Overexpression of CYP2J2 provides protection against doxorubicin-induced cardiotoxicity

10
Overexpression of CYP2J2 provides protection against doxorubicin-induced cardiotoxicity Yunfang Zhang, 1 Haitham El-Sikhry, 1 Ketul R. Chaudhary, 1 Sri Nagarjun Batchu, 1 Anooshirvan Shayeganpour, 1 Taibeh Orujy Jukar, 1 J. Alyce Bradbury, 2 Joan P. Graves, 2 Laura M. DeGraff, 2 Page Myers, 2 Douglas C. Rouse, 2,4 Julie Foley, 2 Abraham Nyska, 2,3 Darryl C. Zeldin, 2 and John M. Seubert 1 1 Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; 2 Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; 3 Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; and 4 Duke University Medical Center, Durham, North Carolina Submitted 9 September 2008; accepted in final form 29 April 2009 Zhang Y, El-Sikhry H, Chaudhary KR, Batchu SN, Shayegan- pour A, Jukar TO, Bradbury JA, Graves JP, DeGraff LM, Myers P, Rouse DC, Foley J, Nyska A, Zeldin DC, Seubert JM. Overexpression of CYP2J2 provides protection against doxorubicin-induced cardiotoxic- ity. Am J Physiol Heart Circ Physiol 297: H37–H46, 2009. First pub- lished May 8, 2009; doi:10.1152/ajpheart.00983.2008.—Human cyto- chrome P-450 (CYP)2J2 is abundant in heart and active in bio- synthesis of epoxyeicosatrienoic acids (EETs). Recently, we demonstrated that these eicosanoid products protect myocardium from ischemia-reperfusion injury. The present study utilized trans- genic (Tr) mice with cardiomyocyte-specific overexpression of human CYP2J2 to investigate protection toward toxicity resulting from acute (0, 5, or 15 mg/kg daily for 3 days, followed by 24-h recovery) or chronic (0, 1.5, or 3.0 mg/kg biweekly for 5 wk, followed by 2-wk recovery) doxorubicin (Dox) administration. Acute treatment re- sulted in marked elevations of serum lactate dehydrogenase and creatine kinase levels that were significantly greater in wild-type (WT) than CYP2J2 Tr mice. Acute treatment also resulted in less activation of stress response enzymes in CYP2J2 Tr mice (catalase 750% vs. 300% of baseline, caspase-3 235% vs. 165% of baseline in WT vs. CYP2J2 Tr mice). Moreover, CYP2J2 Tr hearts exhib- ited less Dox-induced cardiomyocytes apoptosis (measured by TUNEL) compared with WT hearts. After chronic treatment, comparable decreases in body weight were observed in WT and CYP2J2 Tr mice. However, cardiac function, assessed by measure- ment of fractional shortening with M-mode transthoracic echocar- diography, was significantly higher in CYP2J2 Tr than WT hearts after chronic Dox treatment (WT 37 2%, CYP2J2 Tr 47 1%). WT mice also had larger increases in -myosin heavy chain and cardiac ankryin repeat protein compared with CYP2J2 Tr mice. CYP2J2 Tr hearts had a significantly higher rate of Dox metabo- lism than WT hearts (2.2 0.25 vs. 1.6 0.50 ng min 1 100 g protein 1 ). In vitro data from H9c2 cells demonstrated that EETs attenuated Dox-induced mitochondrial damage. Together, these data suggest that cardiac-specific overexpression of CYP2J2 lim- ited Dox-induced toxicity. cytochrome P-450 2J2; heart; function CYTOCHROME P-450 (CYP) epoxygenases are predominant en- zymes responsible for the epoxidation of endogenous arachi- donic acid (AA) to four regioisomeric epoxyeicosatrienoic acids (5,6-, 8,9-, 11,12-, and 14,15-EETs). These epoxy fatty acid products have an important role in cellular signaling and possess numerous biological activities in the cardiovascular system (54). The actions of EETs are terminated by conversion to the corresponding and less biologically active dihydroxyei- cosatrienoic acids (DHETs) by epoxide hydrolases (41). The role of EETs in cardiovascular homeostasis and protection against ischemia-reperfusion injury has been demonstrated in both rodent and dog (19, 48). While a protective role in ischemia-reperfusion injury is known, it remains to be inves- tigated whether EETs mediate myocardial protection against other pathological stressors. Doxorubicin (Dox, Adriamycin) is a quinone-containing anthracycline antibiotic commonly used in treatment of a variety of human neoplastic diseases and solid cancers. Its clinical use is limited by irreversible cardiomyopathy and heart failure (11, 39). Dox-induced cardiotoxicity can be characterized by acute myocardial injury, which occurs immediately after an initial treatment. Alternatively, chronic cardiotoxicity, which can lead to conditions such as conges- tive heart failure, ventricular dysfunction, and arrhythmia, may occur years to decades after treatment (36, 47, 55). It is well established that the adverse effect of Dox treatment is mediated by mechanisms distinct from its therapeutic mode of action, which involves interacting with DNA by interca- lation and thereby inhibiting macromolecular biosynthesis (18). Dox-induced cardiotoxicity results from a series of adverse reactions involving increased oxidative stress, al- teration in cellular energetics, and initiation of cell death pathways (39, 57, 59). Numerous reports have demonstrated moderate protection against Dox-induced cardiotoxicity, yet the exact mechanism(s) and optimal therapy remain un- known (3, 6, 7, 9, 31). Recently, we reported (48, 50) that enhanced postischemic recovery of left ventricular (LV) function and reduced cardiac injury are found in transgenic (Tr) mice that overexpress human CYP2J2 and in mice with targeted deletion of soluble epoxide hydrolase. Whether the overexpression of CYP2J2 can protect against Dox-induced cardiotoxicity has not been inves- tigated. To examine the role of human CYP2J2 isoform in a model of Dox-induced toxicity we utilized Tr mice with cardiac-specific overexpression of CYP2J2. Results of these studies indicate that CYP2J2 overexpression provided signifi- cant protection in both an acute and a chronic model of Dox-induced injury. Address for reprint requests and other correspondence: J. M. Seubert, 3126 Dentistry/Pharmacy Centre, Faculty of Pharmacy and Pharmaceutical Sci- ences, Univ. of Alberta, Edmonton, AB, Canada T6G 2N8 (e-mail: jseubert @pharmacy.ualberta.ca). Am J Physiol Heart Circ Physiol 297: H37–H46, 2009. First published May 8, 2009; doi:10.1152/ajpheart.00983.2008. http://www.ajpheart.org H37

Transcript of Overexpression of CYP2J2 provides protection against doxorubicin-induced cardiotoxicity

Overexpression of CYP2J2 provides protection against doxorubicin-inducedcardiotoxicity

Yunfang Zhang,1 Haitham El-Sikhry,1 Ketul R. Chaudhary,1 Sri Nagarjun Batchu,1

Anooshirvan Shayeganpour,1 Taibeh Orujy Jukar,1 J. Alyce Bradbury,2 Joan P. Graves,2

Laura M. DeGraff,2 Page Myers,2 Douglas C. Rouse,2,4 Julie Foley,2 Abraham Nyska,2,3 Darryl C. Zeldin,2

and John M. Seubert1

1Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; 2Division ofIntramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research TrianglePark, North Carolina; 3Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; and 4Duke University MedicalCenter, Durham, North Carolina

Submitted 9 September 2008; accepted in final form 29 April 2009

Zhang Y, El-Sikhry H, Chaudhary KR, Batchu SN, Shayegan-pour A, Jukar TO, Bradbury JA, Graves JP, DeGraff LM, Myers P,Rouse DC, Foley J, Nyska A, Zeldin DC, Seubert JM. Overexpressionof CYP2J2 provides protection against doxorubicin-induced cardiotoxic-ity. Am J Physiol Heart Circ Physiol 297: H37–H46, 2009. First pub-lished May 8, 2009; doi:10.1152/ajpheart.00983.2008.—Human cyto-chrome P-450 (CYP)2J2 is abundant in heart and active in bio-synthesis of epoxyeicosatrienoic acids (EETs). Recently, wedemonstrated that these eicosanoid products protect myocardiumfrom ischemia-reperfusion injury. The present study utilized trans-genic (Tr) mice with cardiomyocyte-specific overexpression of humanCYP2J2 to investigate protection toward toxicity resulting from acute(0, 5, or 15 mg/kg daily for 3 days, followed by 24-h recovery) orchronic (0, 1.5, or 3.0 mg/kg biweekly for 5 wk, followed by 2-wkrecovery) doxorubicin (Dox) administration. Acute treatment re-sulted in marked elevations of serum lactate dehydrogenase andcreatine kinase levels that were significantly greater in wild-type(WT) than CYP2J2 Tr mice. Acute treatment also resulted in lessactivation of stress response enzymes in CYP2J2 Tr mice (catalase750% vs. 300% of baseline, caspase-3 235% vs. 165% of baselinein WT vs. CYP2J2 Tr mice). Moreover, CYP2J2 Tr hearts exhib-ited less Dox-induced cardiomyocytes apoptosis (measured byTUNEL) compared with WT hearts. After chronic treatment,comparable decreases in body weight were observed in WT andCYP2J2 Tr mice. However, cardiac function, assessed by measure-ment of fractional shortening with M-mode transthoracic echocar-diography, was significantly higher in CYP2J2 Tr than WT heartsafter chronic Dox treatment (WT 37 � 2%, CYP2J2 Tr 47 � 1%).WT mice also had larger increases in �-myosin heavy chain andcardiac ankryin repeat protein compared with CYP2J2 Tr mice.CYP2J2 Tr hearts had a significantly higher rate of Dox metabo-lism than WT hearts (2.2 � 0.25 vs. 1.6 � 0.50 ng �min�1 �100 �gprotein�1). In vitro data from H9c2 cells demonstrated that EETsattenuated Dox-induced mitochondrial damage. Together, thesedata suggest that cardiac-specific overexpression of CYP2J2 lim-ited Dox-induced toxicity.

cytochrome P-450 2J2; heart; function

CYTOCHROME P-450 (CYP) epoxygenases are predominant en-zymes responsible for the epoxidation of endogenous arachi-donic acid (AA) to four regioisomeric epoxyeicosatrienoicacids (5,6-, 8,9-, 11,12-, and 14,15-EETs). These epoxy fatty

acid products have an important role in cellular signaling andpossess numerous biological activities in the cardiovascularsystem (54). The actions of EETs are terminated by conversionto the corresponding and less biologically active dihydroxyei-cosatrienoic acids (DHETs) by epoxide hydrolases (41). Therole of EETs in cardiovascular homeostasis and protectionagainst ischemia-reperfusion injury has been demonstrated inboth rodent and dog (19, 48). While a protective role inischemia-reperfusion injury is known, it remains to be inves-tigated whether EETs mediate myocardial protection againstother pathological stressors.

Doxorubicin (Dox, Adriamycin) is a quinone-containinganthracycline antibiotic commonly used in treatment of avariety of human neoplastic diseases and solid cancers. Itsclinical use is limited by irreversible cardiomyopathy andheart failure (11, 39). Dox-induced cardiotoxicity can becharacterized by acute myocardial injury, which occursimmediately after an initial treatment. Alternatively, chroniccardiotoxicity, which can lead to conditions such as conges-tive heart failure, ventricular dysfunction, and arrhythmia,may occur years to decades after treatment (36, 47, 55). It iswell established that the adverse effect of Dox treatment ismediated by mechanisms distinct from its therapeutic modeof action, which involves interacting with DNA by interca-lation and thereby inhibiting macromolecular biosynthesis(18). Dox-induced cardiotoxicity results from a series ofadverse reactions involving increased oxidative stress, al-teration in cellular energetics, and initiation of cell deathpathways (39, 57, 59). Numerous reports have demonstratedmoderate protection against Dox-induced cardiotoxicity, yetthe exact mechanism(s) and optimal therapy remain un-known (3, 6, 7, 9, 31).

Recently, we reported (48, 50) that enhanced postischemicrecovery of left ventricular (LV) function and reduced cardiacinjury are found in transgenic (Tr) mice that overexpresshuman CYP2J2 and in mice with targeted deletion of solubleepoxide hydrolase. Whether the overexpression of CYP2J2 canprotect against Dox-induced cardiotoxicity has not been inves-tigated. To examine the role of human CYP2J2 isoform in amodel of Dox-induced toxicity we utilized Tr mice withcardiac-specific overexpression of CYP2J2. Results of thesestudies indicate that CYP2J2 overexpression provided signifi-cant protection in both an acute and a chronic model ofDox-induced injury.

Address for reprint requests and other correspondence: J. M. Seubert, 3126Dentistry/Pharmacy Centre, Faculty of Pharmacy and Pharmaceutical Sci-ences, Univ. of Alberta, Edmonton, AB, Canada T6G 2N8 (e-mail: [email protected]).

Am J Physiol Heart Circ Physiol 297: H37–H46, 2009.First published May 8, 2009; doi:10.1152/ajpheart.00983.2008.

http://www.ajpheart.org H37

MATERIALS AND METHODS

Animals. Mice with cardiomyocyte-specific overexpression of hu-man CYP2J2 and their wild-type (WT) littermate controls wereutilized (48). All experiments used male and female mice aged 3–5mo and weighing 25–35 g and were approved by the National Instituteof Environmental Health Sciences/National Institutes of Health andUniversity of Alberta Animal Care and Use Committees.

Treatment protocols. In the acute protocol, mice were randomlydivided into three groups and received 0, 5, or 15 mg/kg Dox byintraperitoneal injections (Fig. 1A). Mice were treated with a singledose each day for 3 days (0, 24, and 48 h) and were killed by CO2

asphyxiation on the fourth day (72 h). Hearts were either isolated andperfused in the Langendorff mode to assess heart function (see below)or collected for histological and biochemical analyses.

In the chronic protocol, mice were randomly divided into threegroups and received 0, 1.5, or 3.0 mg/kg Dox by intraperitonealinjections (Fig. 1B). Dox was administered twice a week for 5 wk fora total of 10 treatments. A 2-wk “washout” period was allowed afterthe last treatment, at which point cardiac function was assessed byechocardiography. Mice were then killed by CO2 asphyxiation, andcardiac specimens were analyzed. All studies were conducted byinvestigators who were blinded to treatment group assignments.

Biochemical analyses. At the end of each protocol, blood wasdrawn from the inferior vena cava to assess levels of lactate dehydro-genase (LDH) and creatine kinase (CK). Serum was collected within2 h from clotted blood by centrifugation and analyzed with end pointassay kits (Sigma Diagnostics, St. Louis, MO). LDH and CK activitieswere expressed as units per liter. Subcellular fractions were preparedby differential centrifugation from frozen hearts as described previ-ously (15). Catalase activity was measured with a spectrophotometricassay that monitored H2O2 disappearance at 240 nm and used themolar extinction coefficient 43.6 M�1cm�1 (2, 15). Caspase-3 activ-ity was assessed with a spectrofluorometric assay as previouslydescribed (49). Briefly, caspase-3 activity was determined in cytosolicfractions by monitoring the release of 7-amino-4-methylcoumarin(AMC) by proteolytic cleavage of the peptide Ac-DEVD-AMC (20

�M) (Sigma-Aldrich, Oakville, ON, Canada). Fluorescence was mon-itored at wavelengths of 380 nm (excitation) and 460 nm (emission).Specific activities were determined to be within the linear range of astandard curve established with AMC. Protein quantities were deter-mined with a Bradford protein assay kit (Bio-Rad Laboratories).

Isolated, perfused hearts. Hearts were perfused in the Langendorffmode as described previously (48, 50). Hearts from CYP2J2 Tr andage-/sex-matched WT littermate control animals were perfused in aretrograde fashion at constant pressure (90 cmH2O) with continuouslyaerated (95% O2-5% CO2) Krebs-Henseleit buffer at 37°C. Heartswere perfused for 40 min (stabilization) and then subjected to 20 minof global no-flow ischemia, followed by 40 min of reperfusion.Recovery of contractile function was taken as LV developed pressure(LVDP) at the end of reperfusion expressed as a percentage ofpreischemic LVDP.

Transthoracic echocardiography. Two-dimensional M-mode echo-cardiography was performed with a Vevo 770 high-resolution imag-ing system (VisualSonics) as described previously (48, 50). LVend-diastolic dimension (LVDd, mm) and LV end-systolic dimension(LVDs, mm) were assessed. Fractional shortening (FS) of the LV wasexpressed as %FS � [(LVDd � LVDs)/LVDd] � 100.

Histology and gene expression. Histological analyses were per-formed on hearts from both CYP2J2 Tr and WT mice as previouslydescribed (48). Briefly, hearts were removed, dissected, fixed in 10%neutral buffered formalin, embedded in paraffin, and sectioned forexamination. Sections were used for a terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labeling (TUNEL) procedure fordetecting apoptotic cardiomyocytes as previously described (16). Thepercentage of TUNEL-positive nuclei was determined by counting 10random fields. All tissues were stored at �80°C until preparation ofRNA. Total RNA was isolated with an RNeasy Midi kit (Qiagen,Valencia, CA) and concentrated with a Microcon YM-30 column(Millipore, Billerica, MA). A formaldehyde agarose gel containingethidium bromide was used to assess the quality of the RNA. Semi-quantitative PCR analysis was performed for alterations in geneexpression. PCR primers for �-myosin heavy chain (�-MHC) were5�-GGA AGA GTG AGC GGC GCA TCA AGG-3� (forward) and5�-CTG CTG GAG AGG TTA TTC CTC G-3� (reverse); for �-myosin heavy chain (�-MHC) were 5�-GCC AAC ACC AAC CTGTCC AAG TTC-3� (forward) and 5�-TGC AAA GGC TCC AGGTCT GAG GGC-3� (reverse); for cardiac ankyrin repeat protein(CARP) were 5�-TGC GAT GAG TAT AAA CGG ACG-3� (forward)and 5�-GTG GAT TCA AGC ATA TCT CGG AA-3� (reverse); andfor glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were 5�-ACC ACA GTC CAT GCC ATC AC-3� (forward) and 5�-TCC ACCACC CTG TTG CTG TA-3� (reverse). Amplification was as follows:1) 50°C for 2 min; 2) 95°C for 5 min; 3) 30 cycles of 94°C for 30 s,55°C for 30 s, 72°C for 2 min; 4) 72°C for 10 min. The products werefractionated on a 2% agarose gel and visualized by ethidium bromidestaining. The ratio of �-MHC to �-MHC and CARP expression werequantitated by assessing relative cDNA levels of the genes comparedwith GAPDH expression from the same sample. Immunoblots wereprepared with S9 fractions (50 �g protein) isolated from hearts andprobed with antibodies to anti-CYP2J2pep1 (1:1,000) and GAPDH(1:1,000) (Santa Cruz Biotechnology, Santa Cruz, CA) (32, 48, 50).

Measurement of Dox metabolism. The ability of WT and CYP2J2Tr mice to metabolize Dox was evaluated in an HPLC assay. Briefly,microsomal proteins from WT and CYP2J2 Tr hearts (1 mg protein/ml) were incubated with Dox (500 nM) at 37°C for 60 min in a buffercontaining 50 mM potassium phosphate, 1.15% KCl, pH 7, and 1 mMNADPH (17, 34). Control experiments were performed with theselective P-450 epoxygenase inhibitor N-methylsulfonyl-6-(propargyl-oxyphenyl)hexanamide (MS-PPOH) (50 �M; generously provided byDr. J. Falck, University of Texas, Dallas, TX). The reactions werestopped by the addition of 300 �l of acetonitrile, and Dox wasextracted with a chloroform and 2-propanol (1:1 vol/vol) procedure,dried, and redissolved in 120 �l of methanol. Samples were injected

Fig. 1. A: acute doxorubicin (Dox) treatment protocol. B: chronic Dox treat-ment protocol.

H38 CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

into a Waters 712 WISP HPLC equipped with a Schima D RF-10AXLfluorescence detector (17, 34, 38). A C18 10-�m Bondapak columnwas utilized with a formic acid (0.05%):acetonitrile gradient mobilephase in reverse mode. All products were identified based on coelu-tion with authentic standards. Standard curves prepared with Dox(0–1,000 ng/ml) were used to determine concentration differences andspecific activity (excitation 470 nm, emission 560 nm).

Mitochondrial assessment. H9c2 cells (American Type CultureCollection, Manassas, VA) were cultured and grown in DMEMsupplemented with 10% bovine serum albumin and antibiotics such aspenicillin and streptomycin at 37°C in an atmosphere of 5% CO2-95%air. Cells were loaded with 150 nM tetramethylrhodamine ethyl ester(TMRE) (Invitrogen) for 30 min and then subjected to time-lapseimaging for 60 min at 37°C and 5% CO2. A Zeiss Axio Observer Z1inverted epifluorescence microscope was used to take z-stack imagesevery minute with 200-ms exposure time. Cells were observed undera �40 objective, fluorescence was excited at the 555-nm line, andemission was recorded with a band-pass filter of 575–640 nm.Changes in fluorescence were recorded in cells treated with vehicle(0.5% ethanol in PBS), Dox (10 �M), 11,12-EET (1 or 10 �M) or14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE) (1 �M; generouslyprovided by Dr. J. Falck, University of Texas). Mitochondrial mor-phology changes were visualized over the 60-min exposure period,and individual mitochondria were assessed for alterations to theelongated and filamentous appearance found in control cells. The term“punctate mitochondria” was used to describe both condensed andfragmented mitochondrial morphology as described elsewhere (35).Measurements were taken from four or five individual experiments,and intensities were quantified relative to background levels. Individ-ual mitochondria were quantified in multiple images taken at similarmagnifications with AxioVision Software (Carl Zeiss Imaging Solu-tions). Changes in fluorescence were expressed as percent changerelative to baseline levels.

Statistical analysis. Values are expressed as means � SE. Statis-tical significance was determined by the unpaired Student’s t-test andone-way ANOVA followed by a Duncan’s test to assess multiplegroup comparisons. Values were considered significant if P 0.05.

RESULTS

Effects of acute Dox administration. Serum CK and LDHactivities increased in a dose-dependent manner after threeconsecutive daily administrations of Dox in both WT andCYP2J2 Tr mice (Fig. 2, A and B). However, overexpression ofCYP2J2 resulted in lower serum CK and LDH activities afterDox treatment, indicative of reduced myocardial injury. Dox-mediated cardiotoxicity has been shown to involve productionof reactive oxygen species (ROS) (1, 8). In turn, increasedintracellular ROS activate endogenous antioxidant enzymes,such as catalase. Catalase is an important antioxidant enzymethat catalyzes the decomposition of hydrogen peroxide to waterand oxygen. Acute Dox treatment resulted in significant in-creases in cardiac catalase activities in both WT and CYP2J2Tr mice, although this increase was less pronounced in thelatter group (Fig. 2C).

Postischemic cardiac performance was decreased in Dox-treated WT mice compared with vehicle-treated mice as mea-sured by LVDP (21.8 � 5% vs. 12.3 � 3% for the 0 and 15mg/kg groups, respectively; P 0.05) (Fig. 2D). CYP2J2 Trmice exhibited improved postischemic cardiac function com-pared with WT mice, consistent with previously publishedresults (48). Importantly, no decrease in LVDP was observedin CYP2J2 Tr hearts after Dox treatment (38 � 3% vs. 36 �11% for the 0 and 15 mg/kg groups, respectively) (Fig. 2D).

Fig. 2. Acute exposure to Dox. A and B: serum creatinekinase (CK; A) and lactate dehydrogenase (LDH; B)levels after 3 consecutive daily administrations of Dox(0, 5, or 15 mg/kg); n � 6–8 per group. *P 0.05treated vs. control of the same genotype; ^P 0.05 vs.wild type (WT). CYP2J2, cytochrome P-450 2J2; Tr,transgenic. C: catalase activity in hearts from mice after3 consecutive daily administrations of Dox (0 or 15mg/kg); n � 5 per group. *P 0.05 treated vs. controlof the same genotype; ^P 0.05 vs. WT. D: postische-mic left ventricular (LV) developed pressure (LVDP)recovery at 40 min of reperfusion (R40) expressed as %of baseline LVDP in hearts from mice after 3 consecutivedaily administrations of Dox (0 or 15 mg/kg); n � 5 pergroup. *P 0.05 vs. control of the same genotype; ^P 0.05 vs. WT.

H39CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

Evidence suggests that cellular apoptotic responses may betriggered subsequent to Dox exposure. Therefore, we furtherassessed cardiac injury by analyzing cytosolic fractions forcaspase-3 activity. Compared with mice treated with vehicle,caspase-3 activity was significantly higher in both WT andCYP2J2 Tr mice treated with Dox (15 mg/kg) (Fig. 3A).Importantly, caspase-3 activity was significantly higher in WThearts than in CYP2J2 Tr hearts after Dox treatment (84 � 8vs. 55 � 9 pmol �min�1 �mg protein�1, respectively; P 0.05)(Fig. 3A). Consistent with these results, significant increases inTUNEL-positive nuclei were observed in WT hearts after acutetreatment with Dox, whereas no significant changes wereobserved in CYP2J2 Tr hearts (Fig. 3B). However, similar to arecent report by Hiraumi et al. (21), we did not observe anysignificant histological changes with hematoxylin and eosinstaining on repeated blinded analyses.

Effects of chronic Dox administration. Chronic treatmentwith Dox resulted in significant dose-dependent decreases inbody weight in both WT and CYP2J2 Tr mice (Fig. 4, A andB). After Dox treatment was stopped and during the “washout”period, body weight stabilized and began to recover in allDox-treated animals. Mice were killed at the end of the

recovery period, after which heart weights were measured andcompared with changes in body weight. As shown in Fig. 4C,heart weight-to-body weight ratios increased in a dose-depen-dent manner after Dox treatment. However, the magnitude ofincrease was significantly larger in WT mice than in CYP2J2Tr mice at the highest Dox dose, suggestive of cardiac hyper-trophy.

In contrast to the acute protocol, serum CK and LDHactivities were not different in Dox-treated mice comparedwith vehicle control animals at the end of the chronic protocol(see Supplemental Data for this article).1 Additionally, nosignificant difference in caspase-3 activity was observed be-tween Dox-treated and vehicle-treated mice in the chronicstudy (see Supplemental Data). To determine whether chronicDox treatment resulted in significant changes to the cardiacstructure, we examined expression levels of MHC and CARPin WT and CYP2J2 Tr mice. Analysis for MHC gene expres-sion following chronic Dox treatment revealed an increased�-MHC-to-�-MHC ratio in WT mice but not in CYP2J2 Trmice (Fig. 4D). CARP is a transcriptional cofactor and struc-tural component of the sarcomere involved in cardiogenesisand muscle injury (46). Studies have demonstrated that Doxtreatment can induce CARP in vivo but repress CARP expres-sion in cell culture systems (5, 60). As shown in Fig. 4E,chronic Dox treatment increased CARP expression only in WTmice. Together the data indicate that CYP2J2 Tr mice had lesscardiac injury than WT mice after chronic Dox treatment.

To examine whether CYP2J2 can metabolize Dox, we in-cubated microsomes from CYP2J2 Tr and WT mouse heartsand measured Dox turnover. As shown in Fig. 4F, CYP2J2 Trmice demonstrated a significantly higher rate of Dox metabo-lism (2.2 � 0.25 ng �min�1 �100 �g protein�1) compared withWT mice (1.6 � 0.50 ng �min�1 �100 �g protein�1). Impor-tantly, the selective epoxygenase inhibitor MS-PPOH abol-ished the improved metabolism observed in CYP2J2 Tr mice(Fig. 4F). Thus the enzymatic activity was comparable in thetwo genotypes after treatment with MS-PPOH. Immunoblotanalysis demonstrated that Dox treatment did reduce the ex-pression level of CYP2J2 protein in hearts after chronic ad-ministration (Supplemental Data).

Cardiac function after chronic administration of Dox. Todetermine whether contractile function was affected by chronicDox administration, LVDd and LVDs were assessed by trans-thoracic echocardiography at the end of the recovery periodand LVFS was calculated. As shown in Fig. 5, there was nosignificant difference in these parameters between WT miceand CYP2J2 Tr mice in the vehicle control groups, indicatingthat CYP2J2 Tr mice had normal chamber dimensions andbasal contractile function. Dox treatment caused significantdose-dependent increases in both LVDd and LVDs in WT mice(Fig. 5, A and B). In contrast, there were no significant changesin LVDd or LVDs in CYP2J2 Tr mice after Dox treatment.DOX caused a reduction in %FS in WT and CYP2J2 Tr mice,although the decrease in %FS was significantly less in CYP2J2Tr mice (Fig. 5C). These data suggest that CYP2J2 Tr heartshad significantly better cardiac function than WT hearts afterchronic Dox treatment. There were no significant changes inheart rate between the groups (Fig. 5D).

1 The online version of this article contains supplemental material.

Fig. 3. Apoptotic response following acute Dox. A: cardiac caspase-3 activity.Cytosolic fractions were prepared from hearts after acute exposure to Dox (0or 15 mg/kg) and analyzed for caspase-3 activity with Ac-DEVD-AMC assubstrate; n � 5. *P 0.05 Dox vs. control of the same genotype; ^P 0.05CYP2J2 Tr vs. respective WT mice. B: terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL)-positive cells. Myocardium apop-tosis was assessed by TUNEL assay in heart slices after acute exposure to Dox(0 or 15 mg/kg). Values represent % increase in TUNEL-positive nuclei abovecontrol groups from the same genotype; n � 10–15 hearts per group. *P 0.05 Dox vs. control of the same genotype.

H40 CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

EETs limit mitochondrial damage caused by Dox in H9C2cells. Recent reports indicate that mitochondria undergo dra-matic fragmentation and dysfunction in response to Dox-induced toxicity (13, 21, 23, 43). To determine whether EETscan limit Dox injury, we investigated the mitochondrial mor-phology and membrane potential in H9c2 cells by real-timeimaging. Mitochondria, which exhibit elongated and filamen-tous morphology in healthy control cells, became dramaticallyshorter and round after Dox exposure in H9c2 cells loaded withTMRE (Fig. 6 and Fig. 7A). Dox exposure resulted in thedissipation of fluorescence from the cells within 30 min (Fig.7B), indicating changes in mitochondrial membrane potential.Pretreatment of H9c2 cells with 11,12-EET significantly atten-uated the fragmentation and conversion of tubular mitochon-dria to punctate mitochondria (Fig. 6 and Fig. 7A). Blindedquantitative analysis revealed significantly higher relative fluo-rescence intensity in cells cotreated with 11,12-EET and Doxcompared with Dox alone (Fig. 7B). Together these datasuggest that EETs attenuated mitochondrial fission andslowed the collapse of the membrane potential. To confirm

that the effect was mediated by EETs, we conducted exper-iments in the presence of 14,15-EEZE. This putative pan-EET receptor antagonist had no effect on mitochondriawhen administered alone and abrogated the effect of 11,12-EET in H9c2 cells (Figs. 6 and 7). In further analysis ofdownstream effects of mitochondrial dysfunction, Dox-induced caspase-3 activity was partially attenuated by co-treatment with EETs (Fig. 7C).

DISCUSSION

Several hypotheses have been put forth to explain the car-diotoxicity that limits the therapeutic use of Dox (37, 52, 53),including generation of free radicals in cardiomyocyte mito-chondria. There is an increasing amount of literature reportingthe functional significance of CYP monooxygenase enzymes inthe heart. This is particularly true for CYP2J2, a primarilycardiac P-450 active in the epoxidation of AA to EETs (61). Inthe present study, we demonstrate that cardiac overexpressionof the human CYP2J2 cDNA limits Dox-induced toxicity in

Fig. 4. Chronic exposure to Dox. A and B: bodyweight change in WT (A) and CYP2J2 Tr (B) mice.Animal weights were taken during the chronic Doxprotocol; n � 12–15. C: analysis of heart weight-to-body weight ratios (HW:BW) in WT andCYP2J2 Tr mice after chronic Dox treatment (0,1.5, or 3.0 mg/kg); n � 10–15. *P 0.05 DOX vs.control of the same genotype; ^P 0.05 CYP2J2Tr vs. respective WT. D: semiquantitative PCRanalysis of myosin heavy chain (MHC) gene ex-pression. �-MHC:�-MHC expression was quanti-tated by assessing relative cDNA of both genesfrom the same individual samples; n � 3. *P 0.05Dox vs. control. E: semiquantitative PCR analysisof cardiac ankyrin repeat protein (CARP) geneexpression. CARP expression was quantitated byassessing relative cDNA to GAPDH expressionfrom the same individual samples; n � 3. *P 0.05Dox vs. control. F: Dox metabolism is increased inCYP2J2 Tr compared with WT hearts and attenu-ated by the P-450 epoxygenase inhibitor N-methyl-sulfonyl-6-(propargyloxyphenyl)hexanamide (MS-PPOH, 50 �M). Values are means � SE; n � 4.*P 0.05 vs. WT.

H41CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

mice by maintaining LV function and increased Dox metabo-lism. Moreover, our in vitro data demonstrate direct protectiveeffects of CYP2J2-derived metabolites, EETs, toward Dox-mediated mitochondrial damage.

Dox-induced injury can be indirectly monitored by therelease of CK and LDH into the serum. No differences inbaseline CK and LDH were observed between CYP2J2 Tr andWT mice, but acute Dox treatment resulted in significant

Fig. 5. Assessment of cardiac function afterchronic Dox treatment. A and B: LV end-dia-stolic dimension (LVDd, mm; A) and LV end-systolic dimension (LVDs, mm; B) after chronicDox treatment (0, 1.5, or 3.0 mg/kg); n �12–17. *P 0.05 Dox vs. control of the samegenotype; ^P 0.05 CYP2J2 Tr vs. respectiveWT. C: fractional shortening (FS) after chronicDox treatment (0, 1.5, or 3.0 mg/kg). FS of theLV is expressed as %FS � (LVDd � LVDs)/LVDd � 100; n � 12–17. *P 0.05 Dox vs.control of the same genotype; ^P 0.05CYP2J2 Tr vs. respective WT. D: heart rate(HR) after chronic DOX treatment (0, 1.5, or 3.0mg/kg); n � 12–17. bpm, beats/min.

Fig. 6. Assessment of mitochondrial mor-phology in H9c2 cells. Representativeframes from time-lapse series show H9c2cells treated with vehicle (0.5% EtOH inPBS), 14,15-epoxyeicosa-5(Z)-enoic acid(14,15-EEZE, 1 �M), Dox (10 �M), Dox(10 �M) 11,12-epoxyeicosatrienoic acid(11,12-EET, 10 �M), or Dox (10 �M) 11,12-EET (10 �M) 14,15-EEZE (1 �M)at 0, 30, and 60 min. Mitochondrial mor-phology, filamentous and tubular shape, ofthe control cells remains unaltered duringthis time period. In contrast, Dox-treatedcells exhibit significant punctate and frag-mented mitochondrial morphology, markedby arrows, which is attenuated in EET-treated cells.

H42 CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

increases in serum LDH and CK levels suggestive of tissuedamage, consistent with other reports (56). Importantly, theselevels were significantly lower in mice overexpressing CYP2J2than in WT mice. Although Dox toxicity occurs more selec-tively in heart than in other tissues, serum LDH and CK canarise from multiple organs. However, as CYP2J2 was only

overexpressed in cardiomyocytes of CYP2J2 Tr mice andcirculating EETs levels are similar between WT and CYP2J2Tr mice (48), the data presented here infer a reduction incardiac-specific CK and LDH as a result of CYP2J2 overex-pression.

Evidence indicates that cardiomyocyte apoptosis plays asignificant role in cardiac dysfunction in Dox-induced cardio-myopathy (39, 59). Here we observed that hearts from CYP2J2Tr mice had reduced activation of caspase-3 and reducedTUNEL-positive cells after acute Dox administration, consis-tent with reports demonstrating the antiapoptotic effects ofCYP2J2-derived EETs in other cell types (12, 26, 63). It isplausible that the increased level of EETs in the hearts ofCYP2J2 Tr mice mediated this response or that CYP2J2 wasinvolved in the metabolism of Dox. Although the exact antiapop-totic mechanism(s) of EETs is not known, it appears to involvep42/p44-MAPK and phosphatidylinositol 3-kinase/Akt path-ways (12, 26, 63). Manifestation of acute injury results infunctional decline in cardiac performance following Dox tox-icity. In this regard, cardiac dysfunction was marginally evi-dent during baseline perfusions as evidenced by decline in bothinotropy and lusitropy before ischemic insult, consistent withother reports (58). However, the cardioprotective effect ofCYP2J2 was prominent after ischemia-reperfusion, when theLVDP of Dox-treated CYP2J2 Tr mice did not differ signifi-cantly from that of vehicle-treated mice whereas Dox-treatedWT mice had a significant decline in LVDP compared withvehicle-treated mice. These data are strongly suggestive of aprotective effect of CYP2J2 in maintaining cardiac functionafter Dox administration.

To assess the influence of CYP2J2 on late events in Dox-mediated cardiotoxicity, we used a chronic Dox administrationprotocol. Our results revealed a general toxicity that occurredin both WT and CYP2J2 Tr mice and manifested as a signif-icant decrease in body weight, most likely stemming fromsevere anorexia, poor oral intake, and dehydration. Interest-ingly, Dox-induced increases in heart weight-to-body weightratios, �-MHC-to-�-MHC ratios, and CARP expression weregreater in WT mice than in CYP2J2 Tr mice. While thefunctional role of CARP is not fully understood, evidencesuggests its involvement in mechanical or stress responses,where it contributes to tissue repair (46). CARP appears tohave a role in structural organization of sarcomeres as well asin the transcriptional machinery of cardiomyocytes, striatedmuscles, and vasculature (51). Increased expression of CARPhas been observed in several cardiovascular injuries, such asLV dilated cardiomyopathy and pressure-overload hypertrophy(4, 46). Interestingly, opposing data are available regardingDox-mediated regulation of CARP expression, with increasedexpression reported in vivo (60) and repression reported invitro; these differences have been attributed to differences intiming and models utilized (46). Data presented here indicatethat cardiac overexpression of CYP2J2 resulted in maintenanceof control levels of CARP expression after chronic Dox ad-ministration, although the significance of this finding for themaintenance of overall cardiac function is unclear. Consistentwith adverse effects, there was a decrease in cardiac function inWT mice that was either not apparent (LVDd and LVDs) or notas severe (LVFS) in CYP2J2 Tr mice. As such changes havebeen well documented in various models of DOX-inducedheart failure (28, 29, 42, 44), the results found here are

Fig. 7. Assessment of mitochondrial morphology and membrane potential(��m) in H9c2 cells. A: mitochondrial morphology. Histograms represent %of punctate mitochondria present in individual cells relative to total mitochon-dria. Cells were treated with vehicle (0.5% EtOH in PBS), Dox (10 �M),14,15-EEZE (1 �M), Dox (10 �M) 11,12-EET (10 �M), or Dox (10 �M) 11,12-EET (10 �M) 14,15-EEZE (1 �M). Values represent means � SE;n � 4 or 5. *P 0.05 vs. vehicle control; ^P 0.05 vs. DoxEET treated;�P 0.05 vs. DoxEETEEZE treated. B: ��m. Histograms representing %of tetramethylrhodamine ethyl ester (TMRE) fluorescence lost in H9c2 cellsafter collapse of ��m. Cells were treated with vehicle (EtOH), Dox (10 �M),14,15-EEZE (1 �M), Dox (10 �M) 11,12-EET (10 �M), or Dox (10 �M) 11,12-EET (10 �M) 14,15-EEZE (1 �M). Values are mean � SE % changein relative fluorescence from baseline; n � 4 or 5. *P 0.05 vs. vehiclecontrol; ^P 0.05 vs. DoxEET treated. C: caspase-3 activity in H9c2 cellsusing Ac-DEVD-AMC as substrate. Cells were treated with vehicle (EtOH),Dox (10 �M), 11,12-EET (10 �M), or Dox (10 �M) 11,12-EET (10 �M)for 24 h. Values represent means � SE; n � 4 or 5. *P 0.05 vs. vehiclecontrol; ^P 0.05 vs. DoxEET treated.

H43CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

indicative of a significant protection against Dox-induced car-diac dysfunction by CYP2J2 overexpression.

Evidence demonstrates that CYP enzymes are expressed inthe heart, where they may participate in the metabolism oftherapeutic agents, environmental toxicants, and endogenouscompounds (10, 14, 48). Currently limited information isavailable regarding the regulation and role CYP enzymes playin the pathophysiology of heart diseases and cardiac drugmetabolism. Our present results demonstrate that CYP2J2 Trmice can limit the Dox-induced injury. Recent studies docu-menting that P-450-derived eicosanoids can affect cardiomyo-cyte function in vitro (25, 27, 32, 33, 40, 45, 62) and protectagainst ischemia-reoxygenation injury (19, 20, 48, 50) have ledto the hypothesis that these metabolites may have importantendogenous functions in the heart. However, the reduced injuryobserved in our CYP2J2 Tr mice may be partially attributed tothe increased Dox metabolism compared with WT mice. In-teresting data from H9c2 cell culture experiments show thatDox can induce CYP enzymes, notably CYP2J2 isoforms (65).The increased turnover of Dox in CYP2J2 Tr mice suggests apotential mechanism for the reduced toxicity observed in theseanimals. Considering that CYP enzymes can produce bioactivemetabolites and metabolize foreign compounds, many impor-tant questions remain regarding the role of cardiac CYP.

Cellular excitation-contraction and mitochondrial energeticsare tightly regulated in cardiac cells to meet the high energeticflux during cardiac work. Importantly, cellular stress condi-tions can result in distinct morphological changes that reducemitochondrial dynamics influencing the energetic state of thecell (24). We recently demonstrated that a marked disorgani-zation of cardiomyocyte ultrastructure following ischemia-reperfusion was significantly reduced in CYP2J2 Tr mice;moreover, EETs can minimize adverse effects of stress onmitochondrial function (30). Mitochondria are dynamic or-ganelles that migrate through the cell and undergo continuousfusion or fission processes to maintain proper function andmeet cellular demands (22). Significant decreases in fusion orincreases in fission resulting from disease or toxicity can leadto punctate, fragmented mitochondria, which are thought toplay a critical role in cellular dysfunction and death (22, 35,64). Dox-induced toxicity can result in mitochondrial swellingand ultrastructural changes and alter function. Recently,Hiraumi et al. (21) demonstrated that Dox-induced mitochon-drial damage can begin to occur at an early phase in cardiacinjury before apoptotic changes. In the present study using anin vitro model, we demonstrate that Dox-increased mitochon-drial fragmentation and membrane depolarization occur within1 h before caspase-3 activation at 24 h in H9c2 cells. Consis-tent with our previous data, our present study demonstrates thatEETs minimize the adverse effects of Dox on mitochondrialfunction. While these results are limited to our in vitro model,the implication that EETs can attenuate formation of punctatemitochondria is of particular significance. Indeed, the factEETs can inhibit apoptotic events and maintain mitochondrialfunction highlights an interesting dichotomy, where elevatedEETs may provide benefit to reduction in cellular injury butcan also be detrimental for cancer therapy. Further studies areneeded to investigate how EETs might influence mitochondrialfission and fusion and, moreover, how this might affect in vivocardiac function and cardioprotection and what implicationsthere are for oncogenesis.

In summary, the results obtained here illustrate that cardiacCYP2J2 overexpression limits the progression of cardiac injuryand preserves cardiac function in mice after treatment withDox under two different administration protocols. Indeed, asassessed by various biochemical and functional end points, amore advanced progression toward development of cardiacinjury was observed in WT mice compared with CYP2J2 Trmice after treatment with Dox. These studies are the first todocument a protective effect of CYP2J2 in Dox-induced car-diotoxicity and may have implications for treatment of cardiacinjury.

GRANTS

This work was supported by a Canadian Institutes of Health Research Grant(MOP79465, J. M. Seubert) and by the Intramural Research Program of theNational Institute of Environmental Health Sciences (Z01-ES-025034). J. M.Seubert is the recipient of a New Investigator Award from the Heart and StrokeFoundation of Canada and a Health Scholar Award from the Alberta HeritageFoundation for Medical Research.

REFERENCES

1. Adachi T, Nagae T, Ito Y, Hirano K, Sugiura M. Relation betweencardiotoxic effect of adriamycin and superoxide anion radical. J Pharma-cobiodyn 6: 114–123, 1983.

2. Aebi H. Catalase in vitro. Methods Enzymol 105: 121–126, 1984.3. Ahmed HH, Mannaa F, Elmegeed GA, Doss SH. Cardioprotective

activity of melatonin and its novel synthesized derivatives on doxorubicin-induced cardiotoxicity. Bioorg Med Chem 13: 1847–1857, 2005.

4. Aihara Y, Kurabayashi M, Saito Y, Ohyama Y, Tanaka T, Takeda S,Tomaru K, Sekiguchi K, Arai M, Nakamura T, Nagai R. Cardiacankyrin repeat protein is a novel marker of cardiac hypertrophy: role ofM-CAT element within the promoter. Hypertension 36: 48–53, 2000.

5. Aihara Y, Kurabayashi M, Tanaka T, Takeda SI, Tomaru K, Sekigu-chi KI, Ohyama Y, Nagai R. Doxorubicin represses CARP gene tran-scription through the generation of oxidative stress in neonatal rat cardiacmyocytes: possible role of serine/threonine kinase-dependent pathways. JMol Cell Cardiol 32: 1401–1414, 2000.

6. Arafa HM, Abd-Ellah MF, Hafez HF. Abatement by naringenin ofdoxorubicin-induced cardiac toxicity in rats. J Egypt Natl Canc Inst 17:291–300, 2005.

7. Chularojmontri L, Wattanapitayakul SK, Herunsalee A, Charu-chongkolwongse S, Niumsakul S, Srichairat S. Antioxidative and car-dioprotective effects of Phyllanthus urinaria L. on doxorubicin-inducedcardiotoxicity. Biol Pharm Bull 28: 1165–1171, 2005.

8. D’Alessandro N, Nicotra C, Crescimanno M, Rausa L. Effects ofdoxorubicin on mouse heart catalase. Drugs Exp Clin Res 13: 601–606,1987.

9. Daosukho C, Chen Y, Noel T, Sompol P, Nithipongvanitch R, VelezJM, Oberley TD, St Clair DK. Phenylbutyrate, a histone deacetylaseinhibitor, protects against Adriamycin-induced cardiac injury. Free RadicBiol Med 42: 1818–1825, 2007.

10. Delozier TC, Kissling GE, Coulter SJ, Dai D, Foley JF, Bradbury JA,Murphy E, Steenbergen C, Zeldin DC, Goldstein JA. Detection ofhuman CYP2C8, CYP2C9, and CYP2J2 in cardiovascular tissues. DrugMetab Dispos 35: 682–688, 2007.

11. Deng S, Kruger A, Kleschyov AL, Kalinowski L, Daiber A,Wojnowski L. Gp91phox-containing NAD(P)H oxidase increases super-oxide formation by doxorubicin and NADPH. Free Radic Biol Med 42:466–473, 2007.

12. Dhanasekaran A, Gruenloh SK, Buonaccorsi JN, Zhang R, Gross GJ,Falck JR, Patel PK, Jacobs ER, Medhora M. Multiple antiapoptotictargets of the PI3K/Akt survival pathway are activated by epoxyeicosa-trienoic acids to protect cardiomyocytes from hypoxia/anoxia. Am JPhysiol Heart Circ Physiol 294: H724–H735, 2008.

13. Diotte NM, Xiong Y, Gao J, Chua BH, Ho YS. Attenuation of doxoru-bicin-induced cardiac injury by mitochondrial glutaredoxin 2. BiochimBiophys Acta 1793: 427–438, 2009.

14. Elbekai RH, El-Kadi AO. Cytochrome P450 enzymes: central players incardiovascular health and disease. Pharmacol Ther 112: 564–587, 2006.

H44 CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

15. Ellerby LM, Bredesen DE. Measurement of cellular oxidation, reactiveoxygen species, and antioxidant enzymes during apoptosis. MethodsEnzymol 322: 413–421, 2000.

16. Falk R, Hacham M, Nyska A, Foley JF, Domb AJ, Polacheck I.Induction of interleukin-1beta, tumour necrosis factor-alpha and apoptosisin mouse organs by amphotericin B is neutralized by conjugation witharabinogalactan. J Antimicrob Chemother 55: 713–720, 2005.

17. Fang C, Gu J, Xie F, Behr M, Yang W, Abel ED, Ding X. Deletion ofthe NADPH-cytochrome P450 reductase gene in cardiomyocytes does notprotect mice against doxorubicin-mediated acute cardiac toxicity. DrugMetab Dispos 36: 1722–1728, 2008.

18. Gewirtz DA. A critical evaluation of the mechanisms of action proposedfor the antitumor effects of the anthracycline antibiotics adriamycin anddaunorubicin. Biochem Pharmacol 57: 727–741, 1999.

19. Gross GJ, Falck JR, Gross ER, Isbell M, Moore J, Nithipatikom K.Cytochrome P450 and arachidonic acid metabolites: role in myocardialischemia/reperfusion injury revisited. Cardiovasc Res 68: 18–25, 2005.

20. Gross GJ, Gauthier KM, Moore J, Falck JR, Hammock BD, Camp-bell WB, Nithipatikom K. Effects of the selective EET antagonist,14,15-EEZE, on cardioprotection produced by exogenous or endogenousEETs in the canine heart. Am J Physiol Heart Circ Physiol 294: H2838–H2844, 2008.

21. Hiraumi Y, Iwai-Kanai E, Baba S, Yui Y, Kamitsuji Y, Mizushima Y,Matsubara H, Watanabe M, Watanabe K, Toyokuni S, Nakahata T,Adachi S. Granulocyte colony-stimulating factor protects cardiac mito-chondria in the early phase of cardiac injury. Am J Physiol Heart CircPhysiol 296: H823–H832, 2009.

22. Hom J, Sheu SS. Morphological dynamics of mitochondria—a specialemphasis on cardiac muscle cells. J Mol Cell Cardiol 46: 811–820, 2009.

23. Huigsloot M, Tijdens IB, Mulder GJ, van de Water B. Differentialregulation of doxorubicin-induced mitochondrial dysfunction and apopto-sis by Bcl-2 in mammary adenocarcinoma (MTLn3) cells. J Biol Chem277: 35869–35879, 2002.

24. Jendrach M, Mai S, Pohl S, Voth M, Bereiter-Hahn J. Short- andlong-term alterations of mitochondrial morphology, dynamics and mtDNAafter transient oxidative stress. Mitochondrion 8: 293–304, 2008.

25. Jeyaseelan R, Poizat C, Baker RK, Abdishoo S, Isterabadi LB, LyonsGE, Kedes L. A novel cardiac-restricted target for doxorubicin. CARP, anuclear modulator of gene expression in cardiac progenitor cells andcardiomyocytes. J Biol Chem 272: 22800–22808, 1997.

26. Jiang JG, Chen CL, Card JW, Yang S, Chen JX, Fu XN, Ning YG,Xiao X, Zeldin DC, Wang DW. Cytochrome P450 2J2 promotes theneoplastic phenotype of carcinoma cells and is up-regulated in humantumors. Cancer Res 65: 4707–4715, 2005.

27. Kanai H, Tanaka T, Aihara Y, Takeda S, Kawabata M, Miyazono K,Nagai R, Kurabayashi M. Transforming growth factor-beta/Smads sig-naling induces transcription of the cell type-restricted ankyrin repeatprotein CARP gene through CAGA motif in vascular smooth muscle cells.Circ Res 88: 30–36, 2001.

28. Kang YJ, Chen Y, Epstein PN. Suppression of doxorubicin cardiotox-icity by overexpression of catalase in the heart of transgenic mice. J BiolChem 271: 12610–12616, 1996.

29. Kang YJ, Chen Y, Yu A, Voss-McCowan M, Epstein PN. Overexpres-sion of metallothionein in the heart of transgenic mice suppresses doxo-rubicin cardiotoxicity. J Clin Invest 100: 1501–1506, 1997.

30. Katragadda D, Batchu SN, Cho WJ, Chaudhary KR, Falck JR,Seubert JM. Epoxyeicosatrienoic acids limit damage to mitochondrialfunction following stress in cardiac cells. J Mol Cell Cardiol 46: 867–875,2009.

31. Kim DS, Kim HR, Woo ER, Kwon DY, Kim MS, Chae SW, Chae HJ.Protective effect of calceolarioside on adriamycin-induced cardiomyocytetoxicity. Eur J Pharmacol 541: 24–32, 2006.

32. King LM, Ma J, Srettabunjong S, Graves J, Bradbury JA, Li L,Spiecker M, Liao JK, Mohrenweiser H, Zeldin DC. Cloning of CYP2J2gene and identification of functional polymorphisms. Mol Pharmacol 61:840–852, 2002.

33. Lee HC, Lu T, Weintraub NL, VanRollins M, Spector AA, ShibataEF. Effects of epoxyeicosatrienoic acids on the cardiac sodium channelsin isolated rat ventricular myocytes. J Physiol 519: 153–168, 1999.

34. Licata S, Saponiero A, Mordente A, Minotti G. Doxorubicin metabo-lism and toxicity in human myocardium: role of cytoplasmic deglycosi-dation and carbonyl reduction. Chem Res Toxicol 13: 414–420, 2000.

35. Liot G, Bossy B, Lubitz S, Kushnareva Y, Sejbuk N, Bossy-Wetzel E.Complex II inhibition by 3-NP causes mitochondrial fragmentation and

neuronal cell death via an NMDA- and ROS-dependent pathway. CellDeath Differ (March 20, 2009); doi:10.1038/cdd.2009.22.

36. Lipshultz SE, Colan SD, Gelber RD, Perez-Atayde AR, Sallan SE,Sanders SP. Late cardiac effects of doxorubicin therapy for acute lym-phoblastic leukemia in childhood. N Engl J Med 324: 808–815, 1991.

37. Lou H, Kaur K, Sharma AK, Singal PK. Adriamycin-induced oxidativestress, activation of MAP kinases and apoptosis in isolated cardiomyo-cytes. Pathophysiology 13: 103–109, 2006.

38. Minotti G, Licata S, Saponiero A, Menna P, Calafiore AM, DiGiammarco G, Liberi G, Animati F, Cipollone A, Manzini S, MaggiCA. Anthracycline metabolism and toxicity in human myocardium: com-parisons between doxorubicin, epirubicin, and a novel disaccharide ana-logue with a reduced level of formation and [4Fe-4S] reactivity of itssecondary alcohol metabolite. Chem Res Toxicol 13: 1336–1341, 2000.

39. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracy-clines: molecular advances and pharmacologic developments in antitumoractivity and cardiotoxicity. Pharmacol Rev 56: 185–229, 2004.

40. Moffat MP, Ward CA, Bend JR, Mock T, Farhangkhoee P,Karmazyn M. Effects of epoxyeicosatrienoic acids on isolated hearts andventricular myocytes. Am J Physiol Heart Circ Physiol 264: H1154–H1160, 1993.

41. Morisseau C, Hammock BD. Epoxide hydrolases: mechanisms, inhibitordesigns, and biological roles. Annu Rev Pharmacol Toxicol 45: 311–333,2005.

42. Mukhopadhyay P, Batkai S, Rajesh M, Czifra N, Harvey-White J,Hasko G, Zsengeller Z, Gerard NP, Liaudet L, Kunos G, Pacher P.Pharmacological inhibition of CB1 cannabinoid receptor protects againstdoxorubicin-induced cardiotoxicity. J Am Coll Cardiol 50: 528–536,2007.

43. Mukhopadhyay P, Rajesh M, Batkai S, Yoshihiro K, Hasko G,Liaudet L, Szabo C, Pacher P. Role of superoxide, nitric oxide andperoxynitrite in doxorubicin-induced cell death in vivo and in vitro. Am JPhysiol Heart Circ Physiol 296: H1466–H1483, 2009.

44. Pacher P, Liaudet L, Bai P, Virag L, Mabley JG, Hasko G, Szabo C.Activation of poly(ADP-ribose) polymerase contributes to development ofdoxorubicin-induced heart failure. J Pharmacol Exp Ther 300: 862–867,2002.

45. Rastaldo R, Paolocci N, Chiribiri A, Penna C, Gattullo D, Pagliaro P.Cytochrome P-450 metabolite of arachidonic acid mediates bradykinin-induced negative inotropic effect. Am J Physiol Heart Circ Physiol 280:H2823–H2832, 2001.

46. Samaras SE, Shi Y, Davidson JM. CARP: fishing for novel mechanismsof neovascularization. J Investig Dermatol Symp Proc 11: 124–131, 2006.

47. Schwartz RG, McKenzie WB, Alexander J, Sager P, D’Souza A,Manatunga A, Schwartz PE, Berger HJ, Setaro J, Surkin L. Conges-tive heart failure and left ventricular dysfunction complicating doxorubicintherapy. Seven-year experience using serial radionuclide angiocardio-graphy. Am J Med 82: 1109–1118, 1987.

48. Seubert J, Yang B, Bradbury JA, Graves J, Degraff LM, Gabel S,Gooch R, Foley J, Newman J, Mao L, Rockman HA, Hammock BD,Murphy E, Zeldin DC. Enhanced postischemic functional recovery inCYP2J2 transgenic hearts involves mitochondrial ATP-sensitive K chan-nels and p42/p44 MAPK pathway. Circ Res 95: 506–514, 2004.

49. Seubert JM, Darmon AJ, El-Kadi AO, D’Souza SJ, Bend JR. Apop-tosis in murine hepatoma hepa 1c1c7 wild-type, C12, and C4 cellsmediated by bilirubin. Mol Pharmacol 62: 257–264, 2002.

50. Seubert JM, Sinal CJ, Graves J, DeGraff LM, Bradbury JA, Lee CR,Goralski K, Carey MA, Luria A, Newman JW, Hammock BD, FalckJR, Roberts H, Rockman HA, Murphy E, Zeldin DC. Role of solubleepoxide hydrolase in postischemic recovery of heart contractile function.Circ Res 99: 442–450, 2006.

51. Shi Y, Reitmaier B, Regenbogen J, Slowey RM, Opalenik SR, Wolf E,Goppelt A, Davidson JM. CARP, a cardiac ankyrin repeat protein, isup-regulated during wound healing and induces angiogenesis in experi-mental granulation tissue. Am J Pathol 166: 303–312, 2005.

52. Singal PK, Iliskovic N. Doxorubicin-induced cardiomyopathy. N EnglJ Med 339: 900–905, 1998.

53. Singal PK, Li T, Kumar D, Danelisen I, Iliskovic N. Adriamycin-induced heart failure: mechanism and modulation. Mol Cell Biochem 207:77–86, 2000.

54. Spector AA, Fang X, Snyder GD, Weintraub NL. Epoxyeicosatrienoicacids (EETs): metabolism and biochemical function. Prog Lipid Res 43:55–90, 2004.

H45CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org

55. Steinherz LJ, Steinherz PG, Tan CT, Heller G, Murphy ML. Cardiactoxicity 4 to 20 years after completing anthracycline therapy. JAMA 266:1672–1677, 1991.

56. Tesoriere L, Ciaccio M, Valenza M, Bongiorno A, Maresi E, AlbieroR, Livrea MA. Effect of vitamin A administration on resistance of ratheart against doxorubicin-induced cardiotoxicity and lethality. J Pharma-col Exp Ther 269: 430–436, 1994.

57. Tokarska-Schlattner M, Wallimann T, Schlattner U. Alterations inmyocardial energy metabolism induced by the anti-cancer drug doxoru-bicin. C R Biol 329: 657–668, 2006.

58. Tokarska-Schlattner M, Zaugg M, da Silva R, Lucchinetti E, SchaubMC, Wallimann T, Schlattner U. Acute toxicity of doxorubicin onisolated perfused heart: response of kinases regulating energy supply.Am J Physiol Heart Circ Physiol 289: H37–H47, 2005.

59. Tokarska-Schlattner M, Zaugg M, Zuppinger C, Wallimann T,Schlattner U. New insights into doxorubicin-induced cardiotoxicity: thecritical role of cellular energetics. J Mol Cell Cardiol 41: 389–405, 2006.

60. Torrado M, Lopez E, Centeno A, Castro-Beiras A, Mikhailov AT.Left-right asymmetric ventricular expression of CARP in the piglet heart:

regional response to experimental heart failure. Eur J Heart Fail 6:161–172, 2004.

61. Wu S, Moomaw CR, Tomer KB, Falck JR, Zeldin DC. Molecularcloning and expression of CYP2J2, a human cytochrome P450 arachidonicacid epoxygenase highly expressed in heart. J Biol Chem 271: 3460–3468,1996.

62. Xiao YF, Huang L, Morgan JP. Cytochrome P450: a novel systemmodulating Ca2 channels and contraction in mammalian heart cells.J Physiol 508: 777–792, 1998.

63. Yang S, Lin L, Chen JX, Lee CR, Seubert JM, Wang Y, Wang H,Chao ZR, Tao DD, Gong JP, Lu ZY, Wang DW, Zeldin DC. Cyto-chrome P-450 epoxygenases protect endothelial cells from apoptosisinduced by tumor necrosis factor-alpha via MAPK and PI3K/Akt signalingpathways. Am J Physiol Heart Circ Physiol 293: H142–H151, 2007.

64. Yu T, Sheu SS, Robotham JL, Yoon Y. Mitochondrial fission mediateshigh glucose-induced cell death through elevated production of reactiveoxygen species. Cardiovasc Res 79: 341–351, 2008.

65. Zordoky BN, El-Kadi AO. Induction of several cytochrome P450 genesby doxorubicin in H9c2 cells. Vascul Pharmacol 49: 166–172, 2008.

H46 CYP2J2-MEDIATED CARDIAC PROTECTION

AJP-Heart Circ Physiol • VOL 297 • JULY 2009 • www.ajpheart.org