Cardiac Metabolism Diabetic Cardiomyopathy

19
doi: 10.1152/ajpheart.00278.2006 291:H1489-H1506, 2006. First published 2 June 2006; Am J Physiol Heart Circ Physiol Ding An and Brian Rodrigues diabetic cardiomyopathy Role of changes in cardiac metabolism in development of You might find this additional info useful... 264 articles, 166 of which you can access for free at: This article cites http://ajpheart.physiology.org/content/291/4/H1489.full#ref-list-1 37 other HighWire-hosted articles: This article has been cited by http://ajpheart.physiology.org/content/291/4/H1489#cited-by including high resolution figures, can be found at: Updated information and services http://ajpheart.physiology.org/content/291/4/H1489.full can be found at: Physiology American Journal of Physiology - Heart and Circulatory about Additional material and information http://www.the-aps.org/publications/ajpheart This information is current as of November 20, 2012. ESSN: 1522-1539. Visit our website at http://www.the-aps.org/. Pike, Bethesda MD 20814-3991. Copyright © 2006 by the American Physiological Society. ISSN: 0363-6135, molecular levels. It is published 12 times a year (monthly) by the American Physiological Society, 9650 Rockville cardiovascular function at all levels of organization ranging from the intact animal to the cellular, subcellular, and physiology of the heart, blood vessels, and lymphatics, including experimental and theoretical studies of publishes original investigations on the American Journal of Physiology - Heart and Circulatory Physiology by guest on November 20, 2012 http://ajpheart.physiology.org/ Downloaded from

description

Cardiac Metabolism

Transcript of Cardiac Metabolism Diabetic Cardiomyopathy

Page 1: Cardiac Metabolism Diabetic Cardiomyopathy

doi: 10.1152/ajpheart.00278.2006291:H1489-H1506, 2006. First published 2 June 2006;Am J Physiol Heart Circ Physiol 

Ding An and Brian Rodriguesdiabetic cardiomyopathyRole of changes in cardiac metabolism in development of

You might find this additional info useful...

 264 articles, 166 of which you can access for free at: This article citeshttp://ajpheart.physiology.org/content/291/4/H1489.full#ref-list-1

 37 other HighWire-hosted articles: This article has been cited by http://ajpheart.physiology.org/content/291/4/H1489#cited-by

including high resolution figures, can be found at: Updated information and serviceshttp://ajpheart.physiology.org/content/291/4/H1489.full

can be found at: PhysiologyAmerican Journal of Physiology - Heart and Circulatory about Additional material and information

http://www.the-aps.org/publications/ajpheart

This information is current as of November 20, 2012.

ESSN: 1522-1539. Visit our website at http://www.the-aps.org/. Pike, Bethesda MD 20814-3991. Copyright © 2006 by the American Physiological Society. ISSN: 0363-6135,molecular levels. It is published 12 times a year (monthly) by the American Physiological Society, 9650 Rockville cardiovascular function at all levels of organization ranging from the intact animal to the cellular, subcellular, andphysiology of the heart, blood vessels, and lymphatics, including experimental and theoretical studies of

publishes original investigations on theAmerican Journal of Physiology - Heart and Circulatory Physiology

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 2: Cardiac Metabolism Diabetic Cardiomyopathy

Invited Review

Role of changes in cardiac metabolism in developmentof diabetic cardiomyopathy

Ding An and Brian RodriguesDivision of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences,The University of British Columbia, Vancouver, British Columbia, Canada

An, Ding, and Brian Rodrigues. Role of changes in cardiac metabolism in devel-opment of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 291: H1489–H1506, 2006. First published June 2, 2006; doi:10.1152/ajpheart.00278.2006.—Inpatients with diabetes, an increased risk of symptomatic heart failure usually developsin the presence of hypertension or ischemic heart disease. However, a predisposition toheart failure might also reflect the effects of underlying abnormalities in diastolicfunction that can occur in asymptomatic patients with diabetes alone (termed diabeticcardiomyopathy). Evidence of cardiomyopathy has also been demonstrated in animalmodels of both Type 1 (streptozotocin-induced diabetes) and Type 2 diabetes (Zuckerdiabetic fatty rats and ob/ob or db/db mice). During insulin resistance or diabetes, theheart rapidly modifies its energy metabolism, resulting in augmented fatty acid anddecreased glucose consumption. Accumulating evidence suggests that this alteration ofcardiac metabolism plays an important role in the development of cardiomyopathy.Hence, a better understanding of this dysregulation in cardiac substrate utilizationduring insulin resistance and diabetes could provide information as to potential targetsfor the treatment of cardiomyopathy. This review is focused on evaluating the acute andchronic regulation and dysregulation of cardiac metabolism in normal and insulin-resistant/diabetic hearts and how these changes could contribute toward the develop-ment of cardiomyopathy.

heart disease; diabetes; insulin resistance; lipoprotein lipase; fatty acid transporters;PPARs; lipotoxicity; mitochondria dysfunction

HEART DISEASE is a leading cause of death in diabetic patients(226, 253), with coronary vessel disease and atherosclerosisbeing primary reasons for the increased incidence of cardio-vascular dysfunction (151, 225, 253). However, a predisposi-tion to heart failure might also reflect the effects of underlyingabnormalities in diastolic function that can be detected inasymptomatic patients with diabetes alone (24, 61, 74). Theseobservations suggest a specific impairment of heart muscle(termed diabetic cardiomyopathy). Rodent models of chronicdiabetes demonstrate abnormalities in diastolic left ventricularfunction, with or without systolic left ventricular dysfunction(73, 205, 219). Because diastolic dysfunction is also seen inpatients with both Type 1 and Type 2 diabetes, it can beproposed that the diabetic state can directly induce abnormal-ities in cardiac tissue independent of vascular defects. Severaletiological factors have been put forward to explain the devel-opment of diabetic cardiomyopathy, including an increasedstiffness of the left ventricular wall associated with accumula-tion of connective tissue and insoluble collagen (9, 204, 212)and abnormalities of various proteins that regulate ion flux,specifically intracellular calcium (90, 235). More recently, theview that diabetic cardiomyopathy could also occur as aconsequence of metabolic alterations has been put forward(146, 147, 149).

Because uninterrupted contraction is a unique feature of theheart, cardiac muscle has a high demand for provision ofenergy. Under normal physiological conditions, hearts canutilize multiple substrates, including fatty acids (FA), carbo-hydrate, amino acids, and ketones (15). Among these sub-strates, carbohydrates and FA are the major sources fromwhich the heart derives most of its energy. In a normal heart,70% of ATP generation is through FA oxidation, whereasglucose and lactate account for �30% of energy provided tothe cardiac muscle (86, 173, 210). It should be noted that theheart can rapidly switch its substrate selection to accommodatedifferent physiological and pathphysiological conditions in-volving altered extracellular hormones, substrate availability, andworkload (energy demand) (11, 130, 204, 217, 227). Acutely orchronically, this regulation occurs through various mechanisms.Although substrate switching is essential to ensure continuousATP generation to maintain heart function, it has also beenassociated with deleterious consequences (17, 267).

In Type 1 and Type 2 diabetes mellitus, glucose uptake,glycolysis, and pyruvate oxidation are impaired. Additionally,lack of insulin function augments lipolysis and release of FAfrom adipose tissue. Under these conditions, the heart rapidlyadapts to using FA exclusively for ATP generation. Chroni-cally, this maladaptation is believed to play a key role in thedevelopment of cardiomyopathy. Hence, it is essential to identifyand understand the mechanisms that control substrate utilizationin the diabetic heart. This review discusses the acute and chroniccellular changes that modify cardiac metabolism and elucidatetheir roles in the development of cardiomyopathy.

Address for reprint requests and other correspondence: B. Rodrigues, Div. ofPharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The Univ.of British Columbia, 2146 East Mall, Vancouver, BC, Canada V6T 1Z3(e-mail: [email protected]).

Am J Physiol Heart Circ Physiol 291: H1489–H1506, 2006.First published June 2, 2006; doi:10.1152/ajpheart.00278.2006.

0363-6135/06 $8.00 Copyright © 2006 the American Physiological Societyhttp://www.ajpheart.org H1489

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 3: Cardiac Metabolism Diabetic Cardiomyopathy

DIABETIC CARDIOMYOPATHY

The central basis for diabetic heart failure is coronarydisease associated with atherosclerosis (151, 225, 253). Be-cause diabetes is often accompanied by hypertension and/orischemic heart disease, which often causes diastolic dysfunc-tion, it is difficult to attribute heart failure in these patients todiabetes alone (23). On the other hand, Type 1 or Type 2diabetic patients have also been diagnosed with reduced orlow-normal diastolic function and left ventricular hypertrophyin the absence of coronary heart disease or hypertension,identified as cardiomyopathy (24, 81, 192, 195, 200, 231).These abnormalities in asymptomatic diabetic patients poten-tially might predispose them to symptomatic heart failure whenthey develop hypertension or ischemic heart disease, particu-larly after myocardial infarction.

In animal models of Type 1 diabetes, either streptozotocin(STZ)-induced rats or genetic nonobese diabetic mice, systolicand diastolic dysfunction have been demonstrated using in vivoor in vitro measurements (177, 185, 232, 239). For example,systolic and diastolic dysfunction have been detected by usingechocardiography in STZ diabetic animals (123, 177). With theuse of in vivo catheterization of these animals, elevated leftventricular end-diastolic pressure and reduced left ventricularsystolic pressure with diminished increase or decrease pressureover time (�dP/dt) were observed (124). Reduced peak leftventricular pressure and �dP/dt were confirmed in ex vivoperfused STZ hearts (239). Recently, evidence of cardiomyop-athy has also been reported in animal models of insulinresistance and Type 2 diabetes (ZDF rats and ob/ob or db/dbmice) (5, 21, 30, 266). In db/db mice, systolic and diastolicdysfunction were detected by echocardiography (218). Withthe use of ex vivo perfused hearts from db/db mice, augmentedleft ventricular end-diastolic pressure and reduced cardiacoutput and cardiac power have also been observed (21). Giventhat rodents are resistant to atherosclerosis, these models pro-vided strong evidence for the occurrence of diabetic cardiomy-opathy. However, in another study, no evidence of impairmentin cardiac function was detected using in ex vivo Langendorffperfusions of hearts from insulin resistant and obese Zuckerrats (224) and Type 2 diabetic Zucker diabetic fatty (ZDF) rats(250). It is likely that such inconsistencies arose due to therodent models used and the methods employed for evaluatingheart function in the different studies.

It should be acknowledged that the features of cardiomyop-athy between human diabetic patients and rodent diabeticmodels might be different. The development of diabetic car-diomyopathy is influenced by the severity and duration ofalterations in plasma parameters such as insulin, leptin, glu-cose, and lipids. Given the discrepancy in changes in thesehormones and substrates between human diabetic patients androdent models of Type 1 and Type 2 diabetes, the etiology andseverity of diabetic cardiomyopathy may vary (196). For ex-ample, STZ-induced diabetes exhibit severe hypoinsulinemia,whereas ob/ob mice show leptin deficiency, which are rare inhuman diabetes. Additionally, it has been suggested that STZcan directly impair the cardiac contractile function of isolatedventricular myocytes (256). Finally, although db/db mice andZucker and ZDF rats exhibit hyperinsulinemia, hyperleptine-mia, and hyperglycemia, the severity of these alterations varybetween animal models and patients with diabetes (196).

Taken together, experimental data obtained using animalsmodels of diabetes should be used with caution when extrap-olating to the human condition.

Cardiomyopathy is a complicated disorder, and several fac-tors have been associated with its development. These includeincreased stiffness of the left ventricular wall associated withaccumulation of connective tissue and insoluble collagen (204,212), depressed autonomic function (73), impaired endothe-lium function and sensitivity to various ligands (e.g., �-ago-nists) (23), and abnormalities of various proteins that regulateion flux, specifically intracellular calcium (90, 235). Morerecently, there is a generalized view that diabetic cardiomyop-athy also occurs as a consequence of altered fuel metabolism.During insulin resistance or diabetes, glucose utilization iscompromised. This alteration, together with increased FA sup-ply, switches cardiac energy generation to utilization of FA.High FA uptake and metabolism not only augment accumula-tion of FA intermediates and triglycerides but also increaseoxygen demand and generation of reactive oxygen species(ROS), leading to cardiac damage. Interestingly, increasing FAuptake through overexpression of cardiac human lipoproteinlipase (LPL) (260) or FA transport protein (47), or augmentingFA oxidation through overexpression of cardiac peroxisomeproliferator-activated receptor (PPAR)-� (79) or long-chainacyl CoA synthase (48), results in a cardiac phenotype resem-bling diabetic cardiomyopathy. Conversely, normalizing car-diac metabolism in diabetic animals reverses the developmentof cardiomyopathy (21, 272). Taken together, these studiesstrongly support the role of altered metabolism in the devel-opment of diabetic cardiomyopathy.

CARDIAC METABOLISM

Glucose Uptake and Oxidation

Under normal physiological conditions, glucose is one of themajor carbohydrates utilized by the heart. Glucose metabolismis regulated through multiple steps, including uptake, glycoly-sis, and pyruvate decarboxylation (Fig. 1). Cardiac glucoseuptake is dependent on the transmembrane glucose gradientand the content of sarcolemmal glucose transporters (GLUT1and GLUT4) (132, 154, 189). GLUT1 has a more pronouncedsarcolemmal localization and represents basal cardiac uptake.When compared with GLUT1, GLUT4 is the dominant trans-porter in the adult heart, and under basal conditions, a majorityof this transporter is located in an intracellular pool (154). Itstranslocation from an intracellular compartment to the sar-colemmal membrane requires insulin (154). Other than recruit-ing GLUT to the sarcolemmal membrane, insulin also influ-ences glucose transport through its regulation of GLUT geneexpression (137, 183). It should be noted that GLUT-mediatedglucose uptake could also be stimulated through insulin-inde-pendent mechanisms. For example, recent studies have dem-onstrated that AMP-activated protein kinase (AMPK) alsopromotes GLUT4 redistribution to the sarcolemmal membrane(143, 261).

Once inside the cardiomyocyte, glucose is broken downthrough glycolysis, a sequence of reactions that convert glu-cose into pyruvate. Phosphofructokinase-1 (PFK1), the enzymethat catalyzes the generation of fructose 1,6-bisphosphate fromfructose 6-phosphate, is a rate-limiting enzyme controllingglycolysis (57, 202). PFK-1 is inhibited by low pH, high

Invited Review

H1490 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 4: Cardiac Metabolism Diabetic Cardiomyopathy

intracellular citrate, or ATP and is activated by ADP, AMP,phosphate, and fructose 2,6-bisphosphate (57, 202, 228). Fruc-tose 2,6-bisphosphate is formed from fructose 6-phosphatecatalyzed by PFK-2 (110). Given that PFK-2 is phosphorylatedand activated by hormones, such as insulin (25, 58) andglucagon (228) or kinases such as AMPK (109, 161), stimu-lation of PFK-2 through these mechanism increases fructose2,6-bisphosphate generation, activates PFK-1, and subse-quently promotes glycolysis. In an aerobic heart, glycolysiscontributes �10% of total ATP generated, with no oxygenconsumption.

After glycolysis, the pyruvate generated has three destina-tions: carboxylation to oxaloacetate or malate, reduction tolactate, and most importantly, decarboxylation to acetyl-CoA.To be oxidized, pyruvate is transported into the mitochondriaand decarboxylated to acetyl-CoA through pyruvate dehydro-genase (PDH), a multienzyme complex. PDH is phosphory-lated and inactivated by pyruvate dehydrogenase kinase (PDK)

(107, 258), which is inhibited by pyruvate and stimulated byhigh mitochondrial acetyl-CoA-to-CoA and NADH-to-NAD(�) ratios (29, 104). Acetyl-CoA then enters the tricar-boxylic acid cycle and is eventually broken down to H2O andCO2 for ATP generation. Oxidation of one glucose provides 30ATP with 6 oxygen consumed, which combined with 2 glyco-lytic ATPs produces a phosphate-to-oxygen ratio of 2.58 ATP/oxygen atom. Although glucose is the focus in this review,other carbohydrates like lactate also make an importantcontribution toward cardiac energy generation (39). In fact,as greater lactate compared with glucose utilization is ob-served in the isolated working heart (119, 127, 148), theinclusion of lactate in the perfusion buffer is recommendedin future studies.

FA Uptake and Utilization

When compared with glucose, FA is the preferred substrateand accounts for �70% of ATP generated in an aerobic heart.FA metabolism includes multiple steps and can be regulated byboth acute and chronic mechanisms, with or without modula-tion of gene expression (Fig. 2).

Lipoprotein lipase. Because the heart has limited capacity tosynthesize and store FA, it relies on continuous exogenoussupply. FA supplied to the heart from the circulation is fromtwo sources: albumin-bound FA and triglyceride (TG)-richlipoproteins. It should be noted that the molar concentration ofFA in lipoprotein-TG is �10-fold larger than FA bound toalbumin (165). Lipoprotein lipase (LPL) is the key enzyme thathydrolyzes lipoproteins to release FA. Thus, when lipoproteinsare hydrolyzed by LPL, a large amount of FA are released andare believed to be the principle source of FA supplied to theheart (14, 237). LPL is produced in cardiomyocytes and sub-sequently secreted onto heparan sulfate proteoglycan bindingsites on the myocyte cell surface (33, 68, 70). From here, LPLis transported onto comparable binding sites on the luminalsurface of coronary endothelial cells (193). At these sites, LPLhydrolyzes TG-rich lipoproteins, such as very-low-density li-poprotein (VLDL) and chylomicrons, to release FA. Perfusionof working hearts with VLDL or chylomicrons, in the absenceor presence of FA, has revealed that, compared with chylomi-crons, VLDL is a poor substrate for LPL (100). In addition,utilization of chylomicrons was inhibited by free FA, whichfailed to affect cardiac VLDL utilization (100). Given theimportant role that LPL plays in regulating FA delivery,alteration in its level is able to change FA delivery andsubsequent oxidation. Indeed, overexpression of LPL in theheart or skeletal muscle accelerates FA uptake (141, 260).Conversely, tissue-specific knockout of LPL in the heartswitches the cardiac substrate selection preference to glucose(12, 13). Other than hydrolysis by LPL, lipoproteins can alsobe transported into the cardiac tissue with help of the lipopro-tein receptor, with VLDL uptake through this mechanismbeing more substantial compared with chylomicrons (178). Inperfused working hearts, inhibition of the lipoprotein receptorsignificantly reduced FA oxidation without altering FA uptake,suggesting that lipoprotein receptor mediated FA uptake chan-nels FA toward oxidation, whereas LPL mediated FA uptakechannels FA toward both oxidation and TG storage (178).

FA transporters. Although FA can translocate into the cellthrough passive diffusion across the plasma membrane, FA

Fig. 1. Glucose utilization in the cardiomyocyte. Glucose uptake into cardio-myocyte occurs through GLUT1 and GLUT4 transporters. Once inside, glu-cose is broken down through glycolysis, a sequence of reactions that convertglucose into pyruvate. Phosphofructokinase-1 (PFK1), the enzyme that cata-lyze the generation of fructose 1,6-bisphosphate from fructose 6-phosphate, isa rate-limiting enzyme controlling glycolysis. PFK1 is activated by 2,6-bisphosphate, which is formed from fructose 6-phosphate catalyzed by PFK-2.After glycolysis, the pyruvate generated is transported into mitochondria anddecarboxylated to acetyl-CoA through pyruvate dehydrogenase (PDH), amultienzyme complex. PDH is phosphorylated and inactivated by pyruvatedehydrogenase kinase (PDK). Acetyl-CoA then enters tricarboxylic acid cycle(citric acid cycle) and is eventually broken down to H2O and CO2 for ATPgeneration.

Invited Review

H1491ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 5: Cardiac Metabolism Diabetic Cardiomyopathy

uptake shows saturation kinetics and is inhibited by proteases(7, 158, 159, 230). Thus FA transporters are also likely re-quired to support this process. In the heart, three FA transport-ers have been identified and these include CD36, FA transportprotein (FATP), and FA binding protein plasma membrane(FABPpm) (157). Given that 55–80% of FA transport wasblocked using general transporter inhibitors (131, 158) and thatoverexpression of CD36 or FATP has been found to dramati-cally increase FA metabolism (47, 113), these FA transportersare believed to play a key role in FA delivery to the cardiactissue.

Regulation of FA transport proteins occurs through differentmechanisms. In severe STZ-induced diabetes, expression ofcardiac CD36 and FABPpm were augmented (152), suggestingtranscriptional modification of these transporters. At present,the mechanisms that induce this change have yet to be eluci-dated. Additionally, because muscle contraction or acute insu-lin treatment does not change protein but simply relocatesCD36 from an intracellular pool to the sarcolemmal membrane(155, 156), posttranslational regulation of this transporter hasalso been suggested.

Acyl-CoA synthase. Acyl-CoA synthase (ACS) catalyzes theesterification of FA to fatty acyl-CoA, the initial step of FAmetabolism. Fatty acyl-CoA can be transported into the mito-chondria for oxidation or used for intracellular TG synthesis.The fate of fatty acyl-CoA is influenced by the location ofdifferent ACS isoforms, energy demand, and the availability ofFA (37). Moreover, ACS not only functions as an enzyme-catalyzing esterification but is also actively involved in con-trolling FA homeostasis (48). Recent studies have demon-strated that ACS is associated with CD36 or FATP on thecytosolic side of the sarcolemmal membrane (84, 201, 214),suggesting that ACS also influences FA uptake. Several studiesalso suggest that FATP1 is a very long chain ACS (53).Overexpression of ACS in the heart or fibroblast causes dra-matically augmented FA uptake and intracellular TG accumu-lation (48).

Under normal conditions, 70–90% of the esterified FA thatenters cardiomyocytes is oxidized for ATP generation, whereas10–30% is converted to TG (228). The TG pool is not static,with lipolysis and esterification taking place continuously(240). In a normal heart, intracellular TG level is constant,indicating a balance of lipogenesis and lipolysis (210). Insituations where FA supply supercedes the cellular oxidativecapacity, such as obesity or diabetes, intracellular TG accumu-lates and is associated with lipotoxicity (240). Although TG isunlikely to be a direct mediator of cell apoptosis, its augmentedlipolysis expands fatty acyl-CoA levels, which may be a keyfactor mediating cell apoptosis (117, 240). Thus TG is oftenused as a marker of lipotoxicity.

PPARs. PPARs are a group of ligand-activated transcrip-tional factors belonging to the superfamily of nuclear recep-tors. They are activated by either natural ligands like FA ornumerous pharmacological ligands (80). Once activated,PPARs form complexes with retinoid X receptors and bind tothe promoter regions of a number of target genes that encodethe proteins involved in controlling FA metabolism (76, 78).Through regulation of expression of these genes, PPARs mod-ulate FA utilization at the transcriptional level. PPARs havethree isoforms: PPAR-�, PPAR-� (or -�), and PPAR-�.PPAR-� is extensively expressed in tissues with high FAmetabolism like the heart (17). Activated by elevated intracel-lular FA levels, PPAR-� promotes expression of genes thatregulate FA oxidation at various steps, such as FA uptake andbinding (LPL, CD36, and FA binding protein), FA esterifica-tion (ACS), and FA oxidation (carnitine palmitoyltrans-ferase-1, acyl-CoA oxidase, long-chain acyl-CoA dehydroge-nase, and very-long-chain acyl-CoA dehydrogenase) (76, 78,111). Knocking out cardiac PPAR-� abolishes fasting-inducedoverexpression of FA metabolic genes and switches substrateselection from FA to glucose (140, 172). Overexpression ofcardiac PPAR-� augments FA uptake and oxidation (77, 79).Taken together, PPAR-� is believed to be the primary regula-tor of FA metabolism in the heart. Similar to PPAR-�, PPAR-�

Fig. 2. Control of fatty acid (FA) delivery andutilization in the cardiomyocyte. FA, either fromadipose tissue or released from triglyceride(TG)-rich lipoproteins through hydrolysis by li-poprotein lipase (LPL), is taken up into thecardiomyocyte by three FA transporters: CD36,FA transport protein (FATP), and FA bindingprotein plasma membrane (FABPpm). FA is con-verted to fatty acyl-CoA, which is transportedinto the mitochondria through CPT1/CPT2. In-side the mitochondria, fatty acyl-CoA undergoes�-oxidation to generate acetyl-CoA, which isfurther oxidized in the tricarboxylic acid cycle.Utilization of FA is regulated through differentmechanisms. FA, through activation of peroxi-some prolferator-activated receptor-� (PPAR-�), increases the expression of a number ofenzymes involved in FA oxidation. Malonyl-CoA, which is generated through carboxylationof acetyl-CoA catalyzed by acetyl-CoA carbox-ylase (ACC), inhibits CPT-1 and FA oxidation.AMP-activated kinase (AMPK) inhibits ACC,relieves its inhibition on CPT-1, and promotesFA oxidation. Similarly, malonyl-CoA decar-boxylase (MCD), through decreasing malonyl-CoA by decarboxylating it to acetyl-CoA, en-hances CPT-1 and FA oxidation.

Invited Review

H1492 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 6: Cardiac Metabolism Diabetic Cardiomyopathy

(or -�) is expressed abundantly in the cardiac tissue (17).Activated by elevated intracellular FA (45), PPAR-� (or -�)augments expression of a group of genes that promote FAutilization (63, 169). Cardiac-specific knockout of PPAR-�also decreases FA oxidative gene expression and FA oxidation(46). Although the targets of PPAR-� and PPAR-� are par-tially overlapping (169), their unique roles and interactionremains unclear in the heart. PPAR-�, the third number of thePPAR family, is highly expressed in adipose tissue. Throughpromoting lipogenic gene expression, PPAR-� controls lipo-genesis. Loss and gain of function experiments have demon-strated that PPAR-� is necessary for adipose tissue prolifera-tion and differentiation (16, 238). In isolated cardiomyocytes,the expression of PPAR-� is barely detectable (89), suggestinga limited direct role for this nuclear receptor in regulatingcardiac metabolism. However, the ability of PPAR-� agoniststo normalize elevated plasma concentrations of glucose and FAin diabetic animals will have a profound indirect effect oncardiac metabolism.

AMP-activated protein kinase. As an energy sensor, AMP-activated protein kinase (AMPK) is activated following a risein the intracellular AMP-to-ATP ratio (97). Once stimulated,AMPK switches off energy-consuming processes like proteinsynthesis, whereas ATP-generating mechanisms, such as FAoxidation and glycolysis, are turned on (96, 105). In heart andskeletal muscle, AMPK facilitates FA utilization through itscontrol of acetyl-CoA carboxylase (ACC) (133, 134). As ACCcatalyzes the conversion of acetyl-CoA to malonyl-CoA,AMPK by inhibiting ACC is able to decrease malonyl-CoAand minimize its inhibition of CPT-1, the rate-limiting en-zyme-controlling FA oxidation. AMPK has also been impli-cated in FA delivery to cardiomyocytes through its regulationof the FA transporter CD36 (155). Additionally, results fromour laboratory have demonstrated a strong correlation betweenactivation of whole heart AMPK and increases in coronary

lumen LPL activity (8). Interestingly, recent studies usingtransgenic mice with a dominant negative form of AMPK hasdemonstrated that the lack of AMPK does not affect cardiacmetabolism under physiological conditions (209, 259). Atpresent, it is unclear as to what compensatory mechanisms areactivated following knockout of AMPK. Additionally, it isunknown whether overexpression of AMPK could affect car-diac lipid homeostasis and metabolism.

Malonyl-CoA decarboxylase. In addition to AMPK, malo-nyl-CoA decarboxylase (MCD) is also known to promote FAoxidation through its lowering of malonyl-CoA. MCD cata-lyzes the degradation of malonyl-CoA to acetyl-CoA, leadingto reduction of malonyl-CoA (65). This action relieves theinhibition of CPT-1 by malonyl-CoA and favors FA oxidation.Recent studies have suggested that inhibition of cardiac MCDleads to accumulation of malonyl-CoA and reduced FA oxida-tion (66).

Interaction Between Glucose and FA Metabolism

Regulation of glucose and FA metabolism does not occurindependently, and numerous studies have reported a “cross-talk” between the utilization of these substrates (Fig. 3) (199,211, 233). Randle et al. (199) demonstrated that FA impairsbasal and insulin-stimulated glucose uptake and oxidation, anevent that is known as the “Randle cycle” (199). FA influencesglucose utilization at multiple levels. Accumulation of FAimpairs insulin-mediated glucose uptake through inhibition ofinsulin receptor substrate and protein kinase B (94, 115, 187).Accumulation of FA leads to augmented intracellular FAderivatives, such as fatty acyl CoA, diacylglycerol, and cer-amide. These FA metabolites activate a serine kinase cascade,which involves protein kinase C- and IB kinase-� (inhibitorof NF-B kinase-�), leading to serine phosphorylation of IRS(129, 269). Serine phosphorylation of IRS-1 reduces tyrosine

Fig. 3. Inhibition of glucose oxidation by FAutilization. Accumulation of FA impairs in-sulin-mediated glucose uptake through inhi-bition of insulin receptor substrate (IRS) andprotein kinase-B (PKB). Increased intracellu-lar FA also activates PPAR-�. As a conse-quence, PPAR-� promotes the expression ofgenes involved in FA oxidation, as well asPDK4, known to inhibit PDH and pyruvateflux. Increased acetyl-CoA and NADHcaused by the high rate of FA oxidationactivate PDK4, leading to further inactivationof PDH. Augmented acetyl-CoA also causesaccumulation of citrate in the cytosol, whichsubsequently inhibits PFK1 and glycolysis.

Invited Review

H1493ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 7: Cardiac Metabolism Diabetic Cardiomyopathy

phosphorylation and interferes with its ability to phosphorylateand activate phosphatidylinositol 3-kinase and protein kinase B(206). Increased intracellular FA also activates PPAR-�. As aconsequence, PPAR-� promotes the expression of genes in-volved in FA oxidation, as well as pyruvate dehydrogenasekinase-4 (PDK4), which is known to inhibit PDH and pyruvateflux (257). Moreover, increased acetyl-CoA-to-free CoA andNADH-to-NAD� ratios caused by the high rate of FA oxida-tion are also known to activate PDK4, leading to inactivationof PDH (29, 104). Augmented acetyl-CoA-to-free CoA ratioalso causes accumulation of citrate in the cytosol, whichsubsequently inhibits PFK and glycolysis (85, 175). Con-versely, inhibition of FA oxidation through elevation of mal-onyl-CoA levels, or using pharmacological inhibitors, favorsglucose oxidation.

ALTERATION OF CARDIAC METABOLISM DURING DIABETES

Changes in Plasma Substrates

Glucose and lipids are the major substrates affected bydiabetes. Hyperglycemia is a consequence of decreased glu-cose clearance and augmented hepatic glucose production,whereas enhanced lipolysis in adipose tissue and higher li-poprotein synthesis in the liver dramatically increases circulat-ing free FA and TG. Because glucose entry into the cell islargely dependent on insulin, whereas FA transport acrossplasma membrane does not require any hormone, the aug-mented circulating lipids increase FA delivery to the cardio-myocyte. With the increase of intracellular FA, cardiac tissuerapidly adapts to promote FA utilization. In addition to diabe-tes, augmentation of plasma lipids by fasting or intralipid-heparin infusion also increases cardiac FA oxidation (52, 266).Under conditions where FA supply supercedes the oxidativecapacity of the heart, the FA is converted to lipids like TG orceramide, with an end result being lipotoxicity (272). In obeseZDF rats, lowering of plasma lipids with a PPAR-� agonistreduced cardiac TG and ceramide and improved heart function(272). Overall, these studies suggest that alterations in plasmalipids may drive changes in cardiac metabolism. It should benoted that a recent study using 4-wk-old ob/ob and db/db micedemonstrated altered cardiac metabolism without any changein plasma substrates (30). Hence, in these genetic mice models,intrinsic changes in the cardiomyocyte, rather than changes incirculating substrates, initiate alterations in cardiac metabolismat this early time point.

Defects in Cardiac Carbohydrate Utilization

In the obese or diabetic heart, myocardial glucose utilizationis compromised at several points. In noninsulin-controlledType 1 diabetic animals, reduced GLUT gene and proteinexpression compromises cardiac glucose uptake and oxidation(34). In obese or Type 2 diabetic animals, although there ishyperglycemia and hyperinsulinemia, cardiac glucose uptake isreduced as a consequence of reduced GLUT4 protein andimpaired insulin signaling (38, 266). With the use of ob/ob anddb/db mice, a recent study (30) reported that cardiac glucoseoxidation is reduced at 4 wk of age and was associated withincreased FA oxidation. Interestingly, this lower glucose oxi-dation occurred before the onset of impaired insulin signalingin the heart and development of hyperglycemia. Thus this early

reduction in glucose utilization is likely due to suppression byhigh FA oxidation rather than impaired cardiac-specific insulinsignaling. In another study that used db/db mice at differentages, increased cardiac FA oxidation preceded the reduction inglucose oxidation (5). Higher FA oxidation increases citrate,which is known to inhibit PFK, the rate-limiting enzyme inglycolysis. Elevated intracellular FA is also known to increasePDK-4 expression, which phosphorylates and inhibits PDH.Finally, high rates of FA oxidation augment acetyl-CoA thatinhibits PDH either allosterically or through activation of PDK.

When compared with animal models, the use of carbohy-drates in the human diabetic hearts is more controversial. InType 1 diabetic patients, decreased myocardial carbohydrateuptake is reported (15, 62). With the use of an euglycemicinsulin clamp, another study (179) has shown that cardiacglucose uptake is normal in these patients, suggesting thatinsulin is the major limiting factor that influences cardiacglucose uptake and no cardiac-specific insulin resistance isevident, as observed in skeletal muscle. In Type 2 diabetes,cardiac expression of GLUT4 is compromised likely due toelevated FA (10). However, a number of studies have alsoreported that cardiac tissue from Type 2 diabetic patientsrespond normally to insulin and show regular glucose uptake,suggesting that myocardial insulin resistance is not a commonfeature of Type 2 diabetes (118, 160, 242). Interestingly, arecent study has suggested that impaired myocardial glucoseuptake is only observed in Type 2 diabetic patients withhypertriglyceridemia, suggesting that myocardial insulin resis-tance in these patients is associated with hypertriglyceridemiaand augmented plasma FA levels (168).

Cardiac lactate utilization is also compromised followingdiabetes. In STZ-induced Type 1 diabetes, cardiac utilizationof lactate is reduced to a greater extent when compared withglucose oxidation (41, 42). Moreover, hearts from 12-wk-oldZDF rats also showed lower carbohydrate oxidation, and thiswas almost entirely due to a reduction in lactate rather thanglucose oxidation (43, 250). The mechanisms that mediate theinhibition of lactate utilization are unclear but is likely inde-pendent of any alterations in lactate dehyrogenase or lactatetransporter (41).

The shift of cardiac energy substrate utilization from carbo-hydrate to lipids increases the intracellular glycogen pool,probably through augmented glycogen synthesis, or impairedglycogenolysis, or a combination of both processes (103, 136,167, 223). Emerging evidence indicates that glycogen, inaddition to its role as energy storage, is also able to regulatemetabolism. At least in skeletal muscle, accumulation of gly-cogen acutely alters glycogen synthesis and glucose metabo-lism (59, 120–122). Chronically, glycogen accumulation mayalso impair insulin signaling in skeletal muscle (121). Whethercardiac glycogen accumulation also influences insulin signal-ing and metabolism is unknown. In addition, recent studieshave identified a glycogen-binding domain in the �-subunit ofAMPK and suggests that this domain links AMPK and glyco-gen (108). Although an association between high glycogenlevels and repressed AMPK activity in human and rat skeletalmuscle have been documented (254, 255), it is still unclearwhether glycogen is able to regulate metabolism throughAMPK.

Invited Review

H1494 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 8: Cardiac Metabolism Diabetic Cardiomyopathy

Alterations in FA Utilization

Utilization of FA by cardiac tissue increases followingobesity and diabetes. This change occurs not only as a conse-quence of increased FA supply but also through an intrinsicadaptation/maladaptation to elevated FA (234, 267). It must beacknowledged that some studies have also observed a reduced,rather than augmented, FA oxidation in Zucker fatty or ZDFrats (266, 272). In human diabetic patients, obese womendemonstrate increased FA utilization, associated with aug-mented cardiac oxygen consumption, and reduced cardiacefficiency (190). Moreover, elevated cardiac TG and increasedexpression of PPAR-� target genes have been observed inpatients without ischemic heart disease, very similar to lipo-toxic ZDF rat hearts (220).

Lipoprotein lipase. The amount of FA supplied to the heartthrough LPL is influenced by multiple factors, including LPLactivity and plasma lipoprotein concentration and composition.The relative contribution of cardiac LPL activity to the deliveryof free FA to the diabetic heart is inconclusive. Thus LPLimmunoreactive protein or activity has been reported to beunchanged, increased, or decreased in the diabetic rat heart. Inpart, this variability between different studies could be due tothe diversity in the rat strains used, the dosage of STZ used toinduce diabetes, and the duration of the diabetic state (203). Inaddition, many of the above investigations utilized proceduresthat did not distinguish between functional (i.e., heparin-releas-able component localized on capillary endothelial cells that isimplicated in the hydrolysis of circulating TG) and cellular(i.e., non-heparin-releasable pool that represents a storage formof the functional enzyme) pools of cardiac LPL because cel-lular LPL activity or protein levels have largely been obtainedusing whole heart homogenates. Rodrigues et al. (203) havepreviously reported that in STZ (55 mg/kg)-induced moderatediabetic Wistar rat hearts, HR-LPL activity is significantlyincreased. Induction of more severe diabetes using 100 mg/kgSTZ did not influence HR-LPL (203). In another study usinghearts from 65 mg/kg STZ-induced diabetic rats, HR-LPLdecreased and was associated with reduced VLDL-TG lipoly-sis (182). Acute treatment of these diabetic rats with insulinenhanced both HR-LPL and VLDL-TG lipolysis (182). Inmouse models of diabetes, although there is no change ofcardiac HR-LPL activity in either STZ or db/db hearts, utili-zation of chylomicron-TG increases (174). Hence, the mecha-nisms that control cardiac LPL during diabetes are complexand have yet to be completely resolved.

It should be noted that even though LPL may not changefollowing obesity or diabetes, increased circulating lipopro-teins through LPL cleavage could still elevate FA supply to thecardiac tissue. However, increased concentration could beoffset by changes in lipoprotein composition or lipoproteinreceptor number. Binding of lipoproteins containing apoli-poprotein (apo) CII to LPL enhances lipolysis (181), whereasbinding of lipoproteins containing apoCIII or apoE suppressesLPL activity (1, 2). A recent study has shown that overexpres-sion of HDL-associated apo AV accelerates hydrolysis ofTG-rich lipoproteins, indicating the importance of this apolipoprotein in stimulating LPL (166, 188). O’Looney et al.(181) have shown that induction of diabetes by STZ changeslipoprotein composition, with reduced apoCII levels, leading toimpaired VLDL-TG lipolysis. A recent study has reported that

expression of VLDL receptor decreases following STZ-in-duced diabetes (116).

FA transporters. During obesity and diabetes, the high rateof FA uptake is facilitated by FA transporters, leading toaugmented FA oxidation and TG storage. In STZ-induceddiabetes, the increase in plasmalemmal CD36 and FABPpm

amplifies FA uptake, an effect resulting from increased CD36and FABPpm protein expression (152). In Zucker fatty rats,without a change in total protein, permanent relocation ofCD36 and FABPpm to the cardiomyocyte plasmalemmal mem-brane augments FA uptake (55, 56, 153). The mechanism forthis permanent repositioning of FA transporters at the plasmamembrane is still unknown. Interestingly, in young (4 wk)ob/ob or db/db mice, cardiac FA oxidation increases withoutany change in circulating substrates (30). Given that the highrate of FA oxidation requires coordinated FA uptake, augmen-tation of CD36-mediated FA supply is suggested to be in-volved in facilitating FA oxidation at this early stage of insulinresistance.

PPAR-�. As the principle regulator of cardiac FA metabo-lism, PPAR-� plays an important role in controlling FA oxi-dation during obesity and diabetes. Activation of this nuclearreceptor in the heart has been reported in almost all obese ordiabetic animal models, including STZ-induced diabetic rats,ZDF rats, and ob/ob and db/db mice (30, 79, 220). Given thatFA and its derivatives activate PPAR-�, higher cardiacPPAR-� activity is always observed when circulating lipids areaugmented (30, 171). Hence, in ob/ob and db/db mice, activa-tion of PPAR-�, evidenced by elevated expression of itsdownstream targets, is only observed at 12 wk and is associ-ated with augmented plasma lipid (30). A similar associationbetween plasma lipids and PPAR-� is also seen in hearts fromSTZ-induced diabetic rats, with only chronic (6 wk) but notacute (4 days) diabetes demonstrating activation of PPAR-�(unpublished data). Interestingly, in acute STZ diabetes or4-wk ob/ob or db/db mice, increased FA oxidation is observedeven in the absence of any change in cardiac PPAR-� and itsdownstream targets, suggesting PPAR-� independent controlof FA oxidation. Identification of these mechanisms requiresfurther studies.

After activation, cardiac PPAR-� promotes the expressionof a group of genes involved in various steps of FA oxidation.Simultaneously, activation of PPAR-� also reduces expressionof genes involved in glucose uptake, glycolysis, and oxidationthrough direct or indirect mechanisms (77, 79). With the use oftransgenic mice, a recent study (186) demonstrated that knock-ing out cardiac PPAR-� prevented suppression of GLUT4expression and glucose uptake by elevated plasma FA andimproved myocardial recovery from ischemia following STZdiabetes induction, high-fat feeding, or fasting. Taken together,activation of cardiac PPAR-� not only favors FA oxidation butalso inhibits glucose uptake and utilization, leading to aug-mented susceptibility to ischemic damage.

MCD. In STZ-induced diabetes, overexpression of cardiacMCD protein was observed and could contribute to the highrate of FA oxidation (213). It is unknown whether MCD alsoplays a role in augmenting cardiac FA oxidation in obese andType 2 diabetes. Nevertheless, given that PPAR-� increasesMCD expression (138, 265), higher MCD protein levels areexpected following PPAR-� activation during obesity andType 2 diabetes.

Invited Review

H1495ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 9: Cardiac Metabolism Diabetic Cardiomyopathy

Limitations of Metabolic Measurements UsingRadiolabeled Substrate

The majority of metabolic data in the above studies wereobtained by using radiolabeled substrate perfusion of isolatedhearts. In these methods, [3H]glucose and [14C]glucose wereused to evaluate glycolysis and glucose oxidation, whereas[3H]FA or [14C]FA was used to estimate FA oxidation bymeasuring the generation of 3H2O and 14CO2. Although thismeasurement allows for the manipulation of substrate concen-tration, there are limitations. First, the substrates used inperfused hearts do not truly reflect what the heart receives invivo. For example, lipoproteins, lactate, and ketone bodies thatare important substrates for the heart in vivo (39, 98, 165) arerarely used to examine metabolism in perfused hearts. Inaddition, most metabolic data of diabetic hearts are obtained byusing normal concentrations of glucose and/or palmitate,which differ from the elevated concentrations seen in thediabetic conditions in vivo. Another drawback is that diabetichearts always have an elevated intracellular TG pool. Aug-mented intracellular TG turnover in diabetic hearts could diluteutilization of exogenous radiolabeled palmitate oxidation, re-sulting in an underestimation of FA oxidation (180). Finally,hormonal effects on energy metabolism are usually not con-sidered (20), and lack of hormones in the in vitro perfusate maycontribute to functional abnormalities that are not recapitulatedin intact models.

CONSEQUENCES OF ALTERED METABOLISM INDIABETIC HEARTS

Impaired Cardiac Function

Emerging evidence supports the concept that alterations inmetabolism contribute toward cardiac contractile dysfunction.In STZ-induced diabetes, a larger number of studies haveimplicated metabolic abnormalities (FA oxidation providesalmost 100% of ATP, with a dramatic decrease of glucoseutilization) in cardiac contractile dysfunction (219, 227, 234,267). In these animals, contractile failure begins as diastolicdysfunction, followed by severe systolic dysfunction (196,219). Normalizing energy metabolism in these hearts reversesthe impaired contractility (40, 176, 248). Animal models ofobesity and Type 2 diabetes also exhibit cardiac dysfunctionassociated with altered cardiac metabolism (5, 21, 218). Somestudies have observed both impaired diastolic and systolicfunction (3, 218), whereas other studies argue that no change insystolic function is present (6, 18, 164, 197). This discrepancycould be due to the severity of diabetes or the methods used toevaluate cardiac function. In diabetic patients, left ventricularhypertrophy and impaired isovolumic relaxation and ventricu-lar filling are the most common abnormalities diagnosed (196).

During diabetes, changes in cardiac metabolism occur earlyand precede the development of cardiomyopathy. For example,in STZ-induced diabetes, altered cardiac metabolism is ob-served as early as 4 days following diabetes induction (88),whereas evidence of cardiomyopathy is only apparent after4–6 wk. Similarly, in ob/ob and db/db mice, changes incardiac metabolism are evident much before confirmation ofcardiac dysfunction (5, 30). To validate the role of alteredmetabolism in provoking cardiac dysfunction, several studieshave treated 6- or 9-wk-old db/db mice with either PPAR-� or

PPAR-� agonists for 3–6 wk (3, 4, 36). Even though thesetreatments normalized cardiac metabolism, they failed to im-prove cardiac function in these mice, suggesting that metabo-lism may be unrelated to heart failure or that the treatment wasnot initiated in time. Interestingly, when ZDF rats were treatedwith a PPAR-� agonist at 6 wk of age [when insulin resistancecompared with 6-wk-old db/db mice is milder (37)], improve-ments of cardiac metabolism and heart function were observed(272). In a different study, changing cardiac metabolic profileby treating ZDF rats with PPAR-� agonist also improvedcontractile function (91). Additionally, overexpression ofGLUT4 in db/db mice not only normalized cardiac metabolismbut also improved heart function (21, 218). These studiessuggest that acute changes in metabolism likely induce earlyand reversible damage to cardiac tissue. Even though theseearly alterations are inadequate to produce cardiac functionalchanges, early interventions would be favored to provideprotection against development of cardiomyopathy in the laterstages of the disease.

The role of abnormal cardiac metabolism in cardiac dys-function is also supported by studies using transgenic mice.Overexpression of cardiac PPAR-� increased FA uptake andoxidation (79). The hearts from these transgenic mice exhibit ametabolic phenotype similar to diabetic hearts (79). Measure-ment of heart function revealed systolic dysfunction and ven-tricular hypertrophy in these hearts, indicating that, in theabsence of systemic metabolic disturbances, alteration of car-diac metabolism is sufficient to induce cardiac contractiledysfunction (79). This concept is further substantiated bytransgenic mice overexpressing cardiac ACS, FATP1, or LPL(47, 48, 260). These transgenic mice have shown increased FAuptake, utilization, or lipid accumulation, and these changescorrelated well with contractile dysfunction.

In contrast, several studies have reported that altered metab-olism has no impact on contractile function (224, 250). Itshould be noted that, in these studies, heart function wasevaluated using a Langendorff heart perfusion. Given thatdiabetic hearts exhibit reduced mitochondrial oxidative capac-ity and cardiac efficiency (28, 106), it is possible that withincreased workload and energy requirement, cardiac functionwould be compromised.

In addition to heart dysfunction per se, diabetic patients alsohave a greater incidence and severity of angina and acutemyocardial infarction (147). After a myocardial infarction,diabetic patients have almost twice the rate of mortality com-pared with nondiabetics (229). Alterations in myocardial en-ergy metabolism during diabetes are probably an importantcontributing factor in explaining this increased susceptibility toischemic damage. In obese or diabetic animals, increasedischemic damage is also observed (5, 60, 93, 147), and nor-malization of cardiac metabolism in hearts from these animalshas been shown to improve functional recovery followingischemia-reperfusion (147, 224, 270). Interestingly, a numberof studies have also reported contradictory results, with de-creased susceptibility to ischemia-reperfusion damage beingreported in STZ or ZDF hearts (145, 150, 236, 249). Althoughthe mechanisms for this observation are still unclear, decreasedglycolysis and glycolytic products, a lower Na�-Ca2� activity,and a decreased clearance of protons via the Na�/H� ex-changer have been proposed to explain this inconsistency (75).

Invited Review

H1496 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 10: Cardiac Metabolism Diabetic Cardiomyopathy

Although changes in metabolism have been implicated indiabetic cardiac dysfunction, the mechanisms responsible arestill unclear. Several factors have been proposed, and theseinclude changes in Ca2� homeostasis, decreased cardiac effi-ciency, lipotoxicity, and myocardial mitochondrial damage(Fig. 4).

Effects on Ca2� Homeostasis

Within the cell, enzymes that catalyze glycolysis are locatedclose to the sarcoplasmic reticulum and sarcolemma (71, 251),and ATP generated through glycolysis is preferentially used byion transporters [like Ca2�-ATPase (SERCA2a) and Na�-K�-ATPase] in these membrane fractions (71, 228, 252). Thusinhibition of cardiac glycolysis by high rates of FA oxidationduring obesity and diabetes may impair intracellular Ca2�

homeostasis, a defect that has been proposed to contributetoward the development of cardiomyopathy. Altered Ca2�

homeostasis can also result from glycolysis-independent mech-anisms. For example, decreased cardiac expression ofSERCA2a or Na�/Ca2� exchanger have been observed inType 1 (99, 126) and Type 2 diabetic animals (22). Althoughthe mechanisms for this reduction in gene expression areunclear, several studies have suggested that accumulation ofglucose metabolites due to dissociation of glucose influx andpyruvate oxidation plays a role (51). Conversely, decreasingglucose metabolites has been shown to prevent the decrease inSERCA2a following diabetes (207). In other studies, a de-crease in the activity of SERCA2a and Na�/Ca2� exchanger,without any change in expression or protein levels, have also

been documented (215, 271). Irrespective of the mechanism,suppression of SERCA2a and the Na�/Ca2� exchanger resultsin poor calcium handling associated with impaired heart func-tion (6, 22, 49). Interestingly, overexpression of SERCA2a inthe diabetic heart improved Ca2� handling (243) and cardiacfunction (239).

Decreased Cardiac Efficiency

When compared with glucose, oxidation of FA consumesmore oxygen (2.58 vs. 2.33 ATP/oxygen atom). Cardiac effi-ciency, the ratio of cardiac work to myocardial oxygen con-sumption, changes with the type of substrate. Thus, in perfusedhearts, provision of FA decreases cardiac efficiency comparedwith when glucose is the sole substrate (31, 162, 245). Addi-tionally, decreased cardiac efficiency is also observed in hu-man or experimental animals with obesity or diabetes (106,162, 191). This reduction in cardiac efficiency and increasedoxygen demand makes the heart especially vulnerable to dam-age following increased workload or ischemia. Interestingly,even though oxidation of FA requires 10% more oxygencompared with glucose, cardiac oxygen consumption is over30% higher in ob/ob or db/db mice compared with controlhearts, suggesting that other mechanisms also contribute tohigher oxygen consumption and lower cardiac efficiency (37).A recent study reported oxygen wasting for noncontractilepurposes in the diabetic heart (106). The mechanisms for thisoxygen wasting are unknown, and uncoupled proteins (UCPs)are suggested as a potential target. In hearts from STZ-induceddiabetic rats and ob/ob and db/db mice, augmented geneexpression or protein levels of UCP2 or UCP3 have beenreported (30, 102, 170, 171, 268). Other studies demonstrate noassociation between change in cardiac UCP protein levels andthe onset of cardiac inefficiency (28, 30). Hence, additionalstudies are required to identify the targets that cause oxygenwasting observed in hearts from obese and diabetic animals.

Lipotoxicity

A number of studies have suggested that excessive FAoverload induces lipotoxicity and contributes to the initiationand development of cardiomyopathy (194, 272). With the useof transgenic mice, studies have shown that elevation of FAuptake or utilization induces lipotoxicity in the absence of anysystemic metabolic disturbance. Cardiac-specific overexpres-sion of LPL or FATP1 significantly increased FA delivery withensuing lipid storage, lipotoxic cardiomyopathy, and contrac-tile dysfunction (47, 260). Moreover, elevating FA utilizationby cardiac-specific overexpression of PPAR-� or ACS alsocauses cardiomyopathy and cardiac dysfunction, similar to thatseen during diabetes (48, 79). Conversely, reducing FA supplyor utilization prevented the development of cardiomyopathy inobese or diabetic animals. In ZDF or transgenic mice withcardiac overexpression of LPL, a PPAR-� agonist decreasedplasma and cardiac intracellular lipids and ameliorated cardio-myopathy (244, 272). A recent study also demonstrated thatincreasing lipoprotein excretion by overexpressing humanapoB reduced cardiac lipids and improved cardiomyopathy(264). Taken together, these studies provide convincing evi-dence that augmented FA supply during obesity or diabetesimpairs cardiac lipid homeostasis and leads to lipotoxicitycardiomyopathy.

Fig. 4. Consequences of changes in cardiac metabolism during diabetes. Afterinsulin resistance or diabetes, augmented cardiac FA uptake promotes FAutilization and storage, which contributes toward a reduction in glycolysis andglucose oxidation. As ATP generated through glycolysis is preferentially usedby ion transporters (like Ca2�-ATPase), inhibition of cardiac glycolysis im-pairs intracellular Ca2� homeostasis. High rate of FA oxidation increasesreactive oxygen species (ROS) generation, along with augmented lipid storage,leading to lipotoxicity and mitochondrial dysfunction. High rate of FA oxida-tion also increases oxygen demand and reduces cardiac efficiency. All of thesechanges can eventually contribute to the development of diabetic cardiomy-opathy.

Invited Review

H1497ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 11: Cardiac Metabolism Diabetic Cardiomyopathy

Although limited evidence is available of the existence oflipotoxicity in human diabetic patients, some studies havedocumented similarities between rodent lipotoxicity and thehuman metabolic syndrome (240). In a subgroup of patientswith heart failure, Sharma et al. (220) have identified severemetabolic dysregulation characterized by intracellular TG ac-cumulation and alterations in gene expression, similar to thelipotoxic rat heart. Increased apoptosis was also detected inventricular biopsies from Type 2 diabetic patients (82). Addi-tionally, cardiac PCr-to-ATP ratio was decreased in Type 2diabetic patients and correlated well with plasma FA concen-trations (216).

The mechanisms that mediate cardiac lipotoxicity are stillnot completely understood. One potential target is over pro-duction of ROS (79). High rate of FA oxidation increasesmitochondrial action potential, leading to augmented ROSgeneration. Under normal physiological conditions, ROS isremoved by cellular antioxidants. In the event of excessivegeneration of ROS, as observed in STZ-induced diabetic rats,ZDF rats, and db/db mice (19, 32, 272), it causes cardiomyo-cyte cell damage and augmented apoptosis. Another potentialmechanism for lipotoxicity is accumulation of lipids, when FAuptake supercedes its oxidation. Regarding accumulation ofTG, the role of this neutral lipid in inducing contractile dys-function is still unknown, although a strong association be-tween TG storage and lipotoxicity has been established in bothanimal models and human studies (50, 220, 272). Interestingly,a recent study suggested that TG formation actually providesprotection against the deleterious effects of fatty acyl-CoA(144). Besides TG, excessive FA also leads to ceramide gen-eration, an intracellular messenger known to trigger apoptosis(272). Accumulation of ceramide has been found in ZDF rats(272) or in isolated cardiomyocytes incubated with high fat(67, 101). Ceramide upregulates inducible nitric oxide synthasethrough activation of NF-B, leading to increased generationof nitric oxide and peroxynitrite (241, 272). As a highlyreactive molecule, peroxynitrite causes opening of the mito-chondrial permeability transition pore and release of cyto-chrome c. Additionally, ceramide directly interacts with cyto-chrome c, leading to its release from the mitochondria (87). Asa consequence, caspase is activated, which initiates the apop-totic pathway in cells. Additionally, with the use of isolatedcardiomyocytes, high FA impairs cardiolipin and leads toceramide-independent cell apoptosis (184). Finally, accumula-tion of FA metabolites has also been associated with insulinresistance. FA derivatives, such as fatty acyl CoA, diacylglyc-erol, or ceramide may activate a serine kinase cascade, whichinvolves protein kinase C- and IB kinase-� (inhibitor ofNF-B kinase-�), leading to serine phosphorylation of IRS(44, 114, 128, 129, 269). Following this, tyrosine phosphory-lation of IRS and its ability to activate phosphatidylinositol3-kinase and protein kinase B are compromised (206). Futurestudies are required to substantiate the roles of these FAderivatives in the development of insulin resistance, especiallyin the heart.

Hyperglycemia

In addition to hyperlipidemia-induced lipotoxicity, hyper-glycemia also provokes glucotoxicity, which could contributeto cardiac tissue injury. Hyperglycemia promotes the produc-

tion of reactive oxygen and nitrogen species, leading to cyto-chrome c-mediated caspase 3 activation and myocardial apop-tosis (32). Hyperglycemia-induced oxidative stress also acti-vates poly(ADP-ribose) polymerase-1 (PARP) (64). Mildactivation of PARP regulates multiple cellular reactions suchas DNA repair, gene expression, and cell survival (246).However, overactivation of PARP could initiate a series ofcellular processes, leading to cellular damage. Through deple-tion of NAD�, an ATP-consuming process, PARP may causeenergy deficit and cell death (69). PARP, through inhibition ofglyceraldehyde phosphate dehydrogenase (GAPDH), divertsglucose from glycolytic pathways into alternative fates, includ-ing advanced glycation end product (AGE) formation, hex-osamine, polyol pathway flux, and protein kinase C (PKC)activation, which are believed to mediate hyperglycemia-in-duced cardiac tissue damage (64, 196). In this regard, increasedformation of AGE forms irreversible cross-links with manymacromolecules such as collagen, leading to tissue fibrosis,inactivation of SERCA2a, and ryanodine receptor calcium-release channel, impaired cardiac relaxation, contractility, andventricular stiffness (26, 27, 35, 54). Increased hexosamineinflux reduces SERCA2a protein levels, resulting in prolongedcalcium transients and delayed myocardial relaxation (51).Increased hexosamine influx also impairs insulin signal trans-duction and contributes to insulin resistance (163). Increasedpolyol flux is associated with a decrease in intracellular gluta-thione levels and an augmentation in cell apoptosis (83).Conversely, inhibition of polyol flux protects hearts fromischemic injury (125, 198). Finally, activation of PKC-�2 leadsto left ventricular hypertrophy, cardiac myocyte necrosis, mul-tifocal fibrosis, and decreased left ventricular performance,resulting in cardiomyopathy (247). Taken together, hypergly-cemia, through multiple pathways, causes cardiac cellular andfunctional changes, possibly contributing to the developmentof cardiomyopathy.

Mitochondrial Dysfunction

Mitochondria are the primary source of energy generationwithin cells. Acetyl-CoA generated from FA �-oxidation orglycolysis is used by the tricarboxylic acid cycle for productionof NADH and FADH2. These electron carriers transfer elec-trons to the mitochondria electron transport chain, where ATPis ultimately generated. Thus cardiac mitochondrial dysfunc-tion is expected to induce deleterious cellular effects, ulti-mately leading to heart disease. Indeed, humans with inheritedor acquired mitochondrial defects develop cardiomyopathy(112, 208). With the use of transgenic mice models, mitochon-drial dysfunction is also associated with heart disease. Forinstance, knock out of Ant1 (which controls exchange ofmitochondrial ATP with cytosolic ADP) or Tfam (a mitochon-drial transcription factor that regulates mitochondrial biosyn-thesis and gene expression) results in a cardiac energy deficitand cardiomyopathy (92, 142). Mice with cardiac-specificdeletion of mitochondrial genes involved in FA oxidation alsodisplay a cardiomyopathic phenotype (72, 135). Interestingly,the opposite is also true with overexpression of PPAR-�(which regulates the expression of mitochondrial enzymesinvolved in FA oxidation) (79) or PGC1 (which controlsmitochondrial biosynthesis) (139) resulting in mitochondrialdysfunction and cardiomyopathy. In Type 1 or Type 2 diabetic

Invited Review

H1498 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 12: Cardiac Metabolism Diabetic Cardiomyopathy

rats, impaired mitochondrial function has been demonstrated(222, 262), an observation similar to hearts from transgenicmice. Given the similarity between transgenic mice and modelsof diabetes, these studies strongly suggest that impairment ofmitochondrial function contributes to the development of car-diomyopathy.

The mechanisms by which mitochondrial dysfunction con-tributes toward cardiomyopathy are still unclear. One potentialtarget is ROS. Augmented ROS generation following high FAoxidation induces oxidative stress and cell damage. Inhibitionof ROS by overexpression of metallothionein, MnSOD, orcatalase has been shown to protect against mitochondrial dys-function and cardiomyopathy (221, 262, 263). Another target isceramide, a sphingolipid that has been shown to accumulatefollowing FA overload. Ceramide, through inhibition of mito-chondrial respiratory chain, or inducing cytochrome c releasefrom mitochondria and apoptosis, is suggested to provoke thedevelopment of cardiomyopathy (67, 95). Augmented expres-sion of PGC1 in hearts from obese or diabetic animals mayincrease mitochondrial proliferation, mitochondrial ultrastruc-tural abnormalities and dysfunction, and ultimately cardiomy-opathy (139). Finally, following end stage diabetes, dysfunc-tion of mitochondria causes energetic starvation, leading toheart failure (208).

Summary

In this review, we document that both insulin resistance andType 1 and Type 2 diabetes exhibit similar alterations incardiac metabolism through extrinsic and intrinsic mecha-nisms. The predominant change is a suppression of cardiacglucose utilization and a switch to excessive FA utilizationassociated with lipid storage. It should be noted that, in themajority of these studies, metabolic outcomes were obtainedusing ex vivo perfused working hearts. Additionally, only twosubstrates (glucose and fatty acid) were present in the perfusionbuffer, and lactate, lipoproteins, ketone bodies, and relevanthormones were excluded. Hence, cardiac metabolism mea-sured through this method may not reflect the in vivo situation.Despite these drawbacks, many studies have documented astrong correlation between altered cardiac metabolism andcardiac dysfunction observed during insulin resistance or dia-betes. This correlation was further substantiated using trans-genic mice with lipid oversupply in the absence of systemicinterference. Regarding heart function, both diastolic and sys-tolic dysfunction are observed in Type 1 diabetic animals. Thisis likely a consequence of prolonged hypoinsulinemia andhyperglycemia. With Type 2 diabetic models, there are con-flicting reports, with some studies documenting both impairedsystolic and diastolic function, whereas other studies argueagainst a change in systolic function. It is possible with Type2 diabetic models, this discrepancy could be due to the severityor duration of hyperinsulinemia, and eventual hyperglycemia,or the techniques used to measure cardiac function. Finally,during diabetes, changes in cardiac metabolism occur early andprecede the development of cardiomyopathy. Even thoughaltered metabolism is inadequate to produce cardiac functionalchanges at this early time, it is likely that early metabolicdamage is occurring at the cellular or subcellular levels. Over-time, these cumulative defects could be contributing to diabeticcardiomyopathy. Identification of this early damage could

facilitate proper interventions and provide protection againstdevelopment of diabetic cardiomyopathy in the later stages ofthe disease.

GRANTS

This work was supported by operating grants from Heart and StrokeFoundation of BC and Yukon and Canadian Diabetes Association.

REFERENCES

1. Aalto-Setala K, Fisher EA, Chen X, Chajek-Shaul T, Hayek T,Zechner R, Walsh A, Ramakrishnan R, Ginsberg HN, and BreslowJL. Mechanism of hypertriglyceridemia in human apolipoprotein (apo)C.III transgenic mice Diminished very low density lipoprotein fractionalcatabolic rate associated with increased apo CIII and reduced apo E onthe particles. J Clin Invest 90: 1889–1900, 1992.

2. Aalto-Setala K, Weinstock PH, Bisgaier CL, Wu L, Smith JD, andBreslow JL. Further characterization of the metabolic properties oftriglyceride-rich lipoproteins from human and mouse apoC-I.I.I. trans-genic mice. J Lipid Res 37: 1802–1811, 1996.

3. Aasum E, Belke DD, Severson DL, Riemersma RA, Cooper M,Andreassen M, and Larsen TS. Cardiac function and metabolism inType 2 diabetic mice after treatment with B.M 170744, a novel PPAR-alpha activator. Am J Physiol Heart Circ Physiol 283: H949–H957,2002.

4. Aasum E, Cooper M, Severson DL, and Larsen TS. Effect of BM 170744, a PPARalpha ligand, on the metabolism of perfused hearts fromcontrol and diabetic mice. Can J Physiol Pharmacol 83: 183–190, 2005.

5. Aasum E, Hafstad AD, Severson DL, and Larsen TS. Age-dependentchanges in metabolism, contractile function, and ischemic sensitivity inhearts from db/db mice. Diabetes 52: 434–441, 2003.

6. Abe T, Ohga Y, Tabayashi N, Kobayashi S, Sakata S, Misawa H,Tsuji T, Kohzuki H, Suga H, Taniguchi S, and Takaki M. Leftventricular diastolic dysfunction in type 2 diabetes mellitus model rats.Am J Physiol Heart Circ Physiol 282: H138–H148, 2002.

7. Abumrad NA, Park JH, and Park CR. Permeation of long-chain fattyacid into adipocytes. Kinetics, specificity, and evidence for involvementof a membrane protein. J Biol Chem 259: 8945–8953, 1984.

8. An D, Pulinilkunnil T, Qi D, Ghosh S, Abrahani A, and Rodrigues B.The metabolic “switch” AMPK regulates cardiac heparin-releasablelipoprotein lipase. Am J Physiol Endocrinol Metab 288: E246–E253,2005.

9. Anguera I, Magrina J, Setoain FJ, Esmatges E, Pare C, Vidal J,Azqueta M, Garcia A, Grau JM, Vidal-Sicart S, and Betriu A.Anatomopathological bases of latent ventricular dysfunction in insulin-dependent diabetics. Rev Esp Cardiol 51: 43–50, 1998.

10. Armoni M, Harel C, Bar-Yoseph F, Milo S, and Karnieli E. Free fattyacids repress the GLUT4 gene expression in cardiac muscle via novelresponse elements. J Biol Chem 280: 34786–34795, 2005.

11. Atkinson LL, Fischer MA, and Lopaschuk GD. Leptin activatescardiac fatty acid oxidation independent of changes in the AMP-activatedprotein kinase-acetyl-CoA carboxylase-malonyl-CoA axis. J Biol Chem277: 29424–29430, 2002.

12. Augustus A, Yagyu H, Haemmerle G, Bensadoun A, Vikrama-dithyan RK, Park SY, Kim JK, Zechner R, and Goldberg IJ.Cardiac-specific knock-out of lipoprotein lipase alters plasma lipoproteintriglyceride metabolism and cardiac gene expression. J Biol Chem 279:25050–25057, 2004.

13. Augustus AS, Buchanan J, Park TS, Hirata K, Noh HL, Sun J,Homma S, D’Armiento J, Abel ED, and Goldberg IJ. Loss oflipoprotein lipase-derived fatty acids leads to increased cardiac glucosemetabolism and heart dysfunction. J Biol Chem 281: 8716–8723, 2006.

14. Augustus AS, Kako Y, Yagyu H, and Goldberg IJ. Routes of FAdelivery to cardiac muscle: modulation of lipoprotein lipolysis altersuptake of TG-derived FA. Am J Physiol Endocrinol Metab 284: E331–E339, 2003.

15. Avogaro A, Nosadini R, Doria A, Fioretto P, Velussi M, Vigorito C,Sacca L, Toffolo G, Cobelli C, Trevisan R, E. Duner, R. Razzolini, F.Rengo, and G. Crepaldi. Myocardial metabolism in insulin-deficientdiabetic humans without coronary artery disease. Am J Physiol Endocri-nol Metab 258: E606–E618, 1990.

16. Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR,Koder A, and Evans RM. PPARgamma is required for placental,cardiac, and adipose tissue development. Mol Cell 4: 585–595, 1999.

Invited Review

H1499ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 13: Cardiac Metabolism Diabetic Cardiomyopathy

17. Barger PM and Kelly DP. PPARsignaling in the control of cardiacenergy metabolism. Trends Cardiovasc Med 10: 238–245, 2000.

18. Barouch LA, Berkowitz DE, Harrison RW, O’Donnell CP, and HareJM. Disruption of leptin signaling contributes to cardiac hypertrophyindependently of body weight in mice. Circulation 108: 754–759, 2003.

19. Barouch LA, Gao D, Chen L, Miller KL, Xu W, Phan AC, KittlesonMM, Minhas KM, Berkowitz DE, Wei C, and Hare JM. Cardiacmyocyte apoptosis is associated with increased DNA damage and de-creased survival in murine models of obesity. Circ Res 98: 119–124,2006.

20. Barr RL and Lopaschuk GD. Methodology for measuring in vitro/exvivo cardiac energy metabolism. J Pharmacol Toxicol Methods 43:141–152, 2000.

21. Belke DD, Larsen TS, Gibbs EM, and Severson DL. Altered metab-olism causes cardiac dysfunction in perfused hearts from diabetic (db/db)mice. Am J Physiol Endocrinol Metab 279: E1104–E1113, 2000.

22. Belke DD, Swanson EA, and Dillmann WH. Decreased sarcoplasmicreticulum activity and contractility in diabetic db/db mouse heart. Dia-betes 53: 3201–3208, 2004.

23. Bell DS. Diabetic cardiomyopathy. A unique entity or a complication ofcoronary artery disease? Diabetes Care 18: 708–714, 1995.

24. Bertoni AG, Tsai A, Kasper EK, and Brancati FL. Diabetes andidiopathic cardiomyopathy: a nationwide case-control study. DiabetesCare 26: 2791–2795, 2003.

25. Bertrand L, Alessi DR, Deprez J, Deak M, Viaene E, Rider MH, andHue L. Heart 6-phosphofructo-2-kinase activation by insulin results fromSer-466 and Ser-483 phosphorylation and requires 3-phosphoinositide-dependent kinase-1, but not protein kinase B. J Biol Chem 274: 30927–30933, 1999.

26. Bidasee KR, Nallani K, Yu Y, Cocklin RR, Zhang Y, Wang M,Dincer UD, and Besch HR Jr. Chronic diabetes increases advancedglycation end products on cardiac ryanodine receptors/calcium-releasechannels. Diabetes 52: 1825–1836, 2003.

27. Bidasee KR, Zhang Y, Shao CH, Wang M, Patel KP, Dincer UD, andBesch HR Jr. Diabetes increases formation of advanced glycation endproducts on Sarco(endo)plasmic reticulum Ca2�-ATPase. Diabetes 53:463–473, 2004.

28. Boudina S, Sena S, O’Neill BT, Tathireddy P, Young ME, and AbelED. Reduced mitochondrial oxidative capacity and increased mitochon-drial uncoupling impair myocardial energetics in obesity. Circulation112: 2686–2695, 2005.

29. Bowker-Kinley MM, Davis WI, Wu P, Harris RA, and Popov KM.Evidence for existence of tissue-specific regulation of the mammalianpyruvate dehydrogenase complex. Biochem J 329: 191–196, 1998.

30. Buchanan J, Mazumder PK, Hu P, Chakrabarti G, Roberts MW,Yun UJ, Cooksey RC, Litwin SE, and Abel ED. Reduced cardiacefficiency and altered substrate metabolism precedes the onset of hyper-glycemia and contractile dysfunction in two mouse models of insulinresistance and obesity. Endocrinology 146: 5341–5349, 2005.

31. Burkhoff D, Weiss RG, Schulman SP, Kalil-Filho R, Wannenburg T,and Gerstenblith G. Influence of metabolic substrate on rat heartfunction and metabolism at different coronary flows. Am J Physiol HeartCirc Physiol 261: H741–H750, 1991.

32. Cai L, Li W, Wang G, Guo L, Jiang Y, and Kang YJ. Hyperglycemia-induced apoptosis in mouse myocardium: mitochondrial cytochromeC-mediated caspase-3 activation pathway. Diabetes 51: 1938–1948,2002.

33. Camps L, Reina M, Llobera M, Vilaro S, and Olivecrona T. Lipopro-tein lipase: cellular origin and functional distribution. Am J Physiol CellPhysiol 258: C673–C681, 1990.

34. Camps M, Castello A, Munoz P, Monfar M, Testar X, Palacin M,and Zorzano A. Effect of diabetes and fasting on GLUT-4 (muscle/fat)glucose-transporter expression in insulin-sensitive tissues Heterogeneousresponse in heart, red and white muscle. Biochem J 282: 765–772, 1992.

35. Candido R, Forbes JM, Thomas MC, Thallas V, Dean RG, BurnsWC, Tikellis C, Ritchie RH, Twigg SM, Cooper ME, and BurrellLMA. breaker of advanced glycation end products attenuates diabetes-induced myocardial structural changes. Circ Res 92: 785–792, 2003.

36. Carley AN, Semeniuk LM, Shimoni Y, Aasum E, Larsen TS, BergerJP, and Severson DL. Treatment of type 2 diabetic db/db mice with anovel PPARgamma agonist improves cardiac metabolism but not con-tractile function. Am J Physiol Endocrinol Metab 286: E449–E455,2004.

37. Carley AN and Severson DL. Fatty acid metabolism is enhanced in type2 diabetic hearts. Biochim Biophys Acta 1734: 112–126, 2005.

38. Carroll R, Carley AN, Dyck JR, and Severson DL. Metabolic effectsof insulin on cardiomyocytes from control and diabetic db/db mousehearts. Am J Physiol Endocrinol Metab 288: E900–E906, 2005.

39. Chatham JC. Lactate–the forgotten fuel! J Physiol 542: 333, 2002.40. Chatham JC and Forder JR. Relationship between cardiac function

and substrate oxidation in hearts of diabetic rats. Am J Physiol Heart CircPhysiol 273: H52–H58, 1997.

41. Chatham JC, Gao ZP, Bonen A, and Forder JR. Preferential inhibi-tion of lactate oxidation relative to glucose oxidation in the rat heartfollowing diabetes. Cardiovasc Res 43: 96–106, 1999.

42. Chatham JC, Gao ZP, and Forder JR. Impact of 1 wk of diabetes onthe regulation of myocardial carbohydrate and fatty acid oxidation. Am JPhysiol Endocrinol Metab 277: E342–E351, 1999.

43. Chatham JC and Seymour AM. Cardiac carbohydrate metabolism inZucker diabetic fatty rats. Cardiovasc Res 55: 104–112, 2002.

44. Chavez JA, Knotts TA, Wang LP, Li G, Dobrowsky RT, Florant GL,and Summers SAA. Role for ceramide, but not diacylglycerol, in theantagonism of insulin signal transduction by saturated fatty acids. J BiolChem 278: 10297–10303, 2003.

45. Chawla A, Lee CH, Barak Y, He W, Rosenfeld J, Liao D, Han J,Kang H, and Evans RM. PPARdelta is a very low-density lipoproteinsensor in macrophages. Proc Natl Acad Sci USA 100: 1268–1273, 2003.

46. Cheng L, Ding G, Qin Q, Huang Y, Lewis W, He N, Evans RM,Schneider MD, Brako FA, Xiao Y, Chen YE, and Yang Q. Cardio-myocyte-restricted peroxisome proliferator-activated receptor-delta dele-tion perturbs myocardial fatty acid oxidation and leads to cardiomyopa-thy. Nat Med 10: 1245–1250, 2004.

47. Chiu HC, Kovacs A, Blanton RM, Han X, Courtois M, WeinheimerCJ, Yamada KA, Brunet S, Xu H, Nerbonne JM, Welch MJ, FettigNM, Sharp TL, Sambandam N, Olson KM, Ory DS, and Schaffer JE.Transgenic expression of fatty acid transport protein 1 in the heart causeslipotoxic cardiomyopathy. Circ Res 96: 225–233, 2005.

48. Chiu HC, Kovacs A, Ford DA, Hsu FF, Garcia R, Herrero P, SaffitzJE, and Schaffer JEA. novel mouse model of lipotoxic cardiomyopathy.J Clin Invest 107: 813–822, 2001.

49. Choi KM, Zhong Y, Hoit BD, Grupp IL, Hahn H, Dilly KW,Guatimosim S, Lederer WJ, and Matlib MA. Defective intracellularCa2� signaling contributes to cardiomyopathy in Type 1 diabetic rats.Am J Physiol Heart Circ Physiol 283: H1398–H1408, 2002.

50. Christoffersen C, Bollano E, Lindegaard ML, Bartels ED, Goetze JP,Andersen CB, and Nielsen LB. Cardiac lipid accumulation associatedwith diastolic dysfunction in obese mice. Endocrinology 144: 3483–3490, 2003.

51. Clark RJ, McDonough PM, Swanson E, Trost SU, Suzuki M,Fukuda M, and Dillmann WH. Diabetes and the accompanying hyper-glycemia impairs cardiomyocyte calcium cycling through increased nu-clear O-GlcNAcylation. J Biol Chem 278: 44230–44237, 2003.

52. Clerk LH, Rattigan S, and Clark MG. Lipid infusion impairs physi-ologic insulin-mediated capillary recruitment and muscle glucose uptakein vivo. Diabetes 51: 1138–1145, 2002.

53. Coe NR, Smith AJ, Frohnert BI, Watkins PA, and Bernlohr DA. Thefatty acid transport protein (FATP1) is a very long chain acyl-CoAsynthetase. J Biol Chem 274: 36300–36304, 1999.

54. Cooper ME. Importance of advanced glycation end products in diabetes-associated cardiovascular, and renal disease. Am J Hypertens 17: 31S–38S, 2004.

55. Coort SL, Hasselbaink DM, Koonen DP, Willems J, Coumans WA,Chabowski A, van der Vusse GJ, Bonen A, Glatz JF, and Luiken JJ.Enhanced sarcolemmal FAT/CD36 content and triacylglycerol storage incardiac myocytes from obese zucker rats. Diabetes 53: 1655–1663, 2004.

56. Coort SL, Luiken JJ, van der Vusse GJ, Bonen A, and Glatz JF.Increased FAT (fatty acid translocase)/CD36-mediated long-chain fattyacid uptake in cardiac myocytes from obese Zucker rats. Biochem SocTrans 32: 83–85, 2004.

57. Depre C, Veitch K, and Hue L. Role of fructose 2,6-bisphosphate in thecontrol of glycolysis. Stimulation of glycogen synthesis by lactate in theisolated working rat heart. Acta Cardiol 48: 147–164, 1993.

58. Deprez J, Vertommen D, Alessi DR, Hue L, and Rider MH. Phos-phorylation and activation of heart 6-phosphofructo-2-kinase by proteinkinase B. and other protein kinases of the insulin signaling cascades.J Biol Chem 272: 17269–17275, 1997.

Invited Review

H1500 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 14: Cardiac Metabolism Diabetic Cardiomyopathy

59. Derave W, Hansen BF, Lund S, Kristiansen S, and Richter EA.Muscle glycogen content affects insulin-stimulated glucose transport andprotein kinase B activity. Am J Physiol Endocrinol Metab 279: E947–E955, 2000.

60. Di Filippo C, Marfella R, Cuzzocrea S, Piegari E, Petronella P,Giugliano D, Rossi F, and D’Amico M. Hyperglycemia in streptozo-tocin-induced diabetic rat increases infarct size associated with low levelsof myocardial HO-1 during ischemia/reperfusion. Diabetes 54: 803–810,2005.

61. Diamant M, Lamb HJ, Groeneveld Y, Endert EL, Smit JW, Bax JJ,Romijn JA, de Roos A, and Radder JK. Diastolic dysfunction isassociated with altered myocardial metabolism in asymptomatic normo-tensive patients with well-controlled type 2 diabetes mellitus. J Am CollCardiol 42: 328–335, 2003.

62. Doria A, Nosadini R, Avogaro A, Fioretto P, and Crepaldi G.Myocardial metabolism in type 1 diabetic patients without coronaryartery disease. Diabet Med 8: S104–S107, 1991.

63. Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, and Muscat GE.The peroxisome proliferator-activated receptor beta/delta agonist,GW501516, regulates the expression of genes involved in lipid catabo-lism and energy uncoupling in skeletal muscle cells. Mol Endocrinol 17:2477–2493, 2003.

64. Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, Szabo C,and Brownlee M. Inhibition of GAPDH activity by poly(ADP-ribose)polymerase activates three major pathways of hyperglycemic damage inendothelial cells. J Clin Invest 112: 1049–1057, 2003.

65. Dyck JR, Barr AJ, Barr RL, Kolattukudy PE, and Lopaschuk GD.Characterization of cardiac malonyl-CoA decarboxylase and its putativerole in regulating fatty acid oxidation. Am J Physiol Heart Circ Physiol275: H2122–H2129, 1998.

66. Dyck JR, Cheng JF, Stanley WC, Barr R, Chandler MP, Brown S,Wallace D, Arrhenius T, Harmon C, Yang G, Nadzan AM, andLopaschuk GD. Malonyl coenzyme a decarboxylase inhibition protectsthe ischemic heart by inhibiting fatty acid oxidation and stimulatingglucose oxidation. Circ Res 94: e78–e84, 2004.

67. Dyntar D, Eppenberger-Eberhardt M, Maedler K, Pruschy M, Ep-penberger HM, Spinas GA, and Donath MY. Glucose and palmiticacid induce degeneration of myofibrils and modulate apoptosis in ratadult cardiomyocytes. Diabetes 50: 2105–2113, 2001.

68. Eckel RH. Lipoprotein lipase. A multifunctional enzyme relevant tocommon metabolic diseases. N Engl J Med 320: 1060–1068, 1989.

69. Eliasson MJ, Sampei K, Mandir AS, Hurn PD, Traystman RJ, BaoJ, Pieper A, Wang ZQ, Dawson TM, Snyder SH, and Dawson VL.Poly(ADP-ribose) polymerase gene disruption renders mice resistant tocerebral ischemia. Nat Med 3: 1089–1095, 1997.

70. Enerback S and Gimble JM. Lipoprotein lipase gene expression:physiological regulators at the transcriptional and post-transcriptionallevel. Biochim Biophys Acta 1169: 107–125, 1993.

71. Entman ML, Bornet EP, Van Winkle WB, Goldstein MA, andSchwartz A. Association of glycogenolysis with cardiac sarcoplasmicreticulum: I. Effect of glycogen depletion, deoxycholate solubilizationand cardiac ischemia: evidence for a phorphorylase kinase membranecomplex. J Mol Cell Cardiol 9: 515–528, 1977.

72. Exil VJ, Roberts RL, Sims H, McLaughlin JE, Malkin RA, GardnerCD, Ni G, Rottman JN, and Strauss AW. Very-long-chain acyl-coenzyme a dehydrogenase deficiency in mice. Circ Res 93: 448–455,2003.

73. Fang ZY, Prins JB, and Marwick TH. Diabetic cardiomyopathy:evidence, mechanisms, and therapeutic implications. Endocr Rev 25:543–567, 2004.

74. Fein FS. Diabetic cardiomyopathy. Diabetes Care 13: 1169–1179, 1990.75. Feuvray D and Lopaschuk GD. Controversies on the sensitivity of the

diabetic heart to ischemic injury: the sensitivity of the diabetic heart toischemic injury is decreased. Cardiovasc Res 34: 113–120, 1997.

76. Finck BN. The role of the peroxisome proliferator-activated receptoralpha pathway in pathological remodeling of the diabetic heart. CurrOpin Clin Nutr Metab Care 7: 391–396, 2004.

77. Finck BN, Han X, Courtois M, Aimond F, Nerbonne JM, Kovacs A,Gross RW, and Kelly DPA. Critical role for PPARalpha-mediatedlipotoxicity in the pathogenesis of diabetic cardiomyopathy: modulationby dietary fat content. Proc Natl Acad Sci USA 100: 1226–1231, 2003.

78. Finck BN and Kelly DP. Peroxisome proliferator-activated receptoralpha (PPARalpha) signaling in the gene regulatory control of energy

metabolism in the normal and diseased heart. J Mol Cell Cardiol 34:1249–1257, 2002.

79. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A,Han X, Gross RW, Kozak R, Lopaschuk GD, and Kelly DP. Thecardiac phenotype induced by PPARalpha overexpression mimics thatcaused by diabetes mellitus. J Clin Invest 109: 121–130, 2002.

80. Forman BM, Chen J, and Evans RM. Hypolipidemic drugs, polyun-saturated fatty acids, and eicosanoids are ligands for peroxisome prolif-erator-activated receptors alpha and delta. Proc Natl Acad Sci USA 94:4312–4317, 1997.

81. Francis GS. Diabetic cardiomyopathy: fact or fiction? Heart 85: 247–248, 2001.

82. Frustaci A, Kajstura J, Chimenti C, Jakoniuk I, Leri A, Maseri A,Nadal-Ginard B, and Anversa P. Myocardial cell death in humandiabetes. Circ Res 87: 1123–1132, 2000.

83. Galvez AS, Ulloa JA, Chiong M, Criollo A, Eisner V, Barros LF, andLavandero S. Aldose reductase induced by hyperosmotic stress mediatescardiomyocyte apoptosis: differential effects of sorbitol and mannitol.J Biol Chem 278: 38484–38494, 2003.

84. Gargiulo CE, Stuhlsatz-Krouper SM, and Schaffer JE. Localizationof adipocyte long-chain fatty acyl-CoA synthetase at the plasma mem-brane. J Lipid Res 40: 881–892, 1999.

85. Garland PB, Randle PJ, and Newsholme EA. Citrate as an interme-diary in the inhibition of phosphofructokinase in rat heart muscle by fattyacids, ketone bodies, pyruvate, diabetes, and starvation. Nature 200:169–170, 1963.

86. Gertz EW, Wisneski JA, Stanley WC, and Neese RA. Myocardialsubstrate utilization during exercise in humans. Dual carbon-labeledcarbohydrate isotope experiments. J Clin Invest 82: 2017–2025, 1988.

87. Ghafourifar P, Klein SD, Schucht O, Schenk U, Pruschy M, Rocha S,and Richter C. Ceramide induces cytochrome c release from isolatedmitochondria. Importance of mitochondrial redox state. J Biol Chem 274:6080–6084, 1999.

88. Ghosh S, An D, Pulinilkunnil T, Qi D, Lau HC, Abrahani A, InnisSM, and Rodrigues B. Role of dietary fatty acids and acute hypergly-cemia in modulating cardiac cell death. Nutrition 20: 916–923, 2004.

89. Gilde AJ, van der Lee KA, Willemsen PH, Chinetti G, van der LeijFR, van der Vusse GJ, Staels B, and van Bilsen M. Peroxisomeproliferator-activated receptor (PPAR) alpha and PPARbeta/delta, butnot PPARgamma, modulate the expression of genes involved in cardiaclipid metabolism. Circ Res 92: 518–524, 2003.

90. Golfman LS, Takeda N, and Dhalla NS. Cardiac membrane Ca(2�)-transport in alloxan-induced diabetes in rats. Diabetes Res Clin Pract 31,Suppl: S73–S77, 1996.

91. Golfman LS, Wilson CR, Sharma S, Burgmaier M, Young ME,Guthrie PH, Van Arsdall M, Adrogue JV, Brown KK, and Taegt-meyer H. Activation of PPARgamma enhances myocardial glucoseoxidation and improves contractile function in isolated working hearts ofZDF rats. Am J Physiol Endocrinol Metab 289: E328–E336, 2005.

92. Graham BH, Waymire KG, Cottrell B, Trounce IA, MacGregor GR,and Wallace DCA. Mouse model for mitochondrial myopathy andcardiomyopathy resulting from a deficiency in the heart/muscle isoformof the adenine nucleotide translocator. Nat Genet 16: 226–234, 1997.

93. Greer JJ, Ware DP, and Lefer DJ. Myocardial infarction and heartfailure in the db/db diabetic mouse. Am J Physiol Heart Circ Physiol 290:H146–H153, 2006.

94. Grundleger ML and Thenen SW. Decreased insulin binding, glucosetransport, and glucose metabolism in soleus muscle of rats fed a high fatdiet. Diabetes 31: 232–237, 1982.

95. Gudz TI, Tserng KY, and Hoppel CL. Direct inhibition of mitochon-drial respiratory chain complex III by cell-permeable ceramide. J BiolChem 272: 24154–24158, 1997.

96. Hardie DG. Minireview: the AMPactivated protein kinase cascade: thekey sensor of cellular energy status. Endocrinology 144: 5179–5183,2003.

97. Hardie DG and Carling D. The AMPactivated protein kinase–fuelgauge of the mammalian cell? Eur J Biochem 246: 259–273, 1997.

98. Hasselbaink DM, Glatz JF, Luiken JJ, Roemen TH, and Van derVusse GJ. Ketone bodies disturb fatty acid handling in isolated cardio-myocytes derived from control and diabetic rats. Biochem J 371: 753–760, 2003.

99. Hattori Y, Matsuda N, Kimura J, Ishitani T, Tamada A, Gando S,Kemmotsu O, and Kanno M. Diminished function and expression of

Invited Review

H1501ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 15: Cardiac Metabolism Diabetic Cardiomyopathy

the cardiac Na�-Ca2� exchanger in diabetic rats: implication in Ca2�

overload. J Physiol 527: 85–94, 2000.100. Hauton D, Bennett MJ, and Evans RD. Utilisation of triacylglycerol

and non-esterified fatty acid by the working rat heart: myocardial lipidsubstrate preference. Biochim Biophys Acta 1533: 99–109, 2001.

101. Hickson-Bick DL, Buja ML, and McMillin JB. Palmitate-mediatedalterations in the fatty acid metabolism of rat neonatal cardiac myocytes.J Mol Cell Cardiol 32: 511–519, 2000.

102. Hidaka S, Kakuma T, Yoshimatsu H, Sakino H, Fukuchi S, andSakata T. Streptozotocin treatment upregulates uncoupling protein 3expression in the rat heart. Diabetes 48: 430–435, 1999.

103. Higuchi M, Miyagi K, Nakasone J, and Sakanashi M. Role of highglycogen in underperfused diabetic rat hearts with added norepinephrine.J Cardiovasc Pharmacol 26: 899–907, 1995.

104. Holness MJ and Sugden MC. Regulation of pyruvate dehydrogenasecomplex activity by reversible phosphorylation. Biochem Soc Trans 31:1143–1151, 2003.

105. Horman S, Browne G, Krause U, Patel J, Vertommen D, Bertrand L,Lavoinne A, Hue L, Proud C, and Rider M. Activation of AMPacti-vated protein kinase leads to the phosphorylation of elongation factor 2and an inhibition of protein synthesis. Curr Biol 12: 1419–1423, 2002.

106. How OJ, Aasum E, Severson DL, Chan WY, Essop MF, and LarsenTS. Increased myocardial oxygen consumption reduces cardiac effi-ciency in diabetic mice. Diabetes 55: 466–473, 2006.

107. Huang B, Wu P, Popov KM, and Harris RA. Starvation and diabetesreduce the amount of pyruvate dehydrogenase phosphatase in rat heartand kidney. Diabetes 52: 1371–1376, 2003.

108. Hudson ER, Pan DA, James J, Lucocq JM, Hawley SA, Green KA,Baba O, Terashima T, and Hardie DG. A novel domain in AMP-activated protein kinase causes glycogen storage bodies similar to thoseseen in hereditary cardiac arrhythmias. Curr Biol 13: 861–866, 2003.

109. Hue L, Beauloye C, Marsin AS, Bertrand L, Horman S, and RiderMH. Insulin and ischemia stimulate glycolysis by acting on the sametargets through different and opposing signaling pathways. J Mol CellCardiol 34: 1091–1097, 2002.

110. Hue L and Rider MH. Role of fructose 2,6-bisphosphate in the controlof glycolysis in mammalian tissues. Biochem J 245: 313–324, 1987.

111. Huss JM and Kelly DP. Nuclear receptor signaling and cardiac ener-getics. Circ Res 95: 568–578, 2004.

112. Huss JM and Kelly DP. Mitochondrial energy metabolism in heartfailure: a question of balance. J Clin Invest 115: 547–555, 2005.

113. Ibrahimi A, Bonen A, Blinn WD, Hajri T, Li X, Zhong K, CameronR, and Abumrad NA. Muscle-specific overexpression of FAT/CD36enhances fatty acid oxidation by contracting muscle, reduces plasmatriglycerides and fatty acids, and increases plasma glucose and insulin.J Biol Chem 274: 26761–26766, 1999.

114. Itani SI, Ruderman NB, Schmieder F, and Boden G. Lipid-inducedinsulin resistance in human muscle is associated with changes in diac-ylglycerol, protein kinase C., and IkappaB-alpha. Diabetes 51: 2005–2011, 2002.

115. Itani SI, Zhou Q, Pories WJ, MacDonald KG, and Dohm GL.Involvement of protein kinase C in human skeletal muscle insulinresistance and obesity. Diabetes 49: 1353–1358, 2000.

116. Iwasaki T, Takahashi S, Takahashi M, Zenimaru Y, Kujiraoka T,Ishihara M, Nagano M, Suzuki J, Miyamori I, Naiki H, Sakai J,Fujino T, Miller NE, Yamamoto TT, and Hattori H. Deficiency of thevery low-density lipoprotein (VLDL) receptors in streptozotocin-induceddiabetic rats: insulin dependency of the VLDL receptor. Endocrinology146: 3286–3294, 2005.

117. Jagasia D and McNulty PH. Diabetes mellitus and heart failure.Congest Heart Fail 9: 133–139; quiz 140–131, 2003.

118. Jagasia D, Whiting JM, Concato J, Pfau S, and McNulty PH. Effectof non-insulin-dependent diabetes mellitus on myocardial insulin respon-siveness in patients with ischemic heart disease. Circulation 103: 1734–1739, 2001.

119. Jeffrey FM, Diczku V, Sherry AD, and Malloy CR. Substrate selectionin the isolated working rat heart: effects of reperfusion, afterload, andconcentration. Basic Res Cardiol 90: 388–396, 1995.

120. Jensen J, Aslesen R, Ivy JL, and Brors O. Role of glycogen concen-tration and epinephrine on glucose uptake in rat epitrochlearis muscle.Am J Physiol Endocrinol Metab 272: E649–E655, 1997.

121. Jensen J, Jebens E, Brennesvik EO, Ruzzin J, Soos MA, EngebretsenEM, O’Rahilly S, and Whitehead JP. Muscle glycogen inharmoniously

regulates glycogen synthase activity, glucose uptake, and proximal insu-lin signaling. Am J Physiol Endocrinol Metab 290: E154–E162, 2006.

122. Jensen J, Ruzzin J, Jebens E, Brennesvik EO, and Knardahl S.Improved insulin-stimulated glucose uptake and glycogen synthase acti-vation in rat skeletal muscles after adrenaline infusion: role of glycogencontent and PKB phosphorylation. Acta Physiol Scand 184: 121–130,2005.

123. Joffe II, Travers KE, Perreault-Micale CL, Hampton T, Katz SE,Morgan JP, and Douglas PS. Abnormal cardiac function in the strep-tozotocin-induced non-insulin-dependent diabetic rat: noninvasive as-sessment with doppler echocardiography and contribution of the nitricoxide pathway. J Am Coll Cardiol 34: 2111–2119, 1999.

124. Kajstura J, Fiordaliso F, Andreoli AM, Li B, Chimenti S, MedowMS, Limana F, Nadal-Ginard B, Leri A, and Anversa P. IGF-1overexpression inhibits the development of diabetic cardiomyopathy andangiotensin II-mediated oxidative stress. Diabetes 50: 1414–1424, 2001.

125. Kaneko M, Bucciarelli L, Hwang YC, Lee L, Yan SF, Schmidt AM,and Ramasamy R. Aldose reductase and AGE-RAGE pathways: keyplayers in myocardial ischemic injury. Ann NY Acad Sci 1043: 702–709,2005.

126. Kashihara H, Shi ZQ, Yu JZ, McNeill JH, and Tibbits GF. Effects ofdiabetes and hypertension on myocardial Na�-Ca2� exchange. CanJ Physiol Pharmacol 78: 12–19, 2000.

127. Khairallah M, Labarthe F, Bouchard B, Danialou G, Petrof BJ, andDes Rosiers C. Profiling substrate fluxes in the isolated working mouseheart using 13C-labeled substrates: focusing on the origin and fate ofpyruvate and citrate carbons. Am J Physiol Heart Circ Physiol 286:H1461–H1470, 2004.

128. Kim JK, Fillmore JJ, Chen Y, Yu C, Moore IK, Pypaert M, Lutz EP,Kako Y, Velez-Carrasco W, Goldberg IJ, Breslow JL, and ShulmanGI. Tissue-specific overexpression of lipoprotein lipase causes tissue-specific insulin resistance. Proc Natl Acad Sci USA 98: 7522–7527, 2001.

129. Kim JK, Fillmore JJ, Sunshine MJ, Albrecht B, Higashimori T, KimDW, Liu ZX, Soos TJ, Cline GW, O’Brien WR, Littman DR, andShulman GI. PKCtheta knockout mice are protected from fat-inducedinsulin resistance. J Clin Invest 114: 823–827, 2004.

130. King KL, Okere IC, Sharma N, Dyck JR, Reszko AE, McElfresh TA,Kerner J, Chandler MP, Lopaschuk GD, and Stanley WC. Regula-tion of cardiac malonyl-CoA content and fatty acid oxidation duringincreased cardiac power. Am J Physiol Heart Circ Physiol 289: H1033–H1037, 2005.

131. Koonen DP, Glatz JF, Bonen A, and Luiken JJ. Long-chain fatty aciduptake and FAT/CD36 translocation in heart and skeletal muscle. Bio-chim Biophys Acta 1736: 163–180, 2005.

132. Kraegen EW, Sowden JA, Halstead MB, Clark PW, Rodnick KJ,Chisholm DJ, and James DE. Glucose transporters and in vivo glucoseuptake in skeletal and cardiac muscle: fasting, insulin stimulation andimmunoisolation studies of GLUT1 and GLUT4. Biochem J 295: 287–293, 1993.

133. Kudo N, Barr AJ, Barr RL, Desai S, and Lopaschuk GD. High ratesof fatty acid oxidation during reperfusion of ischemic hearts are associ-ated with a decrease in malonyl-CoA levels due to an increase in5�-AMP-activated protein kinase inhibition of acetyl-CoA carboxylase.J Biol Chem 270: 17513–17520, 1995.

134. Kudo N, Gillespie JG, Kung L, Witters LA, Schulz R, Clanachan AS,and Lopaschuk GD. Characterization of 5�AMP activated protein ki-nase activity in the heart and its role in inhibiting acetyl-CoA carboxylaseduring reperfusion following ischemia. Biochim Biophys Acta 1301:67–75, 1996.

135. Kurtz DM, Rinaldo P, Rhead WJ, Tian L, Millington DS, Vockley J,Hamm DA, Brix AE, Lindsey JR, Pinkert CA, O’Brien WE, andWood PA. Targeted disruption of mouse long-chain acyl-CoA dehydro-genase gene reveals crucial roles for fatty acid oxidation. Proc Natl AcadSci USA 95: 15592–15597, 1998.

136. Laughlin MR, Petit WA Jr, Shulman RG and Barrett EJ. Measure-ment of myocardial glycogen synthesis in diabetic and fasted rats. Am JPhysiol Endocrinol Metab 258: E184–E190, 1990.

137. Laybutt DR, Thompson AL, Cooney GJ, and Kraegen EW. Selectivechronic regulation of GLUT1 and GLUT4 content by insulin, glucose,and lipid in rat cardiac muscle in vivo. Am J Physiol Heart Circ Physiol273: H1309–H1316, 1997.

138. Lee GY, Kim NH, Zhao ZS, Cha BS, and Kim YS. Peroxisomal-proliferator-activated receptor alpha activates transcription of the rat

Invited Review

H1502 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 16: Cardiac Metabolism Diabetic Cardiomyopathy

hepatic malonyl-CoA decarboxylase gene: a key regulation of malonyl-CoA level. Biochem J 378: 983–990, 2004.

139. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, andKelly DP. Peroxisome proliferator-activated receptor gamma coactiva-tor-1 promotes cardiac mitochondrial biogenesis. J Clin Invest 106:847–856, 2000.

140. Leone TC, Weinheimer CJ, and Kelly DPA. critical role for theperoxisome proliferator-activated receptor alpha (PPARalpha) in thecellular fasting response: the PPARalpha-null mouse as a model of fattyacid oxidation disorders. Proc Natl Acad Sci USA 96: 7473–7478, 1999.

141. Levak-Frank S, Radner H, Walsh A, Stollberger R, Knipping G,Hoefler G, Sattler W, Weinstock PH, Breslow JL, and Zechner R.Muscle-specific overexpression of lipoprotein lipase causes a severemyopathy characterized by proliferation of mitochondria and peroxi-somes in transgenic mice. J Clin Invest 96: 976–986, 1995.

142. Li H, Wang J, Wilhelmsson H, Hansson A, Thoren P, Duffy J, RustinP, and Larsson NG. Genetic modification of survival in tissue-specificknockout mice with mitochondrial cardiomyopathy. Proc Natl Acad SciUSA 97: 3467–3472, 2000.

143. Li J, Hu X, Selvakumar P, Russell RR 3rd, Cushman SW, HolmanGD, and Young LH. Role of the nitric oxide pathway in AMPK-mediated glucose uptake and GLUT4 translocation in heart muscle. Am JPhysiol Endocrinol Metab 287: E834–E841, 2004.

144. Listenberger LL, Han X, Lewis SE, Cases S, Farese RV Jr, Ory DS,and Schaffer JE. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc Natl Acad Sci USA 100: 3077–3082, 2003.

145. Liu Y, Thornton JD, Cohen MV, Downey JM, and Schaffer SW.Streptozotocin-induced non-insulin-dependent diabetes protects the heartfrom infarction. Circulation 88: 1273–1278, 1993.

146. Lopaschuk GD. Alterations in myocardial fatty acid metabolism con-tribute to ischemic injury in the diabetic. Can J Cardiol 5: 315–320,1989.

147. Lopaschuk GD. Metabolic abnormalities in the diabetic heart. HeartFail Rev 7: 149–159, 2002.

148. Lopaschuk GD and Barr RL. Measurements of fatty acid and carbo-hydrate metabolism in the isolated working rat heart. Mol Cell Biochem172: 137–147, 1997.

149. Lopaschuk GD, Belke DD, Gamble J, Itoi T, and Schonekess BO.Regulation of fatty acid oxidation in the mammalian heart in health anddisease. Biochim Biophys Acta 1213: 263–276, 1994.

150. Lopaschuk GD, Saddik M, Barr R, Huang L, Barker CC, andMuzyka RA. Effects of high levels of fatty acids on functional recoveryof ischemic hearts from diabetic rats. Am J Physiol Endocrinol Metab263: E1046–E1053, 1992.

151. Lteif AA, Mather KJ, and Clark CM. Diabetes and heart disease anevidence-driven guide to risk factors management in diabetes. CardiolRev 11: 262–274, 2003.

152. Luiken JJ, Arumugam Y, Bell RC, Calles-Escandon J, Tandon NN,Glatz JF, and Bonen A. Changes in fatty acid transport and transportersare related to the severity of insulin deficiency. Am J Physiol EndocrinolMetab 283: E612–E621, 2002.

153. Luiken JJ, Arumugam Y, Dyck DJ, Bell RC, Pelsers MM, TurcotteLP, Tandon NN, Glatz JF, and Bonen A. Increased rates of fatty aciduptake and plasmalemmal fatty acid transporters in obese Zucker rats.J Biol Chem 276: 40567–40573, 2001.

154. Luiken JJ, Coort SL, Koonen DP, van der Horst DJ, Bonen A,Zorzano A, and Glatz JF. Regulation of cardiac long-chain fatty acidand glucose uptake by translocation of substrate transporters. PflugersArch 448: 1–15, 2004.

155. Luiken JJ, Coort SL, Willems J, Coumans WA, Bonen A, van derVusse GJ, and Glatz JF. Contraction-induced fatty acid translocase/CD36 translocation in rat cardiac myocytes is mediated through AMP-activated protein kinase signaling. Diabetes 52: 1627–1634, 2003.

156. Luiken JJ, Koonen DP, Willems J, Zorzano A, Becker C, Fischer Y,Tandon NN, Van Der Vusse GJ, Bonen A, and Glatz JF. Insulinstimulates long-chain fatty acid utilization by rat cardiac myocytesthrough cellular redistribution of FAT/CD36. Diabetes 51: 3113–3119,2002.

157. Luiken JJ, Schaap FG, van Nieuwenhoven FA, van der Vusse GJ,Bonen A, and Glatz JF. Cellular fatty acid transport in heart and skeletalmuscle as facilitated by proteins. Lipids 34, Suppl: S169–S175, 1999.

158. Luiken JJ, Turcotte LP, and Bonen A. Protein-mediated palmitateuptake and expression of fatty acid transport proteins in heart giantvesicles. J Lipid Res 40: 1007–1016, 1999.

159. Luiken JJ, van Nieuwenhoven FA, America G, van der Vusse GJ,and Glatz JF. Uptake and metabolism of palmitate by isolated cardiacmyocytes from adult rats: involvement of sarcolemmal proteins. J LipidRes 38: 745–758, 1997.

160. Maki M, Nuutila P, Laine H, Voipio-Pulkki LM, Haaparanta M,Solin O, and Knuuti JM. Myocardial glucose uptake in patients withNIDDM and stable coronary artery disease. Diabetes 46: 1491–1496,1997.

161. Marsin AS, Bertrand L, Rider MH, Deprez J, Beauloye C, VincentMF, Van den Berghe G, Carling D, and Hue L. Phosphorylation andactivation of heart PFK-2 by AMPK has a role in the stimulation ofglycolysis during ischaemia. Curr Biol 10: 1247–1255, 2000.

162. Mazumder PK, O’Neill BT, Roberts MW, Buchanan J, Yun UJ,Cooksey RC, Boudina S, and Abel ED. Impaired cardiac efficiency andincreased fatty acid oxidation in insulin-resistant ob/ob mouse hearts.Diabetes 53: 2366–2374, 2004.

163. McClain DA and Crook ED. Hexosamines and insulin resistance.Diabetes 45: 1003–1009, 1996.

164. McDonagh PF and Hokama JY. Microvascular perfusion and transportin the diabetic heart. Microcirculation 7: 163–181, 2000.

165. Merkel M, Eckel RH, and Goldberg IJ. Lipoprotein lipase: genetics,lipid uptake, and regulation. J Lipid Res 43: 1997–2006, 2002.

166. Merkel M, Loeffler B, Kluger M, Fabig N, Geppert G, PennacchioLA, Laatsch A, and Heeren J. Apolipoprotein AV accelerates plasmahydrolysis of triglyceride-rich lipoproteins by interaction with proteogly-can-bound lipoprotein lipase. J Biol Chem 280: 21553–21560, 2005.

167. Montanari D, Yin H, Dobrzynski E, Agata J, Yoshida H, Chao J, andChao L. Kallikrein gene delivery improves serum glucose and lipidprofiles and cardiac function in streptozotocin-induced diabetic rats.Diabetes 54: 1573–1580, 2005.

168. Monti LD, Landoni C, Setola E, Galluccio E, Lucotti P, Sandoli EP,Origgi A, Lucignani G, Piatti P, and Fazio F. Myocardial insulinresistance associated with chronic hypertriglyceridemia and increasedFFA levels in Type 2 diabetic patients. Am J Physiol Heart Circ Physiol287: H1225–H1231, 2004.

169. Muoio DM, MacLean PS, Lang DB, Li S, Houmard JA, Way JM,Winegar DA, Corton JC, Dohm GL, and Kraus WE. Fatty acidhomeostasis and induction of lipid regulatory genes in skeletal muscles ofperoxisome proliferator-activated receptor (PPAR) alpha knock-out miceEvidence for compensatory regulation by PPAR delta. J Biol Chem 277:26089–26097, 2002.

170. Murray AJ, Anderson RE, Watson GC, Radda GK, and Clarke K.Uncoupling proteins in human heart. Lancet 364: 1786–1788, 2004.

171. Murray AJ, Panagia M, Hauton D, Gibbons GF, and Clarke K.Plasma free fatty acids and peroxisome proliferator-activated receptor{alpha} in the control of myocardial uncoupling protein levels. Diabetes54: 3496–3502, 2005.

172. Nagao M, Parimoo B, and Tanaka K. Developmental, nutritional, andhormonal regulation of tissue-specific expression of the genes encodingvarious acyl-CoA dehydrogenases and alpha-subunit of electron transferflavoprotein in rat. J Biol Chem 268: 24114–24124, 1993.

173. Neely JR, Rovetto MJ, and Oram JF. Myocardial utilization ofcarbohydrate and lipids. Prog Cardiovasc Dis 15: 289–329, 1972.

174. Neitzel AS, Carley AN, and Severson DL. Chylomicron and palmitatemetabolism by perfused hearts from diabetic mice. Am J Physiol Endo-crinol Metab 284: E357–E365, 2003.

175. Newsholme EA and Randle PJ. Regulation of glucose uptake bymuscle. 7 Effects of fatty acids, ketone bodies and pyruvate, and ofalloxan-diabetes, starvation, hypophysectomy and adrenalectomy, on theconcentrations of hexose phosphates, nucleotides and inorganic phos-phate in perfused rat heart. Biochem J 93: 641–651, 1964.

176. Nicholl TA, Lopaschuk GD, and McNeill JH. Effects of free fatty acidsand dichloroacetate on isolated working diabetic rat heart. Am J PhysiolHeart Circ Physiol 261: H1053–H1059, 1991.

177. Nielsen LB, Bartels ED, and Bollano E. Overexpression of apolipopro-tein B in the heart impedes cardiac triglyceride accumulation and devel-opment of cardiac dysfunction in diabetic mice. J Biol Chem 277:27014–27020, 2002.

178. Niu YG, Hauton D, and Evans RD. Utilization of triacylglycerol-richlipoproteins by the working rat heart: routes of uptake and metabolicfates. J Physiol 558: 225–237, 2004.

179. Nuutila P, Knuuti J, Ruotsalainen U, Koivisto VA, Eronen E, TerasM, Bergman J, Haaparanta M, Voipio-Pulkki LM, Viikari J, T.Ronnemaa, U. Wegelius, and H. Yki-Jarvinen. Insulin resistance is

Invited Review

H1503ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 17: Cardiac Metabolism Diabetic Cardiomyopathy

localized to skeletal but not heart muscle in type 1 diabetes. Am J PhysiolEndocrinol Metab 264: E756–E762, 1993.

180. O’Donnell JM, Zampino M, Alpert NM, Fasano MJ, Geenen DL,and Lewandowski ED. Accelerated triacylglycerol turnover kinetics inhearts of diabetic rats include evidence for compartmented lipid storage.Am J Physiol Endocrinol Metab 290: E448–E455, 2006.

181. O’Looney P, Irwin D, Briscoe P, and Vahouny GV. Lipoproteincomposition as a component in the lipoprotein clearance defect inexperimental diabetes. J Biol Chem 260: 428–432, 1985.

182. O’Looney P, Vander Maten M, and Vahouny GV. Insulin-mediatedmodifications of myocardial lipoprotein lipase and lipoprotein metabo-lism. J Biol Chem 258: 12994–13001, 1983.

183. Olson AL and Pessin JE. Transcriptional regulation of the humanGLUT4 gene promoter in diabetic transgenic mice. J Biol Chem 270:23491–23495, 1995.

184. Ostrander DB, Sparagna GC, Amoscato AA, McMillin JB, andDowhan W. Decreased cardiolipin synthesis corresponds with cyto-chrome c release in palmitate-induced cardiomyocyte apoptosis. J BiolChem 276: 38061–38067, 2001.

185. Pacher P, Liaudet L, Soriano FG, Mabley JG, Szabo E, and Szabo C.The role of poly(ADP-ribose) polymerase activation in the developmentof myocardial and endothelial dysfunction in diabetes. Diabetes 51:514–521, 2002.

186. Panagia M, Gibbons GF, Radda GK, and Clarke K. PPARalphaactivation required for decreased glucose uptake and increased suscep-tibility to injury during ischemia. Am J Physiol Heart Circ Physiol 288:H2677–H2683, 2005.

187. Paz K, Hemi R, LeRoith D, Karasik A, Elhanany E, Kanety H, andZick YA. molecular basis for insulin resistance. Elevated serine/threo-nine phosphorylation of IRS-1 and IRS-2 inhibits their binding to thejuxtamembrane region of the insulin receptor and impairs their ability toundergo insulin-induced tyrosine phosphorylation. J Biol Chem 272:29911–29918, 1997.

188. Pennacchio LA, Olivier M, Hubacek JA, Cohen JC, Cox DR, Fru-chart JC, Krauss RM, and Rubin EM. An apolipoprotein influencingtriglycerides in humans and mice revealed by comparative sequencing.Science 294: 169–173, 2001.

189. Pessin JE and Bell GI. Mammalian facilitative glucose transporterfamily: structure and molecular regulation. Annu Rev Physiol 54: 911–930, 1992.

190. Peterson LR, Herrero P, Schechtman KB, Racette SB, WaggonerAD, Kisrieva-Ware Z, Dence C, Klein S, Marsala J, Meyer T, andGropler RJ. Effect of obesity and insulin resistance on myocardialsubstrate metabolism and efficiency in young women. Circulation 109:2191–2196, 2004.

191. Peterson LR, Waggoner AD, Schechtman KB, Meyer T, Gropler RJ,Barzilai B, and Davila-Roman VG. Alterations in left ventricularstructure and function in young healthy obese women: assessment byechocardiography and tissue Doppler imaging. J Am Coll Cardiol 43:1399–1404, 2004.

192. Picano E. Diabetic cardiomyopathy the importance of being earliest.J Am Coll Cardiol 42: 454–457, 2003.

193. Pillarisetti S, Paka L, Sasaki A, Vanni-Reyes T, Yin B, Parthasar-athy N, Wagner WD, and Goldberg IJ. Endothelial cell heparanasemodulation of lipoprotein lipase activity. Evidence that heparan sulfateoligosaccharide is an extracellular chaperone. J Biol Chem 272: 15753–15759, 1997.

194. Pillutla P, Hwang YC, Augustus A, Yokoyama M, Yagyu H,Johnston TP, Kaneko M, Ramasamy R, and Goldberg IJ. Perfusionof hearts with triglyceride-rich particles reproduces the metabolic abnor-malities in lipotoxic cardiomyopathy. Am J Physiol Endocrinol Metab288: E1229–E1235, 2005.

195. Poirier P, Bogaty P, Garneau C, Marois L, and Dumesnil JG.Diastolic dysfunction in normotensive men with well-controlled type 2diabetes: importance of maneuvers in echocardiographic screening forpreclinical diabetic cardiomyopathy. Diabetes Care 24: 5–10, 2001.

196. Poornima IG, Parikh P, and Shannon RP. Diabetic cardiomyopathy:the search for a unifying hypothesis. Circ Res 98: 596–605, 2006.

197. Raev DC. Left ventricular function, and specific diabetic complicationsin other target organs in young insulin-dependent diabetics: an echocar-diographic study. Heart Vessels 9: 121–128, 1994.

198. Ramasamy R, Oates PJ, and Schaefer S. Aldose reductase inhibitionprotects diabetic and nondiabetic rat hearts from ischemic injury. Dia-betes 46: 292–300, 1997.

199. Randle PJ, Garland PB, Hales CN, and Newsholme EA. The glucosefatty-acid cycle. Its role in insulin sensitivity and the metabolic distur-bances of diabetes mellitus. Lancet 1: 785–789, 1963.

200. Redfield MM, Jacobsen SJ, Burnett JC Jr, Mahoney DW, Bailey KR,and Rodeheffer RJ. Burden of systolic and diastolic ventricular dys-function in the community: appreciating the scope of the heart failureepidemic. JAMA 289: 194–202, 2003.

201. Richards MR, Harp JD, Ory DS, and Schaffer JE. Fatty acid transportprotein 1 and long-chain acyl coenzyme A synthetase 1 interact inadipocytes. J Lipid Res 47: 665–672, 2006.

202. Rider MH, Bertrand L, Vertommen D, Michels PA, Rousseau GG,and Hue L. 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase:head-to-head with a bifunctional enzyme that controls glycolysis. Bio-chem J 381: 561–579, 2004.

203. Rodrigues B, Cam MC, Jian K, Lim F, Sambandam N, and Shep-herd G. Differential effects of streptozotocin-induced diabetes on car-diac lipoprotein lipase activity. Diabetes 46: 1346–1353, 1997.

204. Rodrigues B, Cam MC, and McNeill JH. Myocardial substrate metab-olism: implications for diabetic cardiomyopathy. J Mol Cell Cardiol 27:169–179, 1995.

205. Rodrigues B, Cam MC, and McNeill JH. Metabolic disturbances indiabetic cardiomyopathy. Mol Cell Biochem 180: 53–57, 1998.

206. Rui L, Aguirre V, Kim JK, Shulman GI, Lee A, Corbould A, DunaifA, and White MF. Insulin/IGF1 and TNFalpha stimulate phosphoryla-tion of IRS-1 at inhibitory Ser307 via distinct pathways. J Clin Invest107: 181–189, 2001.

207. Rupp H, Elimban V, and Dhalla NS. Modification of myosin isozymesand SR Ca(2�)-pump ATPase of the diabetic rat heart by lipid-loweringinterventions. Mol Cell Biochem 132: 69–80, 1994.

208. Russell LK, Finck BN, and Kelly DP. Mouse models of mitochondrialdysfunction and heart failure. J Mol Cell Cardiol 38: 81–91, 2005.

209. Russell RR 3rd, Li J, Coven DL, Pypaert M, Zechner C, Palmeri M,Giordano FJ, Mu J, Birnbaum MJ, and Young LH. AMP activatedprotein kinase mediates ischemic glucose uptake and prevents postische-mic cardiac dysfunction, apoptosis, and injury. J Clin Invest 114: 495–503, 2004.

210. Saddik M and Lopaschuk GD. Myocardial triglyceride turnover andcontribution to energy substrate utilization in isolated working rat hearts.J Biol Chem 266: 8162–8170, 1991.

211. Saha AK, Vavvas D, Kurowski TG, Apazidis A, Witters LA, ShafrirE, and Ruderman NB. Malonyl-CoA regulation in skeletal muscle: itslink to cell citrate and the glucose-fatty acid cycle. Am J PhysiolEndocrinol Metab 272: E641–E648, 1997.

212. Saito F, Kawaguchi M, Izumida J, Asakura T, Maehara K, andMaruyama Y. Alteration in haemodynamics and pathological changes inthe cardiovascular system during the development of Type 2 diabetesmellitus in OLETF rats. Diabetologia 46: 1161–1169, 2003.

213. Sakamoto J, Barr RL, Kavanagh KM, and Lopaschuk GD. Contri-bution of malonyl-CoA decarboxylase to the high fatty acid oxidationrates seen in the diabetic heart. Am J Physiol Heart Circ Physiol 278:H1196–H1204, 2000.

214. Schaffer JE. Fatty acid transport: the roads taken. Am J Physiol Endo-crinol Metab 282: E239–E246, 2002.

215. Schaffer SW, Ballard-Croft C, Boerth S, and Allo SN. Mechanismsunderlying depressed Na�/Ca2� exchanger activity in the diabeticheart. Cardiovasc Res 34: 129–136, 1997.

216. Scheuermann-Freestone M, Madsen PL, Manners D, Blamire AM,Buckingham RE, Styles P, Radda GK, Neubauer S, and Clarke K.Abnormal cardiac and skeletal muscle energy metabolism in patientswith type 2 diabetes. Circulation 107: 3040–3046, 2003.

217. Schonekess BO. Competition between lactate, and fatty acids as sourcesof ATP in the isolated working rat heart. J Mol Cell Cardiol 29:2725–2733, 1997.

218. Semeniuk LM, Kryski AJ, and Severson DL. Echocardiographicassessment of cardiac function in diabetic db/db and transgenic db/db-hGLUT4 mice. Am J Physiol Heart Circ Physiol 283: H976–H982,2002.

219. Severson DL. Diabetic cardiomyopathy: recent evidence from mousemodels of type 1, and type 2 diabetes. Can J Physiol Pharmacol 82:813–823, 2004.

220. Sharma S, Adrogue JV, Golfman L, Uray I, Lemm J, Youker K,Noon GP, Frazier OH, and Taegtmeyer H. Intramyocardial lipidaccumulation in the failing human heart resembles the lipotoxic rat heart.FASEB J 18: 1692–1700, 2004.

Invited Review

H1504 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 18: Cardiac Metabolism Diabetic Cardiomyopathy

221. Shen X, Zheng S, Metreveli NS, and Epstein PN. Protection of CardiacMitochondria by Overexpression of MnSOD Reduces Diabetic Cardio-myopathy. Diabetes 55: 798–805, 2006.

222. Shen X, Zheng S, Thongboonkerd V, Xu M, Pierce WM Jr. Klein JBand Epstein PN Cardiac mitochondrial damage and biogenesis in achronic model of type 1 diabetes. Am J Physiol Endocrinol Metab 287:E896–E905, 2004.

223. Shipp JC and Murthy VK. The heart and diabetes. Vascular andmetabolic aspects. Adv Myocardiol 2: 81–84, 1980.

224. Sidell RJ, Cole MA, Draper NJ, Desrois M, Buckingham RE, andClarke K. Thiazolidinedione treatment normalizes insulin resistance andischemic injury in the zucker Fatty rat heart. Diabetes 51: 1110–1117,2002.

225. Sowers JR, Epstein M, and Frohlich ED. Diabetes, hypertension, andcardiovascular disease: an update. Hypertension 37: 1053–1059, 2001.

226. Stamler J, Vaccaro O, Neaton JD, and Wentworth D. Diabetes, otherrisk factors, and 12-yr cardiovascular mortality for men screened in theMultiple Risk Factor Intervention Trial. Diabetes Care 16: 434–444,1993.

227. Stanley WC, Lopaschuk GD, and McCormack JG. Regulation ofenergy substrate metabolism in the diabetic heart. Cardiovasc Res 34:25–33, 1997.

228. Stanley WC, Recchia FA, and Lopaschuk GD. Myocardial substratemetabolism in the normal and failing heart. Physiol Rev 85: 1093–1129,2005.

229. Stone PH, Muller JE, Hartwell T, York BJ, Rutherford JD, ParkerCB, Turi ZG, Strauss HW, Willerson JT, Robertson T, et al. Theeffect of diabetes mellitus on prognosis and serial left ventricular func-tion after acute myocardial infarction: contribution of both coronarydisease and diastolic left ventricular dysfunction to the adverse prognosisThe MILIS Study Group. J Am Coll Cardiol 14: 49–57, 1989.

230. Stremmel W. Fatty acid uptake by isolated rat heart myocytes representsa carrier-mediated transport process. J Clin Invest 81: 844–852, 1988.

231. Struthers AD and Morris AD. Screening for and treating left-ventric-ular abnormalities in diabetes mellitus: a new way of reducing cardiacdeaths. Lancet 359: 1430–1432, 2002.

232. Suarez J, Belke DD, Gloss B, Dieterle T, McDonough PM, Kim YK,Brunton LL, and Dillmann WH. In vivo adenoviral transfer of sorcinreverses cardiac contractile abnormalities of diabetic cardiomyopathy.Am J Physiol Heart Circ Physiol 286: H68–H75, 2004.

233. Taegtmeyer H, Hems R, and Krebs HA. Utilization of energy-provid-ing substrates in the isolated working rat heart. Biochem J 186: 701–711,1980.

234. Taegtmeyer H, McNulty P, and Young ME. Adaptation and maladap-tation of the heart in diabetes: Part I: general concepts. Circulation 105:1727–1733, 2002.

235. Tahiliani AG and McNeill JH. Diabetes-induced abnormalities in themyocardium. Life Sci 38: 959–974, 1986.

236. Tani M and Neely JR. Hearts from diabetic rats are more resistant to invitro ischemia: possible role of altered Ca2� metabolism. Circ Res 62:931–940, 1988.

237. Teusink B, Voshol PJ, Dahlmans VE, Rensen PC, Pijl H, Romijn JA,and Havekes LM. Contribution of fatty acids released from lipolysis ofplasma triglycerides to total plasma fatty acid flux and tissue-specificfatty acid uptake. Diabetes 52: 614–620, 2003.

238. Tontonoz P, Hu E, and Spiegelman BM. Stimulation of adipogenesis infibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell79: 1147–1156, 1994.

239. Trost SU, Belke DD, Bluhm WF, Meyer M, Swanson E, andDillmann WH. Overexpression of the sarcoplasmic reticulum Ca(2�)-ATPase improves myocardial contractility in diabetic cardiomyopathy.Diabetes 51: 1166–1171, 2002.

240. Unger RH. Lipotoxic diseases. Annu Rev Med 53: 319–336, 2002.241. Unger RH and Orci L. Lipoapoptosis: its mechanism and its diseases.

Biochim Biophys Acta 1585: 202–212, 2002.242. Utriainen T, Takala T, Luotolahti M, Ronnemaa T, Laine H, Ruot-

salainen U, Haaparanta M, Nuutila P, and Yki-Jarvinen H. Insulinresistance characterizes glucose uptake in skeletal muscle but not in theheart in NIDDM. Diabetologia 41: 555–559, 1998.

243. Vetter R, Rehfeld U, Reissfelder C, Weiss W, Wagner KD, GuntherJ, Hammes A, Tschope C, Dillmann W, and Paul M. Transgenicoverexpression of the sarcoplasmic reticulum Ca2�ATPase improvesreticular Ca2� handling in normal and diabetic rat hearts. FASEB J 16:1657–1659, 2002.

244. Vikramadithyan RK, Hirata K, Yagyu H, Hu Y, Augustus A,Homma S, and Goldberg IJ. Peroxisome proliferator-activated receptoragonists modulate heart function in transgenic mice with lipotoxiccardiomyopathy. J Pharmacol Exp Ther 313: 586–593, 2005.

245. Vincent G, Bouchard B, Khairallah M, and Des Rosiers C. Differ-ential modulation of citrate synthesis and release by fatty acids inperfused working rat hearts. Am J Physiol Heart Circ Physiol 286:H257–H266, 2004.

246. Virag L and Szabo C. The therapeutic potential of poly(ADP-ribose)polymerase inhibitors. Pharmacol Rev 54: 375–429, 2002.

247. Wakasaki H, Koya D, Schoen FJ, Jirousek MR, Ways DK, Hoit BD,Walsh RA, and King GL. Targeted overexpression of protein kinaseCbeta2 isoform in myocardium causes cardiomyopathy. Proc Natl AcadSci USA 94: 9320–9325, 1997.

248. Wall SR and Lopaschuk GD. Glucose oxidation rates in fatty acid-perfused isolated working hearts from diabetic rats. Biochim BiophysActa 1006: 97–103, 1989.

249. Wang P and Chatham JC. Onset of diabetes in Zucker diabetic fatty(ZDF) rats leads to improved recovery of function after ischemia in theisolated perfused heart. Am J Physiol Endocrinol Metab 286: E725–E736, 2004.

250. Wang P, Lloyd SG, Zeng H, Bonen A, and Chatham JC. Impact ofaltered substrate utilization on cardiac function in isolated hearts fromZucker diabetic fatty rats. Am J Physiol Heart Circ Physiol 288: H2102–H2110, 2005.

251. Weiss J and Hiltbrand B. Functional compartmentation of glycolyticversus oxidative metabolism in isolated rabbit heart. J Clin Invest 75:436–447, 1985.

252. Weiss JN and Lamp ST. Glycolysis preferentially inhibits ATP-sensi-tive K� channels in isolated guinea pig cardiac myocytes. Science 238:67–69, 1987.

253. Wilson PW. Diabetes mellitus, and coronary heart disease. EndocrinolMetab Clin North Am 30: 857–881, 2001.

254. Wojtaszewski JF, Jorgensen SB, Hellsten Y, Hardie DG, and RichterEA. Glycogen-dependent effects of 5-aminoimidazole-4-carboxamide(AICA)-riboside on AMP-activated protein kinase and glycogen synthaseactivities in rat skeletal muscle. Diabetes 51: 284–292, 2002.

255. Wojtaszewski JF, MacDonald C, Nielsen JN, Hellsten Y, Hardie DG,Kemp BE, Kiens B, and Richter EA. Regulation of 5�AMPactivatedEA protein kinase activity and substrate utilization in exercising humanskeletal muscle. Am J Physiol Endocrinol Metab 284: E813–E822, 2003.

256. Wold LE and Ren J. Streptozotocin directly impairs cardiac contractilefunction in isolated ventricular myocytes via a p38 map kinase-dependentoxidative stress mechanism. Biochem Biophys Res Commun 318: 1066–1071, 2004.

257. Wu P, Inskeep K, Bowker-Kinley MM, Popov KM, and Harris RA.Mechanism responsible for inactivation of skeletal muscle pyruvatedehydrogenase complex in starvation and diabetes. Diabetes 48: 1593–1599, 1999.

258. Wu P, Sato J, Zhao Y, Jaskiewicz J, Popov KM, and Harris RA.Starvation and diabetes increase the amount of pyruvate dehydrogenasekinase isoenzyme 4 in rat heart. Biochem J 329: 197–201, 1998.

259. Xing Y, Musi N, Fujii N, Zou L, Luptak I, Hirshman MF, GoodyearLJ, and Tian R. Glucose metabolism and energy homeostasis in mousehearts overexpressing dominant negative alpha2 subunit of AMP-acti-vated protein kinase. J Biol Chem 278: 28372–28377, 2003.

260. Yagyu H, Chen G, Yokoyama M, Hirata K, Augustus A, Kako Y, SeoT, Hu Y, Lutz EP, Merkel M, Bensadoun A, Homma S, and Gold-berg IJ. Lipoprotein lipase (LpL) on the surface of cardiomyocytesincreases lipid uptake and produces a cardiomyopathy. J Clin Invest 111:419–426, 2003.

261. Yang J and Holman GD. Insulin and contraction stimulate exocytosis,but increased AMP-activated protein kinase activity resulting from oxi-dative metabolism stress slows endocytosis of GLUT4 in cardiomyo-cytes. J Biol Chem 280: 4070–4078, 2005.

262. Ye G, Metreveli NS, Donthi RV, Xia S, Xu M, Carlson EC, andEpstein PN. Catalase protects cardiomyocyte function in models of type1 and type 2 diabetes. Diabetes 53: 1336–1343, 2004.

263. Ye G, Metreveli NS, Ren J, and Epstein PN. Metallothionein preventsdiabetes-induced deficits in cardiomyocytes by inhibiting reactive oxy-gen species production. Diabetes 52: 777–783, 2003.

264. Yokoyama M, Yagyu H, Hu Y, Seo T, Hirata K, Homma S, andGoldberg IJ. Apolipoprotein B production reduces lipotoxic cardiomy-

Invited Review

H1505ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from

Page 19: Cardiac Metabolism Diabetic Cardiomyopathy

opathy: studies in heart-specific lipoprotein lipase transgenic mouse.J Biol Chem 279: 4204–4211, 2004.

265. Young ME, Goodwin GW, Ying J, Guthrie P, Wilson CR, Laws FA,and Taegtmeyer H. Regulation of cardiac and skeletal muscle malonyl-CoA decarboxylase by fatty acids. Am J Physiol Endocrinol Metab 280:E471–E479, 2001.

266. Young ME, Guthrie PH, Razeghi P, Leighton B, Abbasi S, Patil S,Youker KA, and Taegtmeyer H. Impaired long-chain fatty acid oxida-tion and contractile dysfunction in the obese Zucker rat heart. Diabetes51: 2587–2595, 2002.

267. Young ME, McNulty P, and Taegtmeyer H. Adaptation and maladap-tation of the heart in diabetes: Part II: potential mechanisms. Circulation105: 1861–1870, 2002.

268. Young ME, Patil S, Ying J, Depre C, Ahuja HS, Shipley GL,Stepkowski SM, Davies PJ, and Taegtmeyer H. Uncoupling protein 3transcription is regulated by peroxisome proliferator-activated receptor(alpha) in the adult rodent heart. FASEB J 15: 833–845, 2001.

269. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, andShoelson SE. Reversal of obesity- and diet-induced insulin resistancewith salicylates or targeted disruption of Ikkbeta. Science 293: 1673–1677, 2001.

270. Yue TL, Bao W, Gu JL, Cui J, Tao L, Ma XL, Ohlstein EH, andJucker BM. Rosiglitazone treatment in Zucker diabetic Fatty rats isassociated with ameliorated cardiac insulin resistance and protectionfrom ischemia/reperfusion-induced myocardial injury. Diabetes 54: 554–562, 2005.

271. Zarain-Herzberg A, Yano K, Elimban V, and Dhalla NS. Cardiacsarcoplasmic reticulum Ca(2�)-ATPase expression in streptozotocin-induced diabetic rat heart. Biochem Biophys Res Commun 203: 113–120,1994.

272. Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, BaetensD, Orci L, and Unger RH. Lipotoxic heart disease in obese rats:implications for human obesity. Proc Natl Acad Sci USA 97: 1784–1789,2000.

Invited Review

H1506 ABNORMAL REGULATION OF CARDIAC SUBSTRATE METABOLISM IN DIABETES

AJP-Heart Circ Physiol • VOL 291 • OCTOBER 2006 • www.ajpheart.org

by guest on Novem

ber 20, 2012http://ajpheart.physiology.org/

Dow

nloaded from