• Nenhum resultado encontrado

Isoflurane preconditioning confers cardioprotection by activation of ALDH2.

N/A
N/A
Protected

Academic year: 2017

Share "Isoflurane preconditioning confers cardioprotection by activation of ALDH2."

Copied!
11
0
0

Texto

(1)

Activation of ALDH2

Xiao-E Lang1*, Xiong Wang1, Ke-Rang Zhang2, Ji-Yuan Lv1, Jian-Hua Jin3, Qing-Shan Li4

1Department of Cardiology, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China,2Department of Psychiatry, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China,3Department of Nuclear Medicine, The First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China,4School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, China

Abstract

The volatile anesthetic, isoflurane, protects the heart from ischemia/reperfusion (I/R) injury. Aldehyde dehydrogenase 2 (ALDH2) is thought to be an endogenous mechanism against ischemia-reperfusion injury possibly through detoxification of toxic aldehydes. We investigated whether cardioprotection by isoflurane depends on activation of ALDH2.Anesthetized rats underwent 40 min of coronary artery occlusion followed by 120 min of reperfusion and were randomly assigned to the following groups: untreated controls, isoflurane preconditioning with and without an ALDH2 inhibitor, the direct activator of ALDH2 or a protein kinase C (PKCe) inhibitor. Pretreatment with isoflurane prior to ischemia reduced LDH and CK-MB levels and infarct size, while it increased phosphorylation of ALDH2, which could be blocked by the ALDH2 inhibitor, cyanamide. Isolated neonatal cardiomyocytes were treated with hypoxia followed by reoxygenation. Hypoxia/ reoxygenation (H/R) increased cardiomyocyte apoptosis and injury which were attenuated by isoflurane and forced the activation of ALDH2. In contrast, the effect of isoflurane-induced protection was almost abolished by knockdown of ALDH2. Activation of ALDH2 and cardioprotection by isoflurane were substantially blocked by the PKCe inhibitor. Activation of ALDH2 by mitochondrial PKCeplays an important role in the cardioprotection of isoflurane in myocardium I/R injury.

Citation:Lang X-E, Wang X, Zhang K-R, Lv J-Y, Jin J-H, et al. (2013) Isoflurane Preconditioning Confers Cardioprotection by Activation of ALDH2. PLoS ONE 8(2): e52469. doi:10.1371/journal.pone.0052469

Editor:Shree Ram Singh, National Cancer Institute, United States of America

ReceivedAugust 25, 2012;AcceptedNovember 13, 2012;PublishedFebruary 28, 2013

Copyright:ß2013 Lang et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Funding:This study is supported by National Natural Science Foundation of China (81172938). The funder’s website is www.nsfc.gov.cn. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Competing Interests:The authors have declared that no competing interests exist. * E-mail: langxiaoe2007@126.com

Introduction

Acute myocardial infarction (AMI) is responsible for the death of millions of persons worldwide each year [1]. Murry et al. demonstrated that a succession of short periods of myocardial ischemia and reperfusion prior to the continuous maintenance of coronary reperfusion protects the myocardium against subsequent prolonged ischemic insults, which has been termed ‘ischemic preconditioning’ (IPC) [2]. This phenomenon is achieved by several pharmacological agents, including volatile anesthetics. Volatile anesthetics such as isoflurane have cardioprotective effects when administered before a period of myocardial ischemia and reperfusion, and this phenomenon is referred to as anesthetic preconditioning (APC) [3,4]. APC is a cardioprotective strategy that increases resistance to ischemia and reperfusion (I/R) by eliciting innate protective mechanisms, and was described in various animal models [3–6], as well as in humans [7,8]. APC has been shown to reduce infarct size, and attenuate contractile dysfunction and serum CK-MB concentration caused by myocar-dial ischemia. Cellular signaling during APC is complex, and in many aspects, comparable to that of IPC. The intracellular mechanisms involved in APC have not been completely identified. It has become clear that multiple cellular pathways participate in the establishment of a cellular phenotype that makes the heart more resistant to ischemic damage. Mechanisms reported to date involve inhibition of mitochondrial permeability transition pore (mPTP) opening [9], the activation of kinases such as protein

kinase C (PKC) [10,11], the generation of reactive oxygen species (ROS) [12,13], and opening of adenosine triphosphate-sensitive potassium channels (KATP) [3,14,15].

(2)

role of ALDH2 in isoflurane-induced APC has not been investigated. Thus, the current study tested the hypothesis that PKCe-mediated activation of mitochondrial ALDH2 plays a critical role in isoflurane preconditioning.

Materials and Methods

Animals

Male Sprague-Dawley (SD) rats, weighing 200–220 g, were used in this study. The animals were provided by Experimental Animal Center of Tsinghua University. They were placed in a

quiet, temperature (2363uC) and humidity (6065%) controlled room with a 12:12 hours light-dark cycle (light beginning at 8 a.m.). This study was conducted in accordance with the Guide for the Care and Use of Laboratory Animals of the China National Institutes of Health.

Ischemia/reperfusion Injury Experimental Protocol

The acute myocardial I/R injury model was performed as previously described [25]. Male SD rats were anesthetized with pentobarbital sodium (30 mg/kg, i.p.). After a tracheotomy had been performed, rats’ lungs were ventilated mechanically with Figure 1. The influence of anesthetic-induced preconditioning with 1.0 MAC of isoflurane on leakage of LDH and CK-MB, and infarct area in rat hearts.A, B. Serum LDH and CK-MB concentrations were analyzed. The increase in LDH and CK-MB concentrations was lower in rats pretreated with isoflurane than in I/R group. C. Isoflurane preconditioning significantly decreased infarct area compared with I/R group animals. Representative cross-sectional slices derived from a single heart with and without isoflurane. The infarct size normalized to the area at risk. Values are means6S.E.M., n = 5 in each group. *P,0.05, **P,0.01 vs. sham group, and#P

,0.05 vs. the I/R control group. doi:10.1371/journal.pone.0052469.g001

(3)

Figure 2. Phosphorylation of ALDH2 associated with isoflurane-induced cardioprotection.A. Effects of isoflurane preconditioning with and without the ALDH2 inhibitor (cyanamide), and direct activator of ALDH2 (Alda-44) on phosphorylation of ALDH2. (a) Representative of western blot analysis of the phosphorylation of ALDH2 (phos-ALDH2 top lanes) and total ALDH2 (middle lanes).b-actin (lower lanes) was used to demonstrate equal protein loading. (b) Quantification of phos-ALDH2 to the total ALDH2 from 3 independent experiments. B, C. Serum LDH and CK-MB concentrations were analyzed. D. The effects of ALDH inhibitor (cyanamide), and direct activator of ALDH2 (Alda-44) on infarct area in rat hearts. (a) Representative cross-sectional slices derived from a single heart. (b) The infarct size normalized to the area at risk. Values are means6S.E.M., n = 8 in each group.##

(4)
(5)

positive pressure ventilation using 30–40% air/oxygen mixture to maintain arterial blood gas pH within a physiological range by adjusting the respiratory rate and tidal volume throughout the experiment. Myocardial infarction (MI) was created by ligation of the left anterior descending (LAD).The thorax was opened at the fourth or fifth left intercostal space. After left thoracotomy and pericardiotomy, MI was induced by LAD ligation 2–3 mm from the origin with a 6–0 silk suture. All animals (except for the rats in the sham groups) were subjected to 40 min of regional myocardial ischemia followed by 120 min of reperfusion. To confirm isoflurane-induced APC, a minimal alveolar concentration of isoflurane of 1.0 (2.1%) was started at the end of the stabilization period and administered for 30 min, followed by 30 min of washout before coronary occlusion.

Rats were randomly assigned to one of the following groups subjected to different protocols: Sham group, non-ischemic control group of operated rats; non-ischemic control group of sham-operated rats with isoflurane; I/R group with isoflurane; I/R group without isoflurane (n = 5, respectively). To evaluate the role of ALDH2 in isoflurane-induced APC, the direct activator of ALDH2, Alda-44 (40mM) [18], was given 5 min prior to ischemia

in the groups without and with isoflurane (n = 8, respectively), and the ALDH2 inhibitor, cyanamide (5 mM) [20], was given without and with isoflurane (5 min prior to isoflurane) (n = 8, respectively). To verify that PKCe participates in the phosphorylation of ALDH2, the PKCe inhibitor, PKCe v1–2 (1mM), was given

5 min prior to ischemia without and with isoflurane (n = 8, respectively, 5 min prior to isoflurane).

After taking blood samples, the heart was removed and perfused with Langendorff apparatus for 10 min to wash out the blood. The coronary artery was re-occluded and Evans Blue was infused into the aortic root to label the normally perfused zone with a deep blue colour. The heart was sectioned into transverse slices, which were incubated with 1% trimethyl tetrazolium chloride (TTC), and photographed by a digital camera. Since TTC stains viable tissue a deep red colour, non-stained tissue was presumed to be infarcted. Area at risk (negative for Evans Blue) and infarct area (negative for TTC) were quantified using Imageproplus software (Version 4. 1, Media Cybernetics, LP, USA) and infarct size was expressed as percentage of the area at risk (infarct area/ AAR)6100 (%).

Levels of Lactate Dehydrogenase and Creatine Kinase-MB in Plasma

Serum CK-MB analysis is a widely used biomarker to detect cardiac injury. Proportionally greater serum CK-MB, relative to the total CK activity can evaluate acute myocardial injury [8]. Before removing the heart, 5 ml blood samples were taken. Serum was separated by centrifugation at 5000 g for 5 min on a tabletop centrifuge; the supernatant was stored in liquid nitrogen. The samples were thawed for analysis. Lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) were assayed using commercial kits (Roche, Germany) by an automatic analyzer 7600 (Hitachi, Japan).

Western Blotting Analysis

Upon completion of the experimental period, the myocardium and cardiomyocytes were lysed in ice-cold radioimmunoprecipi-tation assay (RIPA) lysis buffer containing 1 mmol/L phenyl-methylsulfonyl fluoride (PMSF), 1mg/ml leupeptin, 1mg/ml aprotinin and 1mg/ml pepstatin at 4uC for 15 min. The homogenate was incubated and centrifuged. The supernatant was collected and the protein concentration was determined using the bicinchoninic acid (BCA) protein assay kit according to the manufacturer’s protocol (Pierce). The detergent soluble superna-tant was frozen with liquid N2, and stored at270uC.

The supernatant was mixed with 56loading buffer and heated for 5 min at 100uC. Soluble extracts (50mg) were loaded in each lane and separated by SDS-polyacrylamide gel electrophoresis (PAGE). After electrophoresis, proteins were electrophoretically transferred to a polyvinylidene difluoride (PVDF) filter membrane (0.45mm, Gehealthcare). The membrane was blocked in Tris-buffered saline Tween-20 (TBST) with 5% non-fat milk and incubated overnight with the corresponding primary antibodies at 4uC. The membrane was then incubated for 1 h with secondary antibody (horseradish peroxidase-conjugated antirabbit IgG) diluted with TBST (1:2000). The signals of detected proteins were visualized by an enhanced chemiluminescence reaction (ECL) system (Millipore, Billerica, MA, USA). The staining was quantified by scanning the films and the band density was determined with Image-Pro software.

Adenovirus Construction and Infection

Previous studies [18,26] demonstrated that the constitutively active ALDH2 amino acid 487 must be Glu not Lys, and Thr185, Thr412 and Ser 279 must be constitutively phosphorylated. Accordingly, we obtained constitutively active mutant ALDH2 (CA-ALDH2) by nucleotide substitutions leading to the mutations Lys487Glu, Thr185Asp, Thr412Asp, and Ser279Asp introduced into the wild-type rat Aldh2 cDNA.

Short RNA hybrids (siRNAs) of 19 bp were formed by annealing two 21-mer oligoribonucleotides (Eurogentec, Belgium), each having two thymidines at their 39end. Rat-Aldh2 siRNA sequence was GCAACCAGATTCATTAATT [27]. The sense and antisense oligonucleotides were incubated together (1.5 nmol each) in 75mL

of 50 mM Tris (pH 7.5) and 100 mM NaCl for 2 min at 94uC, 5 min at 78uC and 5 min at 65uC. Finally, the annealed siRNAs were cooled to 20uC, aliquoted, and stored at280uC. The cooling transitions was carried out at a rate of 2uC/min.

Viral vectors that expressed RFP, si-ALDH2 and RFP, the constitutively active mutant of ALDH2 (ALDH2), and CA-ALDH2 and RFP were generated using the AdEasy system (Stratagene) [23]. Cardiomyocytes were prepared from ventricles and cultured in 60-mm dishes at a density of 16105cells/cm2in NCS-DMEM. After 24 h incubation at 37uC and 5% CO2, cell density reached approximately 70%. Cells were cultured overnight in 10% FBS-containing medium and infected with adenovirus for 6 h at a multiplicity of infection (MOI) of 20, then cultured in serum-free medium for an additional 24 h, before the addition of reagents.

examined using TUNEL assay for DNA fragmentation. Cardiomyoctes were photographed by fluorescence microscopy after 24 hours of hypoxia followed by 12 hours of reoxygenation. TUNEL-positive nuclei are shown in green and transfection efficiency of adenoviruses in red. (b) Quantitative analysis (percentage of apoptotic cells versus total) is shown in histogram. C. Representative Western blots of cleaved caspase-3 and full length caspase-3 (FL caspase-3) in (a), and 3 independent experiments were quantitated in (b).b-actin was used as a loading control. D. Cell lysates under each condition were quantitatively assayed for caspase-3 activity. E. LDH concentrations in cell culture media were analyzed. Values are means6 S.E.M., n = 5 in each group. *P,0.05, **P,0.01 vs. Control group, and#

(6)
(7)

Cell Culture and Hypoxia/reoxygenation Treatment

Hearts were obtained from one-day old neonatal Sprague– Dawley rats, retaining the ventricles only, and kept in cold PBS without Ca2+

and Mg2+

on ice. The ventricles were rapidly minced and dissociated with 0.1% trypsin enzyme solution. The cells released after the first digestion were discarded, whereas the cells from subsequent digestions were added to NCS-DMEM (DMEM supplemented with 20% NCS, 100 U/ml penicillin, and 100mg/ ml streptomycin). After stepwise trypsin dissociation (10 min, 4–5 times), the mixture was centrifuged (1500 r/m, 5 min). The cells were resuspended in NCS-DMEM and first transferred to tissue culture dishes for 1 h in a 37uC incubator to plate out the fibroblasts [28]. The suspended cells were then replated at a density of 16104 cells/cm22 and incubated under the same conditions as above. Bromodeoxyuridine (BrdU, 0.1 mM) was added to the medium for the first 2 days after plating to inhibit the growth of fibroblasts.

Simulated I/R was achieved by culturing the cells in 0.5% FBS DMEM in a hypoxia chamber, saturated with 5%CO2/95%N2

and supplemented with an anaerobic pouch (Mitsubishi Gas Chemical Company, Inc.) at 37uC for 24 h and following reoxygenation for 12 h using 0.5% FBS DMEM in the normal incubating condition [29]. Exposure to isoflurane was carried out by incubating the cells for 5 min in 0.5 mM isoflurane (approx-imately 1.0 minimum alveolar concentration) in 0.5% FBS DMEM. The isoflurane-containing medium was removed imme-diately before the onset of hypoxic conditions and the cells were washed with phosphate-buffered saline (PBS) [9]. Anesthetic concentrations were measured by gas chromatography (Gas chromatograph GC-8A; Shimadzu, Kyoto, Japan). Cells were grouped as follows: vector-infected with and without isoflurane, Adeasy-Si-ALDH2-treated with and without isoflurane; vector-infected with and without isoflurane, Adeasy-CA -ALDH2-treated with and without isoflurane.

Apoptosis Assay

To determine cardiomyocyte apoptosis in a quantitative manner, the in situ detection of apoptotic cardiomyocytes was performed using terminal deoxyribonucleotide transferase-medi-ated dUTP nick end labeling (TUNEL) with an in situ cell death detection kit, Fluorescein (Roche, Germany) according to the manufacturer’s protocol for cultured cells. Cells (105cells/ml) from

different treatment groups were cultured in a 6-well chamber slide and fixed in 4% paraformaldehyde followed by digestion with proteinase K (10mg/ml) for 15 min at 37uC and permeabilization

with 0.1% Triton X-100 for 5 min at 4uC. After washing twice with PBS, the cardiomyocytes were incubated with 50ml TUNEL

reaction mixture that contains TdT and fluorescein-dUTP for 1 h at 37uC. The percentage of TUNEL positive cells was determined by randomly chosen fields in each slide. In each group, at least 500 cells were counted. Sample evaluation was performed in a blinded manner, and samples from 3 independent experiments were scored per group.

Measurement of Caspase 3 Activity

The Caspase 3 Colorimetric assay kit (MBL, MA, USA) was used to measure the activity of caspase 3 according to the manufacturer’s protocol. Cells were grown in a 6 well plate. After the appropriate treatment, cells were resuspended in lysis buffer and centrifuged. The supernatant was diluted with 50 ul cell lysis buffer for each assay. Then, the reaction buffer containing DTT and DEVD-pNA substrate were added to each assay and incubated at 37uC for 2 hours. After the correct incubation time, each sample was transferred to each well in a 96 well plate, and read at 405 nm using a microplate reader. Cell lysates were also analyzed by Western blotting with an antibody (Cell Signaling, Beverly, MA, USA) which allowed detection of inactive procaspase 3 and activated cleaved caspase 3.

Statistical Analysis

Continuous values are expressed as mean6standard error of the mean (SEM). Comparisons between multiple-group means were performed using one-way analysis of variance (one-way ANOVA) and comparisons between groups were performed using the least significant difference test (LSD-test). The number of animals/group and statistical significance for all data are listed in the figures and figure legends. P values,0.05 were considered to be statistically significant. All statistical analyses were performed using SPSS version 15.0.

Results

Isoflurane Preconditioning Attenuated the Release of LDH and CK-MB and Reduced Infarct Size in vivo I/R Injury

Regional myocardial ischemia for 40 min by LAD ligation followed by 120 min of reperfusion markedly increased the leakage of LDH (Figure 1A) and CK-MB (Figure 1B) compared to sham controls. Isoflurane-induced APC significantly reduced the I/R-induced increase in LDH and CK-MB release in rat heart.

As shown in Figure 1C, regional myocardial ischemia for 40 min by LAD ligation followed by 120 min of reperfusion significantly increased myocardial infarct size compared with sham groups. Isoflurane preconditioning substantially decreased I/R-induced myocardial infarct size.

Phosphorylation of ALDH2 Participated in

Cardioprotection Induced by Isoflurane Pretreatment

Representative gels for the different treatment groups are shown in Figure 2Aa. Figure 2Ab summarizes the quantitative data on the ratio of phosphoALDH2 to total ALDH2, and shows that pretreatment with isoflurane prior to ischemia increased the phosphorylation of ALDH2. The ALDH2 inhibitor, cyanamide, significantly inhibited isoflurane-induced activation of ALDH2. The direct activator of ALDH2, Alda-44, substantially increased the phosphorylation of ALDH2, but did not enhance the phosphorylation of ALDH2 by isoflurane.

Figure 4. ALDH2 activation induced protection against hypoxia/reoxygenation.A. Schematic representation of adenoviruses encoding RFP (Ad-RFP) and CA-ALDH2 (Ad-CA-ALDH2-RFP, left panel); right panel, immunoblotting analysis of ALDH2 protein level.b-actin was used as a control. B. Expression of constitutively active mutant of ALDH2 attenuated TUNEL positive staining level, which was not reinforced by isoflurane. (a) Apoptotic cells were examined using TUNEL assay for DNA fragmentation. Cardiomyoctes were photographed by fluorescence microscopy after 24 hours of hypoxia followed by 12 hours of reoxygenation. TUNEL-positive nuclei are shown in green and transfection efficiency of adenoviruses in red. (b) Quantitative analysis (percentage of apoptotic cells versus total) is shown in histogram. C. Representative Western blots of cleaved caspase-3 and FL caspase-3 in (a), and 3 independent experiments are quantitated in (b).b-actin was used as a loading control. D. Cell lysates under each condition were quantitatively assayed for caspase-3 activity. E. LDH concentrations in cell culture media were analyzed. Values are means6S.E.M., n = 5 in each group. *P,0.05, **P,0.01 vs. Control group, and#

(8)
(9)

Consistent with ALDH2 phosphorylation, we observed that isoflurane and Alda-44 markedly attenuated I/R-induced leakage of LDH and CK-MB in plasma, as well as myocardial infarct size. However, the isoflurane-induced decrease in LDH and CK-MB release, and reduction of infarct size was significantly blocked by the ALDH2 inhibitor cyanamide (Figure 2B–D). These findings suggest that isoflurane-mediated cardioprotection is mainly mediated by activation of ALDH2.

Isoflurane Preconditioning Alleviated in vitro H/R Injury by Activation of ALDH2

To further confirm the critical role of ALDH2 in isoflurane-induced cardioprotection, we constructed an ALDH2 knockdown adenovirus and constitutively active ALDH2 mutant adenovirus. We used TUNEL, caspase 3 activity, and LDH release as quantitative assays to determine the functional significance of manipulating ALDH2 expression.

After 24 h of hypoxia followed by 12 h of reoxygenation we observed significant cardiomyocyte apoptosis demonstrated by increased DNA fragmentation using TUNEL staining, by laser scanning cytometry (LSC;) and caspase 3 activity in the vector control group. Pretreatment with isoflurane significantly inhibited the H/R-induced increase in TUNEL positive staining, caspase 3

activity and leakage of LDH (Figure 3B–E). However, when ALDH2 was downregulated (Figure 3A) in cardiomyocytes by Ad-Si-ALDH2, increased TUNEL positive staining level (Figure 3B), more intense cleaved caspase-3 staining (Figure 3C), caspase 3 activity (Figure 3D) and LDH release (Figure 3E) were observed, which supports the hypothesis that phosphorylation of ALDH2 might play a critical role in isoflurane-induced cardioprotection. Immunoblotting analysis showed a substantial increase in ALDH2 level in Ad-CA-ALDH2-RFP-infected cells compared to Ad-RFP (Figure 4A). Constitutively active ALDH2 significantly inhibited the H/R-induced increase in TUNEL positive staining, caspase 3 activity and leakage of LDH (Figure 4 B–E), which were not further increased by isoflurane treatment.

PKCeis Involved in Isoflurane-induced Phosphorylation of ALDH2 and Cardioprotection

PKCetranslocation to mitochondria and then phosphorylation of ALDH2 is required to protect the heart from I/R injury. Here we demonstrate that pretreatment with isoflurane resulted in elevated mitochondrial levels of PKCe accompanied by phos-phorylation of ALDH2. Isoflurane-induced phosphos-phorylation of ALDH2 was inhibited by the PKCe inhibitor, PKCe V1–2. Because mitochondrial translocation of PKCeoccurs rapidly, with a corresponding decline in cytosolic PKCelevels, and because the total cellular PKCe levels do not change (Figure 5A), our data suggest that isoflurane enables dynamic mitochondrial transloca-tion of PKCe in response to I/R. Consistent with PKCe translocation to mitochondria, PKCe V1–2 had a detrimental effect on isoflurane-induced attenuation of LDH and CK-MB leakage (Figure 5B, 5C), and the decrease in myocardial infarct size (Figure 5D).

Discussion

In the present study, we observed that (1) isoflurane pretreat-ment reduced I/R injury in vivo and stimulated H/R insult in vitro associated with phosphorylation of ALDH2; (2) isoflurane-induced phosphorylation of ALDH2 and cardioprotection was mediated by PKCe translocation from the cytosol to mitochon-dria. Thus, phosphorylation of ALDH2 is critical for the cardioprotective effects of isoflurane preconditioning.

Volatile anesthetics have a long history in the clinical management of anesthesia. Consistent with our results, numerous studies have shown that volatile anesthetics can protect the myocardium when applied before a harmful ischemic event and at the beginning of reperfusion, and that the characteristics of this protection are similar to those observed during classic IPC. Studies have attempted to characterize the mechanisms involved. Cardio-protective mechanisms produced by APC were shown to involve activation of phosphoinositide 3-kinase [25], extracellular regulat-ed kinases 1 and 2 (ERK1/2) [30], the 70-kDa ribosomal protein S6 kinase, endothelial ROS (eNOS) [31], mitochondrial KATP

channels [3,14,15] and inhibition of glycogen synthase kinase 3-b [32], but the precise mechanism responsible for APC remains undefined. However, it is unlikely that stimulation of pro-survival Figure 5. PKCe translocation is involved in isoflurane preconditioning. A PKCe translocation was associated with isoflurane-induced phosphorylation of ALDH2. (a) Representative of western blot analysis of phos-ALDH2, total ALDH2,b-actin, mitochondria PKCe(mito-PKCe) and total PKCe(from top lanes to bottom lanes).b-actin was used to demonstrate equal protein loading. (b) Quantification of the phos-ALDH2, normalized to the total ALDH2, and PKCetranslocation to the mitochondria from 3 different experiments. B, C. Isoflurane-induced inhibition of LDH and CK-MB release by I/R was restored by PKCev1–2. Serum LDH and CK-MB concentrations were analyzed. D. PKCev1–2 inhibits the decrease in heart infarct size caused by isoflurane following I/R. Representative cross-sectional slices derived from a single heart. The infarct size normalized to the area at risk. Values are means6S.E.M., n = 8 in each group. *P,0.05, **P,0.01 vs. the saline control group and#P

,0.05,##P

,0.01 with PKCev1–2 vs. the corresponding group without PKCev1–2.

doi:10.1371/journal.pone.0052469.g005

Figure 6. Hypothetical scheme demonstrate that the phos-phorylation of ALDH2 through mitochondrial translocation of PKCe plays an important role in the cardioprotection of isoflurane preconditioning in myocardium I/R injury.Ischemic preconditioning/postconditioning and pharmacological agents result in the activation of the RISK pathway, which lead to the phosphorylation and mitochondrial translocation of PKCe.

(10)

signaling pathways occurs rapidly enough to prevent damage resulting from the initial injury during reperfusion. Recently, attention has focused on mitochondria as a target of cardioprotec-tion by volatile anesthetics [11,33,34].

Mitochondria are essential for cell survival and play important roles in the complex signaling pathways leading to cardioprotec-tion by volatile anesthetics, and in the produccardioprotec-tion of adenosine triphosphate and the regulation of cell death [35]. The mecha-nisms by which isoflurane ultimately limits infarct size are not known. Apoptosis and inflammation have been implicated in cardiac I/R injury [36–39]. In agreement with our results, isoflurane-treated mice subjected to ischemia and 2 weeks of reperfusion showed reduced expression of proapoptotic genes, significantly decreased expression of cleaved caspase-3, and TUNEL staining [5].

ALDH2 is best known for its role in metabolizing the ethanol intermediate, acetaldehyde. These highly toxic, reactive aldehydes can create aldehydic adducts with proteins, causing protein dysfunction and tissue injury, and have been linked to various diseases, such as cancer and MI, in humans [40]. It has been reported that overexpression of the ALDH2 transgene may alleviate I/R injury, post-I/R and ischemic ventricular dysfunc-tion [21,41]. Consistent with this, I/R injury may be exacerbated by ALDH2 knockout [23,41]. These data support our results that ALDH2 plays an essential role in isoflurane-induced cardioprotec-tion against I/R injury. It was shown that overexpression of ALDH2 significantly attenuated acetaldehyde and ethanol-in-duced oxidative stress (ROS generation), activation of stress signal molecules and apoptosis in fetal human cardiac myocytes [21]. Here, we showed that isoflurane preconditioning increased the phosphorylation of ALDH2. These data also support our notion that isoflurane pretreatment attenuated I/R-induced apoptosis which is associated with phosphorylation and activation of ALDH2.

We observed that isoflurane pretreatment led to PKCe translocation to mitochondria. Although phosphorylation and translocation of PKC are thought to be pivotal steps in cardioprotection by IPC, and PKCeseems to play a critical role in the signaling cascade underlying preconditioning [11,16], there are few data suggesting the involvement of PKC in APC [11,42]. It has been shown that phosphorylation and translocation of PKCedepends on the concentration of the volatile anesthetic and that alternative pathways may exist at higher concentrations [30]. Recent studies reported that PKCetargeted the inner

mitochon-drial membrane and phosphorylated a number of intra-mitochon-drial proteins [18,43,44]. Mitochonintra-mitochon-drial ALDH2 has been identified as a PKCe substrate, whose activity correlates with cardioprotection against I/R [18,20]. Our results showed that isoflurane-induced ALDH2 activation was accompanied by translocation of PKCe from the cytosolic to the mitochondria fraction, which was inhibited by the PKCeinhibitor. It was shown that ERK1/2 blockade abolished PKCe activation, suggesting ERK pathway was involved in activation of PKCe, during desflurane-induced preconditioning [30]. Opening of mitochon-drial adenosine triphosphate-sensitive potassium channels and generation of reactive oxygen species were upstream events of PKCe activation in isoflurane-induced preconditioning [45]. Activation of ALDH2 can attenuate ROS production [21], indicating that ALDH2 might be a critical mediator of isoflur-ane-induced protection. Further research needs to be carried out to identify the ALDH2 mechanism in mitochondria.

Although polymorphism in ALDH2 gene is an independent risk factor for myocardial infarction [46,47], a recent study showed that inhibited ALDH2 activity during cardiac surgery got less I/R injury and better cardiac function [48]. The contradicted clinical results might need larger sample and stronger evidence to testify. However, from point of view of clinical application of APC in the future, in patients with lower Aldh2 activity who experience an ischemic event, the use of isoflurane may need to be reconsidered. In summary, our results demonstrate that isoflurane precondi-tioning increased the phosphorylation of mitochondrial ALDH2 which was mediated by mitochondrial PKCeand is required for cardiac protection against I/R (Figure 6). This work suggests a possible mechanism by which isoflurane can access cytoprotective substrates located within the mitochondria to confer cardioprotec-tion [49]. Our data provide an insight into the mitochondrial-dependent basis of isoflurane-induced, and PKCeand ALDH2-mediated protection against cardiac ischemia, in vivo and in vitro [19]. The current study extends our understanding of APC cardiac protection, which is relevant for extrapolation to the clinic.

Author Contributions

Conceived and designed the experiments: X-EL. Performed the experi-ments: X-EL XW K-RZ J-HJ. Analyzed the data: X-EL XW J-YL. Contributed reagents/materials/analysis tools: X-EL Q-SL XW K-RZ J-YL J-HJ. Wrote the paper: X-EL.

References

1. Keeley EC, Boura JA, Grines CL (2003) Primary angioplasty versus intravenous thrombolytic therapy for acute myocardial infarction: a quantitative review of 23 randomised trials. Lancet 361: 13–20.

2. Murry CE, Jennings RB, Reimer KA (1986) Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation 74: 1124–1136. 3. Kersten JR, Schmeling TJ, Pagel PS, Gross GJ, Warltier DC (1997) Isoflurane mimics ischemic preconditioning via activation of K(ATP) channels: reduction of myocardial infarct size with an acute memory phase. Anesthesiology 87: 361– 370.

4. Cason BA, Gamperl AK, Slocum RE, Hickey RF (1997) Anesthetic-induced preconditioning: previous administration of isoflurane decreases myocardial infarct size in rabbits. Anesthesiology 87: 1182–1190.

5. Tsutsumi YM, Patel HH, Lai NC, Takahashi T, Head BP, et al. (2006) Isoflurane produces sustained cardiac protection after ischemia-reperfusion injury in mice. Anesthesiology 104: 495–502.

6. Toller WG, Kersten JR, Pagel PS, Hettrick DA, Warltier DC (1999) Sevoflurane reduces myocardial infarct size and decreases the time threshold for ischemic preconditioning in dogs. Anesthesiology 91: 1437–1446.

7. Penta de PA, Polisca P, Tomai F, De PR, Turani F, et al. (1999) Recovery of LV contractility in man is enhanced by preischemic administration of enflurane. Ann Thorac Surg 68: 112–118.

8. De Hert SG, ten Broecke PW, Mertens E, Van Sommeren EW, Stockman BA, et al. (2002) Sevoflurane but not propofol preserves myocardial function in coronary surgery patients. Anesthesiology 97: 42–49.

9. Pravdic D, Mio Y, Sedlic F, Pratt PF, Warltier DC, et al. (2010) Isoflurane protects cardiomyocytes and mitochondria by immediate and cytosol-indepen-dent action at reperfusion. Br J Pharmacol 160: 220–232.

10. Novalija E, Kevin LG, Camara AK, Bosnjak ZJ, Kampine JP, et al. (2003) Reactive oxygen species precede the epsilon isoform of protein kinase C in the anesthetic preconditioning signaling cascade. Anesthesiology 99: 421–428. 11. Uecker M, Da SR, Grampp T, Pasch T, Schaub MC, et al. (2003) Translocation

of protein kinase C isoforms to subcellular targets in ischemic and anesthetic preconditioning. Anesthesiology 99: 138–147.

12. Tanaka K, Weihrauch D, Kehl F, Ludwig LM, LaDisa JF, et al. (2002) Mechanism of preconditioning by isoflurane in rabbits: a direct role for reactive oxygen species. Anesthesiology 97: 1485–1490.

13. Mullenheim J, Ebel D, Frassdorf J, Preckel B, Thamer V, et al. (2002) Isoflurane preconditions myocardium against infarction via release of free radicals. Anesthesiology 96: 934–940.

14. Pain T, Yang XM, Critz SD, Yue Y, Nakano A, et al. (2000) Opening of mitochondrial K(ATP) channels triggers the preconditioned state by generating free radicals. Circ Res 87: 460–466.

(11)

triphosphate-regulated potassium (KATP) channels. Anesthesiology 85: 794– 807.

16. Liu GS, Cohen MV, Mochly-Rosen D, Downey JM (1999) Protein kinase C-epsilon is responsible for the protection of preconditioning in rabbit cardiomyocytes. J Mol Cell Cardiol 31: 1937–1948.

17. Dorn GW, Souroujon MC, Liron T, Chen CH, Gray MO, et al. (1999) Sustained in vivo cardiac protection by a rationally designed peptide that causes epsilon protein kinase C translocation. Proc Natl Acad Sci U S A 96: 12798– 12803.

18. Chen CH, Budas GR, Churchill EN, Disatnik MH, Hurley TD, et al. (2008) Activation of aldehyde dehydrogenase-2 reduces ischemic damage to the heart. Science 321: 1493–1495.

19. Churchill EN, Disatnik MH, Mochly-Rosen D (2009) Time-dependent and ethanol-induced cardiac protection from ischemia mediated by mitochondrial translocation of varepsilonPKC and activation of aldehyde dehydrogenase 2. J Mol Cell Cardiol 46: 278–284.

20. Budas GR, Disatnik MH, Chen CH, Mochly-Rosen D (2010) Activation of aldehyde dehydrogenase 2 (ALDH2) confers cardioprotection in protein kinase C epsilon (PKCe) knockout mice. J Mol Cell Cardiol 48: 757–764. 21. Li SY, Li Q, Shen JJ, Dong F, Sigmon VK, et al. (2006) Attenuation of

acetaldehyde-induced cell injury by overexpression of aldehyde dehydrogenase-2 (ALDH2) transgene in human cardiac myocytes: role of MAP kinase signaling. J Mol Cell Cardiol 40: 283–294.

22. Sun L, Ferreira JC, Mochly-Rosen D (2011) ALDH2 Activator Inhibits Increased Myocardial Infarction Injury by Nitroglycerin Tolerance. Sci Transl Med 3: 107ra111.

23. Endo J, Sano M, Katayama T, Hishiki T, Shinmura K, et al. (2009) Metabolic remodeling induced by mitochondrial aldehyde stress stimulates tolerance to oxidative stress in the heart. Circ Res 105: 1118–1127.

24. Lagranha CJ, Deschamps A, Aponte A, Steenbergen C, Murphy E (2010) Sex differences in the phosphorylation of mitochondrial proteins result in reduced production of reactive oxygen species and cardioprotection in females. Circ Res 106: 1681–1691.

25. Raphael J, Rivo J, Gozal Y (2005) Isoflurane-induced myocardial precondition-ing is dependent on phosphatidylinositol-3-kinase/Akt signallprecondition-ing. Br J Anaesth 95: 756–763.

26. Kimura M, Kimura S, Matsushita S, Kashima H, Higuchi S (2006) ALDH2 promoter polymorphism has no effect on the risk for alcoholism. Alcohol Alcohol 41: 368–371.

27. Cortinez G, Sapag A, Israel Y (2009) RNA interference against aldehyde dehydrogenase-2: development of tools for alcohol research. Alcohol 43: 97–104. 28. Singh KK, Shukla PC, Quan A, Lovren F, Pan Y, et al. (2011) Herceptin, a recombinant humanized anti-ERBB2 monoclonal antibody, induces cardiomy-ocyte death. Biochem Biophys Res Commun 411: 421–426.

29. Park M, Youn B, Zheng XL, Wu D, Xu A, et al. (2011) Globular adiponectin, acting via AdipoR1/APPL1, protects H9c2 cells from hypoxia/reoxygenation-induced apoptosis. PLoS One 6: e19143.

30. Toma O, Weber NC, Wolter JI, Obal D, Preckel B, et al. (2004) Desflurane preconditioning induces time-dependent activation of protein kinase C epsilon and extracellular signal-regulated kinase 1 and 2 in the rat heart in vivo. Anesthesiology 101: 1372–1380.

31. Lamberts RR, Onderwater G, Hamdani N, Vreden MJ, Steenhuisen J, et al. (2009) Reactive oxygen species-induced stimulation of 59AMP-activated protein kinase mediates sevoflurane-induced cardioprotection. Circulation 120: S10– S15.

32. Bouwman RA, Vreden MJ, Hamdani N, Wassenaar LE, Smeding L, et al. (2010) Effect of bupivacaine on sevoflurane-induced preconditioning in isolated rat hearts. Eur J Pharmacol 647: 132–138.

33. Mio Y, Bienengraeber MW, Marinovic J, Gutterman DD, Rakic M, et al. (2008) Age-related attenuation of isoflurane preconditioning in human atrial cardio-myocytes: roles for mitochondrial respiration and sarcolemmal adenosine triphosphate-sensitive potassium channel activity. Anesthesiology 108: 612–620. 34. Ljubkovic M, Mio Y, Marinovic J, Stadnicka A, Warltier DC, et al. (2007) Isoflurane preconditioning uncouples mitochondria and protects against hypoxia-reoxygenation. Am J Physiol Cell Physiol 292: C1583–C1590. 35. Hu ZY, Liu J (2009) Mechanism of cardiac preconditioning with volatile

anaesthetics. Anaesth Intensive Care 37: 532–538.

36. Suzuki K, Murtuza B, Smolenski RT, Sammut IA, Suzuki N, et al. (2001) Overexpression of interleukin-1 receptor antagonist provides cardioprotection against ischemia-reperfusion injury associated with reduction in apoptosis. Circulation 104: I308–I3I3.

37. Fliss H, Gattinger D (1996) Apoptosis in ischemic and reperfused rat myocardium. Circ Res 79: 949–956.

38. Gottlieb RA, Burleson KO, Kloner RA, Babior BM, Engler RL (1994) Reperfusion injury induces apoptosis in rabbit cardiomyocytes. J Clin Invest 94: 1621–1628.

39. Meldrum DR, Dinarello CA, Shames BD, Cleveland JC, Banerjee A, et al. (1998) Ischemic preconditioning decreases postischemic myocardial tumor necrosis factor-alpha production. Potential ultimate effector mechanism of preconditioning. Circulation 98: II214–II218.

40. Chen CH, Sun L (2010) Mochly-Rosen D. Mitochondrial aldehyde dehydro-genase and cardiac diseases. Cardiovasc Res 88: 51–57.

41. Ma H, Guo R, Yu L, Zhang Y, Ren J (2011) Aldehyde dehydrogenase 2 (ALDH2) rescues myocardial ischaemia/reperfusion injury: role of autophagy paradox and toxic aldehyde. Eur Heart J 32: 1025–1038.

42. Obal D, Weber NC, Zacharowski K, Toma O, Dettwiler S, et al. (2005) Role of protein kinase C-epsilon (PKCepsilon) in isoflurane-induced cardioprotection. Br J Anaesth 94: 166–173.

43. Ogbi M, Chew CS, Pohl J, Stuchlik O, Ogbi S, et al. (2004) Cytochrome c oxidase subunit IV as a marker of protein kinase Cepsilon function in neonatal cardiac myocytes: implications for cytochrome c oxidase activity. Biochem J 382: 923–932.

44. Baines CP, Zhang J, Wang GW, Zheng YT, Xiu JX, et al. (2002) Mitochondrial PKCepsilon and MAPK form signaling modules in the murine heart: enhanced mitochondrial PKCepsilon-MAPK interactions and differential MAPK activa-tion in PKCepsilon-induced cardioprotecactiva-tion. Circ Res 90: 390–397. 45. Dorn GW 2nd, Souroujon MC, Liron T, Chen CH, Gray MO, et al. (1999)

Sustained in vivo cardiac protection by a rationally designed peptide that causes epsilon protein kinase C translocation. Proc Natl Acad Sci U S A 96: 12798– 12803.

46. Xu F, Chen YG, Geng YJ, Zhang H, Jiang CX, et al. (2007) The polymorphism in acetaldehyde dehydrogenase 2 gene, causing a substitution of Glu.Lys (504), is not associated with coronary atherosclerosis severity in Han Chinese. Tohoku J Exp Med 213: 215–220.

47. Bian Y, Chen YG, Xu F, Xue L, Ji WQ, et al. (2010) The polymorphism in aldehyde dehydrogenase-2 gene is associated with elevated plasma levels of high-sensitivity C-reactive protein in the early phase of myocardial infarction. Tohoku J Exp Med 221: 107–112.

48. Zhang H, Gong DX, Zhang YJ, Li SJ, Hu S (2012) Effect of mitochondrial aldehyde dehydrogenase-2 genotype on cardioprotection in patients with congenital heart disease. Eur Heart J 33: 1606–1614.

Referências

Documentos relacionados

We observed lipid peroxidation and NF-κB activation in soleus muscle, accompanied by increased plasma levels of IL-6 and reduced levels of IL-10, following a single bout of

While overexpression of ECM regulators by tumor cells confers aggressiveness to SqCC and AD, overexpression of growth factors and ECM regulators by stroma cells confers a

Knockdown of the taurine transporter (TAUT) or treatment with the ERK-specific inhibitor PD98059 (10 μM) blocked the activation of ERK induced by taurine (1 0 mM) and abolished

Cardioprotection did not reduce infarct size in the hypercholesterolemic animals (pre: 13.55% and post: 8% compared to 7.95% observed in the group subjected only to ischemia

We observed lipid peroxidation and NF-κB activation in soleus muscle, accompanied by increased plasma levels of IL-6 and reduced levels of IL-10, following a single bout of

Results: Alprostadil treatment significantly reduced myocardial infarct size, serum troponin T levels, and CK-MB and LDH activity (P<0.05).. Furthermore, treatment with

Background and objectives: The aim of this study was to evaluate the effects of remote ischemic preconditioning by brief ischemia of unilateral hind limb when combined

In 1.5 and 2.5% isoflurane exposure groups, the decay time of OPS was significantly prolonged and the ratio of OPS recovery and the amplitude of recovery were increased as compared