Abstract

HomeCirculationVol. 106, No. 16Switching Metabolic Genes to Build a Better Heart Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBSwitching Metabolic Genes to Build a Better Heart Heinrich Taegtmeyer, MD, DPhil Heinrich TaegtmeyerHeinrich Taegtmeyer From the University of Texas Houston Medical School, Department of Internal Medicine, Division of Cardiology, Houston, Tex. Search for more papers by this author Originally published15 Oct 2002https://doi.org/10.1161/01.CIR.0000036760.42319.3FCirculation. 2002;106:2043–2045The heart makes its living by liberating energy from a variety of oxidizable substrates, either simultaneously or vicariously.1 Because of built-in mechanisms that choose the most efficient substrate for a given physiological environment, the heart is a true metabolic omnivore.2 The link between metabolism and function of the heart was discovered by Langendorff3 when he demonstrated that the mammalian heart receives oxygen and nutrients through the coronary circulation and not through the endocardium, as it had been assumed until then. Early investigators also knew already that the heart oxidizes fatty acids and glucose,4 and myocardial fuel economy became a focus of biochemical investigation in the 1960s. Biochemists “discovered” the heart as a convenient bag of enzymes to study muscle metabolism and found that fatty acids suppress glucose oxidation, chiefly at the level of the pyruvate dehydrogenase complex.5 Conversely, we later found that glucose suppresses fatty acid oxidation,1 chiefly at the level of fatty acid entry into the mitochondria. In short, fuel metabolism in the heart is highly regulated, allowing the heart to respond to substrate availability, circulating hormones (such as insulin or catecholamines), coronary flow, and workload by choosing the “right” substrate at the right moment. Unless blood supply is curtailed, as it is the case in ischemia, the heart is never short of fuel to burn.See p 2125Control and RegulationWhat is, then, the principle that underlies substrate switching? As every nutritionist knows, fat has a higher caloric value than carbohydrates; at the same time, the oxidation of carbohydrates results in more efficient energy production than the oxidation of fat. The heart readily oxidizes both substances. Substrate switching in the heart is determined by an interaction of control and regulation of the metabolism of energy providing substrates. According to the metabolic control theory,6 metabolic control is the power to change the state of metabolism in response to an external signal, whereas metabolic regulation defines the way a metabolic system responds to environmental changes. For example, changes in workload control the rate of substrate oxidation, while coordinated changes in enzyme activities regulate substrate metabolism. The terms control and regulation are gaining new importance with the advent of metabolically relevant models of genetic manipulation. There are three ways by which the heart changes its fuel efficiency: first, in response to change in the levels of specific fuels or hormones in the blood; second, in response to change in either the workload or blood supply of the heart; and third, in response to a change in the metabolic make-up of the heart in genetically manipulated models of overexpression or deletion of metabolically relevant proteins (receptors, regulators, transporters, enzymes). The first two examples refer to ways that control cardiac performance; the third example refers to ways that regulate cardiac performance.A key regulator of substrate switching in the heart is thought to be the nuclear receptor peroxisome proliferator-activated receptor alpha (PPARα).7 PPARs are a family of ligand-activated transcription factors within the broad nuclear receptor superfamily that heterodimerize with retinoid X receptors and are subject to a wide array of regulatory mechanisms, including the formation of co-activator complexes and phosphorylation.8 PPARα is deactivated and downregulated with hypertrophy7,9 and heart failure,10 as are the enzymes of fatty acid metabolism controlled by PPARα. It is reasonable to assume that PPARα is a major switch that regulates glucose and fatty acid metabolism.Lastly, there is also a temporal component to the system of control and regulation. Cardiac metabolism adapts both to acute and to chronic changes in its environment. In the first instance, this adaptation is regulated by the activation and inactivation of pathways switching metabolic fuels to the most efficient pathway of energy production for a given environment. For example, the heart responds to an acute increase in workload by oxidizing glycogen, lactate, and glucose.11 In the second instance, metabolic regulation occurs at a transcriptional level, resulting in adaptation and, in the extreme case, maladaptation of the heart. For example, the hypertrophied and atrophied heart switches its genetic make-up to a “fetal” pattern that favors glucose over fatty acids.12Defective Energy Substrate Metabolism in Failing HeartAs the process of metabolic remodeling progresses from adaptation to maladaptation, regulated pathways become misregulated pathways, and the failing heart loses its ability to switch to the most efficient fuel for energy production. Energy transfer is impaired at the level of intermediary metabolism.13 Most importantly, the failing heart develops an impaired capacity to oxidize fatty acids14,15 without a sufficiently high compensatory increase in glucose oxidation.16 At a molecular level, glucose transporter 1 (GLUT 1) expression is severely downregulated in failing human heart,10 as is PPARα. Thus, two key regulators of glucose and of fatty acid metabolism are downregulated in failing heart. While substrate supply remains unimpaired, the heart fails in the midst of plenty.Compensating Defective Energy Substrate MetabolismThe study by Liao et al17 reported in this issue of Circulation demonstrates that the overexpression of one regulatory protein in glucose uptake and metabolism augments glucose uptake. The study sheds new light on the importance of substrate switching and the compensatory increase in glucose oxidation in the stressed heart. The investigators generated mice that overexpress the constitutively expressed glucose transporter GLUT1 in the heart and then induced hypertrophy by pressure-overload on the left ventricle. Pressure overload itself controls function and metabolism of the heart and induces the expression of fetal genes in rodent hearts, and the switching of metabolic genes is part of the heart’s adaptation to sustained increases in workload. Adaptive metabolic switching from fatty acids to glucose can precede the development of hypertrophy18 and may become maladaptive when glucose transporter expression and activity is down-regulated in failing heart.19 However, the present study by Liao et al17 shows that up-regulation of GLUT1 alone can prevent the decline in contractile function by preventing misregulation of glucose metabolism. Whether PPARα plays a role in this system remains to be seen.Several lessons can be learned from the elegant experiments of Liao et al.17 First, the constitutively expressed glucose transporter GLUT1 is a regulator of glucose uptake and metabolism, as demonstrated by the phosphorylation of the glucose tracer analog 2-deoxyglucose. Secondly, the combined action of pressure overload-induced hypertrophy and overexpression of GLUT1 has far-reaching (beneficial) consequences for the performance of the heart. The process involves signaling pathways further downstream at the level of metabolism, growth, and gene expression. At this time, very little is known about the cross-talk between metabolism and the signaling pathways of cell growth and survival in the heart. However, we know already that the activation of fatty acid metabolism in the hypertrophied rat heart results in contractile failure,9 which suggests that metabolic switches are a prerequisite for the successful adaptation of the heart to an altered environment.Glucose, Glycogen, and Other Potential MechanismsWhat are the potential mechanisms for the observed phenomena? The first and most direct explanation is increased glucose uptake and oxidation to make up for the energy deficit of the hypertrophied heart. The second, somewhat more complex explanation would link GLUT1 to the concomitant upregulation of the serine-threonine kinase Akt. This speculation is a testable hypothesis. Akt activation reduces myocardial cell death and induces cardiac hypertrophy20 while raising cardiac glycogen levels and protecting the heart from injury. All these features are also present when GLUT1 is overexpressed in the heart. It is reasonable to assume that overexpression of GLUT1 is accompanied by a host of transcriptional and phenotypic changes similar to the spectrum of changes caused by overexpression of activated Akt in the heart.20,21 A third explanation is linked to increased levels of myocardial glycogen in the present model, suggesting that rates of glucose uptake exceed rates of glycolysis and glucose oxidation. The protective effect of glycogen in the heart probably extends beyond the role of glycogen as an endogenous fuel. For example, in fatigued skeletal muscle, glycogen repletion restores Ca2+ sensitivity and maximum Ca2+-activated force.22 Lastly, high intracellular glucose concentrations have been shown to cause a rise in intracellular [Ca2+] of isolated cardiac myocytes.23 The plot begins to thicken, and it is very tempting to link glucose metabolism to the regulation of Ca2+ homeostasis in the cardiac myocyte.ConclusionIntermediary metabolism, with its complex maze of pathways, has long been regarded as “obsolete” and of little relevance for the understanding of cardiac physiology. The report by Liao et al17 in this issue of Circulation shows exactly the opposite. Increasing the amount and activity of one key regulator of myocardial glucose metabolism prevents the progression from adaptation to maladaptation, from compensatory hypertrophy to heart failure. Transcriptional responses in transgenic models are complex and by no means limited to the gene in question. Much more needs to be learned about these very complex mechanisms, but the basic concept is very simple: targeting metabolic interventions may indeed be the foundation for building a better heart. My teacher Hans Krebs wrote in his memoirs, “When studying a biological phenomenon, it is always important to examine the whole process and not merely a fragment in a damaged tissue.”24 The article by Liao et al17 is proof of this principle, as well as another important concept: metabolism is not an innocent bystander in the control of cardiac gene expression.The opinions expressed in this editorial are not necessarily those of the editors or of the American Heart Association.Work in my laboratory is supported by grants from the National Heart, Lung and Blood Institute and the American Heart Association, National Center. I thank Christopher Wilson, a student in the MD/PhD program at the University of Texas Houston Medical School, for help with the preparation of the manuscript.FootnotesCorrespondence to Heinrich Taegtmeyer, MD, DPhil, University of Texas Houston Medical School, Division of Cardiology, 6431 Fannin, MSB 1.246, Houston TX 77030. E-mail [email protected] References 1 Taegtmeyer H, Hems R, Krebs HA. Utilization of energy providing substrates in the isolated working rat heart. Biochem J. 1980; 186: 701–711.CrossrefMedlineGoogle Scholar2 Taegtmeyer H. Carbohydrate interconversions and energy production. Circulation. 1985; 72: 1–8.CrossrefMedlineGoogle Scholar3 Langendorff O. Untersuchungen am uberlebenden Saugethierherzen. Arch Ges Physiol Menschen Tiere. 1895; 61: 291–332.CrossrefGoogle Scholar4 Evans CL. The metabolism of cardiac muscle. In: Newton W, ed. Evan’s Recent Advances in Physiology. 6 ed. Philadelphia, Pa: Blackinston’s Sons and Co; 1939:157–182.Google Scholar5 Randle PJ, Garland PB, Hales CN, et al. The glucose fatty-acid cycle: its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963; 1: 785–789.CrossrefMedlineGoogle Scholar6 Fell D. Understanding the Control of Metabolism. Miami, Fla: Portland Press; 1997.Google Scholar7 Barger PM, Kelly DP. PPAR signaling in the control of cardiac energy metabolism. Trends Cardiovasc Med. 2000; 10: 238–245.CrossrefMedlineGoogle Scholar8 Kelly DP. Peroxisome proliferator-activated receptor alpha as a genetic determinant of cardiac hypertrophic growth: culprit or innocent bystander? Circulation. 2002; 105: 1025–1027.LinkGoogle Scholar9 Young ME, Laws FA, Goodwin GW, et al. Reactivation of peroxisome proliferator-activated receptor alpha is associated with contractile dysfunction in hypertrophied rat heart. J Biol Chem. 2001; 276: 44390–44395.CrossrefMedlineGoogle Scholar10 Razeghi P, Young ME, Cockrill TC, et al. Downregulation of myocardial myocyte enhancer factor 2C and myocyte enhancer factor 2C regulated gene expression in diabetic patients with non-ischemic heart failure. Circulation. 2002; 106: 407–411.LinkGoogle Scholar11 Goodwin GW, Taylor CS, Taegtmeyer H. Regulation of energy metabolism of the heart during acute increase in heart work. J Biol Chem. 1998; 273: 29530–29539.CrossrefMedlineGoogle Scholar12 Depre C, Shipley GL, Chen W, et al. Unloaded heart in vivo replicates fetal gene expression of cardiac hypertrophy. Nat Med. 1998; 4: 1269–1275.CrossrefMedlineGoogle Scholar13 Razeghi P, Young ME, Alcorn JL, et al. Metabolic gene expression in fetal and failing human heart. Circulation. 2001; 104: 2923–2931.CrossrefMedlineGoogle Scholar14 Sack MN, Rader TA, Park S, et al. Fatty acid oxidation enzyme gene expression is downregulated in the failing heart. Circulation. 1996; 94: 2837–2842.CrossrefMedlineGoogle Scholar15 Osorio JC, Stanley WC, Linke A, et al. Impaired myocardial fatty acid oxidation and reduced protein expression of retinoid X receptor-alpha in pacing-induced heart failure. Circulation. 2002; 106: 606–612.LinkGoogle Scholar16 Davila-Roman VG, Vedala G, Herrero P, et al. Altered myocardial fatty acid and glucose metabolism in idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2002; 40: 271–277.CrossrefMedlineGoogle Scholar17 Liao R, Mohit J, Lei C, et al. Cardiac-specific overexpression of GLUT1 prevents the development of heart failure due to pressure overload in mice. Circulation. 2002; 106: 2124–2130.Google Scholar18 Taegtmeyer H, Overturf ML. Effects of moderate hypertension on cardiac function and metabolism in the rabbit. Hypertension. 1988; 11: 416–426.LinkGoogle Scholar19 Friehs I, Moran AM, Stamm C, et al. Impaired glucose transporter activity in pressure-overload hypertrophy is an early indicator of progression to failure. Circulation. 1999; 100: II187–II193.MedlineGoogle Scholar20 Cook SA, Matsui T, Li L, et al. Transcriptional effects of chronic Akt activation in the heart. J Biol Chem. 2002; 277: 22528–22533.CrossrefMedlineGoogle Scholar21 Matsui T, Li L, Wu J C, et al. Phenotypic spectrum caused by transgenic overexpression of activated Akt in the heart. J Biol Chem. 2002; 277: 22896–22901.CrossrefMedlineGoogle Scholar22 Chin E, Allen D. Effects of reduced muscle glycogen concentration on force, Ca2+ release and contractile protein function in intact mouse skeletal muscle. J Physiol (Lond). 1997; 498: 17–29.CrossrefGoogle Scholar23 Smogorzewski M, Galfayan V, G. MS. High glucose concentration causes a rise in [Ca2+]i of cardiac myocytes. Kidney Int. 1998; 54: 1206–1213.CrossrefMedlineGoogle Scholar24 Krebs HA, Martin A. Hans Krebs: Reminiscences and Reflections. Oxford, UK: Clarendon Press; 1981.Google Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Gorgis N and Desai M (2023) Cardiovascular dysfunction in liver diseases: pediatric perspectives Cardio-Hepatology, 10.1016/B978-0-12-817394-7.00013-9, (247-273), . Ware S (2022) Cardiac Disease in Patients With Mitochondrial Defects, Journal of the American College of Cardiology, 10.1016/j.jacc.2022.08.719, 80:15, (1444-1446), Online publication date: 1-Oct-2022. Silva J and da Costa Martins P (2022) Non-Coding RNAs in the Therapeutic Landscape of Pathological Cardiac Hypertrophy, Cells, 10.3390/cells11111805, 11:11, (1805) Wang X, Zhu X, Jiao S, Qi D, Yu B, Xie G, Liu Y, Song Y, Xu Q, Xu Q, Gonzalez F, Du J, Wang X and Qu A (2021) Cardiomyocyte peroxisome proliferator-activated receptor α is essential for energy metabolism and extracellular matrix homeostasis during pressure overload-induced cardiac remodeling, Acta Pharmacologica Sinica, 10.1038/s41401-021-00743-z, 43:5, (1231-1242), Online publication date: 1-May-2022. Shao-mei W, Li-fang Y and Li-hong W (2022) Traditional Chinese medicine enhances myocardial metabolism during heart failure, Biomedicine & Pharmacotherapy, 10.1016/j.biopha.2021.112538, 146, (112538), Online publication date: 1-Feb-2022. Desai M (2022) Mechanistic insights into the pathophysiology of cirrhotic cardiomyopathy, Analytical Biochemistry, 10.1016/j.ab.2021.114388, 636, (114388), Online publication date: 1-Jan-2022. Angelini A, Saha P, Jain A, Jung S, Mynatt R, Pi X and Xie L (2021) PHDs/CPT1B/VDAC1 axis regulates long-chain fatty acid oxidation in cardiomyocytes, Cell Reports, 10.1016/j.celrep.2021.109767, 37:1, (109767), Online publication date: 1-Oct-2021. Wang N, Xu H, Zhou W, Yang H, Wang J, Ma Z and Gao Y (2021) Aconitine attenuates mitochondrial dysfunction of cardiomyocytes via promoting deacetylation of cyclophilin-D mediated by sirtuin-3, Journal of Ethnopharmacology, 10.1016/j.jep.2020.113765, 270, (113765), Online publication date: 1-Apr-2021. Evans L, Stratton M and Ferguson B (2020) Dietary natural products as epigenetic modifiers in aging-associated inflammation and disease, Natural Product Reports, 10.1039/C9NP00057G, 37:5, (653-676) Eid R, Al-Shraim M, Eleawa S, Zaki M, El-kott A, Eldeen M, Alkhateeb M, Alassiri M and Alderah H (2019) Fish oil protects against corn oil-induced cardiac insulin resistance and left ventricular dysfunction in rats via upregulation of PPAR-β/γ and inhibition of diacylglycerol/PCK axis activation, Journal of Functional Foods, 10.1016/j.jff.2019.03.027, 56, (342-352), Online publication date: 1-May-2019. Sousa Fialho M, Abd Jamil A, Stannard G and Heather L (2019) Hypoxia-inducible factor 1 signalling, metabolism and its therapeutic potential in cardiovascular disease, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2018.09.024, 1865:4, (831-843), Online publication date: 1-Apr-2019. Nguyen T, Shingu Y, Amorim P, Schenkl C, Schwarzer M and Doenst T (2018) GLP-1 Improves Diastolic Function and Survival in Heart Failure with Preserved Ejection Fraction, Journal of Cardiovascular Translational Research, 10.1007/s12265-018-9795-z, 11:3, (259-267), Online publication date: 1-Jun-2018. Uchihashi M, Hoshino A, Okawa Y, Ariyoshi M, Kaimoto S, Tateishi S, Ono K, Yamanaka R, Hato D, Fushimura Y, Honda S, Fukai K, Higuchi Y, Ogata T, Iwai-Kanai E and Matoba S (2017) Cardiac-Specific Bdh1 Overexpression Ameliorates Oxidative Stress and Cardiac Remodeling in Pressure Overload–Induced Heart Failure, Circulation: Heart Failure, 10:12, Online publication date: 1-Dec-2017. Gupta A and Houston B (2017) A comprehensive review of the bioenergetics of fatty acid and glucose metabolism in the healthy and failing heart in nondiabetic condition, Heart Failure Reviews, 10.1007/s10741-017-9623-6, 22:6, (825-842), Online publication date: 1-Nov-2017. Hinton R and Ware S (2017) Heart Failure in Pediatric Patients With Congenital Heart Disease, Circulation Research, 120:6, (978-994), Online publication date: 17-Mar-2017. Sheeran F and Pepe S (2017) Mitochondrial Bioenergetics and Dysfunction in Failing Heart Mitochondrial Dynamics in Cardiovascular Medicine, 10.1007/978-3-319-55330-6_4, (65-80), . Vaillant F, Lauzier B, Ruiz M, Shi Y, Lachance D, Rivard M, Bolduc V, Thorin E, Tardif J and Des Rosiers C (2016) Ivabradine and metoprolol differentially affect cardiac glucose metabolism despite similar heart rate reduction in a mouse model of dyslipidemia, American Journal of Physiology-Heart and Circulatory Physiology, 10.1152/ajpheart.00789.2015, 311:4, (H991-H1003), Online publication date: 1-Oct-2016. Nguyen T, Shingu Y, Amorim P, Schwarzer M and Doenst T (2015) Triheptanoin Alleviates Ventricular Hypertrophy and Improves Myocardial Glucose Oxidation in Rats With Pressure Overload, Journal of Cardiac Failure, 10.1016/j.cardfail.2015.07.009, 21:11, (906-915), Online publication date: 1-Nov-2015. Tham Y, Bernardo B, Ooi J, Weeks K and McMullen J (2015) Pathophysiology of cardiac hypertrophy and heart failure: signaling pathways and novel therapeutic targets, Archives of Toxicology, 10.1007/s00204-015-1477-x, 89:9, (1401-1438), Online publication date: 1-Sep-2015. Koentges C, Pfeil K, Schnick T, Wiese S, Dahlbock R, Cimolai M, Meyer-Steenbuck M, Cenkerova K, Hoffmann M, Jaeger C, Odening K, Kammerer B, Hein L, Bode C and Bugger H (2015) SIRT3 deficiency impairs mitochondrial and contractile function in the heart, Basic Research in Cardiology, 10.1007/s00395-015-0493-6, 110:4, Online publication date: 1-Jul-2015. Desai M, Eblimit Z, Thevananther S, Kosters A, Moore D, Penny D and Karpen S (2014) Cardiomyopathy reverses with recovery of liver injury, cholestasis and cholanemia in mouse model of biliary fibrosis, Liver International, 10.1111/liv.12438, 35:4, (1464-1477), Online publication date: 1-Apr-2015. Li J, Cousin C, Tinkler H, Touhami J, Petit V and Thomas N (2014) Profiling of Nutrient Transporter Expression in Human Stem Cell–Derived Cardiomyocytes Exposed to Tyrosine Kinase Inhibitor Anticancer Drugs Using RBD Ligands, SLAS Discovery, 10.1177/1087057114533724, 19:8, (1185-1192), Online publication date: 1-Sep-2014. Quinones Q, Ma Q, Zhang Z, Barnes B and Podgoreanu M (2014) Organ Protective Mechanisms Common to Extremes of Physiology: A Window through Hibernation Biology, Integrative and Comparative Biology, 10.1093/icb/icu047, 54:3, (497-515), Online publication date: 1-Sep-2014. Mailloux R, Xuan J, McBride S, Maharsy W, Thorn S, Holterman C, Kennedy C, Rippstein P, deKemp R, da Silva J, Nemer M, Lou M and Harper M (2014) Glutaredoxin-2 Is Required to Control Oxidative Phosphorylation in Cardiac Muscle by Mediating Deglutathionylation Reactions, Journal of Biological Chemistry, 10.1074/jbc.M114.550574, 289:21, (14812-14828), Online publication date: 1-May-2014. Afanasiev S, Egorova M, Kondratyeva D, Batalov R and Popov S (2014) Comparative Analysis of Changes of Myocardial Angiogenesis and Energy Metabolism in Postinfarction and Diabetic Damage of Rat Heart, Journal of Diabetes Research, 10.1155/2014/827896, 2014, (1-4), . Wang K, Lim C, McMillen I, Duffield J, Brooks D and Morrison J (2013) Alteration of cardiac glucose metabolism in association to low birth weight: Experimental evidence in lambs with left ventricular hypertrophy, Metabolism, 10.1016/j.metabol.2013.06.013, 62:11, (1662-1672), Online publication date: 1-Nov-2013. Osterholt M, Nguyen T, Schwarzer M and Doenst T (2012) Alterations in mitochondrial function in cardiac hypertrophy and heart failure, Heart Failure Reviews, 10.1007/s10741-012-9346-7, 18:5, (645-656), Online publication date: 1-Sep-2013. Ware S and Towbin J (2013) Nuclear Genes Causing Mitochondrial Cardiomyopathy Mitochondrial Disorders Caused by Nuclear Genes, 10.1007/978-1-4614-3722-2_20, (319-335), . Witteles R, Keu K, Quon A, Tavana H and Fowler M (2012) Dipeptidyl Peptidase 4 Inhibition Increases Myocardial Glucose Uptake in Nonischemic Cardiomyopathy, Journal of Cardiac Failure, 10.1016/j.cardfail.2012.07.009, 18:10, (804-809), Online publication date: 1-Oct-2012. Horman S, Beauloye C, Vanoverschelde J and Bertrand L (2012) AMP-activated Protein Kinase in the Control of Cardiac Metabolism and Remodeling, Current Heart Failure Reports, 10.1007/s11897-012-0102-z, 9:3, (164-173), Online publication date: 1-Sep-2012. Gao F, Ni Y, Luo Z, Liang Y, Yan Z, Xu X, Liu D, Wang J, Zhu S and Zhu Z (2012) Atorvastatin Attenuates TNF-α–induced Increase of Glucose Oxidation Through PGC-1α Upregulation in Cardiomyocytes, Journal of Cardiovascular Pharmacology, 10.1097/FJC.0b013e31824c853c, 59:6, (500-506), Online publication date: 1-Jun-2012. Liu D, Huang L, Wang Y, Wang W, Wehrens X, Belousova T, Abdelrahim M, DiMattia G, Sheikh-Hamad D and Peng T (2012) Human Stanniocalcin-1 Suppresses Angiotensin II-Induced Superoxide Generation in Cardiomyocytes through UCP3-Mediated Anti-Oxidant Pathway, PLoS ONE, 10.1371/journal.pone.0036994, 7:5, (e36994) Gurm G, Danik S, Shoup T, Weise S, Takahashi K, Laferrier S, Elmaleh D and Gewirtz H (2012) 4-[18F]-Tetraphenylphosphonium as a PET Tracer for Myocardial Mitochondrial Membrane Potential, JACC: Cardiovascular Imaging, 10.1016/j.jcmg.2011.11.017, 5:3, (285-292), Online publication date: 1-Mar-2012. Pulakat L, DeMarco V, Ardhanari S, Chockalingam A, Gul R, Whaley-Connell A and Sowers J (2011) Adaptive mechanisms to compensate for overnutrition-induced cardiovascular abnormalities, American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 10.1152/ajpregu.00316.2011, 301:4, (R885-R895), Online publication date: 1-Oct-2011. Ventura-Clapier R, Garnier A, Veksler V and Joubert F (2011) Bioenergetics of the failing heart, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 10.1016/j.bbamcr.2010.09.006, 1813:7, (1360-1372), Online publication date: 1-Jul-2011. Yeih D, Yeh H, Lin L, Tsay Y, Chiang F, Tseng C and Tseng Y (2011) Enhanced activity and subcellular redistribution of myocardial hexokinase after acute myocardial infarction, International Journal of Cardiology, 10.1016/j.ijcard.2009.12.002, 149:1, (74-79), Online publication date: 1-May-2011. Sinha S and Shannon R (2011) Insulin resistance and progression of heart failure in the older adult, Aging Health, 10.2217/ahe.10.92, 7:2, (297-309), Online publication date: 1-Apr-2011. Gewirtz H (2011) Cardiac PET: A Versatile, Quantitative Measurement Tool for Heart Failure Management, JACC: Cardiovascular Imaging, 10.1016/j.jcmg.2010.12.006, 4:3, (292-302), Online publication date: 1-Mar-2011. Chilton R, Wyatt J, Nandish S, Oliveros R and Lujan M (2011) Cardiovascular Comorbidities of Type 2 Diabetes Mellitus: Defining the Potential of Glucagonlike peptide–1-Based Therapies, The American Journal of Medicine, 10.1016/j.amjmed.2010.11.004, 124:1, (S35-S53), Online publication date: 1-Jan-2011. Juang J, de las Fuentes L, Waggoner A, Gu C and Dávila-Román V (2010) Association and interaction of PPAR-complex gene variants with latent traits of left ventricular diastolic function, BMC Medical Genetics, 10.1186/1471-2350-11-65, 11:1, Online publication date: 1-Dec-2010. Pillai V, Sundaresan N, Jeevanandam V and Gupta M (2010) Mitochondrial SIRT3 and heart disease, Cardiovascular Research, 10.1093/cvr/cvq250, 88:2, (250-256), Online publication date: 1-Nov-2010. Camara A, Lesnefsky E and Stowe D (2010) Potential Therapeutic Benefits of Strategies Directed to Mitochondria, Antioxidants & Redox Signaling, 10.1089/ars.2009.2788, 13:3, (279-347), Online publication date: 1-Aug-2010. Bhalla V, Kalogeropoulos A, Georgiopoulou V and Butler J (2010) Serum resistin: physiology, pathophysiology and implications for heart failure, Biomarkers in Medicine, 10.2217/bmm.10.17, 4:3, (445-452), Online publication date: 1-Jun-2010. Doenst T, Pytel G, Schrepper A, Amorim P, Farber G, Shingu Y, Mohr F and Schwarzer M (2009) Decreased rates of substrate oxidation ex vivo predict the onset of heart failure and contractile dysfunction in rats with pressure overload, Cardiovascular Research, 10.1093/cvr/cvp414, 86:3, (461-470), Online publication date: 1-Jun-2010. Desai M, Shabier Z, Taylor M, Lam F, Thevananther S, Kosters A and Karpen S (2010) Hypertrophic cardiomyopathy and dysregulation of cardiac energetics in a mouse model of biliary fibrosis, Hepatology, 10.1002/hep.23585, 51:6, (2097-2107), Online publication date: 1-Jun-2010. Lux T and Taegtmeyer H (2009) All things considered-including glucose control in the ICU, Current Hypertension Reports, 10.1007/s11906-009-0066-x, 11:6, (383-384), Online publication date: 1-Dec-2009. Hue L and Taegtmeyer H (2009) The Randle cycle revisited: a new head for an old hat, American Journal of Physiology-Endocrinology and Metabolism, 10.1152/ajpendo.00093.2009, 297:3, (E578-E591), Online publication date: 1-Sep-2009. Gewirtz H (2009) Regulating myocardial blood flow in health and disease, Current Cardiology Reports, 10.1007/s11886-009-0018-8, 11:2, (117-124), Online publication date: 1-Mar-2009. Sulistio M, Carothers C, Mangat M, Lujan M, Oliveros R and Chilton R (2009) GLP-1 agonist-based therapies: An emerging new class of antidiabetic drug with potential cardioprotective effects, Current Atherosclerosis Reports, 10.1007/s11883-009-0015-9, 11:2, (93-99), Online publication date: 1-Mar-2009. Wong A, ALZadjali M, Choy A and Lang C (2008) Insulin Resistance: A Potential New Target for Therapy in Patients with Heart Failure, Cardiovascular Therapeutics, 10.1111/j.1755-5922.2008.00053.x, 26:3, (203-213), Online publication date: 1-Sep-2008. Witteles R and Fowler M (2008) Insulin-Resistant Cardiomyopathy, Journal of the American College of Cardiology, 10.1016/j.jacc.2007.10.021, 51:2, (93-102), Online publication date: 1-Jan-2008. B

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call