventing mitochondrial permeability transition pore opening.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Apoptosis/drug effects , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Guanidines/therapeutic use , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/therapeutic use , Angiotensin II/pharmacology , Animals , Blotting, Western , Cell Nucleus/pathology , Cell Nucleus/ultrastructure , In Situ Nick-End Labeling , Male , Mitochondria, Heart/drug effects , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Rats , Rats, Inbred SHR , Sodium-Hydrogen Exchanger 1 , Superoxides/metabolism , bcl-2-Associated X Protein/metabolism
15.
Hypertens Res ; 31(7): 1465-76, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18957818

ABSTRACT

The aim of this work was to assess the possible correlation between oxidative damage and the development of cardiac hypertrophy in heart tissue from young (40-d-old) and older (4-, 11- and 19-month-old) spontaneously hypertensive rats (SHR) in comparison with age-matched Wistar (W) rats. To this end, levels of thiobarbituric acid reactive substances (TBARS), nitrotyrosine contents, NAD(P)H oxidase activity, superoxide production, and the activities of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) were determined. Compared to age-matched normotensive rats, SHR showed a significant increase in systolic blood pressure from 40 d of age and left ventricular hypertrophy (LVH) was significantly evident from 4 months of age. W rats (11- and 19-month-old) also showed an increase in LVH with aging. TBARS and nitrotyrosine levels were similar in young rats from both strains and were significantly increased with age in both strains, with the values in SHR being significantly higher than those in age-matched W rats. NAD(P)H activity was similar in young SHR and W rats, whereas it was higher in aged SHR compared with age-matched W rats. Compared to W rats, superoxide production was higher in aged SHR, and was abolished by NAD(P)H inhibition with apocynin. CAT activity was increased in the hearts of 4-month-old SHR compared to age-matched W rats and was decreased in the hearts of the oldest SHR compared to the oldest W rats. SOD and GPx activities decreased in both rat strains with aging. Moreover, an increase in collagen deposition with aging was evident in both rat strains. Taken together, these data showed that aged SHR exhibited higher cardiac hypertrophy and oxidative damage compared to W rats, indicating that the two undesirable effects are associated. That is, oxidative stress appears to be a cause and/or consequence of hypertrophy development in this animal model.


Subject(s)
Cardiomegaly/metabolism , Oxidative Stress , Animals , Cardiomegaly/etiology , Catalase/metabolism , Collagen/analysis , Glutathione Peroxidase/metabolism , Lipid Peroxidation , Male , NADPH Oxidases/metabolism , Rats , Rats, Inbred SHR , Rats, Wistar , Superoxide Dismutase/metabolism , Superoxides/metabolism
16.
J Appl Physiol (1985) ; 105(6): 1706-13, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18801963

ABSTRACT

The possibility of a direct mitochondrial action of Na(+)/H(+) exchanger-1 (NHE-1) inhibitors decreasing reactive oxygen species (ROS) production was assessed in cat myocardium. Angiotensin II and endothelin-1 induced an NADPH oxidase (NOX)-dependent increase in anion superoxide (O(2)(-)) production detected by chemiluminescence. Three different NHE-1 inhibitors [cariporide, BIIB-723, and EMD-87580] with no ROS scavenger activity prevented this increase. The mitochondria appeared to be the source of the NOX-dependent ROS released by the "ROS-induced ROS release mechanism" that was blunted by the mitochondrial ATP-sensitive potassium channel blockers 5-hydroxydecanoate and glibenclamide, inhibition of complex I of the electron transport chain with rotenone, and inhibition of the permeability transition pore (MPTP) by cyclosporin A. Cariporide also prevented O(2)(-) production induced by the opening of mK(ATP) with diazoxide. Ca(2+)-induced swelling was evaluated in isolated mitochondria as an indicator of MPTP formation. Cariporide decreased mitochondrial swelling to the same extent as cyclosporin A and bongkrekic acid, confirming its direct mitochondrial action. Increased O(2)(-) production, as expected, stimulated ERK1/2 and p90 ribosomal S6 kinase phosphorylation. This was also prevented by cariporide, giving additional support to the existence of a direct mitochondrial action of NHE-1 inhibitors in preventing ROS release. In conclusion, we report a mitochondrial action of NHE-1 inhibitors that should lead us to revisit or reinterpret previous landmark observations about their beneficial effect in several cardiac diseases, such as ischemia-reperfusion injury and cardiac hypertrophy and failure. Further studies are needed to clarify the precise mechanism and site of action of these drugs in blunting MPTP formation and ROS release.


Subject(s)
Mitochondria, Heart/metabolism , Myocardium/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Superoxides/metabolism , Angiotensin II/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Calcium Chloride/pharmacology , Cats , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanidines/pharmacology , In Vitro Techniques , Mitochondria, Heart/drug effects , Mitochondrial Swelling/drug effects , NADPH Oxidases/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sulfones/pharmacology
17.
Front Biosci ; 13: 7096-114, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508719

ABSTRACT

The enhanced activity of the cardiac Na+/H+ exchanger (NHE-1) after myocardial stretch is considered a key step of the intracellular signaling pathway leading to the slow force response to stretch as well as an early signal for the development of cardiac hypertrophy. We propose that the chain of events triggered by stretch begins with the release of small amounts of Angiotensin II (Ang II)/endothelin (ET) and ends with the increase in intracellular Ca2+ concentration ([Ca2+]i) through the Na+/Ca2+ exchanger in reverse mode (NCX(rev)), which triggers cardiac hypertrophy by activation of widely recognized Ca2+-dependent intracellular signaling pathways.


Subject(s)
Cardiomegaly/physiopathology , Sodium-Hydrogen Exchangers/physiology , Adult , Angiotensin II/physiology , Biomechanical Phenomena , Cardiomegaly/genetics , Heart Ventricles/physiopathology , Humans , Hypertrophy, Right Ventricular/physiopathology , Obesity/genetics , Pressoreceptors/physiology , Reactive Oxygen Species/metabolism , Signal Transduction , Ventricular Function
18.
J Physiol ; 584(Pt 3): 895-905, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17823205

ABSTRACT

When the length of the myocardium is increased, a biphasic response to stretch occurs involving an initial rapid increase in force followed by a delayed slow increase called the slow force response (SFR). Confirming previous findings involving angiotensin II in the SFR, it was blunted by AT1 receptor blockade (losartan). The SFR was accompanied by an increase in reactive oxygen species (ROS) of approximately 30% and in intracellular Na(+) concentration ([Na(+)](i)) of approximately 2.5 mmol l(-1) over basal detected by H(2)DCFDA and SBFI fluorescence, respectively. Abolition of ROS by 2-mercapto-propionyl-glycine (MPG) and EUK8 suppressed the increase in [Na(+)](i) and the SFR, which were also blunted by Na(+)/H(+) exchanger (NHE-1) inhibition (HOE642). NADPH oxidase inhibition (apocynin or DPI) or blockade of the ATP-sensitive mitochondrial potassium channels (5HD or glybenclamide) suppressed both the SFR and the increase in [Na(+)](i) after stretch, suggesting that endogenous angiotensin II activated NADPH oxidase leading to ROS release by the ATP-sensitive mitochondrial potassium channels, which promoted NHE-1 activation. Supporting the notion of ROS-mediated NHE-1 activation, stretch increased the ERK1/2 and p90rsk kinases phosphorylation, effect that was cancelled by losartan. In agreement, the SFR was cancelled by inhibiting the ERK1/2 signalling pathway with PD98059. Angiotensin II at a dose that mimics the SFR (1 nmol l(-1)) induced an increase in .O(2)(-) production of approximately 30-40% detected by lucigenin in cardiac slices, an effect that was blunted by losartan, MPG, apocynin, 5HD and glybenclamide. Taken together the data suggest a pivotal role of mitochondrial ROS in the genesis of the SFR to stretch.


Subject(s)
Mitochondria, Heart/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , Papillary Muscles/physiology , Reactive Oxygen Species/metabolism , Animals , Cats , Mechanotransduction, Cellular , Stress, Mechanical
19.
Can J Physiol Pharmacol ; 85(3-4): 301-10, 2007.
Article in English | MEDLINE | ID: mdl-17612638

ABSTRACT

Na+/H+ exchanger-1 (NHE-1) inhibition induces cardiac hypertrophy regression and (or) prevention in several experimental models, although the intracellular events involved remain unclarified. We aimed to determine whether the calcineurin/NFAT pathway mediates this effect of NHE-1 inhibitors. Spontaneously hypertensive rats (SHR) with cardiac hypertrophy were treated with the NHE-1 inhibitors cariporide and BIIB723 for 30 days. Wistar rats served as normotensive controls. Their hearts were studied by echocardiography, immunoblotting, and real-time RT-PCR. Cytoplasmic Ca2+ and calcineurin Abeta expression were measured in cultured neonatal rat ventricular myocytes (NRVM) stimulated with endothelin-1 for 24 h. NHE-1 blockade induced cardiac hypertrophy regression (heart mass/body mass=3.63+/-0.07 vs. 3.06+/-0.05 and 3.02+/-0.13 for untreated vs. cariporide- and BIIB723-treated SHR, respectively; p<0.05) and decreased myocardial brain natriuretic peptide, calcineurin Abeta, and nuclear NFAT expressions. Echocardiographic evaluation demonstrated a reduction in left ventricular wall thickness without changes in cavity dimensions or a significant decrease in blood pressure. NHE-1-inhibitor treatment did not affect myocardial stiffness or endocardial shortening, but increased mid-wall shortening, suggesting that a positive inotropic effect develops after hypertrophy regression. Cariporide normalized the increased diastolic Ca2+ and calcineurin Abeta expression observed in ET-1-stimulated NRVM. In conclusion, our data suggest that inactivation of calcineurin/NFAT pathway may underlie the regression of cardiac hyper-trophy induced by NHE-1 inhibition.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/metabolism , Hypertension/metabolism , NFATC Transcription Factors/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Animals , Animals, Newborn , Anti-Arrhythmia Agents/pharmacology , Calcium/metabolism , Cells, Cultured , Guanidines/pharmacology , Male , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Wistar , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/metabolism , Sulfones/pharmacology
20.
Hypertension ; 49(5): 1095-103, 2007 May.
Article in English | MEDLINE | ID: mdl-17339532

ABSTRACT

Acute phosphodiesterase 5A inhibition by sildenafil or EMD360527/5 promoted profound inhibition of the cardiac Na(+)/H(+) exchanger (NHE-1), detected by the almost null intracellular pH recovery from an acute acid load (ammonium prepulse) in isolated papillary muscles from Wistar rats. Inhibition of phosphoglycerate kinase-1 (KT5823) restored normal NHE-1 activity, suggesting a causal link between phosphoglycerate kinase-1 increase and NHE-1 inhibition. We then tested whether the beneficial effects of NHE-1 inhibitors against the deleterious postmyocardial infarction (MI) remodeling can be detected after sildenafil-mediated NHE-1 inhibition. MI was induced by left anterior descending coronary artery ligation in Wistar rats, which were randomized to placebo or sildenafil (100 mg kg(-1) day(-1)) for 6 weeks. Sildenafil significantly increased left ventricular phosphoglycerate kinase-1 activity in the post-MI group without affecting its expression. MI increased heart weight/body weight ratio, left ventricular myocyte cross-sectional area, interstitial fibrosis, and brain natriuretic peptide and NHE-1 expression. Sildenafil blunted these effects. Neither a significant change in infarct size nor a change in arterial or left ventricular systolic pressure was detected after sildenafil. MI decreased fractional shortening and the ratio of the maximum rate of rise of LVP divided by the pressure at the moment such maximum occurs, effects that were prevented by sildenafil. Intracellular pH recovery after an acid load was faster in papillary muscles from post-MI hearts (versus sham), whereas sildenafil significantly inhibited NHE-1 activity in both post-MI and sildenafil-treated sham groups. We conclude that increased phosphoglycerate kinase-1 activity after acute phosphodiesterase 5A inhibition blunts NHE-1 activity and protects the heart against post-MI remodeling and dysfunction.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Myocardial Infarction/prevention & control , Phosphodiesterase Inhibitors/pharmacology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Acids/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5 , Hydrogen-Ion Concentration , Male , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Phosphoglycerate Kinase/metabolism , Piperazines/pharmacology , Purines/pharmacology , Rats , Rats, Wistar , Sildenafil Citrate , Sulfones/pharmacology , Ventricular Remodeling/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...