<<

Proteomic Profiling Reveals Adaptive Responses to Surgical Myocardial Ischemia–Reperfusion in Hibernating Arctic Ground Squirrels Compared to Rats PERIOPERATIVE MEDICINE Quintin J. Quinones, M.D., Ph.D., Zhiquan Zhang, Ph.D., Qing Ma, M.D., Michael P. Smith, M.S., Erik Soderblom, Ph.D., M. Arthur Moseley, Ph.D., James Bain, Ph.D., Christopher B. Newgard, Ph.D., Michael J. Muehlbauer, Ph.D., Matthew Hirschey, Ph.D., Kelly L. Drew, Ph.D., Brian M. Barnes, Ph.D., Mihai V. Podgoreanu, M.D.

ABSTRACT

Background: Hibernation is an adaptation to extreme environments known to provide organ protection against ischemia– Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 reperfusion (I/R) injury. An unbiased systems approach was utilized to investigate hibernation-induced changes that are char- acteristic of the hibernator cardioprotective phenotype, by comparing the myocardial proteome of winter hibernating arctic ground squirrels (AGS), summer active AGS, and rats subjected to I/R, and further correlating with targeted metabolic changes. Methods: In a well-defined rodent model of I/R by deep hypothermic circulatory arrest followed by 3 or 24 h of reperfusion or sham, myocardial abundance in AGS (hibernating summer active) and rats (n = 4 to 5/group) was quantified by label-free proteomics (n = 4 to 5/group) and correlated with metabolic changes. Results: Compared to rats, hibernating AGS displayed markedly reduced plasma levels of troponin I, myocardial apoptosis, and left ventricular contractile dysfunction. Of the 1,320 rat and 1,478 AGS identified, 545 were differentially expressed between hibernating AGS and rat hearts (47% up-regulated and 53% down-regulated). ontology analysis revealed down-regulation in hibernating AGS hearts of most proteins involved in mitochondrial energy transduction, includ- ing transport chain complexes, acetyl CoA biosynthesis, Krebs cycle, glycolysis, and ketogenesis. Conversely, fatty acid oxidation and sirtuin-3 were up-regulated in hibernating AGS, with preserved peroxisome proliferator–activated receptor-α activity and reduced tissue levels of acylcarnitines and ceramides after I/R. Conclusions: Natural cardioprotective adaptations in hibernators involve extensive metabolic remodeling, featuring increased expression of fatty acid metabolic proteins and reduced levels of toxic lipid metabolites. Robust up-regula- tion of sirtuin-3 suggests that posttranslational modifications may underlie organ protection in hibernating mammals. (Anesthesiology 2016; 124:1296-310)

SCHEMIA–reperfusion (I/R) injury, a con- What We Already Know about This Topic I sequential component of cardiac surgery and organ • Myocardial ischemia–reperfusion injury remains a major transplantation, is a major determinant of morbidity and source of perioperative morbidity and mortality mortality. Cardioprotection after I/R remains an elusive tar- • The study of hibernation, an adaptive response to extreme get despite significant research and numerous compounds environments, has potential to address novel mechanisms with promising preclinical data.1 Mammalian hibernation and potential therapies for perioperative myocardial ischemia– reperfusion injury is a seasonal phenomenon characterized by prolonged bouts (days to weeks) of winter torpor, a state of decreased physi- What This Article Tells Us That Is New ologic activity, reduced body temperature, and decreased • Applying analysis to hibernating arctic ground metabolic rate, interspersed with brief (hours to a day) squirrels, the authors have found that the natural cardiac pro- periods of arousal when and temperature tem- tective adaptations of hibernators involve metabolic shifts in fatty acid metabolism proteins, reductions in toxic lipid me- 2 porarily return to normal. The entry into and exit from tabolites, and up-regulation of sirtuins torpor is akin to I/R, in that blood flow is greatly reduced and then restored. During arousal from torpor, hiberna- consumption, while accelerating their heart rate 100-fold tors rewarm using a combination of shivering and non- with corresponding increases in organ perfusion.3 Hiber- shivering thermogenesis associated with peaks in nating mammals like arctic ground squirrels (AGS) show

This article is featured in “This Month in Anesthesiology,” page 1A. Corresponding article on page 1215. Supplemental Digital Content is available for this article. Direct URL citations appear in the printed text and are available in both the HTML and PDF versions of this article. Links to the digital files are provided in the HTML text of this article on the Journal’s Web site (www.anesthesiology.org). Submitted for publication May 27, 2015. Accepted for publication February 2, 2016. From the Department of Anesthesiology, Duke Uni- versity, Durham, North Carolina (Q.J.Q., Z.Z., Q.M., M.P.S., M.V.P.); Center for Genomic and Computational Biology – Proteomics Shared Resource, Duke University, Durham, North Carolina (E.S., M.A.M.); Molecular Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University, Durham, North Carolina (J.B., C.B.N., M.J.M., M.H.); and Institute of Arctic Biology, University of Alaska, Fairbanks, Alaska (K.L.D., B.M.B., M.V.P.). Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. All Rights Reserved. Anesthesiology 2016; 124:1296-310

Anesthesiology, V 124 • No 6 1296 June 2016

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

natural resistance to experimental I/R in multiple organs,4–7 which remains yet uncharacterized for the heart. Moreover, although a suite of adaptations associated with the seasonal entry into torpor have been reported, including metabolic rate depression,8 increased reliance on lipid oxidation for energy production,3,9–11 and immune suppression,12 the mechanisms underlying this naturally evolved organ protec- tion remain unknown. In the current study, we hypothesized that experimen- tal surgical I/R will result in reduced severity of myocardial

injury and dysfunction in hibernating AGS compared to a Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 nonhibernator (rat); furthermore, we postulated that such hibernator cardioprotective phenotype will be attenuated in summer active (SA) AGS. As a first step to delineate the metabolic regulatory events involved, we employed an unbi- ased proteomics approach to identify and quantify differ- ences in myocardial protein abundance between AGS and rats and further correlate with targeted metabolic changes. The potential cardioprotective role of sirtuin-3 up-regulation Fig. 1. Experimental design and perfusion apparatus for identified in hibernating AGS hearts was further assessed in deep hypothermic circulatory arrest (DHCA). B = blood specimen; C = cooling; CPB = cardiopulmonary bypass; vitro using gain-of-function (rat) and loss-of-function (AGS) E = echocardiographic measurements; H = hemodynam- experiments in adult primary ventricular cardiomyocyte ic measurements; M = myocardial specimen collection; (APVC) models of hypoxia–reoxygenation. P = surgical preparation; RR = rewarming and reperfusion; W = weaning from cardiopulmonary bypass. Materials and Methods and n = 7 females. As these animals are wild caught, exact age Animal Ischemia–Reperfusion Protocol—Anesthetic, cannot be determined; however, juvenile and aged animals Surgical, and Perfusion Management were excluded. Adult male Brown Norway and Dahl/Salt Sensitive rat Rats and AGS underwent an identical experimental strains (Charles River, USA) were randomly assigned to protocol, as previously described.14,15 Briefly, animals were three experimental groups: anesthetized and cannulated anesthetized with isoflurane, endotracheally intubated and without undergoing cardiopulmonary bypass (CPB; sham mechanically ventilated; anesthesia was maintained with group) and deep hypothermic circulatory arrest (DHCA; isoflurane, fentanyl, and pancuronium. Surgical prepara- 45 min at 18°C), followed by either 3 h (DHCA R3h group) tion involved cannulation of the ventral tail artery (rats) and or 24 h (DHCA 24h group) of reperfusion (fig. 1). right femoral artery (AGS; 20 gauge, arterial inflow), inser- Arctic ground squirrels (Urocitellus parryii) were trapped tion of a multiorifice dual-stage cannula through the right in July near the Toolik Field Station in northern Alaska and internal jugular vein and advanced into the right atrium (4.5 transported to the University of Alaska Fairbanks (Fairbanks, French, venous outflow), and insertion of a 3.5-mm bal- Alaska). In the fall, animals assigned to the hibernating group loon catheter (Sprinter OTW, Medtronic, USA) via right were housed in temperature- and photoperiod-controlled carotid artery cutdown, positioned above the aortic valve facilities (5°C, photoperiod, 4L:20D), after being implanted under echocardiographic guidance. Electrocardiogram, with intraperitoneal temperature-sensitive radio transmit- pulse oximetry, end-tidal capnography, pericranial and rec- ters to monitor their body temperature and precisely deter- tal temperatures, and invasive arterial blood pressure were mine stages of torpor and arousal, as described in the study continuously recorded. Arterial blood gases were monitored by Yan et al.13 Preoperatively, torpid animals (hibernating throughout the experimental protocol (GEM Premier 3000, AGS) were moved to a warm, lighted room and aroused to Instrumentation Lab, USA). After systemic heparinization, euthermia before each experiment. Winter experiments were CPB was initiated and animals were cooled to 18°C. Car- conducted in January to February 2012 and 2013. Postre- dioplegic arrest was achieved by endoaortic balloon inflation productive summer euthermic animals were used as nonhi- and antegrade administration of blood cardioplegia, with bernating controls, with experiments conducted in August electromechanical arrest confirmed electrocardiographi- to September 2012 and 2013. Summer animals were housed cally and echocardiographically. DHCA was initiated for for at least 1 month to complete quarantine and infection 45 min, at the conclusion of which the endoaortic balloon and parasite testing. Summer animals had normal room- was deflated and CPB resumed. Animals were rewarmed to temperature housing and light at least 12 h/d. AGS included normothermia (1 h), weaned from CPB, and recovered until in these experimental groups were composed of n = 16 males the time of euthanasia, for either 3 (R3h) or 24 h (R24h)

Anesthesiology 2016; 124:1296-310 1297 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator

postreperfusion. CPB, ischemia, and rewarming times are (for rat) and against a curated AGS database representing identical between R3h and R24h groups, with only dura- refined thirteen-lined ground squirrel (Ictidomys tridecemlin- tion of reperfusion being different. All experiments were eatus) and protein sequences from Ensembl release approved by the Institutional Animal Care and Use Com- 69, as described.10 After individual peptide scoring using the mittees of Duke University and University of Alaska Fair- Peptide Prophet algorithm, the data were annotated at a less banks, and compliant with the Guide for the Care and Use than 1% peptide false discovery rate (FDR). Relative peptide of Laboratory Animals. and protein abundance were calculated using Rosetta Elu- cidator. Details for mass spectrometry data collection, pro- Myocardial Injury Phenotypes cessing, and quality control are presented in Supplemental Phenotypic characterization of myocardial injury severity Methods, Supplemental Digital Content 1, http://links.lww.

was performed using biochemical, echocardiographic, and com/ALN/B269. Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 histologic measurements at baseline and at the 3- and 24-h reperfusion time points. Levels of cardiac troponin I were Arctic Ground Squirrel to Rat Orthology Mapping measured at each time point in EDTA plasma using a mul- A key component of our analytical strategy to compare tiplex immunoassay system (Meso Scale Discovery, USA). changes in myocardial protein expression across species Serial transthoracic echocardiographic measurements of left involves the establishment of orthologous relationships ventricular (LV) systolic function were conducted using a among the proteins identified and quantified using label- Vivid i ultrasound system (GE Healthcare, USA) equipped free proteomic approaches in rat versus AGS. Commonly with an M12L transducer and analyzed using Xcelera R3.3 used methods for orthology assessment typically use Basic software (Phillips, USA). LV fractional area change (LV- Local Alignment Search Tool to search for pairs of recipro- FAC) was calculated as (LV end diastolic area − LV end sys- cal best hits in the (or proteomes) being compared. tolic area)/LV end diastolic area × 100. Percent change in However, these methods are easily misled when additional LV-FAC at each reperfusion time point compared to baseline gene duplications occur after the species in question have is presented for all groups. Myocardial apoptosis was assessed diverged (“in-paralogs”), leading to the presence of co-ortho- in LV tissue sections by terminal deoxynucleotidyl trans- logs in cross- comparisons. We have defined sets ferase dUTP nick-end labeling assays (Roche Diagnostics, of putatively orthologous proteins among those identified USA) and double immunofluorescence staining for activated within each species by five-fraction 2D-LC/LC-MS/MS pro- caspase-3 and troponin I, as well as by determining the levels teomics. These orthologies were established by adapting the of cleaved caspase 3 in myocardial tissue (Western blot). EnsemblCompara GeneTrees resource developed by Vilella et al.,16 a system that automates the clustering, multiple Sample Preparation for Mass Spectrometry alignment, and phylogenetic analysis necessary to build gene At the time of euthanasia (3- or 24-h reperfusion), animals trees for large numbers of gene families represented in the were completely exsanguinated via the jugular venous can- Ensembl project. Given the known one-to-one, one-to- nula and perfused with 60 ml warm phosphate-buffered many, and many-to-many orthology relationships, we used saline. The heart was rapidly removed, divided using a rat the BioMart data-mining tool and Ensembl Release heart slicer matrix into axial sections for histology, and snap 7317 (Rnor_5.0 for rat, Spetri2 for squirrel) to conduct a frozen in liquid nitrogen. Details of sample preparation pro- bidirectional orthology matching using first the rat as the ref- tocols for proteomic and metabolic profiling are presented erence genome and then the AGS as the reference genome. in Supplemental Methods, Supplemental Digital Content 1, The resulting orthology map—containing 702 unique rat http://links.lww.com/ALN/B269. GenInfo Identifier protein IDs, 700 unique squirrel Ensembl Protein IDs, and 697 unique gene names—was used for Proteomic Data Collection and Processing subsequent cross-species comparative analyses of myocardial We employed a five-fraction two-dimensional liquid chro- protein abundance. Details of the computational approach matography, tandem mass spectrometry (2D-LC/LC-MS/ to establish the orthology map are presented in Supplemental MS) method to identify and quantitate proteins expressed Methods, Supplemental Digital Content 1, http://links.lww. in LV myocardium of AGS (hibernating SA) and Brown com/ALN/B269. Norway rats subjected to DHCA followed by 3 or 24 h of reperfusion or in the sham group. Proteins extracted from Mitochondrial DNA Analysis LV homogenates were normalized, reduced, alkyl- Total DNA (mitochondrial and nuclear) was extracted from ated, and trypsin digested, followed by label-free quan- rat and AGS LV myocardium samples using a QIAamp titative 2D-LC/LC-MS/MS. Robust peak detection and DNA Mini Kit (Qiagen, USA). The content of mtDNA label-free alignment of individual peptides across all sample was calculated using quantitative real-time polymerase chain

injections were performed using Rosetta Elucidator v3.3 reaction (PCR) by measuring the threshold cycle ratio (ΔCT) (Rosetta Biosoftware, USA) with PeakTeller algorithm. MS/ of a mitochondrial-encoded gene ( oxidase, sub- MS data were searched against the NCBI RefSeq Rattus unit I, Mt-Co1) versus a nuclear-encoded gene (Cyclophilin

Anesthesiology 2016; 124:1296-310 1298 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

A, Ppia) in a StepOnePlus reverse transcription (RT)-PCR analysis are provided in Supplemental Methods, Supplemen- System (Life Technologies USA) equipped with StepOne tal Digital Content 1, http://links.lww.com/ALN/B269. v2.2.3 software (see Supplemental Methods, Supplemental Digital Content 1, http://links.lww.com/ALN/B269, for Statistical Analysis details on primers used and RT-PCR protocol). Experimental sample sizes are based on previous reports of differences in protein abundance in hibernators,3,10 com- Western Blot Analysis parative responses to I/R across species,20 as well as efforts to To validate mass spectrometry findings, Western blot imag- conserve scant resources (wild-caught AGS). ing analysis was conducted for specific electron transport Principal component analysis (PCA) was performed to chain (ETC) proteins (NADH subunit explore sources of variation in relative protein abundance

[NDUF] S8, succinate dehydrogenase complex [SDH] A, across the experimental data set, visualize whether biologic Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 cytochrome c reductase [UQCR] Q, cytochrome replicate samples resemble each other, and identify any outli- c oxidase [COX] 5A, and adenosine triphosphate [ATP] 5A) ers. PCA uses orthogonal transformation to convert relative and sirtuin-3, and all results were normalized using ratios protein abundance into a set of linearly uncorrelated principal to the constitutively expressed protein COXIV, HSP60, or components.21 The top three principal components are pre- β-actin depending on protein size (see Supplemental Meth- sented as 3D scatter plots in Supplemental Results, Supple- ods, Supplemental Digital Content 1, http://links.lww.com/ mental Digital Content 1, http://links.lww.com/ALN/B269, ALN/B269, for details). with each point representing the proteomic data from one sample and samples colored by species and hibernation state. Functional Sirtuin-3 Analyses Differentially expressed proteins in this multifactor experi- To characterize the functional relevance of differences in ment were identified using mixed model ANOVA, which myocardial sirtuin-3 abundance identified by proteomic included species, hibernation state (winter hibernating, SA), analysis, we conducted additional in vitro sirtuin-3 gain- reperfusion time (sham, reperfusion 3 h, and reperfusion 24 h), and loss-of-function studies in APVCs isolated from rat and and hibernation state × reperfusion time interaction (to test AGS,18 respectively, and assessed the effects on viability whether seasonal hibernation states would result in temporal after simulated I/R. Rat APVCs were treated with increasing differences in myocardial protein abundance postreperfusion). concentrations of resveratrol (Sigma-Aldrich, USA), an acti- Differentially expressed proteins were defined based on a fold- vator of sirtuin-3, or nicotinamide, an inhibitor of sirtuin-3 change more than or equal to 1.5 or less than or equal to −1.5 (control, 5 μM, and 10 μM) for 1 h. AGS APVCs were and a Benjamini–Hochberg FDR-adjusted P < 0.05. For this transfected with sirtuin-3–siRNA (60 pmol) or scrambled exploratory analysis, we chose FDR to adjust for multiple RNA control (Santa Cruz, USA) overnight, according to comparisons over the more conservative familywise error rate manufacturer’s protocols. Cells were subjected to 2 h of oxy- approach (e.g., Bonferroni correction), which would likely gen and glucose deprivation followed by 24 h of reoxygen- miss a number of potentially novel differences.22 Exploratory ation and determination of apoptosis (Cell Death Detection PCA analysis and differential protein expression analysis were ELISA kit, Roche) and necrosis (cardiac troponin I levels conducted using Partek Genomics Suite v6.6 (Partek, USA). in supernatant, Life Diagnostics, USA). Sirtuin-3 expression Differences in perioperative myocardial injury endpoints (Western blot) and activity (fluorometric sirtuin-3 activity (biochemical, echocardiographic), as well as differences in assay kit, Abcam, Cambridge, MA) were measured in APVC abundance of individual proteins or metabolites between lysates. All experiments were run in triplicate. hibernating AGS, SA AGS, and rats, or between time points were assessed by one-way ANOVA with Tukey post hoc test PPAR-α Activity Assay for pairwise comparisons. Two-way ANOVA was further The nuclear fraction was isolated from cryopreserved myo- used to test whether differences in individual protein abun- cardial samples using a commercial nuclear extraction kit dance between species and reperfusion time points were (Panomics, USA) according to manufacturer’s specifications more than expected by chance. Mann–Whitney U test was and assayed for peroxisome proliferator–activated receptor used for two group comparisons of apoptosis and necrosis (PPAR)-α nuclear receptor activity using ELISA (Affyme- endpoints. Statistical analyses were conducted using Graph- trix, USA; see Supplemental Methods, Supplemental Digital Pad Prism v6 (GraphPad Software, Inc, USA). Content 1, http://links.lww.com/ALN/B269, for details). Results Targeted Metabolic Profiling Cryopreserved myocardial samples were homogenized in The Hibernator Cardioprotective Phenotype 50% acetonitrile supplemented with 0.3% formic acid and Compared to rats, AGS exhibited significantly reduced used to determine levels of acylcarnitines, organic acids, myocardial injury at both reperfusion time points (mean ± amino acids, and ceramides via stable isotope dilution tech- SD of plasma troponin I concentrations after 3 h of reper- niques, as described previously.19 Details of the metabolomic fusion were 0.9 ± 0.2, 2.5 ± 2.1, 4.8 ± 0.8, and 6.7 ± 1.3 and

Anesthesiology 2016; 124:1296-310 1299 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator

after 24 h of reperfusion were 0.2 ± 0.1, 0.1 ± 0.1, 2.1 ± 0.9, percent changes in LV-FAC compared to preoperative base- and 6.3 ± 3.3 for hibernating AGS, SA AGS, Brown Nor- line after 3 h of reperfusion were 7 ± 6, −16 ± 14, and −25 ± 3 way rats, and Dahl-Salt Sensitive rats, respectively; fig. 2A). and after 24 h of reperfusion were 4 ± 14, −17 ± 32, and Reflecting known differences in susceptibility to myocardial −15 ± 4 for hibernating AGS, SA AGS, and rats, respectively; I/R between the rat strains used as nonhibernator experi- fig. 2B). Myocardial apoptosis was attenuated in hibernating mental model , severity of myocardial injury in AGS compared to rats after I/R, as evidenced by lower frac- hibernating AGS was approximately 5-fold lower compared tion of terminal deoxynucleotidyl dUTP nick- to Brown Norway rats (a strain characterized by resistance end labeling–positive nuclei (fig. 2C) and reduced tissue against myocardial I/R) and approximately 8-fold lower than levels of active caspase-3 (fig. 2D). in Dahl/salt-sensitive rats (a strain more susceptible to myo-

cardial I/R). Hibernating AGS displayed the most robust car- Cross-species Quantitative LV Myocardial Proteomic Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 dioprotection, with SA AGS demonstrating an intermediate Analyses level of protection (fig. 2A). The biochemical results were After quality control, the final quantitative data set for rat corroborated echocardiographically, with hibernating AGS consisted of 8,482 peptides/1,320 proteins (786 proteins showing preserved LV systolic function after I/R, whereas contained at least 2 unique peptides) and for AGS, 9,292 LV-FAC declined in both SA AGS and rats (mean ± SD of peptides/1,478 proteins (806 proteins contained at least

Fig. 2. The cardioprotective hibernator phenotype. Winter (hibernating [HIB]) and summer active (SA) arctic ground squirrels (AGSs) exhibit reduced myocardial injury after ischemia–reperfusion (I/R) compared to Brown Norway (BN) or Dahl salt-sensitive (SS) rats, as evidenced by reduced plasma markers of myonecrosis at both the 3-h (R3h) and 24-h (R24h) reperfusion time points (#P < 0.01, *P < 0.05 by ANOVA with Tukey post hoc correction; A). Winter (HIB) AGS further demonstrated preserved echocar- diographic left ventricular systolic function (LV-FAC) after I/R compared to SA AGS or rats (*P < 0.05, ANOVA with Tukey post hoc correction; B) and reduced myocardial apoptosis as evidenced by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining (C) corroborated by cleaved caspase-3 levels (D) at the 3-h reperfusion time point (*P < 0.05, Mann–Whitney U test). Data are presented as mean ± SD, N = 8/group.

Anesthesiology 2016; 124:1296-310 1300 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

2 unique peptides). Differential expression analysis across Western blot analyses confirmed mass spectrometry species was conducted based on an orthology map consisting findings in select ETC proteins of interest. Down-regu- of 697 proteins. PCA was performed to identify high-level lation of complex II protein SDHA in hibernating AGS differences between sample groups (fig. S3, Supplemental and SA AGS compared with rat at all time points is shown Digital Content 1, http://links.lww.com/ALN/B269). Sig- in ­figure 3. Results for complex I protein NDUFS8, com- nal versus noise across all orthologous proteins for each of plex III protein UQCRQ, complex IV protein COX5A, the factors and interactions in the ANOVA model identified and complex V protein ATP5A are shown in figure S11 that all factors contributed significant variation to the data and table S7, Supplemental Digital Content 1, http://links. across the set, with species (rat vs. AGS) representing the lww.com/ALN/B269. dominant source of variation (fig. S4, Supplemental Digital

Content 1, http://links.lww.com/ALN/B269). Increased Myocardial Sirtuin-3 Levels and the Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 A total of 545 proteins were differentially expressed Cardioprotective Hibernator Phenotype between hibernating AGS and rat based on a fold change Sirtuin-3, the major mitochondrial protein deacetylase with more than or equal to 1.5 or less than or equal to −1.5 and multiple cardioprotective targets including ETC proteins, an FDR-adjusted P < 0.05, with 255 up-regulated and is significantly up-regulated in both hibernating and SA 290 down-regulated proteins (fig. S5, Supplemental Digi- AGS compared to rat by proteomic profiling (fig. 4A). Spe- tal Content 1, http://links.lww.com/ALN/B269). Of these, cies differences also exist in the dynamic expression changes 191 proteins had more than a ±6-fold change between hiber- after I/R—unlike rats, AGS recover their baseline sirtuin-3 nating AGS and rat, and 498 had a Bonferroni-adjusted levels by R24h (fig. 4A); Western blot analysis shows a sir- P < 0.05. By using pathway analysis, these proteins were tuin-3 expression pattern consistent with the proteomic mapped onto 77 canonical pathways. Top canonical path- findings (fig. 4B). We also corroborated the sirtuin-3 result ways differentially expressed between hibernating and rat with changes in target proteins known to be regulated by included oxidative phosphorylation (p = 1.1e-20), protein sirtuin-3, including an up-regulation in AGS hearts of man- ubiquitination (1.4e-13), tricarboxylic acid (TCA) cycle ganese dismutase (total MnSOD)23 accompanied (6e-9), branched chain amino acid metabolism (7.3e-7), and by posttranslational deacetylation of MnSOD24 (fig. 4B), cytoskeletal remodeling (7.8e-7). known to increase its ROS scavenging activity.25 To further explore the potential mechanistic links Protein Down-regulation between sirtuin-3 expression, activity, and the hiberna- The most prominent proteomic finding is a general down- tor cardioprotective phenotype, we conducted compara- regulation of ETC proteins in the hibernator hearts tive gain- and loss-of-function experiments in APVCs cell (table 1). Significant reductions in levels of subunits of all 5 models of hypoxia–reoxygenation. In rat APVCs, resveratrol ETC complexes were found in hibernating AGS compared increased sirtuin-3 expression and activity at both 5 and to rats, with only complex V showing increased expression 10 μM doses (fig. 5, A and B), which was associated with of some subunits after I/R. Complex I subunits NADH- reduced apoptotic and necrotic cell death (fig. 5, C and D). ubiquinone (NDUFs) 1 to 13 were reduced Conversely, nicotinamide reduced sirtuin-3 activity at both 1.14- to 17.68-fold. Complex II subunits SDHA, SDHB, 5 and 10 μM (fig. 5B) and was associated with increased and SDHC declined 2.47- to 5.1-fold. Complex III subunits cardiomyocyte death (fig. 5, C and D). On the other hand, UQCRQ, UQCRFS1, UQCRC2, and UQCR10 declined siRNA sirtuin-3 silencing in AGS cardiomyocytes resulted 2.04- to 3.57-fold. Complex IV subunit COX5A declined in significant increases of apoptotic and necrotic death com- 1.23-fold. Complex V subunits ATP synthase mitochondrial pared to scrambled RNA–treated cells (fig. 5, E and F).

F1/F0 complex (ATP) AF2, ATP5F1, and ATP5B declined Furthermore, the observed increase in AGS cardiomyocyte 1.75- to 19.11-fold; however, subunits ATP5A1-ATP5J2 viability after hypoxia–reoxygenation stress (fig. S12, Sup- increased in abundance 1.41- to 15.94-fold (table 1). plemental Digital Content 1, http://links.lww.com/ALN/ To ensure that lower quantities of ETC proteins detected B269) is accompanied by reduced acetylation of mitochon- in AGS were not secondary to lower mitochondrial density drial proteins compared to the rat (fig. S13, Supplemental in the myocardium, mtDNA quantity was calculated as the Digital Content 1, http://links.lww.com/ALN/B269). ratio of COX1 to cyclophillin A DNA levels determined by RT-PCR and found to be higher for AGS compared to Metabolic Correlates of the Hibernator Cardioprotective rat (fig. S10, Supplemental Digital Content 1, http://links. Phenotype lww.com/ALN/B269), consistent with an increase in mito- The proteomic analyses revealed a sharp contrast between a chondrial biogenesis in the hibernator heart. A decrease in general down-regulation of key metabolic enzymes associ- mtDNA copy number, however, was seen in both species ated with glycolysis, TCA cycle, ketolysis, amino acid catab- under the R3h condition compared to sham, coincident olism, and ETC, whereas a significant proportion of proteins with peak reperfusion injury (fig. S10, Supplemental Digital involved in fatty acid uptake, transport, and catabolism were Content 1, http://links.lww.com/ALN/B269). up-regulated in the hibernator hearts (table 2). Accordingly,

Anesthesiology 2016; 124:1296-310 1301 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator

Table 1. Expression Differences in Hibernating AGS versus Rat Hearts for Members of the Electron Transport Chain

Fold Change HIB-AGS FDR-adjusted Protein Symbol Protein Name vs. Rat P Value

Complex I (NADH-ubiquinone reductase) Ndufa10 NADH dehydrogenase [ubiquinone] 1 alpha subcomplex 10-like protein −1.14 1.61E-02 Ndufa13 NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13 −2.76 7.76E-10 Ndufa5 NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 5 −5.26 4.19E-07 Ndufab1 NADH dehydrogenase [ubiquinone] 1, alpha/beta subcomplex, 1 −16.51 1.06E-12 Ndufb10 NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 10 −1.87 2.60E-11 Ndufb5 NADH dehydrogenase [ubiquinone] 1 beta subcomplex, 5 −1.49 1.68E-05 Ndufb6 NADH dehydrogenase [ubiquinone] 1 beta subcomplex, 6 −6.28 6.15E-13 Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 Ndufs1 NADH-ubiquinone oxidoreductase 75 kDa subunit, mitochondrial −3.62 1.27E-12 Ndufs2 NADH dehydrogenase [ubiquinone] - protein 2, mitochondrial −2.18 4.12E-11 Ndufs3 NADH dehydrogenase [ubiquinone] Fe-S protein 3 −1.49 6.15E-05 Ndufs4 NADH dehydrogenase [ubiquinone] iron-sulfur protein 4, mitochondrial −1.20 1.88E-02 Ndufs5 NADH dehydrogenase [ubiquinone] Fe-S protein 5b, 15kDa (NADH-coenzyme Q −3.31 1.58E-16 reductase) Ndufs6 NADH dehydrogenase [ubiquinone] Fe-S protein 6 −1.20 1.58E-02 Ndufs7 NADH dehydrogenase [ubiquinone] Fe-S protein 7 −7.11 5.89E-25 Ndufs8 NADH dehydrogenase [ubiquinone] Fe-S protein 8 −17.68 7.93E-12 Ndufv2 NADH dehydrogenase [ubiquinone] 2, mitochondrial −1.37 1.16E-05 Ndufv3 NADH dehydrogenase [ubiquinone] flavoprotein 3, mitochondrial −2.69 9.07E-12 Complex II (succinate dehydrogenase-CoQ reductase) Sdha Succinate dehydrogenase complex subunit A −5.10 3.20E-16 Sdhb Succinate dehydrogenase complex subunit B −2.60 3.05E-11 Sdhc Succinate dehydrogenase complex subunit C −2.47 3.91E-08 Complex III (cytochrome reductase) Uqcrc2 -c1 complex subunit 2, mitochondrial −2.48 8.02E-08 Uqcrfs1 Cytochrome b-c1 complex subunit Rieske, mitochondrial −2.92 4.79E-21 Uqcrq Cytochrome b-c1 complex subunit 8 −3.57 7.23E-12 Uqcr10 Ubiquinol-cytochrome c reductase, complex III subunit X −2.04 3.75E-06 Complex IV () Cox5a Cytochrome c oxidase subunit 5A, mitochondrial −1.23 1.21E-03 Complex V (ATP synthase) Atp5b ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide −19.11 3.66E-23 Atp5f1 ATP synthase, H+ transporting, mitochondrial F0 complex, subunit B1 −3.52 1.51E-09 Atpaf2 ATP synthase mitochondrial F1 complex assembly factor 2 −1.75 4.79E-04 Atp5a1 ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit 1, car- 2.24 9.07E-10 diac muscle Atp5c1 ATP synthase, H+ transporting, mitochondrial F1 complex, gamma polypeptide 1 1.41 1.49E-06 Atp5d ATP synthase, H+ transporting, mitochondrial F1 complex, delta subunit 1.59 4.02E-11 Atp5e ATP synthase, H+ transporting, mitochondrial F1 complex, epsilon subunit 3.19 9.70E-13 Atp5h ATP synthase, H+ transporting, mitochondrial Fo complex, subunit d 2.17 8.17E-10 Atp5i ATP synthase, H+ transporting, mitochondrial Fo complex, subunit E 2.02 1.07E-07 Atp5j2 ATP synthase, H+ transporting, mitochondrial Fo complex, subunit F2 3.16 5.43E-05 Atp5l ATP synthase, H+ transporting, mitochondrial Fo complex, subunit G 15.94 1.02E-18 Atp5o ATP synthase, H+ transporting, mitochondrial F1 complex, O subunit 2.46 1.07E-10 Atpif1 ATPase inhibitory factor 1 1.50 8.26E-04 Other Etfa Electron transfer flavoprotein, alpha polypeptide −1.83 3.15E-12 Etfdh Electron transfer flavoprotein dehydrogenase −1.62 1.06E-05

AGS = arctic ground squirrel; ATP = adenosine triphosphate; CoQ = ; FDR = false discovery rate; HIB-AGS = hibernating arctic ground squirrel; NADH = nicotinamide adenine dinucleotide, reduced form.

we used mass spectrometry–based targeted metabolomic the acylcarnitine profile. Accummulation of toxic intermedi- profiling to provide additional insight into the energy meta- ates of lipid metabolism (acylcarnitines and ceramides) was bolic changes occurring in the hibernator heart during I/R found in LV myocardial tissue homogenates from rat com- stress. The most striking distinguishing metabolite signature pared to hibernating AGS at both reperfusion time points in the AGS compared to rat myocardium was exhibited by (fig. 6).

Anesthesiology 2016; 124:1296-310 1302 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

Fig. 3. Western blot analysis for succinate dehydrogenase A (SDHA), a protein in electron transport chain complex II. Blots are Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 shown for rat, summer active arctic ground squirrel (AGS Sum or SA-AGS), and hibernating arctic ground squirrel (AGS Win or HIB-AGS) at sham (S), 3 h (R3h), and 24 h of reperfusion (R24h) time points. Heat-shock protein 60 (HSP60) is shown as a mito- chondrial protein loading control (A). Densitometric analysis of HSP60 normalized SDHA bands demonstrating down-regulation in SA and HIB-AGS compared to rat under sham, R3h, and R24h conditions, consistent with the mass spectrometry findings (B). Results are displayed as mean ± SEM. Two-way ANOVA analysis identified both species and time point as being significant factors (P < 0.0001) affecting SDHA expression.

significantly different between rat and hibernating AGS in our analyses. We, therefore, conducted comparative analy- ses of PPAR-α activity, a master regulator of mitochondrial fatty acid utilization, in the hearts of rats and hibernating AGS in response to I/R. While PPAR-α activity levels sig- nificantly declined with I/R in rats, hibernating AGS were able to maintain baseline levels of PPAR-α activity despite I/R and may be involved in the up-regulation of fatty acid oxidation enzymes observed in AGS through its transactiva- tion effects (fig. 7).

Discussion Here, we report for the first time a cross-species analysis demonstrating cardioprotection in hibernating AGS com- pared with summer AGS or rat. After a clinically relevant I/R model and employing robust endpoint measures, hiber- nating AGS displayed significantly attenuated myocardial injury, as assessed by plasma levels of troponin I, myocar- Fig. 4. Proteomic analyses identified a significant increase in ex- dial apoptosis, and preservation of LV contractile function. pression of myocardial sirtuin-3 (SIRT3) in arctic ground squirrel Our comparative analyses employed two different strains of (AGS; both summer active [SA] and hibernating [HIB]) compared rat with known differences in susceptibility to I/R (Brown to rat, at every time point. Species differences also exist in the Norway and Dahl Salt Sensitive) and revealed a further sig- dynamic expression changes after ischemia–reperfusion injury nificant reduction in susceptibility in the hibernator heart. (I/R): unlike rats, AGS recover their baseline SIRT3 levels by 24 h of reperfusion (R24h). Data are shown as mean ± SD, N = 6 to Moreover, we were able to demonstrate a seasonal difference 8/group (A). Results were validated by Western blot and were in responses to I/R in AGS, with increased protection in corroborated with changes in proteins known to be regulated hibernating AGS. While AGS as a species seem to be resis- by SIRT3—up-regulation of manganese superoxide dismutase tant to I/R, many of the adaptations that allow maximum (MnSOD, T-total levels increased in AGS) and deacetylation of protection appear to maximize in the winter.26 Our experi- MnSOD (Ac-MnSOD levels decreased in AGS), which increase mental model of I/R injury employs standardized tempera- its scavenging activity. Extracellular ture (18°C), anesthetic, and cannulation strategies between signal–regulated kinase (ERK)1/2 is used as a loading control (B). *P < 0.05 by ANOVA with Tukey post hoc correction, rela- rat, summer AGS, and hibernating AGS, and therefore, tive to respective time point in the rat. R3h = 24 h of reperfusion; the observed cardioprotection in hibernating AGS can be T-MnSOD = total manganese superoxide dismutase. ascribed to adaptations that accompany the hibernator state, not to protective effects of hypothermia. While the prime regulatory mechanism underlying We undertook an unbiased approach to identify changes the hibernator phenotype remains unclear, key in metabolic enzymes and metabolite pools between species known to play a role in lipid metabolism and energy flux are and hibernation states in response to I/R stress that may shed

Anesthesiology 2016; 124:1296-310 1303 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021

Fig. 5. Functional sirtuin-3 (SIRT3) analyses in adult primary ventricular cardiomyocytes isolated from rat and arctic ground squirrel (AGS) and subjected to 2 h of oxygen and glucose deprivation followed by 24 h of reoxygenation. In rat cardiomyocytes, resveratrol (RES), an activator of SIRT3 (A, B), was associated with reduced apoptosis (C) and a trend toward reduced necrosis (D; *P < 0.05, **P < 0.01, ANOVA with Tukey post hoc correction), whereas nicotinamide (NAM), an inhibitor of SIRT3 (B), had the opposite effects (C, D, #P < 0.05, ##P < 0.01, ANOVA with Tukey post hoc correction). Two-way ANOVA analyses identified sig- nificant (P < 0.0001) treatment (RES vs. NAM) and concentration (5 and 10 μM) effects on SIRT3 activity (B) and cardiomyocyte apoptosis (C). Conversely, in AGS cardiomyocytes, short inhibitory RNA (siRNA)–mediated SIRT3 knockdown was associated with increased apoptosis, but not scrambled RNA (scrRNA). (E) Necrotic cell death (F) after simulated ischemia–reperfusion injury (**P < 0.01, Mann–Whitney U test). Data are presented as mean ± SD, and all experiments were conducted in triplicate. cTnI = cardiac troponin I.

light onto the mechanisms underlying cardioprotection in state28 (and a confusing term for hibernation biologists), hibernators. The most striking difference is the profound was found to have decreased quantities of all five ETC decline in expression of ETC proteins in hibernating AGS complex proteins, including NADH-ubiquinone oxidore- compared to rat, a pattern also associated with the most sig- ductase (NDUFs), SDH, UQCR, COX, and ATP synthase

nificant cardioprotection of all three groups. Recent work mitochondrial F1/F0 complex subunits. This quiescent state by an independent laboratory has characterized proteomic allows for the preservation of viable myocardium that can differences in adaptation to myocardial ischemia among regain function if blood supply is restored.29 Ironically, our nonhibernating mammals. Cabrera et al.27 examined the data demonstrate that the cardioprotective strategy adopted proteomic signatures of myocardial hibernation and isch- by hibernating mammals in response to I/R closely approxi- emic preconditioning in swine. Hibernating myocardium, mates the proteomic features of hibernating myocardium in an adaptation to chronic ischemia in which the myocardium nonhibernators, suggesting that at some level the regulatory decreases its oxygen consumption by adopting a quiescent components of the hibernator cardioprotective phenotype

Anesthesiology 2016; 124:1296-310 1304 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

Table 2. Changes in Protein Abundance for Key Regulators of Intermediary Metabolism

Fold Change HIB-AGS FDR-adjusted Protein Symbol Protein Name vs. Rat P Value

Acetyl CoA biosynthesis—PDH complex Pdha1 (lipoamide) alpha 1 −2.91 6.31E-19 Pdhb Pyruvate dehydrogenase (lipoamide) beta −4.58 1.02E-14 Dlat Dihydrolipoamide S-acetyltransferase −15.01 1.55E-16 Dld Dihydrolipoamide dehydrogenase −2.04 2.88E-14 Ketogenesis—ketolysis Acat1 Acetyl-CoA acetyltransferase 1 −3.12 9.54E-14 Bdh1 3-Hydroxybutyrate dehydrogenase, type 1 −1.46 1.60E-04 Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 Hadhb Hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase −1.74 7.89E-06 (trifunctional protein), beta subunit Oxct1 3-Oxoacid CoA transferase 1 −2.57 3.93E-15 Branched chain amino acid metabolism Acat1 Acetyl-CoA acetyltransferase, mitochondrial −3.12 9.54E-14 Aldh6a1 Methylmalonate-semialdehyde dehydrogenase [acylating], mitochondrial −5.55 4.17E-16 Bckdha Branched chain ketoacid dehydrogenase E1, alpha polypeptide −2.84 2.93E-19 Hadhb Trifunctional subunit beta, mitochondrial −1.74 7.89E-06 Hibch 3-hydroxyisobutyryl-CoA , mitochondrial −1.27 4.94E-04 Ivd Isovaleryl-CoA dehydrogenase, mitochondrial −4.60 1.90E-20 Mccc2 Methylcrotonoyl-CoA carboxylase beta chain, mitochondrial −185.75 3.58E-14 Oxct1 Succinyl-CoA:3-ketoacid coenzyme A transferase 1, mitochondrial −2.57 3.93E-15 Pcca Propionyl-CoA carboxylase alpha chain, mitochondrial −12.07 6.07E-15 Pccb Propionyl-CoA carboxylase beta chain, mitochondrial −5.40 5.11E-18 TCA cycle Aco2 Aconitate hydratase, mitochondrial −1.63 4.00E-07 Cs , mitochondrial −1.56 7.66E-04 Dld Dihydrolipoyl dehydrogenase, mitochondrial −2.04 2.88E-14 Dlst Dihydrolipoyllysine-residue succinyltransferase component of 2-oxoglutarate −1.75 1.57E-06 dehydrogenase complex, mitochondrial Idh2 [NADP], mitochondrial −5.69 4.07E-20 Idh3B Isocitrate dehydrogenase [NAD] subunit beta, mitochondrial −2.06 1.17E-11 Sdha Succinate dehydrogenase complex, subunit A −5.10 3.20E-16 Sdhb Succinate dehydrogenase complex, subunit B −2.60 3.05E-11 Sdhc Succinate dehydrogenase complex, subunit C −2.47 3.91E-08 Sucla2 Succinate-Coenzyme A , ADP-forming, beta subunit −1.50 1.27E-08 Suclg1 Succinyl-CoA ligase [ADP/GDP-forming] subunit alpha, mitochondrial −4.52 1.54E-19 Glycolysis and gluconeogenesis Aldoa Fructose-bisphosphate aldolase A −2.62 5.97E-12 Eno1 Alpha-enolase −40.94 9.81E-18 Eno3 Beta-enolase −31.90 2.67E-18 Gapdhs Glyceraldehyde-3-phosphate dehydrogenase, testis-specific −38.11 3.14E-17 Mdh1 , cytoplasmic −1.94 1.35E-11 Pgam1 Phosphoglycerate mutase 1 −2.41 1.83E-11 Pgk1 Phosphoglycerate kinase 1 −1.52 5.83E-08 Pgk2 phosphoglycerate kinase 2 −5.66 5.32E-04 Fatty acid metabolism Acaa2 Acetyl-CoA acyltransferase 2 1.20 3.97E-02 Acadsb Acyl-CoA dehydrogenase, short/branched chain 1.45 4.04E-02 Adh5 Alcohol dehydrogenase 5 (class III), chi polypeptide 695.23 3.41E-19 Aldh2 Aldehyde dehydrogenase 1 family, member A1 1.79 4.63E-07 Aldh5a1 Aldehyde dehydrogenase 4 family, member A1 1.93 1.52E-03 Cd36 Fatty acid 5.08 1.82E-09 Cpt1b Carnitine palmitoyltransferase 1b, muscle 4.88 3.71E-10 Dhrs4 Dehydrogenase/reductase (SDR family) member 4 2.78 2.04E-11 Echs1 Enoyl CoA hydratase, short chain, 1, mitochondrial 1.36 8.39E-06 Fabp4 Fatty acid binding protein 4, adipocyte 1.78 5.72E-11 Hadha Hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase 2.49 7.40E-11 (trifunctional protein), alpha subunit Hsd17b8 Hydroxysteroid (17-beta) dehydrogenase 8 5.39 7.01E-17 Slc25a3 Solute carrier family 25 (, phosphate) carrier), member 3 21.60 3.02E-26 Slc25a4 Solute carrier family 25 (mitochondrial carrier; adenine) nucleotide translocator), member 4 4.13 1.01E-14

ADP = adenosine diphosphate; AGS = arctic ground squirrel; CoA = coenzyme A; FDR = false discovery rate; GDP = guanosine diphosphate; HIB-AGS = hibernating arctic ground squirrel; NAD = nicotinamide adenine dinucleotide, oxidized form; NADP = nicotinamide adenine dinucleotide phosphate, oxidized form; PDH = pyruvate dehydrogenase; SDR = short-chain /reductases; TCA = tricarboxylic acid.

Anesthesiology 2016; 124:1296-310 1305 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator

Fig. 6. Metabolic correlates of hibernator cardioprotective phenotype. We used mass spectrometry–based metabolomics to quantify ischemia–reperfusion injury induced changes in targeted metabolites between hibernating arctic ground squirrels (HIB Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 AGS) and rats. Accumulation of toxic intermediates of lipid metabolism, acylcarnitine (C4DC acylcarnitine; A) and ceramide species (C18-ceramide and d18:1/C18-ceramide; B, C), was found in left ventricular myocardial tissue homogenates from rats compared to arctic ground squirrels, at both the 3-h (R3h) and 24-h (R24h) reperfusion time points, sham surgery (S) is shown for comparison (*P < 0.05, ANOVA with Tukey post hoc correction). Data are presented as mean ± SD, N = 6/group.

may elicit a conserved response to chronic ischemia in all body temperature is much lower), mitochondrial respira- mammals. However, hibernating AGS hearts also down-reg- tion increases significantly. This pattern suggests that body ulate the molecular machinery involved in glycolysis, TCA temperature–sensitive, enzyme-mediated posttranslational cycle, and the metabolism of ketones and amino acids. This modifications of oxidative phosphorylation complexes like is in contrast to what is known about hibernating myocar- differential acetylation could be involved. Although the dium in mammals that do not seasonally hibernate such as heart accounts for less than 0.5% of basal metabolic rate, rats or , which is more reliant on glycolysis.30 Our cardiac mitochondria from AGS display 60% reductions in comparative analyses revealed that rats suffer more myocar- respiration rates during torpor.38 Inhibition of succinate- dial injury and have a proteomic profile characterized by fueled state 3 mitochondrial respiration has been observed maintenance of ETC protein abundance. This more closely in three different tissues (heart, skeletal muscle, and liver), approximates the adaptations seen in ischemic precondition- appearing to be a hallmark of hibernation in AGS. The ing. This occurs when the myocardium is exposed to brief underlying mechanisms remain unclear, but do not appear nonlethal periods of ischemia and becomes resistant to fur- to involve inhibitions of either succinate oxidation (SDH ther ischemic insults,31–33 yet it does not significantly reduce activity)39 or transport (dicarboxylate transporter). None- ETC proteins.27 In our model, rats demonstrate inferior theless, the report of sirtuin-3–mediated deacetylation of cardioprotection to AGS associated with a pattern of main- SDHA regulating ETC complex II activity40 is of particu- tained expression of ETC proteins and increased glycoly- lar relevance to hibernators, given the profound suppres- sis, ketolysis, and amino acid metabolism. Consistent with sion of succinate oxidation during torpor. Although we these findings, reversible pharmacologic blockade of electron did not specifically assess acetylation of ETC complexes in transport at the onset of reperfusion was recently reported to our study, increased myocardial expression of sirtuin-3 in decrease cardiac injury in aged hearts by improving the inner AGS was associated with activation of key cardioprotective mitochondrial membrane potential.34 targets like MnSOD and consequently reduced oxidative The cardioprotective adaptations in hibernators were stress. We are providing a preliminary characterization of associated with robust up-regulation of sirtuin-3 in AGS sirtuin-3 involvement in the hibernator cardioprotective hearts, the main mitochondrial protein deacetylase, and phenotype, by reporting in cardiomyocyte models of I/R key regulator of metabolic (including fatty acid oxidation) the association between sirtuin-3 gain- and loss-of-function stress-response pathways in the heart.35 Increasing evidence and changes in cell survival. Furthermore, consistent with also suggests sirtuin involvement in regulating the hypomet- the differences in sirtuin-3 expression, rat cardiomyocytes abolic states associated with hibernation. During entrance had hyperacetylated mitochondrial proteins compared to into torpor, reductions in metabolism precede reductions AGS, which was further increased after I/R. in body temperature (even when thermogenesis is not A precisely controlled fuel shift from myocardial car- active) suggesting active mechanisms regulating metabolic bohydrate to fatty acid metabolism in hibernating AGS suppression rather than passive thermal effects.8,36 Mito- appears to be orchestrated through down-regulation of key chondrial respiration decreases quickly during entrance metabolic enzymes associated with glycolysis, TCA cycle, into torpor when body temperature is high (e.g., by 70% ketolysis, and branched-chain amino acid catabolism, while between 37°C during interbout arousal and 30°C during increasing enzymes involved in fatty acid catabolism. This torpor entrance).36 This occurs faster than transcriptional represents something of a contradiction to classic teaching or translational changes; moreover, peptide elongation in for myocardial substrate utilization during ischemic stress, hibernators ceases below 18°C,37 yet in early arousal (when when the heart’s ability to process a wide variety of substrates

Anesthesiology 2016; 124:1296-310 1306 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

A 20 10 ) 0 3h 24h -10

-20

-30

-40 Rat ARα acvity changes (%sham -50 Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 PP -60 AGS

-70 Reperfusion mepoint (h)

Fatty Acid Fatty Acid Binding Extracellular B Protein Fatty Acid Cell Membrane Translocase Fatty Acid Intracellular Acyl Carnitine Palmitoyl Acyl Transferase Fatty Acid Acyl- Carnitine CoA CoA Synthetase

Carnitine Acyl Carnitine Palmitoyl Increased in Hib AGS vs. Rat Transferase Transferase Acyl Decreased in Hib AGS vs. Rat Carnitine Acyl

CoA Dicarboxylacylcarnitines Acyl CoA leak out of mitochondria Dehydrogenase

Enoyl CoA Hydratase

Hydroxyacyl CoA α

Dehydrogenase Transactivation AR PP

Acetyl CoA -oxidatio n PPRE Cytosol Acyltransferase β Intermembrane Space

Electron Transport Chain

Fig. 7. The hibernator cardioprotective phenotype is associated with preservation of peroxisome proliferator–activated receptor-α (PPAR-α) activity by transcription factor–binding assay in hibernating arctic ground squirrel (HIB-AGS) compared to rat, at both reperfusion time points (A). A diagram of lipid metabolism focused on lipid transport into the is given in B. Increased expression of multiple fatty acid transporters and enzymes of fatty acid metabolism may contribute to increased metabolic efficiency in HIB-AGS hearts. PPAR-α-regulated transactivation effects maintain lipid metabolism under conditions of physiologic stress. AGS = arctic ground squirrel.

as fuel would appear to be an advantage; moreover, oxida- levels of fatty acid oxidation enzymes is associated with pre- tion of carbohydrate, ketone, and amino acid fuel sources served activity of the PPAR-α transcription factor after I/R requires less oxygen than the catabolism of fatty acids.41 in AGS but not in rats. The mechanism of increased myocar- However, increased expression of enzymes involved in fatty dial injury in nonhibernators exposed to high fat may in part acid catabolism may help explain why hibernating AGS are be due to incomplete oxidation of fatty acids. Accumulation able to catabolize fats without suffering ill effects, even with of partially oxidized lipids and sphingolipids can give rise to the addition of I/R injury. We show that increased expression lipotoxicity as these reactive intermediates accumulate in the

Anesthesiology 2016; 124:1296-310 1307 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator

mitochondria of organisms unable to metabolize a large fat and other master regulators of metabolism may be key to the load.42 To investigate the role of lipotoxicity in our model, metabolic reprogramming associated with cardioprotection we used targeted metabolomic profiling to measure levels of in hibernators. Further understanding this unique model partially oxidized lipid intermediates previously implicated of extreme metabolic plasticity and developing strategies to as deleterious in I/R injury, like dicarboxylacylcarnitines and “switch” myocardial metabolism to resemble that naturally ceramides.43–45 Compared to rats, winter AGS have signifi- occurring in mammalian hibernators represent a transforma- cantly lower myocardial levels of C4-dicarboxylacylcarnitine tive approach that could ultimately have a positive impact and C-18 ceramide (fig. 6). During early reperfusion (R3h), in patients undergoing cardiac surgery and transplantation rats experience a rise in C4-dicarboxylacylcarnitines, indica- and victims of cardiac arrest, trauma, and hypothermia, in tive of mitochondrial dysfunction, whereas winter AGS addition to fundamentally advancing cardiovascular biology.

maintain their baseline levels. Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 In an attempt to employ an unbiased analysis of dif- Acknowledgments ferences in myocardial proteins between hibernators ver- The authors thank Jeanette Moore, Ph.D., Franziska sus nonhibernators, we have utilized a label-free proteomic Kohl, M.Sc., and Lori Bogren, Ph.D. (University of Alaska profiling method. This technique has the limitation of Fairbanks, Fairbanks, Alaska), for technical assistance with deep hypothermic circulatory arrest (DHCA) experiments; detecting high abundance proteins over rare ones. One Jun Yan, Ph.D. (Chinese Academy of Sciences-German shortcoming of our study is that regulatory proteins may Max Planck Society, Shanghai, China), for help in building not be adequately represented. Also, in this experiment, we phylogenetic orthology maps for cross-species analyses; have focused on the identification and abundance of pro- and G. Burkhard Mackensen, M.D., Ph.D. (Department of teins as a first step, and many regulatory steps are undoubt- Anesthesiology, University of Washington, Seattle, Washing- ton), for his role in developing the DHCA model in rodents. edly controlled by posttranslational modifications including This study was supported by the Foundation for Anesthe- phosphorylation, acetylation, neddylation, modification sia Education and Research (Schaumburg, Illinois) Research with small ubiquitin-like modifier, and ubiquitinylation. Fellowship Grant, Duke University Department of Anesthe- These modifications may exert effects by modifying protein siology, primary investigator (PI): Dr. Quinones, mentor: function or simply marking a protein for degradation. Oxi- Dr. Podgoreanu; National Institute of General Medical Sci- ences (Bethesda, Maryland) T32 GM08600, Duke University dation and partial degradation of protein might also not be Medical Center, fellow: Dr. Quinones; National Institutes of adequately detected by our technique. While we have been Health (Bethesda, Maryland) R01-HL092071, Duke Inno- able to demonstrate quantitative changes in ETC proteins vation Grant Award, Duke University Medical Center, PI: in whole myocardial tissue, we did not perform the analysis Dr. Podgoreanu; and American Heart Association (Dallas, in isolated mitochondria and thus cannot speak to changes Texas) 11BGIN7620055, Beginning Grant in Aid, Duke University Medical Center, PI: Dr. Zhang. in protein abundance within the mitochondria specifically. Our data relate to proteomic changes within a specific Competing Interests quantity of myocardial protein. We did, however, demon- strate that the ratio of mitochondrial to nuclear DNA is in The authors declare no competing interests. fact higher in AGS than rat, thus eliminating the possibil- ity that the decline in ETC proteins is secondary to falling Correspondence numbers of mitochondria. Address correspondence to Dr. Quinones: Department In summary, we present the first comparative proteomics of Anesthesiology, 2301 Erwin Rd, Box 3094, Duke University Medical Center, Durham, North Carolina 27710. study of myocardial protein expression changes after experi- [email protected]. This article may be mental I/R in hibernating versus nonhibernating mammals. accessed for personal use at no charge through the We show that several prominent features of myocardial I/R ­Journal Web site, www.anesthesiology.org. injury like myocardial necrosis, apoptosis, and mechani- cal stunning were significantly reduced in the hibernator References heart and accompanied by differential expression of proteins 1. Schwartz Longacre L, Kloner RA, Arai AE, Baines CP, Bolli R, mainly involved in regulation of mitochondrial fuel and Braunwald E, Downey J, Gibbons RJ, Gottlieb RA, Heusch G, Jennings RB, Lefer DJ, Mentzer RM, Murphy E, Ovize M, energy metabolism. Our data support the idea that mammals Ping P, Przyklenk K, Sack MN, Vander Heide RS, Vinten- have a dichotomous response to I/R injury, one pathway Johansen J, Yellon DM: New horizons in cardioprotection: leading toward the development of hibernating myocardium Recommendations from the 2010 National Heart, Lung, and and the other to ischemic preconditioning. Ironically, hiber- Blood Institute Workshop. Circulation 2011; 124:1172–9 2. Carey HV, Andrews MT, Martin SL: Mammalian hibernation: nating animals exhibit a seasonal cardioprotective phenotype Cellular and molecular responses to depressed metabolism that resembles hibernating myocardium with respect to the and low temperature. Physiol Rev 2003; 83:1153–81 decline of ETC protein abundance. In contrast, rats main- 3. Grabek KR, Karimpour-Fard A, Epperson LE, Hindle A, tain levels of ETC proteins consistent with what has been Hunter LE, Martin SL: Multistate proteomics analysis reveals novel strategies used by a hibernator to precondition the observed in models of ischemic preconditioning. Enhanced heart and conserve ATP for winter heterothermy. Physiol lipid metabolism possibly regulated by sirtuin-3, PPARs, Genomics 2011; 43:1263–75

Anesthesiology 2016; 124:1296-310 1308 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. PERIOPERATIVE MEDICINE

4. Kurtz CC, Lindell SL, Mangino MJ, Carey HV: Hibernation Canadian Critical Care Trials Group: Impact of hypotension confers resistance to intestinal ischemia-reperfusion injury. and low cerebral perfusion pressure on outcomes in children Am J Physiol Gastrointest Liver Physiol 2006; 291:G895–901 treated with hypothermia therapy following severe trau- 5. Lindell SL, Klahn SL, Piazza TM, Mangino MJ, Torrealba matic brain injury: A post hoc analysis of the Hypothermia JR, Southard JH, Carey HV: Natural resistance to liver cold Pediatric Head Injury Trial. Dev Neurosci 2010; 32:406–12 ischemia-reperfusion injury associated with the hibernation 21. Sydenham E, Roberts I, Alderson P: Hypothermia for trau- phenotype. Am J Physiol Gastrointest Liver Physiol 2005; matic head injury. The Cochrane Database of Systematic 288:G473–80 Reviews 2009:CD001048 6. Dave KR, Anthony Defazio R, Raval AP, Dashkin O, Saul 22. Glickman ME, Rao SR, Schultz MR: False discovery rate con- I, Iceman KE, Perez-Pinzon MA, Drew KL: Protein kinase trol is a recommended alternative to Bonferroni-type adjust- C epsilon activation delays neuronal depolarization dur- ments in health studies. J Clin Epidemiol 2014; 67:850–7 ing cardiac arrest in the euthermic arctic ground squirrel. 23. Kozinski M, Pstragowski K, Kubica JM, Fabiszak T, Kasprzak J Neurochem 2009; 110:1170–9 M, Kuffel B, Paciorek P, Navarese EP, Grzesk G, Kubica J: ACS

7. Dave KR, Prado R, Raval AP, Drew KL, Perez-Pinzon MA: The network-based implementation of therapeutic hypothermia Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021 arctic ground squirrel brain is resistant to injury from cardiac for the treatment of comatose out-of-hospital cardiac arrest arrest during euthermia. Stroke 2006; 37:1261–5 survivors improves clinical outcomes: The first European 8. Heldmaier G, Ortmann S, Elvert R: Natural hypometabolism experience. Scand J Trauma Resusc Emerg Med 2013; 21:22 during hibernation and daily torpor in mammals. Respir 24. Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Physiol Neurobiol 2004; 141:317–29 Gutteridge G, Smith K: Treatment of comatose survivors of 9. Frank CL, Karpovich S, Barnes BM: Dietary fatty acid com- out-of-hospital cardiac arrest with induced hypothermia. position and the hibernation patterns in free-ranging arctic N Engl J Med 2002; 346:557–63 ground squirrels. Physiol Biochem Zool 2008; 81:486–95 25. Engler RL: Lidoflazine in the treatment of comatose survivors 10. Shao C, Liu Y, Ruan H, Li Y, Wang H, Kohl F, Goropashnaya of cardiac arrest. N Engl J Med 1991; 325:1046–7 AV, Fedorov VB, Zeng R, Barnes BM, Yan J: Shotgun pro- 26. Barnes BM: Freeze avoidance in a mammal: Body tempera- teomics analysis of hibernating arctic ground squirrels. Mol tures below 0 degree C in an Arctic hibernator. Science 1989; Cell Proteomics 2010; 9:313–26 244:1593–5 11. Williams DR, Epperson LE, Li W, Hughes MA, Taylor R, Rogers 27. Cabrera JA, Butterick TA, Long EK, Ziemba EA, Anderson LB, J, Martin SL, Cossins AR, Gracey AY: Seasonally hibernating Duffy CM, Sluiter W, Duncker DJ, Zhang J, Chen Y, Ward HB, phenotype assessed through transcript screening. Physiol Kelly RF, McFalls EO: Reduced expression of mitochondrial Genomics 2005; 24:13–22 electron transport chain proteins from hibernating hearts 12. Bouma HR, Carey HV, Kroese FG: Hibernation: The immune relative to ischemic preconditioned hearts in the second win- system at rest? J Leukoc Biol 2010; 88:619–24 dow of protection. J Mol Cell Cardiol 2013; 60:90–6 13. Yan J, Barnes BM, Kohl F, Marr TG: Modulation of gene 28. Rahimtoola SH: The hibernating myocardium. Am Heart J expression in hibernating arctic ground squirrels. Physiol 1989; 117:211–21 Genomics 2008; 32:170–81 29. Ceconi C, La Canna G, Alfieri O, Cargnoni A, Coletti G, 14. de Lange F, Yoshitani K, Podgoreanu MV, Grocott HP, Curello S, Zogno M, Parrinello G, Rahimtoola SH, Ferrari R: Mackensen GB: A novel survival model of cardioplegic arrest Revascularization of hibernating myocardium: Rate of meta- and cardiopulmonary bypass in rats: A methodology paper. bolic and functional recovery and occurrence of oxidative J Cardiothorac Surg 2008; 3:51 stress. Eur Heart J 2002; 23:1877–85 15. Jungwirth B, Mackensen GB, Blobner M, Neff F, Reichart B, 30. Vogt AM, Elsässer A, Nef H, Bode C, Kübler W, Schaper J: Kochs EF, Nollert G: Neurologic outcome after cardiopulmo- Increased glycolysis as protective adaptation of energy nary bypass with deep hypothermic circulatory arrest in rats: depleted, degenerating human hibernating myocardium. Mol Description of a new model. J Thorac Cardiovasc Surg 2006; Cell Biochem 2003; 242:101–7 131:805–12 31. Sumeray MS, Yellon DM: Ischaemic preconditioning reduces 16. Vilella AJ, Severin J, Ureta-Vidal A, Heng L, Durbin R, Birney infarct size following global ischaemia in the murine myocar- E: EnsemblCompara GeneTrees: Complete, duplication-aware dium. Basic Res Cardiol 1998; 93:384–90 phylogenetic trees in vertebrates. Genome Res 2009; 19:327–35 32. Sack S, Mohri M, Arras M, Schwarz ER, Schaper W: Ischaemic 17. Flicek P, Ridwan Amode M, Barrell D, Beal K, Billis K, Brent preconditioning—Time course of renewal in the pig. S, Carvalho-Silva D, Clapham P, Coates G, Fitzgerald S, Gil Cardiovasc Res 1993; 27:551–5 L, García Girón C, Gordon L, Hourlier T, Hunt S, Johnson 33. Schott RJ, Rohmann S, Braun ER, Schaper W: Ischemic pre- N, Juettemann T, Kähäri AK, Keenan S, Kulesha E, Martin conditioning reduces infarct size in swine myocardium. Circ FJ, Maurel T, McLaren WM, Murphy DN, Nag R, Overduin B, Pignatelli M, Pritchard B, Pritchard E, Riat HS, Ruffier Res 1990; 66:1133–42 M, Sheppard D, Taylor K, Thormann A, Trevanion SJ, Vullo 34. Chen Q, Ross T, Hu Y, Lesnefsky EJ: Blockade of electron A, Wilder SP, Wilson M, Zadissa A, Aken BL, Birney E, transport at the onset of reperfusion decreases cardiac injury Cunningham F, Harrow J, Herrero J, Hubbard TJP, Kinsella R, in aged hearts by protecting the inner mitochondrial mem- Muffato M, Parker A, Spudich G, Yates A, Zerbino DR, Searle brane. J Aging Res 2012; 2012:753949 SMJ: Ensembl 2014. Nucl Acids Res 2014; 42:D749–55 35. Hirschey MD, Shimazu T, Goetzman E, Jing E, Schwer B, 18. Li XC, Wei L, Zhang GQ, Bai ZL, Hu YY, Zhou P, Bai SH, Chai Lombard DB, Grueter CA, Harris C, Biddinger S, Ilkayeva OR, Z, Lakatta EG, Hao XM, Wang SQ: Ca2+ cycling in heart cells Stevens RD, Li Y, Saha AK, Ruderman NB, Bain JR, Newgard from ground squirrels: Adaptive strategies for intracellular CB, Farese RV Jr, Alt FW, Kahn CR, Verdin E: SIRT3 regu- Ca2+ homeostasis. PLoS One 2011; 6:e24787 lates mitochondrial fatty-acid oxidation by reversible enzyme 19. Turer AT, Stevens RD, Bain JR, Muehlbauer MJ, van der deacetylation. Nature 2010; 464:121–5 Westhuizen J, Mathew JP, Schwinn DA, Glower DD, Newgard 36. Tøien Ø, Blake J, Edgar DM, Grahn DA, Heller HC, Barnes CB, Podgoreanu MV: Metabolomic profiling reveals distinct BM: Hibernation in black bears: Independence of metabolic patterns of myocardial substrate use in humans with coro- suppression from body temperature. Science 2011; 331:906–9 nary artery disease or left ventricular dysfunction during sur- 37. van Breukelen F, Martin SL: Translational initiation is uncou- gical ischemia/reperfusion. Circulation 2009; 119:1736–46 pled from elongation at 18 degrees C during mammalian 20. Hutchison JS, Frndova H, Lo TY, Guerguerian AM; hibernation. Am J Physiol Regul Integr Comp Physiol 2001; Hypothermia Pediatric Head Injury Trial Investigators; 281:R1374–9

Anesthesiology 2016; 124:1296-310 1309 Quinones et al. Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited. Proteomics Cardioprotection in Hibernator

38. Chung D, Lloyd GP, Thomas RH, Guglielmo CG, Staples JF: 42. Drosatos K, Schulze PC: Cardiac lipotoxicity: Molecular path- Mitochondrial respiration and succinate dehydrogenase are ways and therapeutic implications. Curr Heart Fail Rep 2013; suppressed early during entrance into a hibernation bout, 10:109–21 but membrane remodeling is only transient. J Comp Physiol 43. Park TS, Hu Y, Noh HL, Drosatos K, Okajima K, Buchanan J, B 2011; 181:699–711 Tuinei J, Homma S, Jiang XC, Abel ED, Goldberg IJ: Ceramide 39. Brown JC, Staples JF: Substrate-specific changes in mitochon- is a cardiotoxin in lipotoxic cardiomyopathy. J Lipid Res drial respiration in skeletal and cardiac muscle of hibernat- 2008; 49:2101–12 ing thirteen-lined ground squirrels. J Comp Physiol B 2014; 44. Park TS, Goldberg IJ: Sphingolipids, lipotoxic cardiomyopa- 184:401–14 thy, and cardiac failure. Heart Fail Clin 2012; 8:633–41 40. Cimen H, Han MJ, Yang Y, Tong Q, Koc H, Koc EC: Regulation 45. Shah SH, Sun JL, Stevens RD, Bain JR, Muehlbauer MJ, of succinate dehydrogenase activity by SIRT3 in mammalian Pieper KS, Haynes C, Hauser ER, Kraus WE, Granger CB, mitochondria. Biochemistry 2010; 49:304–11 Newgard CB, Califf RM, Newby LK: Baseline metabolomic 41. Grynberg A, Demaison L: Fatty acid oxidation in the heart. profiles predict cardiovascular events in patients at risk for

J Cardiovasc Pharmacol 1996; 28(suppl 1):S11–7 coronary artery disease. Am Heart J 2012; 163:844–850.e1 Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/124/6/1296/487075/20160600_0-00023.pdf by guest on 02 October 2021

Anesthesiology 2016; 124:1296-310 1310 Quinones et al.

Copyright © 2016, the American Society of Anesthesiologists, Inc. Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.